Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

EGFR-upregulated LIFR promotes SUCLG2-dependent castration resistance and neuroendocrine differentiation of prostate cancer

Abstract

Neuroendocrine (NE) differentiation is a well-recognized phenotypic change of prostate cancer after androgen deprivation therapy (ADT), and it ultimately develops into an aggressive subset of this disease. However, the contribution of signaling pathways that lead to metabolic disorders and NE differentiation of prostate cancer remains unclear. In this study, we identified that ADT induced upregulation of the succinate-CoA ligase GDP-forming beta subunit (SUCLG2), which regulates succinate metabolism and NE differentiation of prostate cancer. We demonstrated a connection that upregulation of epidermal growth factor receptor (EGFR)-leukemia inhibitory factor receptor (LIFR) signaling induced SUCLG2 expression in prostate cancer cells. The LIFR is upregulated by nuclear EGFR, which acts as a transcriptional regulator, directly binds to the LIFR promoter, and drives NE differentiation and glycolysis of prostate cancer. LIFR upregulation is associated with SUCLG2, which increased succinate synthesis and enzymatic activities of mitochondrial nucleoside diphosphate kinase (NDPK) in prostate cancer cells. Knockdown of SUCLG2 suppressed NE differentiation in cultured cells and reduced prostate tumor growth in a xenograft model. Analysis of prostate tissue samples showed increased intensity of nuclear EGFR associated with the LIFR and SUCLG2 in castration-resistant prostate cancer tumors. Our study provides a mechanism whereby ADT upregulates EGFR–LIFR signaling that activates SUCLG2, which subsequently stimulates the metabolic changes associated with NE differentiation and aggressive prostate cancer phenotype.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: LIFR upregulation is involved in the NE differentiation of prostate cancer.
Fig. 2: ADT upregulates the LIFR, which promotes the NE differentiation of prostate cancer.
Fig. 3: Activated EGFR pathway enhances LIFR expression.
Fig. 4: ADT-induced SUCLG2 is regulated by LIF–LIFR–STAT3 signaling.
Fig. 5: Activation of LIF–LIFR signaling promotes SUCLG2-driven mtDNA content and NDPK enzymatic activity in prostate cancer cells.
Fig. 6: Targeting SUCLG2 reduces the tumor growth of AR-negative or ADT-resistant prostate cancer cells.
Fig. 7: Induced SUCLG2 is positively associated with prostate cancer progression.

Similar content being viewed by others

References

  1. Dai C, Heemers H, Sharifi N. Androgen Signaling in Prostate Cancer. Cold Spring Harb Perspect Med 2017;7:a030452.

  2. Sasaki T, Sugimura Y. The importance of time to prostate-specific antigen (PSA) nadir after primary androgen deprivation therapy in hormone-naïve prostate cancer patients. J Clin Med. 2018;7:565.

    Google Scholar 

  3. Choueiri TK, Xie W, D’Amico AV, Ross RW, Hu JC, Pomerantz M, et al. Time to prostate-specific antigen nadir independently predicts overall survival in patients who have metastatic hormone-sensitive prostate cancer treated with androgen-deprivation therapy. Cancer. 2009;115:981–7.

    CAS  Google Scholar 

  4. Heidenreich A, Bastian PJ, Bellmunt J, Bolla M, Joniau S, van der Kwast T, et al. EAU guidelines on prostate cancer. Part II: treatment of advanced, relapsing, and castration-resistant prostate cancer. Eur Urol. 2014;65:467–79.

    CAS  Google Scholar 

  5. Li Q, Deng Q, Chao HP, Liu X, Lu Y, Lin K, et al. Linking prostate cancer cell AR heterogeneity to distinct castration and enzalutamide responses. Nat Commun. 2018;9:3600.

    Google Scholar 

  6. Beltran H, Prandi D, Mosquera JM, Benelli M, Puca L, Cyrta J, et al. Divergent clonal evolution of castration-resistant neuroendocrine prostate cancer. Nat Med. 2016;22:298–305.

    CAS  Google Scholar 

  7. di Sant’Agnese PA. Neuroendocrine differentiation in human prostatic carcinoma. Hum Pathol. 1992;23:287–96.

    Google Scholar 

  8. Mosquera JM, Beltran H, Park K, MacDonald TY, Robinson BD, Tagawa ST, et al. Concurrent AURKA and MYCN gene amplifications are harbingers of lethal treatment-related neuroendocrine prostate cancer. Neoplasia. 2013;15:1–10.

    CAS  Google Scholar 

  9. Williamson SR, Zhang S, Yao JL, Huang J, Lopez-Beltran A, Shen S, et al. ERG-TMPRSS2 rearrangement is shared by concurrent prostatic adenocarcinoma and prostatic small cell carcinoma and absent in small cell carcinoma of the urinary bladder: evidence supporting monoclonal origin. Mod Pathol. 2011;24:1120–7.

    CAS  Google Scholar 

  10. Aggarwal R, Huang J, Alumkal JJ, Zhang L, Feng FY, Thomas GV, et al. Clinical and genomic characterization of treatment-emergent small-cell neuroendocrine prostate cancer: a multi-institutional prospective study. J Clin Oncol. 2018;36:2492–503.

    CAS  Google Scholar 

  11. Huang J, Yao JL, di Sant’Agnese PA, Yang Q, Bourne PA, Na Y. Immunohistochemical characterization of neuroendocrine cells in prostate cancer. Prostate. 2006;66:1399–406.

    CAS  Google Scholar 

  12. Tanaka T, Narazaki M, Kishimoto T. IL-6 in inflammation, immunity, and disease. Cold Spring Harb Perspect Biol. 2014;6:a016295.

    Google Scholar 

  13. Nicola NA, Babon JJ. Leukemia inhibitory factor (LIF). Cytokine Growth Factor Rev. 2015;26:533–44.

    CAS  Google Scholar 

  14. Dziennis S, Alkayed NJ. Role of signal transducer and activator of transcription 3 in neuronal survival and regeneration. Rev Neurosci. 2008;19:341–61.

    CAS  Google Scholar 

  15. Onishi K, Zandstra PW. LIF signaling in stem cells and development. Development. 2015;142:2230–6.

    CAS  Google Scholar 

  16. Wu HX, Cheng X, Jing XQ, Ji XP, Chen XZ, Zhang YQ, et al. LIFR promotes tumor angiogenesis by up-regulating IL-8 levels in colorectal cancer. Biochim Biophys Acta Mol Basis Dis. 2018;1864:2769–84.

    CAS  Google Scholar 

  17. Shao J, Zhu W, Ding Y, Zhu H, Jing X, Yu H, et al. Phosphorylation of LIFR promotes prostate cancer progression by activating the AKT pathway. Cancer Lett. 2019;451:110–21.

    CAS  Google Scholar 

  18. Li M, Sendtner M, Smith A. Essential function of LIF receptor in motor neurons. Nature. 1995;378:724–7.

    CAS  Google Scholar 

  19. Bressy C, Lac S, Nigri J, Leca J, Roques J, Lavaut MN, et al. LIF drives neural remodeling in pancreatic cancer and offers a new candidate biomarker. Cancer Res. 2018;78:909–21.

    CAS  Google Scholar 

  20. Rimawi MF, Shetty PB, Weiss HL, Schiff R, Osborne CK, Chamness GC, et al. Epidermal growth factor receptor expression in breast cancer association with biologic phenotype and clinical outcomes. Cancer. 2010;116:1234–42.

    Google Scholar 

  21. Thomas R, Weihua Z. Rethink of EGFR in cancer with its kinase independent function on board. Front Oncol. 2019;9:800.

    Google Scholar 

  22. Peraldo-Neia C, Migliardi G, Mello-Grand M, Montemurro F, Segir R, Pignochino Y, et al. Epidermal growth factor receptor (EGFR) mutation analysis, gene expression profiling and EGFR protein expression in primary prostate cancer. BMC Cancer. 2011;11:31.

