1,296
Views
0
CrossRef citations to date
0
Altmetric
Research Article

Five cuprotosis-related lncRNA signatures for prognosis prediction in acute myeloid leukaemia

Article: 2231737 | Received 10 Oct 2022, Accepted 27 Jun 2023, Published online: 07 Jul 2023

References

  • Short NJ, Rytting ME, Cortes JE. Acute myeloid leukaemia. Lancet. 2018;392(10147):593-606. doi:10.1016/S0140-6736(18)31041-9
  • Riva L, Luzi L, Pelicci PG. Genomics of acute myeloid leukemia: the next generation. Front Oncol. 2012;2:40. doi:10.3389/fonc.2012.00040
  • Zhang R, Xia LQ, Lu WW, Zhang J, Zhu JS. LncRNAs and cancer (Review). Oncol Lett. 2016;12(2):1233-1239. doi:10.3892/ol.2016.4770
  • Tan YT, Lin JF, Li T, et al. LncRNA-mediated posttranslational modifications and reprogramming of energy metabolism in cancer. Cancer Commun (Lond). 2021;41(2):109–120. doi:10.1002/cac2.12108
  • Schwerdtfeger M, Desiderio V, Kobold S, et al. Long non-coding RNAs in cancer stem cells. Transl Oncol. 2021;14(8):101134. doi:10.1016/j.tranon.2021.101134
  • Alvarez-Dominguez JR, Lodish HF. Emerging mechanisms of long noncoding RNA function during normal and malignant hematopoiesis. Blood. 2017;130(18):1965-1975. doi:10.1182/blood-2017-06-788695
  • Dahariya S, Paddibhatla I, Kumar S, et al. Long non-coding RNA: classification, biogenesis and functions in blood cells. Mol Immunol. 2019;112:82–92. doi:10.1016/j.molimm.2019.04.011
  • Gourvest M, Brousset P, Bousquet M. Long noncoding RNAs in acute myeloid leukemia: functional characterization and clinical relevance. Cancers (Basel). 2019;11(11):1638. doi:10.3390/cancers11111638
  • Banci L, Bertini I, Cantini F, et al. Cellular copper distribution: a mechanistic systems biology approach. Cell Mol Life Sci. 2010;67(15):2563–2589. doi:10.1007/s00018-010-0330-x
  • Scheiber I, Dringen R, Mercer JF. Copper: effects of deficiency and overload. Met Ions \Life Sci. 2013;13:359-387. doi:10.1007/978-94-007-7500-8_11
  • Tsvetkov P, Coy S, Petrova B, Dreishpoon M, Verma A, Abdusamad M, Rossen J, Joesch-Cohen L, Humeidi R, Spangler RD, Eaton JK, Frenkel E, Kocak M, Corsello SM, Lutsenko S, Kanarek N, Santagata S, Golub TR. Copper induces cell death by targeting lipoylated TCA cycle proteins. Science. 2022;375(6591):eabq4855. doi:10.1126/science.abf0529
  • Brady DC, Crowe MS, Turski ML, et al. Copper is required for oncogenic BRAF signalling and tumorigenesis. Nature. 2014;509(7501):492–6. doi:10.1038/nature13180
  • Li Y. Copper homeostasis: emerging target for cancer treatment. IUBMB Life. 2020;72(9):1900–1908. doi:10.1002/iub.2341
  • De Luca A, Barile A, Arciello M, et al. Copper homeostasis as target of both consolidated and innovative strategies of anti-tumor therapy. J Trace Elem Med Biol. 2019;55:204–213. doi:10.1016/j.jtemb.2019.06.008
  • Kaiafa GD, Saouli Z, Diamantidis MD, Kontoninas Z, Voulgaridou V, Raptaki M, Arampatzi S, Chatzidimitriou M, Perifanis V. Copper levels in patients with hematological malignancies. Eur J Intern Med. 2012;23(8):738-741. doi:10.1016/j.ejim.2012.07.009
