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.

Substrate-biased activity-based probes identify proteases that cleave receptor CDCP1

Abstract

CUB domain-containing protein 1 (CDCP1) is an oncogenic orphan transmembrane receptor and a promising target for the detection and treatment of cancer. Extracellular proteolysis of CDCP1 by poorly defined mechanisms induces pro-metastatic signaling. We describe a new approach for the rapid identification of proteases responsible for key proteolytic events using a substrate-biased activity-based probe (sbABP) that incorporates a substrate cleavage motif grafted onto a peptidyl diphenyl phosphonate warhead for specific target protease capture, isolation and identification. Using a CDCP1-biased probe, we identify urokinase (uPA) as the master regulator of CDCP1 proteolysis, which acts both by directly cleaving CDCP1 and by activating CDCP1-cleaving plasmin. We show that coexpression of uPA and CDCP1 is strongly predictive of poor disease outcome across multiple cancers and demonstrate that uPA-mediated CDCP1 proteolysis promotes metastasis in disease-relevant preclinical in vivo models. These results highlight CDCP1 cleavage as a potential target to disrupt cancer and establish sbABP technology as a new approach to identify disease-relevant proteases.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Design of an sbABP targeting CDCP1-extracellular domain (ECD) cleavage sites.
Fig. 2: ABPs targeting CDCP1-ECD cleavage sites inhibit CDCP1 proteolysis.
Fig. 3: Identification of proteases reacting with sbABPs targeting CDCP1-ECD cleavage sites.
Fig. 4: uPA controls CDCP1 proteolysis in cancer cells.
Fig. 5: uPA-mediated CDCP1 proteolysis modulates aggressiveness of pancreatic cancer cells.

Similar content being viewed by others

Data availability

The authors declare that the main data supporting the findings of this study are available within the article and its Supplementary Information and source data files. Specialized reagents and MS data are available from J.D.H. Gene expression data from TCGA and ICGC cohorts are available online at https://xenabrowser.net/ and https://icgc.org/. Source data are provided with this paper.

References

  1. Rawlings, N. D. & Barrett, A. J. Evolutionary families of peptidases. Biochem. J. 290, 205–218 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Ten Cate, H. et al. Coagulation factor and protease pathways in thrombosis and cardiovascular disease. Thromb. Haemost. 117, 1265–1271 (2017).

    Article  PubMed  Google Scholar 

  3. Weidmann, H. et al. The plasma contact system, a protease cascade at the nexus of inflammation, coagulation and immunity. Biochim. Biophys. Acta Mol. Cell. Res. 1864, 2118–2127 (2017).

    Article  CAS  PubMed  Google Scholar 

  4. Prassas, I. et al. Unleashing the therapeutic potential of human kallikrein-related serine proteases. Nat. Rev. Drug Discov. 14, 183–202 (2015).

    Article  CAS  PubMed  Google Scholar 

  5. Hermann, M. et al. Deficits in developmental neurogenesis and dendritic spine maturation in mice lacking the serine protease inhibitor neuroserpin. Mol. Cell Neurosci. 102, 103420 (2020).

    Article  CAS  PubMed  Google Scholar 

  6. Toriseva, M. & Kahari, V. M. Proteinases in cutaneous wound healing. Cell. Mol. Life Sci. 66, 203–224 (2009).

    Article  CAS  PubMed  Google Scholar 

  7. Mitschke, J. et al. The role of proteases in epithelial-to-mesenchymal cell transitions in cancer. Cancer Metastasis Rev. 38, 431–444 (2019).

    Article  PubMed  Google Scholar 

  8. Stepanova, V. V. & Tkachuk, V. A. Urokinase as a multidomain protein and polyfunctional cell regulator. Biochem. (Mosc.) 67, 109–118 (2002).

    Article  CAS  Google Scholar 

  9. Kryza, T. et al. The kallikrein-related peptidase family: dysregulation and functions during cancer progression. Biochimie 122, 283–299 (2016).

    Article  CAS  PubMed  Google Scholar 

  10. Millet, J. K. & Whittaker, G. R. Host cell proteases: critical determinants of coronavirus tropism and pathogenesis. Virus Res. 202, 120–134 (2015).

    Article  CAS  PubMed  Google Scholar 

  11. Mohajeri, M. et al. Mast cell tryptase—marker and maker of cardiovascular diseases. Pharmacol. Ther. 199, 91–110 (2019).

    Article  CAS  PubMed  Google Scholar 

  12. Santibanez, J. F. & Krstic, J. Transforming growth factor-β and urokinase type plasminogen interplay in cancer. Curr. Protein Pept. Sci. 19, 1155–1163 (2018).

    Article  CAS  PubMed  Google Scholar 

  13. Vasiljeva, O. et al. The multifaceted roles of tumor-associated proteases and harnessing their activity for prodrug activation. Biol. Chem. 400, 965–977 (2019).

  14. Luo, S. Y. et al. Protease substrate identification using N-terminomics. ACS Chem. Biol. 14, 2361–2371 (2019).

    Article  CAS  PubMed  Google Scholar 

  15. Silva, L. M. et al. Integration of two in-depth quantitative proteomics approaches determines the kallikrein-related peptidase 7 (KLK7) degradome in ovarian cancer cell secretome. Mol. Cell Proteom. 18, 818–836 (2019).

    Article  CAS  Google Scholar 

  16. Song, J. et al. PROSPERous: high-throughput prediction of substrate cleavage sites for 90 proteases with improved accuracy. Bioinformatics 34, 684–687 (2018).

    Article  CAS  PubMed  Google Scholar 

  17. Deng, H. et al. Activity-based protein profiling: recent advances in medicinal chemistry. Eur. J. Med. Chem. 191, 112151 (2020).

    Article  CAS  PubMed  Google Scholar 

  18. Powers, J. C. et al. Irreversible inhibitors of serine, cysteine, and threonine proteases. Chem. Rev. 102, 4639–4750 (2002).

    Article  CAS  PubMed  Google Scholar 

  19. Maslanka, M. & Mucha, A. Recent developments in peptidyl diaryl phoshonates as inhibitors and activity-based probes for serine proteases. Pharmaceuticals 12, 86 (2019).

  20. Maluch, I. et al. Applications of unnatural amino acids in protease probes. Chem. Asian J. 14, 4103–4113 (2019).

    Article  CAS  PubMed  Google Scholar 

  21. Khan, T. et al. The CDCP1 signalling hub: a target for cancer detection and therapeutic intervention. Cancer Res. https://doi.org/10.1158/0008-5472.CAN-20-2978 (2021).

  22. Chen, Y. et al. Development of an enzyme-linked immunosorbent assay for detection of CDCP1 shed from the cell surface and present in colorectal cancer serum specimens. J. Pharm. Biomed. Anal. 139, 65–72 (2017).

    Article  CAS  PubMed  Google Scholar 

  23. He, Y. et al. Evidence that cell surface localization of serine protease activity facilitates cleavage of the protease activated receptor CDCP1. Biol. Chem. 399, 1091–1097 (2018).

    Article  CAS  PubMed  Google Scholar 

  24. He, Y. et al. Proteolysis-induced N-terminal ectodomain shedding of the integral membrane glycoprotein CUB domain-containing protein 1 (CDCP1) is accompanied by tyrosine phosphorylation of its C-terminal domain and recruitment of Src and PKCδ. J. Biol. Chem. 285, 26162–26173 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Kryza, T. et al. Effective targeting of intact and proteolysed CDCP1 for imaging and treatment of pancreatic ductal adenocarcinoma. Theranostics 10, 4116–4133 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Casar, B. et al. In vivo cleaved CDCP1 promotes early tumor dissemination via complexing with activated β1 integrin and induction of FAK/PI3K/Akt motility signaling. Oncogene 33, 255–268 (2014).

    Article  CAS  PubMed  Google Scholar 

  27. Wright, H. J. et al. CDCP1 cleavage is necessary for homodimerization-induced migration of triple-negative breast cancer. Oncogene 35, 4762–4772 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Law, M. E. et al. Glucocorticoids and histone deacetylase inhibitors cooperate to block the invasiveness of basal-like breast cancer cells through novel mechanisms. Oncogene 32, 1316–1329 (2013).

    Article  CAS  PubMed  Google Scholar 

  29. Serim, S. et al. Tuning activity-based probe selectivity for serine proteases by on-resin ‘click’ construction of peptide diphenyl phosphonates. Org. Biomol. Chem. 11, 5714–5721 (2013).

    Article  CAS  PubMed  Google Scholar 

  30. Jaiswal, R. K. et al. Diversity and functional evolution of the plasminogen activator system. Biomed. Pharmacother. 98, 886–898 (2018).

    Article  CAS  PubMed  Google Scholar 

  31. Madunic, J. The urokinase plasminogen activator system in human cancers: an overview of its prognostic and predictive role. Thromb. Haemost. 118, 2020–2036 (2018).

    Article  PubMed  Google Scholar 

  32. Mahmood, N. et al. Multifaceted role of the urokinase-type plasminogen activator (uPA) and its receptor (uPAR): diagnostic, prognostic, and therapeutic applications. Front. Oncol. 8, 24 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Marino, G. et al. Protein termini and their modifications revealed by positional proteomics. ACS Chem. Biol. 10, 1754–1764 (2015).

    Article  CAS  PubMed  Google Scholar 

  34. Dallas, D. C. et al. Current peptidomics: applications, purification, identification, quantification, and functional analysis. Proteomics 15, 1026–1038 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Rut, W. et al. Activity profiling and crystal structures of inhibitor-bound SARS-CoV-2 papain-like protease: a framework for anti-COVID-19 drug design. Sci. Adv. 6, eabd4596 (2020).

  36. Casar, B. et al. Blocking of CDCP1 cleavage in vivo prevents Akt-dependent survival and inhibits metastatic colonization through PARP1-mediated apoptosis of cancer cells. Oncogene 31, 3924–3938 (2012).

    Article  CAS  PubMed  Google Scholar 

  37. Bhatt, A. S. et al. Adhesion signaling by a novel mitotic substrate of src kinases. Oncogene 24, 5333–5343 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Brown, T. A. et al. Adhesion or plasmin regulates tyrosine phosphorylation of a novel membrane glycoprotein p80/gp140/CUB domain-containing protein 1 in epithelia. J. Biol. Chem. 279, 14772–14783 (2004).

    Article  CAS  PubMed  Google Scholar 

  39. Ulisse, S. et al. The urokinase plasminogen activator system: a target for anti-cancer therapy. Curr. Cancer Drug Targets 9, 32–71 (2009).

    Article  CAS  PubMed  Google Scholar 

  40. Sieber, S. A. et al. Proteomic profiling of metalloprotease activities with cocktails of active-site probes. Nat. Chem. Biol. 2, 274–281 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Pollan, S. G. et al. Regulation of inside-out β1-integrin activation by CDCP1. Oncogene 37, 2817–2836 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Chou, A. et al. Tailored first-line and second-line CDK4-targeting treatment combinations in mouse models of pancreatic cancer. Gut 67, 2142–2155 (2018).

    Article  CAS  PubMed  Google Scholar 

  43. He, Y. et al. Elevated CDCP1 predicts poor patient outcome and mediates ovarian clear cell carcinoma by promoting tumor spheroid formation, cell migration and chemoresistance. Oncogene 35, 468–478 (2016).

    Article  CAS  PubMed  Google Scholar 

  44. Harrington, B. S. et al. Cell line and patient-derived xenograft models reveal elevated CDCP1 as a target in high-grade serous ovarian cancer. Br. J. Cancer 114, 417–426 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Harrington, B. S. et al. Anti-CDCP1 immuno-conjugates for detection and inhibition of ovarian cancer. Theranostics 10, 2095–2114 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by grants from the National Health and Medical Research Council of Australia (NHMRC; grant APP1121970), the Avner Pancreatic Cancer Foundation (grant R3 JH 2.1) and the Cancer Council Queensland (grant APP1082040) to J.D.H., grants from the NHMRC, a Cancer Institute Career Development Fellowship and a Philip Hemstritch Fellowship in Pancreatic Cancer to M.P. and a joint grant from the German Academic Exchange Service (DAAD; German-Australian Network on Personalized Medicine to B.S.H., T.D., V.M. and J.D.H.). H.K. is supported by the Sigrid Juselius Foundation. J.D.H. is supported by the Mater Foundation. S.L. is supported by the EPSRC Centre for Doctoral Training in Physical Sciences Innovation in Chemical Biology for Bioindustry and Healthcare (grant EP/LO15498/1). We acknowledge the Translational Research Institute, which receives support from the Australia Federal Government, for providing the core facilities that enabled this research, particularly the Biological Resources Facility and Preclinical Imaging Facility.

Author information

Authors and Affiliations

Authors

Contributions

The study was conceived and designed by T. Kryza, T. Khan, S.L., E.W.T. and J.D.H.. Methodology was developed by T. Kryza, T. Khan and S.L. and data were acquired by T. Kryza, T. Khan, S.L., Y.H. and J.Y. Data analysis and interpretation (for example, statistical analysis, biostatistics and computational analysis) was performed by T. Kryza, T. Khan, S.L., Y.H. and J.Y.; M.P., B.S.H., J.Y., S.P., H.K., J.K.R., T.D., V.M., U.R., Y.H., E.W.T. and J.D.H. were all involved in administrative, technical or material support (that is, reporting or organizing data and constructing databases). All authors were involved in writing, reviewing and revising the manuscript. The study was supervised by J.D.H.

Corresponding author

Correspondence to John D. Hooper.

Ethics declarations

Competing interests

E.W.T. is a founder, Director and shareholder in Myricx Pharma Ltd (UK). No potential conflicts of interest were disclosed by the other authors.

Additional information

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

Extended data

Extended Data Fig. 1 Structures of CDCP1-sbABPs.

Chemical structure of CDCP1-sbABP-1 to 4.

Extended Data Fig. 2 uPA and CDCP1 expression in uPA knock-down cells.

Gene expression analysis of uPA and CDCP1 by RT-qPCR on cDNA samples from TKCC05, PC3 and CAOV3 cells stably expressing control shRNA (shNT) or uPA-targeting ShRNA (shuPA#1 and shuPA#2). Results are expressed as mean of relative expression +/- SD compared to ACTB gene expression from three independent experiments. Statistical analysis was performed using One way non-parametric Kruskal-Wallis test with *p=0.0146, #p=0.0141, &p=0.0146.

Source data

Extended Data Fig. 3 uPA, uPAR, PAI1 and CDCP1 expression in cancer cells.

a, Gene expression analysis of uPA, uPAR, PAI1 and CDCP1 by RT-qPCR on cDNA samples from TKCC10, TKCC05, CAOV3, PC3 and OVMZ6 cells. Results are expressed as mean of relative expression +/- SD compared to ACTB gene expression from three independent experiments. b, Western blot analysis under reducing conditions of patient-derived PDAC TKCC10 and TKCC05 cells, ovarian cancer CAOV3 cells and prostate cancer PC3 cells using anti-CDCP1 antibody 4115 (reacts with CDCP1-FL and CTF) and an anti-GAPDH antibody.

Source data

Extended Data Fig. 4 uPA, uPAR and CDCP1 expression in bioengineered cancer cells.

TKCC05 and CAOV3 cells were bioengineered to stably express control ShRNA control (NT), uPA-targeting ShRNA (ShuPA#1 and ShuPA#2) or uPAR-targeting ShRNA (ShuPAR#1 and ShuPAR#2). Gene expression analysis of uPA, uPAR and CDCP1 was performed by RT-qPCR. Results are expressed as mean +/- SD of relative expression compared to ACTB gene expression from three independent experiments. Statistical analysis was performed using One way non-parametric Kruskal-Wallis test with *p=0.0185, #p=0.0325, $p=0.0479, &p=0.0325.

Source data

Extended Data Fig. 5 uPA, uPAR and PAI1 expression in bioengineered OVMZ6 cells.

Gene expression analysis of uPA, uPAR and PAI-1 by RT-qPCR on cDNA samples from OVMZ6 cells stably expressing WT- CDCP1 and transiently transfected with plasmid encoding uPA, uPAR or PAI-1. Results are expressed as mean +/- SD of relative expression compared to ACTB gene expression from three independent experiment.

Source data

Extended Data Fig. 6 Correlation analysis between uPAR expression and PDAC patient survival.

a, Kaplan-Meier analysis showing correlation between uPAR mRNA expression levels and PDAC survival in the ICGC patient dataset (n=267). For this analysis patients with expression below the first quartile in the entire population were segregated into ‘Bottom 25%’ and those with expression above the third quartile of expression level in the entire population were segregated into the ‘Top 25%’ expression group. Statistical differences between Kaplan-Meier curves were determined by Mantel-Cox test. b, Kaplan-Meier analysis showing correlation between gene pair (CDCP1 and uPAR; uPA and uPAR) expression levels and PDAC survival in the ICGC patient dataset (n=267). For this analysis patients with expression below the first quartile in the entire population were segregated into ‘Low’ expression group and those with expression above the third quartile in the entire population were segregated into the ‘High’ expression group. Statistical differences between Kaplan-Meier curves were determined by Mantel-Cox test.

Source data

Extended Data Fig. 7 Correlation analysis between CDCP1, uPA and uPAR expression and PDAC patient survival in TCGA dataset.

a, Kaplan-Meier analysis showing correlation between CDCP1, uPA and uPAR mRNA expression levels and PDAC survival in the TCGA patient dataset (n=170). For this analysis patients with expression below the first quartile in the entire population were segregated into ‘Bottom 25%’ and those with expression above the third quartile of expression level in and PDAC survival in the TCGA patient dataset (n=170). For this analysis patients with expression below the first quartile in the entire population were segregated into ‘Low’ expression group and those with expression above the third quartile in the entire population were segregated into the ‘High’ expression group. Statistical differences between Kaplan-Meier curves were determined by Mantel-Cox test.

Source data

Extended Data Fig. 8 Integration of sbABP technology with other technologies available to study proteolytic networks.

Conceptual workflow indicating how sbABP technology (a cleavage-centric approach) could be integrated with substrate-centric and protease-centric approaches to identify proteolytic mediators and develop inhibitors/ABPs to control a particular proteolytic event. TAILS, Terminal amine isotopic labeling of substrates; COFRADIC, Combined fractional diagonal chromatography; sbABP, substrate-biased activity-based probe.

Supplementary information

Supplementary Information

Supplementary Note (Synthetic procedures), Supplementary Tables 1and 2 and Supplementary Figs. 1–3

Reporting Summary

Supplementary Dataset 1

List of proteins identified in MS analysis (Human proteome database)

Supplementary Dataset 2

List of peptides corresponding to uPA identified in MS analysis (Human proteome database)

Supplementary Dataset 3

List of proteins identified in MS analysis (Bovin proteome database)

Supplementary Dataset 4

List of peptides corresponding to Plasminogen identified in MS analysis (Bovin proteome database)

Source data

Source Data Fig. 2

Unprocessed Western Blots

Source Data Fig. 2

Statistical Source Data

Source Data Fig. 3

Unprocessed Western Blots

Source Data Fig. 4

Unprocessed Western Blots

Source Data Fig. 4

Statistical Source Data

Source Data Fig. 5

Unprocessed Western Blots

Source Data Fig. 5

Statistical Source Data

Source Data Extended Data Fig. 2

Statistical Source Data

Source Data Extended Data Fig. 3

Unprocessed Western Blots

Source Data Extended Data Fig. 3

Statistical Source Data

Source Data Extended Data Fig. 4

Statistical Source Data

Source Data Extended Data Fig. 5

Statistical Source Data

Source Data Extended Data Fig. 6

Statistical Source Data

Source Data Extended Data Fig. 7

Statistical Source Data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kryza, T., Khan, T., Lovell, S. et al. Substrate-biased activity-based probes identify proteases that cleave receptor CDCP1. Nat Chem Biol 17, 776–783 (2021). https://doi.org/10.1038/s41589-021-00783-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-021-00783-w

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer