p27Kip1 is a cell cycle inhibitor that prevents cyclin dependent kinase (CDK)/cyclin complexes from phosphorylating their targets. p27Kip1 is a known tumor suppressor, as the germline loss of p27Kip1 results in sporadic pituitary formation in aged rodents, and its presence in human cancers is indicative of a poor prognosis. In addition to its role in cancer, loss of p27Kip1 results in regenerative phenotypes in some tissues and maintenance of stem cell pluripotency, suggesting that p27Kip1 inhibitors could be beneficial for tissue regeneration. Because p27Kip1 is an intrinsically disordered protein, identifying direct inhibitors of the p27Kip1 protein is difficult. Therefore, we pursued a high-throughput screening strategy to identify novel p27Kip1 transcriptional inhibitors. We utilized a luciferase reporter plasmid driven by the p27Kip1 promoter to transiently transfect HeLa cells and used cyclohexamide as a positive control for non-specific inhibition. We screened a “bioactive” library consisting of 8,904 (4,359 unique) compounds, of which 830 are Food and Drug Administration (FDA) approved. From this screen, we successfully identified 111 primary hits with inhibitory effect against the promoter of p27Kip1. These hits were further refined using a battery of secondary screens. Here we report four novel p27Kip1 transcriptional inhibitors, and further demonstrate that our most potent hit compound (IC50 = 200 nM) Alsterpaullone 2-cyanoethyl, inhibits p27Kip1 transcription by preventing FoxO3a from binding to the p27Kip1 promoter. This screen represents one of the first attempts to identify inhibitors of p27Kip1 and may prove useful for future tissue regeneration studies.
References
[1]
Sherr CJ, Roberts JM (1995) Inhibitors of mammalian G1 cyclin-dependent kinases. Genes Dev 9: 1149–1163. doi: 10.1101/gad.9.10.1149
[2]
Bertagnolli MM, Warren RS, Niedzwiecki D, Mueller E, Compton CC, et al. (2009) p27Kip1 in stage III colon cancer: implications for outcome following adjuvant chemotherapy in cancer and leukemia group B protocol 89803. Clin Cancer Res 15: 2116–2122. doi: 10.1158/1078-0432.ccr-08-2674
[3]
Bottini C, Platini F, Rinaldi M, Leutner M, Alabiso O, et al. (2009) p27Kip1 is inactivated in human colorectal cancer by cytoplasmic localization associated with activation of Akt/PKB. Int J Oncol 34: 69–77. doi: 10.3892/ijo_00000130
[4]
Chang BL, Zheng SL, Isaacs SD, Wiley KE, Turner A, et al. (2004) A polymorphism in the CDKN1B gene is associated with increased risk of hereditary prostate cancer. Cancer Res 64: 1997–1999. doi: 10.1158/0008-5472.can-03-2340
[5]
Slingerland J, Pagano M (2000) Regulation of the cdk inhibitor p27 and its deregulation in cancer. J Cell Physiol 183: 10–17. doi: 10.1002/(sici)1097-4652(200004)183:1<10::aid-jcp2>3.0.co;2-i
[6]
Pellegata NS, Quintanilla-Martinez L, Siggelkow H, Samson E, Bink K, et al. (2006) Germ-line mutations in p27Kip1 cause a multiple endocrine neoplasia syndrome in rats and humans. Proc Natl Acad Sci U S A 103: 15558–15563. doi: 10.1073/pnas.0603877103
[7]
Chu IM, Hengst L, Slingerland JM (2008) The Cdk inhibitor p27 in human cancer: prognostic potential and relevance to anticancer therapy. Nat Rev Cancer 8: 253–267. doi: 10.1038/nrc2347
[8]
Rabbani F, Koppie TM, Charytonowicz E, Drobnjak M, Bochner BH, et al. (2007) Prognostic significance of p27Kip1 expression in bladder cancer. BJU Int 100: 259–263. doi: 10.1111/j.1464-410x.2007.06927.x
[9]
Menchon C, Edel MJ, Izpisua Belmonte JC (2011) The cell cycle inhibitor p27Kip(1) controls self-renewal and pluripotency of human embryonic stem cells by regulating the cell cycle, Brachyury and Twist. Cell Cycle 10: 1435–1447. doi: 10.4161/cc.10.9.15421
[10]
Zhang S, Huan W, Wei H, Shi J, Fan J, et al. (2013) FOXO3a/p27kip1 expression and essential role after acute spinal cord injury in adult rat. J Cell Biochem 114: 354–365. doi: 10.1002/jcb.24371
[11]
Karnezis AN, Dorokhov M, Grompe M, Zhu L (2001) Loss of p27(Kip1) enhances the transplantation efficiency of hepatocytes transferred into diseased livers. J Clin Invest 108: 383–390. doi: 10.1172/jci200111933
[12]
Liu Z, Walters BJ, Owen T, Brimble MA, Steigelman KA, et al. (2012) Regulation of p27Kip1 by Sox2 maintains quiescence of inner pillar cells in the murine auditory sensory epithelium. J Neurosci 32: 10530–10540. doi: 10.1523/jneurosci.0686-12.2012
[13]
Oesterle EC, Chien WM, Campbell S, Nellimarla P, Fero ML (2011) p27(Kip1) is required to maintain proliferative quiescence in the adult cochlea and pituitary. Cell Cycle 10: 1237–1248. doi: 10.4161/cc.10.8.15301
[14]
White PM, Doetzlhofer A, Lee YS, Groves AK, Segil N (2006) Mammalian cochlear supporting cells can divide and trans-differentiate into hair cells. Nature 441: 984–987. doi: 10.1038/nature04849
[15]
Mantela J, Jiang Z, Ylikoski J, Fritzsch B, Zacksenhaus E, et al. (2005) The retinoblastoma gene pathway regulates the postmitotic state of hair cells of the mouse inner ear. Development 132: 2377–2388. doi: 10.1242/dev.01834
[16]
Chen P, Segil N (1999) p27(Kip1) links cell proliferation to morphogenesis in the developing organ of Corti. Development 126: 1581–1590.
[17]
Lee YS, Liu F, Segil N (2006) A morphogenetic wave of p27Kip1 transcription directs cell cycle exit during organ of Corti development. Development 133: 2817–2826. doi: 10.1242/dev.02453
[18]
Maass JC, Berndt FA, Canovas J, Kukuljan M (2013) p27Kip1 knockdown induces proliferation in the organ of Corti in culture after efficient shRNA lentiviral transduction. J Assoc Res Otolaryngol 14: 495–508. doi: 10.1007/s10162-013-0383-2
[19]
Walters BJ, Zuo J (2013) Postnatal development, maturation and aging in the mouse cochlea and their effects on hair cell regeneration. Hear Res 297: 68–83. doi: 10.1016/j.heares.2012.11.009
[20]
Cheng Y, LeGall T, Oldfield CJ, Mueller JP, Van YY, et al. (2006) Rational drug design via intrinsically disordered protein. Trends Biotechnol 24: 435–442. doi: 10.1016/j.tibtech.2006.07.005
[21]
Denicourt C, Saenz CC, Datnow B, Cui XS, Dowdy SF (2007) Relocalized p27Kip1 tumor suppressor functions as a cytoplasmic metastatic oncogene in melanoma. Cancer Res 67: 9238–9243. doi: 10.1158/0008-5472.can-07-1375
[22]
McAllister SS, Becker-Hapak M, Pintucci G, Pagano M, Dowdy SF (2003) Novel p27(kip1) C-terminal scatter domain mediates Rac-dependent cell migration independent of cell cycle arrest functions. Mol Cell Biol 23: 216–228. doi: 10.1128/mcb.23.1.216-228.2003
[23]
Stahl M, Dijkers PF, Kops GJPL, Lens SMA, Coffer PJ, et al. (2002) The forkhead transcription factor FoxO regulates transcription of p27(Kip1) and bim in response to IL-2. Journal of Immunology 168: 5024–5031. doi: 10.4049/jimmunol.168.10.5024
[24]
Wang C, Hou X, Mohapatra S, Ma Y, Cress WD, et al. (2005) Activation of p27Kip1 Expression by E2F1. A negative feedback mechanism. J Biol Chem 280: 12339–12343. doi: 10.1074/jbc.c400536200
[25]
Wang F, Nguyen M, Qin FX, Tong Q (2007) SIRT2 deacetylates FOXO3a in response to oxidative stress and caloric restriction. Aging Cell 6: 505–514. doi: 10.1111/j.1474-9726.2007.00304.x
[26]
Liu X, Ory V, Chapman S, Yuan H, Albanese C, et al. (2012) ROCK inhibitor and feeder cells induce the conditional reprogramming of epithelial cells. Am J Pathol 180: 599–607. doi: 10.1016/j.ajpath.2011.10.036
[27]
Minami S, Ohtani-Fujita N, Igata E, Tamaki T, Sakai T (1997) Molecular cloning and characterization of the human p27Kip1 gene promoter. FEBS Lett 411: 1–6. doi: 10.1016/s0014-5793(97)00660-1
[28]
Leonardi R, Zhang YM, Yun MK, Zhou R, Zeng FY, et al. (2010) Modulation of pantothenate kinase 3 activity by small molecules that interact with the substrate/allosteric regulatory domain. Chem Biol 17: 892–902. doi: 10.1016/j.chembiol.2010.06.006
[29]
Vrijens K, Lin W, Cui J, Farmer D, Low J, et al. (2013) Identification of small molecule activators of BMP signaling. PLoS One 8: e59045. doi: 10.1371/journal.pone.0059045
[30]
Herbst J, Anthony M, Stewart J, Connors D, Chen T, et al. (2009) Multiplexing a high-throughput liability assay to leverage efficiencies. Assay Drug Dev Technol 7: 294–303. doi: 10.1089/adt.2008.184
[31]
Lin W, Wu J, Dong H, Bouck D, Zeng FY, et al. (2008) Cyclin-dependent kinase 2 negatively regulates human pregnane X receptor-mediated CYP3A4 gene expression in HepG2 liver carcinoma cells. J Biol Chem 283: 30650–30657. doi: 10.1074/jbc.m806132200
[32]
Trapp J, Jochum A, Meier R, Saunders L, Marshall B, et al. (2006) Adenosine mimetics as inhibitors of NAD+-dependent histone deacetylases, from kinase to sirtuin inhibition. J Med Chem 49: 7307–7316. doi: 10.1021/jm060118b
[33]
Minoda R, Izumikawa M, Kawamoto K, Zhang H, Raphael Y (2007) Manipulating cell cycle regulation in the mature cochlea. Hear Res 232: 44–51. doi: 10.1016/j.heares.2007.06.005
[34]
Zaharevitz DW, Gussio R, Leost M, Senderowicz AM, Lahusen T, et al. (1999) Discovery and initial characterization of the paullones, a novel class of small-molecule inhibitors of cyclin-dependent kinases. Cancer Res 59: 2566–2569.
[35]
Duronio RJ, Xiong Y (2013) Signaling pathways that control cell proliferation. Cold Spring Harb Perspect Biol 5: a008904. doi: 10.1101/cshperspect.a008904
[36]
Tolle N, Kunick C (2011) Paullones as inhibitors of protein kinases. Curr Top Med Chem 11: 1320–1332. doi: 10.2174/156802611795589601
[37]
Lyssiotis CA, Foreman RK, Staerk J, Garcia M, Mathur D, et al. (2009) Reprogramming of murine fibroblasts to induced pluripotent stem cells with chemical complementation of Klf4. Proc Natl Acad Sci U S A 106: 8912–8917. doi: 10.1073/pnas.0903860106
[38]
Motta MC, Divecha N, Lemieux M, Kamel C, Chen D, et al. (2004) Mammalian SIRT1 represses forkhead transcription factors. Cell 116: 551–563. doi: 10.1016/s0092-8674(04)00126-6