Chemical Proteomics Methods and Protocols 1st Edition Marcus Bantscheff (Auth.)
Chemical Proteomics Methods and Protocols 1st Edition Marcus Bantscheff (Auth.)
Chemical Proteomics Methods and Protocols 1st Edition Marcus Bantscheff (Auth.)
Chemical Proteomics Methods and Protocols 1st Edition Marcus Bantscheff (Auth.)
1. Visit https://ebookultra.com to download the full version and
explore more ebooks
Chemical Proteomics Methods and Protocols 1st
Edition Marcus Bantscheff (Auth.)
_____ Click the link below to download _____
https://ebookultra.com/download/chemical-proteomics-
methods-and-protocols-1st-edition-marcus-bantscheff-
auth/
Explore and download more ebooks at ebookultra.com
2. Here are some suggested products you might be interested in.
Click the link to download
Chemical Proteomics Methods and Protocols Methods in
Molecular Biology v803 2012th Edition Gerard Drewes
https://ebookultra.com/download/chemical-proteomics-methods-and-
protocols-methods-in-molecular-biology-v803-2012th-edition-gerard-
drewes/
Heart Proteomics Methods and Protocols 1st Edition Jake
Cosme
https://ebookultra.com/download/heart-proteomics-methods-and-
protocols-1st-edition-jake-cosme/
Chemical Genomics and Proteomics Reviews and Protocols 1st
Edition Edward D. Zanders (Auth.)
https://ebookultra.com/download/chemical-genomics-and-proteomics-
reviews-and-protocols-1st-edition-edward-d-zanders-auth/
Cancer Genomics and Proteomics Methods and Protocols
Methods in Molecular Biology 1st Edition Paul B. Fisher
https://ebookultra.com/download/cancer-genomics-and-proteomics-
methods-and-protocols-methods-in-molecular-biology-1st-edition-paul-b-
fisher/
3. Proteomics Methods Applications and Limitations Methods
Applications and Limitations 1st Edition Giselle C.
Rancourt
https://ebookultra.com/download/proteomics-methods-applications-and-
limitations-methods-applications-and-limitations-1st-edition-giselle-
c-rancourt/
Computational methods for mass spectrometry proteomics 1st
Edition Ingvar Eidhammer
https://ebookultra.com/download/computational-methods-for-mass-
spectrometry-proteomics-1st-edition-ingvar-eidhammer/
Genomics Proteomics and Clinical Bacteriology Methods and
Reviews 1st Edition Mark J. Pallen (Auth.)
https://ebookultra.com/download/genomics-proteomics-and-clinical-
bacteriology-methods-and-reviews-1st-edition-mark-j-pallen-auth/
Chemical Genomics Reviews and Protocols 1st Edition Paul
R. Caron (Auth.)
https://ebookultra.com/download/chemical-genomics-reviews-and-
protocols-1st-edition-paul-r-caron-auth/
Polyadenylation Methods and Protocols 1st Edition Joanna
Rorbach
https://ebookultra.com/download/polyadenylation-methods-and-
protocols-1st-edition-joanna-rorbach/
5. Chemical Proteomics Methods and Protocols 1st Edition
Marcus Bantscheff (Auth.) Digital Instant Download
Author(s): Marcus Bantscheff (auth.), Gerard Drewes, Marcus Bantscheff
(eds.)
ISBN(s): 9781617793646, 1617793647
Edition: 1
File Details: PDF, 4.02 MB
Year: 2012
Language: english
6. Me t h o d s i n Mo l e c u l a r Bi o l o g y ™
Series Editor
John M. Walker
School of Life Sciences
University of Hertfordshire
Hatfield, Hertfordshire, AL10 9AB, UK
For further volumes:
http://www.springer.com/series/7651
10. v
Preface
An Overview of Chemical Proteomics: Methods and Applications
The multidisciplinary science of chemical proteomics studies how small molecules of
synthetic or natural origin bind to proteins and modulate their function. Scientists in the
field have different backgrounds including molecular and cell biology, biochemistry, phar-
macology, organic chemistry, and physics. Chemical Proteomics: Methods and Applications is
directed at molecular biologists and biochemists with either an interest in small molecules
themselves, e.g., in drug discovery projects, or in using small-molecule probes as research
tools to study protein function. The book may also be useful for organic chemists with an
interest in biology and for specialists in protein mass spectrometry.
In the introductory chapters, we discuss analytical strategies for chemical proteomics
projects, with a focus on the current state-of-the-art in protein mass spectrometry, and
describe several examples how chemical proteomics can impact the field of drug discovery.
The consecutive chapters provide detailed experimental protocols. Most chemical proteom-
ics projects consist of three parts. In the first part, the chemical probe is selected or designed,
and then synthesized. In the second part, the probe compound is exposed to the protein
sample or cell extract. In the final part, proteins binding to the probe compound are identi-
fied and often quantified. In simple applications, this is often achieved by antibody-based
detection, but if the aim is the discovery of targets in an unbiased fashion, mass spectrom-
etry is the method of choice. The following chapters cover all of these aspects.
The first set of chapters describes how probes are generated from commercially available
reagents without elaborate chemical synthesis procedures, and how the proteins binding to
the probes can be analyzed by immunodetection or by mass spectrometry. Rix et al. and
Saxena provide protocols for direct noncovalent affinity capture using protein kinase inhibi-
tors as an example, which serves to profile the targets of these compounds and provides
probes for kinase expression and activity. Ge and Sem have developed a target class-specific
probe for the labeling of dehydrogenase enzymes. Kawamura and Mihai, and Codreanu
et al. describe the use of biotin-conjugated probes which form covalent adducts with defined
subproteomes, here consisting of adenine-binding proteins and targets of lipid electrophiles.
Lenz et al. combine features of noncovalent and covalent capturing strategies in their bifunc-
tional ligands designed to target methyltransferases, an emerging class of drug targets.
The second set of chapters is concerned with techniques for the discovery of small-
molecule targets and the probing of target function. Ong et al. describe the use of stable
isotope labeling of amino acids in cell culture (SILAC) in identifying proteins that bind
small-molecule probes in cell extracts. Hopf et al. perform affinity enrichment of target pro-
teins on a probe matrix in the presence of competing free test compound in solution, thus
enabling determination of binding potencies of the free test compound to affinity-captured
proteins from cell extracts. The method employs quantitative mass spectrometry with iso-
baric mass tags to determine the potencies for a large number of targets in a single analysis.
Ge and Sem have developed a protocol for the detection and purification of dehydrogenase
11. vi Preface
enzymes, which may represent targets, but also unwanted off-targets, for certain types of
drugs. Kovanich et al. describe a combination of cAMP-based affinity chromatography with
quantitative mass spectrometry to investigate protein kinase A complexes in extracts of cells
and tissues. De Jong et al. use activity-based chemical probes to profile the activity of the
proteasome, which has recently emerged as an important cancer target, in cells and tissues.
The next three chapters provide innovative protocols for the study of potential drug targets
by chemical cross-linking and mass spectrometry. Mueller et al. provide a method to study
protein–drug interactions, and Gasilova et al. employ cross-linking and MALDI-mass spec-
trometry to study ligand modulation of protein–protein interactions. Jeon et al. provide a
protocol for in vivo cross-linking via time-controlled transcardiac perfusion, which in prin-
ciple enables the direct analysis of protein targets in animal models.
The final set of chapters is concerned with small-molecule ligand and drug discovery.
Casalena et al. describe the discovery of probe compounds by utilizing compound libraries
immobilized on microarrays. Wolf et al. delineate general guidelines for working with small
molecules, including aspects like storage, the preparation of solutions, and the determina-
tion of solubility. The chapter by de Matos et al. provides guidelines for the use of the
ChEBI database, which should be very helpful for researchers tasked with the selection of
a particular probe or with building a small molecule collection to purpose. They describe
the Chemical Entities of Biological Interest (ChEBI) database which helps to find probe
molecules with the desired structural or biological features. Finally, many researchers will
consider whether their research tool compound might have the potential to be developed
into a drug. Zhang delivers a lucid analysis of the features that distinguish drugs from probe
molecules, and lays out a set of rules for “drug likeness.”
Affinity- and activity-based chemical probes, combined with quantitative immunodetec-
tion and mass spectrometry techniques, are increasingly gaining appreciation as powerful
strategies for the molecular analysis of complex biological systems in homeostasis and dis-
ease. We hope that the methodologies described in this volume will contribute to a wider
application of chemical proteomics methods in biochemical and cell biological laboratories.
Heidelberg, Germany Gerard Drewes
Marcus Bantscheff
12. vii
Contents
Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Contributors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ix
Part I Introduction
1 Mass Spectrometry-Based Chemoproteomic Approaches . . . . . . . . . . . . . . . . . . . . 3
Marcus Bantscheff
2 Chemical Proteomics in Drug Discovery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
Gerard Drewes
Part II Small Molecules and Probe Design
3 Compound Immobilization and Drug-Affinity Chromatography . . . . . . . . . . . . . . 25
Uwe Rix, Manuela Gridling, and Giulio Superti-Furga
4 Affinity-Based Chemoproteomics with Small Molecule-Peptide Conjugates . . . . . . 39
Chaitanya Saxena
5 A Chemical Proteomic Probe for Detecting Dehydrogenases:
Catechol Rhodanine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 55
Xia Ge and Daniel S. Sem
6 Probing Proteomes with Benzophenone Photoprobes . . . . . . . . . . . . . . . . . . . . . . 65
Akira Kawamura and Doina M. Mihai
7 Biotinylated Probes for the Analysis of Protein Modification
by Electrophiles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 77
Simona G. Codreanu, Hye-Young H. Kim, Ned A. Porter,
and Daniel C. Liebler
8 Profiling of Methyltransferases and Other S-Adenosyl-l-Homocysteine-
Binding Proteins by Capture Compound Mass Spectrometry . . . . . . . . . . . . . . . . 97
Thomas Lenz, Peter Poot, Elmar Weinhold, and Mathias Dreger
Part III Target Discovery and Target Validation
9 Identifying Cellular Targets of Small-Molecule Probes and Drugs
with Biochemical Enrichment and SILAC . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 129
Shao-En Ong, Xiaoyu Li, Monica Schenone, Stuart L. Schreiber,
and Steven A. Carr
10 Determination of Kinase Inhibitor Potencies in Cell Extracts
by Competition Binding Assays and Isobaric Mass Tags . . . . . . . . . . . . . . . . . . . . 141
Carsten Hopf, Dirk Eberhard, Markus Boesche, Sonja Bastuck,
Birgit Dümpelfeld, and Marcus Bantscheff
11 Affinity-Based Profiling of Dehydrogenase Subproteomes . . . . . . . . . . . . . . . . . . . 157
Xia Ge and Daniel S. Sem
13. viii Contents
12 Probing the Specificity of Protein–Protein Interactions
by Quantitative Chemical Proteomics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 167
Duangnapa Kovanich, Thin Thin Aye, Albert J.R. Heck,
and Arjen Scholten
13 Fluorescence-Based Proteasome Activity Profiling . . . . . . . . . . . . . . . . . . . . . . . . . 183
Annemieke de Jong, Karianne G. Schuurman, Boris Rodenko,
Huib Ovaa, and Celia R. Berkers
14 Chemical Cross-Linking and High-Resolution Mass Spectrometry
to Study Protein–Drug Interactions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 205
Mathias Q. Müller and Andrea Sinz
15 Monitoring Ligand Modulation of Protein–Protein Interactions by Chemical
Cross-Linking and High-Mass MALDI Mass Spectrometry . . . . . . . . . . . . . . . . . . 219
Natalia Gasilova and Alexis Nazabal
16 Time-Controlled Transcardiac Perfusion Crosslinking
for In Vivo Interactome Studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 231
Amy Hye Won Jeon and Gerold Schmitt-Ulms
Part IV Ligand Discovery
17 Ligand Discovery Using Small-Molecule Microarrays . . . . . . . . . . . . . . . . . . . . . . 249
Dominick E. Casalena, Dina Wassaf, and Angela N. Koehler
18 Working with Small Molecules: Preparing and Storing Stock
Solutions and Determination of Kinetic Solubility . . . . . . . . . . . . . . . . . . . . . . . . . 265
Andrea Wolf, Satoko Shimamura, and Friedrich B.M. Reinhard
19 A Database for Chemical Proteomics: ChEBI . . . . . . . . . . . . . . . . . . . . . . . . . . . . 273
Paula de Matos, Nico Adams, Janna Hastings, Pablo Moreno,
and Christoph Steinbeck
20 Working with Small Molecules: Rules-of-Thumb of “Drug Likeness” . . . . . . . . . . . 297
Ming-Qiang Zhang
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 309
14. ix
Contributors
Nico Adams • Department of Genetics, University of Cambridge, Cambridge, UK
Thin Thin Aye • Biomolecular Mass Spectrometry and Proteomics Group,
Utrecht University and Netherlands Proteomics Centre, Utrecht, The Netherlands
Marcus Bantscheff • Cellzome AG, Heidelberg, Germany
