Skip to main content

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

  • Review Article
  • Published:

Mechanisms of Disease: protease functions in intestinal mucosal pathobiology

Abstract

Of all our organ systems, the gastrointestinal tract contains the highest levels of endogenous and exogenous proteases (also known as proteinases and peptidases); however, our understanding of their functions and interactions within the gastrointestinal tract is restricted largely to nutrient digestion. The gut epithelium is a sensor of the luminal environment, not only controlling digestive, absorptive and secretory functions, but also relaying information to the mucosal immune, vascular and nervous systems. These functions involve a complex array of cell types that elaborate growth factors, cytokines and extracellular matrix (ECM) proteins, the activity and availability of which are regulated by proteases. Proteolytic activity must be tightly regulated in the face of diverse environmental challenges, because unrestrained or excessive proteolysis leads to pathological gastrointestinal conditions. Moreover, enteric microbes and parasites can hijack proteolytic pathways through 'pathogen host mimicry'. Understanding how the protease balance is maintained and regulated in the intestinal epithelial cell microenvironment and how proteases contribute to physiological and pathological outcomes will undoubtedly contribute to the identification of new potential therapeutic targets for gastrointestinal diseases.

Key Points

  • In addition to their role in nutrient digestion, proteases in the gastrointestinal tract regulate the availability and activity of growth factors, cytokines and extracellular matrix proteins

  • Intestinal epithelial cell biology and function might be controlled by interactions between proteases and their inhibitors and receptors, which are expressed by diverse cells of the intestinal epithelium

  • There are numerous mechanisms in place to ensure that a protease balance is achieved (e.g. synthesis as inactive zymogens, spatial and temporal compartmentalization and termination of activity), because unrestrained or excessive proteolysis can lead to pathological gastrointestinal conditions

  • Enteric microbes and parasites can hijack host proteolytic pathways through 'pathogen host mimicry'

  • To achieve a better understanding of gastrointestinal physiology and disease, it is fundamental that we understand the mechanisms by which proteases function and the ways they are regulated in the complex microenvironment of the cells of the gastrointestinal tract

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

Access options

Buy this article

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

Figure 1: Protease pools relevant to intestinal mucosal pathophysiology
Figure 2: Examples of protease signaling and inter-receptor crosstalk on the cell membrane

Similar content being viewed by others

References

  1. Turk B (2006) Targeting proteases: successes, failures and future prospects. Nat Rev Drug Discov 5: 785–799

    Article  CAS  PubMed  Google Scholar 

  2. Puente XS et al. (2005) A genomic view of the complexity of mammalian proteolytic systems. Biochem Soc Trans 33: 331–334

    Article  CAS  PubMed  Google Scholar 

  3. Neurath H (1999) Proteolytic enzymes, past and future. Proc Natl Acad Sci USA 96: 10962–10963

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Medina C and Radomski MW (2006) Role of matrix metalloproteinases in intestinal inflammation. J Pharmacol Exp Ther 318: 933–938

    Article  CAS  PubMed  Google Scholar 

  5. Neurath H and Walsh KA (1976) Role of proteolytic enzymes in biological regulation (a review). Proc Natl Acad Sci USA 73: 3825–3832

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Eggermont E et al. (1971) Distribution of enterokinase activity in the human intestine. Acta Gastroenterol Belg 34: 655–662

    CAS  PubMed  Google Scholar 

  7. Truninger K et al. (2001) Genetic aspects of chronic pancreatitis: insights into aetiopathogenesis and clinical implications. Swiss Med Wkly 131: 565–574

    CAS  PubMed  Google Scholar 

  8. Netzel-Arnett S et al. (2002) Collagen dissolution by keratinocytes requires cell surface plasminogen activation and matrix metalloproteinase activity. J Biol Chem 277: 45154–45161

    Article  CAS  PubMed  Google Scholar 

  9. Yana I and Weiss SJ (2000) Regulation of membrane type-1 matrix metalloproteinase activation by proprotein convertases. Mol Biol Cell 11: 2387–2401

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Jacob C et al. (2005) Mast cell tryptase controls paracellular permeability of the intestine. Role of protease-activated receptor 2 and beta-arrestins. J Biol Chem 280: 31936–31948

