We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×

Anticancer activities of a benzimidazole compound through sirtuin inhibition in colorectal cancer

    Yi Jer Tan

    Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang, 11800, Malaysia

    ,
    Yeuan Ting Lee

    Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang, 11800, Malaysia

    ,
    Keng Yoon Yeong

    Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang, 11800, Malaysia

    School of Science, Monash University Malaysia Campus, Jalan Lagoon Selatan, Bandar Sunway, 47500, Selangor, Malaysia

    ,
    Sven H Petersen

    Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore

    ,
    Koji Kono

    Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore

    Department of Surgery, National University of Singapore, Singapore, Singapore

    School of Medicine, Fukushima Medical University, Fukushima, Japan

    ,
    Soo Choon Tan

    Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang, 11800, Malaysia

    &
    Chern Ein Oon

    *Author for correspondence: Tel.: +604 653 4879;

    E-mail Address: chern.oon@usm.my

    Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang, 11800, Malaysia

    Published Online:https://doi.org/10.4155/fmc-2018-0052

    Aim: This study aims to investigate the mode of action of a novel sirtuin inhibitor (BZD9L1) and its associated molecular pathways in colorectal cancer (CRC) cells. Materials & methods: BZD9L1 was tested against metastatic CRC cell lines to evaluate cytotoxicity, cell cycle and apoptosis, senescence, apoptosis related genes and protein expressions, as well as effect against major cancer signaling pathways. Results & conclusion: BZD9L1 reduced the viability, cell migration and colony forming ability of both HCT 116 and HT-29 metastatic CRC cell lines through apoptosis. BZD9L1 regulated major cancer pathways differently in CRC with different mutation profiles. BZD9L1 exhibited anticancer activities as a cytotoxic drug in CRC and as a promising therapeutic strategy in CRC treatment.