Sonja Bastuck • Cellzome AG, Heidelberg, Germany
Celia R. Berkers • Division of Cell Biology II, The Netherlands Cancer Institute,
Amsterdam, The Netherlands
Markus Boesche • Cellzome AG, Heidelberg, Germany
Dominick E. Casalena • Chemical Biology Platform, The Broad Institute of MIT
and Harvard, Cambridge, MA, USA
Steven A. Carr • Proteomics platform, The Broad Institute of MIT and Harvard,
Cambridge, MA, USA
Simona G. Codreanu • Vanderbilt University School of Medicine, Nashville, TN, USA
Annemieke de Jong • Division of Cell Biology II, The Netherlands Cancer Institute,
Amsterdam, The Netherlands
Paula de Matos • European Bioinformatics Institute, Hinxton, UK
Mathias Dreger • caprotec bioanalytics GmbH, Berlin, Germany
Gerard Drewes • Cellzome AG, Heidelberg, Germany
Birgit Dümpelfeld • Cellzome AG, Heidelberg, Germany
Dirk Eberhard • Cellzome AG, Heidelberg, Germany
Natalia Gasilova • CovalX AG, Schlieren, Switzerland; Department of Chemistry
and Applied Biosciences, ETH, Zürich, Switzerland
Xia Ge • Chemical Proteomics Facility at Marquette, Department of Chemistry,
Marquette University, Milwaukee, WI, USA
Manuela Gridling • CeMM Research Center for Molecular Medicine of the Austrian
Academy of Sciences, Vienna, Austria
Janna Hastings • European Bioinformatics Institute, Hinxton, UK
Albert J.R. Heck • Biomolecular Mass Spectrometry and Proteomics Group,
Utrecht University and Netherlands Proteomics Centre, Utrecht, The Netherlands
Carsten Hopf • Cellzome AG, Heidelberg, Germany
Amy Hye Won Jeon • Tanz Centre for Research in Neurodegenerative Diseases
and Department of Laboratory Medicine and Pathobiology, University of Toronto,
Toronto, ON, Canada
Akira Kawamura • Department of Chemistry, Hunter College of CUNY, New York,
NY, USA
Hye-Young H. Kim • Vanderbilt University School of Medicine, Nashville, TN, USA
Angela N. Koehler • Chemical Biology Platform, The Broad Institute of MIT
and Harvard, Cambridge, MA, USA
Duangnapa Kovanich • Biomolecular Mass Spectrometry and Proteomics Group,
Utrecht University and Netherlands Proteomics Centre, Utrecht, The Netherlands
15. x Contributors
Thomas Lenz • caprotec bioanalytics GmbH, Berlin, Germany
Xiaoyu Li • Chemical Biology platform, The Broad Institute of MIT and Harvard,
Cambridge, MA, USA
Daniel C. Liebler • Vanderbilt University School of Medicine, Nashville, TN, USA
Doina M. Mihai • Department of Chemistry, Hunter College of CUNY, New York,
NY, USA
Pablo Moreno • European Bioinformatics Institute, Hinxton, UK
Mathias Q. Müller • Department of Pharmaceutical Chemistry and Bioanalytics,
Institute of Pharmacy, Martin-Luther-Universität Halle-Wittenberg, Halle (Saale),
Germany
Alexis Nazabal • CovalX AG, Schlieren, Switzerland
Shao-En Ong • Proteomics Platform, The Broad Institute of MIT and Harvard,
Cambridge, MA, USA
Huib Ovaa • Division of Cell Biology II, The Netherlands Cancer Institute,
Amsterdam, The Netherlands
Peter Poot • Institute of Organic Chemistry, RWTH University, Aachen, Germany
Ned A. Porter • Vanderbilt University School of Medicine, Nashville, TN, USA
Friedrich B.M. Reinhard • Cellzome AG, Heidelberg, Germany
Uwe Rix • CeMM Research Center for Molecular Medicine of the Austrian Academy
of Sciences, Vienna, Austria
Boris Rodenko • Division of Cell Biology II, The Netherlands Cancer Institute,
Amsterdam, The Netherlands
Chaitanya Saxena • Shantani Proteome Analytics Pvt. Ltd., Pune, MH, India
Monica Schenone • Proteomics platform, The Broad Institute of MIT and Harvard,
Cambridge, MA, USA
Gerold Schmitt-Ulms • Tanz Centre for Research in Neurodegenerative Diseases
and Department of Laboratory Medicine and Pathobiology, University of Toronto,
Toronto, ON, Canada
Arjen Scholten • Biomolecular Mass Spectrometry and Proteomics Group,
Utrecht University andNetherlands Proteomics Centre, Utrecht, The Netherlands
Stuart L. Schreiber • Chemical Biology Platform Chemical Biology Program,
The Broad Institute of MIT and Harvard, Cambridge, MA, USA
Karianne G. Schuurman • Division of Cell Biology II, The Netherlands Cancer
Institute, Amsterdam, The Netherlands
Daniel S. Sem • Department of Pharmaceutical Sciences, Concordia University
Wisconsin, Mequon, WI, USA
Satoko Shimamura • Cellzome AG, Heidelberg, Germany
Andrea Sinz • Department of Pharmaceutical Chemistry and Bioanalytics,
Institute of Pharmacy, Martin-Luther-Universität Halle-Wittenberg,
Halle (Saale), Germany
Christoph Steinbeck • European Bioinformatics Institute, Hinxton, UK
Giulio Superti-Furga • CeMM Research Center for Molecular Medicine
of the Austrian Academy of Sciences, Vienna, Austria
Dina Wassaf • Chemical Biology Platform, The Broad Institute of MIT and Harvard,
Cambridge, MA, USA
16. xi
Contributors
Elmar Weinhold • Institute of Organic Chemistry, RWTH University,
Aachen, Germany
Andrea Wolf • Cellzome AG, Heidelberg, Germany
Ming-Qiang Zhang • Merck Sharp Dohme (MSD), Chaoyang District,
Beijing, PR, China
20. 4 M. Bantscheff
For global chemoproteomic profiling, cells or animals are treated
with a drug before system-wide proteome analysis to evaluate the
cellular response in a global way (9–12). This strategy has become
very attractive because of its simplicity and the unbiased nature of
the analysis. Apart from cell permeability, there are no particular
requirements for the small molecule compounds to be tested in
such assays and the required quantitative mass spectrometric tech-
niques are now available in many laboratories (for recent reviews,
see refs. 13–16). However, since typically no protein enrichment is
used, the changes that can be observed are limited by the analytical
depth of the analysis and are often restricted to more abundant
proteins. The analysis is further complicated by the fact that pro-
teins found to be altered in abundance are not necessarily direct
targets or downstream of the affected signaling cascade but often
represent highly abundant proteins involved in stress response
and/or housekeeping functions (12, 17). Hence, it is almost never
possible to distinguish direct drug protein interactions from indi-
rect effects. In examples illustrating the advantages and limitations
2. Global
Chemoproteomic
Profiling
a b
Wash
Digest proteins
LC-MS/MS
Extract proteins
Add Beads
m/z
Purify probes
on beads
Wash
Digest proteins
LC-MS/MS
Extract proteins
Add probe
React probe
c
m/z
Digest proteins
LC-MS/MS
Extract proteins
m/z
+Compound
Control +Compound
Control +Compound
Control
m/z m/z m/z
1
a.i.
1
a.i.
1
a.i.
1
a.i.
1
a.i.
1
a.i.
Fig. 1. Experimental workflows in chemical proteomics. (a) Global proteomics approaches: Cells are treated with a com-
pound, proteins are extracted from the sample, digested to peptides and analyzed by liquid chromatography coupled to
tandem mass spectrometry (LC-MS/MS). (b) Activity-based protein profiling (ABPP) utilizes reactive probes specifically
targeting the active site of an enzyme family. After protein extraction, the lysate is incubated with the probe to covalently
attach to its targets. In the second step, probes and targets are purified using affinity chromatography before digestion and
LC-MS/MS analysis. Pretreatment of cells with a small molecule compound binding to the active site of the investigated
enzyme family leads to reduced capturing of the target enzyme via the reactive probes. (c) Alternatively, the compounds of
interest can be modified and immobilized on a solid support. The immobilized drug is subsequently incubated with a cell
extract to specifically enrich for target proteins that are subsequently identified by mass spectrometry. Competition with
free excess inhibitor reduces the abundance of captured target proteins.
21. 5
1 Mass Spectrometry-Based Chemoproteomic Approaches
of global proteomics approaches, Chen and co-workers (12, 18, 19)
evaluated the differential effect of the R- and S-enantiomers of
atenolol, a β1-selective adrenoreceptor blocker, and the nonsteroi-
dal antiinflammatory drug ibuprofen on two different cell types.
The authors found 27 and 13 proteins to be differentially expressed,
most of which can be classified as highly abundant (17). Yamanaka
and co-workers applied a global proteomics approach for toxico-
logical studies in animals (20). The authors studied the effects of
63 chemical compounds on protein expression in rat liver after 28
daily dosings and employed statistical methods to detect proteins
characteristic for carcinogenicity.
Subcellular and chromatographic fractionation allow for a more
directed analysis of drug-induced changes in protein expression or
posttranslational modifications. Lee and co-authors monitored his-
tone modifications in response to treatment with histone deacety-
lase (HDAC) inhibitors (10). In this study, human colon cancer
cells were treated with HDAC inhibitors of varying degrees of
selectivity followed by a simple prefractionation method to enrich
for histone proteins. By employing label-free quantitative mass
spectrometry, the authors identified HDAC-dependent histone
acetylation patterns and quantified these in response to inhibitor
treatment. Similarly, JmjD2A-dependent demethylation of K9 in
histone H3 was monitored in response to cell treatment with
pyridine-2,4-dicarboxylic acid derivatives (21). In several recent
studies, chromatographic- or antibody-based enrichment methods
have been employed to analyze kinase inhibitor-induced changes in
protein phosphorylation on a global scale, thus mapping the net-
work-level response to inhibitor treatment, and to infer signaling
network topology (2, 11, 22, 23). A study by Mann and co-workers
illustrated the impressive analytical depth that has been achieved in
phosphoproteomics (22). Triple labeling SILAC (stable isotope
labeling by amino acids in cell culture (24)) was used to analyze
phosphorylation levels in growth factor-stimulated cells in the pres-
ence or absence of kinase inhibitors. Among thousands of phospho-
peptides, fewer than 10% were affected by the MAPK inhibitors
U0126 and SB202190. By contrast, almost 1,000 phosphopeptides
were affected by treatment of the leukemia cell line K562 with the
potent but unselective BCR-Abl inhibitor dasatinib. A similar
approach was applied to quantify changes in protein acetylation in
response to the deacetylase inhibitors SAHA and MS-275 (25).
In contrast to global approaches, recent developments in affinity-
based proteomics techniques have enabled to directly determine
protein binding profiles of small molecule drugs under close-to
physiological conditions. These techniques are in principle based
3. Targeted
Chemoproteomics
Approaches
22. 6 M. Bantscheff
on affinity chromatography, typically using immobilized drugs or
tool compounds (1, 8, 26) or covalent active site-labeling probes
(3, 6, 27).
Activity-based protein profiling (ABPP) via reactive probes
designed to specifically bind to active sites of target enzymes was
pioneered by the Cravatt laboratory. In this approach, the reactive
probe is typically fused to an affinity tag, such as biotin, via a spacer.
In a first step, the small molecule probe is incubated with the bio-
logical sample and allowed to covalently attach to proteins it has
affinity for. Subsequently, the formed probe–protein conjugates
are captured using the affinity tag. Probes have been developed for
a variety of enzyme classes including hydrolases (28), proteases
(29, 30), kinases (31, 32), phosphatases (33), histone deacetylases
(34), and glycosidases (35). For example, fluorescent activity-based
probes were reported that enable substrate-free identification of
inhibitors of uncharacterized enzymes. By using fluorescence
polarization as a read-out, the approach is compatible to high-
throughput screening with recombinant enzymes (36). A recent
report demonstrated how ABBP can be used to screen compound
libraries against an entire target class (37). The authors first synthe-
sized a probe to selectively capture 80% of mammalian serine
hydrolases, then screened 70 SHs against 140 structurally diverse
carbamates and assessed the selectivity of hits using the very same
approach. Patricelli and co-workers (31) used acyl phosphate-
containing nucleotides, prepared from a biotin derivative and ATP
or ADP to covalently modify ATP-binding proteins directly in cell
extracts. Activity-based probes can be adapted for in situ and in vivo
labeling by introducing a bio-orthogonal chemical handle, such as
an alkyne. Probe-labeled enzymes are then captured by click chem-
istry conjugation to azide-containing reporter tags (38–40).
Probe design is a crucial step when developing ABPP-based
assays and often requires detailed structural information to ensure
selective binding of the probe to the catalytic site of target enzymes
and to enable the efficient reaction of the active probe with suitable
amino acid residues within or close to the catalytic site. In order to
streamline probe development, a trifunctional probe design has been
suggested consisting of a reversibly interacting selectivity group,
a reactive group, and a sorting function (41). In this strategy, small
molecules generating selectivity for individual enzyme classes are
attached to a common building block containing reactive group and
sorting function (i.e., biotin) that is common to all probes. Recent
reports demonstrated examples for profiling of cAMP-binding pro-
teins (42), kinases (43), and methyltransferases (44).
