The anti-tumor activities of Neferine on cell invasion and oxaliplatin sensitivity regulated by EMT via Snail signaling in hepatocellular carcinoma

Scientific Reports - Tập 7 Số 1
Geng Deng1, Shan Zeng1, Junli Ma2, Yan Zhang1, Yanling Qu1, Ying Han1, Ling Yin1, Changjing Cai2, Cao Guo1, Hong Shen2
1Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
2Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China

Tóm tắt

AbstractTumor invasion and chemotherapy resistance, which are associated with epithelial-mesenchymal transition (EMT), remain as major challenges in hepatocellular carcinoma (HCC) treatment. Neferine, a natural component of Nelumbo nucifera, have been proven the antitumor efficiency in cancer, but the effects of Neferine on HCC invasion and chemosensitivity need to be elucidated. Applying multiple assays of cell proliferation, flow cytometry, immunofluorescence staining, qRT-PCR, Western blot, fluorescence molecular tomography imaging, the influences of Neferine on EMT-regulated viability, apoptosis, invasion, and oxaliplatin (OXA) sensitivity were assessed in HCC cells of HepG2 and Bel-7402, as well as in xenograft animal models in vivo. Here, we reported that Neferine had no obvious effects on HCC cells proliferation, but significantly enhanced cytotoxicity and apoptosis caused by OXA in vitro and in vivo. Through an upregulation of E-cadherin and downregulation of Vimentin, Snail and N-cadherin, Neferine suppressed EMT-induced migration and invasion abilities of HCC cells. TGF-β1 cancelled the effects of Neferine on the migration and invasion of HCC cells. Snail overexpression or TGF-β1-induced EMT attenuated Neferine-mediated OXA sensitization in HCC. Together, our data suggest that Neferine enhances oxaliplatin sensitivity through an inhibition of EMT in HCC cells. Neferine may be used as an OXA sensitizer in HCC chemotherapy.

Từ khóa


Tài liệu tham khảo

Siegel, R., Naishadham, D. & Jemal, A. Cancer statistics, 2013. CA Cancer J Clin 63, 11–30, doi: 10.3322/caac.21166 (2013).

Choi, S. S. & Diehl, A. M. Epithelial-to-mesenchymal transitions in the liver. Hepatology 50, 2007–2013, doi: 10.1002/hep.23196 (2009).

Tao, Y. M. et al. BTB/POZ domain-containing protein 7: epithelial-mesenchymal transition promoter and prognostic biomarker of hepatocellular carcinoma. Hepatology 57, 2326–2337, doi: 10.1002/hep.26268 (2013).

Lu, P., Weaver, V. M. & Werb, Z. The extracellular matrix: a dynamic niche in cancer progression. J Cell Biol 196, 395–406, doi: 10.1083/jcb.201102147 (2012).

Thiery, J. P. Epithelial-mesenchymal transitions in development and pathologies. Curr Opin Cell Biol 15, 740–746, doi: S0955067403001339 (2003).

Fischer, K. R. et al. Epithelial-to-mesenchymal transition is not required for lung metastasis but contributes to chemoresistance. Nature 527, 472–476, doi: 10.1038/nature15748 (2015).

Zheng, X. et al. Epithelial-to-mesenchymal transition is dispensable for metastasis but induces chemoresistance in pancreatic cancer. Nature 527, 525–530, doi: 10.1038/nature16064 (2015).

Ma, J. L., Zeng, S., Zhang, Y., Deng, G. L. & Shen, H. Epithelial-mesenchymal transition plays a critical role in drug resistance of hepatocellular carcinoma cells to oxaliplatin. Tumour Biol 37, 6177–6184, doi: 10.1007/s13277-015-4458-z (2016).

Liu, S., Wang, B., Li, X. Z., Qi, L. F. & Liang, Y. Z. Preparative separation and purification of liensinine, isoliensinine and neferine from seed embryo of Nelumbo nucifera GAERTN using high-speed counter-current chromatography. J Sep Sci 32, 2476–2481, doi: 10.1002/jssc.200800766 (2009).

Qian, J. Q. Cardiovascular pharmacological effects of bisbenzylisoquinoline alkaloid derivatives. Acta Pharmacol Sin 23, 1086–1092 (2002).

Yu, J. & Hu, W. S. [Effects of neferine on platelet aggregation in rabbits]. Yao Xue Xue Bao 32, 1–4 (1997).

Xiong, Y. Q. & Zeng, F. D. Effect of neferine on toxicodynamics of dichlorvos for inhibiting rabbit cholinesterase. Acta Pharmacol Sin 24, 332–336 (2003).

Pan, Y. et al. Neferine enhances insulin sensitivity in insulin resistant rats. J Ethnopharmacol 124, 98–102, doi: 10.1016/j.jep.2009.04.008 (2009).

Cao, J. G., Tang, X. Q. & Shi, S. H. Multidrug resistance reversal in human gastric carcinoma cells by neferine. World J Gastroenterol 10, 3062–3064 (2004).

Poornima, P., Weng, C. F. & Padma, V. V. Neferine from Nelumbo nucifera induces autophagy through the inhibition of PI3K/Akt/mTOR pathway and ROS hyper generation in A549 cells. Food Chem 141, 3598–3605, doi: 10.1016/j.foodchem.2013.05.138 (2013).

Poornima, P., Weng, C. F. & Padma, V. V. Neferine, an alkaloid from lotus seed embryo, inhibits human lung cancer cell growth by MAPK activation and cell cycle arrest. Biofactors 40, 121–131, doi: 10.1002/biof.1115 (2014).

Poornima, P., Kumar, V. B., Weng, C. F. & Padma, V. V. Doxorubicin induced apoptosis was potentiated by neferine in human lung adenocarcima, A549 cells. Food Chem Toxicol 68, 87–98, doi: 10.1016/j.fct.2014.03.008 (2014).

Poornima, P., Quency, R. S. & Padma, V. V. Neferine induces reactive oxygen species mediated intrinsic pathway of apoptosis in HepG2 cells. Food Chem 136, 659–667, doi: 10.1016/j.foodchem.2012.07.112 (2013).

Yoon, J. S. et al. Neferine isolated from Nelumbo nucifera enhances anti-cancer activities in Hep3B cells: molecular mechanisms of cell cycle arrest, ER stress induced apoptosis and anti-angiogenic response. Phytomedicine 20, 1013–1022, doi: 10.1016/j.phymed.2013.03.024 (2013).

Zhang, X. et al. Neferine, an alkaloid ingredient in lotus seed embryo, inhibits proliferation of human osteosarcoma cells by promoting p38 MAPK-mediated p21 stabilization. Eur J Pharmacol 677, 47–54, doi: 10.1016/j.ejphar.2011.12.035 (2012).

Xu, L. et al. Neferine induces autophagy of human ovarian cancer cells via p38 MAPK/JNK activation. Tumour Biol, doi: 10.1007/s13277-015-4737-8 (2016).

Ding, W. et al. Epithelial-to-mesenchymal transition of murine liver tumor cells promotes invasion. Hepatology 52, 945–953, doi: 10.1002/hep.23748 (2010).

Nieto, M. A. The ins and outs of the epithelial to mesenchymal transition in health and disease. Annu Rev Cell Dev Biol 27, 347–376, doi: 10.1146/annurev-cellbio-092910-154036 (2011).

Gavert, N. & Ben-Ze’ev, A. Epithelial-mesenchymal transition and the invasive potential of tumors. Trends Mol Med 14, 199–209, doi: 10.1016/j.molmed.2008.03.004 (2008).

Piedra, M. E. & Ros, M. A. BMP signaling positively regulates Nodal expression during left right specification in the chick embryo. Development 129, 3431–3440 (2002).

Eckert, M. A. et al. Twist1-induced invadopodia formation promotes tumor metastasis. Cancer Cell 19, 372–386, doi: 10.1016/j.ccr.2011.01.036 (2011).

Linder, S. The matrix corroded: podosomes and invadopodia in extracellular matrix degradation. Trends Cell Biol 17, 107–117, doi: 10.1016/j.tcb.2007.01.002 (2007).

Egeblad, M. & Werb, Z. New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer 2, 161–174, doi: 10.1038/nrc745 (2002).

Mejlvang, J. et al. Direct repression of cyclin D1 by SIP1 attenuates cell cycle progression in cells undergoing an epithelial mesenchymal transition. Mol Biol Cell 18, 4615–4624, doi: 10.1091/mbc.E07-05-0406 (2007).

Singh, A. & Settleman, J. EMT, cancer stem cells and drug resistance: an emerging axis of evil in the war on cancer. Oncogene 29, 4741–4751, doi: 10.1038/onc.2010.215 (2010).

Vega, S. et al. Snail blocks the cell cycle and confers resistance to cell death. Genes Dev 18, 1131–1143, doi: 10.1101/gad.294104 (2004).

Ramljak, D. et al. Epidermal growth factor inhibition of c-Myc-mediated apoptosis through Akt and Erk involves Bcl-xL upregulation in mammary epithelial cells. Exp Cell Res 287, 397–410, doi: S0014482703001356 (2003).

Witta, S. E. et al. Restoring E-cadherin expression increases sensitivity to epidermal growth factor receptor inhibitors in lung cancer cell lines. Cancer Res 66, 944–950, doi: 10.1158/0008-5472.CAN-05-1988 (2006).

Franco, D. L. et al. Snail1 suppresses TGF-beta-induced apoptosis and is sufficient to trigger EMT in hepatocytes. J Cell Sci 123, 3467–3477, doi: 10.1242/jcs.068692 (2010).

Mani, S. A. et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell 133, 704–715, doi: 10.1016/j.cell.2008.03.027 (2008).

Polyak, K. & Weinberg, R. A. Transitions between epithelial and mesenchymal states: acquisition of malignant and stem cell traits. Nat Rev Cancer 9, 265–273, doi: 10.1038/nrc2620 (2009).

Hermann, P. C. et al. Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer. Cell Stem Cell 1, 313–323, doi: 10.1016/j.stem.2007.06.002 (2007).

Li, X. et al. Intrinsic resistance of tumorigenic breast cancer cells to chemotherapy. J Natl Cancer Inst 100, 672–679, doi: 10.1093/jnci/djn123 (2008).

Kurrey, N. K. et al. Snail and slug mediate radioresistance and chemoresistance by antagonizing p53-mediated apoptosis and acquiring a stem-like phenotype in ovarian cancer cells. Stem Cells 27, 2059–2068, doi: 10.1002/stem.154 (2009).

Zhou, J. et al. Activation of the PTEN/mTOR/STAT3 pathway in breast cancer stem-like cells is required for viability and maintenance. Proc Natl Acad Sci USA. 104, 16158–16163, doi: 10.1073/pnas.0702596104 (2007).

Dubrovska, A. et al. The role of PTEN/Akt/PI3K signaling in the maintenance and viability of prostate cancer stem-like cell populations. Proc Natl Acad Sci USA. 106, 268–273, doi: 10.1073/pnas.0810956106 (2009).

Gupta, P. B. et al. Identification of selective inhibitors of cancer stem cells by high-throughput screening. Cell 138, 645–659, doi: 10.1016/j.cell.2009.06.034 (2009).

Tallarida, R. J. Drug synergism: its detection and applications. J Pharmacol Exp Ther 298, 865–872 (2001).