Elsevier

Journal of Proteomics

Volume 75, Issue 1, 10 December 2011, Pages 100-115
Journal of Proteomics

Probing small molecule–protein interactions: A new perspective for functional proteomics

https://doi.org/10.1016/j.jprot.2011.07.017Get rights and content

Abstract

The isolation of proteome subsets on the basis of the interactions of small molecules with proteins is an emerging paradigm in proteomics. Depending on the nature of the small molecule used as a bait, entire protein families can be monitored in biological samples, or new functions can be attributed to previously uncharacterized proteins. With pharmaceutical compounds as baits, drug targets and toxicity-relevant off-targets can be discovered in an unbiased proteomic screen. At the heart of this strategy are synthetic bi- or trifunctional small molecule probes. These probes carry the small molecules of interest as baits (selectivity function), as well as a sorting function for the isolation of small molecule–protein complexes or conjugates from complex protein mixtures. In some designs, a covalent linkage of the bound protein to the probe is established through a separate reactivity function or a combined selectivity/reactivity function. The covalent linkage allows for isolation or detection of probe–protein conjugates also under harsh or denaturing conditions. Ultimately, specifically isolated proteins are commonly identified by mass spectrometry. This review summarizes probe designs, workflows, and published applications of the three dominant approaches in the field, namely affinity pulldown, activity-based protein profiling, and Capture Compound Mass Spectrometry.

Graphical abstract

Highlights

► Modular chemical probes enable the isolation of subproteomes defined by functional small molecule-protein interactions. ► Leading strategies are Activity-based protein profiling, affinity pulldown, and Capture Compound mass spectrometry. ► Profiling small molecule-protein interactions contributes to the functional annotation of genomes. ► The approaches enable unbiased direct proteomic screens to identify all proteins that interact with a given drug molecule.

Introduction

Diversification of the experimental approaches with a shift from complex unfractionated protein samples to the investigation of subproteomes has been a general trend in proteomics over the past decade [1]. This allowed identification of lower abundant proteins, and the assignment of some of the functional properties of the proteins, depending on the sample preparation workflow. Examples are the focused analysis of protein sets that share certain types of posttranslational modifications, such as phosphoproteins and their phosphorylation sites in an approach termed “phosphoproteomics” [2]. Other examples are the assignment of proteins to particular subcellular structures, such as nuclei, mitochondria, etc., in an approach termed subcellular proteomics [3], or the large-scale assessment of protein–protein interactions [4]. The common rationale behind these types of analyses was to develop the concept of proteome mapping further towards functional proteomics (see, e.g., [5]). The interactions of metabolites, co-factors, or drug molecules with proteins particularly reveal protein function. It is, therefore, an emerging approach to isolate functional subproteomes on the basis of the interactions of proteins with small molecules (Fig. 1).

Functional subproteome isolation by means of small molecules is associated to the field of chemical proteomics. Chemical proteomics encompasses the profiling of small molecule–protein interactions using small molecule probes (which is the focus of this review article), but also includes molecular biology-driven strategies or small molecule microarrays. Exemplary recent reviews of this wider range of technical approaches, have been provided by Ovaa [6] and Uttamchandani and Yao [7], [8]. Also, chemical proteomics covers the use of synthetic cofactor or substrate analogs to track posttranslational modifications. In this approach, sometimes termed ‘catalomics’ [9], the probes are substrate analogs of the modifying enzymes, and the aim is to identify the acceptor proteins for the respective modification. Published studies addressed methylation [10], [11], palmitoylation [12], acetylation [13], glycosylation [14], phosphorylation [15], [16], or farnesylation [17], [18] (see [9], [19], [20], [21], [22] for recent reviews). Protein targets of disease-relevant modifications such as α, β unsaturated aldehydes generated during free radical-induced damage of polyunsaturated fatty acids have been studied in this way as well [23]. However, a more thorough discussion of this approach is beyond the scope of this review.

Section snippets

Small molecule probes: rationale and design modules

The rationales for designs and functionalities of probes for small molecule–protein interaction profiling comprise elements of affinity chromatography (a key technique in protein purification [24]), photo-affinity labeling (to detect and characterize small molecule binding to target proteins [25]), and active-site chemical labeling of enzymes (see, e.g., [26]). These techniques are traditionally aimed to specifically target and characterize individual proteins. The scope of the newly devised

Small molecule–protein interaction profiling for the functional annotation of the genome and comparative proteomics

In this section, studies are reviewed that aim at the profiling of known and previously unknown members of protein families that share the binding to a particular small molecule. The small molecule of interest is used as the selectivity function of affinity pulldown probes, ABPP or Capture Compounds. ABPP selectivity functions, as outlined above, act as suicide inhibitors through a chemical reaction within enzyme active sites. Thus, the probe is specifically attached via the enzymatic catalysis

Small molecule–protein interaction profiling in drug discovery

One of the most significant foreseeable roles for chemical proteomics and in particular for profiling small molecule–protein interactions is in the field of drug discovery [29], [64], [117]. Through the profiling of interactions between small molecule drugs and proteins, several key questions in drug discovery have been shown to be addressable: (a) Identification of the proteins that underlie the drug mode of action. Drug target proteins can be directly identified and their binding affinities

Conclusion

Already at the present stage of experimental studies that have been conducted, small molecule–protein interaction profiling has emerged as a powerful strategy in proteomics. Functions can be assigned to previously non-annotated gene products, and protein classes assessed in great depth in biological samples. The different technical approaches regarding the design of the small molecule probes each have specific strengths and limitations. While in ABPP, with mechanism-based labeling probes, the

References (133)

  • D. Rotili et al.

    A photoreactive small-molecule probe for 2-oxoglutarate oxygenases

    Chem Biol

    (2011)
  • M. Morak et al.

    Differential activity-based gel electrophoresis for comparative analysis of lipolytic and esterolytic activities

    J Lipid Res

    (2009)
  • F.S. Oppermann et al.

    Large-scale proteomics analysis of the human kinome

    Mol Cell Proteomics

    (2009)
  • J. Wissing et al.

    Proteomics analysis of protein kinases by target class-selective prefractionation and tandem mass spectrometry

    Mol Cell Proteomics

    (2007)
  • H. Daub et al.

    Kinase-selective enrichment enables quantitative phosphoproteomics of the kinome across the cell cycle

    Mol Cell

    (2008)
  • T.T. Aye et al.

    Selectivity in enrichment of cAMP-dependent protein kinase regulatory subunits type I and type II and their interactors using modified cAMP affinity resins

    Mol Cell Proteomics

    (2009)
  • S.E. Hanke et al.

    Cyclic nucleotides as affinity tools: phosphorothioate cAMP analogues address specific PKA subproteomes

    Nat Biotechnol

    (2011)
  • J. Dejardin et al.

    Purification of proteins associated with specific genomic Loci

    Cell

    (2009)
  • M. Bantscheff et al.

    Revealing promiscuous drug–target interactions by chemical proteomics

    Drug Discov Today

    (2009)
  • K.P. Chiang et al.

    An enzyme that regulates ether lipid signaling pathways in cancer annotated by multidimensional profiling

    Chem Biol

    (2006)
  • M.A. Madsen et al.

    Activity-based protein profiling implicates urokinase activation as a key step in human fibrosarcoma intravasation

    J Biol Chem

    (2006)
  • L.C. Gillet et al.

    In-cell selectivity profiling of serine protease inhibitors by activity-based proteomics

    Mol Cell Proteomics

    (2008)
  • D. Greenbaum et al.

    Epoxide electrophiles as activity-dependent cysteine protease profiling and discovery tools

    Chem Biol

    (2000)
  • M.G. Paulick et al.

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

    Curr Opin Genet Dev

    (2008)
  • B.I. Florea et al.

    Activity-based profiling reveals reactivity of the murine thymoproteasome-specific subunit beta5t

    Chem Biol

    (2010)
  • A. Borodovsky et al.

    Chemistry-based functional proteomics reveals novel members of the deubiquitinating enzyme family

    Chem Biol

    (2002)
  • O. Hekmat et al.

    Active-site peptide “fingerprinting” of glycosidases in complex mixtures by mass spectrometry. Discovery of a novel retaining beta-1,4-glycanase in Cellulomonas fimi

    J Biol Chem

    (2005)
  • A.T. Wright et al.

    Chemical proteomic probes for profiling cytochrome p450 activities and drug interactions in vivo

    Chem Biol

    (2007)
  • P.F. van Swieten et al.

    Development of an isotope-coded activity-based probe for the quantitative profiling of cysteine proteases

    Bioorg Med Chem Lett

    (2004)
  • P.A. Everley et al.

    Assessing enzyme activities using stable isotope labeling and mass spectrometry

    Mol Cell Proteomics

    (2007)
  • M. Dreger

    Emerging strategies in mass-spectrometry based proteomics

    Eur J Biochem

    (2003)
  • M. Dreger

    Subcellular proteomics

    Mass Spectrom Rev

    (2003)
  • A. Bauer et al.

    Affinity purification-mass spectrometry. Powerful tools for the characterization of protein complexes

    Eur J Biochem

    (2003)
  • H. Ovaa et al.

    Chemical biology approaches to probe the proteome

    Chembiochem

    (2008)
  • M. Uttamchandani et al.

    The expanding world of small molecule microarrays

    Methods Mol Biol

    (2010)
  • M. Uttamchandani et al.

    Next generation chemical proteomic tools for rapid enzyme profiling

    Acc Chem Res

    (2009)
  • K.A. Kalesh et al.

    The use of click chemistry in the emerging field of catalomics

    Org Biomol Chem

    (2010)
  • C. Dalhoff et al.

    Direct transfer of extended groups from synthetic cofactors by DNA methyltransferases

    Nat Chem Biol

    (2006)
  • O. Binda et al.

    A chemical method for labeling lysine methyltransferase substrates

    Chembiochem

    (2011)
  • B.R. Martin et al.

    Large-scale profiling of protein palmitoylation in mammalian cells

    Nat Methods

    (2009)
  • Y.Y. Yang et al.

    Bioorthogonal chemical reporters for monitoring protein acetylation

    J Am Chem Soc

    (2010)
  • J.A. Prescher et al.

    Chemical remodelling of cell surfaces in living animals

    Nature

    (2004)
  • Y. Chi et al.

    Identification of CDK2 substrates in human cell lysates

    Genome Biol

    (2008)
  • S.E. Lee et al.

    The chemoselective one-step alkylation and isolation of thiophosphorylated Cdk2 substrates in the presence of native cysteine

    Chembiochem

    (2011)
  • A.J. DeGraw et al.

    Evaluation of alkyne-modified isoprenoids as chemical reporters of protein prenylation

    Chem Biol Drug Des

    (2010)
  • L.N. Chan et al.

    A novel approach to tag and identify geranylgeranylated proteins

    Electrophoresis

    (2009)
  • E.W. Tate

    Recent advances in chemical proteomics: exploring the post-translational proteome

    J Chem Biol

    (2008)
  • E.M. Sletten et al.

    Bioorthogonal chemistry: fishing for selectivity in a sea of functionality

    Angew Chem Int Ed Engl

    (2009)
  • J.A. Prescher et al.

    Chemistry in living systems

    Nat Chem Biol

    (2005)
  • O.T. Keppler et al.

    Biochemical engineering of the N-acyl side chain of sialic acid: biological implications

    Glycobiology

    (2001)
  • Cited by (39)

    • Chemical and biology of sulfur fluoride exchange (SuFEx) click chemistry for drug discovery

      2023, Bioorganic Chemistry
      Citation Excerpt :

      Thus fluoride ion is unique base, it achieves strong stabilization in water by catching H+ between two of itself making a centrosymmetric 4-electron-3-center band. (5) Closely related functional groups: Aromatic sulfonyl fluorides are significantly more resistant to hydrolysis than aliphatic derivatives with α-hydrogens, and electron-withdrawing groups on the aromatic ring enhance the electrophilic nature of S (VI) and speed up its reactivity [65]. Sulfonyl fluorides with acidic protons in the α-position pass through elimination process to form sulfene-typeintermediates (R1R2CSO2).

    • Elevated Levels of the Reactive Metabolite Methylglyoxal Recapitulate Progression of Type 2 Diabetes

      2018, Cell Metabolism
      Citation Excerpt :

      These adducts, however, are difficult to isolate biochemically because they are unstable. To circumvent this problem, we used an approach frequently employed to stabilize transient interactions such as ligand-receptor interactions or small molecule-protein interactions, whereby a photoactivatable diazirine cross-linker is added to the molecule of interest (Lenz et al., 2011). Upon photoactivation, the diazirine ring forms a reactive carbene, with a short half-life, causing the molecule of interest to be covalently attached to whichever protein is most proximal.

    • Systematic Targeting of Protein–Protein Interactions

      2016, Trends in Pharmacological Sciences
      Citation Excerpt :

      Several high-throughput mass spectrometry strategies have been implemented to mitigate this challenge. They include, but are not limited to, classic affinity pull-down assays, activity-based protein profiling (ABPP), chemical capture compound assays, stable isotope labeling by amino acids in cell culture (SILAC), isotope-coded affinity tags (ICAT), isobaric tags for relative and absolute quantification (iTRAQ), drug affinity responsive target stability (DARTS), and stability of proteins from rates of oxidation (SPROX) [4,8,9]. In target-based screening, also referred to as ‘reverse chemical genetics’, specific compounds are screened to modulate a particular target or protein of interest [4].

    • Design, synthesis, and evaluation of cell permeable probes for protein kinases

      2016, Tetrahedron
      Citation Excerpt :

      The development and applications of chemical probes have contributed tremendously to the rapid progress in the research of modern chemical biology.1–4

    View all citing articles on Scopus
    1

    T.L. and J.J.F contributed equally to this work.

    View full text