For probes interacting with target enzymes with high affinity,
however, probe design can be drastically simplified by omitting the
reactive group and directly immobilizing the compound of interest
on a solid support. The thus generated probe matrix is then incu-
bated with cell extracts and mass spectrometry can be employed in
23. 7
1 Mass Spectrometry-Based Chemoproteomic Approaches
order to identify capture proteins (5, 45). Recent reports demonstrated
successful applications of this approach for enrichment of protein
kinases and characterization of binding profiles of kinase inhibitors
(13, 23, 46–51), proteins binding to ATP/ADP (52), phosphati-
dylinositols (53, 54), cyclic nucleotides (55, 56), histone deacety-
lases (57), and peptides (58–60). Further, in several recent
mode-of-action studies, immobilized analogs of lead compounds
enabled target identification (61–64). Several large-scale studies
have been performed for target class specific enrichment of kinase
inhibitors using both reactive probes targeting the ATP binding
pocket (31, 43) and immobilized unselective kinase inhibitors (46,
65) for affinity enrichment. The kinome coverage achieved with
kinase inhibitor probes compared quite favorably to results obtained
with reactive probes, indicating a limited impact of the bond-forming
reaction with kinases on target class coverage.
Qualitative binding profiles obtained in simple activity/affinity
enrichment studies give only limited information about binding
potencies of targets and off-targets detected. Consequently, the
pharmacological relevance of detected off-target protein has to be
validated using the standard repertoire of enzymatic assays (13, 47,
49, 50). While this might be feasible for very selective probes, cap-
turing only few proteins, with recent mass spectrometric equip-
ment often hundreds of proteins can be identified from a single
affinity enrichment experiment. Further, the analysis of complex
mass spectrometry data is impaired by the fact that the amounts of
individual proteins captured do not represent the affinities of these
proteins to the immobilized compound. Hence, additional evi-
dence is required to distinguish low-abundant high-affinity inter-
actors from low-affinity abundant ones, e.g., albumin and
hemoglobin are known to have low affinity for a range of small
molecules and many NADH/NADPH binding proteins bind to
immobilized ATP-mimetics (13, 47, 49–51). In addition, proteins
might bind to the resin or additional groups introduced to com-
pounds for probe generation, e.g., linkers, reactive groups, biotin,
etc. Some of these issues can be addressed by excluding those pro-
teins from further analysis that have been frequently observed in
independent experiments using different probe matrices (66). An
elegant way to increase over-all specificity of the experiment is to
design two probes/probe matrices, one containing the active com-
pound of interest and one containing an inactive analog (45).
Experiments with both matrices are then performed in parallel and
candidate target proteins can be shortlisted upon differential display
of results, e.g., using quantitative mass spectrometric methods.
4. Impact
of Experiment
Design and
Quantitative
Read-Out
for Target
Identification
24. 8 M. Bantscheff
However, inactive analogs are often not available and designing
two independent probes is quite laborious.
Utilizing reactive or immobilized analogs of a small molecule
compound in order to identify its targets is a rather indirect
approach and modifications introduced for probe generation might
alter potency and selectivity of the compound under investigation.
This limitation can be addressed by a competition-based experi-
ment design. In this approach, probe/probe matrix binding is per-
formed in the presence or absence of an excess of free, underivatized
compound. Experiments are performed in parallel and quantitative
mass spectrometry can be applied to identify those proteins for
which captured amounts are strongly reduced in the presence of
free compound as compared to experiments performed with vehi-
cle control. In a recent application of this approach, Borawski et al.
generated an affinity matrix consisting of a derivatized, bioactive
PI4KB inhibitor linked to Sepharose beads used to purify cellular
and/or viral proteins from a Huh7 HCV replicon cell lysate (61).
To determine and quantify specific binding, the experiment was
performed in a competition format by adding 10 μM PIK4B inhib-
itor or DMSO alone into the replicon cell lysate prior to affinity
purification. Bound proteins were eluted, digested with trypsin,
and labeled with isobaric mass tags for relative and absolute quan-
tification (iTRAQ, (67)) and combined prior to LC-MS/MS anal-
ysis. This enabled to quantify binding displacement in the presence
of free inhibitor relative to the vehicle. Several hundred proteins
were identified and quantified in this experiment but only the bind-
ing of class III PI4 kinases, PI4KA and PI4KB, was significantly
reduced in the presence of free inhibitor. A similar, SILAC-based
strategy was described by Ong et al. for the identification of targets
of kinase inhibitors and imunophilin binders (68).
Haystead and co-workers suggested a competition-based
approach to estimate relative potencies of target proteins. ATP was
linked to Sepharose-beads through the gamma phosphate group,
thus generating a probe matrix selective for ATP-binding proteins
including protein kinases as well as a variety of other proteins uti-
lizing purine co-factors (52). After enrichment of ATP-binding
proteins from cell extracts, the matrix was incubated with increas-
ing concentrations of a set of antimalarial compounds and target
proteins were eluted in a dose-dependent manner. In a variation of
this approach, Patricelli and co-workers (31) used acyl phosphate-
containing nucleotides, prepared from a biotin derivative and ATP
or ADP to covalently modify ATP-binding proteins in the co-factor
binding site. Biotinylated peptide fragments from labeled pro-
teomes were subsequently captured and identified by mass
spectrometry. Target kinases of kinase inhibitors were then deter-
mined by comparing MS signals of probe-captured kinases in the
absence and presence of excess free kinase inhibitors.
25. 9
1 Mass Spectrometry-Based Chemoproteomic Approaches
Stable isotope labeling-based quantitative mass spectrometry
techniques for precise and accurate relative quantification of proteins
(14, 16) have become an indispensable tool in chemical proteomic
experiments, since identification of target proteins and their bind-
ing affinities (IC50
s) largely depends on the ability to quantify differ-
ences between vehicle control samples and samples incubated with
different amounts of inhibitor. For example, iTRAQ labeling and
LC-MS/MS were combined with a mixed kinase inhibitor probe
matrix (“kinobeads”) for selectivity profiling of three inhibitors of
the tyrosine kinase ABL developed for the treatment of chronic
myeologeneous leukemia (CML); the phase II compound SKI-606
and the marketed drugs imatinib (Glivec) and dasatinib (46). In
this study, cells or cell extracts were treated with inhibitor com-
pounds at varying concentrations followed by incubation with the
mixed kinase inhibitor matrix. Kinase inhibitors blocked the ATP
binding pockets of target and off-target proteins as a function of
concentration and affinity and consequently caused reduced abun-
dance of these target proteins on the kinobeads matrix. Quantitative
mass spectrometric analyses revealed binding profiles comprising
500 proteins including ~150 kinases. While dasatinib and SKI-
606 revealed very broad target profiles (46 and 42 proteins, respec-
tively, showed 50% competition at 1 μM), imatinib was much
more selective. In addition to the primary imatinib targets ABL/
BCR-ABL, ARG two novel target candidates, the receptor tyrosine
kinase DDR1 (90 nM), and the quinone oxidoreductase NQO2
(43 nM) were identified and validated in independent studies (69,
70). Similarly, SILAC (24) has been employed in conjunction with
kinase inhibitor matrices in order to determine the target profile of
gefitinib (71). In a very recent report, such a chemoproteomics
approach was applied to the analysis of inhibitors binding to native
megadalton HDAC complexes. A total of 16 structurally diverse
HDAC inhibitors were characterized for their selectivity in target-
ing multiple HDAC complexes scaffolded by ELM-SANT domain
subunits, including a novel mitotic deacetylase complex (MiDAC).
Inhibitors clustered according to their target profiles, with stronger
binding of aminobenzamides to the HDAC NCoR complex than
to the HDAC Sin3 complex, thus suggesting that the selectivity of
HDAC inhibitors should be evaluated in the context of HDAC
complexes rather than purified catalytic subunits (57).
When cells are treated with an inhibitor, direct targets will be
revealed by their reduced binding to the affinity matrix, however,
e.g., protein kinases downstream of the respective target kinases
will display an altered phosphorylation state due to the reduced
signaling by the target kinase. For example, the case of imatinib-
treated K562 cells, RSK1, and RSK3 were prominent examples for
proteins exhibiting a significant downregulated phosphorylation
state (46, 72).
26. 10 M. Bantscheff
In conclusion, mass spectrometry-based chemoproteomics
approaches provide versatile tools to map direct and indirect tar-
gets of a compound in a single set of experiments. It is anticipated
that these approaches will prove valuable at various stages of drug
discovery including validation and selectivity assessment of screen-
ing hits and of molecules developed during lead optimization phase
as well as in translational studies.
References
1. Bantscheff, M., Scholten, A., and Heck, A. J.
(2009) Revealing promiscuous drug-target
interactions by chemical proteomics, Drug
Discov Today 14, 1021–1029.
2. Carlson, S. M., and White, F. M. (2010) Using
Small Molecules and Chemical Genetics To
Interrogate Signaling Networks, ACS Chem
Biol 6(1):75–85.
3. Cravatt, B. F., Wright, A. T., and Kozarich, J. W.
(2008) Activity-based protein profiling: from
enzyme chemistry to proteomic chemistry,
Annu Rev Biochem 77, 383–414.
4. Kruse, U., Bantscheff, M., Drewes, G., and
Hopf, C. (2008) Chemical and pathway pro-
teomics: powerful tools for oncology drug dis-
covery and personalized health care, Mol Cell
Proteomics 7, 1887–1901.
5. Lolli, G., Thaler, F., Valsasina, B., Roletto, F.,
Knapp, S., Uggeri, M., Bachi, A., Matafora, V.,
Storici, P., Stewart, A., Kalisz, H. M., and
Isacchi, A. (2003) Inhibitor affinity chroma-
tography: profiling the specific reactivity of the
proteome with immobilized molecules,
Proteomics 3, 1287–1298.
6. Sadaghiani, A. M., Verhelst, S. H., and Bogyo, M.
(2007) Tagging and detection strategies for
activity-based proteomics, Curr Opin Chem
Biol 11, 20–28.
7. Yamamoto, K., Yamazaki, A., Takeuchi, M.,
and Tanaka, A. (2006) A versatile method of
identifying specific binding proteins on affinity
resins, Anal Biochem 352, 15–23.
8. Rix, U., and Superti-Furga, G. (2009) Target
profiling of small molecules by chemical pro-
teomics, Nat Chem Biol 5, 616–624.
9. Cohen, A. A., Geva-Zatorsky, N., Eden, E.,
Frenkel-Morgenstern, M., Issaeva, I., Sigal, A.,
Milo, R., Cohen-Saidon, C., Liron, Y., Kam, Z.,
Cohen, L., Danon, T., Perzov, N., and Alon, U.
(2008) Dynamic proteomics of individual can-
cer cells in response to a drug, Science 322,
1511–1516.
10. Lee, A. Y., Paweletz, C. P., Pollock, R. M.,
Settlage, R. E., Cruz, J. C., Secrist, J. P., Miller,
T. A., Stanton, M. G., Kral, A. M., Ozerova, N. D.,
Meng, F., Yates, N. A., Richon, V., and
Hendrickson, R. C. (2008) Quantitative analy-
sis of histone deacetylase-1 selective histone
modifications by differential mass spectrometry,
J Proteome Res 7, 5177–5186.
11. Liang, X., Hajivandi, M., Veach, D., Wisniewski,
D., Clarkson, B., Resh, M. D., and Pope, R. M.
(2006) Quantification of change in phosphory-
lation of BCR-ABL kinase and its substrates in
response to Imatinib treatment in human
chronic myelogenous leukemia cells, Proteomics
6, 4554–4564.
12. Song, D., Chaerkady, R., Tan, A. C., Garcia-
Garcia, E., Nalli, A., Suarez-Gauthier, A.,
Lopez-Rios, F., Zhang, X. F., Solomon, A.,
Tong, J., Read, M., Fritz, C., Jimeno, A.,
Pandey, A., and Hidalgo, M. (2008) Antitumor
activity and molecular effects of the novel heat
shock protein 90 inhibitor, IPI-504, in pancre-
atic cancer, Mol Cancer Ther 7, 3275–3284.
13. Brehmer, D., Godl, K., Zech, B., Wissing, J.,
and Daub, H. (2004) Proteome-wide identifi-
cation of cellular targets affected by bisindolyl-
maleimide-type protein kinase C inhibitors, Mol
Cell Proteomics 3, 490–500.
14. Bantscheff, M., Schirle, M., Sweetman, G.,
Rick, J., and Kuster, B. (2007) Quantitative
mass spectrometry in proteomics: a critical
review, Anal Bioanal Chem 389, 1017–1031.
15. Domon, B., and Aebersold, R. (2010) Options
and considerations when selecting a quantita-
tive proteomics strategy, Nat Biotechnol 28,
710–721.
16. Mallick, P., and Kuster, B. (2010) Proteomics:
a pragmatic perspective, Nat Biotechnol 28,
695–709.
17. Schirle, M., Heurtier, M. A., and Kuster, B.
(2003) Profiling core proteomes of human cell
lines by one-dimensional PAGE and liquid
chromatography-tandem mass spectrometry,
Mol Cell Proteomics 2, 1297–1305.
