Skip to main content
Top
Published in: BMC Cancer 1/2024

Open Access 01-12-2024 | Acute Myeloid Leukemia | Research

UBE2C promotes the proliferation of acute myeloid leukemia cells through PI3K/AKT activation

Authors: Li Wang, Shuqin Zhao, Yongling Wang, Jianying Liu, Xiaoli Wang

Published in: BMC Cancer | Issue 1/2024

Login to get access

Abstract

This study aims to investigate the role and mechanism of tubiquitin-conjugating enzyme E2 C (UBE2C) in acute myeloid leukemia (AML). Initially, UBE2C expression in leukemia was analyzed using the Cancer Genome Atlas database. Further, we silenced UBE2C expression using small-hairpin RNA (sh-RNA). UBE2C expression was detected via the quantitative reverse-transcriptase polymerase chain reaction (qRT-PCR) and Western blot analysis. Apoptotic events and reactive oxygen species (ROS) levels were detected by flow cytometry. A xenograft model of leukemia cells were established, and the protein levels of UBE2C, KI-67, and cleaved-caspase 3 were detected by immunohistochemistry. We reported an overexpression of UBE2C in leukemia patients and cell lines (HL60, THP-1, U937, and KG-1 cells). Moreover, a high expression level of UBE2C was correlated with a dismal prognosis in AML patients. UBE2C knockdown inhibited the viability and promoted apoptosis in AML cells by regulating the phosphatidylinositol-3-kinase (PI3K)/protein kinase B (AKT) signaling pathway. Furthermore, UBE2C knockdown increased cellular Fe2+ and ROS levels, and enhanced erastin-induced ferroptosis in a proteasome-dependent manner. UBE2C knockdown also suppressed the tumor formation of AML cells in the mouse model. In summary, our findings suggest that UBE2C overexpression promotes the proliferation and inhibits ferroptosis in AML cells by activating the PI3K/AKT pathway.
Appendix
Available only for authorised users
Literature
1.
go back to reference Pelcovits A, Niroula R. Acute Myeloid Leukemia: A Review R I Med J (2013), 2020. 103(3): pp. 38–40. Pelcovits A, Niroula R. Acute Myeloid Leukemia: A Review R I Med J (2013), 2020. 103(3): pp. 38–40.
2.
go back to reference Infante MS, Piris M, Hernández-Rivas J. Molecular alterations in acute myeloid leukemia and their clinical and therapeutical implications. Med Clin (Barc). 2018;151(9):362–7.CrossRefPubMed Infante MS, Piris M, Hernández-Rivas J. Molecular alterations in acute myeloid leukemia and their clinical and therapeutical implications. Med Clin (Barc). 2018;151(9):362–7.CrossRefPubMed
3.
go back to reference Cancer Genome Atlas Research Network. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med. 2013;368(22):2059–74.CrossRef Cancer Genome Atlas Research Network. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med. 2013;368(22):2059–74.CrossRef
4.
go back to reference Kirtonia A, et al. A comprehensive review of genetic alterations and molecular targeted therapies for the implementation of personalized medicine in acute myeloid leukemia. J Mol Med (Berl). 2020;98(8):1069–91.CrossRefPubMed Kirtonia A, et al. A comprehensive review of genetic alterations and molecular targeted therapies for the implementation of personalized medicine in acute myeloid leukemia. J Mol Med (Berl). 2020;98(8):1069–91.CrossRefPubMed
7.
go back to reference Kirtonia A, et al. Long noncoding RNAs: a novel insight in the leukemogenesis and drug resistance in acute myeloid leukemia. J Cell Physiol. 2022;237(1):450–65.CrossRefPubMed Kirtonia A, et al. Long noncoding RNAs: a novel insight in the leukemogenesis and drug resistance in acute myeloid leukemia. J Cell Physiol. 2022;237(1):450–65.CrossRefPubMed
11.
go back to reference Yu Y, et al. The ferroptosis inducer erastin enhances sensitivity of acute myeloid leukemia cells to chemotherapeutic agents. Mol Cell Oncol. 2015;2(4):e1054549.CrossRefPubMedPubMedCentral Yu Y, et al. The ferroptosis inducer erastin enhances sensitivity of acute myeloid leukemia cells to chemotherapeutic agents. Mol Cell Oncol. 2015;2(4):e1054549.CrossRefPubMedPubMedCentral
13.
15.
go back to reference Liu Y, et al. UBE2C is upregulated by Estrogen and promotes epithelial-mesenchymal transition via p53 in Endometrial Cancer. Mol Cancer Res. 2020;18(2):204–15.CrossRefPubMed Liu Y, et al. UBE2C is upregulated by Estrogen and promotes epithelial-mesenchymal transition via p53 in Endometrial Cancer. Mol Cancer Res. 2020;18(2):204–15.CrossRefPubMed
16.
go back to reference Jin Z, et al. UBE2C promotes the progression of head and neck squamous cell carcinoma. Biochem Biophys Res Commun. 2020;523(2):389–97.CrossRefPubMed Jin Z, et al. UBE2C promotes the progression of head and neck squamous cell carcinoma. Biochem Biophys Res Commun. 2020;523(2):389–97.CrossRefPubMed
17.
go back to reference Lu ZN, et al. UBE2C affects breast cancer proliferation through the AKT/mTOR signaling pathway. Chin Med J (Engl). 2021;134(20):2465–74.CrossRefPubMed Lu ZN, et al. UBE2C affects breast cancer proliferation through the AKT/mTOR signaling pathway. Chin Med J (Engl). 2021;134(20):2465–74.CrossRefPubMed
18.
go back to reference Zhang S et al. The UBE2C/CDH1/DEPTOR axis is an oncogene and tumor suppressor cascade in lung cancer cells. J Clin Invest, 2023. 133(4). Zhang S et al. The UBE2C/CDH1/DEPTOR axis is an oncogene and tumor suppressor cascade in lung cancer cells. J Clin Invest, 2023. 133(4).
19.
go back to reference Martínez-Canales S, et al. Functional transcriptomic annotation and protein-protein interaction analysis identify EZH2 and UBE2C as key upregulated proteins in ovarian cancer. Cancer Med. 2018;7(5):1896–907.CrossRefPubMedPubMedCentral Martínez-Canales S, et al. Functional transcriptomic annotation and protein-protein interaction analysis identify EZH2 and UBE2C as key upregulated proteins in ovarian cancer. Cancer Med. 2018;7(5):1896–907.CrossRefPubMedPubMedCentral
20.
go back to reference Li J, et al. Depletion of UBE2C reduces ovarian cancer malignancy and reverses cisplatin resistance via downregulating CDK1. Biochem Biophys Res Commun. 2020;523(2):434–40.CrossRefPubMed Li J, et al. Depletion of UBE2C reduces ovarian cancer malignancy and reverses cisplatin resistance via downregulating CDK1. Biochem Biophys Res Commun. 2020;523(2):434–40.CrossRefPubMed
21.
23.
go back to reference Rawat A, et al. Inhibition of ubiquitin conjugating enzyme UBE2C reduces proliferation and sensitizes breast cancer cells to radiation, doxorubicin, tamoxifen and letrozole. Cell Oncol (Dordr). 2013;36(6):459–67.CrossRefPubMed Rawat A, et al. Inhibition of ubiquitin conjugating enzyme UBE2C reduces proliferation and sensitizes breast cancer cells to radiation, doxorubicin, tamoxifen and letrozole. Cell Oncol (Dordr). 2013;36(6):459–67.CrossRefPubMed
24.
go back to reference Bose MV, et al. Dominant negative ubiquitin-conjugating enzyme E2C sensitizes cervical cancer cells to radiation. Int J Radiat Biol. 2012;88(9):629–34.CrossRefPubMed Bose MV, et al. Dominant negative ubiquitin-conjugating enzyme E2C sensitizes cervical cancer cells to radiation. Int J Radiat Biol. 2012;88(9):629–34.CrossRefPubMed
25.
go back to reference Zhou Y, et al. UBE2C mediated radiotherapy resistance of head and neck squamous cell carcinoma by regulating oxidative-stress-relative apoptosis. Aging. 2022;14(17):7003–13.CrossRefPubMedPubMedCentral Zhou Y, et al. UBE2C mediated radiotherapy resistance of head and neck squamous cell carcinoma by regulating oxidative-stress-relative apoptosis. Aging. 2022;14(17):7003–13.CrossRefPubMedPubMedCentral
26.
go back to reference Kim YJ, et al. UBE2C overexpression aggravates patient outcome by promoting Estrogen-Dependent/Independent cell proliferation in early hormone receptor-positive and HER2-Negative breast Cancer. Front Oncol. 2019;9:1574.CrossRefPubMed Kim YJ, et al. UBE2C overexpression aggravates patient outcome by promoting Estrogen-Dependent/Independent cell proliferation in early hormone receptor-positive and HER2-Negative breast Cancer. Front Oncol. 2019;9:1574.CrossRefPubMed
27.
28.
go back to reference Hoang DH et al. Arsenic Trioxide and Venetoclax Synergize against AML progenitors by ROS induction and inhibition of Nrf2 activation. Int J Mol Sci, 2022. 23(12). Hoang DH et al. Arsenic Trioxide and Venetoclax Synergize against AML progenitors by ROS induction and inhibition of Nrf2 activation. Int J Mol Sci, 2022. 23(12).
29.
go back to reference Kirtonia A, Sethi G, Garg M. The multifaceted role of reactive oxygen species in tumorigenesis. Cell Mol Life Sci. 2020;77(22):4459–83.CrossRefPubMed Kirtonia A, Sethi G, Garg M. The multifaceted role of reactive oxygen species in tumorigenesis. Cell Mol Life Sci. 2020;77(22):4459–83.CrossRefPubMed
30.
go back to reference Bhandari A, et al. VASN promotes YAP/TAZ and EMT pathway in thyroid carcinogenesis in vitro. Am J Transl Res. 2019;11(6):3589–99.PubMedPubMedCentral Bhandari A, et al. VASN promotes YAP/TAZ and EMT pathway in thyroid carcinogenesis in vitro. Am J Transl Res. 2019;11(6):3589–99.PubMedPubMedCentral
31.
go back to reference Cao JZ, et al. UBE2C promotes the progression of pancreatic cancer and glycolytic activity via EGFR stabilization-mediated PI3K-Akt pathway activation. J Gastrointest Oncol. 2022;13(3):1444–53.CrossRefPubMedPubMedCentral Cao JZ, et al. UBE2C promotes the progression of pancreatic cancer and glycolytic activity via EGFR stabilization-mediated PI3K-Akt pathway activation. J Gastrointest Oncol. 2022;13(3):1444–53.CrossRefPubMedPubMedCentral
32.
go back to reference Wang F, et al. PD-L1 regulates cell proliferation and apoptosis in acute myeloid leukemia by activating PI3K-AKT signaling pathway. Sci Rep. 2022;12(1):11444.CrossRefPubMedPubMedCentral Wang F, et al. PD-L1 regulates cell proliferation and apoptosis in acute myeloid leukemia by activating PI3K-AKT signaling pathway. Sci Rep. 2022;12(1):11444.CrossRefPubMedPubMedCentral
33.
go back to reference Liu H, et al. Targeting PI3K/AKT/mTOR pathway to enhance the anti-leukemia efficacy of venetoclax. Exp Cell Res. 2022;417(2):113192.CrossRefPubMed Liu H, et al. Targeting PI3K/AKT/mTOR pathway to enhance the anti-leukemia efficacy of venetoclax. Exp Cell Res. 2022;417(2):113192.CrossRefPubMed
34.
go back to reference Xia T, et al. 20(S)-Ginsenoside Rh2 displays efficacy against T-cell acute lymphoblastic leukemia through the PI3K/Akt/mTOR signal pathway. J Ginseng Res. 2020;44(5):725–37.CrossRefPubMed Xia T, et al. 20(S)-Ginsenoside Rh2 displays efficacy against T-cell acute lymphoblastic leukemia through the PI3K/Akt/mTOR signal pathway. J Ginseng Res. 2020;44(5):725–37.CrossRefPubMed
35.
go back to reference Almajali B et al. Gene expression profiling and protein analysis reveal suppression of the C-Myc Oncogene and inhibition JAK/STAT and PI3K/AKT/mTOR signaling by Thymoquinone in Acute myeloid leukemia cells. Pharmaceuticals (Basel), 2022. 15(3). Almajali B et al. Gene expression profiling and protein analysis reveal suppression of the C-Myc Oncogene and inhibition JAK/STAT and PI3K/AKT/mTOR signaling by Thymoquinone in Acute myeloid leukemia cells. Pharmaceuticals (Basel), 2022. 15(3).
Metadata
Title
UBE2C promotes the proliferation of acute myeloid leukemia cells through PI3K/AKT activation
Authors
Li Wang
Shuqin Zhao
Yongling Wang
Jianying Liu
Xiaoli Wang
Publication date
01-12-2024
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2024
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-024-12212-x

Other articles of this Issue 1/2024

BMC Cancer 1/2024 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine