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

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

SIRT7 promotes the proliferation and migration of anaplastic thyroid cancer cells by regulating the desuccinylation of KIF23

Authors: Yongkang Wu, Weijie Chen, Huilai Miao, Tuo Xu

Published in: BMC Cancer | Issue 1/2024

Login to get access

Abstract

Objective

This study was designed to investigate the regulatory effects of kinesin family member (KIF) 23 on anaplastic thyroid cancer (ATC) cell viability and migration and the underlying mechanism.

Methods

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was used to analyze the levels of KIF23 in ATC cells. Besides, the effects of KIF23 and sirtuin (SIRT) 7 on the viability and migration of ATC cells were detected using cell counting kit-8, transwell and wound healing assays. The interaction between SIRT7 and KIF23 was evaluated by co-immunoprecipitation (Co-IP) assay. The succinylation (succ) of KIF23 was analyzed by western blot.

Results

The KIF23 expression was upregulated in ATC cells. Silencing of KIF23 suppressed the viability and migration of 8505C and BCPAP cells. The KIF23-succ level was decreased in ATC cells. SIRT7 interacted with KIF23 to inhibit the succinylation of KIF23 at K537 site in human embryonic kidney (HEK)-293T cells. Overexpression of SIRT7 enhanced the protein stability of KIF23 in HEK-293T cells. Besides, overexpression of KIF23 promoted the viability and migration of 8505C and BCPAP cells, which was partly blocked by silenced SIRT7.

