Embelin Suppresses Growth of Human Pancreatic Cancer Xenografts, and Pancreatic Cancer Cells Isolated from KrasG12D Mice by Inhibiting Akt and Sonic Hedgehog Pathways
Pancreatic cancer is a deadly disease, and therefore effective treatment and/or prevention strategies are urgently needed. The objectives of this study were to examine the molecular mechanisms by which embelin inhibited human pancreatic cancer cell growth in vitro, and xenografts in Balb C nude mice, and pancreatic cancer cell growth isolated from KrasG12D transgenic mice. XTT assays were performed to measure cell viability. AsPC-1 cells were injected subcutaneously into Balb c nude mice and treated with embelin. Cell proliferation and apoptosis were measured by Ki67 and TUNEL staining, respectively. The expression of Akt, and Sonic Hedgehog (Shh) and their target gene products were measured by the immunohistochemistry, and Western blot analysis. The effects of embelin on pancreatic cancer cells isolated from 10-months old KrasG12D mice were also examined. Embelin inhibited cell viability in pancreatic cancer AsPC-1, PANC-1, MIA PaCa-2 and Hs 766T cell lines, and these inhibitory effects were blocked either by constitutively active Akt or Shh protein. Embelin-treated mice showed significant inhibition in tumor growth which was associated with reduced expression of markers of cell proliferation (Ki67, PCNA and Bcl-2) and cell cycle (cyclin D1, CDK2, and CDK6), and induction of apoptosis (activation of caspase-3 and cleavage of PARP, and increased expression of Bax). In addition, embelin inhibited the expression of markers of angiogenesis (COX-2, VEGF, VEGFR, and IL-8), and metastasis (MMP-2 and MMP-9) in tumor tissues. Antitumor activity of embelin was associated with inhibition of Akt and Shh pathways in xenografts, and pancreatic cancer cells isolated from KrasG12D mice. Furthermore, embelin also inhibited epithelial-to-mesenchymal transition (EMT) by up-regulating E-cadherin and inhibiting the expression of Snail, Slug, and ZEB1. These data suggest that embelin can inhibit pancreatic cancer growth, angiogenesis and metastasis by suppressing Akt and Shh pathways, and can be developed for the treatment and/or prevention of pancreatic cancer.
References
[1]
Siegel R, Naishadham D, Jemal A (2013) Cancer statistics, 2013. CA Cancer J Clin 63: 11–30. doi: 10.3322/caac.21166
[2]
Segura PP, Ponce CG, Ramon YCT, Blanch RS, Aranda E (2012) Hereditary pancreatic cancer: molecular bases and their application in diagnosis and clinical management: a guideline of the TTD group. Clin Transl Oncol 14: 553–563. doi: 10.1007/s12094-012-0840-0
[3]
Magee CJ, Ghaneh P, Neoptolemos JP (2002) Surgical and medical therapy for pancreatic carcinoma. Best Pract Res Clin Gastroenterol 16: 435–455. doi: 10.1053/bega.2002.0317
[4]
Li D (2001) Molecular epidemiology of pancreatic cancer. Cancer J 7: 259–265. doi: 10.1007/978-1-4614-6549-2_3
[5]
Gold EB, Goldin SB (1998) Epidemiology of and risk factors for pancreatic cancer. Surg Oncol Clin N Am 7: 67–91.
