AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (3.5 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Original Article | Open Access

Loss of NEIL3 activates radiotherapy resistance in the progression of prostate cancer

Qiong Wang1,2,3,*Zean Li1,3,*Jin Yang3,4Shirong Peng1,3Qianghua Zhou1,3Kai Yao5Wenli Cai6Zhongqiu Xie2Fujun Qin2Hui Li2Xu Chen1,3Kaiwen Li1,3Hai Huang1,3,7
Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
Department of Pathology, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
Department of Radiation Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
Department of Urology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan 511518, China

*These authors contributed equally to this work.

Show Author Information

Abstract

Objective

To explore the genetic changes in the progression of castration-resistant prostate cancer (CRPC) and neuroendocrine prostate cancer (NEPC) and the reason why these cancers resist existing therapies.

Methods

We employed our CRPC cell line microarray and other CRPC or NEPC datasets to screen the target gene NEIL3. Lentiviral transfection and RNA interference were used to construct overexpression and knockdown cell lines. Cell and animal models of radiotherapy were established by using a medical electron linear accelerator. Flow cytometry was used to detect apoptosis or cell cycle progression. Western blot and qPCR were used to detect changes in the protein and RNA levels.

Results

TCGA and clinical patient datasets indicated that NEIL3 was downregulated in CRPC and NEPC cell lines, and NEIL3 was correlated with a high Gleason score but a good prognosis. Further functional studies demonstrated that NEIL3 had no effect on the proliferation and migration of PCa cells. However, cell and animal radiotherapy models revealed that NEIL3 could facilitate the radiotherapy sensitivity of PCa cells, while loss of NEIL3 activated radiotherapy resistance. Mechanistically, we found that NEIL3 negatively regulated the expression of ATR, and higher NEIL3 expression repressed the ATR/CHK1 pathway, thus regulating the cell cycle.

Conclusions

We demonstrated that NEIL3 may serve as a diagnostic or therapeutic target for therapy-resistant patients.

Electronic Supplementary Material

Download File(s)
cbm-19-8-1193_ESM.pdf (655.3 KB)

References

1

Watson PA, Arora VK, Sawyers CL. Emerging mechanisms of resistance to androgen receptor inhibitors in prostate cancer. Nat Rev Cancer. 2015; 15: 701-11.

2

Chandrasekar T, Yang JC, Gao AC, Evans CP. Mechanisms of resistance in castration-resistant prostate cancer (crpc). Transl Androl Urol. 2015; 4: 365-80.

3

Zhao N, Peacock SO, Lo CH, Heidman LM, Rice MA, Fahrenholtz CD, et al. Arginine vasopressin receptor 1a is a therapeutic target for castration-resistant prostate cancer. Sci Transl Med. 2019; 11: eaaw4636.

4

Wang Q, Li W, Zhang Y, Yuan X, Xu K, Yu J, et al. Androgen receptor regulates a distinct transcription program in androgen-independent prostate cancer. Cell. 2009; 138: 245-56.

5

Cornford P, Bellmunt J, Bolla M, Briers E, De Santis M, Gross T, et al. Eau-estro-siog guidelines on prostate cancer. Part Ⅱ: treatment of relapsing, metastatic, and castration-resistant prostate cancer. Eur Urol. 2017; 71: 630-42.

6

Francini E, Gray KP, Shaw GK, Evan CP, Hamid AA, Perry CE, et al. Impact of new systemic therapies on overall survival of patients with metastatic castration-resistant prostate cancer in a hospital-based registry. Prostate Cancer Prostatic Dis. 2019; 22: 420-7.

7

Scher HI, Fizazi K, Saad F, Taplin ME, Sternberg CN, Miller K, et al. Increased survival with enzalutamide in prostate cancer after chemotherapy. N Engl J Med. 2012; 367: 1187-97.

8

Ryan CJ, Smith MR, de Bono JS, Molina A, Logothetis CJ, de Souza P, et al. Abiraterone in metastatic prostate cancer without previous chemotherapy. N Engl J Med. 2013; 368: 138-48.

9

Aparicio A, Logothetis CJ, Maity SN. Understanding the lethal variant of prostate cancer: power of examining extremes. Cancer Discov. 2011; 1: 466-8.

10

Wang W, Epstein JI. Small cell carcinoma of the prostate. A morphologic and immunohistochemical study of 95 cases. Am J Surg Pathol. 2008; 32: 65-71.

11

Nadal R, Schweizer M, Kryvenko ON, Epstein JI, Eisenberger MA. Small cell carcinoma of the prostate. Nat Rev Urol. 2014; 11: 213-9.

12

Komura K, Jeong SH, Hinohara K, Qu F, Wang X, Hiraki M, et al. Resistance to docetaxel in prostate cancer is associated with androgen receptor activation and loss of kdm5d expression. Proc Natl Acad Sci U S A. 2016; 113: 6259-64.

13

Pignot G, Maillet D, Gross E, Barthelemy P, Beauval JB, ConstansSchlurmann F, et al. Systemic treatments for high-risk localized prostate cancer. Nat Rev Urol. 2018; 15: 498-510.

14

Carceles-Cordon M, Kelly WK, Gomella L, Knudsen KE, Rodriguez-Bravo V, Domingo-Domenech J. Cellular rewiring in lethal prostate cancer: The architect of drug resistance. Nat Rev Urol. 2020; 17: 292-307.

15

Gu P, Chen X, Xie R, Han J, Xie W, Wang B, et al. Lncrna hoxd-as1 regulates proliferation and chemo-resistance of castration-resistant prostate cancer via recruiting wdr5. Mol Ther. 2017; 25: 1959-73.

16

Bandaru V, Sunkara S, Wallace SS, Bond JP. A novel human DNA glycosylase that removes oxidative DNA damage and is homologous to escherichia coli endonuclease viii. DNA Repair (Amst). 2002; 1: 517-29.

17

Semlow DR, Zhang J, Budzowska M, Drohat AC, Walter JC. Replication-dependent unhooking of DNA interstrand cross-links by the neil3 glycosylase. Cell. 2016; 167: 498-511.

18

Li N, Wang J, Wallace SS, Chen J, Zhou J, D’Andrea AD. Cooperation of the neil3 and fanconi anemia/brca pathways in interstrand crosslink repair. Nucleic Acids Res. 2020; 48: 3014-28.

19

Olsen MB, Hildrestrand GA, Scheffler K, Vinge LE, Alfsnes K, Palibrk V, et al. Neil3-dependent regulation of cardiac fibroblast proliferation prevents myocardial rupture. Cell Rep. 2017; 18: 82-92.

20

Yadav S, Anbalagan M, Baddoo M, Chellamuthu VK, Mukhopadhyay S, Woods C, et al. Somatic mutations in the DNA repairome in prostate cancers in african americans and caucasians. Oncogene. 2020; 39: 4299-311.

21

Kim YS, Kim Y, Choi JW, Oh HE, Lee JH. Genetic variants and risk of prostate cancer using pathway analysis of a genome-wide association study. Neoplasma. 2016; 63: 629-34.

22

Beltran H, Rickman DS, Park K, Chae SS, Sboner A, MacDonald TY, et al. Molecular characterization of neuroendocrine prostate cancer and identification of new drug targets. Cancer Discov. 2011; 1: 487-95.

23

Yadav SS, Li J, Stockert JA, Herzog B, O’Connor J, Garzon-Manco L, et al. Induction of neuroendocrine differentiation in prostate cancer cells by dovitinib (tki-258) and its therapeutic implications. Transl Oncol. 2017; 10: 357-66.

24

Gu P, Chen X, Xie R, Xie W, Huang L, Dong W, et al. A novel ar translational regulator lncrna lbcs inhibits castration resistance of prostate cancer. Mol Cancer. 2019; 18: 109.