Conclusions

SIRT7 promoted the proliferation and migration of ATC cells by regulating the desuccinylation of KIF23.
Appendix
Available only for authorised users
Literature
7.
go back to reference Liu X, et al. Molecular interactions of Polo-like-kinase 1 with the mitotic kinesin-like protein CHO1/MKLP-1. J Cell Sci. 2004;117(Pt 15):3233–46.CrossRefPubMed Liu X, et al. Molecular interactions of Polo-like-kinase 1 with the mitotic kinesin-like protein CHO1/MKLP-1. J Cell Sci. 2004;117(Pt 15):3233–46.CrossRefPubMed
8.
go back to reference Takahashi S, et al. Downregulation of KIF23 suppresses glioma proliferation. J Neurooncol. 2012;106(3):519–29.CrossRefPubMed Takahashi S, et al. Downregulation of KIF23 suppresses glioma proliferation. J Neurooncol. 2012;106(3):519–29.CrossRefPubMed
9.
go back to reference Li T, et al. Methylation-mediated repression of MiR-424/503 cluster promotes proliferation and migration of ovarian cancer cells through targeting the hub gene KIF23. Cell Cycle. 2019;18(14):1601–18.CrossRefPubMedPubMedCentral Li T, et al. Methylation-mediated repression of MiR-424/503 cluster promotes proliferation and migration of ovarian cancer cells through targeting the hub gene KIF23. Cell Cycle. 2019;18(14):1601–18.CrossRefPubMedPubMedCentral
10.
11.
12.
go back to reference Gao CT, et al. KIF23 enhances cell proliferation in pancreatic ductal adenocarcinoma and is a potent therapeutic target. Ann Transl Med. 2020;8(21):1394.CrossRefPubMedPubMedCentral Gao CT, et al. KIF23 enhances cell proliferation in pancreatic ductal adenocarcinoma and is a potent therapeutic target. Ann Transl Med. 2020;8(21):1394.CrossRefPubMedPubMedCentral
13.
go back to reference Ji Z, et al. Aberrant KIF23 expression is associated with adverse clinical outcome and promotes cellular malignant behavior through the Wnt/beta-catenin signaling pathway in colorectal cancer. J Cancer. 2021;12(7):2030–40.CrossRefPubMedPubMedCentral Ji Z, et al. Aberrant KIF23 expression is associated with adverse clinical outcome and promotes cellular malignant behavior through the Wnt/beta-catenin signaling pathway in colorectal cancer. J Cancer. 2021;12(7):2030–40.CrossRefPubMedPubMedCentral
15.
go back to reference Pande S, Raisuddin S. Molecular and cellular regulatory roles of sirtuin protein. Crit Rev Food Sci Nutr. 2023;63(29):9895–913. Pande S, Raisuddin S. Molecular and cellular regulatory roles of sirtuin protein. Crit Rev Food Sci Nutr. 2023;63(29):9895–913.
17.
go back to reference Mu P, et al. Sirtuin 7 promotes glioma proliferation and invasion through activation of the ERK/STAT3 signaling pathway. Oncol Lett. 2019;17(2):1445–52.PubMed Mu P, et al. Sirtuin 7 promotes glioma proliferation and invasion through activation of the ERK/STAT3 signaling pathway. Oncol Lett. 2019;17(2):1445–52.PubMed
18.
go back to reference De Nigris F, et al. Isolation of a SIR-like gene, SIR-T8, that is overexpressed in thyroid carcinoma cell lines and tissues. Br J Cancer. 2002;87(12):1479.CrossRefPubMedPubMedCentral De Nigris F, et al. Isolation of a SIR-like gene, SIR-T8, that is overexpressed in thyroid carcinoma cell lines and tissues. Br J Cancer. 2002;87(12):1479.CrossRefPubMedPubMedCentral
19.
go back to reference Mu R, et al. Role of succinylation modification in thyroid cancer and breast cancer. Am J Cancer Res. 2021;11(10):4683–99.PubMedPubMedCentral Mu R, et al. Role of succinylation modification in thyroid cancer and breast cancer. Am J Cancer Res. 2021;11(10):4683–99.PubMedPubMedCentral
20.
go back to reference Alleyn M, et al. The dawn of succinylation: a posttranslational modification. Am J Physiol Cell Physiol. 2018;314(2):C228-232.CrossRefPubMed Alleyn M, et al. The dawn of succinylation: a posttranslational modification. Am J Physiol Cell Physiol. 2018;314(2):C228-232.CrossRefPubMed
22.
go back to reference Broekhuis JM, et al. Posttranslational modifications in thyroid cancer: implications for pathogenesis, diagnosis, classification, and treatment. Cancers (Basel). 2022;14(7):1610.CrossRefPubMed Broekhuis JM, et al. Posttranslational modifications in thyroid cancer: implications for pathogenesis, diagnosis, classification, and treatment. Cancers (Basel). 2022;14(7):1610.CrossRefPubMed
23.
go back to reference Jin C, et al. Acetyltransferase NAT10 regulates the Wnt/beta-catenin signaling pathway to promote colorectal cancer progression via Ac(4)C acetylation of KIF23 mRNA. J Exp Clin Cancer Res. 2022;41(1):345.MathSciNetCrossRefPubMedPubMedCentral Jin C, et al. Acetyltransferase NAT10 regulates the Wnt/beta-catenin signaling pathway to promote colorectal cancer progression via Ac(4)C acetylation of KIF23 mRNA. J Exp Clin Cancer Res. 2022;41(1):345.MathSciNetCrossRefPubMedPubMedCentral
24.
go back to reference Yang HB, et al. Biomarker identification of thyroid associated ophthalmopathy using microarray data. Int J Ophthalmol. 2018;11(9):1482–8.PubMedPubMedCentral Yang HB, et al. Biomarker identification of thyroid associated ophthalmopathy using microarray data. Int J Ophthalmol. 2018;11(9):1482–8.PubMedPubMedCentral
25.
go back to reference Li Z, et al. Kinesin family member 23, regulated by FOXM1, promotes triple negative breast cancer progression via activating Wnt/beta-catenin pathway. J Exp Clin Cancer Res. 2022;41(1):168.CrossRefPubMedPubMedCentralADS Li Z, et al. Kinesin family member 23, regulated by FOXM1, promotes triple negative breast cancer progression via activating Wnt/beta-catenin pathway. J Exp Clin Cancer Res. 2022;41(1):168.CrossRefPubMedPubMedCentralADS
26.
go back to reference Bai X, et al. Systematic pan-cancer analysis of KIF23 and a prediction model based on KIF23 in clear cell renal cell carcinoma (ccRCC). Pharmgenomics Pers Med. 2021;14:1717–29.PubMedPubMedCentral Bai X, et al. Systematic pan-cancer analysis of KIF23 and a prediction model based on KIF23 in clear cell renal cell carcinoma (ccRCC). Pharmgenomics Pers Med. 2021;14:1717–29.PubMedPubMedCentral
27.
go back to reference Cheng C, et al. KIF14 and KIF23 promote cell proliferation and Chemoresistance in HCC Cells, and predict worse prognosis of patients with HCC. Cancer Manag Res. 2020;12:13241–57.CrossRefPubMedPubMedCentralADS Cheng C, et al. KIF14 and KIF23 promote cell proliferation and Chemoresistance in HCC Cells, and predict worse prognosis of patients with HCC. Cancer Manag Res. 2020;12:13241–57.CrossRefPubMedPubMedCentralADS
28.
go back to reference Zhao Y, et al. Identification of the hub genes associated with the prognosis of ovarian cancer patients via Integrated bioinformatics analysis and experimental validation. Cancer Manag Res. 2021;13:707–21.CrossRefPubMedPubMedCentralADS Zhao Y, et al. Identification of the hub genes associated with the prognosis of ovarian cancer patients via Integrated bioinformatics analysis and experimental validation. Cancer Manag Res. 2021;13:707–21.CrossRefPubMedPubMedCentralADS
29.
30.
go back to reference Wang C, et al. CPT1A-mediated succinylation of S100A10 increases human gastric cancer invasion. J Cell Mol Med. 2019;23(1):293–305.CrossRefPubMed Wang C, et al. CPT1A-mediated succinylation of S100A10 increases human gastric cancer invasion. J Cell Mol Med. 2019;23(1):293–305.CrossRefPubMed
31.
go back to reference Fesquet D, et al. Binding of Kif23-iso1/CHO1 to 14-3-3 is regulated by sequential phosphorylations at two LATS kinase consensus sites. PLoS One. 2015;10(2):e0117857.CrossRefPubMedPubMedCentral Fesquet D, et al. Binding of Kif23-iso1/CHO1 to 14-3-3 is regulated by sequential phosphorylations at two LATS kinase consensus sites. PLoS One. 2015;10(2):e0117857.CrossRefPubMedPubMedCentral
32.
go back to reference Pu Y, et al. Transcriptome and differential methylation integration analysis identified important differential methylation annotation genes and functional epigenetic modules related to Vitiligo. Front Immunol. 2021;12:587440.CrossRefPubMedPubMedCentral Pu Y, et al. Transcriptome and differential methylation integration analysis identified important differential methylation annotation genes and functional epigenetic modules related to Vitiligo. Front Immunol. 2021;12:587440.CrossRefPubMedPubMedCentral
Metadata
Title
SIRT7 promotes the proliferation and migration of anaplastic thyroid cancer cells by regulating the desuccinylation of KIF23
Authors
Yongkang Wu
Weijie Chen
Huilai Miao
Tuo 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/s12885-024-11965-9

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