|Table of Contents|

Inhibition of triple-negative breast cancer cell proliferation by synergistic induction of nucleolar stress by Flavokawain B and Actinomycin D

Journal Of Modern Oncology[ISSN:1672-4992/CN:61-1415/R]

Issue:
2024 23
Page:
4432-4442
Research Field:
Publishing date:

Info

Title:
Inhibition of triple-negative breast cancer cell proliferation by synergistic induction of nucleolar stress by Flavokawain B and Actinomycin D
Author(s):
LinghuiGAO QixueHE YesongLI Xuesen
Institute for Cancer Medicine,School of Basic Medical Sciences,Southwest Medical University,Sichuan Luzhou 646000,China.
Keywords:
triple-negative breast cancerFlavokawain BActinomycin Dnucleolar stresscell apoptosis
PACS:
R737.9
DOI:
10.3969/j.issn.1672-4992.2024.23.003
Abstract:
Objective:To investigate the synergistic inhibition of triple-negative breast cancer cell proliferation by Flavokawain B(FKB) and Actinomycin D(ActD),as well as their synergistic anticancer mechanism.Methods:Cultivate SUM159PT and HS578T triple-negative breast cancer cells,and employ the MTT assay to assess the inhibitory effects of different concentrations of FKB and ActD monotherapies,as well as appropriate concentrations of combination therapy,on the proliferation of these two types of cancer cells.To utilize flow cytometry to examine the effects of FKB and ActD monotherapies,as well as combination therapy,on cell cycle progression and apoptosis in the two types of triple-negative breast cancer cells.Conduct RT-qPCR experiments to analyze the changes in free rRNA that were not assembed into ribosomes following treatment with FKB and ActD monotherapies,as well as combination therapy,in the two types of triple-negative breast cancer cells.Perform cell immunofluorescence experiments to observe the nuclear localization of NPM1 protein in the two types of triple-negative breast cancer cells after treatment with FKB and ActD monotherapies,as well as combination therapy,in order to identify cellular nucleolar stress induction.Finally,to utilize Western blot experiments to assess changes in cell cycle and apoptosis-related proteins in the two types of triple-negative breast cancer cells following treatment with FKB and ActD monotherapies,as well as combination therapy.Results:FKB and ActD synergistically inhibited cell proliferation in two types of triple-negative breast cancer cells,and flow cytometry results showed that the combination therapy significantly induced more apoptosis in triple-negative breast cancer cells(P<0.01) and affected cell cycle progression,and that the combination’s inhibition of cell proliferation was attenuated by inhibition of apoptotic signaling pathways using apoptosis inhibitors and by controlling the cell cycle using serum deprivation(P<0.01).RT-qPCR results indicated that combination therapy led to a significant accumulation of both mature and immature 18S and 28S rRNA in triple-negative breast cancer cells (P<0.01),suggesting severe disruption of ribosomal RNA splicing or assembly.Cell immunofluorescence results showed that NPM1 protein in triple-negative breast cancer cells exhibited a diffuse distribution throughout the nucleus after combination therapy,indicating that the disruption of ribosomal RNA splicing or assembly triggered a nucleolar stress response in triple-negative breast cancer cells.Western blot results demonstrated that combination therapy significantly upregulated p21 protein,downregulated Bcl2 protein,and induced cleavage of Caspase3 and PARP proteins.Conclusion:FKB and ActD may synergistically disrupt ribosomal RNA splicing or assembly,destabilizing nucleolar homeostasis and triggering a nucleolar stress response.This,in turn,inducing upregulation of downstream p21 protein,leading to apoptosis of triple-negative breast cancer cells and triggering cell cycle arrest,thereby exerting a synergistic inhibitory effect on the proliferation of triple-negative breast cancer cells.

