Fol. Biol. 2022, 68, 189-200

https://doi.org/10.14712/fb2022068050189

Mechanistic Study of Macranthoside B Effects on Apoptotic Cell Death in Human Cervical Adenocarcinoma Cells

Y. Li1, M. Li2,3, K. Ahmed4, J. Yang1, L. Song1, Zheng-Guo Cui2, Yusuke Hiraku2

1School of Life Science and Technology, Henan Institute of Science and Technology, Xinxiang, China
2Department of Environmental Health, University of Fukui School of Medical Sciences, Fukui, Japan
3School of Medicine, Xizang Minzu University, Weicheng District, Xianyang, Shaanxi, China
4Faculty of Eastern Medicine, Hamdard University, Islamabad, Pakistan

Received October 2021
Accepted February 2023

References

1. Avato, P., Migoni, D., Argentieri, M., Fanizzi, F. P., Tava, A. (2017) Activity of saponins from Medicago species against HeLa and MCF-7 cell lines and their capacity to potentiate cisplatin effect. Anticancer Agents Med. Chem. 17, 1508-1518. <https://doi.org/10.2174/1871520617666170727152805>
2. Bagli, E., Stefaniotou, M., Morbidelli, L., Ziche, M., Psillas, K., Murphy, C., Fotsis, T. (2004) Luteolin inhibits vascular endothelial growth factor-induced angiogenesis; inhibition of endothelial cell survival and proliferation by targeting phosphatidylinositol 3’-kinase activity. Cancer Res. 64, 7936-7946. <https://doi.org/10.1158/0008-5472.CAN-03-3104>
3. Bahrami, A., Hasanzadeh, M., Hassanian, S. M., ShahidSales, S., Ghayour-Mobarhan, M., Ferns, G. A., Avan, A. (2017) The potential value of the PI3K/Akt/mTOR signaling pathway for assessing prognosis in cervical cancer and as a target for therapy. J. Cell. Biochem. 118, 4163-4169. <https://doi.org/10.1002/jcb.26118>
4. Bhatla, N., Aoki, D., Sharma, D. N., Sankaranarayanan, R. (2018) Cancer of the cervix uteri. Int. J. Gynaecol. Obstet. 143(Suppl 2), 22-36. <https://doi.org/10.1002/ijgo.12611>
5. Chen, C. Y., Qi, L. W., Yi, L., Li, P., Wen, X. D. (2009) Liquid chromatography-mass spectrometry analysis of macranthoidin B, macranthoidin A, dipsacoside B, and macranthoside B in rat plasma for the pharmacokinetic investigation. J. Chromatogr. B, Analyt. Technol. Biomed. Life Sci. 877, 159-165. <https://doi.org/10.1016/j.jchromb.2008.11.043>
6. Chen, X., Zhou, Y. L., Liang, S. Y., Shi, Y. C., Lin, S., Shu, M. Q. (2019) Overexpression of UHRF1 promoted the proliferation of vascular smooth cells via the regulation of Geminin protein levels. Biosci. Rep. 39, BSR20181341. <https://doi.org/10.1042/BSR20181341>
7. Choi, J. H., Yang, Y. R., Lee, S. K., Kim, S. H., Kim, Y. H., Cha, J. Y., Oh, S. W., Ha, J. R., Ryu, S. H., Suh, P. G. (2008) Potential inhibition of PDK1/Akt signaling by phenothiazines suppresses cancer cell proliferation and survival. Ann. N. Y. Acad. Sci. 1138, 393-403. <https://doi.org/10.1196/annals.1414.041>
8. Cragg, G. M., Newman, D. J. (2005) Plants as a source of anti-cancer agents. J. Ethnopharmacol. 100, 72-79. <https://doi.org/10.1016/j.jep.2005.05.011>
9. Cui, Z. G., Kondo, T., Matsumoto, H. (2006) Enhancement of apoptosis by nitric oxide released from alpha-phenyl-tert-butyl nitrone under hyperthermic conditions. J. Cell. Physiol. 206, 468-476. <https://doi.org/10.1002/jcp.20482>
10. Ferlay, J., Soerjomataram, I., Dikshit, R., Eser, S., Mathers, C., Rebelo, M., Parkin, D. M., Forman, D., Bray, F. (2015) Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int. J. Cancer 136, E359-E386. <https://doi.org/10.1002/ijc.29210>
11. Fonseca-Silva, F., Inacio, J. D., Canto-Cavalheiro, M. M., Almeida-Amaral, E. E. (2011) Reactive oxygen species production and mitochondrial dysfunction contribute to quercetin induced death in Leishmania amazonensis. PLoS One 6, e14666. <https://doi.org/10.1371/journal.pone.0014666>
12. Guan, F., Shan, Y., Zhao, X., Zhang, D., Wang, M., Peng, F., Xia, B., Feng, X. (2011) Apoptosis and membrane permeabilisation induced by macranthoside B on HL-60 cells. Nat. Prod. Res. 25, 332-340. <https://doi.org/10.1080/14786411003752086>
13. Harris, T. K. (2003) PDK1 and PKB/Akt: ideal targets for development of new strategies to structure-based drug design. IUBMB Life 55, 117-126. <https://doi.org/10.1080/1521654031000115951>
14. Jiang, P., Sheng, Y. C., Chen, Y. H., Ji, L. L., Wang, Z. T. (2014) Protection of Flos Lonicerae against acetaminophen-induced liver injury and its mechanism. Environ. Toxicol. Pharmacol. 38, 991-999. <https://doi.org/10.1016/j.etap.2014.10.019>
15. Johnson-Cadwell, L. I., Jekabsons, M. B., Wang, A., Polster, B. M, Nicholls, D. G. (2007) ‘Mild Uncoupling’ does not decrease mitochondrial superoxide levels in cultured cerebellar granule neurons but decreases spare respiratory capacity and increases toxicity to glutamate and oxidative stress. J. Neurochem. 101, 1619-1631. <https://doi.org/10.1111/j.1471-4159.2007.04516.x>
16. Joshi, P., Vishwakarma, R. A., Bharate, S. B. (2017) Natural alkaloids as P-gp inhibitors for multidrug resistance reversal in cancer. Eur. J. Med. Chem. 138, 273-292. <https://doi.org/10.1016/j.ejmech.2017.06.047>
17. Kharbanda, S., Pandey, P., Schofield, L., Israels, S., Roncinske, R., Yoshida, K., Bharti, A., Yuan, Z. M., Saxena, S., Weichselbaum, R., Nalin, C., Kufe, D. (1997) Role for Bcl-xL as an inhibitor of cytosolic cytochrome C accumulation in DNA damage-induced apoptosis. Proc. Natl. Acad. Sci. USA 94, 6939-6942. <https://doi.org/10.1073/pnas.94.13.6939>
18. Lazorchak, A. S., Su, B. (2011) Perspectives on the role of mTORC2 in B lymphocyte development, immunity and tumorigenesis. Protein Cell 2, 523-530. <https://doi.org/10.1007/s13238-011-1077-3>
19. Li, M., Cui, Z. G., Zakki, S. A., Feng, Q., Sun, L., Feril, L. B. Jr., Inadera, H. (2019) Aluminum chloride causes 5-fluorouracil resistance in hepatocellular carcinoma HepG2 cells. J. Cell. Physiol. 234, 20249-20265. <https://doi.org/10.1002/jcp.28625>
20. Li, X. L., Meng, Q. H., Fan, S. J. (2009) Adenovirus-mediated expression of UHRF1 reduces the radiosensitivity of cervical cancer HeLa cells to γ-irradiation. Acta Pharmacol. Sin. 30, 458-466. <https://doi.org/10.1038/aps.2009.18>
21. Liu, P., Cheng, H., Roberts, T. M., Zhao, J. J. (2009) Targeting the phosphoinositide 3-kinase pathway in cancer. Nat. Rev. Drug. Discov. 8, 627-644. <https://doi.org/10.1038/nrd2926>
22. Majumder, D., Das, A., Saha, C. (2017) Catalase inhibition an anti cancer property of flavonoids: a kinetic and structural evaluation. Int. J. Biol. Macromol. 104, 929-935. <https://doi.org/10.1016/j.ijbiomac.2017.06.100>
23. Nitulescu, G. M., Margina, D., Juzenas, P., Peng, Q., Olaru, O. T., Saloustros, E., Fenga, C., Spandidos, D., Libra, M., Tsatsakis, A. M. (2016) Akt inhibitors in cancer treatment: the long journey from drug discovery to clinical use (review). Int. J. Oncol. 48, 869-885. <https://doi.org/10.3892/ijo.2015.3306>
24. Nitulescu, G. M., Van De Venter, M., Nitulescu, G., Ungurianu, A., Juzenas, P., Peng, Q., Olaru, O. T., Gradinaru, D., Tsatsakis, A., Tsoukalas, D., Spandidos, D. A., Margina, D. (2018) The Akt pathway in oncology therapy and beyond (review). Int. J. Oncol. 53, 2319-2331.
25. Ohta, S., Sato, N., Tu, S. H., Shinoda, M. (1993) Protective effects of Taiwan crude drugs on experimental liver injuries. Yakugaku Zasshi 113, 870-880. (in Japanese) <https://doi.org/10.1248/yakushi1947.113.12_870>
26. Prasad, S. B., Yadav, S. S., Das, M., Modi, A., Kumari, S., Pandey, L. K., Singh, S., Pradhan, S., Narayan, G. (2015) PI3K/AKT pathway-mediated regulation of p27(Kip1) is associated with cell cycle arrest and apoptosis in cervical cancer. Cell. Oncol. (Dordr.) 38, 215-225. <https://doi.org/10.1007/s13402-015-0224-x>
27. Prasedya, E. S., Miyake, M., Kobayashi, D., Hazama, A. (2016) Carrageenan delays cell cycle progression in human cancer cells in vitro demonstrated by FUCCI imaging. BMC Complement. Altern. Med. 16, 270. <https://doi.org/10.1186/s12906-016-1199-5>
28. Redza-Dutordoir, M., Averill-Bates, D. A. (2016) Activation of apoptosis signalling pathways by reactive oxygen species. Biochim. Biophys. Acta 1863, 2977-2992. <https://doi.org/10.1016/j.bbamcr.2016.09.012>
29. Riccardi, C., Nicoletti, I. (2006) Analysis of apoptosis by propidium iodide staining and flow cytometry. Nat. Protoc. 1, 1458-1461. <https://doi.org/10.1038/nprot.2006.238>
30. Sambrook, J., Fritsch, T., Maniatis, T. (1989) Molecular Cloning: A Laboratory Manual. 1. Cold Spring Harbor Laboratory Press, New York.
31. Shan, Y., Guan, F., Zhao, X., Wang, M., Chen, Y., Wang, Q., Feng, X. (2016) Macranthoside B induces apoptosis and autophagy via reactive oxygen species accumulation in human ovarian cancer A2780 cells. Nutr. Cancer 68, 280-289. <https://doi.org/10.1080/01635581.2016.1142587>
32. Stevens, M., Oltean, S. (2019) Modulation of the apoptosis gene Bcl-x function through alternative splicing. Front. Genet. 10, 804. <https://doi.org/10.3389/fgene.2019.00804>
33. Sun, L., Zhang, Q., Luan, H., Zhan, Z., Wang, C., Sun, B. (2011) Comparison of KRAS and EGFR gene status between primary non-small cell lung cancer and local lymph node metastases: implications for clinical practice. J. Exp. Clin. Cancer Res. 30, 30. <https://doi.org/10.1186/1756-9966-30-30>
34. Wang, J., Zhao, X. Z., Qi, Q., Tao, L., Zhao, Q., Mu, R., Gu, H. Y., Wang, M., Feng, X., Guo, Q. L. (2009) Macranthoside B, a hederagenin saponin extracted from Lonicera macranthoides and its anti-tumor activities in vitro and in vivo. Food Chem. Toxicol. 47, 1716-1721. <https://doi.org/10.1016/j.fct.2009.04.034>
35. Wang, Y., Yuan, Y., Gao, Y., Li, X., Tian, F., Liu, F., Du, R., Li, P., Wang, F., Xu, S., Wu, X., Wang, C. (2019) MicroRNA-31 regulating apoptosis by mediating the phosphatidylinositol-3 kinase/protein kinase B signaling pathway in treatment of spinal cord injury. Brain Dev. 41, 649-661. <https://doi.org/10.1016/j.braindev.2019.04.010>
36. Webster, K. A. (2012) Mitochondrial membrane permeabilization and cell death during myocardial infarction: roles of calcium and reactive oxygen species. Future Cardiol. 8, 863-884. <https://doi.org/10.2217/fca.12.58>
37. Yun, S. M., Woo, S. H., Oh, S. T., Hong, S. E., Choe, T. B., Ye, S. K., Kim, E. K., Seong, M. K., Kim, H. A., Noh, W. C., Lee, J. K., Jin, H. O., Lee, Y. H., Park, I. C. (2016) Melatonin enhances arsenic trioxide-induced cell death via sustained upregulation of Redd1 expression in breast cancer cells. Mol. Cell. Endocrinol. 422, 64-73. <https://doi.org/10.1016/j.mce.2015.11.016>
38. Zakki, S. A., Cui, Z. G., Sun, L., Feng, Q. W., Li, M. L., Inadera, H. (2018) Baicalin augments hyperthermia-induced apo­ptosis in U937 cells and modulates the MAPK pathway via ROS generation. Cell. Physiol. Biochem. 45, 2444-2460. <https://doi.org/10.1159/000488263>
front cover

ISSN 0015-5500 (Print) ISSN 2533-7602 (Online)

Open access journal

Archive