    Graphical abstract

    Papers of special note have been highlighted as: • of interest

    References

    • 1 Arnold M, Sierra MS, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global patterns and trends in colorectal cancer incidence and mortality. Gut 66(4), 683–691 (2016).
    • 2 Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM. Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int. J. Cancer 127(12), 2893–2917 (2010).
    • 3 Siegel RL, Miller KD, Fedewa SA et al. Colorectal cancer statistics, 2017. CA Cancer J. Clin. 67(3), 177–193 (2017).
    • 4 Longley D, Johnston P. Molecular mechanisms of drug resistance. J. Pathol. 205(2), 275–292 (2005).
    • 5 Aidan JC, Priddee NR, McAleer JJ. Chemotherapy causes cancer! A case report of therapy related acute myeloid leukaemia in early stage breast cancer. Ulster Med. J. 82(2), 97–99 (2013).
    • 6 Dizon DS, Krilov L, Cohen E et al. Clinical cancer advances 2016: annual report on progress against cancer from the American Society of Clinical Oncology. J. Clin. Oncol. 34(9), 987–1011 (2016).
    • 7 Gish RG, Finn RS, Marrero JA. Extending survival with the use of targeted therapy in the treatment of hepatocellular carcinoma. Gastroenterol. Hepatol. (NY) 9(4 Suppl. 2), 1–24 (2013).
    • 8 Keng Yoon Y, Ein Oon C. Sirtuin inhibitors: an overview from medicinal chemistry perspective. Anticancer Agents Med. Chem. 16(8), 1003–1016 (2016).
    • 9 Bradbury C, Khanim F, Hayden R et al. Histone deacetylases in acute myeloid leukaemia show a distinctive pattern of expression that changes selectively in response to deacetylase inhibitors. Leukemia 19(10), 1751–1759 (2005).
    • 10 Huffman DM, Grizzle WE, Bamman MM et al. SIRT1 is significantly elevated in mouse and human prostate cancer. Cancer Res. 67(14), 6612–6618 (2007).
    • 11 Stünkel W, Peh BK, Tan YC et al. Function of the SIRT1 protein deacetylase in cancer. Biotechnol. J. 2(11), 1360–1368 (2007).
    • 12 Schmidt MT, Smith BC, Jackson MD, Denu JM. Coenzyme specificity of Sir2 protein deacetylases implications for physiological regulation. J. Biol. Chem. 279(38), 40122–40129 (2004).
    • 13 Freitag M, Schemies J, Larsen T et al. Synthesis and biological activity of splitomicin analogs targeted at human NAD+-dependent histone deacetylases (sirtuins). Bioorg. Med. Chem. 19(12), 3669–3677 (2011).
    • 14 Oon CE, Strell C, Yeong KY, Östman A, Prakash J. SIRT1 inhibition in pancreatic cancer models: contrasting effects in vitro and in vivo. Eur. J. Pharmacol. 757, 59–67 (2015). • Reports the discovery of sirtuin inhibitor used in this study.
    • 15 Yoon Y, Ali M, Wei A et al. Correction: discovery of a potent and highly fluorescent sirtuin inhibitor. Medchemcomm 6(12), 2235–2235 (2015).
    • 16 Tonelli M, Tasso B, Mina L, Paglietti G, Boido V, Sparatore F. Primary anti-proliferative activity evaluation of 1-(quinolizidin-1’-yl)methyl- and 1-(ω-tert-amino)alkyl-substituted 2-phenyl-, 2-benzyl- and 2-[(benzotriazol-1/2-yl)methyl]benzimidazoles on human cancer cell lines. Mol. Divers. 17(3), 409–419 (2013).
    • 17 Blaszczak-Swiatkiewicz K, Olszewska P, Mikiciuk-Olasik E. Biological approach of anticancer activity of new benzimidazole derivatives. Pharmacol. Rep. 66(1), 100–106 (2014).
    • 18 Mantu D, Antoci V, Moldoveanu C, Zbancioc G, Mangalagiu II. Hybrid imidazole (benzimidazole)/pyridine (quinoline) derivatives and evaluation of their anticancer and antimycobacterial activity. J. Enzyme Inhib. Med. Chem. 31(Suppl. 2), 96–103 (2016).
    • 19 Özdemir A, Uzunoğlu S, Çalışkan B, Banoğlu E, Ark M. Anticancer activity of novel benzimidazole derivatives against MCF-7 cancer cells. FASEB J. 31(Suppl. 1), 934.910 (2017).
    • 20 Chu B, Liu F, Li L et al. A benzimidazole derivative exhibiting antitumor activity blocks EGFR and HER2 activity and upregulates DR5 in breast cancer cells. Cell Death Dis. 6, e1686 (2015).