18. Sui, J., Zhang, J., Tan, T. L., Ching, C. B., and
Chen, W. N. (2008) Comparative proteomics
analysis of vascular smooth muscle cells incu-
bated with S- and R-enantiomers of atenolol
27. 11
1 Mass Spectrometry-Based Chemoproteomic Approaches
using iTRAQ-coupled two-dimensional
LC-MS/MS, Mol Cell Proteomics 7,
1007–1018.
19. Zhang, J., Sui, J., Ching, C. B., and Chen, W. N.
(2008) Protein profile in neuroblastoma
cells incubated with S- and R-enantiomers of
ibuprofen by iTRAQ-coupled 2-D LC-MS/
MS analysis: possible action of induced proteins
on Alzheimer’s disease, Proteomics 8,
1595–1607.
20. Yamanaka, H., Yakabe, Y., Saito, K., Sekijima,
M., and Shirai, T. (2007) Quantitative pro-
teomic analysis of rat liver for carcinogenicity
prediction in a 28-day repeated dose study,
Proteomics 7, 781–795.
21. Mackeen, M. M., Kramer, H. B., Chang, K. H.,
Coleman, M. L., Hopkinson, R. J., Schofield,
C. J., and Kessler, B. M. (2010) Small-molecule-
based inhibition of histone demethylation in
cells assessed by quantitative mass spectrometry,
J Proteome Res 9, 4082–4092.
22. Pan, C., Olsen, J. V., Daub, H., and Mann, M.
(2009) Global effects of kinase inhibitors on
signaling networks revealed by quantitative
phosphoproteomics, Mol Cell Proteomics 8,
2796–2808.
23. Li, J., Rix, U., Fang, B., Bai, Y., Edwards, A.,
Colinge, J., Bennett, K. L., Gao, J., Song, L.,
Eschrich, S., Superti-Furga, G., Koomen, J., and
Haura, E. B. (2010) A chemical and phosphop-
roteomic characterization of dasatinib action in
lung cancer, Nat Chem Biol 6, 291–299.
24. Ong, S. E., Blagoev, B., Kratchmarova, I.,
Kristensen, D. B., Steen, H., Pandey, A., and
Mann, M. (2002) Stable isotope labeling by
amino acids in cell culture, SILAC, as a simple
and accurate approach to expression proteom-
ics, Mol Cell Proteomics 1, 376–386.
25. Choudhary, C., Kumar, C., Gnad, F., Nielsen,
M. L., Rehman, M., Walther, T. C., Olsen, J.
V., and Mann, M. (2009) Lysine acetylation
targets protein complexes and co-regulates
major cellular functions, Science 325,
834–840.
26. Hall, S. E. (2006) Chemoproteomics-driven
drug discovery: addressing high attrition rates,
Drug Discov Today 11, 495–502.
27. Nomura, D. K., Dix, M. M., and Cravatt, B. F.
(2010) Activity-based protein profiling for bio-
chemical pathway discovery in cancer, Nat Rev
Cancer 10, 630–638.
28. Liu, Y., Patricelli, M. P., and Cravatt, B. F.
(1999) Activity-based protein profiling: the ser-
ine hydrolases, Proc Natl Acad Sci USA 96,
14694–14699.
29. Kato, D., Boatright, K. M., Berger, A. B., Nazif,
T., Blum, G., Ryan, C., Chehade, K. A.,
Salvesen, G. S., and Bogyo, M. (2005) Activity-
based probes that target diverse cysteine pro-
tease families, Nat Chem Biol 1, 33–38.
30. Saghatelian, A., Jessani, N., Joseph, A.,
Humphrey, M., and Cravatt, B. F. (2004)
Activity-based probes for the proteomic profiling
of metalloproteases, Proc Natl Acad Sci USA
101, 10000–10005.
31. Patricelli, M. P., Szardenings, A. K., Liyanage,
M., Nomanbhoy, T. K., Wu, M., Weissig, H.,
Aban, A., Chun, D., Tanner, S., and Kozarich,
J. W. (2007) Functional interrogation of the
kinome using nucleotide acyl phosphates,
Biochemistry 46, 350–358.
32. Yee, M. C., Fas, S. C., Stohlmeyer, M. M.,
Wandless, T. J., and Cimprich, K. A. (2005)
A cell-permeable, activity-based probe for protein
and lipid kinases, J Biol Chem 280,
29053–29059.
33. Kumar, S., Zhou, B., Liang, F., Wang, W. Q.,
Huang, Z., and Zhang, Z. Y. (2004) Activity-
based probes for protein tyrosine phosphatases,
Proc Natl Acad Sci USA 101, 7943–7948.
34. Salisbury, C. M., and Cravatt, B. F. (2007)
Activity-based probes for proteomic profiling
of histone deacetylase complexes, Proc Natl
Acad Sci USA 104, 1171–1176.
35. Vocadlo, D. J., and Bertozzi, C. R. (2004)
A strategy for functional proteomic analysis of
glycosidase activity from cell lysates, Angew
Chem Int Ed Engl 43, 5338–5342.
36. Bachovchin, D. A., Brown, S. J., Rosen, H.,
and Cravatt, B. F. (2009) Identification of
selective inhibitors of uncharacterized enzymes
by high-throughput screening with fluorescent
activity-based probes, Nat Biotechnol 27,
387–394.
37. Bachovchin, D. A., Ji, T., Li, W., Simon, G. M.,
Blankman, J. L., Adibekian, A., Hoover, H.,
Niessen, S., and Cravatt, B. F. (2010)
Superfamily-wide portrait of serine hydrolase
inhibition achieved by library-versus-library
screening, Proc Natl Acad Sci USA 107,
20941–20946.
38. Wright, A. T., and Cravatt, B. F. (2007)
Chemical proteomic probes for profiling cyto-
chrome p450 activities and drug interactions
in vivo, Chem Biol 14, 1043–1051.
39. Speers, A. E., Adam, G. C., and Cravatt, B. F.
(2003) Activity-based protein profiling in vivo
using a copper(i)-catalyzed azide-alkyne [3+2]
cycloaddition, J Am Chem Soc 125, 4686–4687.
40. Speers, A. E., and Cravatt, B. F. (2004) Profiling
enzyme activities in vivo using click chemistry
methods, Chem Biol 11, 535–546.
41. Koster, H., Little, D. P., Luan, P., Muller, R.,
Siddiqi, S. M., Marappan, S., and Yip, P. (2007)
28. 12 M. Bantscheff
Capture compound mass spectrometry: a tech-
nology for the investigation of small molecule
protein interactions, Assay Drug Dev Technol 5,
381–390.
42. Luo, Y., Blex, C., Baessler, O., Glinski, M.,
Dreger, M., Sefkow, M., and Koster, H. (2009)
The cAMP capture compound mass spectrom-
etry as a novel tool for targeting cAMP-binding
proteins: from protein kinase A to potassium/
sodium hyperpolarization-activated cyclic
nucleotide-gated channels, Mol Cell Proteomics
8, 2843–2856.
43. Fischer, J. J., Graebner Baessler, O. Y., Dalhoff,
C., Michaelis, S., Schrey, A. K., Ungewiss, J.,
Andrich, K., Jeske, D., Kroll, F., Glinski, M.,
Sefkow, M., Dreger, M., and Koester, H.
(2010) Comprehensive identification of stauro-
sporine-binding kinases in the hepatocyte cell
line HepG2 using Capture Compound Mass
Spectrometry (CCMS), J Proteome Res 9,
806–817.
44. Dalhoff, C., Huben, M., Lenz, T., Poot, P.,
Nordhoff, E., Koster, H., and Weinhold, E.
(2010) Synthesis of S-adenosyl-L-homocysteine
capture compounds for selective photoinduced
isolation of methyltransferases, Chembiochem
11, 256–265.
45. Oda, Y., Owa, T., Sato, T., Boucher, B., Daniels,
S., Yamanaka, H., Shinohara, Y., Yokoi, A.,
Kuromitsu, J., and Nagasu, T. (2003)
Quantitative chemical proteomics for identify-
ing candidate drug targets, Anal Chem 75,
2159–2165.
46. Bantscheff, M., Eberhard, D., Abraham, Y.,
Bastuck, S., Boesche, M., Hobson, S.,
Mathieson, T., Perrin, J., Raida, M., Rau, C.,
Reader, V., Sweetman, G., Bauer, A.,
Bouwmeester, T., Hopf, C., Kruse, U.,
Neubauer, G., Ramsden, N., Rick, J., Kuster,
B., and Drewes, G. (2007) Quantitative chemi-
cal proteomics reveals mechanisms of action of
clinical ABL kinase inhibitors, Nat Biotechnol
25, 1035–1044.
47. Brehmer, D., Greff, Z., Godl, K., Blencke, S.,
Kurtenbach, A., Weber, M., Muller, S., Klebl,
B., Cotten, M., Keri, G., Wissing, J., and Daub,
H. (2005) Cellular targets of gefitinib, Cancer
Res 65, 379–382.
48. Daub, H., Olsen, J. V., Bairlein, M., Gnad, F.,
Oppermann, F. S., Korner, R., Greff, Z., Keri,
G., Stemmann, O., and Mann, M. (2008)
Kinase-selective enrichment enables quantita-
tive phosphoproteomics of the kinome across
the cell cycle, Mol Cell 31, 438–448.
49. Godl, K., Gruss, O. J., Eickhoff, J., Wissing, J.,
Blencke, S., Weber, M., Degen, H., Brehmer,
D., Orfi, L., Horvath, Z., Keri, G., Muller, S.,
Cotten, M., Ullrich, A., and Daub, H. (2005)
Proteomic characterization of the angiogenesis
inhibitor SU6668 reveals multiple impacts on
cellular kinase signaling, Cancer Res 65,
6919–6926.
50. Remsing Rix, L. L., Rix, U., Colinge, J.,
Hantschel, O., Bennett, K. L., Stranzl, T.,
Muller, A., Baumgartner, C., Valent, P.,
Augustin, M., Till, J. H., and Superti-Furga, G.
(2009) Global target profile of the kinase inhib-
itor bosutinib in primary chronic myeloid leu-
kemia cells, Leukemia 23, 477–485.
51. Rix, U., Hantschel, O., Durnberger, G.,
Remsing Rix, L. L., Planyavsky, M., Fernbach,
N. V., Kaupe, I., Bennett, K. L., Valent, P.,
Colinge, J., Kocher, T., and Superti-Furga, G.
(2007) Chemical proteomic profiles of the
BCR-ABL inhibitors imatinib, nilotinib, and
dasatinib reveal novel kinase and nonkinase tar-
gets, Blood 110, 4055–4063.
52. Graves, P. R., Kwiek, J. J., Fadden, P., Ray, R.,
Hardeman, K., Coley, A. M., Foley, M., and
Haystead, T. A. (2002) Discovery of novel tar-
gets of quinoline drugs in the human purine
binding proteome, Mol Pharmacol 62,
1364–1372.
53. Gharbi, S. I., Zvelebil, M. J., Shuttleworth, S.
J., Hancox, T., Saghir, N., Timms, J. F., and
Waterfield, M. D. (2007) Exploring the speci-
ficity of the PI3K family inhibitor LY294002,
Biochem J 404, 15–21.
54. Krugmann, S., Anderson, K. E., Ridley, S. H.,
Risso, N., McGregor, A., Coadwell, J.,
Davidson, K., Eguinoa, A., Ellson, C. D., Lipp, P.,
Manifava, M., Ktistakis, N., Painter, G.,
Thuring, J. W., Cooper, M. A., Lim, Z. Y.,
Holmes, A. B., Dove, S. K., Michell, R. H.,
Grewal, A., Nazarian, A., Erdjument-Bromage,
H., Tempst, P., Stephens, L. R., and Hawkins,
P. T. (2002) Identification of ARAP3, a novel
PI3K effector regulating both Arf and Rho
GTPases, by selective capture on phospho-
inositide affinity matrices, Mol Cell 9, 95–108.
55. Hanke, S. E., Bertinetti, D., Badel, A.,
Schweinsberg, S., Genieser, H. G., and Herberg,
F. W. (2010) Cyclic nucleotides as affinity tools:
phosphorothioate cAMP analogues address
specific PKA subproteomes, NatBiotechnol 28(4):
294–301.
56. Scholten, A., Poh, M. K., van Veen, T. A., van
Breukelen, B., Vos, M. A., and Heck, A. J.
(2006) Analysis of the cGMP/cAMP interac-
tome using a chemical proteomics approach in
mammalian heart tissue validates sphingosine
kinase type 1-interacting protein as a genuine
and highly abundant AKAP, J Proteome Res 5,
1435–1447.
57. Bantscheff, M., Hopf, C., Savitski, M. M.,
Dittmann, A., Grandi, P., Michon, A. M.,
Schlegl, J., Abraham, Y., Becher, I., Bergamini,
G., Boesche, M., Delling, M., Dumpelfeld, B.,
29. 13
1 Mass Spectrometry-Based Chemoproteomic Approaches
Eberhard, D., Huthmacher, C., Mathieson, T.,
Poeckel, D., Reader, V., Strunk, K., Sweetman,
G., Kruse, U., Neubauer, G., Ramsden, N. G.,
and Drewes, G. (2011) Chemoproteomics pro-
filing of HDAC inhibitors reveals selective tar-
geting of HDAC complexes, Nat Biotechnol 29,
255–265.
58. Hanke, S., and Mann, M. (2009) The phos-
photyrosine interactome of the insulin receptor
family and its substrates IRS-1 and IRS-2, Mol
Cell Proteomics 8, 519–534.
59. Schulze, W. X., Deng, L., and Mann, M. (2005)
Phosphotyrosine interactome of the ErbB-
receptor kinase family, Mol Syst Biol 1, 2005
0008.
60. Vermeulen, M., Eberl, H. C., Matarese, F.,
Marks, H., Denissov, S., Butter, F., Lee, K. K.,
Olsen, J. V., Hyman, A. A., Stunnenberg, H.
G., and Mann, M. (2010) Quantitative interac-
tion proteomics and genome-wide profiling of
epigenetic histone marks and their readers, Cell
142, 967–980.
61. Borawski, J., Troke, P., Puyang, X., Gibaja, V.,
Zhao, S., Mickanin, C., Leighton-Davies, J.,
Wilson, C. J., Myer, V., Cornellataracido, I.,
Baryza, J., Tallarico, J., Joberty, G., Bantscheff,
M., Schirle, M., Bouwmeester, T., Mathy, J. E.,
Lin, K., Compton, T., Labow, M., Wiedmann,
B., and Gaither, L. A. (2009) Class III phos-
phatidylinositol 4-kinase alpha and beta are
novel host factor regulators of hepatitis C virus
replication, J Virol 83, 10058–10074.
62. Fleischer, T. C., Murphy, B. R., Flick, J. S.,
Terry-Lorenzo, R. T., Gao, Z. H., Davis, T.,
McKinnon, R., Ostanin, K., Willardsen, J. A.,
and Boniface, J. J. (2010) Chemical proteomics
identifies Nampt as the target of CB30865, an
orphan cytotoxic compound, Chem Biol 17,
659–664.
63. Huang, S. M., Mishina, Y. M., Liu, S., Cheung,
A., Stegmeier, F., Michaud, G. A., Charlat, O.,
Wiellette, E., Zhang, Y., Wiessner, S., Hild, M.,
Shi, X., Wilson, C. J., Mickanin, C., Myer, V.,
Fazal, A., Tomlinson, R., Serluca, F., Shao, W.,
Cheng, H., Shultz, M., Rau, C., Schirle, M.,
Schlegl, J., Ghidelli, S., Fawell, S., Lu, C.,
Curtis, D., Kirschner, M. W., Lengauer, C.,
Finan, P. M., Tallarico, J. A., Bouwmeester, T.,
Porter, J. A., Bauer, A., and Cong, F. (2009)
Tankyrase inhibition stabilizes axin and antago-
nizes Wnt signalling, Nature 461, 614–620.
64. Raijmakers, R., Dadvar, P., Pelletier, S., Gouw, J.,
Rumpel, K., and Heck, A. J. (2010) Target
profiling of a small library of phosphodiesterase
5 (PDE5) inhibitors using chemical proteom-
ics, ChemMedChem 5, 1927–1936.
65. Oppermann, F. S., Gnad, F., Olsen, J. V.,
Hornberger, R., Greff, Z., Keri, G., Mann, M.,
and Daub, H. (2009) Large-scale proteomics
analysis of the human kinome, Mol Cell
Proteomics 8, 1751–1764.
66. Trinkle-Mulcahy, L., Boulon, S., Lam, Y. W.,
Urcia, R., Boisvert, F. M., Vandermoere, F.,
Morrice, N. A., Swift, S., Rothbauer, U.,
Leonhardt, H., and Lamond, A. (2008)
Identifying specific protein interaction partners
using quantitative mass spectrometry and bead
proteomes, J Cell Biol 183, 223–239.
67. Ross, P. L., Huang, Y. N., Marchese, J. N.,
Williamson, B., Parker, K., Hattan, S.,
Khainovski, N., Pillai, S., Dey, S., Daniels, S.,
Purkayastha, S., Juhasz, P., Martin, S., Bartlet-
Jones, M., He, F., Jacobson, A., and Pappin, D. J.
(2004) Multiplexed protein quantitation in
Saccharomyces cerevisiae using amine-reactive
isobaric tagging reagents, Mol Cell Proteomics
3, 1154–1169.
68. Ong, S. E., Schenone, M., Margolin, A. A., Li,
X., Do, K., Doud, M. K., Mani, D. R., Kuai, L.,
Wang, X., Wood, J. L., Tolliday, N. J., Koehler,
A. N., Marcaurelle, L. A., Golub, T. R., Gould,
R. J., Schreiber, S. L., and Carr, S. A. (2009)
Identifying the proteins to which small-molecule
probes and drugs bind in cells, Proc Natl Acad
Sci USA 106, 4617–4622.
69. Day, E., Waters, B., Spiegel, K., Alnadaf, T.,
Manley, P. W., Buchdunger, E., Walker, C., and
Jarai, G. (2008) Inhibition of collagen-induced
discoidin domain receptor 1 and 2 activation by
imatinib, nilotinib and dasatinib, Eur
J Pharmacol 599, 44–53.
70. Winger, J. A., Hantschel, O., Superti-Furga,
G., and Kuriyan, J. (2009) The structure of
the leukemia drug imatinib bound to human
quinone reductase 2 (NQO2), BMC Struct
Biol 9, 7.
71. Sharma, K., Weber, C., Bairlein, M., Greff, Z.,
Keri, G., Cox, J., Olsen, J. V., and Daub, H.
(2009) Proteomics strategy for quantitative
protein interaction profiling in cell extracts,
Nat Methods 6, 741–744.
72. Breitkopf, S. B., Oppermann, F. S., Keri, G.,
Grammel, M., and Daub, H. (2010) Proteomics
analysis of cellular imatinib targets and their
candidate downstream effectors, J Proteome Res 9,
6033–6043.
31. 16 G. Drewes
compound determined in this type of assays is often not predictive
for its pharmacodynamic efficacy. One reason for this discrepancy
is that an isolated recombinant protein, or protein fragment, does
not necessarily reflect the native conformation and activity of the
target in its physiological context, because of the lack of regulatory
domains, expression of alternative splice variants, interacting regu-
latory proteins, or incorrect protein folding or posttranslational
modifications. As a consequence, data generated in such assays may
not be predictive for the effects of a compound or drug in cell-
based or in vivo models. Ideally, assays should be developed to
generate data on native proteins in cell extracts or cell fractions,
under conditions carefully optimized to preserve protein integrity,
folding, posttranslational modification state, and interactions with
other proteins. Both activity-based and affinity-based chemical
proteomics techniques, as described in this volume, should com-
plement, or in some instances replace the traditional recombinant
protein-based assays.
In target-based drug discovery, a project begins with the nomina-
tion of a target. The target is typically defined as a protein which
should be
1. Tractable: Its biochemical activity can be modulated by the
desired therapeutic agent (e.g., a small molecule) in a dose-
dependent fashion.
2. Validated: It mediates a pathophysiological process such that
its modulation reverses a disease-relevant parameter, which can
be measured in disease-related cell-based or animal models,
and is expected to be predictive of human disease.
Targets are often referred to as “druggable” and “clinically
validated” when the modulation of the target was demonstrated to
lead to the desired clinical outcome. Historically almost all drug-
gable targets belong to a small number of target classes, biased
toward cell surface proteins (e.g., G protein-coupled receptors, ion
channels, or transporters) and a small number of intracellular
protein classes (e.g., nuclear receptors, metabolic enzymes, kinases,
or phosphodiesterases). A recent study estimated that the entirety
of approved small molecule drugs acts through approximately 200
human proteins as targets (5), obviously a small number when
compared to the 20,000–25,000 protein-coding genes in the
human genome (6). It has been estimated that ten times as many
suitable drug targets may exist, waiting to be discovered (7). In
fact there are numerous proteins in pathways with a strong disease
2. Chemical
Proteomics Can
Aid More Informed
Selection and
Validation of
Targets
32. 17
2 Chemical Proteomics in Drug Discovery
implication, e.g., based on pathobiochemical and human genetic
evidence, which are not tractable by current small molecule-based
approaches. Chemical proteomics approaches should serve to
expand the number of accessible drug targets by aiding the identi-
fication of tractable targets without the heavy bias toward the
traditional target classes. This type of “target deconvolution”
approaches was pioneered by the Schreiber laboratory in the clas-
sical studies which identified the molecular targets of immunosup-
pressants (8, 9). More recent exemplary approaches employed a
combination of screening of diverse compound libraries in cell-
based assays, which are not biased toward a particular family of
targets, with chemoproteomics-based target identification. Huang
et al. discovered the tankyrase proteins as tractable targets in the
Wnt signaling pathway, which plays a central role in colon cancer
but was characterized by a dearth of tractable drug targets (10).
Using a related strategy, Fleischer et al. found that the potent and
selective cytotoxic agent CB30865 exerts its effects by inhibition
of nicotinamide phosphoribosyltransferase, an enzyme in the NAD
biosynthetic pathway which helps cancer cells to sustain their
increased energy metabolism (11). In another recent study, cell-
based screening was performed for the upregulation of apolipopro-
tein AI production, and the proteomic profiling of hit compounds
led to the unexpected discovery of bromodomain proteins as
tractable targets for the modulation of the expression of apolipo-
protein AI and certain proinflammatory genes (12). These bro-
modomain inhibitors exhibit a novel mechanism of action by
blocking a protein–protein interaction formed between acetylated
histones and BET-family bromodomains, which were not previ-
ously regarded as tractable targets. These and other successful
studies support the notion that there is a general need for small
molecules as research tools to study protein function, particularly
for proteins which are not classical drug targets. Both the Structural
Genomics Consortium (http:/
/www.thesgc.org) and the Center
for Protein Research (http:/
/www.cpr.ku.dk) have recently initi-
ated extensive programs for the development of chemical probes
which will be made available to the scientific community.
Many drug discovery assays rely on the ability to express and purify
the target protein in active form in the substantial amounts – typically
milligrams of pure protein – necessary for the screening of com-
pound libraries. The drug industry has encountered many so-called
“difficult” target classes where this is not easily achieved, for
instance, because the target protein is very large or requires addi-
tional factors like interacting proteins for proper activity. Therefore,
3. Chemical
Proteomics-Based
Screening of
Compound
Libraries
33. 18 G. Drewes
methods based on immobilized probe compounds to capture the
target directly from a cell or tissue extract without further purifica-
tion can represent a viable alternative strategy. This approach was
used by Fadden et al. who captured purine-binding proteins from
porcine tissue with ATP-derivatized Sepharose and performed
affinity elutions with 5,000 different compounds, resulting in the
identification of 463 small molecule compounds eluting a total of
77 distinct proteins. Among these, novel and structurally diverse
inhibitors of the cancer target Hsp90 were identified, which were
further optimized to enter clinical development (13). A different
strategy was used by Bantscheff et al. who screened a compound
library for histone deacetylase (HDAC) inhibitors in a human cell
line extract, using an immobilized hydroxamate-based probe.
Here, compounds were added directly to the cell extract rather
than using them for elution, such that each compound was assayed
for the inhibition of the binding of HDACs to the immobilized
probe (14). An important feature of both approaches is that the
entire complement of proteins binding selectively to the immobi-
lized probe is screened simultaneously. This represents a major
advantage over traditional screening approaches, in particular, for
target classes with a substantial number of structurally related tar-
gets, like protein kinases or deacetylases, because possible “off-
targets” (undesired additional proteins, which typically share a
related active site with the target) are revealed early in the project.
In conventional approaches, one is left to resort to educated guesses
regarding possible “off-targets,” and distinct assays have to be con-
figured for each individual protein.
Despite the fact that drugs are usually optimized against a single
target, many compounds exhibit polypharmacology, i.e., they act
on multiple targets. These “off-targets” can increase the therapeu-
tic potential of a drug, but they might also cause toxic side effects,
which represent a major reason why drugs fail in clinical develop-
ment (15). An important recent example was the chemoproteomics-
based identification of cereblon (CRBN) as a target of the drug
thalidomide which mediates the drug’s teratogenic effects (16).
However, for oncology drugs, polypharmacology is the rule rather
than the exception, as they often target proteins from large target
classes with a high degree of structural conservation around the
active site, like protein and lipid kinases, HDACs, or heat shock
proteins. Compared to a truly selective drug, such a spectrum of
targets is more likely to produce toxic side effects, but in oncology
the increase in therapeutic potential may outweigh this disadvan-
tage (17). Conventional strategies typically rely on assay panels
comprising 10–100 purified enzymes to address compound
4. Chemical
Proteomics
for Drug Target
Profiling
34. 19
2 Chemical Proteomics in Drug Discovery
potency, selectivity, and potential off-target liabilities (18). The
recent progress in affinity-based proteomic techniques has enabled
the direct determination of protein-binding profiles of small mol-
ecule drugs under close to physiological conditions. These tech-
niques utilize immobilized compounds as noncovalent affinity baits
(14, 19–22) or covalent active-site labeling probes (23, 24). The
affinity probes are designed to selectively enrich a larger set of up
to several hundreds of proteins defined by structurally related active
sites, which can be viewed as chemically tractable subproteomes
(25). Noncovalent probes are used either immobilized to an affin-
ity matrix like sepharose or conjugated to biotin, and have been
used successfully for purine-binding proteins (26), protein kinases
including transmembrane receptor kinases (21, 22), lipid kinases
(27, 28), phosphodiesterases (29), and HDACs (14). Covalent
active-site labeling probes are typically biotin conjugates and have
been applied to kinases (30), GTPases (31), methylases (32), dehy-
drogenases (33), serine-, cysteine-, metallo-, and proteasomal pro-
teases (23, 34, 35), and HDACs (36). These methodologies
typically generate protein affinity profiles for the immobilized com-
pounds, which may reveal novel target candidates, but precautions
must be taken to avoid false positives due to the background prob-
lems caused by nonspecific interactions with abundant proteins.
Moreover, for the application to drug discovery, e.g., in screening
or affinity/selectivity profiling assays, the generation of robust
quantitative data for hit and lead compounds is an absolute neces-
sity. These problems can be managed if the affinity capture proto-
cols are formatted as quantitative competition-binding assays. This
can be achieved by adding the compound of interest in its free
form in the tissue extract, before or together with the affinity
matrix or the active site label, such that the free compound binds
to its targets in the lysate, thereby effectively competing with the
capturing probe. By assaying the free compound in the cell extract
over a range of concentrations, dose–response binding curves are
generated for as many proteins as can be captured by the probe
compound and robustly quantified. In case of the “Kinobeads”
matrix for protein kinases and the hydroxamate matrix for HDACs
developed by Bantscheff et al., more than 1,000 proteins were
found to bind to the matrix and were routinely quantified in drug-
profiling experiments using a competition binding assay format
coupled to protein quantification by isobaric tagging and high-
resolution LC-MS/MS peptide sequencing (14, 22). For a more
detailed discussion of qualitative and quantitative small molecule
target profiling, the reader is referred to recent comprehensive
reviews (20, 23, 37). Finally, in addition to the in vitro applications
described above, many chemical proteomics strategies can poten-
tially be adapted to the identification and activity profiling of tar-
gets in living cells and in animal models (38).
In conclusion, the recent advances in chemical proteomics and
in analytical instrumentation have promoted new drug discovery
35. 20 G. Drewes
strategies based on assays with increased content and better
appreciation of the molecular context of the targets. These meth-
odologies are providing complementary approaches to drug screen-
ing, drug target identification, and selectivity profiling, and have
the potential to substantially contribute to in vivo studies and clini-
cal studies of drug–target interactions.
References
1. Brown, D. (2007) Unfinished business: target-
based drug discovery. Drug Discov. Today 12,
1007–1012.
2. Kola, I., and Landis, J. (2004) Can the phar-
maceutical industry reduce attrition rates? Nat.
Rev. Drug Discov. 3, 711–715.
3. Fishman, M. C., and Porter, J. A. (2005)
Pharmaceuticals: a new grammar for drug
discovery. Nature 437, 491–493.
4. Hall, S. E. (2006) Chemoproteomics-driven
drug discovery: addressing high attrition rates.
Drug Discov. Today 11, 495–502.
5. Overington, J. P., Al-Lazikani, B., and Hopkins,
A. L. (2006) How many drug targets are there?
Nat. Rev. Drug Discov. 5, 993–996.
6. Clamp, M., Fry, B., Kamal, M., Xie, X., Cuff,
J., Lin, M. F., Kellis, M., Lindblad-Toh, K.,
and Lander, E. S. (2007) Distinguishing
protein-coding and noncoding genes in the
human genome. Proc. Natl. Acad. Sci. USA
104, 19428–19433.
7. Hopkins, A. L., and Groom, C. R. (2002) The
druggable genome. Nat. Rev. Drug Discov. 1,
727–730.
8. Brown, E. J., Albers, M. W., Shin, T. B.,
Ichikawa, K., Keith, C. T., Lane, W. S., and
Schreiber, S. L. (1994) A mammalian protein
targeted by G1-arresting rapamycin-receptor
complex. Nature 369, 756–758.
9. Harding, M. W., Galat, A., Uehling, D. E., and
Schreiber, S. L. (1989) A receptor for the
immunosuppressant FK506 is a cis-trans pepti-
dyl-prolyl isomerase. Nature 341, 758–760.
10. Huang, S. M., Mishina, Y. M., Liu, S., Cheung,
A., Stegmeier, F., Michaud, G. A., Charlat,
O., Wiellette, E., Zhang, Y., Wiessner, S.,
Hild, M., Shi, X., Wilson, C. J., Mickanin, C.,
Myer, V., Fazal, A., Tomlinson, R., Serluca, F.,
Shao, W., Cheng, H., Shultz, M., Rau, C.,
Schirle, M., Schlegl, J., Ghidelli, S., Fawell, S.,
Lu, C., Curtis, D., Kirschner, M. W., Lengauer,
C., Finan, P. M., Tallarico, J. A., Bouwmeester,
T., Porter, J. A., Bauer, A., and Cong, F.
(2009) Tankyrase inhibition stabilizes axin
and antagonizes Wnt signalling. Nature 461,
614–620.
11. Fleischer, T. C., Murphy, B. R., Flick, J. S.,
Terry-Lorenzo, R. T., Gao, Z. H., Davis, T.,
McKinnon, R., Ostanin, K., Willardsen, J. A.,
and Boniface, J. J. (2010) Chemical proteom-
ics identifies Nampt as the target of CB30865,
an orphan cytotoxic compound. Chem. Biol.
17, 659–664.
12. Nicodeme, E., Jeffrey, K. L., Schaefer, U.,
Beinke,S.,Dewell,S.,Chung,C.W.,Chandwani,
R., Marazzi, I., Wilson, P., Coste, H., White, J.,
Kirilovsky, J., Rice, C. M., Lora, J. M., Prinjha,
R. K., Lee, K., and Tarakhovsky, A. (2010)
Suppression of inflammation by a synthetic his-
tone mimic. Nature 468, 1119–1123.
13. Fadden, P., Huang, K. H., Veal, J. M., Steed, P. M.,
Barabasz, A. F., Foley, B., Hu, M., Partridge, J. M.,
Rice, J., Scott, A., Dubois, L. G., Freed, T. A.,
Silinski, M. A., Barta, T. E., Hughes, P. F.,
Ommen,A.,Ma,W.,Smith,E.D.,Spangenberg,
A. W., Eaves, J., Hanson, G. J., Hinkley, L.,
Jenks, M., Lewis, M., Otto, J., Pronk, G. J.,
Verleysen, K., Haystead, T. A., and Hall, S. E.
(2010) Application of chemoproteomics to
drug discovery: identification of a clinical can-
didate targeting hsp90. Chem. Biol. 17,
686–694.
14. Bantscheff, M., Hopf, C., Savitzki, M. M.,
Dittmann, A., Grandi, P., Michon, A. M. M.,
Schlegl, J., Abraham, Y., Becher, I., Bergamini,
G., Boesche, M., Delling, M., Dümpelfeld, B.,
Eberhard, D., Huthmacher, C., Mathieson, T.,
Poeckel, D., Strunk, K., Sweetman, G., Kruse,
U., Neubauer, G., Ramsden, N., and Drewes, G.
(2011) Chemoproteomics profiling of HDAC
inhibitors reveals selective targeting of HDAC
complexes. Nat. Biotechnol. 29, 255–265.
15. Morphy, R., Kay, C., and Rankovic, Z. (2004)
From magic bullets to designed multiple
ligands. Drug Discov. Today 9, 641–651.
16. Kruse, U., Bantscheff, M., Drewes, G., and
Hopf, C. (2008) Chemical and pathway
proteomics: powerful tools for oncology drug
discovery and personalized health care. Mol.
Cell Proteomics. 7, 1887–1901.
17. Ito, T., Ando, H., Suzuki, T., Ogura, T.,
Hotta, K., Imamura, Y., Yamaguchi, Y., and
Handa H. (2010) Identification of a primary
36. 21
2 Chemical Proteomics in Drug Discovery
target of thalidomide teratogenicity. Science.
327, 1345–1350.
18. Fliri, A. F., Loging, W. T., Thadeio, P. F., and
Volkmann, R. A. (2005) Analysis of drug-
induced effect patterns to link structure and
side effects of medicines. Nat. Chem. Biol. 1,
389–397.
19. Ong, S. E., Schenone, M., Margolin, A. A., Li,
X., Do, K., Doud, M. K., Mani, D. R., Kuai, L.,
Wang, X., Wood, J. L., Tolliday, N. J., Koehler,
A. N., Marcaurelle, L. A., Golub, T. R., Gould,
R. J., Schreiber, S. L., and Carr, S. A. (2009)
Identifying the proteins to which small-mole-
cule probes and drugs bind in cells. Proc. Natl.
Acad. Sci. USA 106, 4617–4622.
20. Rix, U., and Superti-Furga, G. (2009) Target
profiling of small molecules by chemical pro-
teomics. Nat. Chem. Biol. 5, 616–624.
21. Sharma, K., Weber, C., Bairlein, M., Greff, Z.,
Keri, G., Cox, J., Olsen, J. V., and Daub, H.
(2009) Proteomics strategy for quantitative
protein interaction profiling in cell extracts.
Nat. Methods 6, 741–744.
22. Bantscheff, M., Eberhard, D., Abraham, Y.,
Bastuck, S., Boesche, M., Hobson, S.,
Mathieson, T., Perrin, J., Raida, M., Rau, C.,
Reader, V., Sweetman, G., Bauer, A.,
Bouwmeester, T., Hopf, C., Kruse, U.,
Neubauer, G., Ramsden, N., Rick, J., Kuster,
B., and Drewes, G. (2007) Quantitative chemi-
cal proteomics reveals mechanisms of action of
clinical ABL kinase inhibitors. Nat. Biotechnol.
25, 1035–1044.
23. Cravatt, B. F., Wright, A. T., and Kozarich, J.
W. (2008) Activity-based protein profiling:
from enzyme chemistry to proteomic chemis-
try. Annu. Rev. Biochem. 77, 383–414.
24. Paulick, M. G., and Bogyo, M. (2008)
Application of activity-based probes to the
study of enzymes involved in cancer progres-
sion. Curr. Opin. Genet. Dev. 18, 97–106.
25. Haystead, T. A. (2006) The purinome, a com-
plex mix of drug and toxicity targets. Curr. Top.
Med. Chem. 6, 1117–1127.
26. Graves, P. R., Kwiek, J. J., Fadden, P., Ray, R.,
Hardeman, K., Coley, A. M., Foley, M., and
Haystead, T. A. (2002) Discovery of novel targets
of quinoline drugs in the human purine binding
proteome. Mol. Pharmacol. 62, 1364–1372.
27. Borawski, J., Troke, P., Puyang, X., Gibaja, V.,
Zhao, S., Mickanin, C., Leighton-Davies, J.,
Wilson, C. J., Myer, V., Cornellataracido, I.,
Baryza, J., Tallarico, J., Joberty, G., Bantscheff,
M., Schirle, M., Bouwmeester, T., Mathy, J. E.,
Lin, K., Compton, T., Labow, M., Wiedmann,
B., and Gaither, L. A. (2009) Class III phos-
phatidylinositol 4-kinase alpha and beta are
novel host factor regulators of hepatitis C virus
replication. J. Virol. 83, 10058–10074.
28. Gharbi, S. I., Zvelebil, M. J., Shuttleworth, S.
J., Hancox, T., Saghir, N., Timms, J. F., and
Waterfield, M. D. (2007) Exploring the
specificity of the PI3K family inhibitor
LY294002. Biochem. J. 404, 15–21.
29. Raijmakers, R., Dadvar, P., Pelletier, S., Gouw,
J., Rumpel, K., and Heck, A. J. (2010) Target
profiling of a small library of phosphodiesterase
5 (PDE5) inhibitors using chemical proteom-
ics. ChemMedChem. 5, 1927–1936.
30. Patricelli, M. P., Szardenings, A. K., Liyanage,
M., Nomanbhoy, T. K., Wu, M., Weissig, H.,
Aban, A., Chun, D., Tanner, S., and Kozarich,
J. W. (2007) Functional interrogation of the
kinome using nucleotide acyl phosphates.
Biochemistry 46, 350–358.
31. Luo, Y., Fischer, J. J., Baessler, O. Y., Schrey, A.
K., Ungewiss, J., Glinski, M., Sefkow, M.,
Dreger, M., and Koester, H. (2010) GDP-
capture compound – a novel tool for the profil-
ing of GTPases in pro- and eukaryotes by
capturecompoundmassspectrometry(CCMS).
J. Proteomics. 73, 815–819.
32. Dalhoff, C., Huben, M., Lenz, T., Poot, P.,
Nordhoff, E., Koster, H., and Weinhold, E. (2010)
Synthesis of S-adenosyl-L-homocysteine capture
compounds for selective photoinduced isolation of
methyltransferases. Chembiochem. 11, 256–265.