References:

[1]BIANCHINI G,DE ANGELIS C,LICATA L,et al.Treatment landscape of triple-negative breast cancer-expanded options,evolving needs[J].Nat Rev Clin Oncol,2022,19(2):91-113.
[2]PODO F,BUYDENS LM,DEGANI H,et al.Triple-negative breast cancer:Present challenges and new perspectives[J].Mol Oncol,2010,4(3):209-229.
[3]PLANA D,PALMER AC,SORGER PK.Independent drug action in combination therapy:Implications for precision oncology[J].Cancer Discov,2022,12(3):606-624.
[4]刘佳静,黄桂春,王文艺,等.化疗联合PD-(L)1单抗治疗晚期三阴性乳腺癌的研究进展[J].现代肿瘤医学,2023,31(6):1156-1161.LIU JJ,HUANG GC,WANG WY,et al.Research progress of chemotherapy combined with anti-PD-(L)1 treatment in advanced triple-negative breast cancer[J].Modern Oncology,2023,31(6):1156-1161.
[5]OU Y,WANG M,XU Q,et al.Small molecule agents for triple negative breast cancer:Current status and future prospects[J].Transl Oncol,2024,41:101893.
[6]YANG H,LI S,LI W,et al.Actinomycin D synergizes with Doxorubicin in triple-negative breast cancer by inducing P53-dependent cell apoptosis[J].Carcinogenesis,2024,45(4):262-273.
[7]FARBER S,D’ANGIO G,EVANS A,et al.Clinical studies on actinomycin D with special reference to Wilms’ tumor in children[J].Ann N Y Acad Sci,1960,89:421-425.
[8]FOSTER SA,DEMERS GW,ETSCHEID BG,et al.The ability of human papillomavirus E6 proteins to target p53 for degradation in vivo correlates with their ability to abrogate actinomycin D-induced growth arrest[J].J Virol,1994,68(9):5698-5705.
[9]CHOONG ML,YANG H,LEE MA,et al.Specific activation of the p53 pathway by low dose actinomycin D:a new route to p53 based cyclotherapy[J].Cell Cycle,2009,8(17):2810-2818.
[10]YANG K,YANG J,YI J.Nucleolar stress:Hallmarks,sensing mechanism and diseases[J].Cell Stress,2018,2(6):125-140.
[11]SOLLNER-WEBB B,TOWER J.Transcription of cloned eukaryotic ribosomal RNA genes[J].Annu Rev Biochem,1986,55:801-830.
[12]GONZALEZ-ARZOLA K.The nucleolus:Coordinating stress response and genomic stability[J].Biochim Biophys Acta Gene Regul Mech,2024,1867(2):195029.
[13]SIROZH O,SAEZ-MAS A,JUNG B,et al.Nucleolar stress caused by arginine-rich peptides triggers a ribosomopathy and accelerates aging in mice[J].Mol Cell,2024,84(8):1527-1540.
[14]ZENGER K,AGNOLET S,SCHNEIDER B,et al.Biotransformation of Flavokawains A,B,and C,Chalcones from Kava (Piper methysticum),by human liver microsomes[J].J Agric Food Chem,2015,63(28):6376-6385.
[15]MALAMI I,ABDUL AB,ABDULLAH R,et al.Crude extracts,Flavokawain B and Alpinetin compounds from the rhizome of alpinia mutica induce cell death via UCK2 enzyme inhibition and in turn reduce 18S rRNA biosynthesis in HT-29 cells[J].PLoS One,2017,12(1):e0170233.
[16]RAO B,LAIN S,THOMPSON AM.p53-based cyclotherapy:Exploiting the ’guardian of the genome’ to protect normal cells from cytotoxic therapy[J].Br J Cancer,2013,109(12):2954-2958.
[17]DERAKHSHAN F,REIS-FILHO JS.Pathogenesis of triple-negative breast cancer[J].Annu Rev Pathol,2022,17:181-204.
[18]LI Y,ZHANG H,MERKHER Y,et al.Recent advances in therapeutic strategies for triple-negative breast cancer[J].J Hematol Oncol,2022,15(1):121.
[19]CHATTERJEE N,BIVONA TG.Polytherapy and targeted cancer drug resistance[J].Trends Cancer,2019,5(3):170-182.
[20]JIN H,WANG L,BERNARDS R.Rational combinations of targeted cancer therapies:background,advances and challenges[J].Nat Rev Drug Discov,2023,22(3):213-234.
[21]ZAGIDULLIN B,ALDAHDOOH J,ZHENG S,et al.DrugComb:An integrative cancer drug combination data portal[J].Nucleic Acids Res,2019,47(W1):W43-W51.
[22]陈安莉,沈浩元,王舒.三阴性乳腺癌新辅助治疗的临床研究进展[J].现代肿瘤医学,2023,31(3):566-571.CHEN AL,SHEN HY,WANG S.Neoadjuvant therapy for triple negative breast cancer:Clinical progress[J].Modern Oncology,2023,31(3):566-571.
[23]TONG X,TANG R,XIAO M,et al.Targeting cell death pathways for cancer therapy:Recent developments in necroptosis,pyroptosis,ferroptosis,and cuproptosis research[J].J Hematol Oncol,2022,15(1):174.
[24]CHOI HK,RYU H,SON AR,et al.The novel anthraquinone derivative IMP1338 induces death of human cancer cells by p53-independent S and G2/M cell cycle arrest[J].Biomed Pharmacother,2016,79:308-314.
[25]SUSKI JM,BRAUN M,STRMISKA V,et al.Targeting cell-cycle machinery in cancer[J].Cancer Cell,2021,39(6):759-778.
[26]YUAN J,OFENGEIM D.A guide to cell death pathways[J].Nat Rev Mol Cell Biol,2024,25(5):379-395.
[27]RUBBI CP,MILNER J.Disruption of the nucleolus mediates stabilization of p53 in response to DNA damage and other stresses[J].EMBO J,2003,22(22):6068-6077.
[28]YANG K,WANG M,ZHAO Y,et al.A redox mechanism underlying nucleolar stress sensing by nucleophosmin[J].Nat Commun,2016,7:13599.
[29]DE LA TORRE P,FERNANDEZ-DE LA TORRE M,FLORES AI.Premature senescence of placental decidua cells as a possible cause of miscarriage produced by mycophenolic acid[J].J Biomed Sci,2021,28(1):3.
[30]LIU S,BISHOP WR,LIU M.Differential effects of cell cycle regulatory protein p21(WAF1/Cip1) on apoptosis and sensitivity to cancer chemotherapy[J].Drug Resist Updat,2003,6(4):183-195.
[31]ADAMS CM,MITRA R,XIAO Y,et al.Targeted MDM2 degradation reveals a new vulnerability for p53-inactivated triple-negative breast cancer[J].Cancer Discov,2023,13(5):1210-1229.
[32]TORRISI R,BALDUZZI A,GHISINI R,et al.Tailored preoperative treatment of locally advanced triple negative (hormone receptor negative and HER2 negative) breast cancer with epirubicin,cisplatin,and infusional fluorouracil followed by weekly paclitaxel[J].Cancer Chemother Pharmacol,2008,62(4):667-672.
[33]DUFFY MJ,SYNNOTT NC,CROWN J.Mutant p53 in breast cancer:Potential as a therapeutic target and biomarker[J].Breast Cancer Res Treat,2018,170(2):213-219.
[34]MAEHAMA T,NISHIO M,OTANI J,et al.Nucleolar stress:Molecular mechanisms and related human diseases[J].Cancer Sci,2023,114(5):2078-2086.
[35]IAROVAIA OV,MININA EP,SHEVAL EV,et al.Nucleolus:A central hub for nuclear functions[J].Trends Cell Biol,2019,29(8):647-659.
[36]CHEN J,STARK LA.Insights into the relationship between nucleolar stress and the NF-kappaB pathway[J].Trends Genet,2019,35(10):768-780.
[37]JAMES A,WANG Y,RAJE H,et al.Nucleolar stress with and without p53[J].Nucleus,2014,5(5):402-426.

Memo

Memo:
四川省科技计划联合创新专项(编号:2022YFS0623)
Last Update: 2024-11-01