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:

Proteomic analysis reveals a role for PAX8 in peritoneal colonization of high grade serous ovarian cancer that can be targeted with micelle encapsulated thiostrepton

Abstract

High grade serous ovarian cancer (HGSOC) is the fifth leading cause of cancer deaths among women yet effective targeted therapies against this disease are limited. The heterogeneity of HGSOC, including few shared oncogenic drivers and origination from both the fallopian tube epithelium (FTE) and ovarian surface epithelium (OSE), has hampered development of targeted drug therapies. PAX8 is a lineage-specific transcription factor expressed in the FTE that is also ubiquitously expressed in HGSOC where it is an important driver of proliferation, migration, and cell survival. PAX8 is not normally expressed in the OSE, but it is turned on after malignant transformation. In this study, we use proteomic and transcriptomic analysis to examine the role of PAX8 leading to increased migratory capabilities in a human ovarian cancer model, as well as in tumor models derived from the OSE and FTE. We find that PAX8 is a master regulator of migration with unique downstream transcriptional targets that are dependent on the cell’s site of origin. Importantly, we show that targeting PAX8, either through CRISPR genomic alteration or through drug treatment with micelle encapsulated thiostrepton, leads to a reduction in tumor burden. These findings suggest PAX8 is a unifying protein driving metastasis in ovarian tumors that could be developed as an effective drug target to treat HGSOC derived from both the OSE and FTE.

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
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

References

  1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2017. CA Cancer J Clin. 2017;67:7–30.

    Article  Google Scholar 

  2. Ozcan A, Shen SS, Hamilton C, Anjana K, Coffey D, Krishnan B, et al. PAX 8 expression in non-neoplastic tissues, primary tumors, and metastatic tumors: a comprehensive immunohistochemical study. Mod Pathol. 2011;24:751–64.

    Article  CAS  Google Scholar 

  3. McCloskey CW, Goldberg RL, Carter LE, Gamwell LF, Al-Hujaily EM, Collins O, et al. A new spontaneously transformed syngeneic model of high-grade serous ovarian cancer with a tumor-initiating cell population. Front Oncol. 2014;4:53.

    Article  Google Scholar 

  4. Tanwar PS, Mohapatra G, Chiang S, Engler DA, Zhang L, Kaneko-Tarui T, et al. Loss of LKB1 and PTEN tumor suppressor genes in the ovarian surface epithelium induces papillary serous ovarian cancer. Carcinogenesis. 2014;35:546–53.

    Article  CAS  Google Scholar 

  5. Rodgers LH, Ó hAinmhire E, Young AN, Burdette JE. Loss of PAX8 in high-grade serous ovarian cancer reduces cell survival despite unique modes of action in the fallopian tube and ovarian surface epithelium. Oncotarget. 2016;7:32785–95.

    Article  Google Scholar 

  6. Di Palma T, Lucci V, de Cristofaro T, Filippone MG, Zannini M. A role for PAX8 in the tumorigenic phenotype of ovarian cancer cells. Bmc Cancer. 2014;14:292.

    Article  Google Scholar 

  7. Piek JM, van Diest PJ, Zweemer RP, Jansen JW, Poort-Keesom RJ, Menko FH, et al. Dysplastic changes in prophylactically removed Fallopian tubes of women predisposed to developing ovarian cancer. J Pathol. 2001;195:451–6.

    Article  CAS  Google Scholar 

  8. Labidi-Galy SI, Papp E, Hallberg D, Niknafs N, Adleff V, Noe M, et al. High grade serous ovarian carcinomas originate in the fallopian tube. Nat Commun. 2017;8:1093.

    Article  Google Scholar 

  9. Kurman RJ, Shih I-M. Molecular pathogenesis and extraovarian origin of epithelial ovarian cancer–shifting the paradigm. Hum Pathol. 2011;42:918–31.

    Article  CAS  Google Scholar 

  10. Elias KM, Emori MM, Westerling T, Long H, Budina-Kolomets A, Li F, et al. Epigenetic remodeling regulates transcriptional changes between ovarian cancer and benign precursors. JCI Insight. 2016;1. https://doi.org/10.1172/jci.insight.87988.

  11. Adler EK, Corona RI, Lee JM, Rodriguez-Malave N, Mhawech-Fauceglia P, Sowter H, et al. The PAX8 cistrome in epithelial ovarian cancer. Oncotarget. 2017;8:108316–32.

    Article  Google Scholar 

  12. Mansouri A, Chowdhury K, Gruss P. Follicular cells of the thyroid gland require Pax8 gene function. Nat Genet. 1998;19:87–90.

    Article  CAS  Google Scholar 

  13. Li CG, Nyman JE, Braithwaite AW, Eccles MR. PAX8 promotes tumor cell growth by transcriptionally regulating E2F1 and stabilizing RB protein. Oncogene. 2011;30:4824–34.

    Article  CAS  Google Scholar 

  14. Ghannam-Shahbari D, Jacob E, Kakun RR, Wasserman T, Korsensky L, Sternfeld O, et al. PAX8 activates a p53-p21-dependent pro-proliferative effect in high grade serous ovarian carcinoma. Oncogene. 2018;37:2213.

    Article  CAS  Google Scholar 

  15. Haley J, Tomar S, Pulliam N, Xiong S, Perkins SM, Karpf AR, et al. Functional characterization of a panel of high-grade serous ovarian cancer cell lines as representative experimental models of the disease. Oncotarget. 2016;7:32810–20.

    Article  Google Scholar 

  16. King SM, Quartuccio SM, Vanderhyden BC, Burdette JE. Early transformative changes in normal ovarian surface epithelium induced by oxidative stress require akt upregulation, DNA damage, and epithelial-stromal interaction. Carcinogenesis. 2013;34:1125–33.

    Article  CAS  Google Scholar 

  17. Eddie SL, Quartuccio SM, Ó hAinmhir E, Moyle-Heyrman G, Lantvit DD, Wei J-J, et al. Tumorigenesis and peritoneal colonization from fallopian tube epithelium. Oncotarget. 2015;6:20500–12.

    Article  Google Scholar 

  18. Russo A, Czarnecki AA, Dean M, Modi DA, Lantvit DD, Hardy L, et al. PTEN loss in the fallopian tube induces hyperplasia and ovarian tumor formation. Oncogene. 2018;37:1976.

    Article  CAS  Google Scholar 

  19. Ran FA, et al. Genome engineering using the CRISPR-Cas9 system. Nat Protoc. 2013;8:2281–308.

    Article  CAS  Google Scholar 

  20. Endsley MP, Moyle-Heyrman G, Karthikeyan S, Lantvit DD, Davis DA, Wei J-J, et al. Spontaneous transformation of murine oviductal epithelial cells: a model system to investigate the onset of fallopian-derived tumors. Front Oncol. 2015;5:154.

    Article  Google Scholar 

  21. Wiśniewski JR, Zougman A, Nagaraj N, Mann M. Universal sample preparation method for proteome analysis. Nat Methods. 2009;6:359–62.

    Article  Google Scholar 

  22. Batth TS, Francavilla C, Olsen JV. Off-line high-ph reversed-phase fractionation for in-depth phosphoproteomics. J Proteome Res. 2014;13:6176–86.

    Article  CAS  Google Scholar 

  23. Gokce E, Andrews GL, Dean RA, Muddiman DC. Increasing proteome coverage with offline RP HPLC coupled to online RP nanoLC-MS. J Chromatogr B Anal Technol Biomed Life Sci. 2011;879:610–4.

    Article  CAS  Google Scholar 

  24. Adusumilli R, Mallick P. Data conversion with ProteoWizard msConvert. Methods Mol Biol Clifton NJ. 2017;1550:339–68.

    Article  CAS  Google Scholar 

  25. Ong S-E, Mann M. A practical recipe for stable isotope labeling by amino acids in cell culture (SILAC). Nat Protoc. 2006;1:2650–60.

    Article  CAS  Google Scholar 

  26. King SM, et al. The impact of ovulation on fallopian tube epithelial cells: evaluating three hypotheses connecting ovulation and serous ovarian cancer. Endocr Relat Cancer. 2011;18:627–42.

    Article  CAS  Google Scholar 

  27. Martiny-Baron G, Kazanietz MG, Mischak H, Blumberg PM, Kochs G, Hug H, et al. Selective inhibition of protein kinase C isozymes by the indolocarbazole Gö 6976. J Biol Chem. 1993;268:9194–7.

    CAS  PubMed  Google Scholar 

  28. Ke N, Wang X, Xu X, Abassi YA. The xCELLigence system for real-time and label-free monitoring of cell viability. In: Mammalian cell viability. Springer; 2011, pp 33–43.

  29. Esparza K, Onyuksel H. Development of co-solvent freeze-drying method for the encapsulation of water-insoluble thiostrepton in sterically stabilized micelles. Int J Pharm. 2019;556:21–29.

    Article  CAS  Google Scholar 

  30. de Cristofaro T, Di Palma T, Soriano AA, Monticelli A, Affinito O, Cocozza S, et al. Candidate genes and pathways downstream of PAX8 involved in ovarian high-grade serous carcinoma. Oncotarget. 2016;7:41929–47.

    Article  Google Scholar 

  31. Hegde NS, Sanders DA, Rodriguez R, Balasubramanian S. The transcription factor FOXM1 is a cellular target of the natural product thiostrepton. Nat Chem. 2011;3:725–31.

    Article  CAS  Google Scholar 

  32. Wang M, Gartel AL. Micelle-encapsulated thiostrepton as an effective nanomedicine for inhibiting tumor growth and for suppressing FOXM1 in human xenografts. Mol Cancer Ther. 2011;10:2287–97.

    Article  CAS  Google Scholar 

  33. Jiang L, Wu X, Wang P, Wen T, Yu C, Wei L, et al. Targeting FoxM1 by thiostrepton inhibits growth and induces apoptosis of laryngeal squamous cell carcinoma. J Cancer Res Clin Oncol. 2015;141:971–81.

    Article  Google Scholar 

  34. Ashok B, Arleth L, Hjelm RP, Rubinstein I, Onyüksel H. In vitro characterization of PEGylated phospholipid micelles for improved drug solubilization: effects of PEG chain length and PC incorporation. J Pharm Sci. 2004;93:2476–87.

    Article  CAS  Google Scholar 

  35. Lim SB, Banerjee A, Önyüksel H. Improvement of drug safety by the use of lipid-based nanocarriers. J Control Release J Control Release Soc. 2012;163:34–45.

    Article  CAS  Google Scholar 

  36. Laury AR, Hornick JL, Perets R, Krane JF, Corson J, Drapkin R, et al. PAX8 reliably distinguishes ovarian serous tumors from malignant mesothelioma. Am J Surg Pathol. 2010;34:627–35.

    PubMed  Google Scholar 

  37. Cheung HW, Cowley GS, Weir BA, Boehm JS, Rusin S, Scott JA, et al. Systematic investigation of genetic vulnerabilities across cancer cell lines reveals lineage-specific dependencies in ovarian cancer. Proc Natl Acad Sci USA. 2011;108:12372–7.

    Article  CAS  Google Scholar 

  38. Musrap N, Tuccitto A, Karagiannis GS, Saraon P, Batruch I, Diamandis EP. Comparative proteomics of ovarian cancer aggregate formation reveals an increased expression of calcium-activated chloride channel regulator 1 (CLCA1). J Biol Chem 2015;290:17218–27.

    Article  CAS  Google Scholar 

  39. Grassi ML, Palma C, de S, Thomé CH, Lanfredi GP, Poersch A, Faça VM. Proteomic analysis of ovarian cancer cells during epithelial-mesenchymal transition (EMT) induced by epidermal growth factor (EGF) reveals mechanisms of cell cycle control. J Proteom. 2017;151:2–11.

    Article  CAS  Google Scholar 

  40. Waldemarson S, Krogh M, Alaiya A, Kirik U, Schedvins K, Auer G, et al. Protein expression changes in ovarian cancer during the transition from benign to malignant. J Proteome Res. 2012;11:2876–89.

    Article  CAS  Google Scholar 

  41. Wang L-N, Tong S-W, Hu H-D, Ye F, Li S-L, Ren H, et al. Quantitative proteome analysis of ovarian cancer tissues using a iTRAQ approach. J Cell Biochem. 2012;113:3762–72.

    Article  CAS  Google Scholar 

  42. Harms JM, Wilson DN, Schluenzen F, Connell SR, Stachelhaus T, Zaborowska Z, et al. Translational regulation via L11: molecular switches on the ribosome turned on and off by thiostrepton and micrococcin. Mol Cell. 2008;30:26–38.

    Article  CAS  Google Scholar 

  43. Cancer Genome Atlas Research Network. Integrated genomic analyses of ovarian carcinoma. Nature. 2011;474: 609–15.

  44. Zhang X, Cheng L, Minn K, Madan R, Godwin AK, Shridhar V, et al. Targeting of mutant p53-induced FoxM1 with thiostrepton induces cytotoxicity and enhances carboplatin sensitivity in cancer cells. Oncotarget. 2014;5:11365–80.

    PubMed  PubMed Central  Google Scholar 

  45. Cristofaro T, de, Mascia A, Pappalardo A, D’Andrea B, Nitsch L, Zannini M. Pax8 protein stability is controlled by sumoylation. J Mol Endocrinol. 2009;42:35–46.

    Article  Google Scholar 

Download references

Acknowledgements

This work was supported by the Department of Defense Ovarian Cancer Fund 160076, the National Cancer Institute F30CA224986, the National Cancer Center for Research Resources Facilities Improvement Program C06RR15482, the University of Illinois-Chicago Department of Chemistry, the Ara Parsegian Medical Research Foundation, and the Abraham Lincoln Fellowship.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Joanna E. Burdette.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Additional information

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

Supplementary information

Supplemental figure legends

Supplemental Figure 1. Verification of modified cell lines generated in this study by immunoblot

Supplemental Figure 2. Heterozygous deletion of PAX8 in OVCAR8 reduces epithelial markers

Supplemental Figure 3. PAX8 deletion does not affect proliferation in OVCAR8<sup>RFP</sup> cells

Supplemental Figure 4. PAX8 does not affect adherens junctions in OVCAR8<sup>RFP</sup> cel

Supplemental Figure 5. PAX8 does not regulate PKCα in MOSE cells

Supplemental Table 1. Antibodies used in this study

Supplemental Table 2. OVCAR8 proteome with PAX8 alteration

Supplemental Table 3. MOSE proteome with PAX8 alteration

Supplemental Table 4. List of proteins differentially regulated by PAX8 in MOSE and OVCAR8 cell lines

Supplemental Table 5. Transcriptome of MOSE cells with PAX8 overexpressed

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hardy, L.R., Pergande, M.R., Esparza, K. et al. Proteomic analysis reveals a role for PAX8 in peritoneal colonization of high grade serous ovarian cancer that can be targeted with micelle encapsulated thiostrepton. Oncogene 38, 6003–6016 (2019). https://doi.org/10.1038/s41388-019-0842-2

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41388-019-0842-2

This article is cited by

Search

Quick links