33. Ge, X., Wakim, B., and Sem, D. S. (2008)
Chemical proteomics-based drug design: target
and antitarget fishing with a catechol-rhoda-
nine privileged scaffold for NAD(P)(H) bind-
ing proteins. J. Med. Chem. 51, 4571–4580.
34. Kato, D., Boatright, K. M., Berger, A. B.,
Nazif, T., Blum, G., Ryan, C., Chehade, K. A.,
Salvesen, G. S., and Bogyo, M. (2005) Activity-
based probes that target diverse cysteine pro-
tease families. Nat. Chem. Biol. 1, 33–38.
35. Berkers, C. R., Verdoes, M., Lichtman, E.,
Fiebiger, E., Kessler, B. M., Anderson, K. C.,
Ploegh, H. L., Ovaa, H., and Galardy, P. J.
(2005) Activity probe for in vivo profiling of
the specificity of proteasome inhibitor borte-
zomib. Nat. Methods 2, 357–362.
36. Salisbury, C. M., and Cravatt, B. F. (2007)
Activity-based probes for proteomic profiling
of histone deacetylase complexes. Proc. Natl.
Acad. Sci. USA 104, 1171–1176.
37. Nomura, D. K., Dix, M. M., and Cravatt, B. F.
(2010) Activity-based protein profiling for bio-
chemical pathway discovery in cancer. Nat.
Rev. Cancer 10, 630–638.
38. Edgington, L. E., Berger, A. B., Blum, G.,
Albrow, V. E., Paulick, M. G., Lineberry, N.,
and Bogyo, M. (2009) Noninvasive optical
imaging of apoptosis by caspase-targeted
activity-based probes. Nat. Med. 15,
967–973.
39. 26 U. Rix et al.
characterization of its molecular mechanism of action (1, 2). This
approach requires the immobilization of the compound of interest
on solid support as an early step. The strategy for immobilization
has to be designed very carefully as chemical modification of any
bioactive compound harbors the risk of impairing or abrogating its
biological activity through disruption of the protein interaction
interface. Therefore, one should utilize all available structure–
activity relationship information, which can, for instance, be
obtained by activity studies with structural analogs or from X-ray
co-crystal structures. If such information is unavailable, it is recom-
mended to immobilize the bait compound in separate experiments
via two or more different linker attachment points. In some cases,
when a cellular or biochemical readout is at hand, these analogs
can be tested for retention of activity. When employing cellular
assays, though, it has to be kept in mind that modifying a compound
might also alter its cell permeability properties. In addition, some
molecules are sensitive to various commonly employed conditions,
such as acid, base, or heat. Therefore, any broadly applicable
modification or immobilization reaction has to be efficient and
mild. This is the case for several different reactions employing, e.g.,
epoxide or amide chemistry (3). The formation of an amide bond
from an activated N-hydroxy-succinimide (NHS) ester and the
amino group (primary or secondary) of a bioactive compound
(analog) is readily achieved at room temperature and the reaction
usually comes to completion within several hours (4). Compounds
containing hydroxy- or carboxy-groups can be conveniently
converted into amines by Steglich esterification with Boc- or
Fmoc-protected amino acids or mono-protected ethylenediamine,
respectively, and subsequent deprotection (5). The choice of
protecting group should be based on chemical stability of the bait
compound under deprotection conditions (6). Other important
considerations for drug-affinity chromatography are the choice of
buffer and pH. Just as critical is the efficient inhibition of cellular
proteases, which would destroy proteins and prevent affinity enrich-
ment, and possibly enzymes that carry out unscheduled posttrans-
lational modifications in vitro (e.g., protein dephosphorylation), as
well as the choice and concentration of detergent, which aids in the
solubilization of proteins. In the following, we present the detailed
workflow for immobilization and affinity chromatography, which
we have previously applied to the characterization of several clinical
kinase inhibitors by chemical proteomics (7). We exemplify the
approach with a study on the BCR-ABL tyrosine kinase inhibitor
dasatinib (BMS-354825, trade name Sprycel) (8), a drug in clini-
cal use for chronic myeloid leukemia (CML) and several other
malignancies. In this example, drug-affinity chromatography is
performed using the CML cell line KU812 (9).
40. 27
3 Compound Immobilization and Drug-Affinity Chromatography
1. NHS-activated Sepharose 4 Fast Flow (GE Healthcare) (see
Note 1).
2. Dimethyl sulfoxide, absolute over molecular sieve (DMSO,
Fluka).
3. Isopropanol, absolute over molecular sieve (iProp, Fluka).
4. Triethylamine (TEA, Sigma).
5. Ethanolamine (Aldrich).
6. 2 mL microcentrifuge tubes (Eppendorf).
1. Methanol (MeOH), LC-MS grade (Fisher Scientific GmbH).
2. Isopropanol (iProp), LC-MS grade (Fisher Scientific GmbH).
3. Water, LiChrosolv grade (Merck).
4. Formic acid.
5. Chromatography column: SunFire C18
, 5 μm, 3.0×50 mm
(Waters).
6. HPLC 2795 (Waters).
7. Photo diode array detector 2996 (Waters).
8. ZQ 2000 single quadrupole mass spectrometer (Waters/
Micromass).
1. Dulbecco’s modified Eagle’s medium (DMEM) (Gibco) sup-
plemented with 10% fetal bovine serum (Gibco) and penicillin/
streptomycin 100× (PAA Laboratories GmbH).
2. Cell culture dishes, Ø 150 mm (VWR International).
3. 4× Concentrated Laemmli sample buffer containing 10%
β-mercaptoethanol.
4. Lysis buffer 1×: 50 mM Tris–HCl, 100 mM NaCl, 0.2%
NP-40, 5% glycerol, 1.5 mM MgCl2
, 25 mM NaF, 1 mM
Na3
VO4
, 1 mM phenylmethylsulfonyl fluoride, 1 mM dithio-
threitol (DTT), 10 μg/mL TLCK, 1 μg/mL leupeptin, 1 μg/
mL aprotinin, and 10 μg/mL soybean trypsin inhibitor
(Sigma), pH 7.5 (see Note 2).
5. γ-Globin (1 μg/μL) (Bio-Rad).
6. Protein Assay Dye Reagent Concentrate (5×) (Bio-Rad).
7. Hypodermic Needle, 20 G×1½ (Terumo®
), single-use syringe
10 mL (B. Braun Melsungen AG).
8. Cuvettes half-micro, 2.5 mL, PMMA (VWR International).
9. Polycarbonate ultracentrifuge tubes (Beckman Coulter).
10. Ultracentrifuge and fixed-angle rotor (Beckman Coulter).
2. Materials
2.1. Coupling to Solid
Support
2.2. LC-MS Analysis
of the Coupling
Reaction
2.3. Cell Culture,
Harvest, Lysis,
and Protein
Quantification
41. 28 U. Rix et al.
1. Lysis buffer 1× (see above).
2. 4× Concentrated Laemmli sample buffer (containing 10%
β-mercaptoethanol) (see above).
3. Polycarbonate ultracentrifuge tubes (Beckman Coulter).
4. Spin columns Mobicol (MoBiTec GmbH).
5. Spin column lower filters – 90 μm pore size (MoBiTec
GmbH).
6. 1.5 and 2 mL microcentrifuge tubes (Eppendorf).
1. Iodoacetamide (13 μg/μL) (Sigma).
2. Mini-PROTEAN Tetra electrophoresis system (Bio-Rad
Laboratories).
3. 4× Running gel buffer (SDS buffer): 1.5 mM Tris–HCl and
0.54% SDS, pH 8.8.
4. 4× Gel buffer 2: 2 M Tris, 1.6 mM SDS, pH 6.8.
5. Ammoniumperoxide sulfate (APS), acrylamide, tetramethyl-
ethylenediamine (TEMED, Sigma).
6. 5× Running buffer: 602 g Tris, 2,880 g glycine, 200 g SDS,
20 L water, pH 8.3. Do not use HCl!
7. Broad range 7–175 kDa prestained protein marker (New
England Biolabs).
1. Fixing buffer: 40% ethanol, 10% acetic acid.
2. Washing buffer: 35% ethanol or distilled water.
3. Sensitizing buffer: 0.02% Na2
S2
O3
.
4. 0.2% AgNO3
solution (refrigerated).
5. Developing buffer: 3% Na2
CO3
, 0.05% formaldehyde.
6. Quenching solution: 5% acetic acid.
1. 10× Western blot buffer: for 2 L dissolve 60.7 g Tris and
288.4 g glycine in distilled water.
2. 1× Western blot buffer: 10% (v/v) 10× Western blot buffer,
10% (v/v) methanol.
3. Wash buffer: PBS with 1% Tween-20 (PBS-T).
4. Blocking buffer (in this example, see Note 3): 3% bovine serum
albumin (BSA) in PBS-T.
5. Primary antibody dilution (in this example): 1:2,000 mono-
clonal mouse anti-ABL (21–63) (Santa Cruz Biotechnology)
in 3% BSA/PBS-T (see Note 4).
6. Secondary antibody dilution (in this example): 1:2,000 horse
radish peroxidase (HRP)-conjugated anti-mouse (Rockland
Immunochemicals) in PBS-T.
2.4. Affinity Pulldown
2.5. One-Dimensional
SDS-PAGE
2.6. Silver Staining
2.7. Immunoblot
42. 29
3 Compound Immobilization and Drug-Affinity Chromatography
7. Amersham ECL Plus™ Western Blotting Detection Reagents
(GE Healthcare).
8. Nitrocellulose transfer membrane (Protran BA 85,
300 mm×3 m, 0.45 μm).
9. Saran wrap.
10. Amersham Hyperfilm ECL (GE Healthcare).
11. Hoefer Semi-Phor semi-dry transfer unit (Hoefer, Inc.).
This section is illustrated in Fig. 1.
1. The beads are provided in a slurry of approximately 50% iso-
propanol. For one pull down, pipette 100 μL of this slurry into
a 1.5 mL microcentrifuge tube (see Note 5). Compare the
settled bed volume with 50 μL DMSO. Add or remove slurry
until 50 μL bed volume is reached (see Note 6).
3. Methods
3.1. Coupling to Solid
Support
Fig. 1. Immobilization strategy for the BCR-ABL kinase inhibitor dasatinib. The “couple-
able” analog c-dasatinib was designed based on the X-ray co-crystal structure of ABL
with dasatinib, which indicated that the hydroxy group extends into the solvent space (10).
The dasatinib affinity matrix is generated via coupling of c-dasatinib to NHS-activated
sepharose.
43. 30 U. Rix et al.
2. Centrifuge the beads for 3 min at room temperature with
75×g and remove the supernatant.
3. Add 50 μL of DMSO, resuspend gently by inverting several
times, centrifuge (as before), and discard the supernatant.
Repeat this washing step three times with 0.5 mL DMSO.
Then resuspend beads in 50 μL DMSO.
4. Dissolve bait compound to a final concentration of 10 mM in
DMSO and add 5 μL of this stock solution and 0.75 μL TEA
to the 50% bead slurry, mix carefully and incubate on roto-
shaker for 16–24 h (but at least for 8 h) at room temperature
with 10 rpm (see Note 7).
5. Centrifuge the beads (as before) and remove 10 μL (»5 nmol)
of the supernatant for the LC-MS control. This is the T16
time
point for the coupling control.
6. If the drug was successfully coupled to the beads, block the
unreacted NHS-ester groups by adding 25 μL ethanolamine
and incubate on roto-shaker for further 16–24 h (but at least
for 8 h) at room temperature with 10 rpm.
7. Centrifuge beads, remove the supernatant, wash twice with
0.5 mL of DMSO, and discard the supernatant again.
8. Either proceed directly with the drug pulldown and wash the
beads with Lysis buffer or add 50 μL isopropanol, centrifuge,
discard the supernatant, and repeat this washing step twice
with 0.5 mL isopropanol each. The beads can be stored in iso-
propanol at 4°C (away from light) (see Note 8).
This section is illustrated in Fig. 2.
1. Remove 0.5 μL of a 10 mM bait compound stock solution and
add 9.5 μL DMSO. This is the T0
time point. Add 10 μL
MeOH to each sample (T0
and T16
).
2. Inject 5 μL of the samples beginning with the T16
time point to
avoid carryover.
3. The applied solvent system and LC conditions were:
(a) Solvent A=0.1% formic acid in H2
O
(b) Solvent B=80%MeOH/20% iProp
(c) Flow rate=0.5 mL/min
(d) 0–6 min 90% A/10% B to 100% B (linear gradient, curve 6)
(e) 6–8 min 100% B
(f) 8–8.5 min 100% B to 90% A and 10% B (linear gradient,
curve 6)
(g) 8.5–11 min 90% A and 10% B
3.2. LC-MS Analysis
of the Coupling
Reaction
44. 31
3 Compound Immobilization and Drug-Affinity Chromatography
4. The column temp was 40°C.
5. The reaction was analyzed on a photodiode array detector (PDA)
scanning a range of wavelengths between 210 and 500 nm.