[6]
Jaffee EM, Hruban RH, Canto M, Kern SE (2002) Focus on pancreas cancer. Cancer Cell 2: 25–28. doi: 10.1016/s1535-6108(02)00093-4
[7]
Wang Z, Li Y, Ahmad A, Banerjee S, Azmi AS, et al. (2011) Pancreatic cancer: understanding and overcoming chemoresistance. Nat Rev Gastroenterol Hepatol 8: 27–33. doi: 10.1038/nrgastro.2010.188
[8]
Li J, Wientjes MG, Au JL (2010) Pancreatic cancer: pathobiology, treatment options, and drug delivery. AAPS J 12: 223–232. doi: 10.1208/s12248-010-9181-5
[9]
Hu R, Zhu K, Li Y, Yao K, Zhang R, et al. (2011) Embelin induces apoptosis through down-regulation of XIAP in human leukemia cells. Med Oncol 28: 1584–1588. doi: 10.1007/s12032-010-9601-5
[10]
Nikolovska-Coleska Z, Xu L, Hu Z, Tomita Y, Li P, et al. (2004) Discovery of embelin as a cell-permeable, small-molecular weight inhibitor of XIAP through structure-based computational screening of a traditional herbal medicine three-dimensional structure database. J Med Chem 47: 2430–2440. doi: 10.1021/jm030420+
[11]
Xu M, Cui J, Fu H, Proksch P, Lin W, et al. (2005) Embelin derivatives and their anticancer activity through microtubule disassembly. Planta Med 71: 944–948. doi: 10.1055/s-2005-871250
[12]
Huang Y, Lu J, Gao X, Li J, Zhao W, et al. (2012) PEG-derivatized embelin as a dual functional carrier for the delivery of paclitaxel. Bioconjug Chem 23: 1443–1451. doi: 10.1021/bc3000468
[13]
Kim SW, Kim SM, Bae H, Nam D, Lee JH, et al. (2013) Embelin inhibits growth and induces apoptosis through the suppression of Akt/mTOR/S6K1 signaling cascades. Prostate 73: 296–305. doi: 10.1002/pros.22574
[14]
Marsh JL, Jackman CP, Tang SN, Shankar S, Srivastava RK (2014) Embelin suppresses pancreatic cancer growth by modulating tumor immune microenvironment. Front Biosci (Landmark Ed) 19: 113–125. doi: 10.2741/4198
[15]
Heo JY, Kim HJ, Kim SM, Park KR, Park SY, et al. (2011) Embelin suppresses STAT3 signaling, proliferation, and survival of multiple myeloma via the protein tyrosine phosphatase PTEN. Cancer Lett 308: 71–80. doi: 10.1016/j.canlet.2011.04.015
[16]
Siegelin MD, Gaiser T, Siegelin Y (2009) The XIAP inhibitor Embelin enhances TRAIL-mediated apoptosis in malignant glioma cells by down-regulation of the short isoform of FLIP. Neurochem Int 55: 423–430. doi: 10.1016/j.neuint.2009.04.011
[17]
Kennedy AL, Adams PD, Morton JP (2011) Ras, PI3K/Akt and senescence: Paradoxes provide clues for pancreatic cancer therapy. Small GTPases 2: 264–267. doi: 10.4161/sgtp.2.5.17367
[18]
Hill R, Calvopina JH, Kim C, Wang Y, Dawson DW, et al. (2010) PTEN loss accelerates KrasG12D-induced pancreatic cancer development. Cancer Res 70: 7114–7124. doi: 10.1158/0008-5472.can-10-1649
[19]
Shankar S, Nall D, Tang SN, Meeker D, Passarini J, et al. (2011) Resveratrol inhibits pancreatic cancer stem cell characteristics in human and KrasG12D transgenic mice by inhibiting pluripotency maintaining factors and epithelial-mesenchymal transition. PLoS One 6: e16530. doi: 10.1371/journal.pone.0016530