    • 21 Dai X, Wang L, Deivasigamni A et al. A novel benzimidazole derivative, MBIC inhibits tumor growth and promotes apoptosis via activation of ROS-dependent JNK signaling pathway in hepatocellular carcinoma. Oncotarget 8(8), 12831–12842 (2017).
    • 22 Liu JF, Huang YL, Yang WH, Chang CS, Tang CH. 1-benzyl-2-phenylbenzimidazole (BPB), a benzimidazole derivative, induces cell apoptosis in human chondrosarcoma through intrinsic and extrinsic pathways. Int. J. Mol. Sci. 13(12), 16472–16488 (2012).
    • 23 Yoon YK, Ali MA, Wei AC et al. Correction: discovery of a potent and highly fluorescent sirtuin inhibitor. Medchemcomm 6(12), 2235–2235 (2015).
    • 24 Chaitanya GV, Alexander JS, Babu PP. PARP-1 cleavage fragments: signatures of cell-death proteases in neurodegeneration. Cell Commun. Signal. 8(1), 31 (2010).
    • 25 Nosho K, Shima K, Irahara N et al. SIRT1 histone deacetylase expression is associated with microsatellite instability and CpG island methylator phenotype in colorectal cancer. Mod. Pathol. 22(7), 922–932 (2009).
    • 26 Chen X, Sun K, Jiao S et al. High levels of SIRT1 expression enhance tumorigenesis and associate with a poor prognosis of colorectal carcinoma patients. Sci. Rep. 4, 7481 (2014).
    • 27 Wang Q, Zhou Y, Weiss HL, Evers BM. Abstract 4619: Sirt2 protein is required for activation of mTOR signaling in colorectal cancer cells. Cancer Res. 76(Suppl. 14), 4619 (2016).
    • 28 Fong Y, Lin Y-C, Wu C-Y et al. The antiproliferative and apoptotic effects of sirtinol, a sirtuin inhibitor on human lung cancer cells by modulating Akt/β-catenin-Foxo3a axis. ScientificWorldJournal 2014, 1–8 (2014).
    • 29 Lara E, Mai A, Calvanese V et al. Salermide, a sirtuin inhibitor with a strong cancer-specific proapoptotic effect. Oncogene 28(6), 781–791 (2009).
    • 30 Hirai S, Endo S, Saito R et al. Antitumor effects of a sirtuin inhibitor, tenovin-6, against gastric cancer cells via death receptor 5 up-regulation. PLoS ONE 9(7), e102831 (2014).
    • 31 Marques IJ, Weiss FU, Vlecken DH et al. Metastatic behaviour of primary human tumours in a zebrafish xenotransplantation model. BMC Cancer 9(1), 128 (2009).
    • 32 Carafa V, Rotili D, Forgione M et al. Sirtuin functions and modulation: from chemistry to the clinic. Clin. Epigenetics 8(1), 61 (2016). • Highlights the importance of sirtuin modulation in sirtuin inhibitor development.
    • 33 Kozako T, Suzuki T, Yoshimitsu M, Arima N, Honda S-I, Soeda S. Anticancer agents targeted to sirtuins. Molecules 19(12), 20295–20313 (2014). • Accounts the role of SIRT1 and SIRT2 as anticancer therapy targets.
    • 34 Kim H-W, Kim S-A, Ahn S-G. Sirtuin inhibitors, EX527 and AGK2, suppress cell migration by inhibiting HSF1 protein stability. Oncol. Rep. 35(1), 235–242 (2016).
    • 35 Bosch-Presegué L, Vaquero A. The dual role of sirtuins in cancer. Genes Cancer 2(6), 648–662 (2011). • Accounts the detailed information of sirtuin in cancer.
    • 36 Crowley LC, Christensen ME, Waterhouse NJ. Measuring survival of adherent cells with the colony-forming assay. Cold Spring Harb. Protoc. 2016(8), doi:10.1101/pdb.prot087171 (2016).
    • 37 Munshi A, Hobbs M, Meyn RE. Clonogenic cell survival assay. Methods Mol. Med. 110, 21–28 (2005).
    • 38 Chen J, Zhang B, Wong N et al. Sirtuin 1 is upregulated in a subset of hepatocellular carcinomas where it is essential for telomere maintenance and tumor cell growth. Cancer Res. 71(12), 4138–4149 (2011).
    • 39 Hao C, Zhu P-X, Yang X et al. Overexpression of SIRT1 promotes metastasis through epithelial–mesenchymal transition in hepatocellular carcinoma. BMC Cancer 14(1), 978 (2014).
    • 40 Palmirotta R, Cives M, Della-Morte D et al. Sirtuins and cancer: role in the epithelial–mesenchymal transition. Oxid. Med. Cell. Longev. 2016, 1–9 (2016).
    • 41 Zhang L, Wang X, Chen P. MiR-204 down regulates SIRT1 and reverts SIRT1-induced epithelial–mesenchymal transition, anoikis resistance and invasion in gastric cancer cells. BMC Cancer 13(1), 290 (2013).
    • 42 Wang J, Kim TH, Ahn MY et al. Sirtinol, a class III HDAC inhibitor, induces apoptotic and autophagic cell death in MCF-7 human breast cancer cells. Int. J. Oncol. 41(3), 1101–1109 (2012).
    • 43 Stenzinger A, Endris V, Klauschen F et al. High SIRT1 expression is a negative prognosticator in pancreatic ductal adenocarcinoma. BMC Cancer 13(1), 450 (2013).
    • 44 Heltweg B, Gatbonton T, Schuler AD et al. Antitumor activity of a small-molecule inhibitor of human silent information regulator 2 enzymes. Cancer Res. 66(8), 4368–4377 (2006).
    • 45 Bosch-Presegue L, Vaquero A. Sirtuins in stress response: guardians of the genome. Oncogene 33(29), 3764–3775 (2014). • Accounts the detailed information of sirtuin in human genome.
    • 46 Head PE, Zhang H, Bastien AJ et al. Sirtuin 2 mutations in human cancers impair its function in genome maintenance. J. Biol. Chem. 292(24), 9919–9931 (2017).
    • 47 Wlodkowic D, Telford W, Skommer J, Darzynkiewicz Z. Apoptosis and beyond: cytometry in studies of programmed cell death. Methods Cell Biol. 103, 55–98 (2011).
    • 48 Kan WLT, Yin C, Xu HX et al. Antitumor effects of novel compound, guttiferone K, on colon cancer by p21Waf1/Cip1-mediated G0/G1 cell cycle arrest and apoptosis. Int. J. Cancer 132(3), 707–716 (2013).
    • 49 Peck B, Chen C-Y, Ho K-K et al. SIRT inhibitors induce cell death and p53 acetylation through targeting both SIRT1 and SIRT2. Mol. Cancer Ther. 9(4), 844–855 (2010). • Reveals the effect on SIRT1 and SIRT2 inhibition by different sirtuin inhibitors.
    • 50 Karimian A, Ahmadi Y, Yousefi B. Multiple functions of p21 in cell cycle, apoptosis and transcriptional regulation after DNA damage. DNA Repair (Amst.) 42, 63–71 (2016).
    • 51 Ford J, Jiang M, Milner J. Cancer-specific functions of SIRT1 enable human epithelial cancer cell growth and survival. Cancer Res. 65(22), 10457–10463 (2005).
    • 52 Nakano K, Vousden KH. PUMA, a novel proapoptotic gene, is induced by p53. Mol. Cell 7(3), 683–694 (2001).
    • 53 Xia M, Knezevic D, Vassilev LT. p21 does not protect cancer cells from apoptosis induced by nongenotoxic p53 activation. Oncogene 30(3), 346–355 (2011).
    • 54 Chen T, Turner J, McCarthy S, Scaltriti M, Bettuzzi S, Yeatman TJ. Clusterin-mediated apoptosis is regulated by adenomatous polyposis coli and is p21 dependent but p53 independent. Cancer Res. 64(20), 7412–7419 (2004).
    • 55 Dvory-Sobol H, Sagiv E, Kazanov D, Ben-Ze'ev A, Arber N. Targeting the active beta-catenin pathway to treat cancer cells. Mol. Cancer Ther. 5(11), 2861–2871 (2006).
    • 56 Sharma NK, Kumar A, Kumari A et al. Nitric oxide down-regulates topoisomerase i and induces camptothecin resistance in human breast MCF-7 tumor cells. PLoS ONE 10(11), e0141897 (2015).
    • 57 Paul-Samojedny M, Kokocińska D, Samojedny A et al. Expression of cell survival/death genes: Bcl-2 and Bax at the rate of colon cancer prognosis. Biochim. Biophys. Acta 1741(1), 25–29 (2005).
    • 58 Yip K, Reed J. Bcl-2 family proteins and cancer. Oncogene 27(50), 6398–6406 (2008).
    • 59 Wajant H, Scheurich P. Tumor necrosis factor receptor-associated factor (TRAF) 2 and its role in TNF signaling. Int. J. Biochem. Cell Biol. 33(1), 19–32 (2001).
    • 60 Shu HB, Takeuchi M, Goeddel DV. The tumor necrosis factor receptor 2 signal transducers TRAF2 and c-IAP1 are components of the tumor necrosis factor receptor 1 signaling complex. Proc. Natl Acad. Sci. USA 93(24), 13973–13978 (1996).
    • 61 Petersen SL, Chen TT, Lawrence DA, Marsters SA, Gonzalvez F, Ashkenazi A. TRAF2 is a biologically important necroptosis suppressor. Cell Death Differ. 22(11), 1846–1857 (2015).
    • 62 Moskalev AA, Proshkina EN, Shaposhnikov MV. Gadd45 proteins in aging and longevity of mammals and drosophila. In: Life Extension: Lessons from Drosophila. Springer, Cham, Switzerland, 39–65 (2015).
    • 63 Tamura ER, De Vasconcellos FJ, Sarkar D, Libermann AT, Fisher BP, Zerbini FL. GADD45 proteins: central players in tumorigenesis. Curr. Mol. Med. 12(5), 634–651 (2012).