Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2024

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

TrkA promotes MDM2-mediated AGPS ubiquitination and degradation to trigger prostate cancer progression

Authors: Yu Zhang, Zhenlin Huang, Keqiang Li, Guoqing Xie, Yuankang Feng, Zihao Wang, Ningyang Li, Ruoyang Liu, Yinghui Ding, Jun Wang, Jinjian Yang, Zhankui Jia

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2024

Login to get access

Abstract

Background

As a novel necrosis manner, ferroptosis has been increasingly reported to play a role in tumor progression and treatment, however, the specific mechanisms underlying its development in prostate cancer remain unclear. Growing evidence showed that peroxisome plays a key role in ferroptosis. Herein, we identified a novel mechanism for the involvement of ferroptosis in prostate cancer progression, which may provide a new strategy for clinical treatment of prostate cancer.

Methods

Label-Free Mass spectrometry was used to screen and identify candidate proteins after ferroptosis inducer-ML210 treatment. Immunohistochemistry was undertaken to explore the protein expression of AGPS in prostate cancer tissues compared with normal tissues. Co-immunoprecipitation and GST pull-down were used to identify the directly binding of AGPS to MDM2 in vivo and in vitro. CCK8 assay and colony formation assay were used to illustrate the key role of AGPS in the progression of prostate cancer in vitro. The xenograft model was established to verify the key role of AGPS in the progression of prostate cancer in vivo.

Results

AGPS protein expression was downregulated in prostate cancer tissues compared with normal tissues from the first affiliated hospital of Zhengzhou University dataset. Lower expression was correlated with poorer overall survival of patients compared to those with high expression of AGPS. In addition, AGPS can promote ferroptosis by modulating the function of peroxisome-resulting in the lower survival of prostate cancer cells. Furthermore, it was shown that AGPS can be ubiquitinated and degraded by the E3 ligase-MDM2 through the proteasomal pathway. Meanwhile, kinase TrkA can promote the combination of AGPS and MDM2 by phosphorylating AGPS at Y451 site. It was verified that kinase TrkA inhibitor—Larotrectinib can increase the susceptibility of prostate cancer cells to ferroptosis, which leads to the inhibition of prostate cancer proliferation to a great extent in vitro and in vivo.

Conclusion

Based on these findings, we proposed the combination of ferroptosis inducer and TrkA inhibitor to synergistically exert anti-tumor effects, which may provide a new strategy for the clinical treatment of prostate cancer.
Appendix
Available only for authorised users
Literature
1.
go back to reference Clarke N, Wiechno P, Alekseev B, Sala N, Jones R, Kocak I, et al. Olaparib combined with abiraterone in patients with metastatic castration-resistant Prostate cancer: a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Oncol. 2018;19(7):975–86.CrossRefPubMed Clarke N, Wiechno P, Alekseev B, Sala N, Jones R, Kocak I, et al. Olaparib combined with abiraterone in patients with metastatic castration-resistant Prostate cancer: a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Oncol. 2018;19(7):975–86.CrossRefPubMed
2.
go back to reference Antonarakis ES, Piulats JM, Gross-Goupil M, Goh J, Ojamaa K, Hoimes CJ, et al. Pembrolizumab for treatment-refractory metastatic castration-resistant Prostate Cancer: Multicohort, open-label phase II KEYNOTE-199 study. J Clin Oncol. 2020;38(5):395–405.CrossRefPubMed Antonarakis ES, Piulats JM, Gross-Goupil M, Goh J, Ojamaa K, Hoimes CJ, et al. Pembrolizumab for treatment-refractory metastatic castration-resistant Prostate Cancer: Multicohort, open-label phase II KEYNOTE-199 study. J Clin Oncol. 2020;38(5):395–405.CrossRefPubMed
3.
go back to reference Ng K, Smith S, Shamash J. Metastatic hormone-sensitive Prostate Cancer (mHSPC): advances and treatment strategies in the First-Line setting. Oncol Therapy. 2020;8(2):209–30.CrossRef Ng K, Smith S, Shamash J. Metastatic hormone-sensitive Prostate Cancer (mHSPC): advances and treatment strategies in the First-Line setting. Oncol Therapy. 2020;8(2):209–30.CrossRef
4.
go back to reference Shao YH, Demissie K, Shih W, Mehta AR, Stein MN, Roberts CB, et al. Contemporary risk profile of Prostate cancer in the United States. J Natl Cancer Inst. 2009;101(18):1280–3.CrossRefPubMedPubMedCentral Shao YH, Demissie K, Shih W, Mehta AR, Stein MN, Roberts CB, et al. Contemporary risk profile of Prostate cancer in the United States. J Natl Cancer Inst. 2009;101(18):1280–3.CrossRefPubMedPubMedCentral
5.
go back to reference Singer EA, Golijanin DJ, Miyamoto H, Messing EM. Androgen deprivation therapy for Prostate cancer. Expert Opin Pharmacother. 2008;9(2):211–28.CrossRefPubMed Singer EA, Golijanin DJ, Miyamoto H, Messing EM. Androgen deprivation therapy for Prostate cancer. Expert Opin Pharmacother. 2008;9(2):211–28.CrossRefPubMed
6.
go back to reference Voog JC, Smith MR, Efstathiou JA. Radiation with or without androgen deprivation therapy for localized Prostate Cancer. JAMA. 2016;315(10):1054–5.CrossRefPubMed Voog JC, Smith MR, Efstathiou JA. Radiation with or without androgen deprivation therapy for localized Prostate Cancer. JAMA. 2016;315(10):1054–5.CrossRefPubMed
7.
go back to reference Chi KN, Bjartell A, Dearnaley D, Saad F, Schroder FH, Sternberg C, et al. Castration-resistant Prostate cancer: from new pathophysiology to new treatment targets. Eur Urol. 2009;56(4):594–605.CrossRefPubMed Chi KN, Bjartell A, Dearnaley D, Saad F, Schroder FH, Sternberg C, et al. Castration-resistant Prostate cancer: from new pathophysiology to new treatment targets. Eur Urol. 2009;56(4):594–605.CrossRefPubMed
8.
go back to reference Wülfing C, Bgemann M, Goebell PJ, Hammerer P, Schostak M. Erratum zu: Therapiesituation Beim metastasierten kastrationsnaiven Prostatakarzinom (mCNPC) und die Auswirkungen Im Klinischen Alltag. Urologe. 2019;58(8):924.CrossRef Wülfing C, Bgemann M, Goebell PJ, Hammerer P, Schostak M. Erratum zu: Therapiesituation Beim metastasierten kastrationsnaiven Prostatakarzinom (mCNPC) und die Auswirkungen Im Klinischen Alltag. Urologe. 2019;58(8):924.CrossRef
9.
go back to reference Stockwell BR, Friedmann Angeli JP, Bayir H, Bush AI, Conrad M, Dixon SJ, et al. Ferroptosis: a regulated cell death Nexus linking metabolism, Redox Biology, and Disease. Cell. 2017;171(2):273–85.CrossRefPubMedPubMedCentral Stockwell BR, Friedmann Angeli JP, Bayir H, Bush AI, Conrad M, Dixon SJ, et al. Ferroptosis: a regulated cell death Nexus linking metabolism, Redox Biology, and Disease. Cell. 2017;171(2):273–85.CrossRefPubMedPubMedCentral
10.
go back to reference Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell. 2012;149(5):1060–72.CrossRefPubMedPubMedCentral Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell. 2012;149(5):1060–72.CrossRefPubMedPubMedCentral
11.