  • Zuo XL, Chen JM, Zhou X, et al. Levels of selenium, zinc, copper, and antioxidant enzyme activity in patients with leukemia. Biol Trace Elem Res. 2006;114(1-3):41–54. doi:10.1385/BTER:114:1:41
  • Han J, Hu Y, Liu S, et al. A newly established cuproptosis-associated long non-coding RNA signature for predicting prognosis and indicating immune microenvironment features in soft tissue sarcoma. J Oncol. 2022;2022:1. doi:10.1155/2022/8489387
  • Yang L, Yu J, Tao L, et al. Cuproptosis-Related lncRNAs are biomarkers of prognosis and immune microenvironment in head and neck squamous cell carcinoma. Front Genet. 2022;13:947551. doi:10.3389/fgene.2022.947551
  • Goldman MJ, Craft B, Hastie M, et al. Visualizing and interpreting cancer genomics data via the Xena platform. Nat Biotechnol. 2020;38(6):675–678. doi:10.1038/s41587-020-0546-8
  • Raudvere U, Kolberg L, Kuzmin I, et al. g:Profiler: a web server for functional enrichment analysis and conversions of gene lists (2019 update). Nucleic Acids Res. 2019;47(W1):W191–W198. doi:10.1093/nar/gkz369
  • Rath S, Sharma R, Gupta R, et al. MitoCarta3.0: an updated mitochondrial proteome now with sub-organelle localization and pathway annotations. Nucleic Acids Res. 2021;49(D1):D1541–D1547. doi:10.1093/nar/gkaa1011
  • Aran D, Hu Z, Butte AJ. xCell: digitally portraying the tissue cellular heterogeneity landscape. Genome Biol. 2017;18(1):220), doi:10.1186/s13059-017-1349-1
  • Wan J, Guo C, Fang H, et al. Autophagy-related long non-coding RNA is a prognostic indicator for bladder cancer. Front Oncol. 2021; 11:647236. doi:10.3389/fonc.2021.647236
  • Wu B, Wang K, Fei J, Bao Y, Wang X, Song Z, Chen F, Gao J, Zhong Z. Novel three-lncRNA signature predicts survival in patients with pancreatic cancer. Oncol Rep. 2018;40(6):3427-3437. doi:10.3892/or.2018.6761
  • Grubman A, White AR. Copper as a key regulator of cell signalling pathways. Expert Rev Mol Med. 2014;16:e11. doi:10.1017/erm.2014.11
  • Ruiz LM, Libedinsky A, Elorza AA. Role of copper on mitochondrial function and metabolism. Front Mol Biosci. 2021;8:711227. doi:10.3389/fmolb.2021.711227
  • Edmund J, Thaliath LJ, Meleveedu K. Acute myeloid leukemia in the medically unfit elderly patients. Leukemia Res 2023; 130:107306. doi:10.1016/j.leukres.2023.107306
  • Fleischmann M, Scholl S, Frietsch JJ, et al. Clinical experience with venetoclax in patients with newly diagnosed, relapsed, or refractory acute myeloid leukemia. J Cancer Res Clin Oncol. 2022;148(11):3191–3202. doi:10.1007/s00432-022-03930-5
  • Mohamad Anuar NN, Nor Hisam NS, Liew SL, Ugusman A. Clinical review: navitoclax as a pro-apoptotic and anti-fibrotic agent. Front Pharmacol. 2020;11:564108. doi:10.3389/fphar.2020.564108
  • Bradner WT. Mitomycin C: a clinical update. Cancer Treat Rev. 2001;27(1):35–50. doi:10.1053/ctrv.2000.0202
  • Serafin V, Capuzzo G, Milani G, et al. Glucocorticoid resistance is reverted by LCK inhibition in pediatric T-cell acute lymphoblastic leukemia. Blood 2017; 130(25):2750–2761. doi:10.1182/blood-2017-05-784603
  • Wang XS, Zhang Z, Wang HC, et al. Rapid identification of UCA1 as a very sensitive and specific unique marker for human bladder carcinoma. Clin Cancer Res. 2006;12(16):4851–4858. doi:10.1158/1078-0432.CCR-06-0134
