Anticancer effects and underlying mechanism of Colchicine on human gastric cancer cell lines in vitro and in vivo

Tao Zhang, Wei Chen, Xumian Jiang, Lei Liu, Kai Wei, Hansong Du, Hui Wang, Juan Li, Tao Zhang, Wei Chen, Xumian Jiang, Lei Liu, Kai Wei, Hansong Du, Hui Wang, Juan Li

Abstract

The present study investigated the effects of Colchicine on gastric carcinoma (GC) cells and explored its possible mechanisms underlying such effects. The results of MTT and colony formation assays showed that Colchicine (2, 5, and 10 ng/ml) markedly inhibited the proliferation of AGS and NCI-N87 cells in a dose-dependent manner. It also led to a reduction in cell migration in both GC cells as determined by Transwell migration assay. Mover, data form Hoechst 33342 staining and flow cytometry assay indicated that Colchicine (2, 5, and 10 ng/ml) promoted the apoptosis of NCI-N87 cells. In addition, the release of cytochrome c, the activation of bax, and the inhibition of bcl-2 were observed in NCI-N87 cells treated with Colchicine. Furthermore, the in vivo experiment further confirmed that Colchicine administration remarkably suppressed the tumor growth in nude mice via induction of apoptosis at 0.05 and 0.1 mg/kg. In addition, no visible toxicity was observed in liver and renal tissue of mice. This finding suggests that Colchicine-induced apoptosis is associated with caspase-3-mediated mitochondrial apoptotic pathways.

Keywords: Colchicine; apoptosis; caspase-3; gastric cancer; mitochondrial apoptotic pathways.

Conflict of interest statement

The authors declare that there are no competing interests associated with the manuscript.

© 2019 The Author(s).

Figures

Figure 1. The structure of Colchicine
Figure 1. The structure of Colchicine
Figure 2. The effect of Colchicine on…
Figure 2. The effect of Colchicine on cell growth and migration
(A) Effect of Colchicine on the cell viability of human GC AGS and NCI-N87 cells at 48 h. (B) Effect of Colchicine on colony formation of human GC AGS and NCI-N87 cells on day 12. (C) Effect of Colchicine on the migration of human GC AGS and NCI-N87 cells at 48 h. (D) Quantitative analysis of the percentage of migrated cells after treatment with Colchicine at 48 h. Data are means ± S.D. (n=5). **P<0.01, ***P<0.001, compared with control of AGS, ##P<0.01, ###P<0.001, compared with control of NCI-N87.
Figure 3. The effect of Colchicine on…
Figure 3. The effect of Colchicine on cell apoptosis
(A) Morphological analysis of the nuclei of NCI-N87 cells stained by Hoechst 33342 after various concentrations of Colchicine treatment for 48 h (200×). (B) Quantitative analysis of the percentage of apoptotic NCI-N87 cells stained by Hoechst 33342 after various concentrations of Colchicine treatment for 48 h. (C) Flow cytometry analysis of NCI-N87 cells after various concentrations of Colchicine treatment for 48 h. (D) The percentage of apoptotic cells in each group after various concentrations of Colchicine treatment for 48 h. Data are means ± S.D. (n=5). **P<0.01, ***P<0.001, compared with untreated control.
Figure 4. The effect of Colchicine on…
Figure 4. The effect of Colchicine on apoptotic protein expression
(A) The protein expression of bax, bcl-2, cleaved-caspase-3, and cytochrome c in NCI-N87 cells after various concentrations of Colchicine treatment for 48 h. (B) The protein expression of PI3K, p-PI3K, Akt, p-Akt, mTOR, and p-mTOR in NCI-N87 cells after various concentrations of Colchicine treatment for 48 h. Data are means ± S.D. (n=5). *P<0.05, **P<0.01, ***P<0.001, compared with untreated control.
Figure 5. The effect of Colchicine on…
Figure 5. The effect of Colchicine on tumor growth and apoptosis in vivo
(A) The antitumor effects of Colchicine on TV of xenograft model of NCI-N87 cells. (B) The antitumor effects of Colchicine on tumor weight of xenograft model of NCI-N87 cells. (C) Apoptotic cells were detected in xenograft tumor tissue using the TUNEL assay (200×). (D) Quantitated results of the TUNEL assay. Data are means ± S.D. (n=10). *P<0.05, **P<0.01, compared with untreated control.

References

    1. Siegel R., Naishadham D. and Jemal A. (2012) Cancer statistics for hispanics/latinos, 2012. CA Cancer J. Clin. 62, 283–298 10.3322/caac.21153
    1. Hartgrink H.H., Jansen E.P.M., van Grieken N.C.T. and van de Velde C.J. (2009) Gastric cancer. Lancet North Am. Ed. 374, 477–490 10.1016/S0140-6736(09)60617-6
    1. Cervantes A., Roselló S., Roda D. and Rodríguez-Braun E. (2008) The treatment of advanced gastric cancer: current strategies and future perspectives. Ann. Oncol. 19, 103–107 10.1093/annonc/mdn321
    1. Luo H.Y., Wei W., Shi Y.X.. et al. (2010) Cetuximab enhances the effect of oxaliplatin on hypoxic gastric cancer cell lines. Oncol. Rep. 23, 1735–1745
    1. Monsuez J.J., Charniot J.C., Vignat N. and Artigou J.Y. (2010) Cardiac side-effects of cancer chemotherapy. Int. J. Cardiol. 144, 3–15 10.1016/j.ijcard.2010.03.003
    1. D’Angelica M., Gonen M., Brennan M.F., Turnbull A.D., Bains M. and Karpeh M.S. (2004) Patterns of initial recurrence in completely resected gastric adenocarcinoma. Ann. Surg. 240, 808–816 10.1097/01.sla.0000143245.28656.15
    1. Bhat T.A. and Singh R.P. (2008) Tumor angiogenesis-a potential target in cancer chemoprevention. Food Chem. Toxicol. 46, 1334–1345 10.1016/j.fct.2007.08.032
    1. Dudkiewicz I., Brosh T., Perelman M. and Salai M. (2005) Colchicine inhibits fracture union and reduces bone strength—in vivo study. J. Orthop. Res. 23, 877–881 10.1016/j.orthres.2004.11.014
    1. Kaplan M.M., Alling D.W., Zimmerman H.J.. et al. (1986) A prospective trial of colchicine for primary biliary cirrhosis. N. Engl. J. Med. 315, 1448–1454 10.1056/NEJM198612043152304
    1. Lin Z.Y., Kuo C.H., Wu D.C. and Chuang W. (2016) Anticancer effects of clinically acceptable colchicine concentrations on human gastric cancer cell lines. Kaohsiung J. Med. Sci. 32, 68–73 10.1016/j.kjms.2015.12.006
    1. Liu H., Zhu Y., Zhang T.. et al. (2013) Anti-tumor effects of atractylenolide I isolated from Atractylodes macrocephala in human lung carcinoma cell lines. Molecules 18, 13357–13368 10.3390/molecules181113357
    1. Ma Y.S., Weng S.W., Lin M.W.. et al. (2012) Antitumor effects of emodin on LS1034 human colon cancer cells in vitro and in vivo: roles of apoptotic cell death and LS1034 tumor xenografts model. Food Chem. Toxicol. 50, 1271–1278 10.1016/j.fct.2012.01.033
    1. Zhu X., Li Z., Li T.. et al. (2018) Osthole inhibits the PI3K/AKT signaling pathway via activation of PTEN and induces cell cycle arrest and apoptosis in esophageal squamous cell carcinoma. Biomed. Pharmacother. 102, 502–509 10.1016/j.biopha.2018.03.106
    1. Maryam A., Mehmood T., Yan Q., Li Y., Khan M. and Ma T. (2018) Proscillaridin a promotes oxidative stress and ER stress, inhibits STAT3 activation and induces apoptosis in A549 lung adenocarcinoma cells. Oxid. Med. Cell Longev. 2018, 3853409 10.1155/2018/3853409
    1. Hickman J.A. (1996) Apoptosis and chemotherapy resistance. Eur. J. Cancer 32A, 921–926 10.1016/0959-8049(96)00080-9
    1. Sun Y., Lin X. and Chang H. (2016) Proliferation inhibition and apoptosis of breast cancer MCF-7 cells under the influence of colchicine. J. BUON 3, 570–575
    1. Tsuruo T., Naito M., Tomida A.. et al. (2003) Molecular targeting therapy of cancer: drug resistance, apoptosis and survival signal. Cancer Sci. 94, 15–21
    1. Croce C.M. and Reed J.C. (2016) Finally, an apoptosis-targeting therapeutic for cancer. Cancer Res. 76, 5914–5920 10.1158/0008-5472.CAN-16-1248
    1. Jiang C., Wang Z., Ganther H. and Lu J. (2001) Caspases as key executors of methyl selenium-induced apoptosis (anoikis) of DU-145 prostate cancer cells. Cancer Res. 61, 3062–3070
    1. González-Gironès D.M., Moncunill-Massaguer C., Iglesias-Serret D.. et al. (2013) AICAR induces Bax/Bakdependent apoptosis through upregulation of the BH3-only proteins Bim and Noxa in mouse embryonic fibroblasts. Apoptosis 18, 1008–1016 10.1007/s10495-013-0850-6
    1. Reed J.C. (1997) Bcl-2 family proteins: regulators of apoptosis and chemoresistance in hematologic malignancies. Semin. Hematol. 34, 9–19
    1. Chen X.M., Liu J., Wang T. and Shang J. (2012) Colchicine-induced apoptosis in human normal liver L-02 cells by mitochondrial mediated pathways. Toxicol. In Vitro 26, 649–655 10.1016/j.tiv.2012.01.024
    1. Huang Z., Xu Y. and Peng W. (2015) Colchicine induces apoptosis in HT-29 human colon cancer cells via the AKT and c-Jun N-terminal kinase signaling pathways. Mol. Med. Rep. 12, 5939–5944 10.3892/mmr.2015.4222
    1. Yang J., Pi C. and Wang G. (2018) Inhibition of PI3K/Akt/mTOR pathway by apigenin induces apoptosis and autophagy in hepatocellular carcinoma cells. Biomed. Pharmacother. 103, 699–707 10.1016/j.biopha.2018.04.072
    1. Hua H., Zhu Y. and Song Y.H. (2018) Ruscogenin suppressed the hepatocellular carcinoma metastasis via PI3K/Akt/mTOR signaling pathway. Biomed. Pharmacother. 101, 115–122 10.1016/j.biopha.2018.02.031
    1. Engelman J.A. (2009) Targeting PI3K signalling in cancer: opportunities, challenges and limitations. Biomed. Pharmacother. 9, 550–562
    1. Xu C., Wang Q., Feng X. and Bo Y. (2014) Effect of evodiagenine mediates photocytotoxicity on human breast cancer cells MDA-MB-231 through inhibition of PI3K/AKT/mTOR and activation of p38 pathways. Fitoterapia 99, 292–299 10.1016/j.fitote.2014.10.010

Source: PubMed

3
Prenumerera