    Article  CAS  PubMed  Google Scholar 

  11. Hooper JD et al. (2001) Type II transmembrane serine proteases. Insights into an emerging class of cell surface proteolytic enzymes. J Biol Chem 276: 857–860

    Article  CAS  PubMed  Google Scholar 

  12. Netzel-Arnett S et al. (2003) Membrane anchored serine proteases: a rapidly expanding group of cell surface proteolytic enzymes with potential roles in cancer. Cancer Metastasis Rev 22: 237–258

    Article  CAS  PubMed  Google Scholar 

  13. Lorey S et al. (2002) Transcellular proteolysis demonstrated by novel cell surface-associated substrates of dipeptidyl peptidase IV (CD26). J Biol Chem 277: 33170–33177

    Article  CAS  PubMed  Google Scholar 

  14. Plow EF and Miles LA (1990) Plasminogen receptors in the mediation of pericellular proteolysis. Cell Differ Dev 32: 293–298

    Article  CAS  PubMed  Google Scholar 

  15. Antalis TM and Lawrence DA (2004) Serpin mutagenesis. Methods 32: 130–140

    Article  CAS  PubMed  Google Scholar 

  16. Herz J and Strickland DK (2001) LRP: a multifunctional scavenger and signaling receptor. J Clin Invest 108: 779–784

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Gschwind A et al. (2001) Cell communication networks: epidermal growth factor receptor transactivation as the paradigm for interreceptor signal transmission. Oncogene 20: 1594–1600

    Article  CAS  PubMed  Google Scholar 

  18. Ohtsu H et al. (2006) ADAMs as mediators of EGF receptor transactivation by G protein-coupled receptors. Am J Physiol Cell Physiol 291: C1–C10

    Article  CAS  PubMed  Google Scholar 

  19. Steinhoff M et al. (2005) Proteinase-activated receptors: transducers of proteinase-mediated signaling in inflammation and immune response. Endocr Rev 26: 1–43

    Article  CAS  PubMed  Google Scholar 

  20. MacNaughton WK (2005) Epithelial effects of proteinase-activated receptors in the gastrointestinal tract. Mem Inst Oswaldo Cruz 100 (Suppl 1): 211–215

    Article  CAS  PubMed  Google Scholar 

  21. Vergnolle N (2000) Review article: proteinase-activated receptors—novel signals for gastrointestinal pathophysiology. Aliment Pharmacol Ther 14: 257–266

    Article  CAS  PubMed  Google Scholar 

  22. Kawabata A (2003) Gastrointestinal functions of proteinase-activated receptors. Life Sci 74: 247–254

    Article  CAS  PubMed  Google Scholar 

  23. Coughlin SR (1999) How the protease thrombin talks to cells. Proc Natl Acad Sci USA 96: 11023–11027

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Nakanishi-Matsui M et al. (2000) PAR3 is a cofactor for PAR4 activation by thrombin. Nature 404: 609–613

    Article  CAS  PubMed  Google Scholar 

  25. Cottrell GS et al. (2004) Trypsin IV, a novel agonist of protease-activated receptors 2 and 4. J Biol Chem 279: 13532–13539

    Article  CAS  PubMed  Google Scholar 

  26. Cottrell GS et al. (2003) Protease-activated receptor 2: activation, signalling and function. Biochem Soc Trans 31: 1191–1197

    Article  CAS  PubMed  Google Scholar 

  27. Dulon S et al. (2005) Pseudomonas aeruginosa elastase disables proteinase-activated receptor 2 in respiratory epithelial cells. Am J Respir Cell Mol Biol 32: 411–419

    Article  CAS  PubMed  Google Scholar 

  28. Dulon S et al. (2003) Proteinase-activated receptor-2 and human lung epithelial cells: disarming by neutrophil serine proteinases. Am J Respir Cell Mol Biol 28: 339–346

    Article  CAS  PubMed  Google Scholar 

  29. Dery O et al. (1998) Proteinase-activated receptors: novel mechanisms of signaling by serine proteases. Am J Physiol 274: C1429–C1452

    Article  CAS  PubMed  Google Scholar 

  30. Vergnolle N (2005) Clinical relevance of proteinase activated receptors (pars) in the gut. Gut 54: 867–874

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Gloro R et al. (2005) Protease-activated receptors: potential therapeutic targets in irritable bowel syndrome? Expert Opin Ther Targets 9: 1079–1095

    Article  CAS  PubMed  Google Scholar 

  32. Ossovskaya VS and Bunnett NW (2004) Protease-activated receptors: contribution to physiology and disease. Physiol Rev 84: 579–621

    Article  CAS  PubMed  Google Scholar 

  33. Dano K et al. (2005) Plasminogen activation and cancer. Thromb Haemost 93: 676–681

    Article  CAS  PubMed  Google Scholar 

  34. Cenac N et al. (2002) Induction of intestinal inflammation in mouse by activation of proteinase-activated receptor-2. Am J Pathol 161: 1903–1915

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Cenac N et al. (2004) PAR2 activation alters colonic paracellular permeability in mice via IFN-gamma-dependent and -independent pathways. J Physiol 558: 913–925

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Wang W et al. (2000) Human zonulin, a potential modulator of intestinal tight junctions. J Cell Sci 113: 4435–4440

    CAS  PubMed  Google Scholar 

  37. Fasano A et al. (2000) Zonulin, a newly discovered modulator of intestinal permeability, and its expression in coeliac disease. Lancet 355: 1518–1519

    Article  CAS  PubMed  Google Scholar 

  38. El Asmar R et al. (2002) Host-dependent zonulin secretion causes the impairment of the small intestine barrier function after bacterial exposure. Gastroenterology 123: 1607–1615

    Article  CAS  PubMed  Google Scholar 

  39. Clemente MG et al. (2003) Early effects of gliadin on enterocyte intracellular signalling involved in intestinal barrier function. Gut 52: 218–223

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Drago S et al. (2006) Gliadin, zonulin and gut permeability: effects on celiac and non-celiac intestinal mucosa and intestinal cell lines. Scand J Gastroenterol 41: 408–419

    Article  CAS  PubMed  Google Scholar 

  41. Fasano A et al. (1995) Zonula occludens toxin modulates tight junctions through protein kinase C-dependent actin reorganization, in vitro. J Clin Invest 96: 710–720

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Fasano A (2001) Pathological and therapeutical implications of macromolecule passage through the tight junction. In Tight Junctions, 697–722, Boca Raton: CRC Press

    Google Scholar 

  43. Weeks CS et al. (2006) Matrix metalloproteinase-7 activation of mouse paneth cell pro-alpha-defensins: SER43 ILE44 proteolysis enables membrane-disruptive activity. J Biol Chem 281: 28932–28942

    Article  CAS  PubMed  Google Scholar 

  44. Wilson CL et al. (1999) Regulation of intestinal alpha-defensin activation by the metalloproteinase matrilysin in innate host defense. Science 286: 113–117

    Article  CAS  PubMed  Google Scholar 

  45. Li CK et al. (2004) Impaired immunity to intestinal bacterial infection in stromelysin-1 (matrix metalloproteinase-3)-deficient mice. J Immunol 173: 5171–5179

    Article  CAS  PubMed  Google Scholar 

  46. Hansen KK et al. (2005) A major role for proteolytic activity and proteinase-activated receptor-2 in the pathogenesis of infectious colitis. Proc Natl Acad Sci USA 102: 8363–8368

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Shafer WM et al. (1991) Human lysosomal cathepsin G and granzyme B share a functionally conserved broad spectrum antibacterial peptide. J Biol Chem 266: 112–116

    CAS  PubMed  Google Scholar 

  48. Smith PD et al. (2005) Intestinal macrophages: unique effector cells of the innate immune system. Immunol Rev 206: 149–159

    Article  CAS  PubMed  Google Scholar 

  49. Anthony RM et al. (2006) Memory T(H)2 cells induce alternatively activated macrophages to mediate protection against nematode parasites. Nat Med 12: 955–960

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Kokai-Kun JF et al. (2000) Elastase in intestinal mucus enhances the cytotoxicity of Shiga toxin type 2d. J Biol Chem 275: 3713–3721

    Article  CAS  PubMed  Google Scholar 

  51. Sears CL (2001) The toxins of Bacteroides fragilis. Toxicon 39: 1737–1746

    Article  CAS  PubMed  Google Scholar 

  52. Sears CL (2000) Molecular physiology and pathophysiology of tight junctions. V. assault of the tight junction by enteric pathogens. Am J Physiol Gastrointest Liver Physiol 279: G1129–G1134

    Article  CAS  PubMed  Google Scholar 

  53. Dutta PR et al. (2002) Functional comparison of serine protease autotransporters of enterobacteriaceae. Infect Immun 70: 7105–7113

    Article  PubMed  PubMed Central  Google Scholar 

  54. Mistry D and Stockley RA (2006) IgA1 protease. Int J Biochem Cell Biol 38: 1244–1248

    Article  CAS  PubMed  Google Scholar 

  55. Na X et al. (2005) Clostridium difficile toxin B activates the EGF receptor and the ERK/MAP kinase pathway in human colonocytes. Gastroenterology 128: 1002–1011

    Article  CAS  PubMed  Google Scholar 

  56. Kosowska K et al. (2002) The Clostridium ramosum IgA proteinase represents a novel type of metalloendopeptidase. J Biol Chem 277: 11987–11994

    Article  CAS  PubMed  Google Scholar 

  57. Lee JD and Yen CM (2005) Protease secreted by the infective larvae of Angiostrongylus cantonensis and its role in the penetration of mouse intestine. Am J Trop Med Hyg 72: 831–836

    Article  CAS  PubMed  Google Scholar 

  58. Zhao A et al. (2005) Immune regulation of protease-activated receptor-1 expression in murine small intestine during Nippostrongylus brasiliensis infection. J Immunol 175: 2563–2569

    Article  CAS  PubMed  Google Scholar 

  59. Shea-Donohue T et al. (2001) The role of IL-4 in Heligmosomoides polygyrus-induced alterations in murine intestinal epithelial cell function. J Immunol 167: 2234–2239

    Article  CAS  PubMed  Google Scholar 

  60. Madden KB et al. (2004) Enteric nematodes induce stereotypic STAT6-dependent alterations in intestinal epithelial cell function. J Immunol. 172: 5616–5621

    Article  CAS  PubMed  Google Scholar 

  61. Shea-Donohue T and Urban JF Jr (2004) Gastrointestinal parasite and host interactions. Curr Opin Gastroenterol 20: 3–9

    Article  PubMed  Google Scholar 

  62. Rhoads ML et al. (2000) Trichuris suis: a secretory chymotrypsin/elastase inhibitor with potential as an immunomodulator. Exp Parasitol 95: 36–44

    Article  CAS  PubMed  Google Scholar 

  63. Pfaff AW et al. (2002) Litomosoides sigmodontis cystatin acts as an immunomodulator during experimental filariasis. Int J Parasitol 32: 171–178

    Article  CAS  PubMed  Google Scholar 

  64. Dainichi T et al. (2001) Nippocystatin, a cysteine protease inhibitor from Nippostrongylus brasiliensis, inhibits antigen processing and modulates antigen-specific immune response. Infect Immun 69: 7380–7386

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Andersson M et al. (2003) Ascaris nematodes from pig and human make three antibacterial peptides: isolation of cecropin P1 and two ASABF peptides. Cell Mol Life Sci 60: 599–606

    Article  CAS  PubMed  Google Scholar 

  66. Podolsky DK (2002) Inflammatory bowel disease. N Engl J Med 347: 417–429

    Article  CAS  PubMed  Google Scholar 

  67. Uehara A et al. (2003) Neutrophil serine proteinases activate human nonepithelial cells to produce inflammatory cytokines through protease-activated receptor 2. J Immunol 170: 5690–5696

    Article  CAS  PubMed  Google Scholar 

  68. Isozaki Y et al. (2006) Anti-tryptase treatment using nafamostat mesilate has a therapeutic effect on experimental colitis. Scand J Gastroenterol 41: 944–953

    Article  CAS  PubMed  Google Scholar 

  69. Morohoshi Y et al. (2006) Inhibition of neutrophil elastase prevents the development of murine dextran sulfate sodium-induced colitis. J Gastroenterol 41: 318–324

    Article  CAS  PubMed  Google Scholar 

  70. Santana A et al. (2006) Attenuation of dextran sodium sulphate induced colitis in matrix metalloproteinase-9 deficient mice. World J Gastroenterol 12: 6464–6472

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Garg P et al. (2006) Selective ablation of matrix metalloproteinase-2 exacerbates experimental colitis: contrasting role of gelatinases in the pathogenesis of colitis. J Immunol 177: 4103–4112