  • Li JJ, Chen XF, Wang M, et al. Long non-coding RNA UCA1 promotes autophagy by targeting miR-96-5p in acute myeloid leukaemia. Clin Exp Pharmacol Physiol. 2020;47(5):877–885. doi:10.1111/1440-1681.13259
  • Hughes JM, Legnini I, Salvatori B, et al. C/EBPα-p30 protein induces expression of the oncogenic long non-coding RNA UCA1 in acute myeloid leukemia. Oncotarget. 2015;6(21):18534–18544. doi:10.18632/oncotarget.4069
  • Li J, Li Z, Bai X, et al. Lncrna UCA1 promotes the progression of AML by upregulating the expression of CXCR4 and CYP1B1 by affecting the stability of METTL14. J Oncol. 2022;2022:1. doi:10.1155/2022/2756986
  • Zhang Y, Liu Y, Xu X. Knockdown of LncRNA-UCA1 suppresses chemoresistance of pediatric AML by inhibiting glycolysis through the microRNA-125a/hexokinase 2 pathway. J Cell Biochem. 2018;119(7):6296–6308. doi:10.1002/jcb.26899
  • Yao Q, Zhang L, Wang Y, Liu J, Yang L, Wang Y. LncRNA UCA1 elevates the resistance of human leukemia cells to daunorubicin by the PI3K/AKT pathway via sponging miR-613. Biosci Rep. 2021;41(6):BSR20201389. doi:10.1042/BSR20201389
  • Wang Z, Jin D, Zhou S, Dong N, Ji Y, An P, Wang J, Luo Y, Luo J. Regulatory roles of copper metabolism and cuproptosis in human cancers. Front Oncol. 2023;13:1123420. doi:10.3389/fonc.2023.1123420
  • Mehta R, Templeton DM, O'brien PJ. Mitochondrial involvement in genetically determined transition metal toxicity. Chem Biol Interact. 2006;163(1-2):77-85. doi:10.1016/j.cbi.2006.05.011
  • Vriens A, Nawrot TS, Janssen BG, Baeyens W, Bruckers L, Covaci A, De Craemer S, De Henauw S, Den Hond E, Loots I, Nelen V, Schettgen T, Schoeters G, Martens DS, Plusquin M.. Exposure to environmental pollutants and their association with biomarkers of aging: a multipollutant approach. Environ Sci Technol. 2019;53(10):5966-5976. doi:10.1021/acs.est.8b07141
  • Pruchniak MP, Demkow U. Potent NETosis inducers do not show synergistic effects in vitro. Cent Eur J Immunol. 2019;44(1):51-58. doi:10.5114/ceji.2019.84017
  • Demkow U. Neutrophil extracellular traps (NETs) in cancer invasion, evasion and metastasis. Cancers (Basel). 2021;13(17):4495. doi:10.3390/cancers13174495
  • Masucci MT, Minopoli M, Carriero MV. Tumor associated neutrophils. their role in tumorigenesis, metastasis, prognosis and therapy. Front Oncol. 2019;9:1146. doi:10.3389/fonc.2019.01146
  • Lee J, Lee D, Lawler S, et al. Role of neutrophil extracellular traps in regulation of lung cancer invasion and metastasis: structural insights from a computational model. PLoS Comput Biol. 2021;17(2):e1008257. doi:10.1371/journal.pcbi.1008257
  • Shaul ME, Fridlender ZG. Cancer-related circulating and tumor-associated neutrophils – subtypes, sources and function. FEBS J. 2018;285(23):4316–4342. doi:10.1111/febs.14524
  • Percival SS. Copper and immunity. Am J Clin Nutr. 1998;67(5 Suppl):1064S-1068S. doi:10.1093/ajcn/67.5.1064S
  • Liu Q, Hua M, Zhang C, et al. NLRP3-activated bone marrow dendritic cells play antileukemic roles via IL-1β/Th1/IFN-γ in acute myeloid leukemia. Cancer Lett. 2021;520:109–120. doi:10.1016/j.canlet.2021.06.014