GP88 (PC-Cell Derived Growth Factor, progranulin) stimulates proliferation and confers letrozole resistance to aromatase overexpressing breast cancer cells

Tesfom Abrhale, Angela Brodie, Gauri Sabnis, Luciana Macedo, Changsheng Tian, Binbin Yue, Ginette Serrero, Tesfom Abrhale, Angela Brodie, Gauri Sabnis, Luciana Macedo, Changsheng Tian, Binbin Yue, Ginette Serrero

Abstract

Background: Aromatase inhibitors (AI) that inhibit breast cancer cell growth by blocking estrogen synthesis have become the treatment of choice for post-menopausal women with estrogen receptor positive (ER+) breast cancer. However, some patients display de novo or acquired resistance to AI. Interactions between estrogen and growth factor signaling pathways have been identified in estrogen-responsive cells as one possible reason for acquisition of resistance. Our laboratory has characterized an autocrine growth factor overexpressed in invasive ductal carcinoma named PC-Cell Derived Growth Factor (GP88), also known as progranulin. In the present study, we investigated the role GP88 on the acquisition of resistance to letrozole in ER+ breast cancer cells

Methods: We used two aromatase overexpressing human breast cancer cell lines MCF-7-CA cells and AC1 cells and their letrozole resistant counterparts as study models. Effect of stimulating or inhibiting GP88 expression on proliferation, anchorage-independent growth, survival and letrozole responsiveness was examined.

Results: GP88 induced cell proliferation and conferred letrozole resistance in a time- and dose-dependent fashion. Conversely, naturally letrozole resistant breast cancer cells displayed a 10-fold increase in GP88 expression when compared to letrozole sensitive cells. GP88 overexpression, or exogenous addition blocked the inhibitory effect of letrozole on proliferation, and stimulated survival and soft agar colony formation. In letrozole resistant cells, silencing GP88 by siRNA inhibited cell proliferation and restored their sensitivity to letrozole.

Conclusion: Our findings provide information on the role of an alternate growth and survival factor on the acquisition of aromatase inhibitor resistance in ER+ breast cancer.

Figures

Figure 1
Figure 1
GP88 induces AC1 cell proliferation in vitro. 5 × 104 cells/plate (35 mm well plates) MCF-7AC1 cells were plated in PRF medium supplemented with 5% Charcoal treated FBS overnight. The next day, medium was removed and serum free medium was added without or with increasing concentration of GP88: 150 ng/ml (P-150), 300 ng/ml (P-300) and 600 ng/ml (P-600) or 25 nM androstenedione (AD). After 6 days, cells were trypsinized and cell number was determined by counting cells with a hemocytometer. Experiment was performed three times in triplicate.
Figure 2
Figure 2
GP88 confers AC1 cells Letrozole resistance in vitro. 2000 cells/well (96 well plates) MCF-7AC1 cells were plated in PRF medium supplemented with 5% ChX FBS overnight The next day, medium was removed and serum free medium was added with and without 300 ng/ml GP88 (GP88 (P)), 25 nM androstenedione (AD) and letrozole (L) 50 nM and 100 nM, 200 nM. Cell proliferation was measured after 6 days using Cell Titer-Glo assay. Experiment was performed in triplicates with p < 0.01.
Figure 3
Figure 3
GP88 induces anchorage independent colony formation and confers letrozole resistance comparable to E2. A: AC1 cells (1 × 104 cells/well in 6-well plate) were plated in soft agar plates as described in the Method section in (A) PRF-ChX with and without 300 ng/ml GP88 (P), 25 nM androstenedione (AD) and 200 nM letrozole (L) for 2 weeks. (B) in DMEM/F12 supplemented with 5%FBS, E2 = 10-8 M, 25 nM AD, +/- 300 ng/ml GP88 cultured for 3weeks. Fresh medium was added every 3 to 4 days. At the end of the experiment, colonies were fixed with crystal violet and counted, p < 0.01
Figure 4
Figure 4
Overexpression of GP88 in AC1 cells and MCF-7CA cells confer letrozole resistance. A: GP88 overexpressing AC1 cells (AC1-P) cells were developed by stable transfection of GP88 cDNA expression vector as described in the method section. As control, AC1-EV cells were developed by transfecting empty vector (AC1-EV). For measuring anchorage-independent growth of stably transfected AC1-P cells, AC1-P cells were plated at a density of 5 × 103 cells/well in a 12 wells plate and cultivated for 2 weeks with/without 200 nM letrozole, 25 nM AD, 300 ng/ml GP88 for the AC1 and CA1-EV in 5% ChX-FBS supplemented medium containing soft agar. Medium was changed every 3-4 days. B: Stimulation of anchorage independent growth of MCF-7-CA cells overexpressing GP88. The aromatase-overexpressing cells MCF-7CA were transfected with expression vector containing GP88 cDNA (CA-P) or with empty vector (CA-EV). Anchorage independent growth of stable transfectants was examined as described above. C: Comparison of aromatase enzymatic activity in AC1 and AC1-P cells. AC1 and AC1-P cells were plated in 6-well plates at a density of 1.5 × 105 cells/well. Measurement of aromatase activity assay was performed following the protocol described in the method section.
Figure 5
Figure 5
Bcl-2 upregulation by GP88 in AC1 cells. A: The ability of letrozole to down regulate bcl-2 expression was examined for AC1. AC1 cells were plated at 1.5 × 105 cells/well in 6-well plate and incubated in PRF medium with 5% ChX-FBS. The next day, the medium was removed and fresh PRF-5%ChX-FBS medium was added with the indicated treatments 25 nM androstenedione (AD) or AD with 200 nM letrozole (ADL) alone or in the presence of GP88 (150 ng/ml and 300 ng/ml). After 48 hours, total RNA was extracted with Trizol™. Bcl-2 mRNA expression was determined by RT-PCR and examined by agarose gel electrophoresis and staining by ethidium bromide as described in the method section. GAPDH mRNA expression was examined as internal control for equal loading. B: Bcl-2 and GAPDH band intensities of triplicate experiments were determined by densitometry scanning using a UVP gel densitometer. Relative average intensity of bcl-2 bands from the triplicate experiments in each experimental group were then normalized using GAPDH as an internal standard and expressed as bars ± SD, p < 0.05.
Figure 6
Figure 6
Effect of silencing GP88 expression on cell proliferation and letrozole responsiveness. A: Top panel: Comparison of GP88 expression in MCF-7CA cells (letrozole sensitive) and LTLT cells (letrozole resistant cells). Bottom panel: LTLT-CA cells were treated with either siRNA control or with GP88 siRNA for 48 hours before harvesting the cells to determine GP88 expression. GP88 level was determined by IP-Western analysis and normalized to cell number as described previously (Lu and Serrero, 2001). (B) LTLT-CA cells were transfected with GP88 siRNA or control siRNA according to manufacturer's instructions one day before being plated at 2 × 103 cells/well in 96 well plates in PRF-5%ChX medium and incubated for the time indicated. Cell proliferation was then measured by using the cellTiter-GloR assay using a luminometer (Molecular Devices). (C) LTLT-CA cells were transfected with either GP88 siRNA or Control siRNA as described above. The next day, cells were plated at 2 × 103 cells/well in 96-well plate and incubated in PRF-ChX medium for 24 hrs more. After 24 hrs, the medium was removed and cells were washed with PRF medium twice. Cells were then incubated in serum free fresh medium with the indicated concentration of letrozole for 3 days. After 3 days cells were counted using cellTiter-Glo assay, p < 0.05. D) Increase of GP88 expression in AC1-LetR cells derived from AC1 cells rendered letrozole resistant by long term culture in the presence of 5 nM letrozole. E) Effect of silencing GP88 on proliferation of AC1-LetR cells. AC1-LetR cells were transfected with either control siRNA or GP88 siRNA as described in the method section. Proliferation was measured after 24 and 48 hours exposure by cellTiter-GloR assay, p < 0.05.

References

    1. Brueggemeier RW, Hackett JC, Diaz-Cruz ES. Aromatase inhibitors in the treatment of breast cancer. Endocr Rev. 2005;26:331–45. doi: 10.1210/er.2004-0015.
    1. Grodin JM, Siiteri PK, McDonald PC. Source of estrogen production in postmenopausal women. J Clin Endocrinol Metab. 1973;36:207–14. doi: 10.1210/jcem-36-2-207.
    1. Burak WE Jr, Quinn AL, Farrar WB, Brueggemeier RW. Androgens influence estrogen-induced responses in human breast carcinoma cells through cytochrome P450 aromatase. Breast Cancer Res Treat. 1997;44:57–64. doi: 10.1023/A:1005782311558.
    1. Chen S, Zhou D, Okubo T, Kao YC, Yang C. Breast tumor aromatase: functional role and transcriptional regulation. Endocr Relat Cancer. 1999;6:149–56. doi: 10.1677/erc.0.0060149.
    1. Shupnik MA. Crosstalk between steroid receptors and the c-Src-receptor tyrosine kinase pathways: implications for cell proliferation. Oncogene. 2004;23:7979–89. doi: 10.1038/sj.onc.1208076.
    1. Jordan VC, Brodie AM. Development and evolution of therapies targeted to the estrogen receptor for the treatment and prevention of breast cancer. Steroids. 2007;72:7–25. doi: 10.1016/j.steroids.2006.10.009.
    1. Jelovac D, Macedo L, Goloubeva OG, Handratta V, Brodie AM. Activation of mitogen-activated protein kinase in xenografts and cells during prolonged treatment with aromatase inhibitor letrozole. Cancer Res. 2005;65:5380–9. doi: 10.1158/0008-5472.CAN-04-4502.
    1. Chen S, Masri S, Wang X, Phung S, Yuan YC, Wu X. What do we know about the mechanisms of aromatase inhibitor resistance? J Steroid Biochem Mol Biol. 2006;102:232–40. doi: 10.1016/j.jsbmb.2006.09.012.
    1. Dowsett M, Martin LA, Smith I, Johnston S. Mechanisms of resistance to aromatase inhibitors. J Steroid Biochem Mol Biol. 2005;95:167–172. doi: 10.1016/j.jsbmb.2005.04.022.
    1. Nicholson RI, Gee JM. Oestrogen and growth factor cross-talk and endocrine insensitivity and acquired resistance in breast cancer. Br J Cancer. 2000;82:501–13.
    1. Nicholson RI, Hutcheson IR, Britton D, Knowlden JM, Jones HE, Harper ME, Hiscox SE, Barrow D, Gee JM. Growth factor signalling networks in breast cancer and resistance to endocrine agents: new therapeutic strategies. J Steroid Biochem Mol Biol. 2005;93:257–62. doi: 10.1016/j.jsbmb.2004.12.006.
    1. Nicholson RI, Hutcheson IR, Knowlden JM, Jones HE, Harper ME, Jordan N, Hiscox SE, Barrow D, Gee JM. Nonendocrine pathways and endocrine resistance: observations with antiestrogens and signal transduction inhibitors in combination. Clin Cancer Res. 2004;10:346S–54S. doi: 10.1158/1078-0432.CCR-031206.
    1. Pietras RJ, Arboleda J, Reese DM, Wongvipat N, Pegram MD, Ramos L, Gorman CM, Parker MG, Sliwkowski MX, Slamon DJ. HER-2 tyrosine kinase pathway targets estrogen receptor and promotes hormone-independent growth in human breast cancer cells. Oncogene. 1995;10:2435–46.
    1. Lupu R, Cardillo M, Cho C, Harris L, Hijazi M, Perez C, Rosenberg K, Yang D, Tang C. The significance of heregulin in breast cancer tumor progression and drug resistance. Breast Cancer Res Treat. 1996;38:57–66. doi: 10.1007/BF01803784.
    1. Tang CK, Perez C, Grunt T, Waibel C, Cho C, Lupu R. Involvement of heregulin-beta2 in the acquisition of the hormone-independent phenotype of breast cancer cells. Cancer Res. 1996;56:3350–8.
    1. McClelland RA, Barrow D, Madden TA, Dutkowski CM, Pamment J, Knowlden JM, Gee JM, Nicholson RI. Enhanced epidermal growth factor receptor signaling in MCF7 breast cancer cells after long-term culture in the presence of the pure antiestrogen ICI 182,780 (Faslodex) Endocrinology. 2001;142:2776–88. doi: 10.1210/en.142.7.2776.
    1. Knowlden JM, Hutcheson IR, Jones HE, Madden T, Gee JM, Harper ME, Barrow D, Wakeling AE, Nicholson RI. Elevated levels of epidermal growth factor receptor/c-erbB2 heterodimers mediate an autocrine growth regulatory pathway in tamoxifen-resistant MCF-7 cells. Endocrinology. 2003;144:1032–44. doi: 10.1210/en.2002-220620.
    1. Guvakova MA, Surmacz E. Overexpressed IGF-I receptors reduce estrogen growth requirements, enhance survival, and promote E-cadherin-mediated cell-cell adhesion in human breast cancer cells. Exp Cell Res. 1997;231:149–62. doi: 10.1006/excr.1996.3457.
    1. Stephen RL, Shaw LE, Larsen C, Corcoran D, Darbre PD. Insulin-like growth factor receptor levels are regulated by cell density and by long term estrogen deprivation in MCF7 human breast cancer cells. J Biol Chem. 2001;276:40080–6. doi: 10.1074/jbc.M105892200.
    1. Benz CC, Scott GK, Sarup JC, Johnson RM, Tripathy D, Coronado E, Shepard HM, Osborne CK. Estrogen-dependent, tamoxifen-resistant tumorigenic growth of MCF-7 cells transfected with HER2/neu. Breast Cancer Res Treat. 1992;24:85–95. doi: 10.1007/BF01961241.
    1. Liu Y, el-Ashry D, Chen D, Ding IY, Kern FG. MCF-7 breast cancer cells overexpressing transfected c-erbB-2 have an in vitro growth advantage in estrogen-depleted conditions and reduced estrogen-dependence and tamoxifen-sensitivity in vivo. Breast Cancer Res Treat. 1995;34:97–117. doi: 10.1007/BF00665783.
    1. Kurokawa H, Lenferink AE, Simpson JF, Pisacane PI, Sliwkowski MX, Forbes JT, Arteaga CL. Inhibition of HER2/neu (erbB-2) and mitogen-activated protein kinases enhances tamoxifen action against HER2-overexpressing, tamoxifen-resistant breast cancer cells. Cancer Res. 2000;60:5887–94.
    1. Sabnis GJ, Jelovac D, Long B, Brodie A. The role of growth factor receptor pathways in human breast cancer cells adapted to long-term estrogen deprivation. Cancer Res. 2005;65:3903–10. doi: 10.1158/0008-5472.CAN-04-4092.
    1. Oh AS, Lorant LA, Holloway JN, Miller DL, Kern FG, El-Ashry D. Hyperactivation of MAPK induces loss of ERalpha expression in breast cancer cells. Mol Endocrinol. 2001;15:1344–59. doi: 10.1210/me.15.8.1344.
    1. Su B, Wong C, Hong Y, Chen S. Growth factor signaling enhances aromatase activity of breast cancer cells via post-transcriptional mechanisms. J Steroid Biochem Mol Biol. 2010.
    1. Lu R, Serrero G. Mediation of estrogen mitogenic effect in human breast cancer MCF-7 cells by PC-cell-derived growth factor (PCDGF/granulin precursor) Proc Natl Acad Sci USA. 2001;98:142–7. doi: 10.1073/pnas.011525198.
    1. He Z, Bateman A. Progranulin (granulin-epithelin precursor, PC-cell-derived growth factor, acrogranin) mediates tissue repair and tumorigenesis. J Mol Med. 2003;81:600–12. doi: 10.1007/s00109-003-0474-3.
    1. Ong CH, Bateman A. Progranulin (granulin-epithelin precursor, PC-cell derived growth factor, acrogranin) in proliferation and tumorigenesis. Histol Histopathol. 2003;18:1275–88.
    1. Serrero G. Autocrine growth factor revisited: PC-cell-derived growth factor (progranulin), a critical player in breast cancer tumorigenesis. Biochem Biophys Res Commun. 2003;308:409–13. doi: 10.1016/S0006-291X(03)01452-9.
    1. Lu R, Serrero G. Inhibition of PC cell-derived growth factor (PCDGF, epithelin/granulin precursor) expression by antisense PCDGF cDNA transfection inhibits tumorigenicity of the human breast carcinoma cell line MDA-MB-468. Proc Natl Acad Sci USA. 2000;97:3993–8. doi: 10.1073/pnas.97.8.3993.
    1. Serrero G, Ioffe O. Expression of the novel autocrine growth factor PC-Cell Derived Growth Factor in human breast cancer tissue. Human Pathology. 2003;34:1148–54. doi: 10.1016/S0046-8177(03)00425-8.
    1. Tkaczuk KR, Yue B, Zhan M, Tait N, Yarlagadda L, Dai H, Serrero G. Increased circulating level of the survival factor GP88 (Progranulin) in the serum of breast cancer patients when compared to healthy subjects. Breast Cancer Basic and Clinical Research. 2011. in press .
    1. Tangkeangsirisin W, Hayashi J, Serrero G. PC cell-derived growth factor mediates tamoxifen resistance and promotes tumor growth of human breast cancer cells. Cancer Res. 2004;64:1737–43. doi: 10.1158/0008-5472.CAN-03-2364.
    1. Tangkeangsirisin W, Serrero G. PC-Cell Derived Growth Factor (PCDGF/GP88) Stimulates Migration, Invasiveness and VEGF expression in Breast Cancer Cells. Carcinogenesis. 2004;25:1587–89. doi: 10.1093/carcin/bgh171.
    1. Long BJ, Tilghman SL, Yue W, Thiantanawat A, Grigoryev DN, Brodie AM. The steroidal antiestrogen ICI 182,780 is an inhibitor of cellular aromatase activity. J Steroid Biochem Mol Biol. 1998;67:293–304. doi: 10.1016/S0960-0760(98)00122-8.
    1. Macedo LF, Guo Z, Tilghman SL, Sabnis GJ, Qiu Y, Brodie A. Role of androgens on MCF-7 breast cancer cell growth and on the inhibitory effect of letrozole. Cancer Res. 2006;66:7775–7782. doi: 10.1158/0008-5472.CAN-05-3984.
    1. Diel P, Smolnikar K, Michna H. The pure antiestrogen ICI 182780 is more effective in the induction of apoptosis and down regulation of bcl-2 than tamoxifen in MCF-7 cells. Breast Cancer Res Treat. 1999;58:87–97. doi: 10.1023/A:1006338123126.
    1. Brodie A, Jelovac D, Sabnis G, Long B, Macedo L, Goloubeva O. Model systems: mechanisms involved in the loss of sensitivity to letrozole. J Steroid Biochem Mol Biol. 2005;95:41–8. doi: 10.1016/j.jsbmb.2005.04.026.
    1. Brodie A, Jelovac D, Macedo L, Sabnis G, Tilghman S, Goloubeva O. Therapeutic observations in MCF-7 aromatase xenografts. Clin Cancer Res. 2005;11:884s–8s.
    1. Miller TW, Hennessy BT, González-Angulo AM, Fox EM, Mills GB, Chen H, Higham C, García-Echeverría C, Shyr Y, Arteaga CL. Hyperactivation of Phosphatidylinositol-3-kinase promotes escape frpm hormone dependence in estrogen receptor-positive human breast cancer. J Clin Invest. 2010;120:2406–13. doi: 10.1172/JCI41680.
    1. Elkabets M, Gifford AM, Scheel C, Nilsson B, Reinhardt F, Bray MA, Carpenter AE, Jirström K, Magnusson K, Ebert BL, Pontén F, Weinberg RA, McAllister SS. Human tumors instigate granulin-expressing hematopoietic cells that promote malignancy by activating stromal fibroblasts in mice. J Clin Invest. 2011;121:784–99. doi: 10.1172/JCI43757.
    1. Sabnis G, Schayowitz A, Goloubeva O, Macedo L, Brodie A. Trastuzumab reverses letrozole resistance and amplifies the sensitivity of breast cancer cells to estrogen. Cancer Res. 2009;69:1416–1428. doi: 10.1158/0008-5472.CAN-08-0857.
    1. Kim WE, Serrero G. PC cell-derived growth factor (PCDGF/GP88) stimulates proliferation and confers Trastuzumab resistance to Her-2-overexpressing breast cancer cells. Clin Cancer Res. 2006;15:4192–9.

Source: PubMed

3
Sottoscrivi