Elsevier

Biochimie

Volume 92, Issue 11, November 2010, Pages 1705-1714
Biochimie

Review
Proteomic techniques and activity-based probes for the system-wide study of proteolysis

https://doi.org/10.1016/j.biochi.2010.04.027Get rights and content

Abstract

Proteolysis constitutes a major post-translational modification but specificity and substrate selectivity of numerous proteases have remained elusive. In this review, we highlight how advanced techniques in the areas of proteomics and activity-based probes can be used to investigate i) protease active site specificity; ii) protease in vivo substrates; iii) protease contribution to proteome homeostasis and composition; and iv) detection and localization of active proteases. Peptide libraries together with genetical or biochemical selection have traditionally been used for active site profiling of proteases. These are now complemented by proteome-derived peptide libraries that simultaneously determine prime and non-prime specificity and characterize subsite cooperativity. Cell-contextual discovery of protease substrates is rendered possible by techniques that isolate and quantitate protein termini. Here, a novel approach termed Terminal Amine Isotopic Labeling of Substrates (TAILS) provides an integrated platform for substrate discovery and appropriate statistical evaluation of terminal peptide identification and quantification. Proteolytically generated carboxy-termini can now also be analyzed on a proteome-wide level. Proteolytic regulation of proteome composition is monitored by quantitative proteomic approaches employing stable isotope coding or label free quantification. Activity-based probes specifically recognize active proteases. In proteomic screens, they can be used to detect and quantitate proteolytic activity while their application in cellular histology allows to locate proteolytic activity in situ. Activity-based probes – especially in conjunction with positron emission tomography – are also promising tools to monitor proteolytic activities on an organism-wide basis with a focus on in vivo tumor imaging. Together, this array of methodological possibilities enables unveiling physiological protease substrate repertoires and defining protease function in the cellular- and organism-wide context.

Section snippets

Degradomics

Proteolytic enzymes sculpt proteomes through limited, site-specific processing or degradation of proteineous substrates. With over 550 proteases in man, the size of this enzyme class reflects the involvement of proteolysis in almost all physiological processes [1]. For most proteases, the in vivo substrate profiles remain to be elucidated. Limited knowledge of protease substrate repertoires restricts our understanding of protease biology and ultimately hinders to fully exploit the therapeutic

Active site specificity

Numerous experimental strategies have been developed to characterize the active site specificity of proteolytic enzymes. An overview of the methods discussed below is shown in Table 1.

Overview

Mass spectrometry together with gel-based or gel-free separation methods offers a powerful way for the rapid identification of 100s–1000s of proteins in biological systems. These techniques can also determine differential protein abundance in different samples. In gel-based systems, spot-staining intensity serves as a direct measure for protein abundance. In gel-free systems, samples are typically labeled with stable isotopes (such as the commercially available ICAT [23], iTRAQ [24] or SILAC

Overview

Global quantitative comparison of different proteomes is inadequate for the determination of proteolytic cleavage sites in complex biological samples as well as being insufficient to profile subtle changes caused by limited, site-specific proteolysis. Since proteolysis creates neo protein termini, multiple strategies have been designed with the aim to monitor proteolytic events on a proteome-wide scale by identifying and quantifying protein N-termini. Generally, these methods enrich for

Overview

Most proteases are tightly regulated on the post-translational level and aberrant protease activities are characteristics of diseases such as cancer and arthritis. However, techniques monitoring protein abundances do not account for these changes and while giving insight into mechanisms of protease action the identification of substrates and cleavage sites does not detect subtle but detrimental alterations in activity levels. Furthermore, those techniques are based on the analysis of protein

Perspective

No single method alone can uncover the multiple roles of proteases in physiology and pathology. In this review we have highlighted how degradomic techniques offer new routes to uncover key features in protease biology, including specificity, proteome-wide effects, natural substrates, and localization of proteolytic activity. An integrated, multi-facetted approach that appropriately combines these technologies will ultimately provide the best platform to characterize and understand proteolysis

Acknowledgement

O.S. acknowledges support from the Deutsche Forschungsgemeinschaft (DFG) (grant SCHI871/2-1).

References (104)

  • L. Guo et al.

    A proteomic approach for the identification of cell-surface proteins shed by metalloproteases

    Mol. Cell. Proteomics

    (2002)
  • M. Shen et al.

    Isolation and isotope labeling of cysteine- and methionine-containing tryptic peptides: application to the study of cell surface proteolysis

    Mol. Cell. Proteomics

    (2003)
  • R.A. Dean et al.

    Proteomics discovery of metalloproteinase substrates in the cellular context by iTRAQTM labeling reveals a diverse MMP-2 substrate degradome

    Mol. Cell. Proteomics

    (2007)
  • M.M. Dix et al.

    Global mapping of the topography and magnitude of proteolytic events in apoptosis

    Cell

    (2008)
  • T.H. Akiyama et al.

    A method for selective isolation of the amino-terminal peptide from alpha-amino-blocked proteins

    Anal. Biochem

    (1994)
  • G. Coussot et al.

    A method for isolation blocked N-terminal peptides

    Anal. Biochem.

    (2007)
  • H. Kaplan et al.

    Selective isolation of free and blocked amino-terminal peptides from enzymatic digestion of proteins

    Anal. Biochem.

    (1983)
  • S. Mahrus et al.

    Global sequencing of proteolytic cleavage sites in apoptosis by specific labeling of protein N termini

    Cell

    (2008)
  • F.-N. Vögtle et al.

    Global analysis of the mitochondrial N-proteome identifies a processing peptidase critical for protein stability

    Cell

    (2009)
  • U. Auf dem Keller et al.

    A statistics based platform for quantitative N-terminome analysis and identification of protease cleavage products

    Mol. Cell. Proteomics

    (2010)
  • A. Prudova et al.

    Multiplex N-terminome analysis of MMP-2 and MMP-9 substrate degradomes by iTRAQ-TAILS quantitative proteomics

    Mol. Cell. Proteomics

    (2010)
  • J. Vendrell et al.

    Metallocarboxypeptidases and their protein inhibitors. Structure, function and biomedical properties

    Biochim. Biophys. Acta

    (2000)
  • H.P. Bietlot et al.

    Isolation of carboxyl-terminal peptides from proteins by diagonal electrophoresis: application to the entomocidal toxin from Bacillus thuringiensis

    Anal. Biochem.

    (1989)
  • K. Helsens et al.

    Peptizer, a tool for assessing false positive peptide identifications and manually validating selected results

    Mol. Cell. Proteomics

    (2008)
  • A. Baruch et al.

    Defining a link between gap junction communication, proteolysis, and cataract formation

    J. Biol. Chem.

    (2001)
  • M.G. Paulick et al.

    Application of activity-based probes to the study of enzymes involved in cancer progression

    Curr. Opin. Genet. Dev.

    (2008)
  • A.B. Berger et al.

    Identification of early intermediates of caspase activation using selective inhibitors and activity-based probes

    Mol. Cell

    (2006)
  • J.A. Joyce et al.

    Cathepsin cysteine proteases are effectors of invasive growth and angiogenesis during multistage tumorigenesis

    Cancer Cell

    (2004)
  • Q.H. Zheng et al.

    Comparative studies of potential cancer biomarkers carbon-11 labeled MMP inhibitors (S)-2-(4’-[11C]methoxybiphenyl-4-sulfonylamino)-3-methylbutyric acid and N-hydroxy-(R)-2-[[(4’-[11C]methoxyphenyl)sulfonyl]benzylamino]-3-methylbutanamide

    Nucl. Med. Biol.

    (2004)
  • J.E. Sprague et al.

    In vitro and in vivo investigation of matrix metalloproteinase expression in metastatic tumor models

    Nucl. Med. Biol.

    (2006)
  • S. Furumoto et al.

    Tumor detection using 18F-labeled matrix metalloproteinase-2 inhibitor

    Nucl. Med. Biol.

    (2003)
  • X.S. Puente et al.

    Human and mouse proteases: a comparative genomic approach

    Nat. Rev. Genet.

    (2003)
  • L.M. Coussens et al.

    Matrix metalloproteinase inhibitors and cancer: trials and tribulations

    Science

    (2002)
  • B. Turk

    Targeting proteases: successes, failures and future prospects

    Nat. Rev. Drug Discov.

    (2006)
  • O. Schilling et al.

    Proteome-derived, database-searchable peptide libraries for identifying protease cleavage sites

    Nat. Biotechnol.

    (2008)
  • C.M. Overall

    Molecular determinants of metalloproteinase substrate specificity: matrix metalloproteinase substrate binding domains, modules, and exosites

    Mol. Biotechnol.

    (2002)
  • C. López-Otín et al.

    Protease degradomics: a new challenge for proteomics

    Nat. Rev. Mol. Cell. Biol.

    (2002)
  • D.J. Matthews et al.

    Substrate phage: selection of protease substrates by monovalent phage display

    Science

    (1993)
  • S.L. Diamond

    Methods for mapping protease specificity

    Curr. Opin. Chem. Biol.

    (2006)
  • K.T. Boulware et al.

    Protease specificity determination by using cellular libraries of peptide substrates (CLiPS)

    Proc. Natl. Acad. Sci. U.S.A.

    (2006)
  • C.M. Salisbury et al.

    Peptide microarrays for the determination of protease substrate specificity

    J. Am. Chem. Soc.

    (2002)
  • G. Rossé et al.

    Rapid identification of substrates for novel proteases using a combinatorial peptide library

    J. Comb. Chem.

    (2000)
  • D.N. Gosalia et al.

    Profiling serine protease substrate specificity with solution phase fluorogenic peptide microarrays

    Proteomics

    (2005)
  • B.E. Turk et al.

    Determination of protease cleavage site motifs using mixture-based oriented peptide libraries

    Nat. Biotechnol.

    (2001)
  • B.J. Backes et al.

    Synthesis of positional-scanning libraries of fluorogenic peptide substrates to define the extended substrate specificity of plasmin and thrombin

    Nat. Biotechnol.

    (2000)
  • O. Schilling, U. Auf dem Keller, C.M. Overall, Protease specificity profiling by tandem mass spectrometry using...
  • S.P. Gygi et al.

    Quantitative analysis of complex protein mixtures using isotope-coded affinity tags

    Nat. Biotechnol.

    (1999)
  • A.Y. Lee et al.

    Identification of caspase-3 degradome by two-dimensional gel electrophoresis and matrix-assisted laser desorption/ionization-time of flight analysis

    Proteomics

    (2004)
  • A.J. Bredemeyer et al.

    A proteomic approach for the discovery of protease substrates

    Proc. Natl. Acad. Sci. U.S.A.

    (2004)
  • J.J. Bech-Serra et al.

    Proteomic identification of desmoglein 2 and activated leukocyte cell adhesion molecule as substrates of ADAM17 and ADAM10 by difference gel electrophoresis

    Mol. Cell. Biol.

    (2006)
  • Cited by (48)

    • Detection of protease activity in cells and animals

      2016, Biochimica et Biophysica Acta - Proteins and Proteomics
      Citation Excerpt :

      Nowadays, a wide variety of chromogenic, fluorogenic and luminogenic substrates are commercially available or accessible by organic synthesis, allowing sensitive and high-throughput measurement of protease activity (Fig. 2B). The design of substrate reporters is based on the substrate specificity of the target protease, which itself can be uncovered by using libraries of synthetic substrates [15] or proteomics methods [16,17]. The design of a substrate that is only turned over by one particular protease is still a difficult task, since proteases from the same family often have overlapping substrate specificity.

    • Clip-domain serine proteases as immune factors in insect hemolymph

      2015, Current Opinion in Insect Science
      Citation Excerpt :

      Most of the regulatory action in such systems occurs as post-translational modifications (zymogen activation, protease cleavage of substrates, protease interaction with inhibitors) rather than at the level of gene expression. Even for most human plasma proteases, the in vivo substrate profiles have not been elucidated [51], and those which have display astonishing complexity. Thrombin, for example, has at least nine important substrates, with pro-clotting and anti-clotting activities depending on physiological context and interaction with cofactors [52].

    • Proteomic approaches to uncover MMP function

      2015, Matrix Biology
      Citation Excerpt :

      For this purpose, activity-based probes (ABPs) were developed, comprising a warhead that covalently binds to the active site of proteases and that is attached to a reporter group, e.g. a fluorophore or an affinity handle like biotin. This concept was successfully employed to study many proteases, which form covalently bound substrate intermediates, such as serine and cysteine proteases [19,20]. Following the same principles researchers adapted ABPs for activity profiling of metalloproteases [21,22].

    • Proteomic protease specificity profiling of clostridial collagenases reveals their intrinsic nature as dedicated degraders of collagen

      2014, Journal of Proteomics
      Citation Excerpt :

      Residues N-terminal to the scissile peptide bond are designated as the non-prime (P) side, and residues C-terminal as prime (P′) side, both interacting with complementary substrate recognition sites in the protease active site cleft, termed S and S′, respectively. Protease specificity can be profiled using a variety of approaches [44]. Uniquely among these, Proteomic Identification of protease Cleavage Sites (PICS) determines both the full prime and non-prime cleavage site preferences in a single experiment and in a high throughput manner [45].

    • Comparative characterization of direct and indirect substrate probes for on-chip transamidating activity assay of transglutaminases

      2013, Journal of Biotechnology
      Citation Excerpt :

      Activity-based protein profiling (ABPP) has emerged as a powerful strategy for assigning protein functions by identifying and characterizing the catalytic activity of proteins and their complexes that control biological processes of interest (Heal et al., 2010). ABPP is also used for the identification of enzyme substrates or inhibitors, the discovery of diagnostic markers, and the development of therapeutic reagents (auf dem Keller and Schilling, 2010; Gillet et al., 2008; Wiedl et al., 2011). Designing specific probes that either directly or indirectly detect the catalytic activity of enzymes is an essential prerequisite for ABPP (Sadaghiani et al., 2007; Speers and Cravatt, 2004).

    View all citing articles on Scopus
    View full text