53.?" ‘ an. a! .3 :5 .1. .31.‘ .23.. . an .213 a I..I. MI: c 1 .yv THESIS é? QCCQ This is to certify that the dissertation entitled ANTIESTROGEN RESISTANCE AND C1 PHASE CDK REGULATION IN HUMAN BREAST CANCER CELLS presented by ANDREW JOHN SKILDUM has been accepted towards fulfillment of the requirements for Ph.D. Microbiology degree in Major professor July 29, 2002 Date MS U is an Affirmative Action/Equal Opportunity Institution 0-12771 lJBRARY 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 fill'li” P if 2%“ OCT 2 7 2004 r- : i." ’__‘ 5.0 ill - fl; lzosgz 6/01 c:/CIRCIDateDue.pes-p.15 ANTIESTROGEN RESISTANCE AND G] PHASE CDK REGULATION IN HUMAN BREAST CANCER CELLS By Andrew J. Skildum A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Department of Microbiology and Molecular Genetics 2002 ABSTRACT ANTIESTROGEN RESISTANCE AND 6] PHASE CDK REGULATION IN HUMAN BREAST CANCER CELLS By Andrew J. Skildum Advances in detection and treatment have reduced the mortality of breast cancer over the last decade, but it remains the second most lethal cancer for women in the US. Antiestrogens, including Tamoxifen, have been successful therapies, but acquired antiestrogen resistance during treatment remains a significant barrier. A better understanding of how estrogen and antiestrogen signaling controls proliferation in breast cancer model systems may lead to the development of more efficacious therapies which avoid the problem of antiestrogen resistance. Estrogens and antiestrogens affect cell proliferation at G1 phase, passage through which is governed by the sequential activation of two cyclin dependent kinases, Cdk4 and Cdk2. In this report, we investigate the mechanism(s) by which Cdk activity is regulated by estrogen and the steroidal antiestrogen ICI 182780 in the MCF-7 cell line, a model of estrogen dependent, antiestrogen sensitive human breast cancer. The hypothesis of this dissertation is that defects in the regulation of Cdk4 and Cdk2 activities may be a cause of antiestrogen resistant proliferation in in vitro models of human breast cancer. Cdk activity is inhibited by antiestrogen in MCF-7 cells. Through analysis of Cdk4 activity in estrogen and ICI treated MCF-7 cells by lysate mixing experiments, we find that extracts of ICI-treated cells contain a factor capable of inhibiting the Cdk4 activity present in extracts of estrogen-treated cells, and immunodepletion experiments identify this factor as pZIWAFl/Cip'. We show that pZIW’WVCipl is an important ICI mediated physiological regulator of Cdk4 which can inhibit its activity in the absence of changes in levels of Cdk4’s main regulatory subunit, cyclin D1. To examine the potential contribution of cyclin D1 overexpression to antiestrogen resistance, we engineered a sub-clone of the MCF-7 cell line to conditionally express epitope tagged cyclin D1 upon the addition of a synthetic drug. Cyclin D1 overexpression is sufficient to allow cells arrested in G1 phase by antiestrogen to enter S phase. Ectopic cyclin D1 activates Cdk4 in the presence of antiestrogen but does not sustain long term Cdk4 activity or proliferation, and delays, but does not prevent, antiestrogen mediated induction of the Cdk inhibitor p21“"‘mCipl and inhibition of Cdk4 activity in asynchronous cells. Despite containing a normally functioning estrogen receptor, the MCF-7 derivative LCC9 cell line is able to proliferate in ICI and thus serves as a model of acquired antiestrogen resistance. While in MCF-7 cells Cdk2 and Cdk4 are inactivated by ICI treatment, we show that both kinases remain active in antiestrogen treated LCC9 cells. The Cdk4 from LCC9 cells retains sensitivity to inhibition by lysate from antiestrogen treated MCF—7 cells. Compared to MCF-7, LCC9 cells have elevated levels of cyclin D1 protein and unlike MCF-7 cells, p21wam “9' is not highly regulated by antiestrogen in LCC9 cells. These experiments support our hypothesis that deregulated Cdk activity can contribute to antiestrogen resistance, and suggest that altered signaling pathways upstream of cyclin D1 and p21 may converge to maintain the activities of G1 Cdks in a model of acquired antiestrogen resistance. TABLE OF CONTENTS LIST OF FIGURES .................................................................................. v KEY TO ABBREVIATIONS ...................................................................... vi CHAPTER 1: INTRODUCTION ................................................................. 1 Estrogen and breast cancer ................................................................. 3 Estrogen receptor signaling ................................................................ 3 Control of G1-)S phase transition ....................................................... 7 Antiestrogens .............................................................................. 12 Model systems used to study antiestrogen resistance ................................. 14 G1 checkpoint control in breast cancer cells ......................................... 18 Scope and significance of this study ................................................... 20 References ................................................................................ 23 CHAPTER 2: REGULATION OF CDK4 BY ESTROGEN AND ANTIESTROGEN IN MCF-7 CELLS .................................................................................. 38 Introduction .............................................................................. 40 Materials and Methods .................................................................. 43 Results .................................................................................... 46 Discussion ................................................................................. 61 References .................................................................................. 64 CHAPTER 3: INDUCTION OF CYCLIN D1 IN MCF-7 CELLS SUPPORTS SHORT TERM PROLIFERATION IN THE PRESENCE OF ANTIESTROGEN ............... 69 Introduction .............................................................................. 71 Materials and Methods .................................................................. 74 Results .................................................................................... 77 Discussion .................... 88 References .......... 93 CHAPTER 4: CHARACTERIZATION OF THE LCC9 CELL LINE, AN ANTIESTROGEN RESISTANT DERIVATIVE OF MCF-7 CELLS ..................... 97 Introduction .............................................................................. 99 Materials and Methods .................................................................. 102 Results .................................................................................... 106 Discussion .............................................................................. l 17 References .............................................................................. 121 CHAPTER 5: CONCLUSION ............................................................... 128 Introduction .......................................................................... 129 Significance of major findings ......................................................... 130 Summary and model .................................................................. 136 References .............................................................................. l 3 8 iv LIST OF FIGURES CHAPTER 1 Figure 1: Schematic representation of estrogen mediated transcriptional activation...4 Figure 2: General model for progression through G1 phase of the cell cycle ........... 8 Figure 3: Structure of estrogen receptor ligands ........................................... 11 Figure 4: The development of in vitro models of acquired antiestrogen resistance 1 5 CHAPTER 2 Figure 1: Time course of Cdk4 inhibition by ICI in MCF-7 cells ....................... 47 Figure 2: Time course of Cdk4 activation by E2 in MCF-7 cells ....................... 49 Figure 3: Evidence for an ICI—regulated inhibitor of Cdk4 activity in MCF-7 cells ..51 Figure 4: Depletion of p21 but not p27 abolishes the Cdk4 inhibitory activity in ICI treated MCF-7 cells ............................................................................ 54 Figure 5: Effects of p21 immunodepletion on the Cdk4 inhibitory activity in extracts of ICI treated cells ................................................................................. 56 Figure 6: Cdk4 complex composition in E2 and ICI treated cells .......................... 59 CHAPTER 3 Figure 1: Construction of MCF-7-26-D cells .............................................. 78 Figure 2: Effects of cyclin D1 expression on growth arrested cells ..................... 80 Figure 3: Ectopic cyclin D1 activates Cdk4 in the presence of antiestrogen .......... 82 Figure 4: Expression of ectopic cyclin D1 delays, but does not prevent, inhibition of Cdk4 and Cdk2 activity by antiestrogen .................................................... 84 Figure 5: Expression of exogenous cyclin D1 does not support long term proliferation or Cdk4 activation in the presence of antiestrogen ........................................... 86 CHAPTER 4 Figure l: The LCC9 cell line's antiestrogen resistant phenotype is stable in the absence of selective pressure ............................................................................ 107 Figure 2: LCC9 cells' estrogen receptor is regulated normally by E2 and ICI ........ 109 Figure 3: LCC9's G1 Cdks are not inactivated by antiestrogen .......................... 111 Figure 4: Cdk4 activity in LCC9 is sensitive to the inhibitory factor present in ICI treated MCF-7 cells ............................................................................. 113 Figure 5: LCC9 cells have elevated cyclin D levels and deregulated p21 .............. 115 CHAPTER 5 Figure l: Alterations in regulation of G1 Cdks may lead to antiestrogen resistance .......................................................................... 135 KEY TO ABBREVIATIONS Abbreviations used are: AP, AP 1510. CAK, Cdk activating kinase. Cdk, cyclin dependent kinase. Cde, Cdk inhibitor. CRE, CAMP response element. CSS, charcoal stripped serum. DTT, dithiothreotol. EGF, epidermal growth factor. EGF-R, EGF receptor. ER, estrogen receptor. ERE, ER response element. E2, l7B-estradiol. FBS, fetal bovine serum. F KBP, FK506 binding protein. GST, glutathione-S-transferase. Hours, h. ICI, ICI 182,780. IMEM, improved modified Eagle’s medium. IP, immuno recipitation. Minutes, m. PKA, protein kinase A. PR, progesterone receptor. p16, p16 K43. p21, pZIWAFl/C'p‘. p27, p27 “’1. Rb, retinoblastoma protein. SERM, specific estrogen receptor modulator. 26D, MCF-7-26-D. vi CHAPTER ONE Literature Review Chapter 1: Literature Review ABSTRACT Breast cancer is a serious disease, annually accounting for over 1400 deaths in Michigan and 40,000 deaths nationwide. Advances in detection and treatment have reduced mortality over the last decade, but breast cancer remains the second most lethal cancer for women in the US. Antiestrogens, including tamoxifen, have been successful therapies, but acquired antiestrogen resistance during treatment remains a significant barrier to permanently curing breast cancer. A better understanding of the molecular biology of signaling pathways that control proliferation in breast cancer model systems may lead to more accurate diagnosis and the development of more efficacious therapies. This chapter reviews the mechanisms of estrogens’ action and their link to breast cancer, antiestrogens and their limitations in breast cancer treatment, model systems used to study estrogen signaling, and estrogen’s and antiestrogen’s effects on cell cycle regulators. The conclusion provides an overview of the scope and significance of the research presented in the following chapters. Estrogen and breast cancer Estrogen has long been associated with the development and progression of breast cancer (43). The most biologically active estrogen, 17B-estradiol (E2), is the primary female sex hormone, driving neonatal development of the primary sex organs and further differentiation in the genitals and breast during puberty. In addition, after puberty levels of E2 fluctuate over the 28 day menstrual cycle and during pregnancy. E2 is an organic molecule derived from cholesterol; progesterone and testosterone are intermediates in E2 synthesis. E2 is produced primarily in the ovaries, though it is also produced by the placenta during pregnancy, in the brain and in peripheral adipose tissue. In addition to biological sources of E2, exposure to E2 and its analogs can also come from hormone replacement therapy (HRT) and from organic pollutants and pesticides (15, 36, 111). Although inheritance does play a role in the development of breast cancers in some women, most cases occur sporadically. Estrogen is mitogenic in normal breast tissue, and exposure to estrogen is considered to increase the risk of development of breast cancer. Estrogen exposure may underlie other risk factors such as early puberty and high fat diet. The greatest risk factor in the development of female breast cancer is age, which can be considered the net sum of inherited, physiologic and environmental risk and protective factors integrated over time (17, 85, 86). Estrogen Receptor Signaling E2 acts by binding to a nuclear steroid hormone receptor, the estrogen receptor (ER). There are two isoforrns of ER, the well characterized ERa and the more recently described ERB (106, 159). ERoc is a multidomain protein with hormone binding, dimerization, and DNA binding domains, as well as two transcriptional activation domains (AF-1 and AF-2). E2 binds the ligand binding domain and induces a conformational change leading to receptor homodimerization and binding to DNA upstream of E2 regulated genes at estrogen receptor response elements (EREs). ERa then recruits co-activators via protein — protein interactions with the AF -1 and AF-2 domains (79, 87, 88, 91), stimulating transcription (Figure 1). In addition to the classical mechanism described above, more recent investigations suggest variations on this paradigm. ERa homodimers have been shown to bind and activate promoters with non-consensus EREs (127). ERa and ERB also regulate transcription at promoters containing AP-l sites by contacting Jun and F05 transcription factors and recruiting co-activators; this activity is independent of ER’s DNA binding function. While E2 activates, and tamoxifen inhibits, ERa mediated transcription at AP-l sites, ERB transcription at AP-l sites is inhibited by E2 and activated by tamoxifen, and may account for cell type specific differences in the activities of SERMs (92, 120, 166). Finally, ER splicing variants are often expressed in normal and malignant breast tissue, and may serve as an additional level of regulation for ER mediated transcription (83, 117). ERor also can modulate transcription in the absence of ligand. Work by two groups has shown that the cell cycle regulator cyclin D1 (discussed below) can bind ERa and enhance transcriptional activation of EREs by forming a bridge between ER and transcriptional co-activators (95, 110, 172, 173). The interaction between cyclin D1 and A. .l I C0- EZ E2 activator, @ ERa transcription complex I_E§E_J [E2 regulated gene] E2 co- activator transcription complex lEz regulated geneji Figure 1: Schematic representation of estrogen mediated transcriptional activation. Estrogen (E2) acts by binding to the estrogen receptor (ERa) in the nucleus. E2 binding causes receptor dimerization. A) At E2 regulated promoters containing estrogen response elements (ERE), ERa dimers bind to DNA and activate- transcription by recruiting co-activators. B) At E2 regulated promoters which contain AP-l sites, ERa binds jun and fos transcription factors without contacting DNA and recruits co- activators, enhancing transcription. The ER isoforrn ERB has identical activity as ERa at ERE containing promoters, but opposite activity at AP-l containing promoters: ERB bound to E2 inhibits transcription of AP-l controlled genes. ERoc occurs without Cdk4 activity or E2, although it is inhibited by antiestrogens and inhibitors of cAMP signaling pathways. ERa is a substrate for Erkl and 2, mitogen activated protein kinases (MAPKs), and phosphorylation influences the ligand independent recruitment of transcriptional co-activators by AF-l (10, 46, 82, 158). A major problem in the clinical management of breast cancer is the development of antiestrogen resistance. E2 antagonists such as tamoxifen are used to successfully treat ER positive breast cancer, but many patients’ tumors “evolve” to become resistant to the drug; the resistant tumors are more aggressive and less treatable (108). Recent studies have suggested that overexpression of the receptor tyrosine kinase Her2 and phosphorylated, active MAPKs may correlate with relatively poor response to hormonal therapy in ER positive tumors (54, 128). This suggests that overactive MAPK may cause increased activation and ligand insensitivity of ERa, causing transcription and proliferation in the presence of antiestrogen. MAPK overactivation may also cause increases in non-ER regulated signaling pathways, bypassing blockade of ER by antiestrogen and allowing antiestrogen insensitive growth. These findings point to potentially novel ER signaling pathways in breast cancer cells which may influence the outcome of hormonal therapy. Further examination of these topics is necessary to determine which breast cancer cases are likely to develop antiestrogen resistance and to develop alternative treatment modalities for patients with this class of tumors. Control of G1 95 phase transition The effects of E2 and antiestrogens have been studied extensively in ER positive, hormone responsive breast cancer cell lines, particularly the human breast adenocarcinoma derived MCF-7 cell line. Early work showed that E2 acts to promote progression through the G1 phase of the cell cycle, and that antiestrogens arrest sensitive cells in G1 phase (93, 100, 116, 137, 151). During G1 phase (G is for gap), normal mammalian cells biochemically assess intra- and extracellular signals and “decide” whether or not conditions are appropriate to proliferate. If conditions allow proliferation, cells pass a “restriction point”, afier which they are committed to enter S phase, when cells replicate their genomes. If a cell perceives that conditions for growth are unfavorable, for example due to DNA damage or a lack of a required hormonal context, the cell will arrest in G1 phase until conditions improve. Non-transformed, differentiated cells may exit the cell cycle at G1 to enter G0, a quiescent state. A hallmark of cancer cells is their lack of differentiation and appropriate cell cycle control (39, 146). The G1-)S phase transition in normal cells is controlled by two types of serine/threonine kinases, cyclin dependent kinase-2 (Cdk2) and Cdk4/6. As their name implies, catalytic activity of Cdks requires binding of a cyclin protein at one to one stoichiometry. Cdk4, and the related Cdk6, bind to D type cyclins, including cyclins D1, D2 and D3, although in most cell types, including most human breast cancer cells, cyclin D1 is the major regulatory subunit of Cdk4. Cdk2 can bind either cyclin E or cyclin A. Cyclins are proteins with short half lives whose levels peak at distinct points during the cell cycle (12, 68, 69, 113, 121, 146, 154). Mitogenic signals § a @9/ S hose II 90-»@ ,5 \DNA replication genes §l\ Figure 2: General model for progression through GI phase of the cell cycle. Many mitogens activate signaling pathways that activate Cdk4 by increasing levels of regulatory subunits such as cyclin D1 and/or decreasing levels of Cdk inhibitors such as p21. Active Cdk4 phosphorylates Rb, which blocks its function as an inhibitor of E2F family transcription factors. E2F induced genes include cyclins E and A, regulatory subunits of Cdk2, and other genes involved in DNA synthesis. Cdk2 can phosphorylate Rb, providing for amplification of the mitogenic signal, as well as additional substrates that promote the onset of genome duplication. As shown schematically in Figure 2, active Cdk4 and Cdk2 can phosphorylate the retinoblastoma protein (Rb). Rb is a member of the “pocket protein” family, containing a cysteine rich pocket domain which binds numerous cellular proteins and viral antigens. ’ The Rb pocket also contains numerous Cdk2 and Cdk4 phosphorylation sites. While Rb is the only known substrate for D-type cyclin binding kinases, Cdk2 also has additional substrates directly involved in genome duplication, such as proteins in the origin recognition complex (3, 80, 146). In its unphosphorylated state, Rb binds E2F family transcription factors and represses transcription of E2F regulated genes, resulting in growth arrest (61, 96); certain viral proteins such as adenoviral ElA promote growth by binding to the Rb pocket and displacing E2F (29). Similarly, after phosphorylation by Cdks, hyperphosphorylated Rb no longer binds E2F, allowing transcription of genes that control S phase (167). The expression of cyclins E and A is regulated by E2F, providing amplification of the Rb pathway’s signal (13, 60, 68, 72, 80, 102, 146). Certain mammalian cells, such as the SAOS-2 osteosarcoma cell line, do not contain Rb, and no longer require Cdk4 activity for proliferation (14, 63, 99) but still require Cdk2 activity (113). In addition to cyclin binding, Cdk activity is also regulated by binding of small proteins called Cdk inhibitors (Cdes). There are two families of Cdlds, represented by p16ink4a (p16) and p21wafl/“p' (p21). p16, identified as a tumor suppressor protein, is a specific inhibitor of cyclin D containing complexes, binding to Cdk4/6 and preventing their association with an activating subunit (99, 144, 169). In contrast, p21, identified independently as a Cdk interacting and p53 induced protein, forms ternary complexes with all known Cdk/cyclin complexes (44, 45, 58); this fimction is shared by the related pz7kipl (p27) (130, 157). Both p16 and p21 families have Cdk inhibitory activity, and both are reported to block an activating phosphorylation of Cdk by Cdk-activating kinase (CAK) (7). However, in certain experimental contexts, p21 and p27 may also serve as assembly factors that enhance the formation and activity of cyclin D/Cdk4 complexes (30, 94, 168). Whether p21 family Cdes serve as inhibitors or activators may depend on the stoichiometry of their binding to cyclin/Cdk complexes, although there is some controversy in this area (2, 59, 62, 109). Cde function may be regulated by other mechanisms as well, such as proteolysis or phosphorylation (31, 70, 71, 142). Based on numerous studies, a general model for the regulation of passage from G1 to S phase in normal cells has been developed, and deregulation at various stages in the model are proposed to contribute to cellular transformation and tumorigenesis (60, 68, 69, 98, 139, 146). Many mitogens increase cyclin D1 protein levels, either through increased mRNA synthesis and/or stabilization of the protein. Cyclin D1 binds to Cdk4/6, resulting in an increase in Cdk4/6 kinase activity, Rb phosphorylation and synthesis of cyclins E and A. Increasing the cellular concentration of cyclin D1/Cdk4/6 complexes provides additional binding targets for p21 and/or p27, titrating these Cdes from their inhibitory association with Cdk2 complexes (124, 129). Together, the increased levels of cyclins and the decreased association of Cdes lead to Cdk2 activation and progression to S phase (146). While this model is widely accepted, mitogens and anti-mitogens can also affect progression through G1 in other ways, for example by regulating the levels of Cdes (133, 135), the assembly and nuclear import of cyclin/Cdk 10 17B-estradiol ""(CH2)QSO(CH2)3CFZCF3 ICI 182,780 Figure 3: Structure of estrogen receptor ligands. Estrogen receptor ligands affect ER transcription by binding to the ligand binding domain and inducing conformational changes in the receptor, which result in modifications of ER's DNA and co-activator binding behavior. A) The most biologically active estrogen, 17B-estradiol, is representative of endogenous ER ligands. B) Tamoxifen is a triphenylethylene antiestrogen that acts as a selective estrogen receptor modulator, having antagonist and agonist activities in different tissues. C) The steroidal antiestrogen ICI 182,780 is a pure estrogen receptor antagonist. 11 complexes (41), or the levels and activity of Cdk-activating kinases or phosphatases (51, 81,132). Antiestrogens Because of estrogen’s role as a mitogen in breast tissue, systemic estrogen ablation has long been used for the treatment of breast cancer. Initially this was accomplished surgically through ovariectomy (155). With the development of estrogen antagonists (antiestrogens) in the 19605, E2’s effects were blocked chemically at the sites of action in sensitive tissue. Antiestrogens bind ER’s ligand binding domain and inhibit transcription, but individual compounds have their own unique properties. For this reason antiestrogens are often referred to as selective estrogen receptor modulators (SERMs) (75, 101). There are two broad classes, triphenylethylene antiestrogens and steroidal antiestrogens (Figure 3). The drug tamoxifen, a triphenylethylene compound, has been particularly effective in the treatment of breast cancer when used as a supplement to surgery and/or chemotherapy (143, 163). A current major problem in the treatment of breast cancer is that patients who initially respond to Tamoxifen often develop tumors that have acquired antiestrogen resistance and are independent of E2 (55, 76). Tamoxifen has also recently been recommended for the prevention of breast cancer in women considered at high risk for its development, without apparent concern for the potential implications on the effectiveness of hormonal therapy if prevention fails (22). In addition to concerns with resistance, Tamoxifen treatment causes a small increase in the risk of uterine cancers; this may be due to its agonist activity in uterine l2 tissue (6, 49). Agonist activity may also underlie the surprising finding that Tamoxifen appears to mimic E2’s beneficial effects on bone density (9, 141, 147). For these reasons, other triphenylethylene antiestrogens have been developed which reduce the undesirable side effects of Tamoxifen while maximizing the beneficial effects of E2. For example, Raloxifene, which has been shown to be as effective as Tamoxifen in breast cancer prevention without increasing the risk for uterine cancers, also prevents osteoporosis (40, 160). Continued efforts at developing antiestrogen compounds that are only ER antagonists in the breast should improve treatment outcomes for breast cancer patients. Steroidal antiestrogens resemble E2 more closely than triphenylethylenes. Steroidal antiestrogens have the same four ring cholesterol structure as E2, but with carbohydrate substitutions at various positions. The drug ICI 182780 (ICI) is representative of a class of steroidal antiestrogens which act as “pure” antiestrogens, with no known agonist activity. Upon binding to ER’s ligand binding domain, ICI induces a conformation in ER that does not allow interaction with co-activators and that causes receptor degradation. Although acquired resistance to steroidal antiestrogens occurs clinically and in in vitro models, early trials have shown ICI to be effective in breast cancer patients who had previously failed triphenylmethylene treatment, suggesting that resistance to steroidal and non-steroidal antiestrogens occurs by distinct mechanisms (38, 65,123) Several mechanisms have been proposed to explain the acquisition of antiestrogen resistance. These include a loss of ER or selection of ER negative cells from a heterogeneous tumor population, a change in ERa or ERB function, altered metabolism 13 of antiestrogen, or inappropriate activation of estrogen dependent or independent growth stimulatory signaling pathways; these alterations could occur alone or in combination and could result from genetic lesions or epigenetic changes (33, 34, 64, 73, 122, 136). While ER loss may be a cause of antiestrogen resistance prior to treatment, acquired resistance during the course of treatment occurs in most cases without loss or mutation of ER (74), and antiestrogen metabolism has not been shown to be a likely cause of antiestrogen resistance (115). The composition of co-activators and/or co-repressors interacting with antiestrogen bound ER may influence whether the ligand behaves as a transcriptional activator or repressor at any given promoter, and changes in the makeup of ER containing complexes may underlie the acquisition of antiestrogen resistance (57, 149, 153), although clinical evidence has been lacking thus far (28). Much recent attention has therefore been placed on alterations in signal transduction pathways regulating cellular proliferation. Model systems used to study antiestrogen resistance Despite the importance of E2 and the widespread use of antiestrogens in breast cancer treatment, the mechanisms by which these ER ligands affect proliferation in breast tumor cells remain largely undefined. A better understanding of ER signaling pathways may lead to identification of new therapeutic targets to improve breast cancer treatment. Easily malleable model systems of E2 dependent, antiestrogen sensitive cells are required to examine the molecular biology of ER signaling in breast cancer. Most of our current understanding of E2 and antiestrogen action comes from in vitro studies of cell lines derived from pleural effusions of patients with advanced 14 MCF-7 Estrogen dependent tumor formation LCC1 ICI/Tam sensitive in OVX nude mice ESt rogen Independent ICI/Tam sensitive selection in vitro selection in vitro selection in vitro again st LY1 17018 against tamoxifen against ICI 182780 LY2 Estrogen independent Estrogen dependent Estrogen independent Tam re$1$tant ICI/Tam resistant Tam resistant ICI sensitive Figure 4: The development of in vitro models of acquired antiestrogen resistance. The MCF-7 cell line was derived from a metastatic breast adenocarcinoma. MCF-7 cells require estrogen for proliferation and are inhibited by both triphenylethylene and steroidal antiestrogens. Through selection of MCF-7 cells for tumorigenicity in ovariectomized nude mice, the estrogen independent but antiestrogen sensitive LCCl cell line was derived. Compared to MCF-7, LCCl cells have increased expression of matrix metalloproteinases and nucleophosmin. The LCC2 cell line was developed by selection of LCCl cells for growth in tamoxifen in vitro. LCC2 cells are resistant to triphenylethylenes, but remain sensitive to steroidal antiestrogens, and have increased ratio of anti-apoptotic Bcl-2 to the pro-apoptotic Bax, and have increased expression of TGFB. LCC9 cells were selected from LCCl cells by selection for growth in ICI 182780, and are resistant to both types of antiestrogens; aside from their growth phenotype LCC9 cells remain largely uncharacterized. Independently, LY2 cells were selected directly from MCF-7 cells for resistance to a Raloxifene analog in vitro. LY2 cells show hyperactive MAPK, leading to phosphorylation and degradation of the Cde p27. 