    Article  CAS  PubMed  Google Scholar 

  72. Naito Y and Yoshikawa T (2005) Role of matrix metalloproteinases in inflammatory bowel disease. Mol Aspects Med 26: 379–390

    Article  CAS  PubMed  Google Scholar 

  73. Kirkegaard T et al. (2004) Tumour necrosis factor-alpha converting enzyme (TACE) activity in human colonic epithelial cells. Clin Exp Immunol 135: 146–153

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Wang J et al. (2003) Up-regulation and activation of proteinase-activated receptor 2 in early and delayed radiation injury in the rat intestine: influence of biological activators of proteinase-activated receptor 2. Radiat Res 160: 524–535

    Article  CAS  PubMed  Google Scholar 

  75. Yoshida N et al. (2006) Review article: anti-tryptase therapy in inflammatory bowel disease. Aliment Pharmacol Ther 24 (Suppl 4): 249–255

    Google Scholar 

  76. Broughton G et al. (2006) The basic science of wound healing. Plast Reconstr Surg 117: 12S–S34

    Article  CAS  PubMed  Google Scholar 

  77. Salmela MT et al. (2004) Collagenase-1 (MMP-1), matrilysin-1 (MMP-7), and stromelysin-2 (MMP-10) are expressed by migrating enterocytes during intestinal wound healing. Scand J Gastroenterol 39: 1095–1104

    Article  CAS  PubMed  Google Scholar 

  78. Cao C et al. (2006) Endocytic receptor LRP together with tPA and PAI-1 coordinates Mac-1-dependent macrophage migration. EMBO J 25: 1860–1870

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Roy R et al. (2006) Making the cut: protease-mediated regulation of angiogenesis. Exp Cell Res 312: 608–622

    Article  CAS  PubMed  Google Scholar 

  80. Danese S et al. (2006) Angiogenesis as a novel component of inflammatory bowel disease pathogenesis. Gastroenterology 130: 2060–2073

    Article  CAS  PubMed  Google Scholar 

  81. Itoh H and Kataoka H (2002) Roles of hepatocyte growth factor activator (HGFA) and its inhibitor HAI-1 in the regeneration of injured gastrointestinal mucosa. J Gastroenterol 37 (Suppl 14): 15–21

    Article  PubMed  Google Scholar 

  82. Itoh H et al. (2004) Regeneration of injured intestinal mucosa is impaired in hepatocyte growth factor activator-deficient mice. Gastroenterology 127: 1423–1435

    Article  CAS  PubMed  Google Scholar 

  83. Satomi S et al. (2001) A role for membrane-type serine protease (MT-SP1) in intestinal epithelial turnover. Biochem Biophys Res Commun 287: 995–1002

    Article  CAS  PubMed  Google Scholar 

  84. Lee SL et al. (2000) Activation of hepatocyte growth factor and urokinase/plasminogen activator by matriptase, an epithelial membrane serine protease. J Biol Chem 275: 36720–36725

    Article  CAS  PubMed  Google Scholar 

  85. Netzel-Arnett S et al. (2006) Evidence for a matriptase-prostasin proteolytic cascade regulating terminal epidermal differentiation. J Biol Chem 281: 32941–32945

    Article  CAS  PubMed  Google Scholar 

  86. Jin X et al. (2006) Matriptase activates stromelysin (MMP-3) and promotes tumor growth and angiogenesis. Cancer Sci 97: 1327–1334

    Article  CAS  PubMed  Google Scholar 

  87. Takeuchi T et al. (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. J Biol Chem 275: 26333–26342

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Work by the authors was supported in part by grants from the National Institutes of Health: DK48373 (AF), AI/DK49316 (TSD), CA098369 and HL084387 (TMA), AI18797 (SNV), and DK45496 (CS).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Alessio Fasano.

Ethics declarations

Competing interests

AF and SV have economic interests in Alba Therapeutics, a company that works on the treatment of autoimmune diseases, including type 1 diabetes and celiac disease. The other authors declared they have no competing interests.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Antalis, T., Shea-Donohue, T., Vogel, S. et al. Mechanisms of Disease: protease functions in intestinal mucosal pathobiology. Nat Rev Gastroenterol Hepatol 4, 393–402 (2007). https://doi.org/10.1038/ncpgasthep0846

Download citation

  • Received:

  • Accepted:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncpgasthep0846

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing