Skip to main content

Advertisement

Log in

Cell surface–anchored serine proteases in cancer progression and metastasis

  • Published:
Cancer and Metastasis Reviews Aims and scope Submit manuscript

Abstract

Over the last two decades, a novel subgroup of serine proteases, the cell surface–anchored serine proteases, has emerged as an important component of the human degradome, and several members have garnered significant attention for their roles in cancer progression and metastasis. A large body of literature describes that cell surface–anchored serine proteases are deregulated in cancer and that they contribute to both tumor formation and metastasis through diverse molecular mechanisms. The loss of precise regulation of cell surface–anchored serine protease expression and/or catalytic activity may be contributing to the etiology of several cancer types. There is therefore a strong impetus to understand the events that lead to deregulation at the gene and protein levels, how these precipitate in various stages of tumorigenesis, and whether targeting of selected proteases can lead to novel cancer intervention strategies. This review summarizes current knowledge about cell surface–anchored serine proteases and their role in cancer based on biochemical characterization, cell culture–based studies, expression studies, and in vivo experiments. Efforts to develop inhibitors to target cell surface–anchored serine proteases in cancer therapy will also be summarized.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2

Similar content being viewed by others

Abbreviations

ADC :

antibody drug conjugate

ALL :

acute lymphocytic leukemia

AML :

acute myeloid leukemia

APC :

adenomatous polyposis coli

AR :

androgen receptor

ARIH :

autosomal recessive icthyosis with hypotrichosis

BODIPY :

borondipyrromethene

CAF :

cancer-associated fibroblast

CAP1 :

channel-activating protease-1

CLL :

chronic lymphocytic leukemia

CML :

chronic myeloid leukemia

COX-2 :

cyclooxygenase-2

CRC :

colorectal cancer

CT :

computed tomography

DESC :

differentially expressed in squamous cell carcinoma

DMBA :

dimethylbenzanthracene

DHT :

dihydrotestosterone

ECM :

extracellular matrix

ECRG :

esophageal cancer-related gene

EGFR :

epidermal growth factor receptor

EMT :

epithelial to mesenchymal transition

EOC :

epithelial ovarian cancer

ERG :

estrogen-regulated gene

ERK :

extracellular signal-regulated kinase

ESCC :

esophageal squamous cell carcinoma

ETS :

erythroblast transformation specific

FAP :

fibroblast activation protein

GnT-V :

beta1,6-n-acetylglucosaminyltransferase-V

GPI :

glycosyl-phosphatidylinositol

HAI-1 :

hepatocyte growth factor inhibitor-1

HAI-2 :

hepatocyte growth factor inhibitor-2

HAT :

human airway trypsin-like

HGF :

hepatocyte growth factor

HGFA :

hepatocyte growth factor activator

HIF-1a :

hypoxia-inducible factor 1-alpha

HNSCC :

head and neck squamous cell carcinoma

IHC :

immunohistochemistry

IN-1 :

matriptase inhibitor-1

K5 :

keratin-5

K14 :

keratin-14

kbt :

ketobenzothiazole

KD :

knock-down

KD1-PEG :

PEGylated form of the Kunitz domain-1

KO :

knock-out

kt :

ketothiazole

Ln-332 :

laminin-332

LPB :

large probasin

LMP :

low malignant potential

Mab :

monoclonal antibody

MAPK :

mitogen-activated protein kinase

MCL :

mantle cell lymphoma

MCOTI-II :

Momordica cochinchinensis trypsin inhibitor-II

MMAE :

monomethyl auristatin-E

MMP :

matrix metalloprotease

MMTV :

mouse mammary tumor virus

MSP :

macrophage stimulating protein

mTor :

mammalian/mechanistic target of rapamycin

NF :

nuclear factor

NIR :

near-infrared

NPC :

nasopharyngeal carcinoma

NSCLC :

nonsmall cell lung cancer

OSCC :

oral squamous cell carcinoma

PAR-2 :

proteinase-activated receptor-2

PB :

probasin

PCI :

protein C inhibitor

PDAC :

pancreatic ductal adenocarcinoma

PDX :

patient-derived xenograft

PDGF :

platelet-derived growth factor

PDGFR :

platelet-derived growth factor receptor

PEG :

polyethylene glycol

PGE 2 :

prostaglandin E2

PI3K :

Phosphoinositide 3-kinase

PIN :

prostatic intraepithelial neoplasia

PN-1 :

protease nextin-1

PrAg :

protective antigen

PSA :

prostate-specific antigen

PymT :

polyomavirus middle T

RON :

Recepteur d’Origine Nantais

S1p :

sphingosine-1-phosphate

SAM :

S-adenosyl-l-methionine

SAR :

structure–activity relationship

SCC :

squamous cell carcinoma

SCID :

severe combined immunodeficiency

scFv :

single-chain variable fragment

SFTI-1 :

unflower-derived trypsin inhibitor

SNP :

single nucleotide polymorphism

SPECT :

single-photon emission computed tomography

Sphk1 :

sphingosine kinase 1

STAT3 :

signal transducer and activator of transcription 3

Tag :

T antigen

TMPRSS :

transmembrane protease, serine

TNBC :

triple-negative breast cancer

TRAMP :

transgenic adenocarcinoma of the mouse prostate

TTSP :

type II transmembrane serine protease

UPA :

urokinase type plasminogen activator

uPAR :

urokinase-type plasminogen activator receptor

WT :

wild-type

References

  1. Puente, X. S., Sanchez, L. M., Gutierrez-Fernandez, A., Velasco, G., & Lopez-Otin, C. (2005). A genomic view of the complexity of mammalian proteolytic systems. Biochemical Society Transactions, 33(Pt 2), 331–334. https://doi.org/10.1042/bst0330331.

    Article  CAS  PubMed  Google Scholar 

  2. Caughey, G. H. (2007). Mast cell tryptases and chymases in inflammation and host defense. Immunological Reviews, 217, 141–154. https://doi.org/10.1111/j.1600-065X.2007.00509.x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Netzel-Arnett, S., Hooper, J. D., Szabo, R., Madison, E. L., Quigley, J. P., Bugge, T. H., et al. (2003). Membrane anchored serine proteases: a rapidly expanding group of cell surface proteolytic enzymes with potential roles in cancer. Cancer Metastasis Reviews, 22(2–3), 237–258.

    Article  CAS  PubMed  Google Scholar 

  4. Busek, P., Mateu, R., Zubal, M., Kotackova, L., & Sedo, A. (2018). Targeting fibroblast activation protein in cancer—prospects and caveats. Frontiers in Bioscience (Landmark Ed), 23, 1933–1968.

    Article  CAS  Google Scholar 

  5. Zi, F., He, J., He, D., Li, Y., Yang, L., & Cai, Z. (2015). Fibroblast activation protein alpha in tumor microenvironment: recent progression and implications (review). Molecular Medicine Reports, 11(5), 3203–3211. https://doi.org/10.3892/mmr.2015.3197.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Antalis, T. M., Buzza, M. S., Hodge, K. M., Hooper, J. D., & Netzel-Arnett, S. (2010). The cutting edge: membrane-anchored serine protease activities in the pericellular microenvironment. The Biochemical Journal, 428(3), 325–346. https://doi.org/10.1042/BJ20100046.

    Article  CAS  PubMed  Google Scholar 

  7. Hooper, J. D., Clements, J. A., Quigley, J. P., & Antalis, T. M. (2001). Type II transmembrane serine proteases. Insights into an emerging class of cell surface proteolytic enzymes. The Journal of Biological Chemistry, 276(2), 857–860. https://doi.org/10.1074/jbc.R000020200.

    Article  CAS  PubMed  Google Scholar 

  8. Bugge, T. H., Antalis, T. M., & Wu, Q. (2009). Type II transmembrane serine proteases. The Journal of Biological Chemistry, 284(35), 23177–23181. https://doi.org/10.1074/jbc.R109.021006.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Bugge, T. H., List, K., & Szabo, R. (2007). Matriptase-dependent cell surface proteolysis in epithelial development and pathogenesis. Frontiers in Bioscience, 12, 5060–5070.

    Article  CAS  PubMed  Google Scholar 

  10. Szabo, R., & Bugge, T. H. (2008). Type II transmembrane serine proteases in development and disease. The International Journal of Biochemistry & Cell Biology, 40(6–7), 1297–1316. https://doi.org/10.1016/j.biocel.2007.11.013.

    Article  CAS  Google Scholar 

  11. Szabo, R., & Bugge, T. H. (2011). Membrane-anchored serine proteases in vertebrate cell and developmental biology. Annual Review of Cell and Developmental Biology, 27, 213–235. https://doi.org/10.1146/annurev-cellbio-092910-154247.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Murray, A. S., Varela, F. A., & List, K. (2016). Type II transmembrane serine proteases as potential targets for cancer therapy. Biological Chemistry, 397(9), 815–826. https://doi.org/10.1515/hsz-2016-0131.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Tanabe, L. M., & List, K. (2017). The role of type II transmembrane serine protease-mediated signaling in cancer. The FEBS Journal, 284(10), 1421–1436. https://doi.org/10.1111/febs.13971.

    Article  CAS  PubMed  Google Scholar 

  14. Antalis, T. M., Bugge, T. H., & Wu, Q. (2011). Membrane-anchored serine proteases in health and disease. Progress in Molecular Biology and Translational Science, 99, 1–50. https://doi.org/10.1016/b978-0-12-385504-6.00001-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Takeuchi, T., Harris, J. L., Huang, W., Yan, K. W., Coughlin, S. R., & Craik, C. S. (2000). Cellular localization of membrane-type serine protease 1 and identification of protease-activated receptor-2 and single-chain urokinase-type plasminogen activator as substrates. The Journal of Biological Chemistry, 275(34), 26333–26342. https://doi.org/10.1074/jbc.M002941200.

    Article  CAS  PubMed  Google Scholar 

  16. Jiang, J., Yang, J., Feng, P., Zuo, B., Dong, N., Wu, Q., et al. (2014). N-glycosylation is required for matriptase-2 autoactivation and ectodomain shedding. The Journal of Biological Chemistry, 289(28), 19500–19507. https://doi.org/10.1074/jbc.M114.555110.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Qiu, D., Owen, K., Gray, K., Bass, R., & Ellis, V. (2007). Roles and regulation of membrane-associated serine proteases. Biochemical Society Transactions, 35(Pt 3), 583–587. https://doi.org/10.1042/bst0350583.

    Article  CAS  PubMed  Google Scholar 

  18. Afar, D. E., Vivanco, I., Hubert, R. S., Kuo, J., Chen, E., Saffran, D. C., et al. (2001). Catalytic cleavage of the androgen-regulated TMPRSS2 protease results in its secretion by prostate and prostate cancer epithelia. Cancer Research, 61(4), 1686–1692.

    CAS  PubMed  Google Scholar 

  19. Guipponi, M., Vuagniaux, G., Wattenhofer, M., Shibuya, K., Vazquez, M., Dougherty, L., et al. (2002). The transmembrane serine protease (TMPRSS3) mutated in deafness DFNB8/10 activates the epithelial sodium channel (ENaC) in vitro. Human Molecular Genetics, 11(23), 2829–2836. https://doi.org/10.1093/hmg/11.23.2829.

    Article  CAS  PubMed  Google Scholar 

  20. Andreasen, D., Vuagniaux, G., Fowler-Jaeger, N., Hummler, E., & Rossier, B. C. (2006). Activation of epithelial sodium channels by mouse channel activating proteases (mCAP) expressed in Xenopus oocytes requires catalytic activity of mCAP3 and mCAP2 but not mCAP1. Journal of the American Society of Nephrology, 17(4), 968–976. https://doi.org/10.1681/asn.2005060637.

    Article  CAS  PubMed  Google Scholar 

  21. Murray, A. S., Varela, F. A., Hyland, T. E., Schoenbeck, A. J., White, J. M., Tanabe, L. M., et al. (2017). Phosphorylation of the type II transmembrane serine protease, TMPRSS13, in hepatocyte growth factor activator inhibitor-1 and -2-mediated cell-surface localization. The Journal of Biological Chemistry, 292(36), 14867–14884. https://doi.org/10.1074/jbc.M117.775999.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Oberst, M. D., Williams, C. A., Dickson, R. B., Johnson, M. D., & Lin, C. Y. (2003). The activation of matriptase requires its noncatalytic domains, serine protease domain, and its cognate inhibitor. The Journal of Biological Chemistry, 278(29), 26773–26779. https://doi.org/10.1074/jbc.M304282200.

    Article  CAS  PubMed  Google Scholar 

  23. Tseng, I. C., Xu, H., Chou, F. P., Li, G., Vazzano, A. P., Kao, J. P., et al. (2010). Matriptase activation, an early cellular response to acidosis. The Journal of Biological Chemistry, 285(5), 3261–3270. https://doi.org/10.1074/jbc.M109.055640.

    Article  CAS  PubMed  Google Scholar 

  24. Godiksen, S., Soendergaard, C., Friis, S., Jensen, J. K., Bornholdt, J., Sales, K. U., et al. (2013). Detection of active matriptase using a biotinylated chloromethyl ketone peptide. PLoS One, 8(10), e77146. https://doi.org/10.1371/journal.pone.0077146.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Tamberg, T., Hong, Z., De Schepper, D., Skovbjerg, S., Dupont, D. M., Vitved, L., et al. (2019). Blocking the proteolytic activity of zymogen matriptase with antibody-based inhibitors. The Journal of Biological Chemistry, 294(1), 314–326. https://doi.org/10.1074/jbc.RA118.004126.

    Article  CAS  PubMed  Google Scholar 

  26. Inouye, K., Yasumoto, M., Tsuzuki, S., Mochida, S., & Fushiki, T. (2010). The optimal activity of a pseudozymogen form of recombinant matriptase under the mildly acidic pH and low ionic strength conditions. Journal of Biochemistry, 147(4), 485–492. https://doi.org/10.1093/jb/mvp190.

    Article  CAS  PubMed  Google Scholar 

  27. Friis, S., Tadeo, D., Le-Gall, S. M., Jurgensen, H. J., Sales, K. U., Camerer, E., et al. (2017). Matriptase zymogen supports epithelial development, homeostasis and regeneration. BMC Biology, 15(1), 46. https://doi.org/10.1186/s12915-017-0384-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Kawaguchi, T., Qin, L., Shimomura, T., Kondo, J., Matsumoto, K., Denda, K., et al. (1997). Purification and cloning of hepatocyte growth factor activator inhibitor type 2, a Kunitz-type serine protease inhibitor. The Journal of Biological Chemistry, 272(44), 27558–27564. https://doi.org/10.1074/jbc.272.44.27558.

    Article  CAS  PubMed  Google Scholar 

  29. Shimomura, T., Denda, K., Kitamura, A., Kawaguchi, T., Kito, M., Kondo, J., et al. (1997). Hepatocyte growth factor activator inhibitor, a novel Kunitz-type serine protease inhibitor. The Journal of Biological Chemistry, 272(10), 6370–6376.

    Article  CAS  PubMed  Google Scholar 

  30. Kataoka, H., Kawaguchi, M., Fukushima, T., & Shimomura, T. (2018). Hepatocyte growth factor activator inhibitors (HAI-1 and HAI-2): emerging key players in epithelial integrity and cancer. Pathology International, 68(3), 145–158. https://doi.org/10.1111/pin.12647.

    Article  CAS  PubMed  Google Scholar 

  31. Yu, J. X., Chao, L., & Chao, J. (1994). Prostasin is a novel human serine proteinase from seminal fluid. Purification, tissue distribution, and localization in prostate gland. The Journal of Biological Chemistry, 269(29), 18843–18848.

    CAS  PubMed  Google Scholar 

  32. Chen, L. M., Skinner, M. L., Kauffman, S. W., Chao, J., Chao, L., Thaler, C. D., et al. (2001). Prostasin is a glycosylphosphatidylinositol-anchored active serine protease. The Journal of Biological Chemistry, 276(24), 21434–21442. https://doi.org/10.1074/jbc.M011423200.

    Article  CAS  PubMed  Google Scholar 

  33. Szabo, R., Hobson, J. P., List, K., Molinolo, A., Lin, C. Y., & Bugge, T. H. (2008). Potent inhibition and global co-localization implicate the transmembrane Kunitz-type serine protease inhibitor hepatocyte growth factor activator inhibitor-2 in the regulation of epithelial matriptase activity. The Journal of Biological Chemistry, 283(43), 29495–29504. https://doi.org/10.1074/jbc.M801970200.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Chen, Y. W., Wang, J. K., Chou, F. P., Chen, C. Y., Rorke, E. A., Chen, L. M., et al. (2010). Regulation of the matriptase-prostasin cell surface proteolytic cascade by hepatocyte growth factor activator inhibitor-1 during epidermal differentiation. The Journal of Biological Chemistry, 285(41), 31755–31762. https://doi.org/10.1074/jbc.M110.150367.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Fan, B., Wu, T. D., Li, W., & Kirchhofer, D. (2005). Identification of hepatocyte growth factor activator inhibitor-1B as a potential physiological inhibitor of prostasin. The Journal of Biological Chemistry, 280(41), 34513–34520. https://doi.org/10.1074/jbc.M502119200.

    Article  CAS  PubMed  Google Scholar 

  36. Inoue, M., Kanbe, N., Kurosawa, M., & Kido, H. (1998). Cloning and tissue distribution of a novel serine protease esp-1 from human eosinophils. Biochemical and Biophysical Research Communications, 252(2), 307–312. https://doi.org/10.1006/bbrc.1998.9645.

    Article  CAS  PubMed  Google Scholar 

  37. Hooper, J. D., Nicol, D. L., Dickinson, J. L., Eyre, H. J., Scarman, A. L., Normyle, J. F., et al. (1999). Testisin, a new human serine proteinase expressed by premeiotic testicular germ cells and lost in testicular germ cell tumors. Cancer Research, 59(13), 3199–3205.

    CAS  PubMed  Google Scholar 

  38. Tang, T., Kmet, M., Corral, L., Vartanian, S., Tobler, A., & Papkoff, J. (2005). Testisin, a glycosyl-phosphatidylinositol-linked serine protease, promotes malignant transformation in vitro and in vivo. Cancer Research, 65(3), 868–878.

    CAS  PubMed  Google Scholar 

  39. Kempkensteffen, C., Christoph, F., Weikert, S., Krause, H., Kollermann, J., Schostak, M., et al. (2006). Epigenetic silencing of the putative tumor suppressor gene testisin in testicular germ cell tumors. Journal of Cancer Research and Clinical Oncology, 132(12), 765–770. https://doi.org/10.1007/s00432-006-0124-6.

    Article  CAS  PubMed  Google Scholar 

  40. Manton, K. J., Douglas, M. L., Netzel-Arnett, S., Fitzpatrick, D. R., Nicol, D. L., Boyd, A. W., et al. (2015). Hypermethylation of the 5’ CpG island of the gene encoding the serine protease Testisin promotes its loss in testicular tumorigenesis. British Journal of Cancer, 113(11), 1640. https://doi.org/10.1038/bjc.2015.384.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. List, K., Szabo, R., Molinolo, A., Sriuranpong, V., Redeye, V., Murdock, T., et al. (2005). Deregulated matriptase causes ras-independent multistage carcinogenesis and promotes ras-mediated malignant transformation. Genes & Development, 19(16), 1934–1950. https://doi.org/10.1101/gad.1300705.

    Article  CAS  Google Scholar 

  42. Bocheva, G., Rattenholl, A., Kempkes, C., Goerge, T., Lin, C. Y., D'Andrea, M. R., et al. (2009). Role of matriptase and proteinase-activated receptor-2 in nonmelanoma skin cancer. The Journal of Investigative Dermatology, 129(7), 1816–1823. https://doi.org/10.1038/jid.2008.449.

    Article  CAS  PubMed  Google Scholar 

  43. Sales, K. U., Friis, S., Abusleme, L., Moutsopoulos, N. M., & Bugge, T. H. (2015). Matriptase promotes inflammatory cell accumulation and progression of established epidermal tumors. Oncogene, 34(35), 4664–4672. https://doi.org/10.1038/onc.2014.391.

    Article  CAS  PubMed  Google Scholar 

  44. Sales, K. U., Friis, S., Konkel, J. E., Godiksen, S., Hatakeyama, M., Hansen, K. K., et al. (2015). Non-hematopoietic PAR-2 is essential for matriptase-driven pre-malignant progression and potentiation of ras-mediated squamous cell carcinogenesis. Oncogene, 34(3), 346–356. https://doi.org/10.1038/onc.2013.563.

    Article  CAS  PubMed  Google Scholar 

  45. Szabo, R., Rasmussen, A. L., Moyer, A. B., Kosa, P., Schafer, J. M., Molinolo, A. A., et al. (2011). c-Met-induced epithelial carcinogenesis is initiated by the serine protease matriptase. Oncogene, 30(17), 2003–2016. https://doi.org/10.1038/onc.2010.586.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Baba, T., Kawaguchi, M., Fukushima, T., Sato, Y., Orikawa, H., Yorita, K., et al. (2012). Loss of membrane-bound serine protease inhibitor HAI-1 induces oral squamous cell carcinoma cells’ invasiveness. The Journal of Pathology, 228(2), 181–192. https://doi.org/10.1002/path.3993.

    Article  CAS  PubMed  Google Scholar 

  47. Yamamoto, K., Kawaguchi, M., Shimomura, T., Izumi, A., Konari, K., Honda, A., et al. (2018). Hepatocyte growth factor activator inhibitor type-2 (HAI-2)/SPINT2 contributes to invasive growth of oral squamous cell carcinoma cells. Oncotarget, 9(14), 11691–11706. https://doi.org/10.18632/oncotarget.24450.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Friis, S., Uzzun Sales, K., Godiksen, S., Peters, D. E., Lin, C. Y., Vogel, L. K., et al. (2013). A matriptase-prostasin reciprocal zymogen activation complex with unique features: prostasin as a non-enzymatic co-factor for matriptase activation. The Journal of Biological Chemistry, 288(26), 19028–19039. https://doi.org/10.1074/jbc.M113.469932.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Kanemaru, A., Yamamoto, K., Kawaguchi, M., Fukushima, T., Lin, C. Y., Johnson, M. D., et al. (2017). Deregulated matriptase activity in oral squamous cell carcinoma promotes the infiltration of cancer-associated fibroblasts by paracrine activation of protease-activated receptor 2. International Journal of Cancer, 140(1), 130–141. https://doi.org/10.1002/ijc.30426.

    Article  CAS  PubMed  Google Scholar 

  50. Cheng, M. F., Huang, M. S., Lin, C. S., Lin, L. H., Lee, H. S., Jiang, J. C., et al. (2014). Expression of matriptase correlates with tumour progression and clinical prognosis in oral squamous cell carcinoma. Histopathology, 65(1), 24–34. https://doi.org/10.1111/his.12361.

    Article  PubMed  Google Scholar 

  51. Lang, J. C., & Schuller, D. E. (2001). Differential expression of a novel serine protease homologue in squamous cell carcinoma of the head and neck. British Journal of Cancer, 84(2), 237–243. https://doi.org/10.1054/bjoc.2000.1586.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Sedghizadeh, P. P., Mallery, S. R., Thompson, S. J., Kresty, L., Beck, F. M., Parkinson, E. K., et al. (2006). Expression of the serine protease DESC1 correlates directly with normal keratinocyte differentiation and inversely with head and neck squamous cell carcinoma progression. Head & Neck, 28(5), 432–440. https://doi.org/10.1002/hed.20346.

    Article  Google Scholar 

  53. Ng, H. Y., Ko, J. M., Yu, V. Z., Ip, J. C., Dai, W., Cal, S., et al. (2016). DESC1, a novel tumor suppressor, sensitizes cells to apoptosis by downregulating the EGFR/AKT pathway in esophageal squamous cell carcinoma. International Journal of Cancer, 138(12), 2940–2951. https://doi.org/10.1002/ijc.30034.

    Article  CAS  PubMed  Google Scholar 

  54. Chang, Z.-W., Jia, Y.-X., Zhang, W.-J., Song, L.-J., Gao, M., Li, M.-J., et al. (2018). LncRNA-TUSC7/miR-224 affected chemotherapy resistance of esophageal squamous cell carcinoma by competitively regulating DESC1. Journal of Experimental & Clinical Cancer Research: CR, 37(1), 56–56. https://doi.org/10.1186/s13046-018-0724-4.

    Article  CAS  PubMed Central  Google Scholar 

  55. Duhaime, M. J., Page, K. O., Varela, F. A., Murray, A. S., Silverman, M. E., Zoratti, G. L., et al. (2016). Cell surface human airway trypsin-like protease is lost during squamous cell carcinogenesis. Journal of Cellular Physiology, 231(7), 1476–1483. https://doi.org/10.1002/jcp.25173.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Miller, G. S., Zoratti, G. L., Murray, A. S., Bergum, C., Tanabe, L. M., & List, K. (2014). HATL5: a cell surface serine protease differentially expressed in epithelial cancers. PLoS One, 9(2), e87675. https://doi.org/10.1371/journal.pone.0087675.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Wang, J. Y., Jin, X., & Li, X. F. (2018). Knockdown of TMPRSS3, a transmembrane serine protease, inhibits proliferation, migration, and invasion in human nasopharyngeal carcinoma cells. Oncology Research, 26(1), 95–101. https://doi.org/10.3727/096504017x14920318811695.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Bao, Y., Wang, Q., Guo, Y., Chen, Z., Li, K., Yang, Y., et al. (2016). PRSS8 methylation and its significance in esophageal squamous cell carcinoma. Oncotarget, 7(19), 28540–28555. https://doi.org/10.18632/oncotarget.8677.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Lin, C. Y., Wang, J. K., Torri, J., Dou, L., Sang, Q. A., & Dickson, R. B. (1997). Characterization of a novel, membrane-bound, 80-kDa matrix-degrading protease from human breast cancer cells. Monoclonal antibody production, isolation, and localization. The Journal of Biological Chemistry, 272(14), 9147–9152.

    Article  CAS  PubMed  Google Scholar 

  60. Jin, J. S., Cheng, T. F., Tsai, W. C., Sheu, L. F., Chiang, H., & Yu, C. P. (2007). Expression of the serine protease, matriptase, in breast ductal carcinoma of Chinese women: correlation with clinicopathological parameters. Histology and Histopathology, 22(3), 305–309.

    CAS  PubMed  Google Scholar 

  61. Kang, J. Y., Dolled-Filhart, M., Ocal, I. T., Singh, B., Lin, C. Y., Dickson, R. B., et al. (2003). Tissue microarray analysis of hepatocyte growth factor/Met pathway components reveals a role for Met, matriptase, and hepatocyte growth factor activator inhibitor 1 in the progression of node-negative breast cancer. Cancer Research, 63(5), 1101–1105.

    CAS  PubMed  Google Scholar 

  62. Oberst, M., Anders, J., Xie, B., Singh, B., Ossandon, M., Johnson, M., et al. (2001). Matriptase and HAI-1 are expressed by normal and malignant epithelial cells in vitro and in vivo. The American Journal of Pathology, 158(4), 1301–1311. https://doi.org/10.1016/S0002-9440(10)64081-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Siddiqui, S. F., Pawelek, J., Handerson, T., Lin, C. Y., Dickson, R. B., Rimm, D. L., et al. (2005). Coexpression of beta1,6-N-acetylglucosaminyltransferase V glycoprotein substrates defines aggressive breast cancers with poor outcome. Cancer Epidemiology, Biomarkers & Prevention, 14(11 Pt 1), 2517–2523. https://doi.org/10.1158/1055-9965.epi-05-0464.

    Article  CAS  Google Scholar 

  64. Parr, C., Watkins, G., Mansel, R. E., & Jiang, W. G. (2004). The hepatocyte growth factor regulatory factors in human breast cancer. Clinical Cancer Research, 10(1 Pt 1), 202–211.

    Article  CAS  PubMed  Google Scholar 

  65. Zoratti, G. L., Tanabe, L. M., Varela, F. A., Murray, A. S., Bergum, C., Colombo, E., et al. (2015). Targeting matriptase in breast cancer abrogates tumour progression via impairment of stromal-epithelial growth factor signalling. Nature Communications, 6, 6776. https://doi.org/10.1038/ncomms7776.

    Article  CAS  PubMed  Google Scholar 

  66. Zoratti, G. L., Tanabe, L. M., Hyland, T. E., Duhaime, M. J., Colombo, E., Leduc, R., et al. (2016). Matriptase regulates c-Met mediated proliferation and invasion in inflammatory breast cancer. Oncotarget, 7(36), 58162–58173. https://doi.org/10.18632/oncotarget.11262.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Hurst, N. J., Jr., Najy, A. J., Ustach, C. V., Movilla, L., & Kim, H. R. (2012). Platelet-derived growth factor-C (PDGF-C) activation by serine proteases: implications for breast cancer progression. The Biochemical Journal, 441(3), 909–918. https://doi.org/10.1042/bj20111020.

    Article  CAS  PubMed  Google Scholar 

  68. Parr, C., Sanders, A. J., Davies, G., Martin, T., Lane, J., Mason, M. D., et al. (2007). Matriptase-2 inhibits breast tumor growth and invasion and correlates with favorable prognosis for breast cancer patients. Clinical Cancer Research, 13(12), 3568–3576. https://doi.org/10.1158/1078-0432.ccr-06-2357.

    Article  CAS  PubMed  Google Scholar 

  69. Tuhkanen, H., Hartikainen, J. M., Soini, Y., Velasco, G., Sironen, R., Nykopp, T. K., et al. (2013). Matriptase-2 gene (TMPRSS6) variants associate with breast cancer survival, and reduced expression is related to triple-negative breast cancer. International Journal of Cancer, 133(10), 2334–2340. https://doi.org/10.1002/ijc.28254.

    Article  CAS  PubMed  Google Scholar 

  70. Xing, P., Li, J. G., Jin, F., Zhao, T. T., Liu, Q., Dong, H. T., et al. (2011). Clinical and biological significance of hepsin overexpression in breast cancer. Journal of Investigative Medicine, 59(5), 803–810. https://doi.org/10.2310/JIM.0b013e31821451a1.

    Article  CAS  PubMed  Google Scholar 

  71. Tervonen, T. A., Belitškin, D., Pant, S. M., Englund, J. I., Marques, E., Ala-Hongisto, H., et al. (2016). Deregulated hepsin protease activity confers oncogenicity by concomitantly augmenting HGF/MET signalling and disrupting epithelial cohesion. Oncogene, 35(14), 1832–1846. https://doi.org/10.1038/onc.2015.248.

    Article  CAS  PubMed  Google Scholar 

  72. Pant, S. M., Belitskin, D., Ala-Hongisto, H., Klefstrom, J., & Tervonen, T. A. (2018). Analyzing the type II transmembrane serine protease hepsin-dependent basement membrane remodeling in 3D cell culture. Methods in Molecular Biology, 1731, 169–178. https://doi.org/10.1007/978-1-4939-7595-2_16.

    Article  CAS  PubMed  Google Scholar 

  73. Damalanka, V. C., Han, Z., Karmakar, P., O'Donoghue, A. J., La Greca, F., Kim, T., et al. (2019). Discovery of selective matriptase and hepsin serine protease inhibitors: useful chemical tools for cancer cell biology. Journal of Medicinal Chemistry, 62(2), 480–490. https://doi.org/10.1021/acs.jmedchem.8b01536.

    Article  CAS  PubMed  Google Scholar 

  74. Rui, X., Li, Y., Jin, F., & Li, F. (2015). TMPRSS3 is a novel poor prognostic factor for breast cancer. International Journal of Clinical and Experimental Pathology, 8(5), 5435–5442.

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Luo, P., Lu, G., Fan, L. L., Zhong, X., Yang, H., Xie, R., et al. (2017). Dysregulation of TMPRSS3 and TNFRSF11B correlates with tumorigenesis and poor prognosis in patients with breast cancer. Oncology Reports, 37(4), 2057–2062. https://doi.org/10.3892/or.2017.5449.

    Article  CAS  PubMed  Google Scholar 

  76. Pelkonen, M., Luostari, K., Tengström, M., Ahonen, H., Berdel, B., Kataja, V., et al. (2015). Low expression levels of hepsin and TMPRSS3 are associated with poor breast cancer survival. BMC Cancer, 15, 431–431. https://doi.org/10.1186/s12885-015-1440-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Chen, L. M., & Chai, K. X. (2002). Prostasin serine protease inhibits breast cancer invasiveness and is transcriptionally regulated by promoter DNA methylation. International Journal of Cancer, 97(3), 323–329.

    Article  CAS  PubMed  Google Scholar 

  78. Bergum, C., Zoratti, G., Boerner, J., & List, K. (2012). Strong expression association between matriptase and its substrate prostasin in breast cancer. Journal of Cellular Physiology, 227(4), 1604–1609. https://doi.org/10.1002/jcp.22877.

    Article  CAS  PubMed  Google Scholar 

  79. Vogel, L. K., Saebo, M., Skjelbred, C. F., Abell, K., Pedersen, E. D., Vogel, U., et al. (2006). The ratio of matriptase/HAI-1 mRNA is higher in colorectal cancer adenomas and carcinomas than corresponding tissue from control individuals. BMC Cancer, 6, 176. https://doi.org/10.1186/1471-2407-6-176.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Tsai, W. C., Sheu, L. F., Chao, Y. C., Chen, A., Chiang, H., & Jin, J. S. (2007). Decreased matriptase/HAI-1 ratio in advanced colorectal adenocarcinoma of Chinese patients. The Chinese Journal of Physiology, 50(5), 225–231.

    PubMed  Google Scholar 

  81. Forbs, D., Thiel, S., Stella, M. C., Sturzebecher, A., Schweinitz, A., Steinmetzer, T., et al. (2005). In vitro inhibition of matriptase prevents invasive growth of cell lines of prostate and colon carcinoma. International Journal of Oncology, 27(4), 1061–1070.

    PubMed  Google Scholar 

  82. List, K., Haudenschild, C. C., Szabo, R., Chen, W., Wahl, S. M., Swaim, W., et al. (2002). Matriptase/MT-SP1 is required for postnatal survival, epidermal barrier function, hair follicle development, and thymic homeostasis. Oncogene, 21(23), 3765–3779. https://doi.org/10.1038/sj.onc.1205502.

    Article  CAS  PubMed  Google Scholar 

  83. Kosa, P., Szabo, R., Molinolo, A. A., & Bugge, T. H. (2012). Suppression of tumorigenicity-14, encoding matriptase, is a critical suppressor of colitis and colitis-associated colon carcinogenesis. Oncogene, 31(32), 3679–3695. https://doi.org/10.1038/onc.2011.545.

    Article  CAS  PubMed  Google Scholar 

  84. Huang, A., Zhou, H., Zhao, H., Quan, Y., Feng, B., & Zheng, M. (2014). TMPRSS4 correlates with colorectal cancer pathological stage and regulates cell proliferation and self-renewal ability. Cancer Biology & Therapy, 15(3), 297–304. https://doi.org/10.4161/cbt.27308.

    Article  CAS  Google Scholar 

  85. Kim, S., Kang, H. Y., Nam, E. H., Choi, M. S., Zhao, X. F., Hong, C. S., et al. (2010). TMPRSS4 induces invasion and epithelial-mesenchymal transition through upregulation of integrin alpha5 and its signaling pathways. Carcinogenesis, 31(4), 597–606. https://doi.org/10.1093/carcin/bgq024.

    Article  CAS  PubMed  Google Scholar 

  86. Selzer-Plon, J., Bornholdt, J., Friis, S., Bisgaard, H. C., Lothe, I. M., Tveit, K. M., et al. (2009). Expression of prostasin and its inhibitors during colorectal cancer carcinogenesis. BMC Cancer, 9, 201. https://doi.org/10.1186/1471-2407-9-201.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Bao, Y., Li, K., Guo, Y., Wang, Q., Li, Z., Yang, Y., et al. (2016). Tumor suppressor PRSS8 targets Sphk1/S1P/Stat3/Akt signaling in colorectal cancer. Oncotarget, 7(18), 26780–26792. https://doi.org/10.18632/oncotarget.8511.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Bao, Y., Guo, Y., Yang, Y., Wei, X., Zhang, S., Zhang, Y., et al. (2019). PRSS8 suppresses colorectal carcinogenesis and metastasis. Oncogene.

  89. List, K., Szabo, R., Wertz, P. W., Segre, J., Haudenschild, C. C., Kim, S. Y., et al. (2003). Loss of proteolytically processed filaggrin caused by epidermal deletion of matriptase/MT-SP1. The Journal of Cell Biology, 163(4), 901–910. https://doi.org/10.1083/jcb.200304161.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Leyvraz, C., Charles, R. P., Rubera, I., Guitard, M., Rotman, S., Breiden, B., et al. (2005). The epidermal barrier function is dependent on the serine protease CAP1/Prss8. The Journal of Cell Biology, 170(3), 487–496. https://doi.org/10.1083/jcb.200501038.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Tanimoto, H., Underwood, L. J., Wang, Y., Clarke, J., & O’Brien, T. J. Cloning and expression of TADG-15, a novel serine protease expressed in ovarian cancer. 1998 Proc. Am. Assoc. Cancer Res., 39, 648.

  92. Tanimoto, H., Shigemasa, K., Tian, X., Gu, L., Beard, J. B., Sawasaki, T., et al. (2005). Transmembrane serine protease TADG-15 (ST14/matriptase/MT-SP1): expression and prognostic value in ovarian cancer. British Journal of Cancer, 92(2), 278–283. https://doi.org/10.1038/sj.bjc.6602320.

    Article  CAS  PubMed  Google Scholar 

  93. Ji, M., Li, S., Xie, Y., Zhao, Z., Chang, W., Li, Y., et al. (2017). Expression and prognostic value of matriptase in ovarian serous adenocarcinoma. Oncology Letters, 13(3), 1741–1744. https://doi.org/10.3892/ol.2017.5600.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Oberst, M. D., Johnson, M. D., Dickson, R. B., Lin, C. Y., Singh, B., Stewart, M., et al. (2002). Expression of the serine protease matriptase and its inhibitor HAI-1 in epithelial ovarian cancer: correlation with clinical outcome and tumor clinicopathological parameters. Clinical Cancer Research, 8(4), 1101–1107.

    CAS  PubMed  Google Scholar 

  95. Johnson, M. D., Oberst, M. D., Lin, C. Y., & Dickson, R. B. (2003). Possible role of matriptase in the diagnosis of ovarian cancer. Expert Review of Molecular Diagnostics, 3(3), 331–338. https://doi.org/10.1586/14737159.3.3.331.

    Article  CAS  PubMed  Google Scholar 

  96. Nakamura, K., Abarzua, F., Kodama, J., Hongo, A., Nasu, Y., Kumon, H., et al. (2009). Expression of hepatocyte growth factor activator inhibitors (HAI-1 and HAI-2) in ovarian cancer. International Journal of Oncology, 34(2), 345–353.

    CAS  PubMed  Google Scholar 

  97. Sun, P., Jiang, Z., Chen, X., Xue, L., Mao, X., Ruan, G., et al. (2016). Decreasing the ratio of matriptase/HAI-1 by downregulation of matriptase as a potential adjuvant therapy in ovarian cancer. Molecular Medicine Reports, 14(2), 1465–1474. https://doi.org/10.3892/mmr.2016.5435.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Suzuki, M., Kobayashi, H., Kanayama, N., Saga, Y., Lin, C. Y., Dickson, R. B., et al. (2004). Inhibition of tumor invasion by genomic down-regulation of matriptase through suppression of activation of receptor-bound pro-urokinase. The Journal of Biological Chemistry, 279(15), 14899–14908. https://doi.org/10.1074/jbc.M313130200.

    Article  CAS  PubMed  Google Scholar 

  99. Underwood, L. J., Shigemasa, K., Tanimoto, H., Beard, J. B., Schneider, E. N., Wang, Y., et al. (2000). Ovarian tumor cells express a novel multi-domain cell surface serine protease. Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease, 1502(3), 337–350. https://doi.org/10.1016/S0925-4439(00)00058-2.

    Article  CAS  Google Scholar 

  100. Guerrero, K., Wang, Z., Bachvarova, M., Gregoire, J., Renaud, M.-C., Plante, M., et al. (2012). A novel genome-based approach correlates TMPRSS3 overexpression in ovarian cancer with DNA hypomethylation. Gynecologic Oncology, 125(3), 720–726. https://doi.org/10.1016/j.ygyno.2012.03.026.

    Article  CAS  PubMed  Google Scholar 

  101. Zhang, D., Qiu, S., Wang, Q., & Zheng, J. (2016). TMPRSS3 modulates ovarian cancer cell proliferation, invasion and metastasis. Oncology Reports, 35(1), 81–88. https://doi.org/10.3892/or.2015.4356.

    Article  CAS  PubMed  Google Scholar 

  102. Shigemasa, K., Underwood, L. J., Beard, J., Tanimoto, H., Ohama, K., Parmley, T. H., et al. (2000). Overexpression of testisin, a serine protease expressed by testicular germ cells, in epithelial ovarian tumor cells. Journal of the Society for Gynecologic Investigation, 7(6), 358–362.

    CAS  PubMed  Google Scholar 

  103. Bignotti, E., Tassi, R. A., Calza, S., Ravaggi, A., Bandiera, E., Rossi, E., et al. (2007). Gene expression profile of ovarian serous papillary carcinomas: identification of metastasis-associated genes. American Journal of Obstetrics and Gynecology, 196(3), 245.e241–245.e211. https://doi.org/10.1016/j.ajog.2006.10.874.

    Article  CAS  Google Scholar 

  104. Conway, G. D., Buzza, M. S., Martin, E. W., Duru, N., Johnson, T. A., Peroutka, R. J., et al. (2019). PRSS21/testisin inhibits ovarian tumor metastasis and antagonizes proangiogenic angiopoietins ANG2 and ANGPTL4. Journal of Molecular Medicine, 97(5), 691–709. https://doi.org/10.1007/s00109-019-01763-3.

    Article  CAS  PubMed  Google Scholar 

  105. Martin, E. W., Buzza, M. S., Driesbaugh, K. H., Liu, S., Fortenberry, Y. M., Leppla, S. H., et al. (2015). Targeting the membrane-anchored serine protease testisin with a novel engineered anthrax toxin prodrug to kill tumor cells and reduce tumor burden. Oncotarget, 6(32), 33534–33553. https://doi.org/10.18632/oncotarget.5214.

    Article  PubMed  PubMed Central  Google Scholar 

  106. Riddick, A. C., Shukla, C. J., Pennington, C. J., Bass, R., Nuttall, R. K., Hogan, A., et al. (2005). Identification of degradome components associated with prostate cancer progression by expression analysis of human prostatic tissues. British Journal of Cancer, 92(12), 2171–2180. https://doi.org/10.1038/sj.bjc.6602630.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Saleem, M., Adhami, V. M., Zhong, W., Longley, B. J., Lin, C. Y., Dickson, R. B., et al. (2006). A novel biomarker for staging human prostate adenocarcinoma: overexpression of matriptase with concomitant loss of its inhibitor, hepatocyte growth factor activator inhibitor-1. Cancer Epidemiology, Biomarkers & Prevention, 15(2), 217–227. https://doi.org/10.1158/1055-9965.EPI-05-0737.

    Article  CAS  Google Scholar 

  108. Sanders, A. J., Parr, C., Davies, G., Martin, T. A., Lane, J., Mason, M. D., et al. (2006). Genetic reduction of matriptase-1 expression is associated with a reduction in the aggressive phenotype of prostate cancer cells in vitro and in vivo. Journal of Experimental Therapeutics & Oncology, 6(1), 39–48.

    CAS  Google Scholar 

  109. Warren, M., Twohig, M., Pier, T., Eickhoff, J., Lin, C. Y., Jarrard, D., et al. (2009). Protein expression of matriptase and its cognate inhibitor HAI-1 in human prostate cancer: a tissue microarray and automated quantitative analysis. Applied Immunohistochemistry & Molecular Morphology, 17(1), 23–30. https://doi.org/10.1097/PAI.0b013e31817c3334.

    Article  CAS  Google Scholar 

  110. Sanders, A. J., Parr, C., Mason, M. D., & Jiang, W. G. (2007). Suppression of hepatocyte growth factor activator inhibitor-1 leads to a more aggressive phenotype of prostate cancer cells in vitro. International Journal of Molecular Medicine, 20(4), 613–619.

    CAS  PubMed  Google Scholar 

  111. Bergum, C., & List, K. (2010). Loss of the matriptase inhibitor HAI-2 during prostate cancer progression. Prostate, 70(13), 1422–1428. https://doi.org/10.1002/pros.21177.

    Article  CAS  PubMed  Google Scholar 

  112. Tsai, C. H., Teng, C. H., Tu, Y. T., Cheng, T. S., Wu, S. R., Ko, C. J., et al. (2014). HAI-2 suppresses the invasive growth and metastasis of prostate cancer through regulation of matriptase. Oncogene, 33(38), 4643–4652. https://doi.org/10.1038/onc.2013.412.

    Article  CAS  PubMed  Google Scholar 

  113. Wu, S. R., Teng, C. H., Tu, Y. T., Ko, C. J., Cheng, T. S., Lan, S. W., et al. (2017). The Kunitz domain I of hepatocyte growth factor activator inhibitor-2 inhibits matriptase activity and invasive ability of human prostate cancer cells. Scientific Reports, 7(1), 15101. https://doi.org/10.1038/s41598-017-15415-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Owen, K. A., Qiu, D., Alves, J., Schumacher, A. M., Kilpatrick, L. M., Li, J., et al. (2010). Pericellular activation of hepatocyte growth factor by the transmembrane serine proteases matriptase and hepsin, but not by the membrane-associated protease uPA. The Biochemical Journal, 426(2), 219–228. https://doi.org/10.1042/bj20091448.

    Article  CAS  PubMed  Google Scholar 

  115. Najy, A. J., Dyson, G., Jena, B. P., Lin, C. Y., & Kim, H. R. (2016). Matriptase activation and shedding through PDGF-D-mediated extracellular acidosis. American Journal of Physiology. Cell Physiology, 310(4), C293–C304. https://doi.org/10.1152/ajpcell.00043.2015.

    Article  PubMed  Google Scholar 

  116. Huang, W., & Kim, H. R. (2015). Dynamic regulation of platelet-derived growth factor D (PDGF-D) activity and extracellular spatial distribution by matriptase-mediated proteolysis. The Journal of Biological Chemistry, 290(14), 9162–9170. https://doi.org/10.1074/jbc.M114.610865.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Tripathi, M., Potdar, A. A., Yamashita, H., Weidow, B., Cummings, P. T., Kirchhofer, D., et al. (2011). Laminin-332 cleavage by matriptase alters motility parameters of prostate cancer cells. Prostate, 71(2), 184–196. https://doi.org/10.1002/pros.21233.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Zarif, J. C., Lamb, L. E., Schulz, V. V., Nollet, E. A., & Miranti, C. K. (2015). Androgen receptor non-nuclear regulation of prostate cancer cell invasion mediated by Src and matriptase. Oncotarget, 6(9), 6862–6876. https://doi.org/10.18632/oncotarget.3119.

    Article  PubMed  PubMed Central  Google Scholar 

  119. Wu, S. R., Cheng, T. S., Chen, W. C., Shyu, H. Y., Ko, C. J., Huang, H. P., et al. (2010). Matriptase is involved in ErbB-2-induced prostate cancer cell invasion. The American Journal of Pathology, 177(6), 3145–3158. https://doi.org/10.2353/ajpath.2010.100228.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Ko, C. J., Huang, C. C., Lin, H. Y., Juan, C. P., Lan, S. W., Shyu, H. Y., et al. (2015). Androgen-induced TMPRSS2 activates matriptase and promotes extracellular matrix degradation, prostate cancer cell invasion, tumor growth, and metastasis. Cancer Research, 75(14), 2949–2960. https://doi.org/10.1158/0008-5472.CAN-14-3297.

    Article  CAS  PubMed  Google Scholar 

  121. Ko, C. J., Lan, S. W., Lu, Y. C., Cheng, T. S., Lai, P. F., Tsai, C. H., et al. (2017). Inhibition of cyclooxygenase-2-mediated matriptase activation contributes to the suppression of prostate cancer cell motility and metastasis. Oncogene, 36(32), 4597–4609. https://doi.org/10.1038/onc.2017.82.

    Article  CAS  PubMed  Google Scholar 

  122. Magee, J. A., Araki, T., Patil, S., Ehrig, T., True, L., Humphrey, P. A., et al. (2001). Expression profiling reveals hepsin overexpression in prostate cancer. Cancer Research, 61(15), 5692–5696.

    CAS  PubMed  Google Scholar 

  123. Goel, M. M., Agrawal, D., Natu, S. M., & Goel, A. (2011). Hepsin immunohistochemical expression in prostate cancer in relation to Gleason’s grade and serum prostate specific antigen. Indian Journal of Pathology & Microbiology, 54(3), 476–481. https://doi.org/10.4103/0377-4929.85078.

    Article  Google Scholar 

  124. Pal, P., Xi, H., Kaushal, R., Sun, G., Jin, C. H., Jin, L., et al. (2006). Variants in the HEPSIN gene are associated with prostate cancer in men of European origin. Human Genetics, 120(2), 187–192. https://doi.org/10.1007/s00439-006-0204-3.

    Article  CAS  PubMed  Google Scholar 

  125. Holt, S. K., Kwon, E. M., Lin, D. W., Ostrander, E. A., & Stanford, J. L. (2010). Association of hepsin gene variants with prostate cancer risk and prognosis. Prostate, 70(9), 1012–1019. https://doi.org/10.1002/pros.21135.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Ganesan, R., Kolumam, G. A., Lin, S. J., Xie, M. H., Santell, L., Wu, T. D., et al. (2011). Proteolytic activation of pro-macrophage-stimulating protein by hepsin. Molecular Cancer Research, 9(9), 1175–1186. https://doi.org/10.1158/1541-7786.MCR-11-0004.

    Article  CAS  PubMed  Google Scholar 

  127. Tripathi, M., Nandana, S., Yamashita, H., Ganesan, R., Kirchhofer, D., & Quaranta, V. (2008). Laminin-332 is a substrate for hepsin, a protease associated with prostate cancer progression. The Journal of Biological Chemistry, 283(45), 30576–30584. https://doi.org/10.1074/jbc.M802312200.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Kirchhofer, D., Peek, M., Lipari, M. T., Billeci, K., Fan, B., & Moran, P. (2005). Hepsin activates pro-hepatocyte growth factor and is inhibited by hepatocyte growth factor activator inhibitor-1B (HAI-1B) and HAI-2. FEBS Letters, 579(9), 1945–1950. https://doi.org/10.1016/j.febslet.2005.01.085.

    Article  CAS  PubMed  Google Scholar 

  129. Mi, J., Hooker, E., Balog, S., Zeng, H., Johnson, D. T., He, Y., et al. (2018). Activation of hepatocyte growth factor/MET signaling initiates oncogenic transformation and enhances tumor aggressiveness in the murine prostate. The Journal of Biological Chemistry, 293(52), 20123–20136. https://doi.org/10.1074/jbc.RA118.005395.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Li, W., Wang, B. E., Moran, P., Lipari, T., Ganesan, R., Corpuz, R., et al. (2009). Pegylated kunitz domain inhibitor suppresses hepsin-mediated invasive tumor growth and metastasis. Cancer Research, 69(21), 8395–8402. https://doi.org/10.1158/0008-5472.CAN-09-1995.

    Article  CAS  PubMed  Google Scholar 

  131. Klezovitch, O., Chevillet, J., Mirosevich, J., Roberts, R. L., Matusik, R. J., & Vasioukhin, V. (2004). Hepsin promotes prostate cancer progression and metastasis. Cancer Cell, 6(2), 185–195. https://doi.org/10.1016/j.ccr.2004.07.008.

    Article  CAS  PubMed  Google Scholar 

  132. Wittig-Blaich, S. M., Kacprzyk, L. A., Eismann, T., Bewerunge-Hudler, M., Kruse, P., Winkler, E., et al. (2011). Matrix-dependent regulation of AKT in Hepsin-overexpressing PC3 prostate cancer cells. Neoplasia, 13(7), 579–589. https://doi.org/10.1593/neo.11294.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Tang, X., Mahajan, S. S., Nguyen, L. T., Beliveau, F., Leduc, R., Simon, J. A., et al. (2014). Targeted inhibition of cell-surface serine protease Hepsin blocks prostate cancer bone metastasis. Oncotarget, 5(5), 1352–1362. https://doi.org/10.18632/oncotarget.1817.

    Article  PubMed  PubMed Central  Google Scholar 

  134. Valkenburg, K. C., Hostetter, G., & Williams, B. O. (2015). Concurrent Hepsin overexpression and adenomatous polyposis coli deletion causes invasive prostate carcinoma in mice. Prostate, 75(14), 1579–1585. https://doi.org/10.1002/pros.23032.

    Article  CAS  PubMed  Google Scholar 

  135. Nandana, S., Ellwood-Yen, K., Sawyers, C., Wills, M., Weidow, B., Case, T., et al. (2010). Hepsin cooperates with MYC in the progression of adenocarcinoma in a prostate cancer mouse model. Prostate, 70(6), 591–600. https://doi.org/10.1002/pros.21093.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Demichelis, F., Fall, K., Perner, S., Andren, O., Schmidt, F., Setlur, S. R., et al. (2007). TMPRSS2:ERG gene fusion associated with lethal prostate cancer in a watchful waiting cohort. Oncogene, 26(31), 4596–4599. https://doi.org/10.1038/sj.onc.1210237.

    Article  CAS  PubMed  Google Scholar 

  137. Tomlins, S. A., Laxman, B., Dhanasekaran, S. M., Helgeson, B. E., Cao, X., Morris, D. S., et al. (2007). Distinct classes of chromosomal rearrangements create oncogenic ETS gene fusions in prostate cancer. Nature, 448(7153), 595–599. https://doi.org/10.1038/nature06024.

    Article  CAS  PubMed  Google Scholar 

  138. Tomlins, S. A., Rhodes, D. R., Perner, S., Dhanasekaran, S. M., Mehra, R., Sun, X. W., et al. (2005). Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. Science, 310(5748), 644–648. https://doi.org/10.1126/science.1117679.

    Article  CAS  PubMed  Google Scholar 

  139. Vaarala, M. H., Porvari, K., Kyllonen, A., Lukkarinen, O., & Vihko, P. (2001). The TMPRSS2 gene encoding transmembrane serine protease is overexpressed in a majority of prostate cancer patients: detection of mutated TMPRSS2 form in a case of aggressive disease. International Journal of Cancer, 94(5), 705–710.

    Article  CAS  PubMed  Google Scholar 

  140. Wang, Z., Wang, Y., Zhang, J., Hu, Q., Zhi, F., Zhang, S., et al. (2017). Significance of the TMPRSS2:ERG gene fusion in prostate cancer. Molecular Medicine Reports, 16(4), 5450–5458. https://doi.org/10.3892/mmr.2017.7281.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Lucas, J. M., True, L., Hawley, S., Matsumura, M., Morrissey, C., Vessella, R., et al. (2008). The androgen-regulated type II serine protease TMPRSS2 is differentially expressed and mislocalized in prostate adenocarcinoma. The Journal of Pathology, 215(2), 118–125. https://doi.org/10.1002/path.2330.

    Article  CAS  PubMed  Google Scholar 

  142. Lucas, J. M., Heinlein, C., Kim, T., Hernandez, S. A., Malik, M. S., True, L. D., et al. (2014). The androgen-regulated protease TMPRSS2 activates a proteolytic cascade involving components of the tumor microenvironment and promotes prostate cancer metastasis. Cancer Discovery, 4(11), 1310–1325. https://doi.org/10.1158/2159-8290.CD-13-1010.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Takahashi, S., Suzuki, S., Inaguma, S., Ikeda, Y., Cho, Y. M., Hayashi, N., et al. (2003). Down-regulated expression of prostasin in high-grade or hormone-refractory human prostate cancers. Prostate, 54(3), 187–193. https://doi.org/10.1002/pros.10178.

    Article  CAS  PubMed  Google Scholar 

  144. Chen, L. M., Hodge, G. B., Guarda, L. A., Welch, J. L., Greenberg, N. M., & Chai, K. X. (2001). Down-regulation of prostasin serine protease: a potential invasion suppressor in prostate cancer. Prostate, 48(2), 93–103. https://doi.org/10.1002/pros.1085.

    Article  CAS  PubMed  Google Scholar 

  145. Chen, L. M., Zhang, X., & Chai, K. X. (2004). Regulation of prostasin expression and function in the prostate. Prostate, 59(1), 1–12. https://doi.org/10.1002/pros.10346.

    Article  CAS  PubMed  Google Scholar 

  146. Chen, M., Fu, Y. Y., Lin, C. Y., Chen, L. M., & Chai, K. X. (2007). Prostasin induces protease-dependent and independent molecular changes in the human prostate carcinoma cell line PC-3. Biochimica et Biophysica Acta, 1773(7), 1133–1140. https://doi.org/10.1016/j.bbamcr.2007.04.013.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Nakamura, K., Hongo, A., Kodama, J., Abarzua, F., Nasu, Y., Kumon, H., et al. (2009). Expression of matriptase and clinical outcome of human endometrial cancer. Anticancer Research, 29(5), 1685–1690.

    PubMed  Google Scholar 

  148. Santin, A. D., Cane, S., Bellone, S., Bignotti, E., Palmieri, M., De Las Casas, L. E., et al. (2003). The novel serine protease tumor-associated differentially expressed gene-15 (matriptase/MT-SP1) is highly overexpressed in cervical carcinoma. Cancer, 98(9), 1898–1904. https://doi.org/10.1002/cncr.11753.

    Article  CAS  PubMed  Google Scholar 

  149. Lee, J. W., Yong Song, S., Choi, J. J., Lee, S. J., Kim, B. G., Park, C. S., et al. (2005). Increased expression of matriptase is associated with histopathologic grades of cervical neoplasia. Human Pathology, 36(6), 626–633. https://doi.org/10.1016/j.humpath.2005.03.003.

    Article  CAS  PubMed  Google Scholar 

  150. Nakamura, K., Abarzua, F., Hongo, A., Kodama, J., Nasu, Y., Kumon, H., et al. (2009). Hepatocyte growth factor activator inhibitor-2 (HAI-2) is a favorable prognosis marker and inhibits cell growth through the apoptotic pathway in cervical cancer. Annals of Oncology, 20(1), 63–70. https://doi.org/10.1093/annonc/mdn556.

    Article  CAS  PubMed  Google Scholar 

  151. Nakamura, K., Abarzua, F., Hongo, A., Kodama, J., Nasu, Y., Kumon, H., et al. (2009). The role of hepatocyte growth factor activator inhibitor-1 (HAI-1) as a prognostic indicator in cervical cancer. International Journal of Oncology, 35(2), 239–248.

    CAS  PubMed  Google Scholar 

  152. Matsuo, T., Nakamura, K., Takamoto, N., Kodama, J., Hongo, A., Abrzua, F., et al. (2008). Expression of the serine protease hepsin and clinical outcome of human endometrial cancer. Anticancer Research, 28(1a), 159–164.

    CAS  PubMed  Google Scholar 

  153. El-Rebey, H. S., Kandil, M. A., Samaka, R. M., Al-Sharaky, D. R., & El Deeb, K. (2017). The role of hepsin in endometrial carcinoma. Applied Immunohistochemistry & Molecular Morphology, 25(9), 624–631. https://doi.org/10.1097/pai.0000000000000352.

    Article  CAS  Google Scholar 

  154. Nakamura, K., Takamoto, N., Abarzua, F., Hongo, A., Kodama, J., Nasu, Y., et al. (2008). Hepsin inhibits the cell growth of endometrial cancer. International Journal of Molecular Medicine, 22(3), 389–397.

    CAS  PubMed  Google Scholar 

  155. Cheng, H., Wang, W., Zhang, Y., Zhang, B., Cheng, J., Teng, P., et al. (2017). Expression levels and clinical significance of hepsin and HMGB1 proteins in cervical carcinoma. Oncology Letters, 14(1), 159–164. https://doi.org/10.3892/ol.2017.6116.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Uhland, K., Siphos, B., Arkona, C., Schuster, M., Petri, B., Steinmetzer, P., et al. (2009). Use of IHC and newly designed matriptase inhibitors to elucidate the role of matriptase in pancreatic ductal adenocarcinoma. International Journal of Oncology, 35(2), 347–357.

    CAS  PubMed  Google Scholar 

  157. Cheng, H., Fukushima, T., Takahashi, N., Tanaka, H., & Kataoka, H. (2009). Hepatocyte growth factor activator inhibitor type 1 regulates epithelial to mesenchymal transition through membrane-bound serine proteinases. Cancer Research, 69(5), 1828–1835. https://doi.org/10.1158/0008-5472.CAN-08-3728.

    Article  CAS  PubMed  Google Scholar 

  158. Ye, J., Kawaguchi, M., Haruyama, Y., Kanemaru, A., Fukushima, T., Yamamoto, K., et al. (2014). Loss of hepatocyte growth factor activator inhibitor type 1 participates in metastatic spreading of human pancreatic cancer cells in a mouse orthotopic transplantation model. Cancer Science, 105(1), 44–51. https://doi.org/10.1111/cas.12306.

    Article  CAS  PubMed  Google Scholar 

  159. Keppner, A., Andreasen, D., Merillat, A. M., Bapst, J., Ansermet, C., Wang, Q., et al. (2015). Epithelial sodium channel-mediated sodium transport is not dependent on the membrane-bound serine protease CAP2/Tmprss4. PLoS One, 10(8), e0135224. https://doi.org/10.1371/journal.pone.0135224.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Wallrapp, C., Hahnel, S., Muller-Pillasch, F., Burghardt, B., Iwamura, T., Ruthenburger, M., et al. (2000). A novel transmembrane serine protease (TMPRSS3) overexpressed in pancreatic cancer. Cancer Research, 60(10), 2602–2606.

    CAS  PubMed  Google Scholar 

  161. Hooper, J. D., Bowen, N., Marshall, H., Cullen, L. M., Sood, R., Daniels, R., et al. (2000). Localization, expression and genomic structure of the gene encoding the human serine protease testisin. Biochimica et Biophysica Acta, 1492(1), 63–71. https://doi.org/10.1016/s0167-4781(00)00071-3.

    Article  CAS  PubMed  Google Scholar 

  162. Manton, K. J., Douglas, M. L., Netzel-Arnett, S., Fitzpatrick, D. R., Nicol, D. L., Boyd, A. W., et al. (2005). Hypermethylation of the 5’ CpG island of the gene encoding the serine protease Testisin promotes its loss in testicular tumorigenesis. British Journal of Cancer, 92(4), 760–769. https://doi.org/10.1038/sj.bjc.6602373.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Driesbaugh, K. H., Buzza, M. S., Martin, E. W., Conway, G. D., Kao, J. P., & Antalis, T. M. (2015). Proteolytic activation of the protease-activated receptor (PAR)-2 by the glycosylphosphatidylinositol-anchored serine protease testisin. The Journal of Biological Chemistry, 290(6), 3529–3541. https://doi.org/10.1074/jbc.M114.628560.

    Article  CAS  PubMed  Google Scholar 

  164. Gao, L., Liu, M., Dong, N., Jiang, Y., Lin, C. Y., Huang, M., et al. (2013). Matriptase is highly upregulated in chronic lymphocytic leukemia and promotes cancer cell invasion. Leukemia, 27(5), 1191–1194. https://doi.org/10.1038/leu.2012.289.

    Article  CAS  PubMed  Google Scholar 

  165. Chou, F. P., Chen, Y. W., Zhao, X. F., Xu-Monette, Z. Y., Young, K. H., Gartenhaus, R. B., et al. (2013). Imbalanced matriptase pericellular proteolysis contributes to the pathogenesis of malignant B cell lymphomas. The American Journal of Pathology, 183(4), 1306–1317. https://doi.org/10.1016/j.ajpath.2013.06.024.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Chiu, Y.-L., Wu, Y.-Y., Barndt, R. B., Yeo, Y. H., Lin, Y.-W., Sytwo, H.-P., et al. (2019). Aberrant regulation favours matriptase proteolysis in neoplastic B cells that co-express HAI-2. Journal of Enzyme Inhibition and Medicinal Chemistry, 34(1), 692–702. https://doi.org/10.1080/14756366.2019.1577831.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  167. Yan, R., Liu, M., Hu, Y., Wang, L., Wang, C., Jiang, Y., et al. (2019). Ectopic expression of human airway trypsin-like protease 4 in acute myeloid leukemia promotes cancer cell invasion and tumor growth. Cancer Medicine, 8(5), 2348–2359. https://doi.org/10.1002/cam4.2074.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Coussens, L. M., Fingleton, B., & Matrisian, L. M. (2002). Matrix metalloproteinase inhibitors and cancer: trials and tribulations. Science, 295(5564), 2387–2392. https://doi.org/10.1126/science.1067100.

    Article  CAS  PubMed  Google Scholar 

  169. Takeuchi, T., Shuman, M. A., & Craik, C. S. (1999). Reverse biochemistry: use of macromolecular protease inhibitors to dissect complex biological processes and identify a membrane-type serine protease in epithelial cancer and normal tissue. Proceedings of the National Academy of Sciences of the United States of America, 96(20), 11054–11061.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Stoop, A. A., & Craik, C. S. (2003). Engineering of a macromolecular scaffold to develop specific protease inhibitors. Nature Biotechnology, 21(9), 1063–1068. https://doi.org/10.1038/nbt860.

    Article  CAS  PubMed  Google Scholar 

  171. Enyedy, I. J., Lee, S. L., Kuo, A. H., Dickson, R. B., Lin, C. Y., & Wang, S. (2001). Structure-based approach for the discovery of bis-benzamidines as novel inhibitors of matriptase. Journal of Medicinal Chemistry, 44(9), 1349–1355.

    Article  CAS  PubMed  Google Scholar 

  172. Long, Y. Q., Lee, S. L., Lin, C. Y., Enyedy, I. J., Wang, S., Li, P., et al. (2001). Synthesis and evaluation of the sunflower derived trypsin inhibitor as a potent inhibitor of the type II transmembrane serine protease, matriptase. Bioorganic & Medicinal Chemistry Letters, 11(18), 2515–2519.

    Article  CAS  Google Scholar 

  173. Galkin, A. V., Mullen, L., Fox, W. D., Brown, J., Duncan, D., Moreno, O., et al. (2004). CVS-3983, a selective matriptase inhibitor, suppresses the growth of androgen independent prostate tumor xenografts. Prostate, 61(3), 228–235. https://doi.org/10.1002/pros.20094.

    Article  CAS  PubMed  Google Scholar 

  174. Steinmetzer, T., Schweinitz, A., Sturzebecher, A., Donnecke, D., Uhland, K., Schuster, O., et al. (2006). Secondary amides of sulfonylated 3-amidinophenylalanine. New potent and selective inhibitors of matriptase. Journal of Medicinal Chemistry, 49(14), 4116–4126. https://doi.org/10.1021/jm051272l.

    Article  CAS  PubMed  Google Scholar 

  175. Farady, C. J., Sun, J., Darragh, M. R., Miller, S. M., & Craik, C. S. (2007). The mechanism of inhibition of antibody-based inhibitors of membrane-type serine protease 1 (MT-SP1). Journal of Molecular Biology, 369(4), 1041–1051. https://doi.org/10.1016/j.jmb.2007.03.078.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Schneider, E. L., Lee, M. S., Baharuddin, A., Goetz, D. H., Farady, C. J., Ward, M., et al. (2012). A reverse binding motif that contributes to specific protease inhibition by antibodies. Journal of Molecular Biology, 415(4), 699–715. https://doi.org/10.1016/j.jmb.2011.11.036.

    Article  CAS  PubMed  Google Scholar 

  177. Sun, J., Pons, J., & Craik, C. S. (2003). Potent and selective inhibition of membrane-type serine protease 1 by human single-chain antibodies. Biochemistry, 42(4), 892–900. https://doi.org/10.1021/bi026878f.

    Article  CAS  PubMed  Google Scholar 

  178. Kim, M. G., Chen, C., Lyu, M. S., Cho, E. G., Park, D., Kozak, C., et al. (1999). Cloning and chromosomal mapping of a gene isolated from thymic stromal cells encoding a new mouse type II membrane serine protease, epithin, containing four LDL receptor modules and two CUB domains. Immunogenetics, 49(5), 420–428.

    Article  CAS  PubMed  Google Scholar 

  179. Darragh, M. R., Schneider, E. L., Lou, J., Phojanakong, P. J., Farady, C. J., Marks, J. D., et al. (2010). Tumor detection by imaging proteolytic activity. Cancer Research, 70(4), 1505–1512. https://doi.org/10.1158/0008-5472.can-09-1640.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. LeBeau, A. M., Lee, M., Murphy, S. T., Hann, B. C., Warren, R. S., Delos Santos, R., et al. (2013). Imaging a functional tumorigenic biomarker in the transformed epithelium. Proceedings of the National Academy of Sciences of the United States of America, 110(1), 93–98. https://doi.org/10.1073/pnas.1218694110.

    Article  PubMed  Google Scholar 

  181. Rather, G. M., Lin, S. Y., Lin, H., Banach-Petrosky, W., Hirshfield, K. M., Lin, C. Y., et al. (2018). Activated matriptase as a target to treat breast cancer with a drug conjugate. Oncotarget, 9(40), 25983–25992. https://doi.org/10.18632/oncotarget.25414.

    Article  PubMed  PubMed Central  Google Scholar 

  182. Lin, C. Y., Anders, J., Johnson, M., & Dickson, R. B. (1999). Purification and characterization of a complex containing matriptase and a Kunitz-type serine protease inhibitor from human milk. The Journal of Biological Chemistry, 274(26), 18237–18242.

    Article  CAS  PubMed  Google Scholar 

  183. Benaud, C., Dickson, R. B., & Lin, C. Y. (2001). Regulation of the activity of matriptase on epithelial cell surfaces by a blood-derived factor. European Journal of Biochemistry, 268(5), 1439–1447.

    Article  CAS  PubMed  Google Scholar 

  184. Benaud, C., Oberst, M., Hobson, J. P., Spiegel, S., Dickson, R. B., & Lin, C. Y. (2002). Sphingosine 1-phosphate, present in serum-derived lipoproteins, activates matriptase. The Journal of Biological Chemistry, 277(12), 10539–10546. https://doi.org/10.1074/jbc.M109064200.

    Article  CAS  PubMed  Google Scholar 

  185. Rather, G. M., Lin, S. Y., Lin, H., Szekely, Z., & Bertino, J. R. (2019). A novel antibody-toxin conjugate to treat mantle cell lymphoma. Frontiers in Oncology, 9, 258. https://doi.org/10.3389/fonc.2019.00258.

    Article  PubMed  PubMed Central  Google Scholar 

  186. List, K., Kosa, P., Szabo, R., Bey, A. L., Wang, C. B., Molinolo, A., et al. (2009). Epithelial integrity is maintained by a matriptase-dependent proteolytic pathway. The American Journal of Pathology, 175(4), 1453–1463. https://doi.org/10.2353/ajpath.2009.090240.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. List, K., Currie, B., Scharschmidt, T. C., Szabo, R., Shireman, J., Molinolo, A., et al. (2007). Autosomal ichthyosis with hypotrichosis syndrome displays low matriptase proteolytic activity and is phenocopied in ST14 hypomorphic mice. The Journal of Biological Chemistry, 282(50), 36714–36723. https://doi.org/10.1074/jbc.M705521200.

    Article  CAS  PubMed  Google Scholar 

  188. Alef, T., Torres, S., Hausser, I., Metze, D., Tursen, U., Lestringant, G. G., et al. (2009). Ichthyosis, follicular atrophoderma, and hypotrichosis caused by mutations in ST14 is associated with impaired profilaggrin processing. The Journal of Investigative Dermatology, 129(4), 862–869. https://doi.org/10.1038/jid.2008.311.

    Article  CAS  PubMed  Google Scholar 

  189. Avrahami, L., Maas, S., Pasmanik-Chor, M., Rainshtein, L., Magal, N., Smitt, J., et al. (2008). Autosomal recessive ichthyosis with hypotrichosis syndrome: further delineation of the phenotype. Clinical Genetics, 74(1), 47–53. https://doi.org/10.1111/j.1399-0004.2008.01006.x.

    Article  CAS  PubMed  Google Scholar 

  190. Basel-Vanagaite, L., Attia, R., Ishida-Yamamoto, A., Rainshtein, L., Ben Amitai, D., Lurie, R., et al. (2007). Autosomal recessive ichthyosis with hypotrichosis caused by a mutation in ST14, encoding type II transmembrane serine protease matriptase. American Journal of Human Genetics, 80(3), 467–477. https://doi.org/10.1086/512487.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Desilets, A., Beliveau, F., Vandal, G., McDuff, F. O., Lavigne, P., & Leduc, R. (2008). Mutation G827R in matriptase causing autosomal recessive ichthyosis with hypotrichosis yields an inactive protease. The Journal of Biological Chemistry, 283(16), 10535–10542. https://doi.org/10.1074/jbc.M707012200.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Colombo, E., Desilets, A., Duchene, D., Chagnon, F., Najmanovich, R., Leduc, R., et al. (2012). Design and synthesis of potent, selective inhibitors of matriptase. ACS Medicinal Chemistry Letters, 3(7), 530–534. https://doi.org/10.1021/ml3000534.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  193. St-Georges, C., Desilets, A., Beliveau, F., Ghinet, M., Dion, S. P., Colombo, E., et al. (2017). Modulating the selectivity of matriptase-2 inhibitors with unnatural amino acids. European Journal of Medicinal Chemistry, 129, 110–123. https://doi.org/10.1016/j.ejmech.2017.02.006.

    Article  CAS  PubMed  Google Scholar 

  194. Gray, K., Elghadban, S., Thongyoo, P., Owen, K. A., Szabo, R., Bugge, T. H., et al. (2014). Potent and specific inhibition of the biological activity of the type-II transmembrane serine protease matriptase by the cyclic microprotein MCoTI-II. Thrombosis and Haemostasis, 112(2), 402–411. https://doi.org/10.1160/TH13-11-0895.

    Article  CAS  PubMed  Google Scholar 

  195. Quimbar, P., Malik, U., Sommerhoff, C. P., Kaas, Q., Chan, L. Y., Huang, Y. H., et al. (2013). High-affinity cyclic peptide matriptase inhibitors. The Journal of Biological Chemistry, 288(19), 13885–13896. https://doi.org/10.1074/jbc.M113.460030.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Swedberg, J. E., Nigon, L. V., Reid, J. C., de Veer, S. J., Walpole, C. M., Stephens, C. R., et al. (2009). Substrate-guided design of a potent and selective kallikrein-related peptidase inhibitor for kallikrein 4. Chemistry & Biology, 16(6), 633–643. https://doi.org/10.1016/j.chembiol.2009.05.008.

    Article  CAS  Google Scholar 

  197. Clark, R. J., Jensen, J., Nevin, S. T., Callaghan, B. P., Adams, D. J., & Craik, D. J. (2010). The engineering of an orally active conotoxin for the treatment of neuropathic pain. Angewandte Chemie (International Ed. in English), 49(37), 6545–6548. https://doi.org/10.1002/anie.201000620.

    Article  CAS  Google Scholar 

  198. Wong, C. T., Rowlands, D. K., Wong, C. H., Lo, T. W., Nguyen, G. K., Li, H. Y., et al. (2012). Orally active peptidic bradykinin B1 receptor antagonists engineered from a cyclotide scaffold for inflammatory pain treatment. Angewandte Chemie (International Ed. in English), 51(23), 5620–5624. https://doi.org/10.1002/anie.201200984.

    Article  CAS  Google Scholar 

  199. Desilets, A., Longpre, J. M., Beaulieu, M. E., & Leduc, R. (2006). Inhibition of human matriptase by eglin c variants. FEBS Letters, 580(9), 2227–2232. https://doi.org/10.1016/j.febslet.2006.03.030.

    Article  CAS  PubMed  Google Scholar 

  200. Mitchell, A. C., Kannan, D., Hunter, S. A., Parra Sperberg, R. A., Chang, C. H., & Cochran, J. R. (2018). Engineering a potent inhibitor of matriptase from the natural hepatocyte growth factor activator inhibitor type-1 (HAI-1) protein. The Journal of Biological Chemistry, 293(14), 4969–4980. https://doi.org/10.1074/jbc.M117.815142.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Kojima, K., Tsuzuki, S., Fushiki, T., & Inouye, K. (2008). Roles of functional and structural domains of hepatocyte growth factor activator inhibitor type 1 in the inhibition of matriptase. The Journal of Biological Chemistry, 283(5), 2478–2487. https://doi.org/10.1074/jbc.M709073200.

    Article  CAS  PubMed  Google Scholar 

  202. Zhao, B., Yuan, C., Li, R., Qu, D., Huang, M., & Ngo, J. C. (2013). Crystal structures of matriptase in complex with its inhibitor hepatocyte growth factor activator inhibitor-1. The Journal of Biological Chemistry, 288(16), 11155–11164. https://doi.org/10.1074/jbc.M113.454611.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Han, Z., Harris, P. K., Karmakar, P., Kim, T., Owusu, B. Y., Wildman, S. A., et al. (2016). alpha-Ketobenzothiazole serine protease inhibitors of aberrant HGF/c-MET and MSP/RON kinase pathway signaling in cancer. ChemMedChem, 11(6), 585–599. https://doi.org/10.1002/cmdc.201500600.

    Article  CAS  PubMed  Google Scholar 

  204. Han, Z., Harris, P. K., Jones, D. E., Chugani, R., Kim, T., Agarwal, M., et al. (2014). Inhibitors of HGFA, matriptase, and hepsin serine proteases: a nonkinase strategy to block cell signaling in cancer. ACS Medicinal Chemistry Letters, 5(11), 1219–1224. https://doi.org/10.1021/ml500254r.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  205. Damalanka, V. C., & Janetka, J. W. (2019). Recent progress on inhibitors of the type II transmembrane serine proteases, hepsin, matriptase and matriptase-2. Future Medicinal Chemistry, 11(7), 743–769. https://doi.org/10.4155/fmc-2018-0446.

    Article  CAS  PubMed  Google Scholar 

  206. Miyazawa, K., Shimomura, T., Kitamura, A., Kondo, J., Morimoto, Y., & Kitamura, N. (1993). Molecular cloning and sequence analysis of the cDNA for a human serine protease reponsible for activation of hepatocyte growth factor. Structural similarity of the protease precursor to blood coagulation factor XII. The Journal of Biological Chemistry, 268(14), 10024–10028.

    CAS  PubMed  Google Scholar 

  207. Franco, F. M., Jones, D. E., Harris, P. K., Han, Z., Wildman, S. A., Jarvis, C. M., et al. (2015). Structure-based discovery of small molecule hepsin and HGFA protease inhibitors: evaluation of potency and selectivity derived from distinct binding pockets. Bioorganic & Medicinal Chemistry, 23(10), 2328–2343. https://doi.org/10.1016/j.bmc.2015.03.072.

    Article  CAS  Google Scholar 

  208. Owusu, B. Y., Bansal, N., Venukadasula, P. K., Ross, L. J., Messick, T. E., Goel, S., et al. (2016). Inhibition of pro-HGF activation by SRI31215, a novel approach to block oncogenic HGF/MET signaling. Oncotarget, 7(20), 29492–29506. https://doi.org/10.18632/oncotarget.8785.

    Article  PubMed  PubMed Central  Google Scholar 

  209. Owusu, B. Y., Thomas, S., Venukadasula, P., Han, Z., Janetka, J. W., Galemmo, R. A., Jr., et al. (2017). Targeting the tumor-promoting microenvironment in MET-amplified NSCLC cells with a novel inhibitor of pro-HGF activation. Oncotarget, 8(38), 63014–63025. https://doi.org/10.18632/oncotarget.18260.

    Article  PubMed  PubMed Central  Google Scholar 

  210. Venukadasula, P. K., Owusu, B. Y., Bansal, N., Ross, L. J., Hobrath, J. V., Bao, D., et al. (2016). Design and synthesis of nonpeptide inhibitors of hepatocyte growth factor activation. ACS Medicinal Chemistry Letters, 7(2), 177–181. https://doi.org/10.1021/acsmedchemlett.5b00357.

    Article  CAS  PubMed  Google Scholar 

  211. Kwon, H., Kim, Y., Park, K., Choi, S. A., Son, S. H., & Byun, Y. (2016). Structure-based design, synthesis, and biological evaluation of Leu-Arg dipeptide analogs as novel hepsin inhibitors. Bioorganic & Medicinal Chemistry Letters, 26(2), 310–314. https://doi.org/10.1016/j.bmcl.2015.12.023.

    Article  CAS  Google Scholar 

  212. Kim, K., Kwon, H., Choi, D., Lim, T., Minn, I., Son, S. H., et al. (2019). Design and synthesis of dye-conjugated hepsin inhibitors. Bioorganic Chemistry, 89, 102990. https://doi.org/10.1016/j.bioorg.2019.102990.

    Article  CAS  PubMed  Google Scholar 

  213. Shia, S., Stamos, J., Kirchhofer, D., Fan, B., Wu, J., Corpuz, R. T., et al. (2005). Conformational lability in serine protease active sites: structures of hepatocyte growth factor activator (HGFA) alone and with the inhibitory domain from HGFA inhibitor-1B. Journal of Molecular Biology, 346(5), 1335–1349. https://doi.org/10.1016/j.jmb.2004.12.048.

    Article  CAS  PubMed  Google Scholar 

  214. Xuan, J. A., Schneider, D., Toy, P., Lin, R., Newton, A., Zhu, Y., et al. (2006). Antibodies neutralizing hepsin protease activity do not impact cell growth but inhibit invasion of prostate and ovarian tumor cells in culture. Cancer Research, 66(7), 3611–3619. https://doi.org/10.1158/0008-5472.CAN-05-2983.

    Article  CAS  PubMed  Google Scholar 

  215. Ganesan, R., Zhang, Y., Landgraf, K. E., Lin, S. J., Moran, P., & Kirchhofer, D. (2012). An allosteric anti-hepsin antibody derived from a constrained phage display library. Protein Engineering, Design & Selection, 25(3), 127–133. https://doi.org/10.1093/protein/gzr067.

    Article  CAS  Google Scholar 

  216. Koschubs, T., Dengl, S., Durr, H., Kaluza, K., Georges, G., Hartl, C., et al. (2012). Allosteric antibody inhibition of human hepsin protease. The Biochemical Journal, 442(3), 483–494. https://doi.org/10.1042/BJ20111317.

    Article  CAS  PubMed  Google Scholar 

  217. Tully, D. C., Vidal, A., Chatterjee, A. K., Williams, J. A., Roberts, M. J., Petrassi, H. M., et al. (2008). Discovery of inhibitors of the channel-activating protease prostasin (CAP1/PRSS8) utilizing structure-based design. Bioorganic & Medicinal Chemistry Letters, 18(22), 5895–5899. https://doi.org/10.1016/j.bmcl.2008.08.029.

    Article  CAS  Google Scholar 

  218. Klimpel, K. R., Molloy, S. S., Thomas, G., & Leppla, S. H. (1992). Anthrax toxin protective antigen is activated by a cell surface protease with the sequence specificity and catalytic properties of furin. Proceedings of the National Academy of Sciences of the United States of America, 89(21), 10277–10281. https://doi.org/10.1073/pnas.89.21.10277.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  219. Liu, S., Netzel-Arnett, S., Birkedal-Hansen, H., & Leppla, S. H. (2000). Tumor cell-selective cytotoxicity of matrix metalloproteinase-activated anthrax toxin. Cancer Research, 60(21), 6061–6067.

    CAS  PubMed  Google Scholar 

  220. Liu, S., Bugge, T. H., & Leppla, S. H. (2001). Targeting of tumor cells by cell surface urokinase plasminogen activator-dependent anthrax toxin. The Journal of Biological Chemistry, 276(21), 17976–17984. https://doi.org/10.1074/jbc.M011085200.

    Article  CAS  PubMed  Google Scholar 

  221. Peters, D. E., Hoover, B., Cloud, L. G., Liu, S., Molinolo, A. A., Leppla, S. H., et al. (2014). Comparative toxicity and efficacy of engineered anthrax lethal toxin variants with broad anti-tumor activities. Toxicology and Applied Pharmacology, 279(2), 220–229. https://doi.org/10.1016/j.taap.2014.06.010.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  222. Liu, S., Redeye, V., Kuremsky, J. G., Kuhnen, M., Molinolo, A., Bugge, T. H., et al. (2005). Intermolecular complementation achieves high-specificity tumor targeting by anthrax toxin. Nature Biotechnology, 23(6), 725–730. https://doi.org/10.1038/nbt1091.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Funding

This work was supported by NIH/NCI R01CA222359A (KL), Susan G. Komen IBC17511329 (KL) and IBC18511329 (K.L), and an NIH/NCI Training grant (CEM) awarded to Wayne State University Cancer Biology Graduate Program (Ruth L. Kirschstein National Research Service Award T32-CA009531).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Karin List.

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.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Martin, C.E., List, K. Cell surface–anchored serine proteases in cancer progression and metastasis. Cancer Metastasis Rev 38, 357–387 (2019). https://doi.org/10.1007/s10555-019-09811-7

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10555-019-09811-7

Keywords

Navigation