108
Views
4
CrossRef citations to date
0
Altmetric
Original Research

Sinomenine Inhibits the Growth of Ovarian Cancer Cells Through the Suppression of Mitosis by Down-Regulating the Expression and the Activity of CDK1

, ORCID Icon, ORCID Icon, , , , & show all
Pages 823-834 | Published online: 05 Feb 2021

References

  • Bray F, Ferlay J, Soerjomataram I, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394–424.
  • Tew WP. Ovarian cancer in the older woman. J Geriatr Oncol. 2016;7:354–361.
  • Pujade-Lauraine E. New treatments in ovarian cancer. Ann Oncol. 2017;28:viii57–viii60.
  • Cortez AJ, Tudrej P, Kujawa KA, et al. Advances in ovarian cancer therapy. Cancer Chemother Pharmacol. 2018;81:17–38.
  • Leung CS, Yeung TL, Yip KP, et al. Calcium-dependent FAK/CREB/TNNC1 signalling mediates the effect of stromal MFAP5 on ovarian cancer metastatic potential. Nat Commun. 2014;5:5092.
  • Zhang YS, Han JY, Iqbal O, et al. Research advances and prospects on mechanism of sinomenin on histamine release and the binding to histamine receptors. Int J Mol Sci. 2018;20:70.
  • Zhou H, Liu JX, Luo JF, et al. Suppressing mPGES-1 expression by sinomenine ameliorates inflammation and arthritis. Biochem Pharmacol. 2017;142:133–144.
  • Wu Y, Lin Z, Yan Z, et al. Sinomenine contributes to the inhibition of the inflammatory response and the improvement of osteoarthritis in mouse-cartilage cells by acting on the Nrf2/HO-1 and NF-κB signaling pathways. Int Immunopharmacol. 2019;75:105715.
  • Liu S, Chen Q, Liu J, et al. Sinomenine protects against E.coli-induced acute lung injury in mice through Nrf2-NF-κB pathway. Biomed Pharmacother. 2018;107:696‐702.
  • Zhu Q, Sun Y, Zhu J, et al. Antinociceptive effects of sinomenine in a rat model of neuropathic pain. Sci Rep. 2014;4:7270.
  • Feng ZT, Yang T, Hou XQ, et al. Sinomenine mitigates collagen-induced arthritis mice by inhibiting angiogenesis. Biomed Pharmacother. 2019;13:108759.
  • Tang J, Raza A, Chen J, et al. A systematic review on the sinomenine derivatives. Mini Rev Med Chem. 2018;18::906–917.
  • Hong Y, Yang J, Shen X, et al. Sinomenine hydrochloride enhancement of the inhibitory effects of anti-transferrin receptor antibody-dependent on the COX-2 pathway in human hepatoma cells. Cancer Immunol Immunother. 2013;62:447–454.
  • Li X, Wang K, Ren Y, et al. MAPK signaling mediates sinomenine hydrochloride-induced human breast cancer cell death via both reactive oxygen species-dependent and -independent pathways: an in vitro and in vivo study. Cell Death Dis. 2014;5:e1356.
  • Shen KH, Hung JH, Liao YC, et al. Sinomenine inhibits migration and invasion of human lung cancer cell through downregulating expression of miR-21 and MMPs. Int J Mol Sci. 2020;21:3080.
  • Zhao B, Liu L, Mao J, et al. Sinomenine hydrochloride attenuates the proliferation, migration, invasiveness, angiogenesis and epithelial-mesenchymal transition of clear-cell renal cell carcinoma cells via targeting Smad in vitro. Biomed Pharmacother. 2017;96:1036–1044.
  • Jiang Y, Jiao Y, Liu Y, et al. Sinomenine hydrochloride inhibits the metastasis of human glioblastoma cells by suppressing the expression of matrix metalloproteinase-2/-9 and reversing the endogenous and exogenous epithelial-mesenchymal transition. Int J Mol Sci. 2018;19:844.
  • Li H, Lin Z, Bai Y, et al. Sinomenine inhibits ovarian cancer cell growth and metastasis by mediating the Wnt/β-catenin pathway via targeting MCM2. RSC Adv. 2017;7:50017–50026.
  • Xu Y, Jiang T, Wang C, et al. Sinomenine hydrochloride exerts antitumor outcome in ovarian cancer cells by inhibition of long non-coding RNA HOST2 expression. Artif Cells Nanomed Biotechnol. 2019;47:4131–4138.
  • Tang F, Barbacioru C, Wang Y, et al. mRNA-seq whole-transcriptome analysis of a single cell. Nat Methods. 2009;6:377–382.
  • Wang J, Dean DC, Hornicek FJ, et al. RNA sequencing (RNA-Seq) and its application in ovarian cancer. Gynecol Oncol. 2019;152:194–201.
  • Yu B, Li H, Chen J, et al. Extensively expanded murine-induced hepatic stem cells maintain high-efficient hepatic differentiation potential for repopulation of injured livers. Liver Int. 2020. doi:10.1111/liv.14509
  • Trapnell C, Pachter L, Salzberg SL. TopHat: discovering splice junctions with RNA-Seq. Bioinformatics. 2009;25:1105–1111.
  • Li B, Dewey CN. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinform. 2011;12:323.
  • Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26:139–140.
  • Xie C, Mao X, Huang J, et al. KOBAS 2.0: a web server for annotation and identification of enriched pathways and diseases. Nucleic Acids Res. 2011;39:W316–W322.
  • Szklarczyk D, Morris JH, Cook H, et al. The STRING database in 2017: quality-controlled protein-protein association networks, made broadly accessible. Nucleic Acids Res. 2017;45:D362–D368.
  • Shannon P, Markiel A, Ozier O, et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003;13:2498–2504.
  • Yang LH, Wang Y, Qiao S, et al. Liver-enriched activator protein 1 as an isoform of ccaat/enhancer-binding protein beta suppresses stem cell features of hepatocellular carcinoma. Cancer Manag Res. 2018;10:873–885.
  • Liao H, Ji F, Ying S. CDK1: beyond cell cycle regulation. Aging. 2017;9:2465–2466.
  • Qi W, Tang Z, Yu H. Phosphorylation- and polo-box-dependent binding of Plk1 to bub1 is required for the kinetochore localization of Plk1. Mol Biol Cell. 2006;17:3705–3716.
  • Hunt T. Under arrest in the cell cycle. Nature. 1989;342:483–484.
  • Nurse P. Universal control mechanism regulating onset of M-phase. Nature. 1990;344:503–508.
  • Woodbury EL, Morgan DO. Cdk and APC activities limit the spindle-stabilizing function of Fin1 to anaphase. Nat Cell Biol. 2007;9:106–112.
  • Ducommun B, Brambilla P, Félix MA, et al. cdc2 phosphorylation is required for its interaction with cyclin. EMBO J. 1991;10:3311–3319.
  • Atherton-Fessler S, Liu F, Gabrielli B, et al. Cell cycle regulation of the p34cdc2 inhibitory kinases. Mol Biol Cell. 1994;5:989–1001.
  • Gezici S, Şekeroğlu N. Current perspectives in the application of medicinal plants against cancer: novel therapeutic agents. Anticancer Agents Med Chem. 2019;19:101–111.
  • Hall PA. Cell proliferation. J Pathol. 1991;165:349–354.
  • Matson JP, Cook JG. Cell cycle proliferation decisions: the impact of single cell analyses. FEBS J. 2017;284:362–375.
  • Mochida S, Rata S, Hino H, et al. Two bistable switches govern M phase entry. Curr Biol. 2016;26:3361–3367.
  • Norbury C, Blow J, Nurse P. Regulatory phosphorylation of the p34cdc2 protein kinase in vertebrates. EMBO J. 1991;10:3321–3329.
  • Hendzel MJ, Wei Y, Mancini MA, et al. Mitosis-specific phosphorylation of histone H3 initiates primarily within pericentromeric heterochromatin during G2 and spreads in an ordered fashion coincident with mitotic chromosome condensation. Chromosoma. 1997;106:348–360.
  • McIntosh JR. Mitosis. Cold Spring Harb Perspect Biol. 2016;8:a023218.
  • Petronczki M, Lénárt P, Peters JM. Polo on the rise-from mitotic entry to cytokinesis with Plk1. Dev Cell. 2008;14:646–659.
  • Kishi K, van Vugt MA, Okamoto K, et al. Functional dynamics of Polo-like kinase 1 at the centrosome. Mol Cell Biol. 2009;29:3134–3150.
  • Raaijmakers JA, van Heesbeen R, Blomen VA, et al. BUB1 is essential for the viability of human cells in which the spindle assembly checkpoint is compromised. Cell Rep. 2018;22:1424–1438.
  • Amin MA, McKenney RJ, Varma D. Antagonism between the dynein and Ndc80 complexes at kinetochores controls the stability of kinetochore-microtubule attachments during mitosis. J Biol Chem. 2018;293:5755–5765.