25

Xie M, Vesuna F, Tantravedi S, Bol GM, Heerma van Voss MR, Nugent K, et al. Rk-33 radiosensitizes prostate cancer cells by blocking the rna helicase ddx3. Cancer Res. 2016; 76: 6340-50.

26

El Bezawy R, Cominetti D, Fenderico N, Zuco V, Beretta GL, Dugo M, et al. Mir-875-5p counteracts epithelial-to-mesenchymal transition and enhances radiation response in prostate cancer through repression of the egfr-zeb1 axis. Cancer Lett. 2017; 395: 53-62.

27

Pignatta S, Arienti C, Zoli W, Di Donato M, Castoria G, Gabucci E, et al. Prolonged exposure to (r)-bicalutamide generates a lncap subclone with alteration of mitochondrial genome. Mol Cell Endocrinol. 2014; 382: 314-24.

28

Xu P, Zhou Z, Xiong M, Zou W, Deng X, Ganaie SS, et al. Parvovirus b19 ns1 protein induces cell cycle arrest at g2-phase by activating the atr-cdc25c-cdk1 pathway. PLoS Pathog. 2017; 13: e1006266.

29

Ding L, Madamsetty VS, Kiers S, Alekhina O, Ugolkov A, Dube J, et al. Glycogen synthase kinase-3 inhibition sensitizes pancreatic cancer cells to chemotherapy by abrogating the topbp1/atr-mediated DNA damage response. Clin Cancer Res. 2019; 25: 6452-62.

30

Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin. 2018; 68: 7-30.

31

Rohrer Bley C, Furmanova P, Orlowski K, Grosse N, Broggini-Tenzer A, McSheehy PM, et al. Microtubule stabilising agents and ionising radiation: Multiple exploitable mechanisms for combined treatment. Eur J Cancer. 2013; 49: 245-53.

32

Maity A, McKenna WG, Muschel RJ. The molecular basis for cell cycle delays following ionizing radiation: a review. Radiother Oncol. 1994; 31: 1-13.

33

Wallace BD, Berman Z, Mueller GA, Lin Y, Chang T, Andres SN, et al. Ape2 zf-grf facilitates 3’-5’ resection of DNA damage following oxidative stress. Proc Natl Acad Sci U S A. 2017; 114: 304-9.

34

Klattenhoff AW, Thakur M, Chu CS, Ray D, Habib SL, Kidane D. Loss of neil3 DNA glycosylase markedly increases replication associated double strand breaks and enhances sensitivity to atr inhibitor in glioblastoma cells. Oncotarget. 2017; 8: 112942-58.

35

Shinmura K, Kato H, Kawanishi Y, Igarashi H, Goto M, Tao H, et al. Abnormal expressions of DNA glycosylase genes neil1, neil2, and neil3 are associated with somatic mutation loads in human cancer. Oxid Med Cell Longev. 2016; 2016: 1546392.

36

Massaad MJ, Zhou J, Tsuchimoto D, Chou J, Jabara H, Janssen E, et al. Deficiency of base excision repair enzyme neil3 drives increased predisposition to autoimmunity. J Clin Invest. 2016; 126: 4219-36.

Cancer Biology & Medicine
Pages 1193-1210
Cite this article:
Wang Q, Li Z, Yang J, et al. Loss of NEIL3 activates radiotherapy resistance in the progression of prostate cancer. Cancer Biology & Medicine, 2022, 19(8): 1193-1210. https://doi.org/10.20892/j.issn.2095-3941.2020.0550

42

Views

6

Downloads

13

Crossref

17

Web of Science

16

Scopus

Altmetrics

Received: 13 September 2020
Accepted: 13 January 2021
Published: 29 August 2022
©2022 Cancer Biology & Medicine.

Creative Commons Attribution-NonCommercial 4.0 International License

Return