ADAM and ADAMTS Proteases in Hepatic Disorders

Main Article Content

Julia Bolik
Janina E. E. Tirnitz-Parker
Dirk Schmidt-Arras

Keywords

ADAM, ADAMTS, metzincin superfamily, thrombotic thrombocytopenia purpura, von Willebrand Factor

Abstract

Proteolysis is an irreversible post-translational modification that regulates protein function and signal transduction. This includes remodelling of the extracellular matrix, release of membrane-bound cytokines and receptor ectodomains, as well as the initiation of intracellular signalling cues. Members of the adamalysin protease subfamily, in particular the ADAM (a disintegrin and metalloprotease) and ADAMTS (the ADAM containing thrombospondin motif) families, are involved in these processes. This review presents an overview of how ADAM and ADAMTS proteins are involved in liver physiology and pathophysiology.

Abstract 1085 | PDF Downloads 573 HTML Downloads 864 XML Downloads 248

References

1. Gilgenkrantz H, Collin de l’Hortet A. Understanding liver regeneration: From mechanisms to regenerative medicine. Am J Pathol. 2018;188:1316–27. https://doi.org/10.1016/j.ajpath.2018.03.008
2. Köhn-Gaone J, Gogoi-Tiwari J, Ramm GA, Olynyk JK, Tirnitz-Parker JEE. The role of liver progenitor cells during liver regeneration, fibrogenesis, and carcinogenesis. Am J Physiol. 2016;310:G143–54.
3. Ruddell RG, Knight B, Tirnitz-Parker JEE, Akhurst B, Summerville L, Subramaniam VN, et al. Lymphotoxin-beta receptor signaling regulates hepatic stellate cell function and wound healing in a murine model of chronic liver injury. Hepatology. 2009;49:227–39. https://doi.org/10.1002/hep.22597
4. Boulter L, Govaere O, Bird TG, Radulescu S, Ramachandran P, Pellicoro A, et al. Macrophage-derived Wnt opposes Notch signaling to specify hepatic progenitor cell fate in chronic liver disease. Nat Med. 2012;18:572–9. https://doi.org/10.1038/nm.2667
5. Tirnitz-Parker JEE, Olynyk JK, Ramm GA. Role of TWEAK in coregulating liver progenitor cell and fibrogenic responses. Hepatology. 2014;59:1198–201. https://doi.org/10.1002/hep.26701
6. Finkin S, Yuan D, Stein I, Taniguchi K, Weber A, Unger K, et al. Ectopic lymphoid structures function as microniches for tumor progenitor cells in hepatocellular carcinoma. Nat Immunol. 2015;16:1235–44. https://doi.org/10.1038/ni.3290
7. Huxley-Jones J, Clarke T-K, Beck C, Toubaris G, Robertson DL, Boot-Handford RP. The evolution of the vertebrate metzincins; insights from Ciona intestinalis and Danio rerio. BMC Evol Biol. 2007;7:63. https://doi.org/10.1186/1471-2148-7-63
8. Gomis-Rüth FX. Catalytic domain architecture of metzincin metalloproteases. Biol Chem. 2009;284:15353–7. https://doi.org/10.1074/jbc.R800069200
9. Reiss K, Saftig P. The “a disintegrin and metalloprotease” (ADAM) family of sheddases: Physiological and cellular functions. Semin Cell Dev Biol. 2009;20:126–37. https://doi.org/10.1016/j.semcdb.2008.11.002
10. Seegar TCM, Killingsworth LB, Saha N, Meyer PA, Patra D, Zimmerman B, et al. Structural basis for regulated proteolysis by the ?-secretase ADAM10. Cell. 2017;171:1638–48. https://doi.org/10.1016/j.cell.2017.11.014
11. Mosyak L, Georgiadis K, Shane T, Svenson K, Hebert T, McDonagh T, et al. Crystal structures of the two major aggrecan degrading enzymes, ADAMTS4 and ADAMTS5. Protein Sci. 2008;17:16–21. https://doi.org/10.1110/ps.073287008
12. Düsterhöft S, Jung S, Hung C-W, Tholey A, Sönnichsen FD, Grötzinger J, et al. Membrane-proximal domain of a disintegrin and metalloprotease-17 represents the putative molecular switch of its shedding activity operated by protein-disulfide isomerase. J Am Chem Soc. 2013;135:5776–81. https://doi.org/10.1110/ps.073287008
13. Jones JC, Rustagi S, Dempsey PJ. ADAM proteases and gastrointestinal function. Annu Rev Physiol. 2016;78:243–76. https://doi.org/10.1146/annurev-physiol-021014-071720
14. Howard L, Maciewicz RA, Blobel CP. Cloning and characterization of ADAM28: Evidence for autocatalytic pro-domain removal and for cell surface localization of mature ADAM28. Biochem J. 2000;348 Pt 1:21–7. https://doi.org/10.1042/bj3480021
15. Schlomann U, Wildeboer D, Webster A, Antropova O, Zeuschner D, Knight CG, et al. The metalloprotease disintegrin ADAM8. Processing by autocatalysis is required for proteolytic activity and cell adhesion. J Biol Chem. 2002;277:48210–19. https://doi.org/10.1074/jbc.M203355200
16. Wong E, Cohen T, Romi E, Levin M, Peleg Y, Arad U, et al. Harnessing the natural inhibitory domain to control TNF? Converting Enzyme (TACE) activity in vivo. Sci Rep. 2016;6:35598. https://doi.org/10.1038/srep35598
17. Maskos K, Fernandez-Catalan C, Huber R, Bourenkov GP, Bartunik H, Ellestad GA, et al. Crystal structure of the catalytic domain of human tumor necrosis factor-alpha-converting enzyme. Proc Natl Acad Sci U S A. 1998;95:3408–12. https://doi.org/10.1073/pnas.95.7.3408
18. Stawikowska R, Cudic M, Giulianotti M, Houghten RA, Fields GB, Minond D. Activity of ADAM17 (a disintegrin and metalloprotease 17) is regulated by its noncatalytic domains and secondary structure of its substrates. J Biol Chem. 2013;288:22871–9. https://doi.org/10.1074/jbc.M113.462267
19. Janes PW, Saha N, Barton WA, Kolev MV, Wimmer-Kleikamp SH, Nievergall E, et al. Adam meets Eph: An ADAM substrate recognition module acts as a molecular switch for ephrin cleavage in trans. Cell. 2005;123:291–304. https://doi.org/10.1016/j.cell.2005.08.014
20. Xu P, Derynck R. Direct activation of TACE-mediated ectodomain shedding by p38 MAP kinase regulates EGF receptor-dependent cell proliferation. Mol Cell. 2010;37:551–66. https://doi.org/10.1016/j.molcel.2010.01.034
21. Xu P, Liu J, Sakaki-Yumoto M, Derynck R. TACE activation by MAPK-mediated regulation of cell surface dimerization and TIMP3 association. Sci Signal. 2012;5:ra34. https://doi.org/10.1126/scisignal.2002689
22. Liu C, Xu P, Lamouille S, Xu J, Derynck R. TACE-mediated ectodomain shedding of the type I TGF-beta receptor downregulates TGF-beta signaling. Mol Cell. 2009;35:26–36. https://doi.org/10.1016/j.molcel.2009.06.018
23. Scheller J, Chalaris A, Garbers C, Rose-John S. ADAM17: A molecular switch to control inflammation and tissue regeneration Trends Immunol. 2011;32:380–7. https://doi.org/10.1016/j.it.2011.05.005
24. Schmidt-Arras D, Rose-John S. IL-6 pathway in the liver: From physiopathology to therapy. J Hepatol. 2016;64:1403–15. https://doi.org/10.1016/j.jhep.2016.02.004
25. Moss ML, Minond D. Recent advances in ADAM17 research: A promising target for cancer and inflammation. Mediators Inflamm. 2017;2017:9673537.
26. Hartmann D, de Strooper B, Serneels L, Craessaerts K, Herreman A, Annaert W, et al. The disintegrin/metalloprotease ADAM 10 is essential for Notch signalling but not for alpha-secretase activity in fibroblasts. Hum Mol Genet. 2002;11:2615–24. https://doi.org/10.1093/hmg/11.21.2615
27. White JM. ADAMs: Modulators of cell-cell and cell-matrix interactions. Curr Opin Cell Biol. 2003;15:598–606. https://doi.org/10.1016/j.ceb.2003.08.001
28. Oria VO, Lopatta P, Schilling O. The pleiotropic roles of ADAM9 in the biology of solid tumors. Cell Mol Life Sci. 2018;75:2291–301. https://doi.org/10.1007/s00018-018-2796-x
29. Schwettmann L, Wehmeier M, Jokovic D, Aleksandrova K, Brand K, Manns MP, et al. Hepatic expression of A disintegrin and metalloproteinase (ADAM) and ADAMs with thrombospondin motives (ADAM-TS) enzymes in patients with chronic liver diseases. J Hepatol. 2008;49:243–50. https://doi.org/10.1016/j.jhep.2008.03.020
30. Le Pabic H, Bonnier D, Wewer UM, Coutand A, Musso O, Baffet G, et al. ADAM12 in human liver cancers: TGF-beta-regulated expression in stellate cells is associated with matrix remodeling. Hepatology. 2003;37:1056–66. https://doi.org/10.1053/jhep.2003.50205
31. Tannapfel A, Anhalt K, Häusermann P, Sommerer F, Benicke M, Uhlmann D, et al. Identification of novel proteins associated with hepatocellular carcinomas using protein microarrays. J Pathol. 2003;201:238–49. https://doi.org/10.1002/path.1420
32. Kohga K, Takehara T, Tatsumi T, Ishida H, Miyagi T, Hosui A, et al. Sorafenib inhibits the shedding of major histocompatibility complex class I-related chain A on hepatocellular carcinoma cells by down-regulating a disintegrin and metalloproteinase 9. Hepatology. 2010;51:1264–73. https://doi.org/10.1002/hep.23456
33. Mazzocca A, Coppari R, De Franco R, Cho J-Y, Libermann TA, Pinzani M, et al. A secreted form of ADAM9 promotes carcinoma invasion through tumor-stromal interactions. Cancer Res. 2005;65:4728–38. https://doi.org/10.1158/0008-5472.CAN-04-4449
34. Geisler F, Nagl F, Mazur PK, Lee M, Zimber-Strobl U, Strobl LJ, et al. Liver-specific inactivation of Notch2, but not Notch1, compromises intrahepatic bile duct development in mice. Hepatology. 2008;48:607–16. https://doi.org/10.1002/hep.22381
35. Sparks EE, Huppert KA, Brown MA, Washington MK, Huppert SS. Notch signaling regulates formation of the three-dimensional architecture of intrahepatic bile ducts in mice. Hepatology. 2010;51:1391–400. https://doi.org/10.1002/hep.23431
36. Oda T, Elkahloun AG, Pike BL, Okajima K, Krantz ID, Genin A et al., Mutations in the human Jagged1 gene are responsible for Alagille syndrome. Nat Genet. 1997;16:235–42. https://doi.org/10.1038/ng0797-235
37. Müller M, Wetzel S, Köhn-Gaone J, Chalupsky K, Lüllmann-Rauch R, Barikbin R, et al. A disintegrin and metalloprotease 10 (ADAM10) is a central regulator of murine liver tissue homeostasis. Oncotarget. 2016;7:17431–41. https://doi.org/10.18632/oncotarget.7836
38. Bourd-Boittin K, Basset L, Bonnier D, L’helgoualc’h A, Samson M, Théret N. CX3CL1/fractalkine shedding by human hepatic stellate cells: Contribution to chronic inflammation in the liver. J Cell Mol Med. 2009;13:1526–35. https://doi.org/10.1111/j.1582-4934.2009.00787.x
39. Kohga K, Takehara T, Tatsumi T, Miyagi T, Ishida H, Ohkawa K, et al. Anticancer chemotherapy inhibits MHC class I-related chain a ectodomain shedding by downregulating ADAM10 expression in hepatocellular carcinoma. Cancer Res. 2009;69:8050–7. https://doi.org/10.1158/0008-5472.CAN-09-0789
40. Yuan S, Lei S, Wu S. ADAM10 is overexpressed in human hepatocellular carcinoma and contributes to the proliferation, invasion and migration of HepG2 cells. Oncol Rep. 2013;30:1715–22. https://doi.org/10.3892/or.2013.2650
41. Chalupský K, Kanchev I, Žbodáková O, Buryová H, Jiroušková M, Ko?ínek V, et al. ADAM10/17-dependent release of soluble c-Met correlates with hepatocellular damage. Folia Biol (Praha) 2013;59:76–86.
42. Zhang Y-W, Graveel C, Shinomiya N, Vande Woude GF. Met decoys: Will cancer take the bait? Cancer Cell. 2004;6:5–6. https://doi.org/10.1016/j.ccr.2004.07.003
43. Gavert N, Sheffer M, Raveh S, Spaderna S, Shtutman M, Brabletz T, et al. Expression of L1-CAM and ADAM10 in human colon cancer cells induces metastasis. Cancer Res. 2007;67:7703–12. https://doi.org/10.1158/0008-5472.CAN-07-0991
44. Kopitz C, Gerg M, Bandapalli OR, Ister D, Pennington CJ, Hauser S, et al. Tissue inhibitor of metalloproteinases-1 promotes liver metastasis by induction of hepatocyte growth factor signaling. Cancer Res. 2007;67:8615–23. https://doi.org/10.1158/0008-5472.CAN-07-0232
45. Schelter F, Grandl M, Seubert B, Schaten S, Hauser S, Gerg M, et al. Tumor cell-derived Timp-1 is necessary for maintaining metastasis-promoting Met-signaling via inhibition of Adam-10. Clin Exp Metastasis. 2011;28:793–802. https://doi.org/10.1007/s10585-011-9410-z
46. Grünwald B, Harant V, Schaten S, Frühschütz M, Spallek R, Höchst B et al., Pancreatic premalignant lesions secrete tissue inhibitor of metalloproteinases-1, which activates hepatic stellate cells via CD63 signaling to create a premetastatic niche in the liver. Gastroenterology. 2016;151:1011–24.e7. https://doi.org/10.1053/j.gastro.2016.07.043
47. Nyren-Erickson EK, Jones JM, Srivastava D, Mallik S. A disintegrin and metalloproteinase-12 ADAM12): Function, roles in disease progression, and clinical implications Biochim Biophys Acta. 2013;1830:4445–55.
48. Daduang J, Limpaiboon T, Daduang S. Biomarker to distinguish hepatocellular carcinoma from cholangiocarcinoma by serum a disintegrin and metalloprotease 12. Arch Med Sci. 2011;7:1013–16. https://doi.org/10.5114/aoms.2011.26613
49. Fang T, Lin J, Wang Y, Chen G, Huang J, Chen J, et al. Tetraspanin-8 promotes hepatocellular carcinoma metastasis by increasing ADAM12m expression. Oncotarget. 2016;7:40630–43. https://doi.org/10.18632/oncotarget.9769
50. He G, Dhar D, Nakagawa H, Font-Burgada J, Ogata H, Jiang Y, et al. Identification of liver cancer progenitors whose malignant progression depends on autocrine IL-6 signaling. Cell. 2013;155:384–96. https://doi.org/10.1016/j.cell.2013.09.031
51. Argast GM, Campbell JS, Brooling JT, Fausto N. Epidermal growth factor receptor transactivation mediates tumor necrosis factor-induced hepatocyte replication. J Biol Chem. 2004;279:34530–6. https://doi.org/10.1074/jbc.M405703200
52. Murillo MM, del Castillo G, Sánchez A, Fernández M, Fabregat I. Involvement of EGF receptor and c-Src in the survival signals induced by TGF-beta1 in hepatocytes. Oncogene. 2005;24:4580–7. https://doi.org/10.1038/sj.onc.1208664
53. Li Y, Ren Z, Wang Y, Dang Y-Z, Meng B-X, Wang G-D, et al. ADAM17 promotes cell migration and invasion through the integrin ?1 pathway in hepatocellular carcinoma. Exp Cell Res. 2018;370:373–82. https://doi.org/10.1016/j.yexcr.2018.06.039
54. Wang R, Li Y, Tsung A, Huang H, Du Q, Yang M, et al. iNOS promotes CD24+, CD133+ liver cancer stem cell phenotype through a TACE/ADAM17-dependent Notch signaling pathway. Proc Natl Acad Sci U S A. 2018;115:E10127–36. https://doi.org/10.1073/pnas.1722100115
55. Hong SW, Hur W, Choi JE, Kim J-H, Hwang D, Yoon SK. Role of ADAM17 in invasion and migration of CD133-expressing liver cancer stem cells after irradiation. Oncotarget. 2016;7:23482–97. https://doi.org/10.18632/oncotarget.8112
56. Lu J, Ye X, Fan F, Xia L, Bhattacharya R, Bellister S, et al. Endothelial cells promote the colorectal cancer stem cell phenotype through a soluble form of Jagged-1. Cancer Cell. 2013;23:171–85. https://doi.org/10.1016/j.ccr.2012.12.021
57. Bergmann J, Müller M, Baumann N, Reichert M, Heneweer C, Bolik J, et al. IL-6 trans-signaling is essential for the development of hepatocellular carcinoma in mice. Hepatology. 2017;65:89–103. https://doi.org/10.1002/hep.28874
58. Kelwick R, Desanlis I, Wheeler GN, Edwards DR. The ADAMTS (a disintegrin and metalloproteinase with thrombospondin motifs) family. Genome Biol. 2015;16:113. https://doi.org/10.1186/s13059-015-0676-3
59. Stanton H, Melrose J, Little CB, Fosang AJ. Proteoglycan degradation by the ADAMTS family of proteinases. Biochim Biophys Acta. 2011;1812:1616–29. https://doi.org/10.1016/j.bbadis.2011.08.009
60. Gerhardt S, Hassall G, Hawtin P, McCall E, Flavell L, Minshull C, et al. Crystal structures of human ADAMTS-1 reveal a conserved catalytic domain and a disintegrin-like domain with a fold homologous to cysteine-rich domains. J Mol Biol. 2007;373:891–902. https://doi.org/10.1016/j.jmb.2007.07.047
61. Bourd-Boittin K, Bonnier D, Leyme A, Mari B, Tuffery P, Samson M, et al. Protease profiling of liver fibrosis reveals the ADAM metallopeptidase with thrombospondin type 1 motif, 1 as a central activator of transforming growth factor beta. Hepatology. 2011;54:2173–84. https://doi.org/10.1002/hep.24598
62. Colige A, Sieron AL, Li SW, Schwarze U, Petty E, Wertelecki W, et al. Human Ehlers-Danlos syndrome type VII C and bovine dermatosparaxis are caused by mutations in the procollagen I N-proteinase gene. Am J Hum Genet. 1999;65:308–17. https://doi.org/10.1086/302504
63. Kesteloot F, Desmoulière A, Leclercq I, Thiry M, Arrese JE, Prockop DJ, et al. ADAM metallopeptidase with thrombospondin type 1 motif 2 inactivation reduces the extent and stability of carbon tetrachloride-induced hepatic fibrosis in mice. Hepatology. 2007;46:1620–31. https://doi.org/10.1002/hep.21868
64. Sadler JE. von Willebrand factor assembly and secretion. J Thromb Haemost. 2009;7 Suppl 1:24–7. https://doi.org/10.1111/j.1538-7836.2009.03375.x
65. Sadler JE. Von Willebrand factor, ADAMTS13, and thrombotic thrombocytopenic purpura. Blood. 2008;112:11–18. https://doi.org/10.1182/blood-2008-02-078170
66. Levy GG, Nichols WC, Lian EC, Foroud T, McClintick JN, McGee BM, et al. Mutations in a member of the ADAMTS gene family cause thrombotic thrombocytopenic purpura. Nature. 2001;413:488–94. https://doi.org/10.1038/35097008
67. Zheng XL. ADAMTS13 and von Willebrand factor in thrombotic thrombocytopenic purpura. Annu Rev Med. 2015;66:211–25.
68. Bianchi V, Robles R, Alberio L, Furlan M, Lämmle B. Von Willebrand factor-cleaving protease ADAMTS13) in thrombocytopenic disorders: A severely deficient activity is specific for thrombotic thrombocytopenic purpura. Blood. 2002;100:710–13. https://doi.org/10.1182/blood-2002-02-0344
69. Ono T, Mimuro J, Madoiwa S, Soejima K, Kashiwakura Y, Ishiwata A, et al. Severe secondary deficiency of von Willebrand factor-cleaving protease (ADAMTS13) in patients with sepsis-induced disseminated intravascular coagulation: Its correlation with development of renal failure. Blood. 2006;107:528–34. https://doi.org/10.1182/blood-2005-03-1087
70. Azfar MF, Khan MF, Habib SS, Aseri ZA, Zubaidi AM, Aguila DO, et al. Prognostic value of ADAMTS13 in patients with severe sepsis and septic shock. Clin Invest Med. 2017;40:E49–58. https://doi.org/10.25011/cim.v40i2.28195
71. Levy GG, Motto DG, Ginsburg D. ADAMTS13 turns 3. Blood. 2005;106:11–17. https://doi.org/10.1182/blood-2004-10-4097
72. Uemura M, Matsuyama T, Ishikawa M, Fujimoto M, Kojima H, Sakurai S, et al. Decreased activity of plasma ADAMTS13 may contribute to the development of liver disturbance and multiorgan failure in patients with alcoholic hepatitis. Alcohol Clin Exp Res. 2005;29:264S–271S.
73. Hugenholtz GCG, Adelmeijer J, Meijers JCM, Porte RJ, Stravitz RT, Lisman T. An unbalance between von Willebrand factor and ADAMTS13 in acute liver failure: Implications for hemostasis and clinical outcome. Hepatology. 2013;58:752–61. https://doi.org/10.1002/hep.26372
74. Uemura M, Tatsumi K, Matsumoto M, Fujimoto M, Matsuyama T, Ishikawa M, et al. Localization of ADAMTS13 to the stellate cells of human liver. Blood. 2005;106:922–4. https://doi.org/10.1182/blood-2005-01-0152
75. Niiya M, Uemura M, Zheng XW, Pollak ES, Dockal M, Scheiflinger F, et al. Increased ADAMTS-13 proteolytic activity in rat hepatic stellate cells upon activation in vitro and in vivo. J Thromb Haemost. 2006;4:1063–70. https://doi.org/10.1111/j.1538-7836.2006.01893.x
76. Lisman T, Bongers TN, Adelmeijer J, Janssen HLA, de Maat MPM, de Groot PG, et al. Elevated levels of von Willebrand factor in cirrhosis support platelet adhesion despite reduced functional capacity. Hepatology. 2006;44:53–61. https://doi.org/10.1002/hep.21231
77. Mannucci PM, Canciani MT, Forza I, Lussana F, Lattuada A, Rossi E. Changes in health and disease of the metalloprotease that cleaves von Willebrand factor. Blood. 2001;98:2730–5. https://doi.org/10.1182/blood.V98.9.2730
78. Uemura M, Fujimura Y, Matsumoto M, Ishizashi H, Kato S, Matsuyama T, et al. Comprehensive analysis of ADAMTS13 in patients with liver cirrhosis. Thromb Haemost. 2008;99:1019–29. https://doi.org/10.1160/TH08-01-0006
79. Lancellotti S, Basso M, Veca V, Sacco M, Riccardi L, Pompili M, et al. Presence of portal vein thrombosis in liver cirrhosis is strongly associated with low levels of ADAMTS-13: A pilot study. Intern Emerg Med. 2016;11:959–67. https://doi.org/10.1007/s11739-016-1467-x
80. Potze W, Siddiqui MS, Boyett SL, Adelmeijer J, Daita K, Sanyal AJ, et al. Preserved hemostatic status in patients with non-alcoholic fatty liver disease. J Hepatol. 2016;65:980–7. https://doi.org/10.1016/j.jhep.2016.06.001
81. Moss ML, Sklair-Tavron L, Nudelman R. Drug insight: Tumor necrosis factor-converting enzyme as a pharmaceutical target for rheumatoid arthritis Nat Clin Pract Rheumatol. 2008;4:300–9. https://doi.org/10.1038/ncprheum0797

Most read articles by the same author(s)