    CAS  Google Scholar 

  23. Day KC, Lorenzatti Hiles G, Kozminsky M, Dawsey SJ, Paul A, Broses LJ, et al. HER2 and EGFR overexpression support metastatic progression of prostate cancer to bone. Cancer Res. 2017;77:74–85.

    CAS  Google Scholar 

  24. Traish AM, Morgentaler A. Epidermal growth factor receptor expression escapes androgen regulation in prostate cancer: a potential molecular switch for tumour growth. Br J Cancer. 2009;101:1949–56.

    CAS  Google Scholar 

  25. Cai C, Portnoy DC, Wang H, Jiang X, Chen S, Balk SP. Androgen receptor expression in prostate cancer cells is suppressed by activation of epidermal growth factor receptor and ErbB2. Cancer Res. 2009;69:5202–9.

    CAS  Google Scholar 

  26. Humez S, Monet M, Legrand G, Lepage G, Delcourt P, Prevarskaya N. Epidermal growth factor-induced neuroendocrine differentiation and apoptotic resistance of androgen-independent human prostate cancer cells. Endocr Relat Cancer. 2006;13:181–95.

    CAS  Google Scholar 

  27. Cortes MA, Cariaga-Martinez AE, Lobo MV, Martin Orozco RM, Motino O, Rodriguez-Ubreva FJ, et al. EGF promotes neuroendocrine-like differentiation of prostate cancer cells in the presence of LY294002 through increased ErbB2 expression independent of the phosphatidylinositol 3-kinase-AKT pathway. Carcinogenesis. 2012;33:1169–77.

    CAS  Google Scholar 

  28. Lin SY, Makino K, Xia W, Matin A, Wen Y, Kwong KY, et al. Nuclear localization of EGF receptor and its potential new role as a transcription factor. Nat Cell Biol. 2001;3:802–8.

    CAS  Google Scholar 

  29. Lo HW, Hsu SC, Ali-Seyed M, Gunduz M, Xia W, Wei Y, et al. Nuclear interaction of EGFR and STAT3 in the activation of the iNOS/NO pathway. Cancer Cell. 2005;7:575–89.

    CAS  Google Scholar 

  30. Read J, Ingram A, Al Saleh HA, Platko K, Gabriel K, Kapoor A, et al. Nuclear transportation of exogenous epidermal growth factor receptor and androgen receptor via extracellular vesicles. Eur J Cancer. 2017;70:62–74.

    CAS  Google Scholar 

  31. Kharmate G, Hosseini-Beheshti E, Caradec J, Chin MY, Tomlinson, Guns ES. Epidermal growth factor receptor in prostate cancer derived exosomes. PLoS ONE. 2016;11:e0154967.

    Google Scholar 

  32. Brand TM, Iida M, Li C, Wheeler DL. The nuclear epidermal growth factor receptor signaling network and its role in cancer. Disco Med. 2011;12:419–32.

    Google Scholar 

  33. Chang YS, Chen WY, Yin JJ, Sheppard-Tillman H, Huang J, Liu YN. EGF receptor promotes prostate cancer bone metastasis by downregulating miR-1 and activating TWIST1. Cancer Res. 2015;75:3077–86.

    CAS  Google Scholar 

  34. Tsai YC, Chen WY, Siu MK, Tsai HY, Yin JJ, Huang J, et al. Epidermal growth factor receptor signaling promotes metastatic prostate cancer through microRNA-96-mediated downregulation of the tumor suppressor ETV6. Cancer Lett. 2017;384:1–8.

    CAS  Google Scholar 

  35. Xu RH, Pelicano H, Zhou Y, Carew JS, Feng L, Bhalla KN, et al. Inhibition of glycolysis in cancer cells: a novel strategy to overcome drug resistance associated with mitochondrial respiratory defect and hypoxia. Cancer Res. 2005;65:613–21.

    CAS  Google Scholar 

  36. Kim J, DeBerardinis RJ. Mechanisms and implications of metabolic heterogeneity in cancer. Cell Metab. 2019;30:434–46.

    CAS  Google Scholar 

  37. Vlachostergios PJ, Papandreou CN. Targeting neuroendocrine prostate cancer: molecular and clinical perspectives. Front Oncol. 2015;5:6.

    Google Scholar 

  38. Dobolyi A, Bago AG, Gal A, Molnar MJ, Palkovits M, Adam-Vizi V, et al. Localization of SUCLA2 and SUCLG2 subunits of succinyl CoA ligase within the cerebral cortex suggests the absence of matrix substrate-level phosphorylation in glial cells of the human brain. J Bioenerg Biomembr. 2015;47:33–41.

    CAS  Google Scholar 

  39. Dobolyi A, Ostergaard E, Bago AG, Doczi T, Palkovits M, Gal A, et al. Exclusive neuronal expression of SUCLA2 in the human brain. Brain Struct Funct. 2015;220:135–51.

    CAS  Google Scholar 

  40. Miller C, Wang L, Ostergaard E, Dan P, Saada A. The interplay between SUCLA2, SUCLG2, and mitochondrial DNA depletion. Biochim Biophys Acta. 2011;1812:625–9.

    CAS  Google Scholar 

  41. Liu YN, Niu S, Chen WY, Zhang Q, Tao Y, Chen WH, et al. Leukemia inhibitory factor promotes castration-resistant prostate cancer and neuroendocrine differentiation by activated ZBTB46. Clin Cancer Res. 2019;25:4128–40.

    CAS  Google Scholar 

  42. Nelson PS, Clegg N, Arnold H, Ferguson C, Bonham M, White J, et al. The program of androgen-responsive genes in neoplastic prostate epithelium. Proc Natl Acad Sci USA. 2002;99:11890–5.

    CAS  Google Scholar 

  43. Wang G, Jones SJ, Marra MA, Sadar MD. Identification of genes targeted by the androgen and PKA signaling pathways in prostate cancer cells. Oncogene. 2006;25:7311–23.

    CAS  Google Scholar 

  44. Chicas A, Wang X, Zhang C, McCurrach M, Zhao Z, Mert O, et al. Dissecting the unique role of the retinoblastoma tumor suppressor during cellular senescence. Cancer Cell. 2010;17:376–87.

    CAS  Google Scholar 

  45. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA. 2005;102:15545–50.

    CAS  Google Scholar 

  46. Hoshino M, Fukui H, Ono Y, Sekikawa A, Ichikawa K, Tomita S, et al. Nuclear expression of phosphorylated EGFR is associated with poor prognosis of patients with esophageal squamous cell carcinoma. Pathobiology. 2007;74:15–21.

    CAS  Google Scholar 

  47. Madshus IH, Stang E. Internalization and intracellular sorting of the EGF receptor: a model for understanding the mechanisms of receptor trafficking. J Cell Sci. 2009;122:3433–9.

    CAS  Google Scholar 

  48. Chiarugi P, Paoli P, Cirri P. Tumor microenvironment and metabolism in prostate cancer. Semin Oncol. 2014;41:267–80.

    CAS  Google Scholar 

  49. Timofeeva OA, Chasovskikh S, Lonskaya I, Tarasova NI, Khavrutskii L, Tarasov SG, et al. Mechanisms of unphosphorylated STAT3 transcription factor binding to DNA. J Biol Chem. 2012;287:14192–200.

    CAS  Google Scholar 

  50. Chen L, Huang M, Plummer J, Pan J, Jiang YY, Yang Q, et al. Master transcription factors form interconnected circuitry and orchestrate transcriptional networks in oesophageal adenocarcinoma. Gut. 2020;69:630–40.

    CAS  Google Scholar 

  51. Egan K, Kusao I, Troelstrup D, Agsalda M, Shiramizu B. Mitochondrial DNA in residual leukemia cells in cerebrospinal fluid in children with acute lymphoblastic leukemia. J Clin Med Res. 2010;2:225–9.

    CAS  Google Scholar 

  52. Kim MM, Clinger JD, Masayesva BG, Ha PK, Zahurak ML, Westra WH, et al. Mitochondrial DNA quantity increases with histopathologic grade in premalignant and malignant head and neck lesions. Clin Cancer Res. 2004;10:8512–5.

    CAS  Google Scholar 

  53. Wang Y, Liu VW, Xue WC, Cheung AN, Ngan HY. Association of decreased mitochondrial DNA content with ovarian cancer progression. Br J Cancer. 2006;95:1087–91.

    CAS  Google Scholar 

  54. Mizumachi T, Muskhelishvili L, Naito A, Furusawa J, Fan CY, Siegel ER, et al. Increased distributional variance of mitochondrial DNA content associated with prostate cancer cells as compared with normal prostate cells. Prostate. 2008;68:408–17.

    CAS  Google Scholar 

  55. Zhang XQ, Kondrikov D, Yuan TC, Lin FF, Hansen J, Lin MF. Receptor protein tyrosine phosphatase alpha signaling is involved in androgen depletion-induced neuroendocrine differentiation of androgen-sensitive LNCaP human prostate cancer cells. Oncogene. 2003;22:6704–16.

    CAS  Google Scholar 

  56. Marcoux N, Gettinger SN, O’Kane G, Arbour KC, Neal JW, Husain H, et al. EGFR-mutant adenocarcinomas that transform to small-cell lung cancer and other neuroendocrine carcinomas: clinical outcomes. J Clin Oncol. 2019;37:278–85.

    CAS  Google Scholar 

  57. Papouchado B, Erickson LA, Rohlinger AL, Hobday TJ, Erlichman C, Ames MM, et al. Epidermal growth factor receptor and activated epidermal growth factor receptor expression in gastrointestinal carcinoids and pancreatic endocrine carcinomas. Mod Pathol. 2005;18:1329–35.

    CAS  Google Scholar 

  58. Shah T, Hochhauser D, Frow R, Quaglia A, Dhillon AP, Caplin ME. Epidermal growth factor receptor expression and activation in neuroendocrine tumours. J Neuroendocrinol. 2006;18:355–60.

    CAS  Google Scholar 

  59. Angelescu R, Burada F, Angelescu C, Gheonea DI, Iordache S, Mixich F, et al. Expression of vascular endothelial growth factor and epidermal growth factor receptor in pancreatic ductal adenocarcinomas, neuroendocrine tumours and chronic pancreatitis. Endosc Ultrasound. 2013;2:86–91.

    Google Scholar 

  60. Martin-Orozco RM, Almaraz-Pro C, Rodriguez-Ubreva FJ, Cortes MA, Ropero S, Colomer R, et al. EGF prevents the neuroendocrine differentiation of LNCaP cells induced by serum deprivation: the modulator role of PI3K/Akt. Neoplasia. 2007;9:614–24.

    CAS  Google Scholar 

  61. Brand TM, Iida M, Luthar N, Starr MM, Huppert EJ, Wheeler DL. Nuclear EGFR as a molecular target in cancer. Radiother Oncol. 2013;108:370–7.

    CAS  Google Scholar 

  62. Hsing AW, Sakoda LC, Chua S Jr. Obesity, metabolic syndrome, and prostate cancer. Am J Clin Nutr. 2007;86:s843–57.

    Google Scholar 

  63. Spiotto MT, Chung TD. STAT3 mediates IL-6-induced growth inhibition in the human prostate cancer cell line LNCaP. Prostate. 2000;42:88–98.

    CAS  Google Scholar 

  64. Bromberg J. Stat proteins and oncogenesis. J Clin Invest. 2002;109:1139–42.

    CAS  Google Scholar 

  65. Scaltriti M, Baselga J. The epidermal growth factor receptor pathway: a model for targeted therapy. Clin Cancer Res. 2006;12:5268–72.

