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

Open Access 01-12-2024 | Prostate Cancer | Research

HIST3H2A promotes the progression of prostate cancer through inhibiting cell necroptosis

Authors: Lihong Yang, Yong Ruan, Houqiang Xu

Published in: BMC Cancer | Issue 1/2024

Login to get access

Abstract

In recent years, there has been an increase in the incidence and mortality rates of prostate cancer (PCa). However, the specific molecular mechanisms underlying its occurrence and development remain unclear, necessitating the identification of new therapeutic targets. Through bioinformatics analysis, we discovered a previously unstudied differential gene called HIST3H2A in prostate cancer. Our study revealed that HIST3H2A is highly expressed in PCa tissues, as confirmed by analysis of both the GEO and UALCAN databases. Further analysis using the KEGG database demonstrated that HIST3H2A regulates the pathway of programmed necroptosis in cells. Additionally, we observed significant up-regulation of HIST3H2A in PCa tissues and cell lines. HIST3H2A was found to regulate cell proliferation, migration, invasion, and the epithelial-mesenchymal transition (EMT) process in tumors. Notably, HIST3H2A’s role in regulating programmed necroptosis in prostate cancer cells differs from its role in apoptosis. In vitro and in vivo experiments collectively support the key role of HIST3H2A in promoting the development of prostate cancer, highlighting its potential as a therapeutic target for patients with PCa.
Appendix
Available only for authorised users
Literature
27.
go back to reference Fedyuk V, Erez N, Furth N, Beresh O, Andreishcheva E, Shinde A, Jones D, Zakai BB, Mavor Y, Peretz T, Hubert A, Cohen JE, Salah A, Temper M, Grinshpun A, Maoz M, Zick A, Ron G, Shema E. Multiplexed, single-molecule, epigenetic analysis of plasma-isolated nucleosomes for cancer diagnostics. Nat Biotechnol. 2023;41(2):212–21. https://doi.org/10.1038/s41587-022-01447-3.CrossRefPubMed Fedyuk V, Erez N, Furth N, Beresh O, Andreishcheva E, Shinde A, Jones D, Zakai BB, Mavor Y, Peretz T, Hubert A, Cohen JE, Salah A, Temper M, Grinshpun A, Maoz M, Zick A, Ron G, Shema E. Multiplexed, single-molecule, epigenetic analysis of plasma-isolated nucleosomes for cancer diagnostics. Nat Biotechnol. 2023;41(2):212–21. https://​doi.​org/​10.​1038/​s41587-022-01447-3.CrossRefPubMed
32.
go back to reference Ogitani Y, Aida T, Hagihara K, Yamaguchi J, Ishii C, Harada N, Soma M, Okamoto H, Oitate M, Arakawa S, Hirai T, Atsumi R, Nakada T, Hayakawa I, Abe Y, Agatsuma T. DS-8201a, A Novel HER2-Targeting ADC with a novel DNA topoisomerase I inhibitor, demonstrates a Promising Antitumor Efficacy with differentiation from T-DM1. Clin Cancer Res. 2016;22(20):5097–108. https://doi.org/10.1158/1078-0432.CCR-15-2822.CrossRefPubMed Ogitani Y, Aida T, Hagihara K, Yamaguchi J, Ishii C, Harada N, Soma M, Okamoto H, Oitate M, Arakawa S, Hirai T, Atsumi R, Nakada T, Hayakawa I, Abe Y, Agatsuma T. DS-8201a, A Novel HER2-Targeting ADC with a novel DNA topoisomerase I inhibitor, demonstrates a Promising Antitumor Efficacy with differentiation from T-DM1. Clin Cancer Res. 2016;22(20):5097–108. https://​doi.​org/​10.​1158/​1078-0432.​CCR-15-2822.CrossRefPubMed
38.
go back to reference Weinreb O, Amit T, Bar-Am O, Sagi Y, Mandel S, Youdim MB. Involvement of multiple survival signal transduction pathways in the neuroprotective, neurorescue and APP processing activity of rasagiline and its propargyl moiety. J Neural Transm Suppl. 2006;70457–65. https://doi.org/10.1007/978-3-211-45295-0_69. Weinreb O, Amit T, Bar-Am O, Sagi Y, Mandel S, Youdim MB. Involvement of multiple survival signal transduction pathways in the neuroprotective, neurorescue and APP processing activity of rasagiline and its propargyl moiety. J Neural Transm Suppl. 2006;70457–65. https://​doi.​org/​10.​1007/​978-3-211-45295-0_​69.
56.
go back to reference Berta DG, Kuisma H, Välimäki N, Räisänen M, Jäntti M, Pasanen A, Karhu A, Kaukomaa J, Taira A, Cajuso T, Nieminen S, Penttinen RM, Ahonen S, Lehtonen R, Mehine M, Vahteristo P, Jalkanen J, Sahu B, Ravantti J, Mäkinen N, Rajamäki K, Palin K, Taipale J, Heikinheimo O, Bützow R, Kaasinen E, Aaltonen LA. Deficient H2A.Z deposition is associated with genesis of uterine leiomyoma. Nature. 2021;596(7872):398–403. https://doi.org/10.1038/s41586-021-03747-1.CrossRefPubMed Berta DG, Kuisma H, Välimäki N, Räisänen M, Jäntti M, Pasanen A, Karhu A, Kaukomaa J, Taira A, Cajuso T, Nieminen S, Penttinen RM, Ahonen S, Lehtonen R, Mehine M, Vahteristo P, Jalkanen J, Sahu B, Ravantti J, Mäkinen N, Rajamäki K, Palin K, Taipale J, Heikinheimo O, Bützow R, Kaasinen E, Aaltonen LA. Deficient H2A.Z deposition is associated with genesis of uterine leiomyoma. Nature. 2021;596(7872):398–403. https://​doi.​org/​10.​1038/​s41586-021-03747-1.CrossRefPubMed
Metadata
Title
HIST3H2A promotes the progression of prostate cancer through inhibiting cell necroptosis
Authors
Lihong Yang
Yong Ruan
Houqiang Xu
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/s12885-024-12308-4

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