go back to reference Zou Y, Henry WS, Ricq EL, Graham ET, Schreiber SL. Plasticity of ether lipids promotes ferroptosis susceptibility and evasion. Nature. 2020;585(7826):1–6.CrossRef Zou Y, Henry WS, Ricq EL, Graham ET, Schreiber SL. Plasticity of ether lipids promotes ferroptosis susceptibility and evasion. Nature. 2020;585(7826):1–6.CrossRef
14.
go back to reference Cui W, Liu D, Gu W, Chu B. Peroxisome-driven ether-linked phospholipids biosynthesis is essential for ferroptosis. Cell Death Differ. 2021;28(8):2536–51.CrossRefPubMedPubMedCentral Cui W, Liu D, Gu W, Chu B. Peroxisome-driven ether-linked phospholipids biosynthesis is essential for ferroptosis. Cell Death Differ. 2021;28(8):2536–51.CrossRefPubMedPubMedCentral
15.
go back to reference Feng FY, Zhang Y, Kothari V, Evans JR, Jackson WC, Chen W, et al. MDM2 inhibition sensitizes Prostate Cancer cells to androgen ablation and Radiotherapy in a p53-Dependent manner. Neoplasia. 2016;18(4):213–22.CrossRefPubMedPubMedCentral Feng FY, Zhang Y, Kothari V, Evans JR, Jackson WC, Chen W, et al. MDM2 inhibition sensitizes Prostate Cancer cells to androgen ablation and Radiotherapy in a p53-Dependent manner. Neoplasia. 2016;18(4):213–22.CrossRefPubMedPubMedCentral
16.
go back to reference Benjamin DI, Cozzo A, Ji X, Roberts LS, Louie SM, Mulvihill MM, et al. Ether lipid generating enzyme AGPS alters the balance of structural and signaling lipids to fuel cancer pathogenicity. Proc Natl Acad Sci U S A. 2013;110(37):14912–7.CrossRefPubMedPubMedCentral Benjamin DI, Cozzo A, Ji X, Roberts LS, Louie SM, Mulvihill MM, et al. Ether lipid generating enzyme AGPS alters the balance of structural and signaling lipids to fuel cancer pathogenicity. Proc Natl Acad Sci U S A. 2013;110(37):14912–7.CrossRefPubMedPubMedCentral
18.
go back to reference Yang WS, Sriramaratnam R, Welsch ME, Shimada K, Skouta R, Viswanathan VS, et al. Regulation of Ferroptotic Cancer Cell death by GPX4. Cell. 2014;156(1–2):317–31.CrossRefPubMedPubMedCentral Yang WS, Sriramaratnam R, Welsch ME, Shimada K, Skouta R, Viswanathan VS, et al. Regulation of Ferroptotic Cancer Cell death by GPX4. Cell. 2014;156(1–2):317–31.CrossRefPubMedPubMedCentral
19.
go back to reference Koppula P, Zhuang L, Gan B. Cystine transporter SLC7A11/xCT in cancer: ferroptosis, nutrient dependency, and cancer therapy. Protein Cell. 2021;12(8):599–620.CrossRefPubMed Koppula P, Zhuang L, Gan B. Cystine transporter SLC7A11/xCT in cancer: ferroptosis, nutrient dependency, and cancer therapy. Protein Cell. 2021;12(8):599–620.CrossRefPubMed
20.
go back to reference Cui Y, Zhang Y, Zhao X, Shao L, Liu G, Sun C, et al. ACSL4 exacerbates ischemic Stroke by promoting ferroptosis-induced brain injury and neuroinflammation. Brain Behav Immun. 2021;93:312–21.CrossRefPubMed Cui Y, Zhang Y, Zhao X, Shao L, Liu G, Sun C, et al. ACSL4 exacerbates ischemic Stroke by promoting ferroptosis-induced brain injury and neuroinflammation. Brain Behav Immun. 2021;93:312–21.CrossRefPubMed
21.
go back to reference Lin HK, Wang L, Hu YC, Altuwaijri S, Chang C. Phosphorylation-dependent ubiquitylation and degradation of androgen receptor by akt require Mdm2 E3 ligase. EMBO J. 2002;21(15):4037–48.CrossRefPubMedPubMedCentral Lin HK, Wang L, Hu YC, Altuwaijri S, Chang C. Phosphorylation-dependent ubiquitylation and degradation of androgen receptor by akt require Mdm2 E3 ligase. EMBO J. 2002;21(15):4037–48.CrossRefPubMedPubMedCentral
22.
go back to reference Festuccia C, Gravina GL, Muzi P, Pomante R, Ventura L, Ricevuto E, et al. In vitro and in vivo effects of bicalutamide on the expression of TrkA and P75 neurotrophin receptors in prostate carcinoma. Prostate. 2007;67(12):1255–64.CrossRefPubMed Festuccia C, Gravina GL, Muzi P, Pomante R, Ventura L, Ricevuto E, et al. In vitro and in vivo effects of bicalutamide on the expression of TrkA and P75 neurotrophin receptors in prostate carcinoma. Prostate. 2007;67(12):1255–64.CrossRefPubMed
23.
go back to reference Pierotti MA, Greco A. Oncogenic rearrangements of the NTRK1/NGF receptor. Cancer Lett. 2006;232(1):90–8.CrossRefPubMed Pierotti MA, Greco A. Oncogenic rearrangements of the NTRK1/NGF receptor. Cancer Lett. 2006;232(1):90–8.CrossRefPubMed
24.
go back to reference Ghoochani A, Hsu EC, Aslan M, Rice MA, Nguyen HM, Brooks JD, et al. Ferroptosis Inducers are a Novel Therapeutic Approach for Advanced Prostate Cancer. Cancer Res. 2021;81(6):1583–94.CrossRefPubMedPubMedCentral Ghoochani A, Hsu EC, Aslan M, Rice MA, Nguyen HM, Brooks JD, et al. Ferroptosis Inducers are a Novel Therapeutic Approach for Advanced Prostate Cancer. Cancer Res. 2021;81(6):1583–94.CrossRefPubMedPubMedCentral
25.
go back to reference Zhu Y, Zhu L, Lu L, Zhang L, Zhang G, Wang Q, et al. Role and mechanism of the alkylglycerone phosphate synthase in suppressing the invasion potential of human glioma and hepatic carcinoma cells in vitro. Oncol Rep. 2014;32(1):431–6.CrossRefPubMed Zhu Y, Zhu L, Lu L, Zhang L, Zhang G, Wang Q, et al. Role and mechanism of the alkylglycerone phosphate synthase in suppressing the invasion potential of human glioma and hepatic carcinoma cells in vitro. Oncol Rep. 2014;32(1):431–6.CrossRefPubMed
26.
go back to reference Itzkovitz B, Jiralerspong S, Nimmo G, Loscalzo M, Horovitz DD, Snowden A, et al. Functional characterization of novel mutations in GNPAT and AGPS, causing rhizomelic chondrodysplasia punctata (RCDP) types 2 and 3. Hum Mutat. 2012;33(1):189–97.CrossRefPubMed Itzkovitz B, Jiralerspong S, Nimmo G, Loscalzo M, Horovitz DD, Snowden A, et al. Functional characterization of novel mutations in GNPAT and AGPS, causing rhizomelic chondrodysplasia punctata (RCDP) types 2 and 3. Hum Mutat. 2012;33(1):189–97.CrossRefPubMed
27.
go back to reference Hangauer MJ, Viswanathan VS, Ryan MJ, Bole D, Eaton JK, Matov A, et al. Drug-tolerant persister cancer cells are vulnerable to GPX4 inhibition. Nature. 2017;551(7679):247.CrossRefPubMedPubMedCentral Hangauer MJ, Viswanathan VS, Ryan MJ, Bole D, Eaton JK, Matov A, et al. Drug-tolerant persister cancer cells are vulnerable to GPX4 inhibition. Nature. 2017;551(7679):247.CrossRefPubMedPubMedCentral
28.
go back to reference Eaton JK, Furst L, Ruberto RA, Moosmayer D, Hilpmann A, Ryan MJ, et al. Selective covalent targeting of GPX4 using masked nitrile-oxide electrophiles. Nat Chem Biol. 2020;16(5):497–506.CrossRefPubMedPubMedCentral Eaton JK, Furst L, Ruberto RA, Moosmayer D, Hilpmann A, Ryan MJ, et al. Selective covalent targeting of GPX4 using masked nitrile-oxide electrophiles. Nat Chem Biol. 2020;16(5):497–506.CrossRefPubMedPubMedCentral
29.
go back to reference Laetsch TW, DuBois SG, Mascarenhas L, Turpin B, Federman N, Albert CM, et al. Larotrectinib for paediatric solid tumours harbouring NTRK gene fusions: phase 1 results from a multicentre, open-label, phase 1/2 study. Lancet Oncol. 2018;19(5):705–14.CrossRefPubMedPubMedCentral Laetsch TW, DuBois SG, Mascarenhas L, Turpin B, Federman N, Albert CM, et al. Larotrectinib for paediatric solid tumours harbouring NTRK gene fusions: phase 1 results from a multicentre, open-label, phase 1/2 study. Lancet Oncol. 2018;19(5):705–14.CrossRefPubMedPubMedCentral
30.
go back to reference Jiang L, Kon N, Li T, Wang SJ, Su T, Hibshoosh H, et al. Ferroptosis as a p53-mediated activity during tumour suppression. Nature. 2015;520(7545):57–62.CrossRefPubMedPubMedCentral Jiang L, Kon N, Li T, Wang SJ, Su T, Hibshoosh H, et al. Ferroptosis as a p53-mediated activity during tumour suppression. Nature. 2015;520(7545):57–62.CrossRefPubMedPubMedCentral
31.
go back to reference Li Y, Cao Y, Xiao J, Shang J, Tan Q, Ping F, et al. Inhibitor of apoptosis-stimulating protein of p53 inhibits ferroptosis and alleviates intestinal ischemia/reperfusion-induced acute lung injury. Cell Death Differ. 2020;27(9):2635–50.CrossRefPubMedPubMedCentral Li Y, Cao Y, Xiao J, Shang J, Tan Q, Ping F, et al. Inhibitor of apoptosis-stimulating protein of p53 inhibits ferroptosis and alleviates intestinal ischemia/reperfusion-induced acute lung injury. Cell Death Differ. 2020;27(9):2635–50.CrossRefPubMedPubMedCentral
32.
33.
go back to reference Klein AM, de Queiroz RM, Venkatesh D, Prives C. The roles and regulation of MDM2 and MDMX: it is not just about p53. Genes Dev. 2021;35(9–10):575–601.CrossRefPubMedPubMedCentral Klein AM, de Queiroz RM, Venkatesh D, Prives C. The roles and regulation of MDM2 and MDMX: it is not just about p53. Genes Dev. 2021;35(9–10):575–601.CrossRefPubMedPubMedCentral
35.
Metadata
Title
TrkA promotes MDM2-mediated AGPS ubiquitination and degradation to trigger prostate cancer progression
Authors
Yu Zhang
Zhenlin Huang
Keqiang Li
Guoqing Xie
Yuankang Feng
Zihao Wang
Ningyang Li
Ruoyang Liu
Yinghui Ding
Jun Wang
Jinjian Yang
Zhankui Jia
Publication date
01-12-2024
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2024
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-023-02920-w

Other articles of this Issue 1/2024

Journal of Experimental & Clinical Cancer Research 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