[20]
Saqui-Salces M, Merchant JL (2010) Hedgehog signaling and gastrointestinal cancer. Biochim Biophys Acta 1803: 786–795. doi: 10.1016/j.bbamcr.2010.03.008
[21]
Varjosalo M, Taipale J (2008) Hedgehog: functions and mechanisms. Genes Dev 22: 2454–2472. doi: 10.1101/gad.1693608
[22]
Rohatgi R, Scott MP (2007) Patching the gaps in Hedgehog signalling. Nat Cell Biol 9: 1005–1009. doi: 10.1038/ncb435
[23]
Kinzler KW, Ruppert JM, Bigner SH, Vogelstein B (1988) The GLI gene is a member of the Kruppel family of zinc finger proteins. Nature 332: 371–374. doi: 10.1038/332371a0
[24]
Choy SW, Cheng SH (2012) Hedgehog signaling. Vitam Horm 88: 1–23. doi: 10.1016/b978-0-12-394622-5.00001-8
[25]
Fu J, Rodova M, Roy SK, Sharma J, Singh KP, et al. (2013) GANT-61 inhibits pancreatic cancer stem cell growth in vitro and in NOD/SCID/IL2R gamma null mice xenograft. Cancer Lett 330: 22–32. doi: 10.1016/j.canlet.2012.11.018
[26]
Li SH, Fu J, Watkins DN, Srivastava RK, Shankar S (2013) Sulforaphane regulates self-renewal of pancreatic cancer stem cells through the modulation of Sonic hedgehog-GLI pathway. Mol Cell Biochem 373: 217–227. doi: 10.1007/s11010-012-1493-6
[27]
Rodova M, Fu J, Watkins DN, Srivastava RK, Shankar S (2012) Sonic hedgehog signaling inhibition provides opportunities for targeted therapy by sulforaphane in regulating pancreatic cancer stem cell self-renewal. PLoS One 7: e46083. doi: 10.1371/journal.pone.0046083
[28]
Roy SK, Srivastava RK, Shankar S (2010) Inhibition of PI3K/AKT and MAPK/ERK pathways causes activation of FOXO transcription factor, leading to cell cycle arrest and apoptosis in pancreatic cancer. J Mol Signal 5: 10. doi: 10.1186/1750-2187-5-10
[29]
Singh BN, Fu J, Srivastava RK, Shankar S (2011) Hedgehog signaling antagonist GDC-0449 (Vismodegib) inhibits pancreatic cancer stem cell characteristics: molecular mechanisms. PLoS One 6: e27306. doi: 10.1371/journal.pone.0027306
[30]
Singh BN, Kumar D, Shankar S, Srivastava RK (2012) Rottlerin induces autophagy which leads to apoptotic cell death through inhibition of PI3K/Akt/mTOR pathway in human pancreatic cancer stem cells. Biochem Pharmacol 84: 1154–1163. doi: 10.1016/j.bcp.2012.08.007
[31]
Zhao M, Tang SN, Marsh JL, Shankar S, Srivastava RK (2013) Ellagic acid inhibits human pancreatic cancer growth in Balb c nude mice. Cancer Lett 337: 210–217. doi: 10.1016/j.canlet.2013.05.009
[32]
Basanez G, Soane L, Hardwick JM (2012) A new view of the lethal apoptotic pore. PLoS Biol 10: e1001399. doi: 10.1371/journal.pbio.1001399
[33]
Harris MH, Thompson CB (2000) The role of the Bcl-2 family in the regulation of outer mitochondrial membrane permeability. Cell Death Differ 7: 1182–1191. doi: 10.1038/sj.cdd.4400781
[34]
Vander Heiden MG, Thompson CB (1999) Bcl-2 proteins: regulators of apoptosis or of mitochondrial homeostasis? Nat Cell Biol 1: E209–216. doi: 10.1038/70237
[35]
Harada H, Grant S (2003) Apoptosis regulators. Rev Clin Exp Hematol 7: 117–138.
[36]
Maitra A, Hruban RH (2005) A new mouse model of pancreatic cancer: PTEN gets its Akt together. Cancer Cell 8: 171–172. doi: 10.1016/j.ccr.2005.08.007
[37]
Folkman J (2003) Angiogenesis and proteins of the hemostatic system. J Thromb Haemost 1: 1681–1682. doi: 10.1046/j.1538-7836.2003.00344.x
[38]
Hill R, Li Y, Tran LM, Dry S, Calvopina JH, et al. (2012) Cell intrinsic role of COX-2 in pancreatic cancer development. Mol Cancer Ther 11: 2127–2137. doi: 10.1158/1535-7163.mct-12-0342
[39]
Palena C, Hamilton DH, Fernando RI (2012) Influence of IL-8 on the epithelial-mesenchymal transition and the tumor microenvironment. Future Oncol 8: 713–722. doi: 10.2217/fon.12.59
[40]
Steeg PS (2006) Tumor metastasis: mechanistic insights and clinical challenges. Nat Med 12: 895–904. doi: 10.1038/nm1469
[41]
Kraljevic Pavelic S, Sedic M, Bosnjak H, Spaventi S, Pavelic K (2011) Metastasis: new perspectives on an old problem. Mol Cancer 10: 22. doi: 10.1186/1476-4598-10-22
[42]
Jenkins D (2009) Hedgehog signalling: emerging evidence for non-canonical pathways. Cell Signal 21: 1023–1034. doi: 10.1016/j.cellsig.2009.01.033
[43]
Shankar S, Singh TR, Fandy TE, Luetrakul T, Ross DD, et al. (2005) Interactive effects of histone deacetylase inhibitors and TRAIL on apoptosis in human leukemia cells: Involvement of both death receptor and mitochondrial pathways. Int J Mol Med 16: 1125–1138. doi: 10.3892/ijmm.16.6.1125
[44]
Srivastava RK (2000) Intracellular mechanisms of TRAIL and its role in cancer therapy. Mol Cell Biol Res Commun 4: 67–75. doi: 10.1006/mcbr.2001.0265
[45]
Srivastava RK (2001) TRAIL/Apo-2L: mechanisms and clinical applications in cancer. Neoplasia 3: 535–546. doi: 10.1038/sj.neo.7900203
[46]
Thayer SP, di Magliano MP, Heiser PW, Nielsen CM, Roberts DJ, et al. (2003) Hedgehog is an early and late mediator of pancreatic cancer tumorigenesis. Nature 425: 851–856. doi: 10.1038/nature02009
[47]
Tang SN, Singh C, Nall D, Meeker D, Shankar S, et al. (2010) The dietary bioflavonoid quercetin synergizes with epigallocathechin gallate (EGCG) to inhibit prostate cancer stem cell characteristics, invasion, migration and epithelial-mesenchymal transition. J Mol Signal 5: 14. doi: 10.1186/1750-2187-5-14
[48]
Hao K, Tian XD, Qin CF, Xie XH, Yang YM (2013) Hedgehog signaling pathway regulates human pancreatic cancer cell proliferation and metastasis. Oncol Rep 29: 1124–1132. doi: 10.3892/or.2012.2210
[49]
Srivastava RK, Tang SN, Zhu W, Meeker D, Shankar S (2011) Sulforaphane synergizes with quercetin to inhibit self-renewal capacity of pancreatic cancer stem cells. Front Biosci (Elite Ed) 3: 515–528. doi: 10.2741/e266
[50]
Tang SN, Fu J, Nall D, Rodova M, Shankar S, et al. (2012) Inhibition of sonic hedgehog pathway and pluripotency maintaining factors regulate human pancreatic cancer stem cell characteristics. Int J Cancer 131: 30–40. doi: 10.1002/ijc.26323
[51]
Fernando RI, Castillo MD, Litzinger M, Hamilton DH, Palena C (2011) IL-8 signaling plays a critical role in the epithelial-mesenchymal transition of human carcinoma cells. Cancer Res 71: 5296–5306. doi: 10.1158/0008-5472.can-11-0156
[52]
Hingorani SR, Petricoin EF, Maitra A, Rajapakse V, King C, et al. (2003) Preinvasive and invasive ductal pancreatic cancer and its early detection in the mouse. Cancer Cell 4: 437–450. doi: 10.1016/s1535-6108(03)00309-x