Discover the SciOpen Platform and Achieve Your Research Goals with Ease.
Search articles, authors, keywords, DOl and etc.
IL-17 A is a promoter of colorectal cancer initiation and progression. Narciclasine is a polyhydroxy alkaloid compound isolated from Narcissus plants, which has potent anti-inflammatory and antitumor actions. The effects of narciclasine on colorectal tumors were evaluated, with a focus on IL-17 A. Narciclasine reduced the growth of HCT-116 and SW-480 colon cancer cells in vitro and in vivo in murine xenografts. The results of flow cytometry on JC-1 and Annexin V/PI revealed that narciclasine significantly reduced the mitochondrial membrane potential and induced apoptosis, findings confirmed by western blotting results of reduced Bcl-2 and enhanced Bax expression, as well as accumulation of cleaved Caspase-3, Caspase-8, Caspase-9, and cytoplasmic Cytochrome-c. After narciclasine incubation, IL-17 A, Act1, and TRAF6 were down-regulated, while p-P65 (Ser536) accumulated in the cytoplasm, a finding confirmed by laser scanning confocal microscopy. IL17A substitution could partly reverse these narciclasine effects while they were elevated by IL17A silencing. Moreover, IL-17 A, Act1, and TRAF6 were significantly expressed to greater extents in human colorectal cancer compared to normal adjacent tissue specimens and were closely linked with a poor prognosis. This study provided evidence that narciclasine may be a useful therapeutic drug for colorectal cancer treatment through its actions in down-regulating the L-17A/Act1/TRAF6/NF-κB anti-apoptotic signaling pathway.
Xi Y, Xu P. Global colorectal cancer burden in 2020 and projections to 2040. Transl Oncol. 2021;14(10):101174.
Chung AS, Wu X, Zhuang G, et al. An interleukin-17-mediated paracrine network promotes tumor resistance to anti-angiogenic therapy. Nat Med. 2013;19(9):1114-1123.
Liu C, Liu R, Wang B, et al. Blocking IL-17A enhances tumor response to anti-PD-1 immunotherapy in microsatellite stable colorectal cancer. J Immunother Cancer. 2021;9(1):e001895.
Fabre J, Giustiniani J, Garbar C, et al. Targeting the tumor microenvironment: the protumor effects of IL-17 related to cancer type. Int J Mol Sci. 2016;17(9):1433.
Gaffen SL. Structure and signalling in the IL-17 receptor family. Nat Rev Immunol. 2009;9(8):556–567.
Shalom-Barak T, Quach J, Lotz M. Interleukin-17-induced gene expression in articular chondrocytes is associated with activation of mitogen-activated protein kinases and NF-κB. J Biol Chem. 1998;273(42):27467–27473.
Lereclus E, Tout M, Girault A, et al. A possible association of baseline serum IL-17A concentrations with progression-free survival of metastatic colorectal cancer patients treated with a bevacizumab-based regimen. BMC Cancer. 2017;17:220.
Wang D, Yuan W, Wang Y, et al. Serum CCL20 combined with IL-17A as early diagnostic and prognostic biomarkers for human colorectal cancer. J Transl Med. 2019;17:253.
Ren H, Wang Z, Zhang S, et al. IL-17A promotes the migration and invasiveness of colorectal cancer cells through NF-κB-mediated MMP expression. Oncol Res. 2016;23(5):249–256.
Kornienko A, Evidente A. Chemistry, biology, and medicinal potential of narciclasine and its congeners. Chem Rev. 2008;108(6):1982–2014.
Fürst R. Narciclasine - an Amaryllidaceae alkaloid with potent antitumor and anti-inflammatory properties. Planta Med. 2016;82(16):1389–1394.
Cao C, Huang W, Zhang N, et al. Narciclasine induces autophagy-dependent apoptosis in triple-negative breast cancer cells by regulating the AMPK-ULK1 axis. Cell Prolif. 2018;51(6):e12518.
Van Goietsenoven G, Mathieu V, Lefranc F, et al. Narciclasine as well as other Amaryllidaceae isocarbostyrils are promising GTP-ase targeting agents against brain cancers. Med Res Rev. 2013;33(2):439–455.
Bräutigam J, Bischoff I, Schürmann C, et al. Narciclasine inhibits angiogenic processes by activation of Rho kinase and by downregulation of the VEGF receptor 2. J Mol Cell Cardiol. 2019;135:97–108.
Dumont P, Ingrassia L, Rouzeau S, et al. The Amaryllidaceae isocarbostyril narciclasine induces apoptosis by activation of the death receptor and/or mitochondrial pathways in cancer cells but not in normal fibroblasts. Neoplasia. 2007;9(9):766–776.
Deng H, Huang L, Liao Z, et al. Itraconazole inhibits the Hedgehog signaling pathway thereby inducing autophagy-mediated apoptosis of colon cancer cells. Cell Death Dis. 2020;11(7):539.
Vidal S, Puig L, Carrascosa-Carrillo JM, et al. From messengers to receptors in psoriasis: the role of IL-17RA in disease and treatment. Int J Mol Sci. 2021;22(13):6740.
Wu D, Wu P, Huang Q, et al. Interleukin-17: a promoter in colorectal cancer progression. Clin Dev Immunol. 2013;2013:436307.
Kryczek I, Wu K, Zhao E, et al. IL-17+ regulatory T cells in the microenvironments of chronic inflammation and cancer. J Immunol. 2011;186(7):4388–4395.
Yang S, Wang B, Guan C, et al. Foxp3+ IL-17+ T cells promote development of cancer-initiating cells in colorectal cancer. J Leukoc Biol. 2011;89(1):85–91.
Hyun YS, Han DS, Lee AR, et al. Role of IL-17A in the development of colitis-associated cancer. Carcinogenesis. 2012;33(4):931–936.
Housseau F, Wu S, Wick EC, et al. Redundant innate and adaptive sources of IL17 production drive colon tumorigenesis. Cancer Res. 2016;76(8):2115–2124.
Ali ET, Masri MAM, Siddig EE, et al. Immunohistochemical expression of interleukin-17 and hormonal receptors in benign and malignant breast lesions. BMC Res Notes. 2020;13:300.
Chang YH, Yu CW, Lai LC, et al. Up-regulation of interleukin-17 expression by human papillomavirus type 16 E6 in nonsmall cell lung cancer. Cancer. 2010;116(20):4800–4809.
Cao HX, Zhang W, Liang J. Inhibition of IL17A promotes bufalin-induced apoptosis in colon cancer cells via miR-96/DDIT3. Int J Clin Exp Pathol. 2016;9:4360–4367.
Steiner GE, Newman ME, Paikl D, et al. Expression and function of pro-inflammatory interleukin IL-17 and IL-17 receptor in normal, benign hyperplastic, and malignant prostate. Prostate. 2003;56(3):171–182.
Do Thi VA, Park SM, Lee H, et al. The membrane-bound form of IL-17A promotes the growth and tumorigenicity of colon cancer cells. Mol Cell. 2016;39(7):536–542.
Hu C, Zhang X, Wei W, et al. Matrine attenuates oxidative stress and cardiomyocyte apoptosis in doxorubicin-induced cardiotoxicity via maintaining AMPKα/UCP2 pathway. Acta Pharm Sin B. 2019;9(4):690–701.
Wang X. The expanding role of mitochondria in apoptosis. Genes Dev. 2001;15(22):2922–2933.
Green DR, Reed JC. Mitochondria and apoptosis. Science. 1998;281(5381):1309–1312.
Martinou JC, Youle RJ. Mitochondria in apoptosis: Bcl-2 family members and mitochondrial dynamics. Dev Cell. 2011;21(1):92–101.
Asakura T, Ohkawa K. Chemotherapeutic agents that induce mitochondrial apoptosis. Curr Cancer Drug Targets. 2004;4(7):577–590.
Soleimani A, Rahmani F, Ferns GA, et al. Role of the NF-κB signaling pathway in the pathogenesis of colorectal cancer. Gene. 2020;726:144132.
Cui G, Yuan A, Goll R, et al. IL-17A in the tumor microenvironment of the human colorectal adenoma–carcinoma sequence. Scand J Gastroenterol. 2012;47(11):1304–1312.
Sun H, Li X, Fan L, et al. TRAF6 is upregulated in colon cancer and promotes proliferation of colon cancer cells. Int J Biochem Cell Biol. 2014;53:195–201.
This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).