    CAS  Google Scholar 

  66. Chua CY, Liu Y, Granberg KJ, Hu L, Haapasalo H, Annala MJ, et al. IGFBP2 potentiates nuclear EGFR-STAT3 signaling. Oncogene. 2016;35:738–47.

    CAS  Google Scholar 

  67. Mahdavi A, Davey RE, Bhola P, Yin T, Zandstra PW. Sensitivity analysis of intracellular signaling pathway kinetics predicts targets for stem cell fate control. PLoS Comput Biol. 2007;3:e130.

    Google Scholar 

  68. Faraco CCF, Faria J, Kunrath-Lima M, Miranda MC, de Melo MIA, Ferreira ADF, et al. Translocation of epidermal growth factor (EGF) to the nucleus has distinct kinetics between adipose tissue-derived mesenchymal stem cells and a mesenchymal cancer cell lineage. J Struct Biol. 2018;202:61–9.

    CAS  Google Scholar 

  69. Gagliano-Juca T, Burak MF, Pencina KM, Li Z, Edwards RR, Travison TG, et al. Metabolic changes in androgen-deprived nondiabetic men with prostate cancer are not mediated by cytokines or aP2. J Clin Endocrinol Metab. 2018;103:3900–8.

    Google Scholar 

  70. Xue Y, Verhofstad A, Lange W, Smedts F, Debruyne F, de la Rosette J, et al. Prostatic neuroendocrine cells have a unique keratin expression pattern and do not express Bcl-2: cell kinetic features of neuroendocrine cells in the human prostate. Am J Pathol. 1997;151:1759–65.

    CAS  Google Scholar 

  71. Beltran H, Hruszkewycz A, Scher HI, Hildesheim J, Isaacs J, Yu EY, et al. The role of lineage plasticity in prostate cancer therapy resistance. Clin Cancer Res. 2019;25:6916–24.

    CAS  Google Scholar 

  72. Schindelin J, Rueden CT, Hiner MC, Eliceiri KW. The ImageJ ecosystem: an open platform for biomedical image analysis. Mol Reprod Dev. 2015;82:518–29.

    CAS  Google Scholar 

  73. Rooney JP, Ryde IT, Sanders LH, Howlett EH, Colton MD, Germ KE, et al. PCR based determination of mitochondrial DNA copy number in multiple species. Methods Mol Biol. 2015;1241:23–38.

    CAS  Google Scholar 

Download references

Acknowledgements

This work was jointly supported by grants from the Ministry of Science and Technology of Taiwan (MOST108-2320-B-038-047) to WYC, (MOST 109-2314-B-038-105) to YCW, and (MOST109-2326-B-038-001-MY3) to YNL, Taipei Medical University—Wan Fang Hospital (109TMU-WFH-01) to WYC, and the National Health Research Institute of Taiwan (NHRI-EX109-10702BI) to YNL.

Author information

Authors and Affiliations

Authors

Contributions

WYC and YNL designed the experiments and supervised the project. SRL, HLY, KCJ, and WHC performed the experiments. YCW, WYC, and JH provided the human prostate cancer samples. WYC performed the histomorphometric analysis. HLY constructed the databases and performed the statistical and computational analyses. SRL, YCW, WYC, and YNL wrote, reviewed, and/or revised the manuscript. All authors analyzed and interpreted the data.

Corresponding authors

Correspondence to Wei-Yu Chen or Yen-Nien Liu.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Ethical approval

Tissue samples were used in accordance with the Declaration of Helsinki and U.S. Common Rule, and their use was approved by the Taipei Medical University—Joint Institutional Review Board (approval no. N201711067) and the Duke University School of Medicine—Institutional Review Board (protocol ID, Pro00070193).

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lin, SR., Wen, YC., Yeh, HL. et al. EGFR-upregulated LIFR promotes SUCLG2-dependent castration resistance and neuroendocrine differentiation of prostate cancer. Oncogene 39, 6757–6775 (2020). https://doi.org/10.1038/s41388-020-01468-9

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41388-020-01468-9

This article is cited by

Search

Quick links