15 metastatic breast cancer. Well characterized ER positive human breast cancer cell lines include ZR-75-1 (4, 47) and T47D (145), but by far the most widely studied is MCF-7. The MCF-7 cell line was derived from a 69 year old Caucasian female with metastatic breast adenocarcinoma who had failed previous radiation and hormone therapy (21, 150), although the cell line is hormone sensitive. In order to study the clinically relevant acquisition of antiestrogen resistance, investigators have attempted to recreate the phenomena in vitro (Figure 4). Through selection of MCF-7 cells for estrogen independent tumorigenicity in ovariectomized nude mice, Clarke and colleagues derived the LCC] cell line (23, 32), which is independent of estrogen for growth but retains sensitivity to antiestrogens. Further selection of LCCl in vitro with increasing concentrations of Tamoxifen yielded LCC2, which is estrogen independent, resistant to Tamoxifen yet sensitive to growth inhibition by steroidal antiestrogens (25, 35), supporting the clinical observation that patients who had failed Tamoxifen treatment were still effectively treated by ICI. Selection of LCC] in vitro with increasing concentrations of ICI led to the LCC9 cell line, which is resistant to ICI and to triphenylethylenes, despite not being exposed to them during the selection process (24). Together, these four cell lines represent a model of stepwise acquired antiestrogen resistance in human breast cancer. As is often the case with clinical antiestrogen resistance, ER expression is unchanged in the LCC series of cell lines (24, 25, 156). Likewise, antiestrogen metabolism is unchanged during the acquisition of antiestrogen resistance (77). This suggests that other mechanisms besides loss of ER or altered drug metabolism are responsible for the growth phenotypes of these cell lines. 16 Compared with MCF-7 cells, estrogen independent LCCl cells show increased metastatic potential, correlating with increased matrix metalloproteinase expression (138). LCCl cells also have higher levels of nucleophosmin, an inhibitor of interferon regulatory factor-1 (IRF -1), a tumor suppressing transcription factor involved in growth arrest (90, 148), which may contribute to an increased growth potential in the absence of hormone signaling. The tamoxifen resistant LCC2 cells are reported to have increased levels of TGF- [3, a peptide hormone that inhibits the proliferation of normal breast tissue and early stage breast cancer cells, but is often overexpressed and mitogenic in advanced breast cancer (8, 140). LCC2 cells have also been shown to have elevated Bel-2 and decreased Bax expression (97). The pro-apoptotic Bax forms homodimers and heterodimers with Bel-2, and when Bcl-2 - Bax dimers predominate, apoptosis is inhibited in cells (114). Tamoxifen is reported to cause death of breast cancer cells by apoptosis (26, 119), and the LCC2 cell line’s elevated Bel-2 to Bax ratio may increase their growth potential in tamoxifen by making them less sensitive to normal apoptotic pathways. LCC2 cells are also reported to have increased levels of an ERa splice variant, and altered nuclear matrix protein organization compared to their tamoxifen sensitive progenitors (8, 89, 97, 131), although the possible role of these changes in LCC2 cells’ tamoxifen resistant phenotype is not clear. As of the preparation of this manuscript, no reports describe potential mechanisms for the phenotype of the steroidal antiestrogen resistant LCC9 cell line. In contrast to the stepwise selection used to generate the LCC model of acquired antiestrogen resistance, another antiestrogen resistant derivative of MCF-7, the LY2 cell line, was selected directly for in vitro resistance to a Raloxifene analog (20). LY2 cells 17 retain wild type ER yet, compared to MCF-7, LY2 show deregulated E2 mediated expression of the transcription factor P82, but not the progesterone receptor (PR) (37, 107). This cell line has been shown to have hyperactive MAPK signaling leading to hyperphosphorylation and degradation of p27 a, and treatment with specific MAPK- kinase inhibitors restores antiestrogen sensitivity (42). It is apparent that multiple changes accompany the development of estrogen independence and antiestrogen resistance in the above models of human breast cancer. However, the precise molecular determinants of antiestrogen resistance remain undefined. Further study of these and novel models of breast cancer progression may reveal commonalities that will suggest new therapeutic strategies that avoid or delay the development of antiestrogen resistance. GI checkpoint regulation in breast cancer cells The important roles of cyclins and Cdkls in regulating cell proliferation prompted investigation into their expression in primary breast tumors and correlation with other parameters, such as clinical outcomes and ER expression. Approximately 60% of breast tumors are ER positive, and although ER expression correlates with a more benign tumor grade, endocrine therapy achieves complete remission in only one half of patients with ER positive cancer (1, 50, 56, 118). Cyclin D1 protein is overexpressed in one third of breast cancers, and in a third of these cases, the cyclin D1 gene, CCNDl, located on chromosome 11q13, is amplified (48, 53, 78, 126, 171). Cyclin D1 amplification and protein overexpression correlate positively with ERa expression in breast cancer (48, 66, 152, 161 , 171). Although cyclin 18 D1 overexpression has been consistently reported in primary tumors, reports of its prognostic significance vary considerably. The overexpression of cyclin D1 has been reported to improve relapse free survival (18, 125), to be predictive of poor prognosis if in combination with positive immunostaining for ER, epidermal growth factor, or Rb (84, 104), and to have no correlation with tumor grade or prognosis (152, 161). These results suggest that in breast tumors, other factors influence the effects of cyclin D1 overexpression on the course of disease. In addition to cyclin D1, Cdkls have been investigated in primary breast tumors. Although ofien mutated in other cancers, p16 mutation appears to be infrequent in breast cancer, although changes in p16 expression have been reported due to either homozygous deletion or alterations in promoter methylation, and overexpression of pl 6 is associated with increased tumor malignancy (16, 19, 67, 105). p21 mutation appears infrequently in breast tumors as well, although one point mutated p21 gene that encodes a mutant p21 capable of binding, but not inhibiting, Cdk complexes was isolated from primary breast tumors (1 l, 103). Positive staining for p21 correlates with ER and p53 expression, lower tumor grade, and improved clinical outcomes in breast cancer patients (112, 162). These results suggest that in primary breast cancer, maintenance of mechanisms which normally regulate the activities of Cdk4 and Cdk2 is linked to maintenance of ER and to less advanced disease. Based on these observations, E2 and antiestrogens’ effects on Cdk regulation of the G1 -)S phase transition were investigated further using in vitro models of hormone sensitive breast cancer such as the MCF-7 cell line. Like many cancer derived cell lines, the MCF-7 cell line has inherent defects in cell cycle regulation, including deletion of 19 p16 and constitutively expressed cyclin E (170), however other aspects of E2’s mitogenic influence on MCF-7’s cell cycle machinery resemble normal cells (Figure 2). Others have reported that antiestrogen treatment of MCF-7 cells decreases cyclin D1 and cyclin A protein levels, Cdk4 and Cdk2 activity and Rb phosphorylation, and increases Cde levels (27, 52, 129, 164, 165). All of these effects are reversed upon removal of antiestrogen and replacement with estrogen (5, 51, 134). Although both total Cdk2 and cyclin E/Cdk2 activities are highly regulated, cyclin B protein is expressed at constant levels in estrogen and antiestrogen treated MCF-7 cells. The lack of activity of cyclin E/Cdk2 complexes in the presence of antiestrogen is believed to be due to inhibition by p21 and/or p27 in the complex. Cyclin E/Cdk2 activation after estrogen treatment may therefore be a result of decreased levels of these inhibitors and/or sequestration of p21 or p27 into newly formed cyclin D1/Cdk4 complexes (129, 134). These observations suggest that E2 and antiestrogens influence proliferation by signaling through ER to modulate Cdk activity through multiple mechanisms. Disruption of one or more of these mechanisms may account for the acquisition of antiestrogen resistance in breast cancer cells. Further study to address the precise function of cyclins and Cdkls in ER mediated proliferation, as well as to define ER’s signaling pathways upstream of Cdks, is warranted. Scope and significance of this study Acquired antiestrogen resistance is a major problem in the management of breast cancer, and antiestrogens exert their antiproliferative effects in sensitive cells by blocking the activation of G1 phase Cdks. The hypothesis of this Dissertation is that alterations in 20 the regulation of Cdk4 and/or Cdk2 activities are responsible for acquired resistance to the steroidal antiestrogen ICI in in vitro models of human breast cancer. Work described in the subsequent chapters addresses this hypothesis using three approaches: 1) We attempted to identify key mediators of antiestrogen’s antiproliferative effects in the ICI sensitive MCF-7 human breast cancer cell line (Chapter 2). We found that Cdk4 activity can be regulated by E2 and ICI independently of cyclin D levels, and that the Cde p21 is responsible for inhibiting Cdk4 after ICI treatment. 2) MCF-7 cells were engineered to conditionally express cyclin D1, a component of the Rb pathway implicated in ER signaling, and the effects on hormonal responses were tested (Chapter 3). Short term ICI resistance was achieved by overexpressing cyclin D1 in MCF-7 cells; ectopic cyclin D1 activated and co-precipitated with endogenous Cdk4. Cyclin D1 delayed, but did not prevent, ICI’s upregulation of p21, and cyclin D1 overexpression was not sufficient to allow long term proliferation in ICI. 3) The composition and kinase activities of Cdk4 and Cdk2 complexes observed after E2 and ICI treatment of MCF-7 cells were compared with those observed in an ICI resistant MCF-7 derivative, LCC9 (Chapter 4). While Cdk4 and Cdk2 activities were inhibited by ICI in MCF-7 cells, in LCC9 the Cdks remain active, although LCC9’s Cdk4 remained sensitive to inhibition by p21. LCC9 cells express higher levels of cyclin D than MCF-7, and their p21 is not upregulated by ICI. Based on the results of these studies, we propose that the proliferative potential of hormone sensitive breast cancer cells depends on a positive factor, cyclin D1 , and a negative factor, p21, the balance of which determine the activity of G1 Cdks (see model Chapter 5, Figure 1). Expression of ectopic cyclin D1 leads to Cdk4 activation and 21 proliferation in the presence of antiestrogen, and antiestrogen resistant LCC9 cells have overexpressed cyclin D1 and unregulated p21, although the general function of their Rb pathway appears intact. The hypothesis that disruption of the Rb pathway can cause antiestrogen resistance is thus accepted, with the qualification that in our model of acquired antiestrogen resistance, the disruption(s) appears to be caused by factors upstream of cyclin D1 and p21 expression rather than in cyclin D1 and p21 themselves. Future studies will examine the control of cyclin D1 and p21 expression by ER ligands to assess its impact on the development of antiestrogen resistance. 22 References 10. 11. Adami, H. 0., S. Graffman, A. Lindgren, and J. Sallstrom. 1985. Prognostic implication of estrogen receptor content in breast cancer. Breast Cancer Res Treat 5:293-300. Adkins, J. N., and K. J. Lumb. 2000. Stoichiometry of cyclin A-cyclin-dependent kinase 2 inhibition by p21Cip1/Waf1. Biochemistry 39:13925-30. Akiyama, T., T. Ohuchi, S. Sumida, K. Matsumoto, and K. Toyoshima. 1992. Phosphorylation of the retinoblastoma protein by cdk2. Proc Natl Acad Sci U S A 89:7900-4. Allegra, J. C., and M. E. Lippman. 1978. Growth of a human breast cancer cell line in serum-free hormone- supplemented medium. Cancer Res 38:3823-9. Altucci, L., R. Addeo, L. Cicatiello, S. Dauvois, M. G. Parker, M. Truss, M. Beato, V. Sica, F. Bresciani, and A. Weisz. 1996. 17beta-Estradiol induces cyclin D1 gene transcription, p36D1-p34cdk4 complex activation and p105Rb phosphorylation during mitogenic stimulation of G(l)-arrested human breast cancer cells. Oncogene 12:2315-24. Andersson, M., H. H. Storm, and H. T. Mouridsen. 1991. Incidence of new primary cancers after adjuvant tamoxifen therapy and radiotherapy for early breast cancer. J Natl Cancer Inst 83: 1013-7. Aprelikova, 0., Y. Xiong, and E. T. Liu. 1995. Both p16 and p21 families of cyclin-dependent kinase (CDK) inhibitors block the phosphorylation of cyclin- dependent kinases by the CDK- activating kinase. J Biol Chem 270: 18195-7. Arteaga, C. L., K. M. Koli, T. C. Dugger, and R. Clarke. 1999. Reversal of tamoxifen resistance of human breast carcinomas in vivo by neutralizing antibodies to transforming grth factor-beta. J Natl Cancer Inst 91 :46-53. Assikis, V. J., and V. C. Jordan. 1997. Risks and benefits of tamoxifen therapy. Oncology (Huntingt) l 1:21-3. Atanaskova, N., V. G. Keshamouni, J. S. Krueger, J. A. Schwartz, F. Miller, and K. B. Reddy. 2002. MAP kinase/estrogen receptor cross-talk enhances estrogen- mediated signaling and tumor growth but does not confer tamoxifen resistance. Oncogene 21 :4000-8. Balbin, M., G. J. Hannon, A. M. Pendas, A. A. Ferrando, F. Vizoso, A. Fueyo, and C. Lopez-Otin. 1996. Functional analysis of a p21WAFl,CIP1,SDII mutant (Arg94 --> Trp) identified in a human breast carcinoma. Evidence that the mutation impairs the ability of p21 to inhibit cyclin-dependent kinases. J Biol Chem 271 :15782-6. 23 12. 13. 14. 15. 16. 17. 18. 19. 20. 21. 22. 23. 24. Baldin, V., J. Lukas, M. J. Marcote, M. Pagano, and G. Draetta. 1993. Cyclin D1 is a nuclear protein required for cell cycle progression in G1. Genes Dev 7:812- 21. Bates, S., L. Bonetta, D. MacAllan, D. Parry, A. Holder, C. Dickson, and G. Peters. 1994. CDK6 (PLSTIRE) and CDK4 (PSK-J3) are a distinct subset of the cyclin— dependent kinases that associate with cyclin D1. Oncogene 9:71 -9. Bates, S., D. Parry, L. Bonetta, K. Vousden, C. Dickson, and G. Peters. 1994. Absence of cyclin D/cdk complexes in cells lacking functional retinoblastoma protein. Oncogene 9: 1633-40. Bergkvist, L., and I. Persson. 1996. Hormone replacement therapy and breast cancer. A review of current knowledge. Drug Saf 15:360-70. Bems, E. M., J. G. Klijn, M. Smid, I. L. van Staveren, N. A. Gruis, and J. A. F oekens. 1995. Infrequent CDKN2 (MTSl/p16) gene alterations in human primary breast cancer. Br J Cancer 72:964-7. Bernstein, L., and R. K. Ross. 1993. Endogenous hormones and breast cancer risk. Epidemiol Rev 15:48-65. Bieche, 1., M. Olivi, C. Nogues, M. Vidaud, and R. Lidereau. 2002. Prognostic value of CCNDl gene status in sporadic breast tumours, as determined by real- time quantitative PCR assays. Br J Cancer 86:580—6. Bisogna, M., J. E. Calvano, G. H. Ho, 1. Orlow, C. Cordon-Cardo, P. I. Borgen, and K. J. Van Zee. 2001. Molecular analysis of the IN K4A and INK4B gene loci in human breast cancer cell lines and primary carcinomas. Cancer Genet Cytogenet 125:131-8. Bronzert, D. A., G. L. Greene, and M. E. Lippman. 1985. Selection and characterization of a breast cancer cell line resistant to the antiestrogen LY 117018. Endocrinology 117:1409-17. Brooks, S. C., E. R. Locke, and H. D. Soule. 1973. Estrogen receptor in a human cell line (MCF-7) from breast carcinoma. J Biol Chem 248:6251-3. Brown, P. H., and S. M. Lippman. 2000. Chemoprevention of breast cancer. Breast Cancer Res Treat 62:1-17. Brunner, N., V. Boulay, A. F ojo, C. E. Freter, M. E. Lippman, and R. Clarke. 1993. Acquisition of hormone-independent growth in MCF-7 cells is accompanied by increased expression of estrogen-regulated genes but without detectable DNA amplifications. Cancer Res 53:283-90. Brunner, N., B. Boysen, S. Jirus, T. C. Skaar, C. Holst-Hansen, J. Lippman, T. Frandsen, M. Spang-Thomsen, S. A. Fuqua, and R. Clarke. 1997. MCF7/LCC9: 24 25. 26. 27. 28. 29. 30. 31. 32. an antiestrogen-resistant MCF-7 variant in which acquired resistance to the steroidal antiestrogen ICI 182,780 confers an early cross-resistance to the nonsteroidal antiestrogen tamoxifen. Cancer Res 57:3486-93. Brunner, N., T. L. Frandsen, C. Holst-Hansen, M. Bei, E. W. Thompson, A. E. Wakeling, M. E. Lippman, and R. Clarke. 1993. MCF7/LCC2: a 4- hydroxytamoxifen resistant human breast cancer variant that retains sensitivity to the steroidal antiestrogen ICI 182,780. Cancer Res 53:3229-32. Bursch, W., A. Ellinger, H. Kienzl, L. Torok, S. Pandey, M. Sikorska, R. Walker, and R. S. Hermann. 1996. Active cell death induced by the anti-estrogens tamoxifen and ICI 164 384 in human mammary carcinoma cells (MCF-7) in culture: the role of autophagy. Carcinogenesis 17:1595-607. Carroll, J. S., O. W. Prall, E. A. Musgrove, and R. L. Sutherland. 2000. A pure estrogen antagonist inhibits cyclin E-Cdk2 activity in MCF-7 breast cancer cells and induces accumulation of p1 30-E2F4 complexes characteristic of quiescence. J Biol Chem 275:38221-9. Chan, C. M., A. E. Lykkesfeldt, M. G. Parker, and M. Dowsett. 1999. Expression of nuclear receptor interacting proteins TIP -1 , SUG-l , receptor interacting protein 140, and corepressor SMRT in tamoxifen- resistant breast cancer. Clin Cancer Res 5:3460-7. Chellappan, S., V. B. Kraus, B. Kroger, K. Munger, P. M. Howley, W. C. Phelps, and J. R. Nevins. 1992. Adenovirus ElA, simian virus 40 tumor antigen, and human papillomavirus E7 protein share the capacity to disrupt the interaction between transcription factor E2F and the retinoblastoma gene product. Proc Natl Acad Sci U S A 89:4549—53. Cheng, M., P. Olivier, J. A. Diehl, M. Fero, M. F. Roussel, J. M. Roberts, and C. J. Sherr. 1999. The p21(Cip1) and p27(Kip1) CDK 'inhibitors' are essential activators of cyclin D-dependent kinases in murine fibroblasts. Embo J 18:1571- 83. Chiarle, R., L. M. Budel, J. Skolnik, G. Frizzera, M. Chilosi, A. Corato, G. Pizzolo, J. Magidson, A. Montagnoli, M. Pagano, B. Macs, C. De Wolf-Peeters, and G. Inghirami. 2000. Increased proteasome degradation of cyclin-dependent kinase inhibitor p27 is associated with a decreased overall survival in mantle cell lymphoma. Blood 95:61 9-26. Clarke, R., N. Brunner, B. S. Katzenellenbogen, E. W. Thompson, M. J. Norman, C. Koppi, S. Paik, M. E. Lippman, and R. B. Dickson. 1989. Progression of human breast cancer cells from honnone-dependent to hormone-independent growth both in vitro and in vivo. Proc Natl Acad Sci U S A 86:3649-53. 25 33. 34. 35. 36. 37. 38. 39. 40. 41. 42. 43. 44. Clarke, R., T. C. Skaar, K. B. Bouker, N. Davis, Y. R. Lee, J. N. Welch, and F. Leonessa. 2001. Molecular and pharmacological aspects of antiestrogen resistance. J Steroid Biochem Mol Biol 76:71-84. Clarke, R., E. W. Thompson, F. Leonessa, J. Lippman, M. McGarvey, T. L. Frandsen, and N. Brunner. 1993. Hormone resistance, invasiveness, and metastatic potential in breast cancer. Breast Cancer Res Treat 24:227-39. Coopman, P., M. Garcia, N. Brunner, D. Derocq, R. Clarke, and H. Rochefort. 1994. Anti-proliferative and anti-estrogenic effects of ICI 164,3 84 and ICI 182,780 in 4-OH-tamoxifen-resistant human breast-cancer cells. Int J Cancer 56:295-300. Davidson, N. E. 1998. Environmental estrogens and breast cancer risk. Curr Opin Oncol 10:475-8. Davidson, N. E., D. A. Bronzert, P. Chambon, E. P. Gelmann, and M. E. Lippman. 1986. Use of two MCF-7 cell variants to evaluate the growth regulatory potential of estrogen-induced products. Cancer Res 46:1904-8. DeFriend, D. J ., A. Howell, R. I. Nicholson, E. Anderson, M. Dowsett, R. E. Manse], R. W. Blarney, N. J. Bundred, J. F. Robertson, C. Saunders, and et al. 1994. Investigation of a new pure antiestrogen (ICI 182780) in women with primary breast cancer. Cancer Res 54:408-14. DelSal, G., M. Loda, and M. Pagano. 1996. Cell cycle and cancer: critical events at the G1 restriction point. Crit Rev Oncog 7: 127-42. Dickler, M. N., and L. Norton. 2001. The MORE trial: multiple outcomes for raloxifene evaluation--breast cancer as a secondary end point: implications for prevention. Ann N Y Acad Sci 949:134-42. Diehl, J. A., and C. J. Sherr. 1997. A dominant-negative cyclin D1 mutant prevents nuclear import of cyclin- dependent kinase 4 (CDK4) and its phosphorylation by CDK-activating kinase. Mol Cell Biol 17:7362-74. Donovan, J. C., A. Milic, and J. M. Slingerland. 2001. Constitutive MEK/MAPK activation leads to p27(Kip1) deregulation and antiestrogen resistance in human breast cancer cells. J Biol Chem 276:40888-95. Edwards, D. P., and W. L. McGuire. 1982. Estrogen action in human breast cancer (review). Anticancer Res 2:297-308. el-Deiry, W. S., J. W. Harper, P. M. O'Connor, V. E. Velculescu, C. E. Canman, J. Jackman, J. A. Pietenpol, M. Burrell, D. E. Hill, Y. Wang, and et a1. 1994. WAF l/CIPl is induced in p53-mediated G1 arrest and apoptosis. Cancer Res 54:1169-74. 26 45. 46. 47. 48. 49. 50. 51. 52. 53. 54. el-Deiry, W. S., T. Tokino, V. E. Velculescu, D. B. Levy, R. Parsons, J. M. Trent, D. Lin, W. E. Mercer, K. W. Kinzler, and B. Vogelstein. 1993. WAF 1, a potential mediator of p53 tumor suppression. Cell 75:817-25. Endoh, H., K. Maruyama, Y. Masuhiro, Y. Kobayashi, M. Goto, H. Tai, J. Yanagisawa, D. Metzger, S. Hashimoto, and S. Kato. 1999. Purification and identification of p68 RNA helicase acting as a transcriptional coactivator specific for the activation fimction 1 of human estrogen receptor alpha. Mol Cell Biol 19:5363 -72. Engel, L. W., N. A. Young, T. S. Tralka, M. E. Lippman, S. J. O'Brien, and M. J. Joyce. 1978. Establishment and characterization of three new continuous cell lines derived from human breast carcinomas. Cancer Res 38:3352-64. F antl, V., M. A. Richards, R. Smith, G. A. Lammie, G. Johnstone, D. Allen, W. Gregory, G. Peters, C. Dickson, and D. M. Barnes. 1990. Gene amplification on chromosome band 11q13 and oestrogen receptor status in breast cancer. Eur J Cancer 26:423-9. Fisher, B., J. P. Costantino, C. K. Redmond, E. R. Fisher, D. L. Wickerham, and W. M. Cronin. 1994. Endometrial cancer in tamoxifen-treated breast cancer patients: findings from the National Surgical Adjuvant Breast and Bowel Project (NSABP) B-14. J Natl Cancer Inst 86:527-37. Fisher, B., E. R. Fisher, C. Redmond, and A. Brown. 1986. Tumor nuclear grade, estrogen receptor, and progesterone receptor: their value alone or in combination as indicators of outcome following adjuvant therapy for breast cancer. Breast Cancer Res Treat 7:147-60. Foster, J. S., D. C. Henley, A. Bukovsky, P. Seth, and J. Wimalasena. 2001. Multifaceted regulation of cell cycle progression by estrogen: regulation of Cdk inhibitors and Cdc25A independent of cyclin D1-Cdk4 function. Mol Cell Biol 21:794-810. Foster, J. S., and J. Wimalasena. 1996. Estrogen regulates activity of cyclin- dependent kinases and retinoblastoma protein phosphorylation in breast cancer cells. Mol Endocrinol 10:488-98. Frierson, H. F., Jr., M. J. Gaffey, L. R. Zukerberg, A. Arnold, and M. E. Williams. 1996. Immunohistochemical detection and gene amplification of cyclin D1 in mammary infiltrating ductal carcinoma. Mod Pathol 9:725-30. Gee, J. M., J. F. Robertson, 1. 0. Ellis, and R. I. Nicholson. 2001. Phosphorylation of ERK1/2 mitogen-activated protein kinase is associated with poor response to anti-hormonal therapy and decreased patient survival in clinical breast cancer. Int J Cancer 95:247-54. 27 55. 56. 57. 58. 59. 60. 61. 62. 63. 64. 65. 66. 67. Geisler, J ., and P. E. Lonning. 2001. Resistance to endocrine therapy of breast cancer: recent advances and tomorrow's challenges. Clin Breast Cancer 1:297- 308; discussion 309. Gerdes, J., H. Pickartz, J. Brotherton, J. Hammerstein, H. Weitzel, and H. Stein. 1987. Growth fractions and estrogen receptors in human breast cancers as determined in situ with monoclonal antibodies. Am J Pathol 129:486-92. Graham, J. D., D. L. Bain, J. K. Richer, T. A. Jackson, L. Tung, and K. B. Horwitz. 2000. Nuclear receptor conformation, coregulators, and tamoxifen- resistant breast cancer. Steroids 65:579-84. Harper, J. W., G. R. Adami, N. Wei, K. Keyomarsi, and S. J. Elledge. 1993. The p21 Cdk-interacting protein Cipl is a potent inhibitor of G1 cyclin- dependent kinases. Cell 75:805-16. Harper, J. W., S. J. Elledge, K. Keyomarsi, B. Dynlacht, L. H. Tsai, P. Zhang, S. Dobrowolski, C. Bai, L. Connell-Crowley, E. Swindell, and et a1. 1995. Inhibition of cyclin-dependent kinases by p21. Mol Biol Cell 6:387-400. Hatakeyama, M., and R. A. Weinberg. 1995. The role of RB in cell cycle control. Prog Cell Cycle Res 1:9-19. Helin, K., E. Harlow, and A. Fattaey. 1993. Inhibition of E2F-1 transactivation by direct binding of the retinoblastoma protein. Mol Cell Biol 13:6501-8. Hengst, L., U. Gopfert, H. A. Lashuel, and S. 1. Reed. 1998. Complete inhibition of Cdk/cyclin by one molecule of p21(Cipl). Genes Dev 12:3 882-8. Herrera, R. E., V. P. Sah, B. 0. Williams, T. P. Makela, R. A. Weinberg, and T. Jacks. 1996. Altered cell cycle kinetics, gene expression, and G1 restriction point regulation in Rb-deficient fibroblasts. Mol Cell Biol 16:2402-7. Horwitz, K. B. 1993. Mechanisms of hormone resistance in breast cancer. Breast Cancer Res Treat 26:1 19-30. Howell, A., and J. Robertson. 1995. Response to a specific antioestrogen (ICI 182780) in tamoxifen- resistant breast cancer. Lancet 345:989-90. Hui, R., A. L. Cornish, R. A. McClelland, J. F. Robertson, R. W. Blarney, E. A. Musgrove, R. I. Nicholson, and R. L. Sutherland. 1996. Cyclin D1 and estrogen receptor messenger RNA levels are positively correlated in primary breast cancer. Clin Cancer Res 2:923-8. Hui, R., R. D. Macmillan, F. S. Kenny, E. A. Musgrove, R. W. Blarney, R. I. Nicholson, J. F. Robertson, and R. L. Sutherland. 2000. IN K4a gene expression and methylation in primary breast cancer: overexpression of p16INK4a messenger RNA is a marker of poor prognosis. Clin Cancer Res 622777-87. 28 68. 69. 70. 71. 72. 73. 74. 75. 76. 77. 78. 79. Hunter, T., and J. Pines. 1991. Cyclins and cancer. Cell 66:1071-4. Hunter, T., and J. Pines. 1994. Cyclins and cancer. 11: Cyclin D and CDK inhibitors come of age. Cell 79:573-82. Jaumot, M., J. M. Estanol, O. Casanovas, X. Grana, N. Agell, and O. Bachs. 1997. The cell cycle inhibitor p21CIP is phosphorylated by cyclin A-CDK2 complexes. Biochem Biophys Res Commun 241 :434-8. Jin, Y. H., K. J. Yoo, Y. H. Lee, and S. K. Lee. 2000. Caspase 3-mediated cleavage of p21WAF l/CIPl associated with the cyclin A-cyclin-dependent kinase 2 complex is a prerequisite for apoptosis in SK-HEP-l cells. J Biol Chem 275:30256-63. Johnson, D. G. 1995. Regulation of E2F-l gene expression by p130 (Rb2) and D- type cyclin kinase activity. Oncogene 11:1685-92. Johnston, S. R. 1997. Acquired tamoxifen resistance in human breast cancer-- potential mechanisms and clinical implications. Anticancer Drugs 8:911-30. Johnston, S. R., G. Saccani-Jotti, I. E. Smith, J. Salter, J. Newby, M. Coppen, S. R. Ebbs, and M. Dowsett. 1995. Changes in estrogen receptor, progesterone receptor, and pS2 expression in tamoxifen-resistant human breast cancer. Cancer Res 55:3331-8. Jordan, V. C. 1992. The strategic use of antiestrogens to control the development and growth of breast cancer. Cancer 70:977-82. Jordan, V. C. 1995. Tamoxifen: toxicities and drug resistance during the treatment and prevention of breast cancer. Annu Rev Pharmacol Toxicol 35: 195-21 1. Jorgensen, L., N. Brunner, M. Spang-Thomsen, M. R. James, R. Clarke, P. Dombemowsky, and B. Svenstrup. 1997. Steroid metabolism in the hormone dependent MCF-7 human breast carcinoma cell line and its two hormone resistant subpopulations MCF- 7/LCC1 and MCF-7/LCC2. J Steroid Biochem Mol Biol 63:275-81. Karlseder, J ., R. Zeillinger, C. Schneeberger, K. Czerwenka, P. Speiser, E. Kubista, D. Bimbaum, P. Gaudray, and C. Theillet. 1994. Patterns of DNA amplification at band q13 of chromosome 11 in human breast cancer. Genes Chromosomes Cancer 9:42-8. Kastner, P., A. Krust, B. Turcotte, U. Stropp, L. Tora, H. Gronemeyer, and P. Chambon. 1990. Two distinct estrogen-regulated promoters generate transcripts encoding the two functionally different human progesterone receptor forms A and B. Embo J 9:1603-14. 29 80. 81. 82. 83. 84. 85. 86. 87. 88. 89. 90. 91. Kato, J ., H. Matsushime, S. W. Hiebert, M. E. Ewen, and C. J. Sherr. 1993. Direct binding of cyclin D to the retinoblastoma gene product (pr) and pr phosphorylation by the cyclin D-dependent kinase CDK4. Genes Dev 7:331-42. Kato, J. Y., M. Matsuoka, D. K. Strom, and C. J. Sherr. 1994. Regulation of cyclin D-dependent kinase 4 (cdk4) by cdk4-activating kinase. Mol Cell Biol 14:2713-21. Kato, S., H. Endoh, Y. Masuhiro, T. Kitamoto, S. Uchiyama, H. Sasaki, S. Masushige, Y. Gotoh, E. Nishida, H. Kawashima, and et a1. 1995. Activation of the estrogen receptor through phosphorylation by mitogen- activated protein kinase. Science 270: 1491-4. Katzenellenbogen, B. S., M. M. Montano, T. R. Ediger, J. Sun, K. Ekena, G. Lazennec, P. G. Martini, E. M. McInemey, R. Delage-Mourroux, K. Weis, and J. A. Katzenellenbogen. 2000. Estrogen receptors: selective ligands, partners, and distinctive pharmacology. Recent Prog Horm Res 55:163-93. Kenny, F. S., R. Hui, E. A. Musgrove, J. M. Gee, R. W. Blamey, R. I. Nicholson, R. L. Sutherland, and J. F. Robertson. 1999. Overexpression of cyclin D1 messenger RNA predicts for poor prognosis in estrogen receptor-positive breast cancer. Clin Cancer Res 5:2069-76. Key, T. J ., and M. C. Pike. 1988. The role of oestrogens and progestagens in the epidemiology and prevention of breast cancer. Eur J Cancer Clin Oncol 24:29-43. King, R. J. 1993. William L. McGuire Memorial Symposium. Estrogen and progestin effects in human breast carcinogenesis. Breast Cancer Res Treat 27:3- 15. Klein-Hitpass, L., G. U. Ryffel, E. Heitlinger, and A. C. Cato. 1988. A 13 bp palindrome is a functional estrogen responsive element and interacts specifically with estrogen receptor. Nucleic Acids Res 162647-63. Klein-Hitpass, L., S. Y. Tsai, G. L. Greene, J. H. Clark, M. J. Tsai, and B. W. O'Malley. 1989. Specific binding of estrogen receptor to the estrogen response element. Mol Cell Biol 9243-9. Koduri, S., and I. Poola. 2001. Quantitation of alternatively spliced estrogen receptor alpha mRN As as separate gene populations. Steroids 66:17-23. Kondo, T., N. Minamino, T. Nagamura-Inoue, M. Matsumoto, T. Taniguchi, and N. Tanaka. 1997. Identification and characterization of nucleophosmin/B23/numatrin which binds the anti-oncogenic transcription factor IRF-1 and manifests oncogenic activity. Oncogene 15:1275-81. Kumar, V., and P. Chambon. 1988. The estrogen receptor binds tightly to its responsive element as a ligand-induced homodimer. Cell 55:145-56. 30 92. 93. 94. 95. 96. 97. 98. 99. 100. 101. 102. 103. Kushner, P. J ., D. A. Agard, G. L. Greene, T. S. Scanlan, A. K. Shiau, R. M. Uht, and P. Webb. 2000. Estrogen receptor pathways to AP-l. J Steroid Biochem Mol Biol 74:311-7. Kute, T. E., C. Linville, R. G. Mehta, and R. C. Moon. 1985. Cell kinetics in normal and neoplastic mammary tissues by flow cytometric analyses. Cytometry 6:362-7. LaBaer, J., M. D. Garrett, L. F. Stevenson, J. M. Slingerland, C. Sandhu, H. S. Chou, A. Fattaey, and E. Harlow. 1997. New functional activities for the p21 family of CDK inhibitors. Genes Dev 11:847-62. Lamb, J ., M. H. Ladha, C. McMahon, R. L. Sutherland, and M. E. Ewen. 2000. Regulation of the functional interaction between cyclin D1 and the estrogen receptor. Mol Cell Biol 20:8667-75. Lees, J. A., M. Saito, M. Vidal, M. Valentine, T. Look, E. Harlow, N. Dyson, and K. Helin. 1993. The retinoblastoma protein binds to a family of E2F transcription factors. Mol Cell Biol 13:7813-25. Lilling, G., H. Hacohen, J. Nordenberg, T. Livnat, V. Rotter, and Y. Sidi. 2000. Differential sensitivity of MCF-7 and LCC2 cells, to multiple growth inhibitory agents: possible relation to high bcl-2/bax ratio? Cancer Lett 161:27-34. Lukas, J., J. Bartkova, and J. Bartek. 1996. Convergence of mitogenic signalling cascades from diverse classes of receptors at the cyclin D-cyclin-dependent kinase-pr—controlled G1 checkpoint. Mol Cell Biol 16:6917-25. Lukas, J ., D. Parry, L. Aagaard, D. J. Mann, J. Bartkova, M. Strauss, G. Peters, and J. Bartek. 1995. Retinoblastoma-protein-dependent cell-cycle inhibition by the tumour suppressor p16. Nature 375:503-6. Lykkesfeldt, A. E., J. K. Larsen, and I. J. Christensen. 1986. Cell cycle analysis of estrogen stimulation and antiestrogen inhibition of growth of the human breast cancer cell line MCF-7. Breast Cancer Res Treat 7:S83-90. MacGregor, J. 1., and V. C. Jordan. 1998. Basic guide to the mechanisms of antiestrogen action. Pharmacol Rev 50: 151-96. Matsushime, H., D. E. Quelle, S. A. Shurtleff, M. Shibuya, C. J. Sherr, and J. Y. Kato. 1994. D-type cyclin-dependent kinase activity in mammalian cells. Mol Cell Biol 14:2066-76. McClelland, R. A., J. M. Gee, L. O'Sullivan, D. M. Barnes, J. F. Robertson, 1. 0. Ellis, and R. I. Nicholson. 1999. p21(WAF1) expression and endocrine response in breast cancer. J Pathol 188:126-32. 31 104. 105. 106. 107. 108. 109. 110. 111. 112. 113. 114. 115. 116. McIntosh, G. G., J. J. Anderson, 1. Milton, M. Steward, A. H. Parr, M. D. Thomas, J. A. Henry, B. Angus, T. W. Lennard, and C. H. Home. 1995. Determination of the prognostic value of cyclin D1 overexpression in breast cancer. Oncogene 1 1:885-91. Milde-Langosch, K., A. M. Bamberger, G. Rieck, B. Kelp, and T. Loning. 2001. Overexpression of the p16 cell cycle inhibitor in breast cancer is associated with a more malignant phenotype. Breast Cancer Res Treat 67:61-70. Mosselman, S., J. Polman, and R. Dijkema. 1996. ER beta: identification and characterization of a novel human estrogen receptor. F EBS Lett 392:49—53. Mullick, A., and P. Chambon. 1990. Characterization of the estrogen receptor in two antiestrogen-resistant cell lines, LY2 and T47D. Cancer Res 50:333-8. Muss, H. B. 1992. Endocrine therapy for advanced breast cancer: a review. Breast Cancer Res Treat 21:15-26. Nakanishi, M., Y. Kagawa, H. Takahashi, and H. Matsushime. 1997. Two different bindings of p21 Cdk inhibitor to cyclin/Cdk complex. Leukemia 11 Suppl 32356-7. Neuman, E., M. H. Ladha, N. Lin, T. M. Upton, S. J. Miller, J. DiRenzo, R. G. Pestell, P. W. Hinds, S. F. Dowdy, M. Brown, and M. E. Ewen. 1997. Cyclin D1 stimulation of estrogen receptor transcriptional activity independent of cdk4. Mol Cell Biol 17:5338-47. Nilsson, R. 2000. Endocrine modulators in the food chain and environment. Toxicol Pathol 28:420-31. Oh, Y. L., J. S. Choi, S. Y. Song, Y. H. Ko, B. K. Han, S. J. Nam, and J. H. Yang. 2001. Expression of p21Waf1 , p27Kipl and cyclin D1 proteins in breast ductal carcinoma in situ: Relation with clinicopathologic characteristics and with p53 expression and estrogen receptor status. Pathol Int 51 :94-9. Ohtsubo, M., A. M. Theodoras, J. Schumacher, J. M. Roberts, and M. Pagano. 1995. Human cyclin B, a nuclear protein essential for the G1-to-S phase transition. Mol Cell Biol 15:2612-24. Oltvai, Z. N., C. L. Milliman, and S. J. Korsmeyer. 1993. Bel-2 heterodimerizes in vivo with a conserved homolog, Bax, that accelerates programmed cell death. Cell 74:609-19. Osborne, C. K. 1993. Mechanisms for tamoxifen resistance in breast cancer: possible role of tamoxifen metabolism. J Steroid Biochem Mol Biol 47283-9. Osborne, C. K., D. H. Boldt, and P. Estrada. 1984. Human breast cancer cell cycle synchronization by estrogens and antiestrogens in culture. Cancer Res 44: 1433-9. 32 117. 118. 119. 120. 121. 122. 123. 124. 125. 126. 127. 128. Osborne, C. K., R. Schiff, S. A. Fuqua, and J. Shou. 2001. Estrogen receptor: current understanding of its activation and modulation. Clin Cancer Res 7:4338s- 4342s; discussion 441 IS- 44125. Osborne, C. K., H. Zhao, and S. A. F uqua. 2000. Selective estrogen receptor modulators: structure, fimction, and clinical use. J Clin Oncol 18:3172-86. Otto, A. M., R. Paddenberg, S. Schubert, and H. G. Mannherz. 1996. Cell-cycle arrest, micronucleus formation, and cell death in growth inhibition of MCF-7 breast cancer cells by tamoxifen and cisplatin. J Cancer Res Clin Oncol 122:603- 12. Paech, K., P. Webb, G. G. Kuiper, S. Nilsson, J. Gustafsson, P. J. Kushner, and T. S. Scanlan. 1997. Differential ligand activation of estrogen receptors ERalpha and ERbeta at AP] sites. Science 277: 1 508-10. Pagano, M., R. Pepperkok, J. Lukas, V. Baldin, W. Ansorge, J. Bartek, and G. Draetta. 1993. Regulation of the cell cycle by the cdk2 protein kinase in cultured human fibroblasts. J Cell Biol 121:101-11. Paik, S., D. P. Hartmann, R. B. Dickson, and M. E. Lippman. 1994. Antiestrogen resistance in ER positive breast cancer cells. Breast Cancer Res Treat 31 :301-7. Parker, M. G. 1993. Action of "pure" antiestrogens in inhibiting estrogen receptor action. Breast Cancer Res Treat 26:131-7. Parry, D., D. Mahony, K. Wills, and E. Lees. 1999. Cyclin D-CDK subunit arrangement is dependent on the availability of competing INK4 and p21 class inhibitors. Mol Cell Biol 19:1775-83. Pelosio, P., M. Barbareschi, E. Bonoldi, A. Marchetti, P. Verderio, O. Catfo, P. Bevilacqua, P. Boracchi, F. Buttitta, R. Barbazza, P. Dalla Palma, and G. Gasparini. 1996. Clinical significance of cyclin D1 expression in patients with node— positive breast carcinoma treated with adjuvant therapy. Ann Oncol 7:695- 703. Peters, G., V. Fantl, R. Smith, S. Brookes, and C. Dickson. 1995. Chromosome 1 qu3 markers and D-type cyclins in breast cancer. Breast Cancer Res Treat 33: 125-35. Petz, L. N., and A. M. Nardulli. 2000. Spl binding sites and an estrogen response element half-site are involved in regulation of the human progesterone receptor A promoter. Mol Endocrinol 14:972-85. Piccart, M., C. Lohrisch, A. Di Leo, and D. Larsimont. 2001. The predictive value of HER2 in breast cancer. Oncology 61 :73-82. 33 129. 130. 131. 132. 133. 134. 135. 136. 137. 138. 139. 140. Planas-Silva, M. D., and R. A. Weinberg. 1997. Estrogen-dependent cyclin B- cdk2 activation through p21 redistribution. Mol Cell Biol 17:4059-69. Polyak, K., M. H. Lee, H. Erdjument-Bromage, A. Koff, J. M. Roberts, P. Tempst, and J. Massague. 1994. Cloning of p27Kipl, a cyclin-dependent kinase inhibitor and a potential mediator of extracellular antimitogenic signals. Cell 78:5 9-66. Poola, I., S. Koduri, S. Chatra, and R. Clarke. 2000. Identification of twenty alternatively spliced estrogen receptor alpha mRNAs in breast cancer cell lines and tumors using splice targeted primer approach. J Steroid Biochem Mol Biol 72:249-58. Poon, R. Y., and T. Hunter. 1995. Dephosphorylation of Cdk2 Thrl60 by the cyclin-dependent kinase- interacting phosphatase KAP in the absence of cyclin. Science 270:90-3. Poon, R. Y., W. Jiang, H. Toyoshima, and T. Hunter. 1996. Cyclin-dependent kinases are inactivated by a combination of p21 and Thr-14/Tyr-15 phosphorylation after UV-induced DNA damage. J Biol Chem 271:13283-91. Prall, O. W., B. Sarcevic, E. A. Musgrove, C. K. Watts, and R. L. Sutherland. 1997. Estrogen-induced activation of Cdk4 and Cdk2 during Gl-S phase progression is accompanied by increased cyclin D1 expression and decreased cyclin-dependent kinase inhibitor association with cyclin E- Cdk2. J Biol Chem 272: 10882-94. Rao, S., J. Gray-Bablin, T. W. Herliczek, and K. Keyomarsi. 1999. The biphasic induction of p21 and p27 in breast cancer cells by modulators of cAMP is posttranscriptionally regulated and independent of the PKA pathway. Exp Cell Res 252:211-23. Reddel, R. R., L. C. Murphy, and R. L. Sutherland. 1983. Effects of biologically active metabolites of tamoxifen on the proliferation kinetics of MCF-7 human breast cancer cells in vitro. Cancer Res 43 :461 8-24. Reddel, R. R., and R. L. Sutherland. 1987. Effects of pharmacological concentrations of estrogens on proliferation and cell cycle kinetics of human breast cancer cell lines in vitro. Cancer Res 47:5323-9. Ree, A. H., K. Bjomland, N. Brunner, H. T. Johansen, K. B. Pedersen, A. O. Aasen, and O. Fodstad. 1998. Regulation of tissue-degrading factors and in vitro invasiveness in progression of breast cancer cells. Clin Exp Metastasis 16:205-15. Reed, S. I. 1997. Control of the Gl/S transition. Cancer Surv 29:7-23. Reiss, M., and M. H. Barcellos-Hoff. 1997. Transforming grth factor-beta in breast cancer: a working hypothesis. Breast Cancer Res Treat 45:81-95. 34 141. 142. 143. 144. 145. 146. 147. 148. 149. 150. 151. 152. Resch, A., E. Biber, M. Seifert, and H. Resch. 1998. Evidence that tamoxifen preserves bone density in late postmenopausal women with breast cancer. Acta Oncol 37:661-4. Rossig, L., A. S. Jadidi, C. Urbich, C. Badorff, A. M. Zeiher, and S. Dimmeler. 2001. Akt-dependent phosphorylation of p21(Cipl) regulates PCNA binding and proliferation of endothelial cells. Mol Cell Biol 21 :5644-57. Rutqvist, L. E., B. Cederrnark, U. Glas, H. Johansson, B. Nordenskjold, L. Skoog, A. Somell, T. Theve, S. Friberg, and J. Askergren. 1987. The Stockholm trial on adj uvant tamoxifen in early breast cancer. Correlation between estrogen receptor level and treatment effect. Breast Cancer Res Treat 10:255-66. Serrano, M., G. J. Hannon, and D. Beach. 1993. A new regulatory motif in cell- cycle control causing specific inhibition of cyclin D/CDK4. Nature 366:704-7. Sher, E., J. A. Eisman, J. M. Moseley, and T. J. Martin. 1981. Whole-cell uptake and nuclear localization of 1,25- dihydroxycholecalciferol by breast cancer cells (T47 D) in culture. Biochem J 200:315-20. Sherr, C. J. 1996. Cancer cell cycles. Science 274:1672-7. Sismondi, P., N. Biglia, M. Giai, L. Sgro, and C. Campagnoli. 1994. Metabolic effects of tamoxifen in postmenopause. Anticancer Res 14:2237-44. Skaar, T. C., S. C. Prasad, S. Sharareh, M. E. Lippman, N. Brunner, and R. Clarke. 1998. Two-dimensional gel electrophoresis analyses identify nucleophosmin as an estrogen regulated protein associated with acquired estrogen- independence in human breast cancer cells. J Steroid Biochem Mol Biol 67:391-402. Smith, C. L., Z. Nawaz, and B. W. O'Malley. 1997. Coactivator and corepressor regulation of the agonist/antagonist activity of the mixed antiestrogen, 4- hydroxytamoxifen. Mol Endocrinol 11:657-66. Soule, H. D., J. Vazguez, A. Long, S. Albert, and M. Brennan. 1973. A human cell line from a pleural effusion derived from a breast carcinoma. J Natl Cancer Inst 51 : 1409-16. Sutherland, R. L., C. K. Watts, R. E. Hall, and P. C. Ruenitz. 1987. Mechanisms of growth inhibition by nonsteroidal antioestrogens in human breast cancer cells. J Steroid Biochem 27:891-7. Takano, Y., H. Takenaka, Y. Kato, M. Masuda, T. Mikami, M. Saegusa, and I. Okayasu. 1999. Cyclin D1 overexpression in invasive breast cancers: correlation with cyclin-dependent kinase 4 and oestrogen receptor overexpression, and lack of correlation with mitotic activity. J Cancer Res Clin Oncol 125:505-12. 35 153. 154. 155. 156. 157. 158. 159. 160. 161. 162. 163. 164. Takimoto, G. S., J. D. Graham, T. A. Jackson, L. Tung, R. L. Powell, L. D. Horwitz, and K. B. Horwitz. 1999. Tamoxifen resistant breast cancer: coregulators determine the direction of transcription by antagonist-occupied steroid receptors. J Steroid Biochem Mol Biol 69:45-50. Tam, S. W., A. M. Theodoras, J. W. Shay, G. F. Draetta, and M. Pagano. 1994. Differential expression and regulation of Cyclin D1 protein in normal and tumor human cells: association with Cdk4 is required for Cyclin D1 function in G1 progression. Oncogene 9:2663-74. Tengrup, 1., L. T. Nittby, and T. Landberg. 1986. Prophylactic oophorectomy in the treatment of carcinoma of the breast. Surg Gynecol Obstet 162:209-14. Thompson, E. W., N. Brunner, J. Torri, M. D. Johnson, V. Boulay, A. Wright, M. E. Lippman, P. S. Steeg, and R. Clarke. 1993. The invasive and metastatic properties of hormone-independent but horrnone-responsive variants of MCF-7 human breast cancer cells. Clin Exp Metastasis 11:15-26. Toyoshima, H., and T. Hunter. 1994. p27, a novel inhibitor of G1 cyclin-Cdk protein kinase activity, is related to p21. Cell 78:67-74. Tremblay, A., G. B. Tremblay, F. Labrie, and V. Giguere. 1999. Ligand- independent recruitment of SRC-l to estrogen receptor beta through phosphorylation of activation function AF-l. Mol Cell 3:513-9. Tremblay, G. B., A. Tremblay, N. G. Copeland, D. J. Gilbert, N. A. Jenkins, F. Labrie, and V. Giguere. 1997. Cloning, chromosomal localization, and functional analysis of the murine estrogen receptor beta. Mol Endocrinol 11:353-65. Vogel, V. G. 2001. F ollow-up of the breast cancer prevention trial and the future of breast cancer prevention efforts. Clin Cancer Res 7:441 3s-441 8s; discussion 441 IS- 44123. Vos, C. B., N. T. Ter Haar, J. L. Peterse, C. J. Comelisse, and M. J. van de Vijver. 1999. Cyclin D1 gene amplification and overexpression are present in ductal carcinoma in situ of the breast. J Pathol 187:279-84. Wakasugi, E., T. Kobayashi, Y. Tarnaki, Y. Ito, 1. Miyashiro, Y. Komoike, T. Takeda, E. Shin, Y. Takatsuka, N. Kikkawa, T. Monden, and M. Monden. 1997. p21(Wafl /Cipl) and p53 protein expression in breast cancer. Am J Clin Pathol 107:684-91. Wallgren, A., E. Baral, U. Glas, L. Kamstrom, B. Nordenskiold, N. O. Theve, and C. Silfversward. 1985. Adjuvant tamoxifen treatment in postmenopausal patients with operable breast cancer. J Steroid Biochem 23:1161-2. Watts, C. K., A. Brady, B. Sarcevic, A. deFazio, E. A. Musgrove, and R. L. Sutherland. 1995. Antiestrogen inhibition of cell cycle progression in breast 36 165. 166. 167. 168. 169. 170. 171. 172. 173. cancer cells in associated with inhibition of cyclin-dependent kinase activity and decreased retinoblastoma protein phosphorylation. Mol Endocrinol 9: 1 804-13. Watts, C. K., K. J. Sweeney, A. Warlters, E. A. Musgrove, and R. L. Sutherland. 1994. Antiestrogen regulation of cell cycle progression and cyclin D1 gene expression in MCF-7 human breast cancer cells. Breast Cancer Res Treat 31 :95- 105. Webb, P., P. Nguyen, C. Valentine, G. N. Lopez, G. R. Kwok, E. McInemey, B. S. Katzenellenbogen, E. Enmark, J. A. Gustafsson, S. Nilsson, and P. J. Kushner. 1999. The estrogen receptor enhances AP-l activity by two distinct mechanisms with different requirements for receptor transactivation functions. Mol Endocrinol 13 : 1 672-85. Weintraub, S. J ., C. A. Prater, and D. C. Dean. 1992. Retinoblastoma protein switches the E2F site from positive to negative element. Nature 358:259-61. Weiss, R. H., A. Joo, and C. Randour. 2000. p21(Waf1/Cip1) is an assembly factor required for platelet-derived growth factor-induced vascular smooth muscle cell proliferation. J Biol Chem 275:10285-90. Xiong, Y., H. Zhang, and D. Beach. 1993. Subunit rearrangement of the cyclin- dependent kinases is associated with cellular transformation. Genes Dev 7: 1572- 83. Zhou, J. N., and S. Linder. 1996. Expression of CDK inhibitor genes in immortalized and carcinoma derived breast cell lines. Anticancer Res 16:1931-5. Zukerberg, L. R., W. I. Yang, M. Gadd, A. D. Thor, F. C. Koemer, E. V. Schmidt, and A. Arnold. 1995. Cyclin D1 (PRADI) protein expression in breast cancer: approximately one-third of infiltrating mammary carcinomas show overexpression of the cyclin D1 oncogene. Mod Pathol 8:560-7. Zwijsen, R. M., R. S. Buckle, E. M. Hijmans, C. J. Loomans, and R. Bemards. 1998. Ligand-independent recruitment of steroid receptor coactivators to estrogen receptor by cyclin D1. Genes Dev 12:3488-98. Zwijsen, R. M., E. Wientjens, R. Klompmaker, J. van der Sman, R. Bemards, and R. J. Michalides. 1997. CDK-independent activation of estrogen receptor by cyclin D1. Cell 88:405-15. 37 CHAPTER TWO Regulation of Cdk4 by estrogen and antiestrogen in MCF-7 Cells Originally published as Skildum, A. J., S. Mukherjee, and S. E. Conrad. 2001. The cyclin dependent kinase inhibitor p21WAF” C'“ is an antiestrogen regulated inhibitor of Cdk4 in human breast cancer cells. J Biol Chem 277(7):5145-52. 38 Chapter 2: Regulation of Cdk4 by estrogen and antiestrogen in MCF—7 Cells ABSTRACT The MCF-7 cell line is a model of estrogen dependent, antiestrogen sensitive human breast cancer. Antiestrogen treatment of MCF-7 cells causes dramatic decreases in both Cdk4 and Cdk2 activities, which leads to a G1 phase cell cycle arrest. In this report, we investigate the mechanism(s) by which Cdk4 activity is regulated in MCF-7 cells. Through time course analysis, we demonstrate that changes in Cdk4 activity in response to estrogen or antiestrogen treatment do not correlate directly with cyclin D1 protein levels or association. In contrast, Cdk4 activity does correlate with changes in the level of the Cdk inhibitor p21WAF ” Cipl. Furthermore we show that extracts of antiestrogen-treated cells contain a factor capable of inhibiting the Cdk4 activity present in extracts of estrogen-treated cells, and immunodepletion experiments identify this factor as p21WAH/C’p'. These results identify p21‘MF”Cipl as an important physiological regulator of Cdk4 complexes in human breast cancer cells. 39 Introduction Many estrogen receptor (ER)I positive breast tumors require estrogen for growth, and can be successfully treated with antiestrogens (40, 41). The MCF-7 cell line, which was derived from a human breast adenocarcinoma, serves as a model for such estrogen responsive, antiestrogen sensitive, breast tumors (22). MCF-7 cells require estrogen to proliferate, and arrest in the G1 phase of the cell cycle when they are deprived of estrogen or treated with antiestrogens (24, 27, 28, 45). Understanding the mechanisms by which antiestrogens arrest the growth of breast cancer cells is an area of active investigation with potential clinical significance. In normal cells, the transition from G1 to S phase requires the activity of two classes of cyclin dependent kinases (Cdks), Cdk4/6, and Cdk2. Cdk4/6 phosphorylate the tumor suppressor protein pr, which leads to transcription of E2F- regulated genes including cyclins E and A. Cdk2 can also phosphorylate pr, as well as additional substrates necessary for genome duplication (13, 14, 42, 43). Cdk activity is regulated by multiple mechanisms, including phosphorylation (9, 19, 33, 52) and association with both positive and negative regulatory proteins, and each of these mechanisms is potentially modified by ER signaling. Cdk activation requires association with a cyclin partner: Cdk4/6 associate with D type cyclins, while Cdk2 associates with either cyclin E or cyclin A (7, 16). Cdk activity can be inhibited by two different families of cyclin dependent kinase inhibitors (Cdkls). Members of the INK4a family, including p16INK4a (p16), bind specifically to monomeric Cdk4/6 and prevent its association with a D type ' Abbreviations used are: ER, estrogen receptor. p21, WAFl/Cipl. p27, Kipl. p16, INK4a. Cdk, cyclin dependent kinase. Cdkl, Cdk inhibitor. pr, retinoblastoma protein. 152, 17B-estradiol. IP, immunoprecipitation. 1C1, 1C1 182,780. DTT, dithiothreotol. IMEM, improved modified Eagle’s medium. CSS, charcoal stripped serum. Minutes, m. Hours, h. FBS, fetal bovine serum. 40 cyclin (30, 37). Members of the WAF l/Cipl family, which include p21WAF”Cipl (p21) and p27kipl (p27), bind to G1 cyclin/Cdk complexes, not to monomeric cyclins or Cdks (11, 17, 20, 30, 32, 46, 47). While both p21 and p27 can inhibit the activity of Cdk2 and Cdk4/6, p21 can also serve as an assembly factor for cyclinD/Cdk4 complexes, increasing the efficiency of complex formation and Cdk4 activity (5, 12, 21). Based on numerous studies, a general model for the regulation of passage from G1 to S phase in normal cells has been proposed, and deregulation at various stages in the model are proposed to be responsible for cellular transformation and tumorigenesis (14, 16, 17, 23, 39, 42). Many mitogens induce cyclin D expression, either through increased mRN A synthesis and/or stabilization of the protein. Cyclin D binds to Cdk4/6, resulting in an increase in Cdk4/6 kinase activity, pr phosphorylation and synthesis of cyclins E and A. Increasing the cellular concentration of cyclin D/Cdk4/6 complexes provides additional binding targets for p21 and/or p27, thereby titrating these Cdkls from their inhibitory association with Cdk2 complexes (30, 31). Together, the increased levels of cyclins and the decreased association of Cdkls lead to Cdk2 activation and progression to S phase (42). While this model is widely accepted, mitogens and anti-mitogens can also affect progression through G1 in other ways, for example by regulating the levels of Cdkls (34, 38), the assembly and nuclear import of cyclin/Cdk complexes (6), or the levels and activity of Cdk-activating kinases or phosphatases (9, 19, 33). The specific mechanism(s) by which estrogen and antiestrogens regulate MCF-7 cell proliferation are not completely understood. Others have reported that antiestrogen treatment decreases cyclin D1 and cyclin A protein levels, Cdk4 and Cdk2 activity, and pr phosphorylation, and increase Cde levels (4, 10, 31, 48, 49). All of these effects are 41 reversed upon removal of antiestrogen and replacement with estrogen (2, 10, 36). Although both total Cdk2 and cyclin E/Cdk2 activities are highly regulated, cyclin B protein is expressed at constant levels in estrogen and antiestrogen treated MCF-7 cells. The lack of activity of cyclin E/Cdk2 complexes in the presence of antiestrogen is believed to be due to inhibition by p21 and/or p27 (31, 36). Cyclin E/Cdk2 activation after estrogen treatment may therefore be a result of decreased levels of these inhibitors and/or sequestration of p21 or p27 into newly formed cyclin Dl/Cdk4 complexes (31, 36). Previous reports suggest that Cdk4 activity in estrogen or antiestrogen treated MCF-7 cells is regulated primarily by the levels of cyclin D1 protein. Although p21 and p27 are found in association with Cdk4, evidence for a physiological role in inhibiting Cdk4 is lacking. The Cdkl p21 is reported to be both an activator and an inhibitor of Cdk4, so its function in the regulation of Cdk4 activity by antiestrogen is unclear. In the current report, we investigate the mechanisms by which l7B—estradiol (E2) and the pure antiestrogen ICI 182,780 (ICI) regulate Cdk4 activity in MCF-7 cells. We find that Cdk4 activity is not directly correlated with cyclin D1 protein levels or association. Utilizing an in vitro mixing assay, we demonstrate that extracts of ICI-treated cells contain a factor that inhibits the Cdk4 activity present in extracts of estrogen treated cells, and that this inhibitory factor is specifically removed by immunodepletion of p21 but not p27. These studies identify p21 as an important physiological target of antiestrogen action that inhibits both Cdk4 and Cdk2 activities in human breast cancer cells. 42 Materials and Methods Cell lines and culture media. MCF-7 cells were obtained from Dr. Michael Johnson of the Lombardi Cancer Center, Georgetown University. They were routinely passaged in IMEM (BioFluids), supplemented with 5% fetal bovine serum (FBS, HyClone), 100 units/mL penicillin (Gibco) and 100 ug/mL streptomycin (Gibco). For experiments, cells were cultured in IMEM without phenol red (BioFluids) containing 5% charcoal stripped serum (CSS, HyClone) and penicillin / streptomycin, with either 10'8 M E2 (Sigma) or 10'8 M ICI (Astra Zeneca). Cells were cultured at 37° C with 5% C02. Cell cycle analysis. Cells were trypsinized, washed in phosphate buffered saline (PBS), suspended in PBS+10% F BS, fixed with 80% cold ethanol and stored at —20° C. Prior to analysis, cells were washed twice with PBS, then suspended in PBS + 1 mg/ml RNaseA, 0.2 mg/ml propidium iodide, 0.5 mM EDTA and 0.1% Triton-X-lOO. Cells were then analyzed for red fluorescence on a F ACSVantage flow cytometer; cell cycle distribution was determined using ModFit software. Three 60 mm plates were analyzed for each time point. These experiments were performed in part by Shibani Mukherjee. Immunoprecipitations and Cdk4 kinase assays. Immunoprecipitations (IP) and Cdk4 activity measurements were performed using a modification of a published method (25). Unless indicated otherwise, all manipulations were carried out on ice. Cells were washed twice in ice cold PBS, harvested by scraping in PBS and pelleted; the pellets were frozen and stored in liquid nitrogen until the day of analysis. Cells were resuspended and lysed 43 by sonication in IP buffer (50 mM HEPES (pH 7.5), 150 mM NaCl, 1 mM EDTA, 2.5 mM EGTA, 0.1% Tween-20, 10% glycerol, 1 mM DTT, 10 mM B-glycerophosphate, 1 mM NaF, lmM NaVO4, 0.1 mM PMSF, 10 ug/ml leupeptin, and 2 [lg/{1’11 aprotinin). The lysates were cleared by centrifugation, and protein concentrations were quantitated using BioRad’s protein assay reagent. For IPs, 75 pg of total protein was diluted to 500 111 with IP buffer, then incubated for 60 minutes (m) rocking at 4° C with 1.5 pg antibody (anti- Cdk4 H-22-G or normal goat IgG, Santa Cruz) bound to 7.5 111 protein G agarose beads (Boehringer Mannheim). Beads were pelleted and washed four times with 100 pl IP buffer and twice with 100 1.11 kinase buffer (50 mM HEPES pH7.5, 10 mM MgC12, 1 mM DTT, 2.5 mM EGTA, 10 mM B-glycerophosphate, 0.1 mM NaVO4, and 1 mM NaF). Pellets were then suspended in 40 111 kinase buffer containing 10 pCi y-[32P]ATP, 20 11M cold ATP, and 1.0 111 glutathione sepharose 48 (Amersham Pharrnacia) to which approximately 2.0 pg of a fusion protein between glutathione-S-transferase (GST) and amino acids 792 to 928 of human Rb were bound (GST-Rb bacterial expression vector was kindly provided by Dr. William Kaelin of the Dana F arber Cancer Institute). The reactions were incubated at 30° C for 30 m with occasional mixing, after which they were boiled in SDS loading buffer containing mercaptoethanol. Beads were pelleted and the supematants transferred to clean tubes. Aliquots of the reaction products were resolved on 10% SDS polyacrylamide gels, which were then dried and exposed to autoradiography film. The data were quantitated by phosphorimaging with a Storm phosphorimager (Molecular Dynamics) using ImageQuant software. 44 Western blotting. Either total cell lysates (20 pg), IP supematants or IP pellets were resolved on 12% SDS-polyacrylamide gels, transferred to membranes and probed with antibodies for cyclin D1 (UB1 06 137, Upstate Biotechnology), p21wan (p21-c-19, Santa Cruz Biotechnology), p27k’p1 (C-19, Santa Cruz Biotechnology), Cdk4 (H22, Santa Cruz Biotechnology) or actin (Sigma clone AC-40). Membranes were then incubated with horseradish peroxidase conjugated goat anti-rabbit (BioRad) or goat anti-mouse (American Qualex) secondary antibodies, and immunoreactive proteins were detected using Super Signal West Pico Chemiluminescent Substrate (Pierce). Lysate mixing assays. Cells were harvested, stored, and lysed as described above. Seventy five pg of protein from E2 treated cells were mixed with 7.5 to 75 pg of protein from ICI treated cells, and diluted to 500 p1 with IP buffer. The mixtures were incubated at 30° C for 30 m with occasional agitation. The mixed lysates were then subjected to the Cdk4 kinase assay as described above. Phosphorylated substrate band intensities were quantitated by storage phosphorimaging. Immunodepletion. For immunodepletion studies, 300 pg of lysates were diluted to 600 p1 with IP buffer and incubated rocking at 4° C with 22.5 pl protein G agarose beads to which was bound 4.5 pg (Figures 4 and 5) or 9 pg (Figure 6) antibody (p21-C-19-G, p27- C-19-G, Cdk4-H22-G or normal goat IgG, all from Santa Cruz). The beads were pelleted, and the supernatant incubated with fresh antibody bound beads a second time. Aliquots of the supematants and pellets were then analyzed for Cdk4 activity, by Western 45 blotting or used in the lysate mixing assay as described above. Statistical analysis for data presented in Figure 5 was performed using Microsoft Excel. Results ICI and E2 regulate Cdk4 activity independent of cyclin D] levels or association in MCF-7 cells. Cyclin D/Cdk4 complexes are key integrators of positive and negative growth signals (16, 17, 23, 42, 44), and both cyclin D1 levels and Cdk4 activity are regulated by E2 and ICI in MCF-7 cells (10, 31, 36). To determine whether changes in cyclin D1 protein levels and/or association correlate directly with Cdk4 activity and cell cycle progression in these cells, we investigated the effects of ICI and E2 on cell cycle distribution, Cdk4 activity and cyclin D/Cdk4 complex formation. The experiments were performed in the presence of charcoal stripped serum (CSS), which is free of steroid hormones. To study the inhibition of proliferation by ICI, MCF-7 cells were plated at low density and incubated in the presence of E2 for 48 h to generate an asynchronous, proliferating population. The E2 containing medium was then removed and fresh medium containing ICI was added to block ER signaling, and cells were harvested at six hour intervals. As shown in Figure 1A, ICI caused a gradual decrease in the percentage of cells in S phase beginning by 18 h after treatment, and reaching a minimum of 5% by 48 h. Under these conditions, Cdk4 activity was reduced 50% by 12 h, and 90% by 24 h (Figure 1B). The decrease in Cdk4 kinase activity preceded the observed changes in cell cycle phase distribution, consistent with the concept that the loss of Cdk4 activity was responsible for the cell cycle arrest observed. 46 % Cells in S Phase 1?"? C,’ ”7 E ”7 1°: _ I i! 1' I 0h 6h 12h 18h‘24h 30h‘36h 48h Time After ICI Treatment E B. g Hours after E Cdk4 | P lCItreatment: 0 0 6 12 24 3o GST-Rb —> m “ ’ .» ‘é C 6 ' Hours after lysates g Cdk4 |p '0 treatment: 0 612 24 3O 0 0 612 24 30 m- ‘_ Cdk4 W- ‘_ ACtln Figure 1: Time course of Cdk4 inhibition by ICI in MCF-7 cells. MCF-7 cells were cultured in medium containing 10-8 M E2 for 48 h, washed in PBS, then treated with medium conatining 10-8 M ICI and harvested at the times indicated. A) Cells were fixed, stained with propidium iodide, and analyzed by flow cytometry as described in Materials and Methods. The average percentage of cells in S phase at each time point is shown +/- one stande deviation (n = 3). B) Cells were treated as in IA and assayed for Cdk4 activity as described in Materials and Methods. Numbers indicate hours afier ICI treatment. The lane labeled mock IP shows kinase activity precipitating with pre-immune control antibodies. C) Lysates and aliquots of the reaction from (B) were irnmunoblotted for cyclin D1, Cdk4, p21, p27 and actin. Data shown are representative of three independent experiments. 47 To determine if the decrease in Cdk4 activity shown in Figure 1B correlated with changes in cyclin D1 and/or Cdkl protein levels, Western blot analyses were performed (Figure 1C). No changes in Cdk4 protein levels were detected, confirming that the decrease in Cdk4 activity was not due to a decrease in the level of the catalytic subunit. These analyses also indicate that Cdk4 activity did not correlate directly with cyclin D1 protein levels. At 24 and 30 h after antiestrogen treatment, when there was virtually complete inhibition of Cdk4 activity, the cyclin D1 levels in the lysates were similar to those observed in asynchronous MCF-7 cells prior to antiestrogen treatment (0 h). More significantly, the amount of cyclin D1 co-immunoprecipitating with Cdk4 did not change decrease detectably during the time course. While the levels of p27 remained relatively constant during this experiment, the levels of p21 increased afier ICI treatment, and this increase correlated with Cdk4 inactivation (Figure 1C). These data indicate that the inhibition of Cdk4 activity following antiestrogen treatment is independent of changes in cyclin D1 levels, and suggest that increased p21 levels may inhibit Cdk4. To further examine the effects of ICI and E2 on Cdk4 activity, the above experiment was performed in reverse. Cells were pre-arrested with ICI, released by the removal of ICI and the addition of E2, and harvested at 6 h intervals. At each time point, samples were analyzed for cell cycle distribution, for Cdk4 levels and activity and for cyclin D1, p21 and p27 protein levels and Cdk4 association. As shown in Figure 2A, the percentage of cells in S phase remained low until 18 h, then increased to 40—50% at the 24 and 30 h time points. Cdk4 activity was undetectable at 0 h, and increased by 24 and 30 h (Figure 2B). In agreement with the results presented in Figure 1, activation of Cdk4 did not correlate with changes in the levels of cyclin D1 or in its association with Cdk4. 48 50 0) v1 Eh g 40i .f “ I "'1‘ 1:1 2 3° 1 ; Li 1 a 20 ?‘ u i 3 59 10 1 ; :3 0 WELL-fhflfi 011 611 12h 18h 24h 3011 Time After 152 Treatment E x 3 Hours after Cdk4 l P E Eltreatment o 6 12 18 24 30 30 “" "" ‘— GST-Rb & 1? Hours after U 5315 Cdk4 11’ E E2treatment: 0 6 12 18 24 3O 0 6 12 18 24 30 30 I-”""""“** CyclinDI "- ---...__ .""""-H p27 'D..-. ..... ~~~~¢~ ACtln Figure 2: Time course of Cdk4 activation by E2 in MCF-7 cells. MCF-7 cells were arrested for 48 h in medium containing10-8 M ICI, washed, treated with medium containing 10-8 M E2 and harvested at the time points indicated. A) Cell cycle analysis was performed as in Figure 2A. B) Samples were assayed for Cdk4 activity as described in Materials and Methods. C) Aliquots of total cell lysates and of the immunoprecipitates from (B) were Western blotted for cyclin D1, Cdk4, p21, p27 and actin. This experiment has been repeated once with similar results (not shown). 49 A transient increase in cyclin D1 was reproducibly observed at 6 and 12 h after treatment in both the total lysate and the Cdk4 IP, but this increase was not coincident with Cdk4 activation. When Cdk4 activity was highest, at 24 and 30 h after E2 treatment, cyclin D1 protein levels were similar to before treatment (Figure 2C). In this experiment, both p21 and p27 levels decreased after E2 treatment in both the total cellular lysate and Cdk4 IP. These data, taken together with the results of Figure 1, suggest that Cdk4 activity in E2 or ICI treated cells is not directly determined by the total amount of cyclin D1 in the cell or by the amount of cyclin D1 in complex with Cdk4. Both experiments provide correlative data suggesting that p21 and/or p27 might be responsible for the inhibition of Cdk4 activity in response to ICI treatment. ICI regulates Cdk4 activity through an inhibitory factor. The results presented above provide correlative data that accumulation of p21 and/or p27 might be responsible for the inhibition of Cdk4 activity in these ICI treated cells. We therefore designed in vitro mixing experiments to test this possibility directly. MCF-7 cells were pre-arrested in ICI for 48 h, then treated with medium containing either E2 or fresh ICI for an additional 24 h. Lysates were prepared, and increasing amounts of lysate from ICI treated cells were mixed with a constant 75 pg of lysate from E2 treated cells. The single and mixed lysates were diluted to a constant volume, incubated for 30 m, and then subjected to a Cdk4 activity assay as described in Materials and Methods. This experiment was designed to distinguish between three potential mechanisms for regulating Cdk4. If the lack of Cdk4 activity in ICI-treated cells was due to a post- translational modification of the complex, such as a change in phosphorylation state, we 50 2"le Cdk4 IP: Cdk4 mock Cdk4 1:1 _ _ _ 1:10 2 3 LJ 3 LJ 8 L“. Q Qle 111111 a + GST-Rb B. a b c A B c a? ’ i > E E i m . E ..0.5 0 C .9 6 El 3 o vvvvv 110 15121 ICI C. lysate c_dk42 IP mocklP ICI E2 10 £2 ICI £2 Cyclin DI —> ‘ II— ..... .. p2] -> - " -... Actin—> - . Figure 3: Evidence for an ICI-regulated inhibitor of Cdk4 activity in MCF-7 cells. MCF-7 cells were pre-arrested with ICI for 48 h, then treated for 24 h with either ICI or E2. A) Samples were assayed for Cdk4 activity as described in Materials and Methods. Supematants from samples marked with lower case letters were subjected to a second round of Cdk4 IP and kinase assay, shown in lanes with the corresponding upper case letters. For mixes, 75 pg of E2 treated lysate were added to increasing amounts of ICI treated lysate in a constant volume of 500 ml. Unmixed reactions were diluted to 500 p1, and all tubes were incubated at 30° C for 30 In prior to immunoprecipitation for Cdk4 assays. B) Three independent mixing experiments were performed as described in (A) and quantitated by phosphorimaging. For each experiment, the amount of activity in the E2 treated extract was defined as 1, and activities recovered in the mixed extracts are expressed relative to that value. The average activities are shown +/— standard deviation is shown (open bars) compared to the predicted activity expected based on the ratio of active to total lysate in the mixtures (closed bars). C) Lysates and immunoprecipitates from (A) were immunoblotted for cyclin D1 , p21 and actin. 51 would expect the Cdk4 activity in the lysate of E2-treated cells to be unaffected by the addition of the lysate from ICI-treated cells. If the lack of Cdk4 activity was due to the absence of a Cdk4 activator (such as a D type cyclin), we might expect to see an increase in total activity in the mixed lysates. Finally, if the lack of Cdk4 activity was due to the presence an inhibitory factor, we would expect the addition of the extract of ICI-treated cells to lower the activity present in the E2 treated lysates. The results of one representative mixing experiment are shown in Figure 3. As predicted from the time course experiments described above, only background levels of Cdk4 activity were recovered from the ICI treated cells, and a 10 fold increase in Cdk4 activity was observed after E2 treatment (Figure 3A, lanes 1 and 2). Under these experimental conditions, the amount of immunoprecipitating antibody was limiting, so that the Cdk4 activity recovered was not the total activity in the lysate but rather a representative fraction thereof. This is demonstrated by the fact that equal activity was detected in a second sequential Cdk4 IP of the same lysate (Figure 3A, lanes 5-7, marked “2“d 11)”). Because the IPS were performed in “lysate excess”, we predicted that the mixed lysates would exhibit decreased Cdk4 activity in the absence of any activating or inhibitory activities. Since Cdk4 is present at equal levels in active and inactive extracts and IPs (see Western blots in Figure 2C; note the constant level of Cdk4 detected in Cdk4 IPs despite differences in kinase activity), and since the mixed reactions contained a greater amount of total cellular protein, the predicted decrease would be proportional to the ratio of inactive to active lysate in the mixture. For example, at the 1 to 2 mixing ratio, 37.5 pg of ICI treated lysate was mixed with 75 pg of E2 treated lysate, and the 52 predicted Cdk4 activity in the mixture would be 75/(75 + 37.5) = 0.67 = 67% of the activity of the E2 treated lysate; this fraction is represented by black bars in Figure 3B. Three identical mixing experiments were performed, and the activity recovered in mixed lysates was expressed as a fraction of the activity recovered from the E2 treated lysate. As shown in Figure 3B, at each mixing ratio the amount of activity recovered was significantly less than the fractional representation of the E2 treated lysate in the mixtures. These results provide the first direct evidence that ICI treatment induces a factor capable of inhibiting Cdk4 activity, and that this factor is present in excess of cyclin D/Cdk4 complexes in ICI treated lysates. Aliquots of the lysates and Cdk4 IP pellets used in Figure 4A were analyzed for cyclin D1 and p21 protein levels by Western blotting (Figure 3C). Consistent with our previous result, similar amounts of cyclin D1 were present in the lysates from ICI and E2 treated cells, despite the differences in kinase activity. The amount of p21 in both the total lysate and in complex with Cdk4 was slightly higher in the ICI-treated than in the E2-treated lysate. Depletion of p21 , but not p2 7, removes the Cdk4 inhibitory activity from ICI treated lysates. To determine whether a known protein accounted for the inhibition of Cdk4 activity in extracts of ICI-treated cells, we modified the lysate mixing experiment described above. Prior to mixing, aliquots of the ICI treated lysate were subjected to two rounds of immunodepletion with antibodies against p21, p27, Cdk4, or with a normal goat IgG control (mock). The immunodepleted ICI treated lysates were then incubated with E2 treated lysate at a 1:1 mixing ratio and subjected to a Cdk4 kinase as described 53 A' ICI 52 ID: - p21 p27Cdk4 m - . ' E 1' 1- i Hi. I"... v ‘- , k ‘7‘ ' . , A “'4:- J4—p27 ““fl *- -~ In»- <-—Cdk4 - .--.- <—Cyclin DI <—Actin B mocklP '0 E2f‘j Mix: |C|_+E2 V .5: st .2 1— Nx U PK .2 U '0 '3382'- -aaag " '- - - -<—GST-Rb C. 31.2 -- ,1 _ - ,- -- _, --_.,_____j .210 2 E ' ’7 790.8— 30.6— __ o E 0.4“ 60.2 * gooi— D ' D 1:1 ,2: 52 E2+ICI 10 ID: - - p21 p27 Cdk4 mock - Figure 4: Depletion of p21 but not p27 abolishes the Cdk4 inhibitory activity in [CI treated MCF-7 cells. The Cdk4 inhibition assay was carried out as described in Figure 4, but with the ICI treated lysate subjected to two rounds of immunodepletion with anti- p21, anti-p27, anti-Cdk4 or control (goat IgG, "mock") antibodies prior to mixing with the E2 treated lysate. A) Aliquots of the untreated and immunodepleted lysates were immunoblotted for p21, p27, Cdk4, cyclin D1 and actin. Lanes marked " - " contain untreated lysates. M, mock immunodepletion. B) Autoradiogram of the Cdk4 in vitro kinase assay. The first seven lanes show the Cdk4 activity in unmixed lysates; the last five lanes show Cdk4 activity in lysates mixed at 1:1 ratios. C) Bands in (B) were quantitated by phosphorimaging. The Cdk4 activities in the mixed lysates (E2 + ICI) are expressed as a fraction of the activity in the E2 treated lysate. The dashed line represents 50% of the activity in the lysate from E2 treated cells, which is the fractional representation of the E2 treated lysate in the mixtures. This experiment was repeated once with similar results (not shown). 54 above. Aliquots of the untreated and depleted extracts were also analyzed by Western blotting for p21, p27, Cdk4, cyclin D1 and actin levels. As shown in Figure 4A, two rounds of immunodepletion were sufficient to significantly reduce the amount of the target proteins in the ICI treated lysate, although all were detectable by Western blotting with very long exposures. Two rounds of treatment with a control antibody (goat IgG, “mock”) had no effect on the levels of the proteins examined. Immunodepletion of either Cdk4 or p21 removed the majority of cyclin D1 from the lysate of ICI treated cells, suggesting that virtually all the cyclin D1 in 1C1 treated cells is in complex with both Cdk4 and p21. Immunodepletion of Cdk4 reduced the amount of p2 1 , while immunodepletion of p21 resulted in only a slight decrease in the amount of Cdk4 present in lysates from ICI treated cells. Depletion of p27 did not change the levels of p21, Cdk4 or cyclin D1, although we have consistently observed p27 co-precipitating with Cdk4 (see Figure 6), as reported by others (5, 46). Figure 4B shows the autoradiogram of the in vitro kinase assay for this experiment; the quantitation of phosphorylated GST-Rb band intensities is shown in Figure 4C. Immunodepletion of p21 or p27 had no effect on the Cdk4 activity recovered from the ICI treated lysate, indicating that the removal of inhibitors alone is not sufficient to activate Cdk4. Consistent with the results described in Figure 3, mixing the non- immunodepleted ICI treated lysate with the E2 treated lysate resulted in inhibition of Cdk4 activity to less than the 50% predicted by the fractional representation of the E2 treated lysate in the mixture. When the ICI treated lysate was immunodepleted of p27, Cdk4 or treated with control antibodies, a similar inhibition of the Cdk4 activity in the E2 treated lysate was observed. However, immunodepletion of p21 from the 1C1 treated 55 O. A. .3 supematants Cdk4 IP 2 ID: - p21 mock- - p21mock- - Q Q Q a Q Q Q m a p21-y . - - . _ Actin-> "~ -- «- u... m B. E U E NolD 21 ID mocle ”3‘ ‘ P21 "1°C": ' 15 1:1 1': 1:1 15 1:1 9 Q Q a a /l /I /I “ H I? It at at up w .<-GST—Rb C. 1.2 “" Z‘ 1 IE 1.0 l ‘0' 1 _‘_° 0.8 1 N F ‘5 1 3: 0.6 O C .3 0.4 1 1 L“ 0-2 g“ l \‘ ! E2 1:5 1:2 1 1 ICI Figure 5: Eflects of p21 immunodepletion on the Cdk4 inhibitory activity in extracts of ICI treated cells. Mixing assays were carried out as described in Figure 4. Prior to mixing, aliquots of ICI treated extract were subjected to two rounds of immunodepletion with anti-p21 or control antibodies (mock). A) Aliquots of IP supematants and IP pellets were immunoblotted for p21 and actin. B) Representative autoradiogram of a Cdk4 kinase assay. Lanes marked " - " were undepleted. C) Three replicate experiments were quantitated by phosphorimaging, and the fractional activity recovered from the mixed lysates relative to the activity in the E2 treated lysate was plotted as described in Figure 4. Gray bars, Cdk4 activity from undepleted extracts from ICI treated cells and mixtures thereof. Hatched bars, Cdk4 activity in mock depleted ICI treated extracts and mixtures. White bars, Cdk4 activity from p21 immunodepleted extracts and mixtures. Black bars, fractional representation of the E2 treated lysate in the mixed lysates. Error bars represent one standard deviation. 56 lysate resulted in a loss of inhibition; the amount of Cdk4 activity recovered in the mixture was approximately 60% of the amount in unmixed E2 treated lysate (Figure 4B), which is greater than the 50% predicted if no inhibitor was present. We therefore conclude that the Cdk4 inhibitory activity present in ICI-treated MCF-7 cells is the Cdkl p21. To confirm the role of p21 in regulating Cdk4 activity, additional immunodepletion/mixing experiments were performed. MCF-7 cells were pre-arrested with ICI, treated with ICI or E2, and lysates were then prepared as described above. The ICI—treated lysate was immunodepleted of p21 and mixed with the E2 treated lysate at three different mixing ratios. The mixed lysates were then subjected to Cdk4 in vitro kinase assays. Three independent experiments were performed, and the average inhibition observed in the mixed lysates was compared to the predicted value based on the proportional representation of the E2 treated lysate as described above. Aliquots of both the lysates and the Cdk4 immunoprecipitates were also analyzed by Western blotting for p21 and actin. As shown in Figure 5A, two rounds of immunodepletion qualitatively removed p21 from both the total lysate and from Cdk4 immunoprecipitates, while treatment with control antibodies (goat IgG, “mock”) had no effect on p21 levels. A representative autoradiogram from these experiments is shown in Figure 5B, and the average results from the three independent experiments are shown in Figure 5C. Consistent with the results in Figure 4, removal of p21 from the ICI treated lysate abolished the Cdk4 inhibitory activity. At each mixing ratio, the Cdk4 activity recovered was significantly higher when p21 depleted ICI-treated lysate was mixed with E2 treated lysate than when 57 non-depleted or IgG-depleted lysates were used. A one tailed t-test was performed to compare the Cdk4 activities recovered from mixtures of undepleted lysate with mixtures of IgG depleted lysates (51). The p values were 0.31, 0.14, and 0.19 for the 1 to 5, 1 to 2 and 1 to 1 mixing ratios respectively. When undepleted mixtures were compared with p21 depleted mixtures, p values were 0.015, 0.023 and 0.020. When IgG treated mixtures were compared with p21 depleted mixtures, p values were 0.017, 0.011 and 0.0013. These data confirm an important role for p21 in inhibiting Cdk4 kinase activity in antiestrogen treated human breast cancer cells. The majority of cyclin D1 in E2 treated MCF—7 cells is in complex with both p21 and Cdk4. Since cyclin D1 was co-immunodepleted with both Cdk4 and p21 antibodies from extracts of ICI treated cells, we conclude that virtually all of the cyclin D1 was present in complexes containing both Cdk4 and p21 under these conditions (Figure 4A). To investigate whether a pool of p21-free cyclin D1/Cdk4 complexes accounted for the Cdk4 activity in extracts of E2-treated cells, additional immunodepletion experiments were performed on extracts of both E2 and ICI-treated cells using antibodies directed against p21, p27 and Cdk4. As shown in Figure 6A, cyclin D1 was almost completely removed by immunodepletion of either Cdk4 or p21 in extracts of both E2 and ICI treated cells. This confirms that the majority of cyclin D1 in ICI treated MCF-7 cells is in complex with both Cdk4 and p21, and demonstrates that this is also true in E2 treated cells. Thus, even though p21 levels are decreased by E2 treatment, the p21 remaining is sufficient to co- immunoprecipitate the majority of the cyclin D1. Immunodepletion of p27 partially 58 supematants . Pellets d ID: - p21 p27 Cdk4 mock ”fund 21mm" _ ._ _ — — _ LU _. LU 232328128213 ( Cdk4“- .2... M .~ 4-Cdk4 . .- -- . a... Cyclin Dl . -- {-Cyclin DI . ' . Cdk4 < 921 «1...... '1‘ M.w.‘.+p21 p27 “-uw mm-‘ n “ +1327 K ,p27 he - 2... M +actin mock 927 ‘m ‘m g ‘4 Cdk4 “- ~ p21 Cyclin DI ’- 7". p21 -- 1327 Cyclin D1 ~ -- . Figure 6: Cdk4 complex composition in E2 and ICI treated cells. Cells were arrested with ICI for 48 h, then treated for 24 h with either ICI or E2. Extracts were prepared and immunodepleted of p21, p27 or Cdk4 as described in Figure 4, with the exception that the amount of antibody used in each depletion was doubled. Mock-depleted extracts were treated with preimmune goat IgG. A) Cell lysates and supematants from the second immunoprecipitation were analyzed by Western blotting for levels of Cdk4, cyclin D1, p21, p27 and actin. B) Pellets from the sequential immunodepletions were analyzed by Western blotting for Cdk4, cyclin D1, p21 and p27. This experiment was repeated once with similar results. The Cdk4 IP, p27 Western blot marked with an asterisk is from the second experiment. 59 removed cyclin D1 from both lysates (Figure 6A), and immunodepletion of Cdk4 lowered p27 levels. Since most of the cyclin D1 is removed by immunodepletion of p2 1 , this suggests that at least a subset of cyclin Dl/Cdk4 complexes must contain both p21 and p27. Others have suggested that there is a redistribution of p21 and/or p27 from Cdk2 to Cdk4 complexes after E2 treatment (31, 36). As shown in Figure 6A, there appears to be less p27 remaining in the supernatant following Cdk4 immunodepletion in the lysate from E2 treated cells than in lysate from ICI treated cells, which is consistent with this proposal. To test whether there is an increase in Cdkl association with Cdk4 following E2 treatment, the pellets from the sequential immunodepletions were analyzed by Western blot (Figure 6B). In lysates of both ICI and E2 treated cells, equal amounts of p21 co-immunoprecipitated with Cdk4, and equal amounts of Cdk4 and cyclin D1 co- immunoprecipitated with p21, even though the overall level of p21 was decreased in the E2 treated cells. An increase in the amount of p27 co-immunoprecipitating with Cdk4 was detected in lysates from E2 treated cells, although a high background was observed. We investigated the binding of p27 to control antibodies and found that p27 binds non- specifically to pre-immune goat IgG coated beads (Figure 6B, panel labeled “mock” ID, p27 WB). No such non-specific binding was observed for cyclin D1, Cdk4 or p21 (not shown). The increase in p27 association with Cdk4 in E2 treated cells was confirmed in a second independent experiment which is shown in the panel marked with an asterisk in Figure 6B. Since the total amount of p27 in the lysate was not significantly different in lysates prepared from E2 and ICI treated cells, the reduction of p27 in lysates from E2 60 treated cells following immunodepletion of Cdk4 and the increase in p27 co- immunoprecipitating with Cdk4 in E2 treated cells suggests that, in response to E2, p27 is redistributed to Cdk4 containing complexes from Cdk2 or some other cellular pool. Discussion The experiments described herein were designed to investigate the mechanisms by which E2 and ICI regulate Cdk4 activity in MCF-7 cells. As reported by others (2, 10, 31, 36), we observed transient increases in cyclin D1 protein levels between 6 and 18 h after E2 treatment of ICI pre-arrested cells (Figure 2C). However, this increase in cyclin D1 protein did not correlate directly with Cdk4 activity, which remained low until 24 h after E2 treatment (Figure ZB). At 24 and 30 h after E2 treatment, when Cdk4 activity was high, the levels of cyclin D1 in both the total cellular lysates and in complex with Cdk4 were indistinguishable from those seen before E2 treatment, suggesting that under these conditions, Cdk4 activity is not controlled solely by the total level of cyclin D1. In contrast to cyclin D1 , p21 levels did correlate with Cdk4 activity in both ICI and E2 treated cells (Figures 1 and 2), and the functional significance of this correlation was confirmed by a combination of lysate mixing and immunodepletion experiments. The initial mixing experiments shown in Figure 3 indicated that ICI treatment leads to Cdk4 inactivation via the induction of an inhibitor, rather than by the loss of a positive activator or by a post-translational modification. The fact that immunodepletion of p21, but not p27, removed the inhibitory activity from extracts of ICI treated cells (Figures 4 and 5) strongly implicates p21 as this inhibitor. Others have reported that p21 levels are regulated by estrogen and antiestrogens, and shown that active Cdk2 complexes are free 61 of p21 in E2 treated MCF-7 cells (9, 36, 48). In this report, we confirm that p21 levels are regulated by ER signaling, and demonstrate for the first time that p21 inhibits Cdk4 activity in antiestrogen treated MCF-7 cells. Because p21 has been reported to play both activating and inhibitory roles in regulating Cdk4 activity (5, 12, 21), and because we show that high levels of p21 protein are associated with Cdk4 inactivation, it was of interest to investigate the composition of the active Cdk4 complexes in E2 treated cells. The co—immunodepletion experiments shown in Figure 6A demonstrate that the vast majority of the cyclin D1 is present in complexes containing Cdk4 and p21 in both E2 and ICI- treated cells. This argues against a model in which there is a large pool of active, “p21-free” cyclin D1/Cdk4 complexes in E2 treated cells. However, it remains possible that the activity recovered is due to p21 free complexes which represent a small minority of the total Cdk4 complexes in the lysate, and thus have evaded our detection. In fact, a similar situation has been reported for Cdk2 in MCF-7 cells, in which active complexes that lack p21 can only be identified after biochemical fractionation (3 6). If both active and inactive Cdk4 complexes contain p21, there are several alternative mechanisms by which it may regulate Cdk4 activity. Stoichiometry of binding may determine whether p21 acts as a Cdk4 activator or inhibitor. A similar model is proposed for p21’s regulation of cyclin A/Cdk2 complexes, although there is disagreement as to its accuracy (1 , 12, 15, 26). In some of our experiments (see Figure 3C), less p21 appears to be bound to Cdk4 in E2 treated than in ICI treated cells, but this has not been observed in every experiment. For example, in Figure 6B, the amount of p21 co-precipitating with Cdk4 was indistinguishable in E2 and ICI treated cells. Since 62 p21 is a phosphoprotein (18), it is also possible that p21 may be differently modified in E2 and ICI treated cells, and that such a modification may modulate its Cdk4 inhibitory activity. Finally, a p21-associated protein might be responsible for the regulation of Cdk4 activity; a p21 associated factor has been reported which specifically modulates its inhibition of cyclin E/Cdk2 activity (8). Future studies will seek to identify how estrogen and antiestrogens regulate p21 levels, and how p21 regulates the activity of Cdk4 in response to ER signaling. Because we have identified p21 as an important mediator of antiestrogen’s effects in human breast cancer cells, we will also investigate whether a defect in p21’s inhibition of Cdk4 could contribute to the development of antiestrogen resistance. Our results clearly demonstrate that the p21 which accumulates in ICI treated cells inhibits Cdk4 activity. They also suggest that the decrease in p21 levels after E2 treatment is required for sustained Cdk4 activity. The data described above, together with previous reports, suggest that an antiestrogen-induced cell cycle arrest is due to a shift in the balance between activators (cyclins) and inhibitors (p21 or p27) of G1 Cdks. Consistent with this idea, both overexpression of cyclin D1 (29, 35, 50) and ablation of p21 or p27 expression (3) can cause G1 -)S progression in the presence of antiestrogen, at least in the short term, in MCF-7 cells. In different model systems and in clinical cases of breast cancer, different relative levels of activators and inhibitors may affect which factor limits Cdk activity and ultimately proliferation. This in turn may influence the responsiveness of ER positive breast tumors to antiestrogen therapies, and changes in the levels or functions of Cdk activators and inhibitors during treatment may account for the development of antiestrogen resistance. 63 References 10. Adkins, J. N., and K. J. Lumb. 2000. Stoichiometry of cyclin A-cyclin-dependent kinase 2 inhibition by p21Cipl/Waf1. Biochemistry 39:13925-30. Altucci, L., R. Addeo, L. Cicatiello, S. Dauvois, M. G. Parker, M. Truss, M. Beato, V. Sica, F. Bresciani, and A. Weisz. 1996. l7beta-Estradiol induces cyclin D1 gene transcription, p36D1-p34cdk4 complex activation and p105Rb phosphorylation during mitogenic stimulation of G(l )-arrested human breast cancer cells. Oncogene 12:2315-24. Cariou, S., J. C. Donovan, W. M. Flanagan, A. Milic, N. Bhattacharya, and J. M. Slingerland. 2000. Down-regulation of p21WAF1/CIP1 or p27Kip1 abrogates antiestrogen— mediated cell cycle arrest in human breast cancer cells. Proc Natl Acad Sci U S A 97:9042-6. Carroll, J. S., O. W. Prall, E. A. Musgrove, and R. L. Sutherland. 2000. A pure estrogen antagonist inhibits cyclin E-Cdk2 activity in MCF-7 breast cancer cells and induces accumulation of pl 30-E2F4 complexes characteristic of quiescence. J Biol Chem 275:38221-9. Cheng, M., P. Olivier, J. A. Diehl, M. Fero, M. F. Roussel, J. M. Roberts, and C. J. Sherr. 1999. The p21(Cip1) and p27(Kipl) CDK 'inhibitors' are essential activators of cyclin D-dependent kinases in murine fibroblasts. Embo J 18:1571- 83. Diehl, J. A., and C. J. Sherr. 1997. A dominant-negative cyclin D1 mutant prevents nuclear import of cyclin- dependent kinase 4 (CDK4) and its phosphorylation by CDK-activating kinase. Mol Cell Biol 17:7362-74. Draetta, G. F. 1994. Mammalian G1 cyclins. Curr Opin Cell Biol 6:842-6. Estanyol, J. M., M. Jaumot, O. Casanovas, A. Rodriguez-Vilarrupla, N. Agell, and O. Bachs. 1999. The protein SET regulates the inhibitory effect of p21(Cip1) on cyclin E-cyclin-dependent kinase 2 activity. J Biol Chem 274:33161-5. Foster, J. S., D. C. Henley, A. Bukovsky, P. Seth, and J. Wimalasena. 2001. Multifaceted regulation of cell cycle progression by estrogen: regulation of Cdk inhibitors and Cdc25A independent of cyclin Dl-Cdk4 function. Mol Cell Biol 21:794-810. Foster, J. S., and J. Wimalasena. 1996. Estrogen regulates activity of cyclin- dependent kinases and retinoblastoma protein phosphorylation in breast cancer cells. Mol Endocrinol 10:488-98. 64 11. 12. 13. 14. 15. 16. 17. 18. 19. 20. 21. 22. 23. Hall, M., S. Bates, and G. Peters. 1995. Evidence for different modes of action of cyclin-dependent kinase inhibitors: pl 5 and p16 bind to kinases, p21 and p27 bind to cyclins. Oncogene l 1:1581-8. Harper, J. W., S. J. Elledge, K. Keyomarsi, B. Dynlacht, L. H. Tsai, P. Zhang, S. Dobrowolski, C. Bai, L. Connell-Crowley, E. Swindell, and et a1. 1995. Inhibition of cyclin-dependent kinases by p21. Mol Biol Cell 6:387-400. Hatakeyama, M., J. A. Brill, G. R. Fink, and R. A. Weinberg. 1994. Collaboration of G1 cyclins in the functional inactivation of the retinoblastoma protein. Genes Dev 8:1759—71. Hatakeyama, M., and R. A. Weinberg. 1995. The role of RB in cell cycle control. Prog Cell Cycle Res 1:9-19. Hengst, L., U. Gopfert, H. A. Lashuel, and S. 1. Reed. 1998. Complete inhibition of Cdk/cyclin by one molecule of p21(Cip1). Genes Dev 12:3882-8. Hunter, T., and J. Pines. 1991. Cyclins and cancer. Cell 66:1071-4. Hunter, T., and J. Pines. 1994. Cyclins and cancer. 11: Cyclin D and CDK inhibitors come of age. Cell 791573-82. Jaumot, M., J. M. Estanol, O. Casanovas, X. Grana, N. Agell, and O. Bachs. 1997. The cell cycle inhibitor p21CIP is phosphorylated by cyclin A-CDK2 complexes. Biochem Biophys Res Commun 241:434-8. Kato, J. Y., M. Matsuoka, D. K. Strom, and C. J. Sherr. 1994. Regulation of cyclin D-dependent kinase 4 (cdk4) by cdk4-activating kinase. Mol Cell Biol 14:2713-21. Koff, A., and K. Polyak. 1995. p27KIPl , an inhibitor of cyclin-dependent kinases. Prog Cell Cycle Res 1:141-7. LaBaer, J ., M. D. Garrett, L. F. Stevenson, J. M. Slingerland, C. Sandhu, H. S. Chou, A. F attaey, and E. Harlow. 1997. New functional activities for the p21 family of CDK inhibitors. Genes Dev 11:847-62. Lippman, M., G. Bolan, and K. Huff. 1976. The effects of estrogens and antiestrogens on hormone-responsive human breast cancer in long-term tissue culture. Cancer Res 36:4595-601. Lukas, J ., J. Bartkova, and J. Bartek. 1996. Convergence of mitogenic signalling cascades from diverse classes of receptors at the cyclin D-cyclin-dependent kinase-pr-controlled G1 checkpoint. Mol Cell Biol 16:6917-25. 65 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. 35. Lykkesfeldt, A. E., J. K. Larsen, and I. J. Christensen. 1986. Cell cycle analysis of estrogen stimulation and antiestrogen inhibition of growth of the human breast cancer cell line MCF-7. Breast Cancer Res Treat 7:S83-90. Matsushime, H., D. E. Quelle, S. A. Shurtleff, M. Shibuya, C. J. Sherr, and J. Y. Kato. 1994. D-type cyclin-dependent kinase activity in mammalian cells. Mol Cell Biol 14:2066-76. Nakanishi, M., Y. Kagawa, H. Takahashi, and H. Matsushime. 1997. Two different bindings of p21 Cdk inhibitor to cyclin/Cdk complex. Leukemia 11 Suppl 3:356-7. Osborne, C. K., D. H. Boldt, G. M. Clark, and J. M. Trent. 1983. Effects of tamoxifen on human breast cancer cell cycle kinetics: accumulation of cells in early G1 phase. Cancer Res 43:3583-5. Osborne, C. K., D. H. Boldt, and P. Estrada. 1984. Human breast cancer cell cycle synchronization by estrogens and antiestrogens in culture. Cancer Res 44:1433-9. Pacilio, C., D. Germano, R. Addeo, L. Altucci, V. B. Petrizzi, M. Cancemi, L. Cicatiello, S. Salzano, F. Lallemand, R. J. Michalides, F. Bresciani, and A. Weisz. 1998. Constitutive overexpression of cyclin D1 does not prevent inhibition of hormone-responsive human breast cancer cell growth by antiestrogens. Cancer Res 58:871-6. Parry, D., D. Mahony, K. Wills, and E. Lees. 1999. Cyclin D-CDK subunit arrangement is dependent on the availability of competing INK4 and p21 class inhibitors. Mol Cell Biol 19:1775-83. Planas-Silva, M. D., and R. A. Weinberg. 1997. Estrogen-dependent cyclin B- cdk2 activation through p21 redistribution. Mol Cell Biol 17:4059-69. Polyak, K., M. H. Lee, H. Erdjument-Bromage, A. Koff, J. M. Roberts, P. Tempst, and J. Massague. 1994. Cloning of p27Kip1 , a cyclin-dependent kinase inhibitor and a potential mediator of extracellular antimitogenic signals. Cell 78:59-66. Poon, R. Y., and T. Hunter. 1995. Dephosphorylation of Cdk2 Thr160 by the cyclin-dependent kinase- interacting phosphatase KAP in the absence of cyclin. Science 270:90-3. Poon, R. Y., W. Jiang, H. Toyoshima, and T. Hunter. 1996. Cyclin-dependent kinases are inactivated by a combination of p21 and Thr-14/Tyr-15 phosphorylation after UV-induced DNA damage. J Biol Chem 271:13283-91. Prall, O. W., E. M. Rogan, E. A. Musgrove, C. K. Watts, and R. L. Sutherland. 1998. c-Myc or cyclin D1 mimics estrogen effects on cyclin E-Cdk2 activation and cell cycle reentry. Mol Cell Biol 18:4499-508. 66 36. 37. 38. 39. 40. 41. 42. 43. 44. 45. 46. 47. 48. Prall, O. W., B. Sarcevic, E. A. Musgrove, C. K. Watts, and R. L. Sutherland. 1997. Estrogen-induced activation of Cdk4 and Cdk2 during Gl-S phase progression is accompanied by increased cyclin D1 expression and decreased cyclin-dependent kinase inhibitor association with cyclin E- Cdk2. J Biol Chem 272:10882-94. Quelle, D. E., R. A. Ashmun, G. J. Harmon, P. A. Rehberger, D. Trono, K. H. Richter, C. Walker, D. Beach, C. J. Sherr, and M. Serrano. 1995. Cloning and characterization of murine p16INK4a and pl 51NK4b genes. Oncogene 11:635-45. Rao, 8., J. Gray-Bablin, T. W. Herliczek, and K. Keyomarsi. 1999. The biphasic induction of p21 and p27 in breast cancer cells by modulators of cAMP is posttranscriptionally regulated and independent of the PKA pathway. Exp Cell Res 252:211-23. Reed, S. I. 1997. Control of the Gl/S transition. Cancer Surv 29:7-23. Rose, C., S. M. Thorpe, J. Lober, J. L. Daenfeldt, T. Palshof, and H. T. Mouridsen. 1980. Therapeutic effect of tamoxifen related to estrogen receptor level. Recent Results Cancer Res 71 :134-41. Rutqvist, L. E., B. Cederrnark, U. Glas, H. Johansson, B. Nordenskjold, L. Skoog, A. Somell, T. Theve, S. F riberg, and J. Askergren. 1987. The Stockholm trial on adj uvant tamoxifen in early breast cancer. Correlation between estrogen receptor level and treatment effect. Breast Cancer Res Treat 10:255—66. Sherr, C. J. 1996. Cancer cell cycles. Science 274: 1672-7. Sherr, C. J. 2000. The Pezcoller lecture: cancer cell cycles revisited. Cancer Res 60:3689-95. Sherr, C. J ., J. Kato, D. E. Quelle, M. Matsuoka, and M. F. Roussel. 1994. D-type cyclins and their cyclin-dependent kinases: G1 phase integrators of the mitogenic response. Cold Spring Harb Symp Quant Biol 59:11-9. Sutherland, R. L., M. D. Green, R. E. Hall, R. R. Reddel, and I. W. Taylor. 1983. Tamoxifen induces accumulation of MCF 7 human mammary carcinoma cells in the GO/Gl phase of the cell cycle. Eur J Cancer Clin Oncol 19:615-21. Toyoshima, H., and T. Hunter. 1994. p27, a novel inhibitor of G1 cyclin-Cdk protein kinase activity, is related to p21. Cell 78267-74. Vidal, A., and A. Koff. 2000. Cell-cycle inhibitors: three families united by a common cause. Gene 247: 1-15. Watts, C. K., A. Brady, B. Sarcevic, A. deFazio, E. A. Musgrove, and R. L. Sutherland. 1995. Antiestrogen inhibition of cell cycle progression in breast 67 49. 50. 51. 52. cancer cells in associated with inhibition of cyclin-dependent kinase activity and decreased retinoblastoma protein phosphorylation. Mol Endocrinol 9: 1 804-13. Watts, C. K., K. J. Sweeney, A. Warlters, E. A. Musgrove, and R. L. Sutherland. 1994. Antiestrogen regulation of cell cycle progression and cyclin D1 gene expression in MCF-7 human breast cancer cells. Breast Cancer Res Treat 31 :95- 105. Wilcken, N. R., O. W. Prall, E. A. Musgrove, and R. L. Sutherland. 1997. Inducible overexpression of cyclin D1 in breast cancer cells reverses the growth- inhibitory effects of antiestrogens. Clin Cancer Res 3:849-54. Williams, F. 1986. Reasoning With Statistics: How to Read Quantitative Research, 3rd ed. Holt, Rinehart and Winston, New York. Yan, Y., and M. C. Mumby. 1999. Distinct roles for PPl and PP2A in phosphorylation of the retinoblastoma protein. PP2a regulates the activities of G(l) cyclin- dependent kinases. J Biol Chem 274:31917-24. 68 CHAPTER THREE Induction of Cyclin D1 in MCF-7 Cells Supports Short Term Proliferation in the Presence of Antiestrogen 69 Chapter 3: Induction of Cyclin D1 in MCF-7 Cells Supports Short Term Proliferation in the Presence of Antiestrogen ABSTRACT Amplification of the cyclin D1 gene and/or overexpression of its protein product is a common feature of clinical breast cancer, and experimental evidence suggests that cyclin D1 overexpression may allow estrogen receptor positive breast cancer cells to overcome the G1 arrest imposed by antiestrogen treatment in vitro. To determine the mechanisms by which cyclin D1 stimulates proliferation in the presence of antiestrogen and to examine the potential contribution of cyclin D1 overexpression to antiestrogen resistance, we describe a sub-clone of the MCF-7 human breast cancer cell line engineered to conditionally express epitope tagged cyclin D1 upon the addition of a synthetic drug. Using this novel gene regulatory system, we show that ectopic cyclin D1 expression can be tightly regulated, and that cyclin D1 overexpression is sufficient to allow cells arrested in G1 phase by antiestrogen to enter S phase. Ectopic cyclin D1 can activate Cdk4 in the presence of antiestrogen, but does not sustain long term Cdk4 activity or proliferation. Ectopic cyclin D1 expression delayed, but did not prevent, antiestrogen mediated induction of the Cdk inhibitor p21m‘mCipl and inhibition of Cdk4 activity in asynchronous cells. These data suggest that cyclin D1 overexpression may contribute to antiestrogen resistant growth of breast Cancer cells, but that additional alterations, such as bypassing p21wamcipl, may be necessary for the acquisition of a stable antiestrogen resistant phenotype. 70 Introduction Cyclin D1, along with cyclins D2 and D3, are the regulatory partners of cyclin dependent kinase 4 (Cdk4)2, a serine threonine kinase that phosphorylates the retinoblastoma protein (Rb) (24, 28). Cyclin D1 association with Cdk4 facilitates Cdk4 nuclear localization and phosphorylation by the Cdk activating kinase (CAK) (11). Hyperphosphorylated Rb contributes to cellular proliferation by relieving transcriptional repression of E2F regulated genes (4, 46). The cyclin Dl/Cdk4 complex can also associate with one or more cyclin dependent kinase inhibitor (Cdkl) such as p2 lwa'mCipl (p21)(12, 19). Although p21 was originally identified as an inhibitor of cyclin D/Cdk4 activity(49) and it clearly plays this role in the context of ER signaling (41), recent work has suggested that p21 may also facilitate cyclin D association with Cdk4 and enhance catalytic activity (8, 50). Recent investigations have suggested that cyclin D1 may be involved in the development and progression of breast cancer. Cyclin D1 is encoded by the CCNDl gene, located on chromosome 1 1q13, and this locus is amplified in approximately 15% of breast tumors (13, 23, 37). Cyclin D1 protein overexpression is more common, occuring in one third of breast cancers, suggesting that gene amplification is not the only mechanism by which cyclin D1 protein expression could be increased (15, 51). Gene amplification and protein overexpression both correlate positively with estrogen receptor (ER) expression in breast cancer (13, 22, 42, 43, 51). 2Abbreviations used are: Cdk, cyclin dependent kinase. ER, estrogen receptor. p21, WAFl/Cipl. p27, Kipl. p16, INK4a. Cdkl, Cdk inhibitor. Rb, retinoblastoma protein. E2, l7B-estradiol. AP, AP1510. IP, immunoprecipitation. 26D, MCF-7-26D cell line. ICI, 1C1 182,780. DTT, dithiothreotol. IMEM, improved modified Eagle’s medium. EGF-R, epidermal growth factor receptor. CAMP, cyclic adenosine monophosphate. CSS, charcoal stripped serum. Minutes, m. Hours, h. FBS, fetal bovine serum. 71 The significance of cyclin D1 in breast cancer is unclear. Positive immunostaining and cyclin D1 gene amplification are reported both to predict improved relapse free survival (6, 36), and to have no association with tumor grade or prognosis (42, 43). Elevated cyclin D1 mRN A and positive immunostaining correlated with a poor prognosis if in combination with positive immunostaining for ERa, Rb or epidermal growth factor receptor (EGF-R) (25, 29). These inconsistent results suggest that in breast tumors, other factors may influence cyclin D1 ’3 effects on the course of disease, if any. Cell lines whose proliferation is dependent on estrogen have been utilized to examine the relationship between cyclin D1 and ER signaling. Cyclin D1 mRN A and protein are upregulated following E2 treatment of the breast adenocarcinoma derived MCF-7 cell line, pre-arrested either by serum deprivation or antiestrogen treatment (1 , 14, 27, 39), and E2 treatment causes cyclin D1 mRNA induction and Cdk4 activation in E2 responsive tissue in vivo (2). Antiestrogen treatment is reported to reduce cyclin D1 mRN A and protein (44, 45), and this reduction precedes observable effects on Rb phosphorylation and cell cycle, suggesting a causative role in inhibiting proliferation. E2 affects cyclin D1 transcription in MCF-7 and ZR-75 cells in a protein kinase-A dependent manner through a CRE like sequence, and transcription is enhanced by binding of c-Jun/ATF 2 heterodimers to these sequences. Cyclin D1 expression is also controlled by ERa interaction with Spl transcription factors at Spl binding sites upstream of the promoter (7, 35, 40). Interestingly, cyclin D1 has been shown to directly interact with ERoc and enhance its transcriptional activity in the absence of ligand, and this interaction is enhanced by cAMP (26, 32, 52, 53). 72 The link between estrogen and cyclin D1 in human breast cancer, and cyclin D1 ’8 known role as a mediator of proliferation, led some to hypothesize that overexpression of cyclin D1 would confer estrogen independent growth in cell lines derived from breast tissue. In the HBL-100 cell line, derived from normal mammary tissue, stable transduction with cyclin D1 inhibited proliferation and tumorigenicity (18), while in the breast tumor derived MCF-7 and T47D cell lines, conditional expression of ectopic cyclin D1 caused cells arrested by serum deprivation or antiestrogen treatment to activate cyclin E/Cdk2 complexes and enter the cell cycle (31, 38, 47). Although ectopic cyclin D1 expression caused arrested cells to progress from G1 to S phase, it was not sufficient to support long term growth in the presence of antiestrogens (34). In the current work, we address the apparent paradox that cyclin D overexpression causes G1 arrested breast cancer cells to progress to S phase in the presence of antiestrogen but does not support long term proliferation in the presence of antiestrogen. Utilizing a novel gene regulation system (3), Cunjie Zhang developed an MCF-7 derived cell line that expresses an epitope tagged cyclin D1 protein upon addition of an otherwise innocuous drug to the culture media. Consistent with previous reports, we show that induction of cyclin D1 does cause cell cycle progression in antiestrogen arrested cells, but is insufficient to promote long term growth in the presence of antiestrogen. The ectopic cyclin D1 is able to activate Cdk4 initially, however, Cdk4 activity is downregulated and returns to basal levels within 72 h post treatment, despite the presence of abundant ectopic cyclin D1 in stable association with Cdk4. We further demonstrate that the level of the Cdkl p21, which is downregulated after antiestrogen arrested cells are treated with estrogen, remains high in the presence of antiestrogen despite ectopic cyclin D1 73 expression. These data suggest that ER ligands exert control over the passage of MCF-7 cells through the G1 checkpoint by changing the balance between activators and inhibitors of G1 Cdk complexes, and that cyclin D1 overexpression alone is an unlikely cause of antiestrogen resistance in the absence of other changes. Materials and Methods Cell lines and culture conditions. MCF-7-26D cells were generated in two steps from MCF-7 cells using Ariad Pharmaceuticals’ AP1510 gene regulation system (3) by Ms. Cunjie Zhang. MCF-7-7-6 cells were generated by transfection of MCF-7 cells with pCEN-F3p65/Z1F3; this is an expression vector encoding a G418 selectable marker, a DNA binding domain fused to multiple FKBP (FK506 binding protein) domains, and a transactivation domain fused to multiple FKBP repeats. MCF-7-7-6 cells were then stably transfected with pLH-ZlZ-I-CCNDl—F LAG, which encodes hygromycin resistance gene and a FLAG epitope tagged human cyclin D1 gene downstream of repeated unique DNA sequence elements recognized by the DNA binding domain encoded by the first plasmid. After selection in hygromycin, one cell line, designated MCF-7-26D, exhibited tight regulation of ectopic cyclin D1 expression in response to the dimerizing drug AP1510 and was chosen for further charaterization. MCF-7-26D cells were cultured at 37° with 5% C02 in IMEM (BioFluids) with 5% fetal bovine serum (FBS, HyClone) and penicillin/streptomycin (Gibco) and 10 pg/ml hygromycin (Gibco). Experiments were performed in phenol red free IMEM (BioFluids) alone or containing 5% charcoal stripped serum (CSS, HyClone), 10 nM 17B-estradiol (E2, Sigma), 10 nM ICI 182780 (ICI, 74 AstraZeneca), and/or 300 nM AP1510 (AP, Ariad Pharmaceutical) as indicated. For experiments involving long term culture (Figure 4), media were changed at 72 h after initial treatment. Cell cycle analysis. Cells were trypsinized, washed in phosphate buffered saline (PBS), suspended in PBS+10% FBS, fixed with 80% cold ethanol and stored at —20° C. Prior to analysis, cells were washed twice with PBS, then suspended in PBS + 1 mg/ml RNaseA, 0.2 mg/ml propidium iodide, 0.5 mM EDTA and 0.1% Triton-X-lOO. Cells were then analyzed for red fluorescence on a F ACSVantage flow cytometer; cell cycle distribution was determined using ModF it and WinCycle LT software. Immunoprecipitations and kinase assays. Immunoprecipitations (IPs) and Cdk activity measurements were performed using modifications of a published method (28). Unless indicated otherwise, all manipulations were carried out on ice. Cells were washed twice in ice cold PBS, harvested by scraping in PBS and pelleted; the pellets were frozen and stored in liquid nitrogen until the day of analysis. Cells were resuspended and lysed by sonication in IP buffer (50 mM HEPES pH 7.5, 150 mM NaCl, 1 mM EDTA, 2.5 mM EGTA, 0.1% Tween-20, 10% glycerol, 1 mM DTT, 10 mM B-glycerophosphate, 1 mM NaF, lmM NaVO4, 0.1 mM PMSF, 10 pg/ml leupeptin, and 2 pg/ml aprotinin). The lysates were cleared by centrifugation, and protein concentrations were quantitated using BioRad’s protein assay reagent. For IPs, 75 pg of total protein was diluted to 500 pl with IP buffer, then incubated for 60 minutes (m) rocking at 4° C with 1.5 pg antibody (anti- Cdk4 H-22-G, anti-Cdk2 M2, or normal goat IgG, all from Santa Cruz) bound to 7.5 pl 75 protein G agarose beads (Boehringer Mannheim). Beads were pelleted and washed four times with 100 pl 1P buffer and twice with 100 pl kinase buffer (50 mM HEPES pH7.5, 10 mM MgC12, 1 mM DTT, 2.5 mM EGTA, 10 mM B-glycerophosphate, 0.1 mM NaVO4, and 1 mM NaF). Pellets were then suspended in 40 pl kinase buffer containing 10 pCi y-[32P]ATP, 20 pM cold ATP, and 1.0 pg Histone H1 (Sigma) or 1.0 pl glutathione sepharose 4B (Amersham Pharmacia) to which approximately 2.0 pg of a fusion protein between glutathione-S-transferase (GST) and amino acids 792 to 928 of human Rb were bound (GST-Rb bacterial expression vector was kindly provided by Dr. William Kaelin of the Dana Farber Cancer Institute). The reactions were incubated at 30° C for 30 m with occasional mixing, after which they were boiled in SDS loading buffer containing mercaptoethanol. Beads were pelleted and the supematants transferred to clean tubes. Aliquots of the reaction products were resolved on 10% SDS polyacrylamide gels, which were then dried and exposed to autoradiography film. The data were quantitated by phosphorimaging with a Storm phosphorimager (Molecular Dynamics) using ImageQuant software. Western blotting. Either 20 pg total cell lysates or aliquots of IP pellets were resolved on 12% SDS-polyacrylamide gels, transferred to membranes and probed with antibodies for cyclin D1 (UB1 06 137, Upstate Biotechnology), p21wan (p21-c-19, Santa Cruz Biotechnology), cyclin A (14531C, Pharmingen) or actin (Sigma clone AC-40). Membranes were then incubated with horseradish peroxidase conjugated goat anti-rabbit (BioRad) or goat anti-mouse (American Qualex) secondary antibodies, and 76 immunoreactive proteins were detected using Super Signal West Pico Chemiluminescent Substrate (Pierce). Results Generation of MCF - 7-26D cells. Cells which express ectopic, FLAG tagged cyclin D1 from a drug inducible promoter were generated using ARIAD Pharmaceutical’s AP1510 expression regulation kit (3) by Ms. Cunjie Zhang prior to the author’s arrival in the Conrad lab. MCF-7-7-6 cells were generated by stable transfection of MCF-7 cells with a tricistronic expression plasmid encoding a selection marker, a DNA binding domain fused to multiple FKBP domains, and a transactivation domain fused to multiple FKBP domains. Upon addition of the FK506 derivative AP1510 (AP), the two fusion proteins dimerize and become a potent transcriptional activator at promoters containing unique DNA sequences recognized by the DNA binding domain — FKBP fiision protein (Figure 1A). MCF-7-26D cells were generated by stable transfection of MCF-7-7-6 cells with an expression plasmid encoding a FLAG epitope tagged cyclin D1 under control of a minimal IL-2 promoter downstream of repeated unique sequence motifs recognized by the DNA binding domain encoded by the first plasmid. A diagram of the strategy for regulating ectopic cyclin D1 expression is shown in Figure 1A. Upon addition of AP to the culture media, MCF-7-26D cells (26D cells) express ectopic cyclin D1 that is recognized by antibodies to wild type human cyclin D, but distinguishable from endogenous cyclin D due to its shified mobility in electorphoretic gels (Figure 18). 77 It A- 'eguamy —CMVJ:I DNAB H TA H neo I—— plasmid: l l target plasmid: leZFHDl — IL-2 ——lflagl CCNDl l——— addition of dimerizer: 8' FBS FBS+AP c - 2: cyclin D1* cyclin D Figure 1: Construction of MCF - 7-26-D cells. A) Schematic of regulatory system used to induce expression of flag-tagged cyclin D1 in breast cancer cells. The regulatory plasmid encodes two fusion proteins whose drug induced dimerization leads to transcriptional activation of the target gene. CMV, cytomegalovirus promoter. DNAB, DNA binding domain - FKBP fusion protein. TA, transactivation domain - FKBP fusion protein. Neo, G418 resistance cassette. Hygro, hygromycin resistance cassette. 12xZFHD1, unique DNA sequence repeats recognized by DNAB. IL-2, minimal interleukin-2 promoter. Flag, flag epitope tag. CCNDl, human cyclin D1 cDNA. AP, AP1510 dimerizer. RNAP, RNA polymerase. B) Regulated expression of flag-tagged cyclin D1 in MCF-7-26D cells. Cells were treated with either PBS or FBS plus the dimerizer AP1510 for 24 h. Aliquots of lysates were Western blotted with a polyclonal antibody which recognizes human cyclin D. The slower migrating band is ectopic, flag tagged cyclin D1. 78 Cell cycle eflects of cyclin D1 induction. To assess the effects of ectopic cyclin D1 expression on breast cancer cells’ ability to proliferate in the presence of antiestrogen, an experiment was conducted in which 26D cells were pre-arrested with ICI in the presence of charcoal stripped serum (CSS). The arrested cells were then washed and treated with either E2 or ICI+AP1510, harvested at intervals over 30 h, and fixed in ethanol. Cells were then stained with propidium iodide and their DNA content evaluated by flow cytometry (Figure 2A). Prior to treatment (0 h), the cells were effectively arrested in the G1 phase of the cell cycle. Estrogen treatment caused cells to re-enter the cell cycle by 24 h, and induction of cyclin D1 in the continued presence of ICI resulted in a 2.5 fold greater increase in the percentage of cells in S phase. The fraction of cells entering S phase after 24 h AP+ICI treatment varied among numerous experiments, but was generally comparable to that achieved by E2 treatment without ICI. This suggests that the amount of cyclin D1 protein may limit proliferation in the presence of antiestrogen. We next asked whether ectopic cyclin D1 expression could override ICI’s effects in the absence of other growth factors present in serum. Cells were plated in 5% fetal bovine serum (FBS) for 24 h to allow attachment. The cells were then washed and incubated in serum free media (SFM) for 48 h to induce quiescence, after which they were washed and treated with SFM plus ICI, E2, AP and CSS, alone and in combination, for 24 h. Cells were then harvested and their cell cycle phase distributions determined by flow cytometry as described above. As indicated in Figure 2B, serum deprivation caused cell cycle arrest in 26D cells, with only 5% of cells in S phase. The addition of ICI did not further depress the fraction of cells in S phase. Treatment with E2 alone induced 79 .> I E2 [:1 ICI+AP U1 0 b O N 0 percent of cells in S phase (A) C d O 0 h 6 h 24 h 30 h time after treatment percent of cells in S phase SFM SFM SFM SFM SFM SFM C55 C55 +ICI +E2 +AP +AP +AP +E2 +ICI +E2 Figure 2: Effects of cyclin D1 expression on growth arrested cells. A) 26D cells were pre-arrested with ICI for 48 h, then treated with either E2 or ICI+AP. Cells were harvested at intervals over 30 h and analyzed for cell cycle distribution by flow cytometry. The average of two samples is shown; results are representative of three repeated experiments. B) 26D cells were plated in 5% FBS, arrested by serum deprivation for 48 h, then treated for 24 h with serum free media (SFM) alone or with ICI, E2, or AP as indicated. Cells were also treated with 5% charcoal stripped serum (CSS) alone and in combination with E2. Cells were collected and the percentage of cells in S phase determined by flow cytometry. The average of three samples is shown; error bars indicate one standard deviation. 80 proliferation, with nearly 30% of cells in S phase. Induction of ectopic cyclin D1 via treatment with AP caused a lesser degree of proliferation that was unaffected by the addition of ICI, with approximately 17% of cells in S phase in both cases. Samples treated with AP+E2 had a higher percentage of cells in S phase than samples treated with either alone. The addition of CSS alone caused a slight increase in the percentage of cells in S phase, and, as expected, the most robust proliferation was observed with the addition of CSS and E2. These data suggest that in the absence of serum growth factors, cyclin D1 levels limit proliferation, but that both E2 and serum have additional targets which promote proliferation in breast cancer cells. Activation of endogenous Cdk4 by ectopic cyclin D1. Inducing FLAG tagged cyclin D1 in 26D cells caused cell cycle progression in the presence of the antiestrogen ICI. We hypothesized that this was due to the ectopic cyclin D1 binding to and activating Cdk4, which then phosphorylated substrates critical for the initiation of DNA synthesis, such as Rb. To test this hypothesis, 26D cells were pre-arrested in ICI for 48 h, washed, and treated with either E2 or ICI + AP. Cells were harvested at intervals over 30 h, afier which whole cell lysates were prepared and analyzed using a Cdk4 in vitro kinase assay in which Cdk4 is immunoprecipitated and incubated with recombinant Rb and radiolabeled ATP. E2 treatment caused a dramatic increase in Cdk4 activity that was evident by 24 h after treatment (Figure 3A). Induction of cyclin D1 in the continued presence of ICI also activated Cdk4 with somewhat faster kinetics than E2 treatment. To test whether ectopic cyclin D1 could be detected in complex with Cdk4, lysates and reaction products from 81 c_L 2 . 0 hours after E2 L59» ICI+AP 9 treatment: 0 6 12 24 30 24 6 12 24 3O 24 GST-Rb —) .1... u I! I. B. 50 :'__ I E2 40 __ I ICI+AP N O fold induction U0 0 _: O o 6 12 24 30 hours after treatment C. lysates Cdk4 pellets 2 0 1224 12 24 012 24241224 cyclin D13 “M” m .9 actin -> ”was!“ 0 E2 ICI+AP E2 2‘ ICI+AP Figure 3: Ectopic cyclin D1 activates Cdk4 in the presence of antiestrogen. 26D cells were pre-arrested with ICI for 48 h, then treated with either E2 or ICI+AP and harvested at intervals over 30 h. Whole cell lysates were prepared and quantitated. A) Aliquots of lysates were immunoprecipitated (IPd) with anti-Cdk4 or control (IgG) antibodies. Pellets were reacted with radiolabeled ATP and recombinant Rb substrate (GST-Rb). Products were resolved on gels and visualized by autoradiography. B) Substrate bands from (A) were quantitated by storage phosphorimaging. Band intensity is plotted relative to the untreated, 0 h sample. C) Aliquots of lysates (left panels) and Cdk4 co-IPs (right panel) were Western blotted for human cyclin D1 and actin. The slower migrating band in the cyclin D1 panel is ectopic, FLAG tagged cyclin D1. The data shown in this experiment was repeated in a second experiment. 82 the kinase assay were Western blotted for cyclin D1 and actin. E2 treatment caused an increase in cyclin D1 both in total and in complex with Cdk4 at 12 h after treatment, but levels returned to pre-treatment levels by 24 h. Treatment with AP induced expression of the slower migrating ectopic cyclin D1 at 12 h, and expression was sustained until 24 h. The amount of ectopic cyclin D1 in complex with Cdk4 decreased from 12 h to 24 h, and expression of ectopic cyclin D1 appeared to decrease the amount of endogenous cyclin D1 both in total and in complex with Cdk4. Interestingly, although more cyclin D1 was in complex with Cdk4 at 12 h in AP + ICI treated cells, activation was not observed until 24 h, when levels were lower. Although cyclin D1 may be limiting for Cdk4 activation, increasing the amount of cyclin D1 in complex with Cdk4 does not cause immediate activation of the complex, suggesting that further modification of the Cdk4 complex must occur prior to activation. ICI treatment and ectopic cyclin D1 expression in asynchronous cells. The data presented above demonstrate that ectopic cyclin D1 can activate Cdk4 in the presence of ICI after a two day ICI pre-arrest. We next tested whether ectopic cyclin D1 could sustain Cdk4 activity in proliferating cells treated simultaneously with ICI and AP. 26D cells were plated in 5% FBS for one day, then treated with E2 for 48 h. Cells were then washed and treated with either E2, ICI, or ICI + AP and harvested at intervals over 30 h. Cellular lysates were prepared and analyzed by Cdk4 and Cdk2 kinase assay and Western blotting. The addition of fresh E2 to proliferating 26D cells caused a modest increase in both Cdk4 and Cdk2 activities at 30 h (Figure 4). ICI treatment caused a decrease in 83 A' Cdk4 IP hours after ICI ICI+AP E2_ treatments 0 6 18 30 6 18 30 30 303 _ 196 IP drum: W.» rm- IWWW m- . foldchange 101.5 0602 14 1..305 1301 B. Cdk2 IP % 2 hours after 9 g E2 :33 treatment: 0 30 3o 30 30 HistoneH13’ ‘ fold change: ~- 1 O OO 2 O. 5 1.9 O.2 C. ICI ICI+AP E2 9 618 39 618 .3930 cyclinD “mum‘s-4...... p21 «4...... _ 2.12:3. J - ‘.-?~r-,-.\r.w'J‘mr‘-.-.-“i';rx""Hen-w. U. *1 .. ' -- Figure 4: Expression of ectopic cyclin D1 delays, but does not prevent, inhibition of Cdk4 and Cdk2 activity by antiestrogen. 26D cells were treated with E2 for two days to generate an asynchronous population. Cells were then treated with either ICI or ICI+AP and harvested at intervals over 30 h. Whole cell lysates were prepared, quantitated and analyzed for G1 Cdk activity and cyclin D, p21 and actin levels. A) Autoradiogram showing Cdk4 in vitro kinase assay performed as described. Numbers below lanes indicate the fold change relative to the 0 h activity as quantitated by storage phosphor imaging. B) Autoradiogram showing histone H1 kinase activity of Cdk2 or control (IgG) immunoprecipitates. Numbers below lanes indicate fold change relative to the 0 h activity. C) Aliquots of lysates were Western blotted for cyclin D, p21, and actin. 84 Cdk4 activity by 18 h, and both Cdk4 and Cdk2 activities were at background levels by 30 h. Consistent with our earlier observations in MCF-7 cells (41), Cdk4 activity decreased after ICI treatment without a corresponding decrease in cyclin D levels. Cells treated with ICI + AP sustained high levels of Cdk4 activity through 18 h, but activity was reduced to approximately half by 30 h, as was Cdk2 activity. The decrease in Cdk4 activity occurred despite continued high levels of ectopic cyclin D1. However, while levels of the Cdkl p21 increased when cycling 26D cells were treated with ICI alone, correlating with Cdk4 inactivation, there was a delayed and less dramatic increase in p21 when ICI was combined with cyclin D1 induction (Figure 4C). Cyclin D1 delayed, but did not prevent, inhibition of GI Cdks in asynchronous cells treated with ICI, correlating with delayed induction of p21. Failure of ectopic cyclin D1 to support long term proliferation or Cdk4 activity in antiestrogen. Others have reported that ectopic cyclin D1 expressed using a tetracycline inducible system is insufficient to promote long term proliferation in antiestrogen treated breast cancer cells (34). To confirm this finding, and to examine the mechanisms responsible, we examined the effects of inducing cyclin D1 in the presence of ICI over the course of several days treatment. 26D cells were pre-arrested in ICI as described, then treated with either E2 or ICI+AP. Cells were collected at 24 h intervals over four days and analyzed for cell cycle distribution by flow cytometry as described. As indicated in Figure 5A, cells were efficiently arrested prior to treatment, with 78% of cells in G1 phase. Consistent with our earlier results (Figure 2A), both E2 and ICI+AP treated cells entered the cell cycle by 24 h after treatment, with E2 treated cells reduced 85 .> § ; m —~ 5 80 a 301 E2 o. 1 .1: 1 G 70 1 5} j I ICI+AP : 60 1 E Z '=: 50 £203 ' 8 40 3”: . 1 :6 30 8 1 5810 ,, g 1 0 24 72 96 0 24 48 72 96 hours after treatment hours after treatment B E E o 0 K9 E2 9 ICI+AP 9 lCl hours after —— treatment: f 0 24 72 96 120' 24 :1. 72 96.120. 24. ‘29... GST—Rb —) C. lysates Cd k4 Co-IP hours after ,E2 ,2, ,, ,lCliAP ICI 132 ilCl+AP ,, ICI treatment: 0 24 2296120 23.21296 120 120 0 24 72 96 120 24 72 96 120 120 c. actin mu“ , Figure 5: Expression of exogenous cyclin D1 does not support long term proliferation or Cdk4 activation in the presence of antiestrogen. A) 26D cells were pre-arrested in ICI, then treated with either E2 or ICI+AP and harvested at 24 h intervals for 96 h. Cell cycle phase distribution was determined by flow cytometry as described. The average of three samples is shown; error bars indicate standard deviation. B) 26D cells were pre-arrested in ICI, treated with either E2, ICI+AP, or ICI and harvested at the indicated times afier treatment. Lysates were prepared, quantitated, and Cdk4 in vitro kinase activity was determined as described. C) Aliquots of lysates and Cdk4 IP pellets from (B) were Western blotted for cyclin D, p21 and actin. 86 to 46% G1 phase and ICI+AP treated cells to 60% G1 phase; both treatments showed a corresponding increase in the percentage of cells in S phase (not shown). The fraction of G1 phase cells in both treatments increased after 24 h. While the percentage of G1 phase cells leveled off at approximately 60% in E2 treated cells, consistent with an asynchronous population, ICI+AP treated cells appeared to re-arrest in G1 phase by 96 h after treatment, with 80% of cells in G1 phase. These data are consistent with others’ observations that induction of ectopic cyclin D1 in the presence of antiestrogen can cause progression from G1 to S phase, but is insufficient to sustain long term cell growth (34, 38, 47, C. Zhang, unpublished observations). Induction of cyclin D1 did not prevent, but appeared to delay, the inhibition of Cdk4 activity and the increase in p21 levels seen after ICI treatment of proliferating cells (Figure 4). Previously, we showed that p21 acts as an ICI regulated inhibitor of Cdk4 in MCF-7 cells (41). We therefore hypothesized that the failure of ectopic cyclin D1 to support long term proliferation was due to the inability of ectopic cyclin D1 — Cdk4 complexes to overcome inhibition by p21 after an initial activation. To test whether Cdk4 was inactivated after long term induction of ectopic cyclin D1 , 26D cells were pre- arrested in ICI and then treated with either E2 or ICI+AP. Cells were harvested at intervals over five days and analyzed by Cdk4 kinase assay and Western blotting (Figure 5B, C). E2 treatment caused activation of Cdk4 by 24 h after treatment, and activity was sustained for 120 h; this activation was not accompanied by any changes in the level of total cyclin D1 protein, although there was a transient increase in Cdk4 associated cyclin D1 at 24 h post treatment. Levels of p21 in the lysates decreased by 24 h after E2 treatment and continued to decrease through 120 h. 87 Induction of ectopic cyclin D1 in the presence of ICI caused activation of Cdk4 to about half the level of E2 treated cells by 24 h after treatment in this experiment. Unlike E2 treated cells, in which Cdk4 activity was sustained, in ICI+AP treated cells Cdk4 activity began to decrease by 72 h after treatment and was undetectable by 96 h, despite the presence of abundant ectopic cyclin D1 in the lysate and in complex with Cdk4. Levels of p21 decreased to approximately the same level as with E2 treatment by 24 h after treatment, but then increased gradually over the 120 h, coinciding with Cdk4 inactivation. These data suggest that the failure of ectopic cyclin D1 to support long term cell growth in ICI is due to its inability to sustain activation of Cdk4, and that this is due to increased and/or maintained levels of p2 1. This phenomena was observed in two independent experiments. Interestingly, when cells were treated with ICI alone for 120 h (in addition to the 48 h pre-arrest), endogenous cyclin D1 and p21 were not detectable in the lysate, and cyclin D was barely detectable in complex with Cdk4; no Cdk4 activity was detected in this sample (Figure 5B, rightmost lane). This suggests that p21 and cyclin D1 are coordinately regulated in long term ICI treated cells, or, alternatively, that cell death pathways have been activated which cause non-specific protein degradation. Discussion Breast cancer is an important disease whose etiology is associated with exposure to estrogen and with increased expression of cyclin D1. The work described above describes the consequences of expressing FLAG epitope tagged cyclin D1 in estrogen 88 dependent human breast cancer cells. We demonstrate that inducing ectopic cyclin D1 in G1 arrested cells causes progression to S phase in the presence of antiestrogen, in both the presence and absence of serum growth factors, although cells re-arrest in G1 afier several days of treatment. Cdk4 is activated following AP treatment in the presence of ICI, and ectopic cyclin D1 co-precipitates with active Cdk4 by 24 h after induction. However, Cdk4 is inactivated after long term AP+ICI treatment, despite the presence of both ectopic and endogenous cyclin D1 in complex with Cdk4. The inactivation of Cdk4 coincides with a maintenance and/or gradual accumulation of p21 in the cells. We propose that initially, ectopic cyclin D1 can bind Cdk4, forming p21 free cyclin D1-Cdk4 complexes that are active Rb kinases. At later time points, treatment with antiestrogen, p21 levels increase until all cyclin D1-Cdk4 complexes contain and are inactivated by the Cdkl. In the absence of Cdk4 activity, progression through the G1 checkpoint is inhibited, and cells re-arrest. This work provides an explanation for the somewhat disparate results of two groups who have published reports characterizing cyclin D1 induction in stably transfected MCF-7 derivatives. Sutherland et a1, using ectopic cyclin D1 expressed from a zinc inducible promoter, showed that inducing cyclin D1 in the presence of antiestrogen caused activation of Cdk4 and progression through at least one cell cycle, with concurrent activation of cyclin E-Cdk2 complexes (3 8, 47). While Weisz et al also observed progression through the G1 checkpoint when ectopic cyclin D1 expression was induced from a tetracycline inducible promoter, they showed that antiestrogen inhibited long term cell growth despite high levels of cyclin D1 (34). Our data corroborate the work of both groups in an independent system of cyclin D1 induction in human breast cancer cells. In 89 addition, they provide a mechanistic explanation for these results: the failure of p21 to be downregulated in the absence of E2. The regulation of p21 is complex, and numerous signaling pathways converge at the p21 promoter. Growth arrest by interferon-y and differentiation by interleukin-6 is mediated by STAT activation of the p21 promoter (5, 9, l7), and epidermal growth factor (EGF) mediated STAT activation of p21 expression is also reported in cells whose growth is inhibited by EGF (33, 48). Transforming growth factor-B increases p21 levels by acting through Spl and Sp3 transcription factors, while Myc represses p21 levels, possibly by interfering with Spl and Sp3 activity (10, 16, 30). Transcription from the p21 promoter is also controlled by E2F1 (21). Coordinate regulation of p21 and cyclin D1 has been reported (20), and E2 can regulate transcription at the cyclin D1 promoter through Spl sites (7), a potential point at which their regulation may intersect. We suggest that p21 expression is increased or activated by ICI in estrogen dependent breast cancer cells, and that in the presence of ICI sufficient p21 is present to inhibit the activity of all cyclin D1-Cdk4 complexes. This inhibition may be overcome temporarily by expression of ectopic cyclin D1, but over time p21 levels are adjusted so that all cyclin D1-Cdk4 is inactivated. In this model, the activity of Cdk4 and ultimately cell proliferation is controlled by a balance of activating factors (D type cyclins) and inhibitors (e. g. p21). In our system of antiestrogen mediated growth arrest it appears that the cell has some mechanism to re-adjust the balance when it is altered experimentally. This suggests that in human breast cancer, overexpression of cyclin D1 is unlikely to result in the development of antiestrogen resistance in the absence of other changes. The observation that cyclin D1 overexpression is a common feature of breast cancers and that 90 its overexpression often correlates with positive ER status may reflect cyclin D l ’s involvement in other phases of tumorigenesis besides the development of antiestrogen resistance (15, 51). 91 References 1. Altucci, L., R. Addeo, L. Cicatiello, S. Dauvois, M. G. Parker, M. Truss, M. Beato, V. Sica, F. Bresciani, and A. Weisz. 1996. 17beta-Estradiol induces cyclin D1 gene transcription, p36Dl-p34cdk4 complex activation and p105Rb phosphorylation during mitogenic stimulation of G(l )-arrested human breast cancer cells. Oncogene 12:2315-24. 2. Altucci, L., R. Addeo, L. Cicatiello, D. Germano, C. Pacilio, T. Battista, M. Cancemi, V. B. Petrizzi, F. Bresciani, and A. Weisz. 1997. Estrogen induces early and timed activation of cyclin-dependent kinases 4, 5, and 6 and increases cyclin messenger ribonucleic acid expression in rat uterus. Endocrinology 138:978-84. 3. Amara, J. F., T. Clackson, V. M. Rivera, T. Guo, T. Keenan, S. Natesan, R. Pollock, W. Yang, N. L. Courage, D. A. Holt, and M. Gilman. 1997. A versatile synthetic dimerizer for the regulation of protein-protein interactions. Proc Natl Acad Sci U S A 94:10618-23. 4. Arroyo, M., and P. Raychaudhuri. 1992. Retinoblastoma-repression of E2F- dependent transcription depends on the ability of the retinoblastoma protein to interact with E2F and is abrogated by the adenovirus ElA oncoprotein. Nucleic Acids Res 20:5947-54. 5. Bellido, T., C. A. O'Brien, P. K. Roberson, and S. C. Manolagas. 1998. Transcriptional activation of the p21(WAFl,CIP1,SDIl) gene by interleukin-6 type cytokines. A prerequisite for their pro- differentiating and anti-apoptotic effects on human osteoblastic cells. J Biol Chem 273:21137-44. 6. Bieche, I., M. Olivi, C. Nogues, M. Vidaud, and R. Lidereau. 2002. Prognostic value of CCNDl gene status in sporadic breast tumours, as determined by real- time quantitative PCR assays. Br J Cancer 86:580-6. 7. Castro-Rivera, E., I. Samudio, and S. Safe. 2001. Estrogen regulation of cyclin D1 gene expression in ZR-75 breast cancer cells involves multiple enhancer elements. J Biol Chem 276:30853-61. 8. Cheng, M., P. Olivier, J. A. Diehl, M. Fero, M. F. Roussel, J. M. Roberts, and C. J. Sherr. 1999. The p21(Cipl) and p27(Kipl) CDK 'inhibitors' are essential activators of cyclin D-dependent kinases in murine fibroblasts. Embo J 18:1571- 83. 9. Chin, Y. E., M. Kitagawa, K. Kuida, R. A. Flavell, and X. Y. Fu. 1997. Activation of the STAT signaling pathway can cause expression of caspase 1 and apoptosis. Mol Cell Biol 17:5328-37. 92 10. 11. 12. 13. 14. 15. 16. 17. 18. 19. 20. Datto, M. B., Y. Yu, and X. F. Wang. 1995. Functional analysis of the transforming growth factor beta responsive elements in the WAF 1/Cip1/p21 promoter. J Biol Chem 270:28623-8. Diehl, J. A., and C. J. Sherr. 1997. A dominant-negative cyclin D1 mutant prevents nuclear import of cyclin— dependent kinase 4 (CDK4) and its phosphorylation by CDK-activating kinase. Mol Cell Biol 17:7362-74. el-Deiry, W. S., T. Tokino, V. E. Velculescu, D. B. Levy, R. Parsons, J. M. Trent, D. Lin, W. E. Mercer, K. W. Kinzler, and B. Vogelstein. 1993. WAF l, a potential mediator of p53 tumor suppression. Cell 75:817-25. Fantl, V., M. A. Richards, R. Smith, G. A. Lammie, G. Johnstone, D. Allen, W. Gregory, G. Peters, C. Dickson, and D. M. Barnes. 1990. Gene amplification on chromosome band 11q13 and oestrogen receptor status in breast cancer. Eur J Cancer 26:423-9. Foster, J. S., and J. Wimalasena. 1996. Estrogen regulates activity of cyclin- dependent kinases and retinoblastoma protein phosphorylation in breast cancer cells. Mol Endocrinol 102488-98. Frierson, H. F., Jr., M. J. Gaffey, L. R. Zukerberg, A. Arnold, and M. E. Williams. 1996. Immunohistochemical detection and gene amplification of cyclin D1 in mammary infiltrating ductal carcinoma. Mod Pathol 9:725—30. Gartel, A. L., X. Ye, E. Goufrnan, P. Shianov, N. Hay, F. Najmabadi, and A. L. Tyner. 2001. Myc represses the p21(WAF1/CIP1) promoter and interacts with Spl/Sp3. Proc Natl Acad Sci U S A 98:4510-5. Gooch, J. L., R. E. Herrera, and D. Yee. 2000. The role of p21 in interferon gamma-mediated growth inhibition of human breast cancer cells. Cell Growth Differ 11:335-42. Han, E. K., A. Sgambato, W. Jiang, Y. J. Zhang, R. M. Santella, Y. Doki, A. M. Cacace, I. Schieren, and I. B. Weinstein. 1995. Stable overexpression of cyclin D1 in a human mammary epithelial cell line prolongs the S-phase and inhibits growth. Oncogene 10:953-61. Harper, J. W., G. R. Adami, N. Wei, K. Keyomarsi, and S. J. Elledge. 1993. The p21 Cdk-interacting protein Cipl is a potent inhibitor of G1 cyclin- dependent kinases. Cell 75:805-16. Hiyama, H., A. Iavarone, J. LaBaer, and S. A. Reeves. 1997. Regulated ectopic expression of cyclin D1 induces transcriptional activation of the cdk inhibitor p21 gene without altering cell cycle progression. Oncogene 14:2533-42. 93 21. 22. 23. 24. 25. 26. 27. 28. 29. 30. 31. Hiyama, H., A. Iavarone, and S. A. Reeves. 1998. Regulation of the cdk inhibitor p21 gene during cell cycle progression is under the control of the transcription factor E2F. Oncogene 16:1513-23. Hui, R., A. L. Cornish, R. A. McClelland, J. F. Robertson, R. W. Blamey, E. A. Musgrove, R. I. Nicholson, and R. L. Sutherland. 1996. Cyclin D1 and estrogen receptor messenger RNA levels are positively correlated in primary breast cancer. Clin Cancer Res 2:923-8. Karlseder, J ., R. Zeillinger, C. Schneeberger, K. Czerwenka, P. Speiser, E. Kubista, D. Bimbaum, P. Gaudray, and C. Theillet. 1994. Patterns of DNA amplification at band q13 of chromosome 11 in human breast cancer. Genes Chromosomes Cancer 9242-8. Kato, J ., H. Matsushime, S. W. Hiebert, M. E. Ewen, and C. J. Sherr. 1993. Direct binding of cyclin D to the retinoblastoma gene product (pr) and pr phosphorylation by the cyclin D-dependent kinase CDK4. Genes Dev 7:331-42. Kenny, F. S., R. Hui, E. A. Musgrove, J. M. Gee, R. W. Blarney, R. I. Nicholson, R. L. Sutherland, and J. F. Robertson. 1999. Overexpression of cyclin D1 messenger RNA predicts for poor prognosis in estrogen receptor-positive breast cancer. Clin Cancer Res 5:2069-76. Lamb, J ., M. H. Ladha, C. McMahon, R. L. Sutherland, and M. E. Ewen. 2000. Regulation of the fimctional interaction between cyclin D1 and the estrogen receptor. Mol Cell Biol 20:8667-75. Lukas, J ., J. Bartkova, and J. Bartek. 1996. Convergence of mitogenic signalling cascades from diverse classes of receptors at the cyclin D-cyclin-dependent kinase-pr-controlled G1 checkpoint. Mol Cell Biol 16:6917-25. Matsushime, H., D. E. Quelle, S. A. Shurtleff, M. Shibuya, C. J. Sherr, and J. Y. Kato. 1994. D-type cyclin-dependent kinase activity in mammalian cells. Mol Cell Biol 14:2066-76. McIntosh, G. G., J. J. Anderson, 1. Milton, M. Steward, A. H. Parr, M. D. Thomas, J. A. Henry, B. Angus, T. W. Lennard, and C. H. Home. 1995. Determination of the prognostic value of cyclin D1 overexpression in breast cancer. Oncogene 1 1:885-91. Mitchell, K. O., and W. S. El-Deiry. 1999. Overexpression of c-Myc inhibits p21WAF1/CIP1 expression and induces S- phase entry in 12-0- tetradecanoylphorbol-l3-acetate (TPA)-sensitive human cancer cells. Cell Growth Differ 10:223-30. Musgrove, E. A., C. S. Lee, M. F. Buckley, and R. L. Sutherland. 1994. Cyclin D1 induction in breast cancer cells shortens G1 and is sufficient for cells arrested in G1 to complete the cell cycle. Proc Natl Acad Sci U S A 91 :8022-6. 94 32. 33. 34. 35. 36. 37. 38. 39. 40. 41. Neuman, E., M. H. Ladha, N. Lin, T. M. Upton, S. J. Miller, J. DiRenzo, R. G. Pestell, P. W. Hinds, S. F. Dowdy, M. Brown, and M. E. Ewen. 1997. Cyclin D1 stimulation of estrogen receptor transcriptional activity independent of cdk4. Mol Cell Biol 17:5338-47. Ohtsubo, M., A. Takayanagi, S. Gamou, and N. Shimizu. 2000. Interruption of NFkappaB-STATI signaling mediates EGF-induced cell- cycle arrest. J Cell Physiol 1842131-7. Pacilio, C., D. Germano, R. Addeo, L. Altucci, V. B. Petrizzi, M. Cancemi, L. Cicatiello, S. Salzano, F. Lallemand, R. J. Michalides, F. Bresciani, and A. Weisz. 1998. Constitutive overexpression of cyclin D1 does not prevent inhibition of hormone-responsive human breast cancer cell growth by antiestrogens. Cancer Res 58:871-6. Park, Y. G., S. Park, S. O. Lim, M. S. Lee, C. K. Ryu, 1. Kim, and Y. S. Cho- Chung. 2001. Reduction in cyclin Dl/Cdk4/retinoblastoma protein signaling by CRE- decoy oligonucleotide. Biochem Biophys Res Commun 281 :1213-9. Pelosio, P., M. Barbareschi, E. Bonoldi, A. Marchetti, P. Verderio, O. Caffo, P. Bevilacqua, P. Boracchi, F. Buttitta, R. Barbazza, P. Dalla Palma, and G. Gasparini. 1996. Clinical significance of cyclin D1 expression in patients with node- positive breast carcinoma treated with adjuvant therapy. Ann Oncol 7:695- 703. Peters, G., V. Fantl, R. Smith, S. Brookes, and C. Dickson. 1995. Chromosome 11q13 markers and D-type cyclins in breast cancer. Breast Cancer Res Treat 33:125-35. Prall, O. W., E. M. Rogan, E. A. Musgrove, C. K. Watts, and R. L. Sutherland. 1998. c-Myc or cyclin D1 mimics estrogen effects on cyclin E-Cdk2 activation and cell cycle reentry. Mol Cell Biol 18:4499-508. Prall, O. W., B. Sarcevic, E. A. Musgrove, C. K. Watts, and R. L. Sutherland. 1997. Estrogen-induced activation of Cdk4 and Cdk2 during Gl-S phase progression is accompanied by increased cyclin D1 expression and decreased cyclin-dependent kinase inhibitor association with cyclin E- Cdk2. J Biol Chem 272:10882-94. Sabbah, M., D. Courilleau, J. Mester, and G. Redeuilh. 1999. Estrogen induction of the cyclin D1 promoter: involvement of a CAMP response-like element. Proc Natl Acad Sci U S A 96:11217-22. Skildum, A. J ., S. Mukherjee, and S. E. Conrad. 2001. The cyclin dependent kinase inhibitor p21{superWAF1/Cip1} is an antiestrogen regulated inhibitor of Cdk4 in human breast cancer cells. J Biol Chem 7:7. 95 42. 43. 44. 45. 46. 47. 48. 49. 50. 51. 52. 53. Takano, Y., H. Takenaka, Y. Kato, M. Masuda, T. Mikami, M. Saegusa, and I. Okayasu. 1999. Cyclin D1 overexpression in invasive breast cancers: correlation with cyclin-dependent kinase 4 and oestrogen receptor overexpression, and lack of correlation with mitotic activity. J Cancer Res Clin Oncol 125:505-12. Vos, C. B., N. T. Ter Haar, J. L. Peterse, C. J. Comelisse, and M. J. van de Vijver. 1999. Cyclin D1 gene amplification and overexpression are present in ductal carcinoma in situ of the breast. J Pathol 187:279-84. Watts, C. K., A. Brady, B. Sarcevic, A. deFazio, E. A. Musgrove, and R. L. Sutherland. 1995. Antiestrogen inhibition of cell cycle progression in breast cancer cells in associated with inhibition of cyclin-dependent kinase activity and decreased retinoblastoma protein phosphorylation. Mol Endocrinol 9:1804-13. Watts, C. K., K. J. Sweeney, A. Warlters, E. A. Musgrove, and R. L. Sutherland. 1994. Antiestrogen regulation of cell cycle progression and cyclin D1 gene expression in MCF-7 human breast cancer cells. Breast Cancer Res Treat 31 :95- 105. Weintraub, S. J., C. A. Prater, and D. C. Dean. 1992. Retinoblastoma protein switches the E2F site from positive to negative element. Nature 358:259-61. Wilcken, N. R., O. W. Prall, E. A. Musgrove, and R. L. Sutherland. 1997. Inducible overexpression of cyclin D1 in breast cancer cells reverses the growth- inhibitory effects of antiestrogens. Clin Cancer Res 3:849-54. Xie, W., K. Su, D. Wang, A. J. Paterson, and J. E. Kudlow. 1997. MDA468 growth inhibition by EGF is associated with the induction of the cyclin-dependent kinase inhibitor p21WAF]. Anticancer Res 17:2627-33. Xiong, Y., G. J. Harmon, H. Zhang, D. Casso, R. Kobayashi, and D. Beach. 1993. p21 is a universal inhibitor of cyclin kinases. Nature 366:701-4. Zhang, H., G. J. Hannon, D. Casso, and D. Beach. 1994. p21 is a component of active cell cycle kinases. Cold Spring Harb Symp Quant Biol 59:21-9. Zukerberg, L. R., W. I. Yang, M. Gadd, A. D. Thor, F. C. Koemer, E. V. Schmidt, and A. Arnold. 1995. Cyclin D1 (PRADl) protein expression in breast cancer: approximately one-third of infiltrating mammary carcinomas show overexpression of the cyclin D1 oncogene. Mod Pathol 8:560-7. Zwij sen, R. M., R. S. Buckle, E. M. Hijmans, C. J. Loomans, and R. Bemards. 1998. Li gand-independent recruitment of steroid receptor coactivators to estrogen receptor by cyclin D1. Genes Dev 12:3488-98. Zwij sen, R. M., E. Wientjens, R. Klompmaker, J. van der Sman, R. Bemards, and R. J. Michalides. 1997. CDK-independent activation of estrogen receptor by cyclin D1. Cell 88:405-15. 96 CHAPTER FOUR Characterization of LCC9, an Antiestrogen Resistant Derivative of MCF-7 Cells 97 Chapter 4: Characterization of LCC9, an Antiestrogen Resistant Derivative of MCF-7 Cells ABSTRACT Although antiestrogen therapies represent a key advance in the treatment of breast cancer, acquired resistance to antiestrogens is a major cause of treatment failure. The molecular changes underlying the conversion from an antiestrogen sensitive to an antiestrogen resistant phenotype are poorly understood, despite much investigation. The LCC9 cell line, which is resistant to growth arrest by the pure steroidal antiestrogen ICI 182780, is a derivative of the estrogen receptor positive, antiestrogen sensitive human breast cancer cell line MCF-7. Here we compare the molecular events at the G1 -)S phase checkpoint in LCC9 cells with their parental MCF-7 cells. Using transfection with estrogen receptor response element driven luciferase expression vectors, we demonstrate that LCC9’s estrogen receptor is regulated similarly to MCF-7’s. While MCF-7’s G1 phase cyclin dependent kinases Cdk2 and Cdk4 are inactivated following antiestrogen treatment, we show that both kinases remain active in antiestrogen treated LCC9 cells. The Cdk4 complexes from LCC9 cells retain sensitivity to inhibition by lysate from antiestrogen treated MCF-7 cells. We show that compared to MCF-7, LCC9 cells have elevated levels of cyclin D1 protein. In addition, while levels of the Cdk inhibitor p21wamCipl increase after two days of antiestrogen treatment in MCF-7 cells, p21 is not highly regulated by antiestrogen in LCC9 cells. These experiments suggest that in LCC9 cells, altered signaling pathways upstream of cyclin D and p21 may maintain the activities of G1 Cdks, allowing proliferation in the presence of antiestrogen. 98 Introduction Despite advances in detection and treatment, breast cancer remains a common and deadly malignancy in the United States. Because many breast tumor cells require estrogen for growth, drugs that block the effects of estrogen have long been used as therapies for breast cancer patients. Tamoxifen, which acts as an antagonist of the estrogen receptor (ER)3 in breast tissue, has been a successful treatment for ER positive breast cancer and has recently been recommended for the prevention of breast cancer in women at high risk for developing the disease. However, the usefulness of Tamoxifen and other antiestrogens is limited by the development of resistant tumors in patients who initially responded positively to therapy (33, 34, 72). The process by which ER positive tumors acquire an antiestrogen resistant phenotype is unknown, and there are no molecular markers associated with its development, with the possible exception of Her2 overexpression (50). Research in this area may point to diagnostic markers that predict the effectiveness of antiestrogen therapies, or to alternative treatment strategies. In sensitive tissues, the most biologically potent form of estrogen, 17B-estradiol (E2), acts as a mitogen by binding to the ER, a nuclear steroid receptor. There are two forms of ER, the classical ERa and the more recently described ERB (42, 65). Both ER types act as a ligand activated transcription factors by direct binding to DNA at estrogen response elements (EREs) and variations thereof and recruiting transcriptional 3 Abbreviations used are: ER, estrogen receptor. ERE, estrogen receptor response element. p21, WAF l/Cipl. p27, Kipl. Cdk, cyclin dependent kinase. Cdkl, Cdk inhibitor. pr, retinoblastoma protein. E2, 17B-estradiol. IP, immunoprecipitation. ICI, ICI 182,780. DTT, dithiothreotol. IMEM, improved modified Eagle’s medium. CSS, charcoal stripped serum. Luc, luciferase. TK, thymidine kinase minimal promoter. RT-PCR, reverse transcriptase-polymerase chain reaction. Minutes, m. Hours, h. FBS, fetal bovine serum. EGF, epidermal growth factor. STAT, signal transducers and activators of transcription. 99 coactivator proteins such as CBP/p300, p/CAF, p160, and SRCl to the promoter region (9, 44, 48, 64, 70, 71). Additionally, ER can act to modulate transcription in the absence of DNA binding via protein — protein interactions with transcriptional activators such as Apl and Spl (30, 56); in both cases, ER recruits to coactivators to the promoter, thereby enhancing transcription. ERa and B have different activities at AP] sites (45), and evidence suggests that ERB may act to inhibit transcription of some ERa regulated genes (40). The activities of both ERs can be further regulated by phosphorylation by Erk, a MAPK (1, 36, 66). Thus, evolution has produced a complex signaling network through which estrogen mediated gene expression can be finely tuned. In general, antiestrogens mimic the three dimensional conformation of estrogen and bind ER’s ligand binding domain but do not allow transcriptional activation; however, some antiestrogens have tissue and/or promoter specific agonist activity. For example, Tamoxifen acts as an ER antagonist in the breast but has partial agonist activity in the uterus, and has opposite activities at AP-l dependent promoters with the two known ER isoforrns, ERa and ERB (20, 26, 45, 67). The “pure” antiestrogen ICI 182,780 (ICI), used in the present study, acts solely as an ER antagonist and can be an effective second line antiestrogen therapy in patients whose tumors have become Tamoxifen resistant (15, 16, 25 , 47). Resistance to pure ER antagonists occurs clinically and in experimental models, although the effectiveness of pure antagonists in Tamoxifen resistant breast cancer suggests that distinct mechanisms are responsible for resistance to partial agonists and pure antagonists. Several mechanisms have been proposed to explain the acquisition of antiestrogen resistance. These include a loss of ER or selection of ER negative cells from a 100 heterogeneous tumor population, a change in ER function, altered metabolism of antiestrogen, and inappropriate activation of estrogen dependent or independent growth stimulatory signaling pathways. These alterations could occur alone or in combination and could result from genetic lesions or epigenetic changes (13, 14, 23, 31, 46, 54). While ER loss may be a cause of antiestrogen resistance prior to treatment, acquired resistance during the course of treatment occurs in most cases without loss or mutation of ER (32), and antiestrogen metabolism has not been shown to be a likely cause of antiestrogen resistance (43). The composition of co—activators and/or co-repressors interacting with antiestrogen bound ER may influence whether the ligand behaves as a transcriptional activator or repressor at any given promoter, and changes in the makeup of ER containing complexes may underlie the acquisition of antiestrogen resistance (19, 60, 63), although clinical evidence has been lacking thus far (10). Much recent attention has therefore been placed on alterations in signal transduction pathways regulating cellular proliferation as potential causes of antiestrogen resistance. Estrogens and antiestrogens affect proliferation in target cells at the G1 checkpoint (18, 62, 68), passage through which is governed by the Rb pathway (see Chapter 1, Figure 2). In cells containing wild type retinoblastoma protein (Rb), progression through Gl to S phase is controlled by the activity of two types of serine/threonine kinases, cyclin dependent kinase-2 (Cdk2) and Cdk4/6, which integrate the activities of multiple signaling pathways. Cdk2 can form a catalytically active enzyme when bound in one to one stoichiometry with either cyclin E or A, while Cdk4 and the related Cdk6 are activated by association with a D-type cyclin (27, 28, 57). In addition to cyclins, Cdks can stably associate with Cdk inhibitors (Cdkls), including 101 1921“”cipl (p21) and p27k‘P' (p27). Originally described as inhibitors of Cdk activity, recent data suggests that under some conditions some Cdkls can act to increase the activity of cyclin D/Cdk4 complexes (11, 22, 52, 58, 73). Estrogen has been shown to increase cyclin D1 protein levels, Cdk4 and Cdk2 activity, and Rb phosphorylation, and to decrease Cdkl expression and association with Cdk2 in breast cancer cells (1 7, 18, 51, 53), and antiestrogen reverses these effects (7, 59, 68, 69). Inappropriate activation of G1 Cdks could therefore contribute to antiestrogen resistant grth of breast cancer cells. In this work, we examine the molecular events underlying the G1-)S phase transition in a model of acquired resistance to the pure steroidal antiestrogen ICI. The MCF-7 cell line, derived from a human breast adenocarcinoma, is ERa positive, estrogen dependent and antiestrogen sensitive (4, 5, 24, 61). Through in viva selection for estrogen independent tumorigenicity and in vitro selection for grth in the presence of increasing concentrations of ICI, Clarke et al generated the LCC9 cell line. LCC9 cells retain ER, but are able to proliferate in concentrations of ICI that completely inhibit MCF-7 cell growth (6). Below we compare signaling events in ICI and E2 treated MCF- 7 and LCC9 cells, demonstrate that LCC9 cells have numerous changes in the Rb pathway, and that the causes of these changes are likely to be upstream of G1 Cdk activation. Materials and Methods Cell lines and culture media. MCF-7 and LCC9 cells were obtained from Dr. Michael Johnson of the Lombardi Cancer Center, Georgetown University. They were routinely 102 passaged in IMEM (BioFluids), supplemented with 5% fetal bovine serum (F BS, HyClone), 100 units/ml penicillin and 100 ug/ml streptomycin (Gibco). In the experiments described in Figures 1 and 2, early passage LCC9 cells were split into two populations: one was cultured routinely in 5% F BS as described above, while the other was routinely cultured in IMEM without phenol red (BioFluids) containing 5% charcoal stripped serum (CSS, HyClone), penicillin / streptomycin and 10'8 M ICI (Astra Zeneca). These two populations were cultured in this way for 7 passages (28 days) prior to the experiments; the two populations were passaged at the same ratio and intervals. For estrogen and antiestrogen treatments, cells were cultured in IMEM without phenol red (BioFluids) containing 5% charcoal stripped serum (CSS, HyClone) and penicillin / streptomycin, with either 10'8 M E2 (Sigma) or 10'8 M ICI (Astra Zeneca) unless otherwise indicated. Cells were washed twice with PBS before all media changes. Cells were cultured at 37° C with 5% C02. Cell cycle analysis. MCF-7 and LCC9 cells were plated at 2.5 x 105 cells per 60 mm plate in 5% F BS. After the indicated treatments, cells were trypsinized, washed in phosphate buffered saline (PBS), suspended in PBS+10% F BS, fixed with 80% cold ethanol and stored at —20° C. Prior to analysis, cells were washed twice with PBS, then suspended in PBS + 1 mg/ml RNaseA, 0.2 mg/ml propidium iodide, 0.5 mM EDTA and 0.1% Triton-X-IOO. Cells were then analyzed for red fluorescence on a FACSVantage flow cytometer; cell cycle distribution was determined using ModFit software. Three 60 mm plates were analyzed for each time point. 103 Transfections and luciferase assays. This experiment was performed by Dr. Hemant Varma. MCF-7 and LCC9 cells were plated at 5x105 cells/60mm plate and transfected with plasmid DNA using the Lipofectin reagent following the protocol provided by the manufacturer (GIBCO-BRL). 1 ug of ERE-tk-luc or tk-luc (vector) plasmid was cotransfected with 0.1 ug of pCMV-B-galactosidase as a control for transfection efficiency (luciferase plasmid provided by Dr. Richard Miksicek; B-gal plasmid provided by Dr. Richard Schwartz). Cells were kept in 5% charcoal dextran stripped serum (CSS) containing medium for 24 hours followed by CSS alone or with ICI (lOOnM), E2 (lnM) or E2 +ICI for an additional 24 hours. Cells were harvested at 48 h post-transfection and both the luciferase and B-galactosidase activities were measured using the detection reagents and protocols provided by the manufacturer (Clontech) on a Turner TD 20E luminometer (Turner Designs). For each experiment, ERE-luciferase and B-galactosidase transfections were performed in triplicate; tk-luc transfection was performed twice. Immunoprecipitations and kinase assays. Immunoprecipitations (IP) and Cdk activity measurements were performed using a modification of a published method (41). Unless indicated otherwise, all manipulations were carried out on ice. Cells were washed twice in ice cold PBS, harvested by scraping in PBS and pelleted; the pellets were frozen and stored in liquid nitrogen until the day of analysis. Cells were resuspended and lysed by sonication in IP buffer (50 mM HEPES (pH 7.5), 150 mM NaCl, 1 mM EDTA, 2.5 mM EGTA, 0.1% Tween-20, 10% glycerol, 1 mM DTT, 10 mM B-glycerophosphate, 1 mM NaF, lmM NaVO4, 0.1 mM PMSF, 10 ug/ml leupeptin, and 2 ug/ml aprotinin). The lysates were cleared by centrifugation, and protein concentrations were quantitated using 104 BioRad’s protein assay reagent. For IPs, 75 ug of total protein was diluted to 500 1.11 with IP buffer, then incubated for 60 minutes (m) rocking at 4° C with 1.5 ug antibody (anti- Cdk4 H-22-G, anti-Cdk2 M2 or normal goat IgG, Santa Cruz) bound to 7.5 ul protein G agarose beads (Boehringer Mannheim). Beads were pelleted and washed four times with 100 pl IP buffer and twice with 100 ul kinase buffer (50 mM HEPES pH7.5, 10 mM MgC12, 1 mM DTT, 2.5 mM EGTA, 10 mM B-glycerophosphate, 0.1 mM NaVO4, and 1 mM NaF). Pellets were then suspended in 40 ul kinase buffer containing 10 uCi y- [32P]ATP and 20 uM cold ATP. For Cdk4 assays, 1.0 ul glutathione sepharose 48 (Amersham Pharmacia) to which approximately 2.0 ug of a fusion protein between glutathione-S-transferase (GST) and amino acids 792 to 928 of human Rb were bound (GST-Rb bacterial expression vector was kindly provided by Dr. William Kaelin of the Dana Farber Cancer Institute) was added as substrate. For Cdk2 assays, 2.0 ug histone H1 (Sigma) was used as substrate. The reactions were incubated at 30° C for 30 m with occasional mixing, after which they were boiled in SDS loading buffer containing mercaptoethanol. Beads were pelleted and the supematants transferred to clean tubes. Aliquots of the reaction products were resolved on 10% SDS polyacrylamide gels, which were then dried and exposed to autoradiography film. The data were quantitated by phosphorimaging with a Storm phosphorimager (Molecular Dynamics) using ImageQuant software. Western blotting. Either total cell lysates (20 ug), IP supematants or IP pellets were resolved on 12% SDS-polyacrylamide gels, transferred to membranes and probed with antibodies for cyclin D1 (UB1 06 137, Upstate Biotechnology), cyclin E (14761C, 105 Pharmingen), p21wan (p21-c-19, Santa Cruz Biotechnology), p27"ipl (C-19, Santa Cruz Biotechnology), Cdk4 (H22, Santa Cruz Biotechnology), Cdk2 (M2, Santa Cruz Biotechnology), or actin (Sigma clone AC-40). Membranes were then incubated with horseradish peroxidase conjugated goat anti-rabbit (BioRad) or goat anti-mouse (American Qualex) secondary antibodies, and immunoreactive proteins were detected using Super Signal West Pico Chemiluminescent Substrate (Pierce) according to manufacturers’ protocol. Lysate mixing assays. MCF-7 and LCC9 cells were treated as indicated, then harvested, stored, and lysed as described above. Seventy five pg of protein from E2 treated MCF-7 or LCC9 cells were mixed with 7.5 to 75 ug of protein from ICI treated MCF—7 cells, and diluted to 500 ul with IP buffer. The mixtures were incubated at 30° C for 30 m with occasional agitation. Unmixed control reactions were diluted and incubated simultaneously. The samples were then subjected to the Cdk4 kinase assay as described above. Phosphorylated substrate band intensities were quantitated by storage phosphorimaging. Results LC C 9 cells are not arrested by the antiestrogen 1C] 182 780. To verify the reported antiestrogen resistant phenotype of LCC9 cells (6), we compared the cell cycle profile of MCF-7 and LCC9 cells treated with the antiestrogen ICI over a 42 h time course. To evaluate the stability of this phenotype, two parallel populations of LCC9 cells were 106 % cells in 61 phase 18 hours after ICI treatment % cells in S phase O 6 18 24 30 42 hours after ICI treatment Figure 1: The LCC9 cell line's antiestrogen resistant phenotype is stable in the absence of selective pressure. MCF-7 cells (black bars) and LCC9 cells cultured long term (> 1 month) in the presence of FBS (grey bars) or CSS+100 nM ICI (white bars) were cultured in FBS for two days. Cells were then treated with 100 nM ICI and harvested at intervals over 42 h. Cells were fixed in ethanol, stained with propidium iodide, and analyzed by flow cytometry. A) The average percentage of cells in G1 phase; error bars indicate on standard deviation (n = 3). B) The percent of cells in S phase. 107 used, one cultured long term in 10 nM ICI and the other cultured long term in 5% F BS. Cells were plated at low density in FBS, then washed twice with PBS and treated with 10 nM ICI. Cells were harvested and fixed at the times indicated, then DNA content was measured with propidium iodide staining and flow cytometry. Both MCF-7 and LCC9 cells were asynchrous at the beginning of the time course, with 38% of MCF-7 cells and approximately 50% of LCC9 cells containing a G1 phase DNA content (Figure 1). MCF-7 cells began to accumulate in G1 phase after ICI treatment, reaching nearly 90% G1 in phase by 42 h, with a corresponding reduction in the fraction of cells in S phase. In contrast, LCC9 cells were unaffected by ICI treatment. The fraction of cells in G1 phase was about 50% at each time point for both LCC9 populations. These data confirm that LCC9 cells are resistant to the cytostatic effects of the pure ER antagonist ICI, and demonstrate that LCC9’s antiestrogen resistant phenotype is stable in the absence of selective pressure. LC C9 's ER is regulated normally at a consensus ERE. Because LCC9 cells contain EROt but have altered expression of E2 regulated genes (6), we speculated that LCC9’s ERor may have an altered response to antiestrogen, and this might account for its antiestrogen resistant phenotype. This possibility was tested by comparing MCF-7 and LCC9 in a luciferase assay measuring transcriptional activity from a consensus ERE. Cells were transfected with either ERE (ERE-tk-luc) or minimal promoter (tk-luc) plasmids and a B- galactosidase control plasmid, then treated with CSS alone or containing either 100 nM ICI, 1 nM E2, or 100 nM ICI + 1 nM E2. 108 -- ClERE-luc 5 . lTK-luc 1“ J 1 I—-}—-1 luc/b-gal N J —l i css ICI £2 E2+|C| css ICI £2 E2+ICI MCF-7 LCC9 Figure 2: LCC9 cells’ estrogen receptor is regulated normally by E2 and ICI. MCF-7 and LCC9 cells were transfected with vectors encoding luciferase under the control of either a consensus estrogen response element upstream of a minimal promoter (ERE-luc) or the minimal promoter alone (TK-luc). Cells were co- transfected with a B-galactosidase expression vector. Transfected cells were cultured in charcoal/dextran stripped serum (CSS) for 24 h, then treated with either fresh CSS, 100 nM ICI, 1 nM E2, or 100 nM + 1 nM E2 for an additional 24 h. Cells were harvested and luciferase and B-galactosidase activities determined on a Turner luminometer. The graph shows luciferase activity divided by B-galactosidase activity. The experiment was performed three times, and three samples were measured for each experiment. Error bars indicate one standard deviation (n = 9). 109 The luciferase activity divided by B-galactosidase activity to correct for possible differences in transfection efficiency is shown in Figure 2. In MCF-7 cells, E2 treatment led to strong transcriptional activation of the ERE, but not the control promoter. This activation was abolished by co-treatment with ICI, confirming its function as an E2 antagonist. Identical results were obtained in LCC9 cells, suggesting that LCC9’s ability to proliferate in ICI is not due to a failure of ICI to downregulate transcription at consensus EREs. Interestingly, the basal levels of ERE luciferase expression were slightly but reproducibly higher in LCC9 than in MCF-7 cells. C dk2 and Cdk4 are not inactivated by antiestrogen in LCC 9 cells. We and others have shown that in MCF-7 cells, G1 Cdks are inactivated following ICI treatment (7, 59, 68). To test whether LCC9’s ability to proliferate in the presence of ICI could be due to a lack of Cdk inactivation, we compared the Cdk2 and Cdk4 associated kinase activities in ICI treated MCF-7 and the two populations of LCC9 cells described in Figure 1. Cells were plated in FBS for two days, then treated with ICI and harvested at the time points indicated. Cellular lysates were immunoprecipitated with antibodies reactive to Cdk2 or Cdk4; pellets were washed extensively and reacted with radiolabeled ATP and substrate (histone H1 for Cdk2 and GST-Rb for Cdk4). Reaction products were resolved electrophoretically and visualized by autoradiography. As indicated in Figure 3A, Cdk2 in vitro kinase activity was inhibited by ICI treatment by 18 h in MCF-7 cells, and was undetectable by 30 h. ICI inactivated Cdk4 activity in MCF-7 cells with similar kinetics (Figure 33). However, consistent with their growth phenotype observed in Figure 1, neither G1 Cdk activity was downregulated in 110 A- ‘5» MCF-7 LCC-9(FBS) LCC-9(ICI) 00 6182430 0 6182430 0 6182430 -* 8829998!!! B. “.3. MCF-7 LCC-9(FBS) LCC-9(ICI) 0 0 61824300 618 24300 6182430 ”dr- fiflfififimnggw GST-Rb C. MCF—7 LCC-9(FBS) LCC-9(ICI) 01830 01830 01830 .- a- . - up a. - g .- p21(Cdk4co-IP) w “we?” “M” w .1” «but W diam"?! 'rt‘r‘lflfli‘ actin Figure 3: LCC9's GI Cdks are not inactivated by antiestrogen. MCF-7 and LCC9 cells (cultured for one month in either FBS or ICI as in Figure 1) were incubated in FBS for two days, then treated with 100 nM ICI. Cells were harvested at the indicated intervals and lysates were prepared and normalized. A) Aliquots of lysates were immunoprecipitated with anti-Cdk2 antibodies. The pellets were incubated with radiolabeled ATP and histone H1 substrate, and kinase reaction products were resolved on SDS-PAGE and visualized by autoradiography. B) Aliquots of lysates were immunoprecipitated with anti-Cdk4 antibodies, and the pellets were incubated with radiolabeled ATP and recombinant retinoblastoma protein (GST-Rb) substrate. Products were resolved on SDS-PAGE and visualized by autoradiography. C) Aliquots of lysate and reaction products from (B) were analyzed by Western blot using cyclin D, p21, and actin antibodies. 111 LCC9 cells following ICI treatment during the course of this experiment. While the levels of Cdk2 activity were comparable in asynchronous MCF-7 and LCC9 cells (0 h), LCC9 cells had reduced Cdk4 activity compared to MCF-7 cells. There were no differences between the FBS-cultured and ICI-cultured LCC9 populations’ response to ICI treatment. These results suggest that the defect in ICI signaling in LCC9 cells is upstream of both G1 phase Cdks. We next compared the levels of cyclin D1 and p21 protein in whole cell lysates and in Cdk4 immunoprecipitates by Western blotting (Figure 3C). In MCF-7 cells, total and Cdk4 associated cyclin D1 protein decreased slightly following ICI treatment, while p21 levels were unchanged, although p21 is upregulated upon longer treatment in MCF-7 cells (see Figure 5). In LCC9 cells, levels of cyclin D were unchanged over the course of 30 h, and p21 appeared to decrease slightly. However, the amount of both cyclin D1 and p21, in lysates and in Cdk4 pellets, was slightly but consistently higher in LCC9 cells than in MCF-7 over numerous experiments. Cdk4 activity in LCC9 is sensitive to inhibition by lysatefi'om ICI treated MCF-7 cells. We have previously shown that ICI treated MCF-7 cells contain a factor capable of inhibiting active Cdk4 in vitro, and that this factor is lost upon p21 immunodepletion (59). We reasoned that the Cdk4-associated kinase activity present in ICI treated LCC9 cells could be due to the fact that the Cdk4 complex had become refractory to inhibition. To test this possibility, Cdk4 activity in E2 treated MCF-7 and LCC9 cells was compared in a lysate mixing assay. 112 A' ICI MCF7 ICI MCF7 BEL M + E2 MCF7 + E2 LCC9 MCF7 LCC9 MCF7 LCC9 . . ICI E2 10 E2 10 E2 ICI E2 "“X W I MCF-7 El LCC9 — El theoretical fraction of "E2" activity 53 o A O\ 9 N L E2 1t05 1t02 1 to 1 ICI mixing ratio Figure 4: Cdk4 activity in LCC9 is sensitive to the inhibitory factor present in [CI treated MCF-7 cells. Parallel cultures of MCF-7 and LCC9 cells were treated for 48 h in ICI and E2 as described, and harvested. Cellular lysates were prepared and quantitated. A) Representative autoradiogram showing Cdk4 inhbition in MCF-7 and LCC9 cells. In the first eight lanes, equal amounts of the indicated lysates were immunoprecipitated with Cdk4 or control (mock) antibodies. The pellets were then incubated with radiolabeled ATP and substrate (GST-Rb) and the reaction products resolved by SDS-PAGE. In the final six lanes, increasing amounts of lysate from ICI treated MCF-7 cells (20 ug, 50 ug and 100 ug) were added to fixed amounts of lysates from E2 treated MCF-7 or LCC9 cells (100 ug) as indcated, and the mixed lysates were incubated prior to immunoprecipitation with Cdk4 antibodies and kinase reaction. B) The lysate mixing experiment was repeated in triplicate and product bands were quantitated by phosphorimaging. The graph shows the avereage Cdk4 activity in mixed and unmixed lysates as a fraction of the activity recovered from lysates from E2 treated cells; error bars indicate one standard deviation. The white bars ("theoretical") indicate the fractional representation of E2 lysate in the mixtures at each mixing ratio (see text). 113 Cells were cultured in 100 nM ICI or E2 for 48 h, then harvested and cellular lysates prepared. Increasing amounts (20, 50 and 100 ug) of ICI treated MCF-7 cell lysates were added to a fixed amount (100 ug) of E2 treated MCF-7 or LCC9 lysate. The mixed lysates and unmixed controls were then incubated briefly and subjected to Cdk4 kinase assay as described in Material and Methods. A representative autoradiogram is shown in Figure 4A. Cdk4 activity in unmixed lysates is shown in the first four lanes, and kinase activity obtained with pre-immune control antibodies in the next four lanes. Consistent with the results in Figure 2, Cdk4 associated kinase activity is tightly regulated by ICI and E2 in MCF-7 cells. In LCC9 cells Cdk4 is active under both conditions, and the activity is approximately 50% of that in E2 treated MCF-7 cells. The Cdk4 activity recovered from the mixed lysates is shown in the last six lanes. Consistent with our earlier report, Cdk4 in lysates of E2 treated MCF-7 cells was inhibited in a dose dependent manner by the lysate from ICI treated MCF-7 cells. The Cdk4 activity from LCC9 cells was not refractory to inhibition by lysate from ICI treated MCF-7 cells. This experiment was performed three times, and the combined results are shown in Figure 4B. The Cdk4 associated Rb kinase activity in the mixed lysates is expressed relative to the activity in the unmixed lysate from both E2 treated samples. The bars marked “theoretical” indicate the fractional representation of the E2 treated lysate in each mixed lysate; for example at the 1 to 2 ratio, there is 100 ug of E2 treated lysate and 50 ug of ICI treated lysate; the fractional representation of the E2 treated lysate is 100/(100+50) = 0.67. At each mixing ratio, the Cdk4 activity recovered is significantly less than that predicted by dilution of the active enzyme with inactive enzyme, and there was no significant difference between the Cdk4 activity recovered from MCF-7 and 114 LCC9 cells. These data suggest that although Cdk4 is not inactivated by ICI in LCC9 cells, LCC9’s Cdk4 complexes remains sensitive to inhibition by the ICI regulated inhibitory factor present in MCF-7 cells; this inhibitory activity is attributed to p21. To support the generality of this finding, we transfected MCF-7 and LCC9 cells with an expression vector encoding p21 as a fusion protein with green fluorescent protein (GFP) and an empty GFP control vector. Cells were then stained with a cell permeable DNA binding fluorescent dye, and cells were analyzed by dual parameter flow cytometry for GFP expression and DNA content. Proliferation in the presence of E2 was inhibited by expression of GFP-p21 in both MCF-7 and LCC9 cell lines (data not shown), suggesting that Cdk4 and/or Cdk2 in LCC9 cells remains sensitive to p21 inhibition, and that this inhibition leads to cell cycle arrest. One possible explanation for the constitutive Cdk2 and Cdk4 activity in LCC9 cells despite high p21 levels and sensitivity to inhibition by p21 is that LCC9 cells’ p21 is mutant. A mutant p21 that is unable to inhibit Cdk activity while retaining Cdk binding activity was previously isolated from primary human breast cancer (2). To determine if this or some other mutation was responsible for LCC9’s lack of Cdk regulation, we amplified the p21 and p27 coding sequences from MCF-7 and LCC9 cells by reverse transcriptase-polymerase chain reaction using primers directed at their 3’ and 5’ untranslated regions. The entire coding sequences were sequenced, and no differences existed between MCF-7 and LCC9. The p21 and p27 sequences from both cell lines were identical to the published sequences (GenBank accession numbers AF497972 and AF247551, respectively, data not shown). We therefore conclude that a mutant Cdkl could not account for LCC9’s antiestrogen resistant phenotype. 115 MCF7 5523 IC E2 __ .c. E2 cyclin Di ‘_} saw, 2» .uvir. actin 1“ 1~ ~ the Figure 5: LCC9 cells have elevated cyclin D levels and deregulated p21. MCF-7 and LCC9 were plated in FBS, then treated with either 10 nM ICI or 10 nM E2 for 48 h. Lysates were prepared, and quantitated, and aliquots were Western blotted with antibodies recogniziung cyclin D1, cyclin B, Cdk2, p21, p27 and actin. 116 Altered expression of cell cycle regulators in LC C 9 cells. The altered regulation of Cdk4 and Cdk2 activity in LCC9 cells by ICI could be caused by changes in expression levels of proteins which regulate Cdk activity. We compared the expression of a panel of cell cycle regulators in MCF-7 and LCC9 cells in the presence of ICI and E2. Cells were treated for 48 h with ICI or E2, then harvested and analyzed by Western blotting for cyclins D, E and A, Cdk2, p21, p27 and actin. The results from one experiment are shown in Figure 5. Consistent with our earlier observation (Figure 2C), cyclin D protein levels were slightly higher in LCC9 cells than in MCF-7 cells. Cyclin E and Cdk2 were expressed at equal levels and were unregulated in the two cell lines. While expression of the Cdkls p21 and p27 was upregulated after two day ICI treatment of MCF-7 cells, in LCC9 cells p21 was not regulated and p27 was only slightly upregulated by ICI. In both ICI and E2 treated LCC9 cells, Cdkl levels were higher than in E2 treated MCF-7 cells. This experiment was repeated with identical results. Discussion This work describes an analysis of the LCC9 cell line, an antiestrogen resistant derivative of the MCF-7 human breast cancer cell line originally selected and characterized by Brunner et a1 (6). Despite expressing ER, LCC9 exhibit E2 independent and ICI resistant grth in vitro and in nude mice. However, the molecular basis for antiestrogen resistance in this cell line is unknown. Here we compare regulation through 117 the G1 phase cell cycle check point in ICI and E2 treated LCC9 cells with the well characterized responses of MCF-7. As is the case in MCF-7 cells, a canonical ERE is transcriptionally activated by E2 in LCC9 cells and this activation is inhibited by ICI (Figure 2). This suggests that LCC9’s ER functions normally, although this conclusion must be qualified in two ways. First, basal transcription from an ERE containing promoter, in the presence of ICI or in CSS alone, is slightly but reproducibly higher in LCC9 cells than in MCF-7. It is difficult to judge the contribution of this small difference towards LCC9’s antiestrogen resistance, but it is possible that the threshold of transcriptional activity required for proliferation is quite low and LCC9 cells have enough ER activity in ICI to permit growth. Second, this assay only measures transcriptional activity at a consensus ERE. Growing evidence suggests that ER can regulate transcription via many different promoter elements, including non-consensus EREs and ERE half sites (29, 37, 38, 49) and AP-l sites (30, 39, 45). LCC9’s antiestrogen resistance may be due to an alteration in ER mediated transcription of one or more critical genes, although the ER retains wild type activity at a consensus ERE. Future studies will compare the regulation of transcription at alternative ER regulated promoters in MCF-7 and LCC9 cells. Transit through G] into S phase is dependent on the activity of both Cdk4 and Cdk2, and these activities are regulated by posttranslational modification, by association with cyclins, and by association with Cdkls (27, 28, 35, 57). The proliferative potential of a cell in any given context depends on a balance between activators (cyclins) and inhibitors (i.e. p21) of G1 phase Cdks. In MCF-7 cells Cdk4 and Cdk2 are tightly regulated by ICI and E2. In contrast, LCC9 cells’ G1 Cdk activities are not affected by 118 ICI or E2 treatment, although they remain sensitive to an inhibitory activity in ICI treated MCF-7 cells previously attributed to p21 (Figures 3 and 4) (59). Both cyclin D1 and p21 appear to be expressed at higher levels in E2 treated LCC9 compared to MCF-7 cells, and while in MCF-7 cells p21 is highly induced by two days treatment with ICI, p21 is not regulated by ICI in LCC9 cells (Figure 5). A cell in G1 phase decides whether or not to enter S phase based on the balance of Cdk activators and inhibitors. In the presence of E2, the balance of activators and inhibitors favors active Cdks in both MCF-7 and LCC9 cells, and active Cdks phosphorylate Rb and other critical S phase substrates. In MCF-7 cells, ICI treatment increases Cdkl levels until all Cdk4 and Cdk2 is inactive, and proliferation ceases. In contrast, the balance remains in favor of active Cdks in ICI treated LCC9 cells due to a lack of p21 upregulation, allowing antiestrogen resistant proliferation. This work supports a model of acquired antiestrogen resistance in which signaling alterations occur upstream of G1 Cdk activation. Both the increased cyclin D1 expression and the lack of p21 induction upon ICI treatment may contribute to active G1 Cdks, and ultimately proliferation. This points to a number of signal transduction pathways that merit further study for their potential contribution to antiestrogen resistance in LCC9 cells. During the preparation of this manuscript, Clarke et al, using expression and promoter analysis, reported that levels of transcription from a cAMP response element was increased in LCC9 cells compared with their parental cell line, and that levels of epidermal growth factor receptor (EGF-R), which activates STATS, were lower (21). These reported alterations may account for elevated levels of cyclin D1 and the lack of p21 regulation observed in LCC9 cells. Estrogen regulation of the cyclin D1 119 promoter has been demonstrated to involve a CAMP response element (CRE), (8, 55) and the induction of p21 is dependent upon STAT signaling (3, l2). Overactive CRE transcription could lead to increased cyclin D] levels, and decreased levels of EGF-R could depress STAT signaling and account for the lack of p21 regulation in LCC9 cells. Further comparison of these signaling pathways in MCF-7 and LCC9 cells to determine if corruption of one or more pathways could contribute to the development of acquired antiestrogen resistance is warranted. 120 References 10. Atanaskova, N., V. G. Keshamouni, J. S. Krueger, J. A. Schwartz, F. Miller, and K. B. Reddy. 2002. MAP kinase/estrogen receptor cross-talk enhances estrogen- mediated signaling and tumor growth but does not confer tamoxifen resistance. Oncogene 21 14000-8. Balbin, M., G. J. Hannon, A. M. Pendas, A. A. F errando, F. Vizoso, A. Fueyo, and C. Lopez-Otin. 1996. Functional analysis of a p21WAF1,CIP1,SDII mutant (Arg94 --> Trp) identified in a human breast carcinoma. Evidence that the mutation impairs the ability of p21 to inhibit cyclin-dependent kinases. J Biol Chem 271:15782-6. Bellido, T., C. A. O'Brien, P. K. Roberson, and S. C. Manolagas. 1998. Transcriptional activation of the p21(WAF 1,CIP1 ,SDIl) gene by interleukin-6 type cytokines. A prerequisite for their pro- differentiating and anti-apoptotic effects on human osteoblastic cells. J Biol Chem 273:21137-44. Briand, P., and A. E. Lykkesfeldt. 1984. Effect of estrogen and antiestrogen on the human breast cancer cell line MCF-7 adapted to growth at low serum concentration. Cancer Res 44:1114-9. Brooks, S. C., E. R. Locke, and H. D. Soule. 1973. Estrogen receptor in a human cell line (MCF-7) from breast carcinoma. J Biol Chem 248:6251-3. Brunner, N., B. Boysen, S. Jirus, T. C. Skaar, C. Holst-Hansen, J. Lippman, T. Frandsen, M. Spang-Thomsen, S. A. Fuqua, and R. Clarke. 1997. MCF7/LCC9: an antiestrogen-resistant MCF-7 variant in which acquired resistance to the steroidal antiestrogen ICI 182,780 confers an early cross-resistance to the nonsteroidal antiestrogen tamoxifen. Cancer Res 57:3486-93. Carroll, J. S., O. W. Prall, E. A. Musgrove, and R. L. Sutherland. 2000. A pure estrogen antagonist inhibits cyclin E-Cdk2 activity in MCF-7 breast cancer cells and induces accumulation of p1 30-E2F4 complexes characteristic of quiescence. J Biol Chem 275:38221-9. Castro-Rivera, E., 1. Samudio, and S. Safe. 2001. Estrogen regulation of cyclin D1 gene expression in ZR-75 breast cancer cells involves multiple enhancer elements. J Biol Chem 276:30853-61. Cavailles, V., S. Dauvois, F. L'Horset, G. Lopez, S. Hoare, P. J. Kushner, and M. G. Parker. 1995. Nuclear factor RIP140 modulates transcriptional activation by the estrogen receptor. Embo J 14:3741-51. Chan, C. M., A. E. Lykkesfeldt, M. G. Parker, and M. Dowsett. 1999. Expression of nuclear receptor interacting proteins TIF-l , SUG-l , receptor interacting protein 121 ll. 12. l3. 14. 15. 16. 17. 18. 19. 20. 21. 140, and corepressor SMRT in tamoxifen- resistant breast cancer. Clin Cancer Res 5:3460—7. Cheng, M., P. Olivier, J. A. Diehl, M. Fero, M. F. Roussel, J. M. Roberts, and C. J. Sherr. 1999. The p21(Cip1) and p27(Kipl) CDK 'inhibitors' are essential activators of cyclin D-dependent kinases in murine fibroblasts. Embo J 1821571- 83. Chin, Y. E., M. Kitagawa, W. C. Su, Z. H. You, Y. Iwamoto, and X. Y. Fu. 1996. Cell growth arrest and induction of cyclin-dependent kinase inhibitor p21 WAF 1/CIP1 mediated by STATI. Science 272:719-22. Clarke, R., T. C. Skaar, K. B. Bouker, N. Davis, Y. R. Lee, J. N. Welch, and F. Leonessa. 2001. Molecular and pharmacological aspects of antiestrogen resistance. J Steroid Biochem Mol Biol 76:71-84. Clarke, R., E. W. Thompson, F. Leonessa, J. Lippman, M. McGarvey, T. L. Frandsen, and N. Brunner. 1993. Hormone resistance, invasiveness, and metastatic potential in breast cancer. Breast Cancer Res Treat 24:227-39. DeFriend, D. J., B. Anderson, J. Bell, D. P. Wilks, C. M. West, R. E. Mansel, and A. Howell. 1994. Effects of 4-hydroxytamoxifen and a novel pure antioestrogen (ICI 182780) on the clonogenic growth of human breast cancer cells in vitro. Br J Cancer 702204-11. DeFriend, D. J ., A. Howell, R. I. Nicholson, B. Anderson, M. Dowsett, R. E. Mansel, R. W. Blarney, N. J. Bundred, J. F. Robertson, C. Saunders, and et a1. 1994. Investigation of a new pure antiestrogen (ICI 182780) in women with primary breast cancer. Cancer Res 54:408-14. Foster, J. S., D. C. Henley, A. Bukovsky, P. Seth, and J. Wimalasena. 2001. Multifaceted regulation of cell cycle progression by estrogen: regulation of Cdk inhibitors and Cdc25A independent of cyclin D1-Cdk4 function. Mol Cell Biol 21:794-810. Foster, J. S., and J. Wimalasena. 1996. Estrogen regulates activity of cyclin- dependent kinases and retinoblastoma protein phosphorylation in breast cancer cells. Mol Endocrinol 10:488-98. Graham, J. D., D. L. Bain, J. K. Richer, T. A. Jackson, L. Tung, and K. B. Horwitz. 2000. Nuclear receptor conformation, coregulators, and tamoxifen- resistant breast cancer. Steroids 65:579-84. Grese, T. A., and J. A. Dodge. 1998. Selective estrogen receptor modulators (SERMs). Curr Pharm Des 4:71-92. Gu, Z., R. Y. Lee, T. C. Skaar, K. B. Bouker, J. N. Welch, J. Lu, A. Liu, Y. Zhu, N. Davis, F. Leonessa, N. Brunner, Y. Wang, and R. Clarke. 2002. Association of 122 22. 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. Interferon Regulatory F actor-l, Nucleophosmin, Nuclear F actor-kappaB, and Cyclic AMP Response Element Binding with Acquired Resistance to Faslodex (ICI 182,780). Cancer Res 62:3428-37. Harper, J. W., G. R. Adami, N. Wei, K. Keyomarsi, and S. J. Elledge. 1993. The p21 Cdk-interacting protein Cipl is a potent inhibitor of G1 cyclin- dependent kinases. Cell 75:805-16. Horwitz, K. B. 1993. Mechanisms of hormone resistance in breast cancer. Breast Cancer Res Treat 26:119-30. Horwitz, K. B., M. E. Costlow, and W. L. McGuire. 1975. MCF-7; a human breast cancer cell line with estrogen, androgen, progesterone, and glucocorticoid receptors. Steroids 262785-95. Howell, A., C. K. Osborne, C. Morris, and A. E. Wakeling. 2000. ICI 182,780 (Faslodex): development of a novel, "pure" antiestrogen. Cancer 89:817-25. Hunter, D. S., L. C. Hodges, P. M. Vonier, R. F uchs-Young, M. M. Gottardis, and C. L. Walker. 1999. Estrogen receptor activation via activation function 2 predicts agonism of xenoestrogens in normal and neoplastic cells of the uterine myometrium. Cancer Res 59:3090-9. Hunter, T., and J. Pines. 1991. Cyclins and cancer. Cell 66:1071-4. Hunter, T., and J. Pines. 1994. Cyclins and cancer. 11: Cyclin D and CDK inhibitors come of age. Cell 79:573-82. Hyder, S. M., C. Chiappetta, and G. M. Stance]. 1998. The 3-flanking region of the mouse c-fos gene contains a cluster of GGTCA hormone response-like elements. Mol Biol Rep 25: 189-91 . Jakacka, M., M. Ito, J. Weiss, P. Y. Chien, B. D. Gehm, and J. L. Jameson. 2001. Estrogen receptor binding to DNA is not required for its activity through the nonclassical APl pathway. J Biol Chem 276:13615-21. Johnston, S. R. 1997. Acquired tamoxifen resistance in human'breast cancer-- potential mechanisms and clinical implications. Anticancer Drugs 8:911-30. Johnston, S. R., G. Saccani-Jotti, I. E. Smith, J. Salter, J. Newby, M. Coppen, S. R. Ebbs, and M. Dowsett. 1995. Changes in estrogen receptor, progesterone receptor, and p82 expression in tamoxifen-resistant human breast cancer. Cancer Res 55:3331-8. Jordan, V. C. 1983. Laboratory studies to develop general principles for the adjuvant treatment of breast cancer with antiestrogens: problems and potential for future clinical applications. Breast Cancer Res Treat 3:873-86. 123 34. 35. 36. 37. 38. 39. 40. 41. 42. 43. 44. 45. Jordan, V. C. 1992. The strategic use of antiestrogens to control the development and growth of breast cancer. Cancer 70:977-82. Kato, J. Y., M. Matsuoka, D. K. Strom, and C. J. Sherr. 1994. Regulation of cyclin D-dependent kinase 4 (cdk4) by cdk4-activating kinase. Mol Cell Biol 14:2713-21. Kato, S., H. Endoh, Y. Masuhiro, T. Kitamoto, S. Uchiyama, H. Sasaki, S. Masushige, Y. Gotoh, E. Nishida, H. Kawashima, and et al. 1995. Activation of the estrogen receptor through phosphorylation by mitogen— activated protein kinase. Science 270: 1491-4. Kim, J., L. N. Petz, Y. S. Ziegler, J. R. Wood, S. J. Potthoff, and A. M. Nardulli. 2000. Regulation of the estrogen-responsive p82 gene in MCF-7 human breast cancer cells. J Steroid Biochem Mol Biol 74:157-68. Klinge, C. M., D. L. Bodenner, D. Desai, R. M. Niles, and A. M. Traish. 1997. Binding of type 11 nuclear receptors and estrogen receptor to full and half-site estrogen response elements in vitro. Nucleic Acids Res 25:1903-12. Kushner, P. J ., D. A. Agard, G. L. Greene, T. S. Scanlan, A. K. Shiau, R. M. Uht, and P. Webb. 2000. Estrogen receptor pathways to AP-l. J Steroid Biochem Mol Biol 74:311-7. Liu, M. M., C. Albanese, C. M. Anderson, K. Hilty, P. Webb, R. M. Uht, R. H. Price, Jr., R. G. Pestell, and P. J. Kushner. 2002. Opposing Action of Estrogen Receptors alpha and beta on Cyclin D1 Gene Expression. J Biol Chem 277:24353-24360. Matsushime, H., D. E. Quelle, S. A. Shurtleff, M. Shibuya, C. J. Sherr, and J. Y. Kato. 1994. D-type cyclin-dependent kinase activity in mammalian cells. Mol Cell Biol 14:2066-76. Mosselman, S., J. Polman, and R. Dijkema. 1996. ER beta: identification and characterization of a novel human estrogen receptor. FEBS Lett 392:49-53. Osborne, C. K. 1993. Mechanisms for tamoxifen resistance in breast cancer: possible role of tamoxifen metabolism. J Steroid Biochem Mol Biol 47:83-9. Osborne, C. K., R. Schiff, S. A. Fuqua, and J. Shou. 2001. Estrogen receptor: current understanding of its activation and modulation. Clin Cancer Res 7:4338s- 43423; discussion 441 ls- 44128. Paech, K., P. Webb, G. G. Kuiper, S. Nilsson, J. Gustafsson, P. J. Kushner, and T. S. Scanlan. 1997. Differential ligand activation of estrogen receptors ERalpha and ERbeta at AP] sites. Science 277:1508-10. 124 46. 47. 48. 49. 50. 51. 52. 53. 54. 55. 56. 57. 58. 59. Paik, S., D. P. Hartmann, R. B. Dickson, and M. E. Lippman. 1994. Antiestrogen resistance in ER positive breast cancer cells. Breast Cancer Res Treat 31 :301-7. Parker, M. G. 1993. Action of "pure" antiestrogens in inhibiting estrogen receptor action. Breast Cancer Res Treat 26:131-7. Parker, M. G., N. Arbuckle, S. Dauvois, P. Danielian, and R. White. 1993. Structure and function of the estrogen receptor. Ann N Y Acad Sci 684:119-26. Petz, L. N., and A. M. Nardulli. 2000. Spl binding sites and an estrogen response element half-site are involved in regulation of the human progesterone receptor A promoter. Mol Endocrinol 14:972-85. Piccart, M., C. Lohrisch, A. Di Leo, and D. Larsimont. 2001. The predictive value of HER2 in breast cancer. Oncology 61:73-82. Planas-Silva, M. D., and R. A. Weinberg. 1997. Estrogen-dependent cyclin E- cde activation through p21 redistribution. Mol Cell Biol 17:4059-69. Polyak, K., J. Y. Kato, M. J. Solomon, C. J. Sherr, J. Massague, J. M. Roberts, and A. Koff. 1994. p27Kip1, a cyclin-Cdk inhibitor, links transforming growth factor-beta and contact inhibition to cell cycle arrest. Genes Dev 819-22. Prall, O. W., B. Sarcevic, E. A. Musgrove, C. K. Watts, and R. L. Sutherland. 1997. Estrogen-induced activation of Cdk4 and Cdk2 during Gl-S phase progression is accompanied by increased cyclin D1 expression and decreased cyclin-dependent kinase inhibitor association with cyclin E- Cdk2. J Biol Chem 272: 10882-94. Reddel, R. R., L. C. Murphy, and R. L. Sutherland. 1983. Effects of biologically active metabolites of tamoxifen on the proliferation kinetics of MCF-7 human breast cancer cells in vitro. Cancer Res 43 :461 8-24. Sabbah, M., D. Courilleau, J. Mester, and G. Redeuilh. 1999. Estrogen induction of the cyclin D1 promoter: involvement of a cAMP response-like element. Proc Natl Acad Sci U S A 96:11217-22. Safe, S. 2001. Transcriptional activation of genes by 17 beta-estradiol through estrogen receptor-Spl interactions. Vitam Horm 62:231-52. Sherr, C. J. 1996. Cancer cell cycles. Science 274:1672-7. Sherr, C. J ., and J. M. Roberts. 1999. CDK inhibitors: positive and negative regulators of Gl-phase progression. Genes Dev 13:1501-12. Skildum, A. J ., S. Mukherjee, and S. E. Conrad. 2001. The cyclin dependent kinase inhibitor p21WAF ”C'p' is an antiestrogen regulated inhibitor of Cdk4 in human breast cancer cells. J Biol Chem 277(7):5145-52. 125 60. 61. 62. 63. 64. 65. 66. 67. 68. 69. 70. Smith, C. L., Z. Nawaz, and B. W. O'Malley. 1997. Coactivator and corepressor regulation of the agonist/antagonist activity of the mixed antiestrogen, 4- hydroxytamoxifen. Mol Endocrinol 11:657-66. Soule, H. D., J. Vazguez, A. Long, S. Albert, and M. Brennan. 1973. A human cell line from a pleural effusion derived from a breast carcinoma. J Natl Cancer Inst 51:1409-16. Sutherland, R. L., M. D. Green, R. E. Hall, R. R. Reddel, and I. W. Taylor. 1983. Tamoxifen induces accumulation of MCF 7 human mammary carcinoma cells in the GO/Gl phase of the cell cycle. Eur J Cancer Clin Oncol 19:615-21. Takimoto, G. S., J. D. Graham, T. A. Jackson, L. Tung, R. L. Powell, L. D. Horwitz, and K. B. Horwitz. 1999. Tamoxifen resistant breast cancer: coregulators determine the direction of transcription by antagonist-occupied steroid receptors. J Steroid Biochem Mol Biol 69:45-50. Tremblay, A., G. B. Tremblay, F. Labrie, and V. Giguere. 1999. Ligand- independent recruitment of SRC-l to estrogen receptor beta through phosphorylation of activation firnction AF-l. Mol Cell 3:513-9. Tremblay, G. B., A. Tremblay, N. G. Copeland, D. J. Gilbert, N. A. Jenkins, F. Labrie, and V. Giguere. 1997. Cloning, chromosomal localization, and functional analysis of the murine estrogen receptor beta. Mol Endocrinol 112353-65. Wade, C. B., S. Robinson, R. A. Shapiro, and D. M. Dorsa. 2001. Estrogen receptor (ER)alpha and ERbeta exhibit unique pharmacologic properties when coupled to activation of the mitogen-activated protein kinase pathway. Endocrinology 142:2336-42. Wakeling, A. E., B. Valcaccia, E. Newboult, and L. R. Green. 1984. Non- steroidal antioestrogens--receptor binding and biological response in rat uterus, rat mammary carcinoma and human breast cancer cells. J Steroid Biochem 20:111- 20. Watts, C. K., A. Brady, B. Sarcevic, A. deFazio, E. A. Musgrove, and R. L. Sutherland. 1995. Antiestrogen inhibition of cell cycle progression in breast cancer cells in associated with inhibition of cyclin-dependent kinase activity and decreased retinoblastoma protein phosphorylation. Mol Endocrinol 9: 1 804-13. Watts, C. K., K. J. Sweeney, A. Warlters, E. A. Musgrove, and R. L. Sutherland. 1994. Antiestrogen regulation of cell cycle progression and cyclin D1 gene expression in MCF-7 human breast cancer cells. Breast Cancer Res Treat 31:95- 105. Webb, P., P. Nguyen, J. Shinsako, C. Anderson, W. Feng, M. P. Nguyen, D. Chen, S. M. Huang, S. Subramanian, E. McKinemey, B. S. Katzenellenbogen, M. 126 71. 72. 73. R. Stallcup, and P. J. Kushner. 1998. Estrogen receptor activation function 1 works by binding p160 coactivator proteins. Mol Endocrinol 12:1605-18. Webb, P., P. Nguyen, C. Valentine, G. N. Lopez, G. R. Kwok, E. McInemey, B. S. Katzenellenbogen, E. Enmark, J. A. Gustafsson, S. Nilsson, and P. J. Kushner. 1999. The estrogen receptor enhances AP-l activity by two distinct mechanisms with different requirements for receptor transactivation functions. Mol Endocrinol 13:1672-85. Wolmark, N., and B. K. Dunn. 2001. The role of tamoxifen in breast cancer prevention: issues sparked by the NSABP Breast Cancer Prevention Trial (P-l). Ann N Y Acad Sci 949199-108. Xiong, Y., G. J. Harmon, H. Zhang, D. Casso, R. Kobayashi, and D. Beach. 1993. p21 is a universal inhibitor of cyclin kinases. Nature 366:701-4. 127 CHAPTER FIVE Conclusion 128 Chapter 5: Conclusion Introduction. Antiestrogens are drugs that bind to ERa’s ligand binding domain and prevent transcription of some or all E2 regulated genes. Because of E2’s role as a mitogen in normal and malignant breast tissue, antiestrogens such as tamoxifen have been used to successfully treat breast cancer patients with ERa positive tumors since the 1970s (19, 21). One limitation of their use is that during treatment, many patients’ tumors bypass their requirement for E2 and become insensitive to growth arrest by antiestrogens (35). Despite much attention, the molecular mechanisms responsible for this change in phenotype is not well understood. Research in this area may lead to better therapeutic techniques that avoid or delay the problem of antiestrogen resistance. In hormone sensitive cells, E2 promotes passage through the G1 phase of the cell cycle, and antiestrogens cause a G1 phase cell cycle arrest (32). The G198 phase transition is controlled by the sequential activation of Cdk4 and Cdk2, which phosphorylate Rb; Cdk2 also has additional substrates. Cdk activity depends on association with a cyclin, a regulatory subunit. Cdk activity can be further modified by association with Cdkls, such as p21 (16, 17, 29). In clinical breast cancer, the main regulatory subunit of Cdk4, cyclin D1 , is often overexpressed, and overexpression correlates with ERa-positive status (15, 18, 27). Investigations into hormone signaling in models of human breast cancer revealed that E2 activates, and antiestrogens inhibit, Cdk4 and Cdk2 activities. It is proposed that E2 increases cyclin D1 protein levels, which activates Cdk4 and provides additional binding targets for p21, tritrating it from inhibitory association with Cdk2 (24). E2 also decreases p21 protein levels, contributing to further increases in Cdk activity. 129 These observations led us to investigate the possible relationship between the regulation of G1 phase Cdk activity and antiestrogen resistance. The hypothesis of this dissertation is that alterations in signaling pathways leading to inappropriate activation of G1 Cdks contributes to antiestrogen resistant proliferation in in vitro models of human breast cancer cells. This hypothesis was explored by examining the regulation of Cdk4 by E2 and the pure steroidal antiestrogen ICI in the E2 dependent, ICI sensitive MCF-7 cell line (chapter 2). We then studied the effects of expressing ectopic cyclin D1 on nsIA's-I“. um regulation of Cdks and proliferation in an MCF-7 subclone engineered to conditionally express cyclin D1 (chapter 3). Finally, we compared the effects of E2 and ICI on MCF-7 with LCC9 cells, an ICI resistant derivative of MCF-7 (chapter 4). 1E. Significance of major findings. In chapter 2, the regulation of Cdk4 by ICI and E2 in MCF-7 cells was examined. In time course experiments, Cdk4 activity was inhibited by ICI and activated by E2 treatment, consistent with previous reports (8, 26, 33). In contrast to previous reports, no correlation was observed between levels of cyclin D1 protein and Cdk4 activity, although there was an inverse correlation between p21 protein levels and Cdk4 activity. Through lysate mixing experiments, we demonstrated that lysates of ICI treated MCF-7 cells inhibit active Cdk4 from E2 treated MCF-7 cells in a dose dependent manner. Furthermore, immunodepletion of p21, but not p27, from the lysate of ICI treated cells abolished the Cdk4 inhibitory activity in the lysate, suggesting p21 was responsible for Cdk4 inhibition. Although p21 may serve as an activator of cyclin Dl-Cdk4 complexes under some conditions (4, 30, 34), in the context of ICI mediated grth arrest it clearly acts as a Cdk4 inhibitor. 130 We conclude that p21 is an ICI regulated inhibitor of Cdk4 that can act independently of changes in Cdk4’s major regulatory subunit, cyclin D1. These results corroborate the work of Foster et al, who showed that E2 downregulates Cdkl expression in MCF-7 cells even when cell cycle arrest is enforced inhibition of cyclin Dl-Cdk4 by ectopic p16, suggesting that E2 directly regulates Cdkl expression independently of progression through the cell cycle (7). In MCF-7 cells, p21 controls the activity of critical G1 -)S phase regulators, and is a direct target of ICI and E2. The inhibitory activity of p21 can be overcome by ectopic expression of Cdk4’s main regulatory subunit, cyclin D1. MCF-7 cells were engineered to conditionally express epitope tagged human cyclin D1 to assess its potential to contribute to acquired antiestrogen resistance (chapter 3). Consistent with results from other labs, we found that ectopic cyclin D1 expression caused G1 arrested cells to enter S phase in the presence of antiestrogen, but was insufficient to support long term antiestrogen resistant proliferation. In time course experiments, ectopic cyclin D1 activated Cdk4 in the presence of ICI. While E2 treatment resulted in sustained Cdk4 activation, expression of ectopic cyclin D1 in the presence of ICI did not sustain activity beyond 72 h, although ectopic cyclin D1 remained in complex with Cdk4 up to 120 h. Levels of p21 protein increased during antiestrogen treatment, and decreased with E2 treatment. These data provide a possible mechanism for the paradoxical ability of ectopic cyclin D1 to cause ICI arrested cells to enter the cell cycle, but its inability to promote long term antiestrogen resistant proliferation (20, 22, 36). We propose a model whereby in ICI arrested cells, Cdk4 is in excess and the amount of cyclin D1 is limiting for proliferation because all cyclin Dl/Cdk4 complexes are in inhibitory association with a 131 Cdkl. Increasing the amount of cyclin D1 using regulated expression systems transiently increases the amount of p21-free cyclin D1/Cdk4 complexes, which are active kinases and promote proliferation. Upon long term treatment with ICI, p21 levels accumulate such that all cyclin D1-Cdk4 complexes contain p21 and are catalytically inhibited. When cells are treated with ICI, p21 levels may increase up to some fixed level, or, alternatively, cells may have a “sensor” of Cdk4 activity which acts with ICI to increase p21 levels until all cyclin Dl-Cdk4 complexes are inhibited. In support of this possibility, in other systems expression of ectopic cyclin D1 has been shown to increase ("‘12 3 _'I E2F transcription factor binding and activity at the p21 promoter, leading to increased p21 protein levels (13, 14). This model predicts that cyclin D1 amplification alone is an F|—_.,, 9, .. .. unlikely cause of acquired antiestrogen resistance, and that additional changes, such as in Cdkl regulation of Cdk complexes, would be necessary for the development of antiestrogen resistant tumors. To further examine potential changes in G1 -)S phase regulation that may contribute to antiestrogen resistance, the MCF-7 cells were compared with the LCC9 cell line, an MCF-7 derivative selected for ICI resistant growth (chapter 4) (2). LCC9 cells were insensitive to growth arrest by ICI, despite having an apparently normally functioning ER. While Cdk4 and Cdk2 are inactivated following ICI treatment of MCF-7 cells, in LCC9 both Cdks remain active. Active Cdk4 from LCC9 cells was not refractory to inhibition by p21 from lysates prepared from ICI treated MCF-7 cells, suggesting that no change in the Cdk4 complex’s ability to be inhibited by p21 occurred during selection of the LCC9 cell line. However, while levels of p21 and p27 in MCF-7 cells increased after two days ICI treatment, p21 was not regulated by E2 and ICI in 132 LCC9 and p27 was only slightly increased by ICI. In addition, while cyclin D1 protein levels were not regulated by ICI and E2 in either cell line, LCC9 cells showed consistently higher levels of cyclin D1 than the parental MCF-7 cells. In LCC9 cells, increased cyclin D1 levels may increase the total amount of cyclin D1-Cdk4 complexes, and deregulated Cdkls may lead to constitutively active Cdk4 and Cdk2. The unregulated Cdks would lead to increased phosphorylation of Rb and Cdk substrates, contributing to estrogen insensitive and antiestrogen resistant grth (Figure r 1). However, the fundamental causes of the changes in cyclin D1 levels and p21 . regulation in the LCC9 cell line are not known. E2 is reported to regulate the cyclin D1 promoter by ERa’s direct interaction with is. Spl at Spl binding sites, and indirectly by protein kinase A dependent and independent CAMP response element activation (CRE) (3, 28); cyclin D1 transcription is also regulated positively by NF-KB (12). Transcription of p21 is activated by Spl and STAT transcription factors, and Myc association with Spl and Sp3 inhibits p21 expression; Myc levels are increased by E2 treatment in breast cancer cells. (5, 6, 9, 31). STATS are activated by a number of cytokine and tyrosine kinase receptors and are implicated in p21 mediated growth arrest by induced by interferon-y and transforming growth factor-[3 (5, 10). STAT induced expression of p21 is also implicated in the growth arrest caused by epidermal growth factor (EGF) in the MDA468 human breast cancer cell line, which overexpresses EGF-receptor (EGF-R). The transcriptional regulation of cyclin D1 and/or p21 may underlie LCC9s antiestrogen resistant phenotype. One point at which cyclin D1 and p21 regulation by ER ligands may intersect is the Spl transcription factor. Both promoters are activated by Spl 133 (3, 9), and direct interaction between ER and Spl is responsible for E2 mediated transcription of progesterone receptor (23). Myc acts as a transcriptional repressor at Spl sites of the p21 promoter (9). If Myc expression or its activity was increased through mutation or epigenetic change, the induction of p21 by ICI might be diminished or abolished. Myc levels are increased by E2 and decreased by ICI, and ectopic Myc expression in MCF-7 cells has been reported to cause ICI arrested cells to enter the cell cycle and activates cyclin E-Cdk2 without activating cyclin D1-Cdk4 (6, 25), suggesting that Myc may act downstream of cyclin D1. Other alterations in the regulation of cyclin D1 and p21 might also be involved in causing the LCC9 cell line’s antiestrogen resistance. MCF-7 cells’ basal and ICI regulated gene expression was recently compared with LCC9s (11). LCC9 cells have increased levels of basal transcription from CRE and NF-IcB site reporter plasmids, and unlike MCF-7 cells, their NF-KB activity is not inhibited by ICI. The cyclin D1 promoter is controlled by both a CRE and NF-KB binding site (12, 28), and increases in their activity may account for the increased level of cyclin D1 protein observed in LCC9 cells compared to MCF-7 cells. In addition, LCC9 cells are reported to have decreased levels of EGF-R expression and protein (11). EGF-R activates STAT transcription factors, which are implicated in p21’s transcriptional regulation (1 , 5). The reduced levels of EGF-R in LCC9 compared to MCF-7 cells may result in reduced STAT activation and reduced or deregulated expression of p21. Further examination of these signaling pathways for their potential contribution to acquired antiestrogen resistance is warranted. 134 NF-ch Cdk4 Cdk2 —> ""°”"°”'a“°"°' S phase substrates Rb CYClln D1 EZF regulated fl gene transcription I '. E2F / 8p1 CRE O .0 O 0 E2 Figure 1: Alterations in regulation of GI Cdks may lead to antiestrogen resistance. In hormone sensitive human breast cancer cells, proliferation is controlled by the balance between activating (cyclin D1) and inhibitory (p21) factors which regulate the activity of Cdk4 and Cdk2. E2 increases the level of cyclin D1 by activating transcription at Spl and CRE sites in the cyclin D1 promoter; cyclin D1 transcription is also regulated by NF-KB. E2 decreases levels of Cdkls such as p21, although mechanisms of transcriptional regulation are not described. In growth arrest by cytokine and receptor tyrosine kinases, p21 is upregulated by Spl and STAT transcription factors, and p21 is expression is inhibited by Myc. In the antiestrogen resistant LCC9 cell line, cyclin D1 levels are higher than their parental MCF-7 cells, and p21 protein levels are not regulated by ICI. Points at which the regulation of cyclin D1 and p21 may be altered in antiestrogen resistant LCC9 cells are shown with dashed lines. 135 Model of E2 and 1C1 regulation of GI 9S in MCF-7 and implications an acquired antiestrogen resistance. The experiments presented in the preceding chapters tested the hypothesis that inappropriate activation of Cdk4 and Cdk2 contributes to antiestrogen resistant proliferation in an in vitro model of human breast cancer. In addition to being an inhibitor of Cdk2, we have demonstrated that p21 is an ICI regulated inhibitor of Cdk4, and that its expression is deregulated in the LCC9 cell line, an antiestrogen resistant variant of the MCF-7 cell line. Furthermore, ectopic expression of cyclin D1 causes ICI arrested cells to enter S phase, and cyclin D1 levels are higher in LCC9 than MCF-7 cells. The data support the hypothesis, with the following qualifications: 1) The expression of ectopic cyclin D1 did not support long term Cdk4 activation or proliferation, suggesting that this single change contributes towards, but is unlikely to cause, antiestrogen resistance. In this regard, LCC9 cells, selected from the MCF-7 cell line in multiple steps, have multiple alterations, including elevated cyclin D1 protein levels and p21 deregulation. 2) Although the regulation of LCC9 cells’ Cdk activity is dramatically different than in MCF-7 cells, the composition and basic function of Cdk4 complexes appears normal. This suggests that the cause of Cdk deregulation is not due to mutational change of a Cdk complex member, but that the ultimate causes are likely to be changes in upstream regulators, particularly cyclin D1 and p21. These experiments support a model in which breast cancer cell proliferation is determined by balance between positive factors, e.g. cyclin D1, and negative factors, e.g. p21, which regulate the activity of G1 phase Cdks (Figure 1). In antiestrogen sensitive cells, antiestrogen shifts the balance in favor of negative factors, inhibiting G1 Cdk 136 activity and ultimately proliferation. In antiestrogen resistant cells, deregulated expression of cyclins and Cdkls maintains the balance in favor of active G1 Cdks, allowing proliferation in the presence of antiestrogen. Multiple molecular changes upstream of cyclin and Cdkl expression may contribute to antiestrogen resistance in human breast cancer. 137 References 10. Berclaz, G., H. J. Alterrnatt, V. Rohrbach, A. Siragusa, E. Dreher, and P. D. Smith. 2001. EGFR dependent expression of STAT3 (but not STATl) in breast cancer. Int J Oncol 19:1155-60. Brunner, N., B. Boysen, S. Jirus, T. C. Skaar, C. Holst-Hansen, J. Lippman, T. F randsen, M. Spang-Thomsen, S. A. F uqua, and R. Clarke. 1997. MCF7/LCC9: an antiestrogen-resistant MCF-7 variant in which acquired resistance to the steroidal antiestrogen ICI 182,780 confers an early cross-resistance to the nonsteroidal antiestrogen tamoxifen. Cancer Res 57:3486-93. Castro-Rivera, E., I. Samudio, and S. Safe. 2001. Estrogen regulation of cyclin D1 gene expression in ZR-75 breast cancer cells involves multiple enhancer elements. J Biol Chem 276:30853-61. Cheng, M., P. Olivier, J. A. Diehl, M. Fero, M. F. Roussel, J. M. Roberts, and C. J. Sherr. 1999. The p21(Cip1) and p27(Kipl) CDK 'inhibitors' are essential activators of cyclin D-dependent kinases in murine fibroblasts. Embo J 18:1571- 83. Chin, Y. E., M. Kitagawa, W. C. Su, Z. H. You, Y. Iwamoto, and X. Y. Fu. 1996. Cell growth arrest and induction of cyclin-dependent kinase inhibitor p21 WAFl/CIPl mediated by STAT]. Science 272:719-22. Dubik, D., and R. P. Shin. 1988. Transcriptional regulation of c-myc oncogene expression by estrogen in hormone-responsive human breast cancer cells. J Biol Chem 263:12705-8. Foster, J. S., D. C. Henley, A. Bukovsky, P. Seth, and J. Wimalasena. 2001. Multifaceted regulation of cell cycle progression by estrogen: regulation of Cdk inhibitors and Cdc25A independent of cyclin Dl-Cdk4 function. Mol Cell Biol 21 1794-810. Foster, J. S., and J. Wimalasena. 1996. Estrogen regulates activity of cyclin- dependent kinases and retinoblastoma protein phosphorylation in breast cancer cells. Mol Endocrinol 10:488-98. Gartel, A. L., X. Ye, E. Goufinan, P. Shianov, N. Hay, F. Najmabadi, and A. L. Tyner. 2001. Myc represses the p21(WAF1/CIP1) promoter and interacts with Spl/Sp3. Proc Natl Acad Sci U S A 98:4510-5. Gooch, J. L., R. E. Herrera, and D. Yee. 2000. The role of p21 in interferon gamma-mediated growth inhibition of human breast cancer cells. Cell Growth Differ 11:335-42. 138 11. 12. 13. 14. 15. 16. 17. 18. 19. 20. 21. 22. Gu, Z., R. Y. Lee, T. C. Skaar, K. B. Bouker, J. N. Welch, J. Lu, A. Liu, Y. Zhu, N. Davis, F. Leonessa, N. Brunner, Y. Wang, and R. Clarke. 2002. Association of Interferon Regulatory Factor-l , Nucleophosmin, Nuclear Factor-kappaB, and Cyclic AMP Response Element Binding with Acquired Resistance to Faslodex (ICI 182,780). Cancer Res 62:3428-37. Hinz, M., D. Krappmann, A. Eichten, A. Heder, C. Scheidereit, and M. Strauss. 1999. NF-kappaB function in growth control: regulation of cyclin D1 expression and GO/Gl-to-S-phase transition. Mol Cell Biol 19:2690-8. Hiyama, H., A. Iavarone, J. LaBaer, and S. A. Reeves. 1997. Regulated ectopic expression of cyclin D1 induces transcriptional activation of the cdk inhibitor p21 gene without altering cell cycle progression. Oncogene 14:2533-42. Hiyama, H., A. Iavarone, and S. A. Reeves. 1998. Regulation of the cdk inhibitor p21 gene during cell cycle progression is under the control of the transcription factor E2F. Oncogene 16: 1513-23. Hui, R., A. L. Cornish, R. A. McClelland, J. F. Robertson, R. W. Blarney, E. A. Musgrove, R. I. Nicholson, and R. L. Sutherland. 1996. Cyclin D1 and estrogen receptor messenger RNA levels are positively correlated in primary breast cancer. Clin Cancer Res 2:923-8. Hunter, T., and J. Pines. 1991. Cyclins and cancer. Cell 66:1071-4. Hunter, T., and J. Pines. 1994. Cyclins and cancer. 11: Cyclin D and CDK inhibitors come of age. Cell 79:573-82. Jares, P., M. J. Rey, P. L. Fernandez, E. Carnpo, A. Nada], M. Munoz, C. Mallofre, J. Muntane, I. Nayach, J. Estape, and A. Cardesa. 1997. Cyclin D1 and retinoblastoma gene expression in human breast carcinoma: correlation with tumour proliferation and oestrogen receptor status. J Pathol 182:160-6. Jordan, V. C. 1992. The strategic use of antiestrogens to control the development and growth of breast cancer. Cancer 70:977-82. Musgrove, E. A., C. S. Lee, M. F. Buckley, and R. L. Sutherland. 1994. Cyclin D1 induction in breast cancer cells shortens G1 and is sufficient for cells arrested in G1 to complete the cell cycle. Proc Natl Acad Sci U S A 91 :8022-6. Osborne, C. K., H. Zhao, and S. A. F uqua. 2000. Selective estrogen receptor modulators: structure, function, and clinical use. J Clin Oncol 18:3172-86. Pacilio, C., D. Germano, R. Addeo, L. Altucci, V. B. Petrizzi, M. Cancemi, L. Cicatiello, S. Salzano, F. Lallemand, R. J. Michalides, F. Bresciani, and A. Weisz. 1998. Constitutive overexpression of cyclin D1 does not prevent inhibition of hormone-responsive human breast cancer cell growth by antiestrogens. Cancer Res 58:871-6. 139 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. Petz, L. N., and A. M. Nardulli. 2000. Spl binding sites and an estrogen response element half-site are involved in regulation of the human progesterone receptor A promoter. Mol Endocrinol 14:972-85. Planas-Silva, M. D., and R. A. Weinberg. 1997. Estrogen-dependent cyclin E- cde activation through p21 redistribution. Mol Cell Biol 17:4059-69. Prall, O. W., E. M. Rogan, E. A. Musgrove, C. K. Watts, and R. L. Sutherland. 1998. c-Myc or cyclin D1 mimics estrogen effects on cyclin E-Cdk2 activation and cell cycle reentry. Mol Cell Biol 18:4499-508. Prall, O. W., B. Sarcevic, E. A. Musgrove, C. K. Watts, and R. L. Sutherland. 1997. Estrogen-induced activation of Cdk4 and Cdk2 during Gl-S phase progression is accompanied by increased cyclin D1 expression and decreased cyclin-dependent kinase inhibitor association with cyclin E- Cdk2. J Biol Chem 272:10882-94. Reed, W., V. A. Fllrenes, R. Holm, E. Hannisda], and J. M. Nesland. 1999. Elevated levels of p27, p21 and cyclin D1 correlate with positive oestrogen and progesterone receptor status in node-negative breast carcinoma patients. Virchows Arch 435:116-24. Sabbah, M., D. Courilleau, J. Mester, and G. Redeuilh. 1999. Estrogen induction of the cyclin D1 promoter: involvement of a cAMP response-like element. Proc Natl Acad Sci U S A 96:11217-22. Sherr, C. J. 1996. Cancer cell cycles. Science 274:1672-7. Sherr, C. J ., and J. M. Roberts. 1999. CDK inhibitors: positive and negative regulators of Gl-phase progression. Genes Dev 13: 1501-12. Sinibaldi, D., W. Wharton, J. Turkson, T. Bowman, W. J. Pledger, and R. Jove. 2000. Induction of p21WAFl/CIP1 and cyclin D1 expression by the Src oncoprotein in mouse fibroblasts: role of activated STAT3 signaling. Oncogene 19:5419-27. Sutherland, R. L., M. D. Green, R. E. Hall, R. R. Reddel, and I. W. Taylor. 1983. Tamoxifen induces accumulation of MCF 7 human mammary carcinoma cells in the GO/Gl phase of the cell cycle. Eur J Cancer Clin Oncol 19:615-21. Watts, C. K., A. Brady, B. Sarcevic, A. deFazio, E. A. Musgrove, and R. L. Sutherland. 1995. Antiestrogen inhibition of cell cycle progression in breast cancer cells in associated with inhibition of cyclin-dependent kinase activity and decreased retinoblastoma protein phosphorylation. Mol Endocrinol 9: 1 804-13. Weiss, R. H., A. Joo, and C. Randour. 2000. p21(Waf1/Cip1) is an assembly factor required for platelet-derived growth factor-induced vascular smooth muscle cell proliferation. J Biol Chem 275:10285-90. 140 35. 36. Wiebe, V. J ., C. K. Osborne, S. A. Fuqua, and M. W. DeGregorio. 1993. Tamoxifen resistance in breast cancer. Crit Rev Oncol Hematol 14: 173-88. Wilcken, N. R., O. W. Prall, E. A. Musgrove, and R. L. Sutherland. 1997. Inducible overexpression of cyclin D1 in breast cancer cells reverses the growth- inhibitory effects of antiestrogens. Clin Cancer Res 3:849-54. 141 i3129ii