6. The MS was set to scan a mass/charge ratio from 100 to 1,000,
switching continuously between the positive and negative elec-
trospray ionization (ESI) mode.
7. The MS parameters were:
(a) Capillary voltage: 3.2 (ESI+/−)
(b) Cone voltage: 32 (ESI+), 30 (ESI−)
(c) Extractor voltage: 3 (ESI+), 4 (ESI−)
(d) RF lens voltage: 0.3 (ESI+), 1.2 (ESI−)
(e) Source temperature: 150 (ESI+/−)
(f) Desolvation temperature: 400 (ESI+/−)
(g) Desolvation gas (N2
): 450 (ESI+/−)
(h) Cone gas (N2
): 50 (ESI+/−)
(i) HM and LM resolution: 15 (ESI+/−)
(j) Ion energy: 0.6 (ESI+) and 0.1 (ESI−)
Fig. 2. LC-MS analysis of immobilization reaction of c-dasatinib. (a) UV chromatograms of T0
and T16
samples measured at
254 nm. The peak at a retention time of 3.00 min disappears after 16 h of incubation. (b) Positive mode electrospray MS
spectra at 3.00 min retention time of the T0
and T16
samples confirming that the observed peak represents c-dasatinib,
which completely disappears after incubation, i.e., couples to the activated matrix.
45. 32 U. Rix et al.
1. Culture KU812 cells in 20 mL DMEM on a 15 cm dish. After
cells approach confluence, centrifuge the cell suspension for
3 min with 300×g. Aspirate the medium and resuspend cells in
2 mL fresh medium.
2. Fill 20 mL DMEM in four new 15 cm dishes and add 0.5 mL
of the cell suspension. After ca. 2 days cells approach conflu-
ence and then need to be split again.
3. To harvest the cells, centrifuge the cell suspension, aspirate the
medium, and wash with PBS. Shock freeze the samples in liq-
uid nitrogen and store them at −80°C until further use or use
them directly for lysis and the protein quantification.
1. Thaw pellet on ice and resuspend in appropriate amount of
Lysis buffer (depending on pellet size) and pull it through a
0.9 mm syringe ten times (see Note 9).
2. Transfer the homogenate to a 15 mL Falcon tube and incubate
on ice for 30 min.
3. Transfer lysate to an ultracentrifuge tube, balance tubes, and
centrifuge for 10 min at 4°C with 20,000×g.
4. Transfer the supernatant to a fresh polycarbonate ultracentri-
fuge tube, balance tubes, and centrifuge for 1 h at 4°C with
100,000×g.
5. Transfer supernatant to a fresh 15 mL Falcon tube and keep
on ice.
6. Determine the protein concentration by the Bradford method
using a freshly determined standard curve. Apply 0, 5, 10,
15, 20, 25, and 30 μL γ-globin and 0.5, 1, and 2 μL lysate to
the cuvettes. Add 1 mL of the dye reagent (1×) to each
cuvette, mix by careful vortexing, and measure the absor-
bance at 595 nm.
7. Prepare 25 mg aliquots in microcentrifuge tubes. Use the
lysates directly for the drug pulldown or shock freeze the sam-
ples in liquid nitrogen and store them at −80°C until use.
1. Prepare the 1× Lysis buffer freshly and keep it on ice.
2. Dilute the thawed cell lysate (preferably, prepare cell lysate
freshly) with 1× Lysis buffer to a total volume of 3 mL (for
25 mg total protein) transfer to Beckman ultracentrifuge tube,
balance tubes, and centrifuge for 20 min at 4°C with
100,000×g.
3. Collect “total cell lysate” (TCL) sample (100 μg per Western
blot), add the appropriate amount of 4× Laemmli sample buf-
fer, and denature proteins at 100°C for 3–5 min.
4. Pipette 100 μL 50% drug-bead slurry (i.e., 50 μL settled bead
volume=0.1 μmol drug) into a 2 mL Microcentrifuge tube
3.3. Cell Culture
and Harvest
3.4. Preparation
of Total Cell Lysate
and Protein
Quantification
3.5. Affinity Pulldown
46. 33
3 Compound Immobilization and Drug-Affinity Chromatography
(always use cut pipette tips for pipetting beads) and centrifuge
beads for 3 min at 4°C with 75×g, remove supernatant.
5. Add 1 mL 1× Lysis buffer and wash beads by gently resuspend-
ing and inverting several times, centrifuge beads, remove super-
natant, repeat wash step three times, and remove supernatant.
6. Combine and resuspend the beads with the centrifuged cell
lysate.
7. Incubate on roto-shaker for 2 h at 0–4°C with 10 rpm.
8. Centrifuge beads, remove and discard supernatant, but leave
ca. 700 μL buffer in microcentrifuge tube (perform following
steps in cold room at 0–4°C).
9. Resuspend beads gently and transfer to (plugged!) Mobicol
columns, let beads settle by gravity.
10. Drain remaining buffer by gravity flow, fill with fresh 1× Lysis
buffer and connect to 30 mL syringe with luer lock tip.
11. Add 7.5 mL 1× Lysis buffer and let the buffer drain by gravity
flow (possibly apply gentle pressure with syringe plunger).
12. Place the column in a 2 mL microcentrifuge tube and centri-
fuge for 1 min at 4°C with 100×g, plug the column, and place
it in a 1.5 mL microcentrifuge tube.
13. Add 30 μL 4× Laemmli sample buffer and denature proteins at
100°C for 3–5 min.
14. Open first the top lid of the column, then unplug the bottom
(careful!) and place back into the microcentrifuge tube.
15. Centrifuge for 1 min at room temperature with 400×g, collect
eluate (this is the first elution sample) and replug the column.
16. Place column in another 1.5 mL microcentrifuge tube, add
30 μL 4× Laemmli sample buffer and denature proteins at
100°C for 3–5 min.
17. Unplug the column, centrifuge (as before) and collect eluate
(this is the second elution sample) (see Note 10).
18. Store at −20°C until SDS-PAGE analysis.
1. For the 7% separating gel, mix 2.5 mL SDS buffer, 2.35 mL
30% acrylamide, 5.17 mL H2
O, 100 μL 10% APS, and 10 μL
TEMED and fill ca. 4 mL (leave space for 2.5 mL of stacking
gel) of the resulting solution between the two glass plates and
overlay with 1 mL isopropanol, wait until the gel has polymer-
ized (ca. 20 min) and pour off the isopropanol.
2. For the stacking gel, mix at first 25 mL Gel buffer 2 with
16.6 mL 30% acrylamide and 58.4 mL H2
O. Take 10 mL of
this solution and add 100 μL 10% APS and 15 μL TEMED.
Pour ca. 2.5 mL on the polymerized separating gel and insert
a comb.
3.6. One-Dimensional
SDS-PAGE
47. 34 U. Rix et al.
3. After another 20 min remove the comb and place the gel in the
electrophoresis chamber, which is filled with running buffer.
4. For immunoblotting, load 10 μL (5 μl each of first and second
elution) per well, in the first well inject 5 μL of the protein
marker. For silver staining, mix 10 μL of the first and second
elutions and alkylate the sample with iodoacetamide (final con-
centration 13 μg/μL) for 20 min in the dark. Then load the
samples on the gel. If possible, fill the first and the last well of
the gel with 8 μL of Laemmli sample buffer to ensure an even
running profile.
5. Complete the assembly of the gel unit and connect the power
supply. The gel can be run at 120 V or higher, if time is short.
When the lysates are run through the gel it can be transferred
to the nitrocellulose membrane or used for silver staining.
This section is illustrated in Fig 3a.
1. Perform all the following steps in a dust-free cabinet to mini-
mize keratin contamination. First, fix the gel with 40% ethanol
and 10% acetic acid (always add sufficient solution to com-
pletely cover the gel) for 1 h at room temperature on a rocker
or at 4°C overnight.
2. Wash the gel two times with 30% ethanol for 20 min and for
another 20 min with water (pro analysi). In the meantime prepare
the sensitizer, the silver nitrate (refrigerator) and the developer.
3.7. Silver Staining
Fig. 3. SDS-PAGE-based analysis of the dasatinib pulldown eluate from KU812 CML cells (9). (a) Silver staining shows
several bands that subsequent LC-MS/MS analysis reveals to correlate with validated dasatinib target kinases.
(b) Immunoblot with anti-ABL 21–63 antibody shows the enrichment of BCR-ABL and c-ABL by the dasatinib affinity matrix
while an ampicillin control matrix does not display affinity for the dasatinib cognate targets. SFK SRC family kinase.
48. 35
3 Compound Immobilization and Drug-Affinity Chromatography
3. Sensitize the gel with 0.02% Na2
S2
O3
for 1.5 min and wash it
three times for 20 s with water (pro analysi).
4. Incubate the gel with refrigerated 0.2% AgNO3,
for 20 min.
5. Discard the silver nitrate in heavy metal waste and wash the gel
again (three times for 20 s). Transfer the gel into a clean Petri
dish after the second wash step to keep the background as low
as possible.
6. Develop the gel in 3% Na2
CO3
and 0.05% formaldehyde. Rock
the Petri dish until the solution changes to a yellow color.
Remove the liquid and allow the development without liquid.
If the staining is not sufficient, repeat the developing step.
7. When all the bands are visible on the gel, quench the developer
by removing the liquid from the Petri dish and adding 5% ace-
tic acid. Leave the gel in the acetic acid for a minimum of
5 min.
8. Remove the acid, place the gel in the lid of the Petri dish, cover
it with parafilm, and scan the developed gel for the documen-
tation using a conventional picture-scanner.
9. Cover the gel with water if the gel is stored in the refrigerator
for further analysis.
This section is illustrated in Fig. 3b.
1. Take three 8×9 cm filter papers, soak them in 1× Western blot
buffer (containing 10% methanol), and place them in the transfer
unit. Then take a 7×8 cm membrane, soak it in the buffer, and
place it on the filter paper. Cut away the comb of the gel, soak
the gel in the buffer, carefully lay it on top of the membrane,
and cover it with another three soaked filter papers. Finally
ensure that there are no air bubbles in the resulting sandwich
and close the transfer cassette. The transfer can be accom-
plished at 56 mA (1 mA per cm²) for 1.5 h.
2. Incubate the membrane for 1 h in approximately 50 mL
Blocking buffer.
3. Discard the Blocking buffer and incubate the membrane with
the primary antibody dilution over night in the cold room. Use
gentle agitation.
4. Wash the membrane three times for 5 min with PBS-T.
5. Add 10 mL of the secondary antibody dilution (always use a
fresh solution) and incubate for 1 h.
6. Discard the solution and wash the membrane again three times
for 5 min.
7. Mix together the ECL Plus solution (1:40) and cover the
membrane with 2 mL of the resulting solution. Incubate for
4 min and let the redundant ECL solution drip down. Envelope
the membrane with the Saran-Film (ensure that there are no
3.8. Immunoblot
49. 36 U. Rix et al.
bubbles) and place it in an X-ray film cassette. Perform the
remaining steps in a dark room under safe light conditions.
8. Insert a developing film in the cassette and expose the film for
a few seconds to minutes (see Note 11).
1. Always use cut pipette tips for pipetting beads with a clean
scalpel or scissors.
2. Use high-purity distilled water of consistent quality to avoid
contaminations, such as DNase and RNase free distilled water
(Gibco).
3. The primary antibody and therefore the blocking conditions
depends on any prior knowledge of validated target proteins of
the bait compound. For our example, we have chosen dasat-
inib as the bait compound, the cognate target of which is ABL.
Therefore, we will delineate in the following conditions for the
anti-ABL (21–63) antibody (Santa Cruz Biotechnology).
4. The primary antibody can be often reused for subsequent
applications. If used in milk/PBS-T, it should be stored at 4°C
and be used within a few days. If used in BSA, it can usually be
stored at −20°C for several months.
5. Always use aerosol filter pipette tips to avoid cross contamina-
tion of samples, as mass spectrometers are highly sensitive instru-
ments, which will detect even trace amounts of contaminants.
6. Contamination of samples with keratin is one of the most com-
mon and serious complications for proteomics mass spectrom-
etry analysis, due to the abundance of this family of proteins
and dynamic range limits of mass spectrometers. There are sev-
eral sources of keratin contamination, such as dust, hair, skin,
and clothing. It is therefore essential to take the precautions to
reduce keratin levels as much as possible.
(a) Always wear gloves and laboratory coat as much for per-
sonal protection as for avoiding keratin contamination.
(b) Avoid wearing woollen clothing, but rather synthetics or
cotton.
(c) Use a designated dust-free clean bench.
(d) Use a dedicated set of pipetting and electrophoresis
equipment.
(e) Prepare buffers and reagents freshly from designated stock
solutions.
7. The bait compound needs to contain either a primary or sec-
ondary amino group, as does c-dasatinib, which was designed
4. Notes
50. 37
3 Compound Immobilization and Drug-Affinity Chromatography
based on the available X-ray co-crystal structure of dasatinib
with ABL (10). Alternatively, primary or secondary alcohols
can be readily esterified under Steglich conditions, i.e., in the
presence of dimethylaminopyridine (DMAP) and dicyclohexy-
lcarbodiimide (DCC) (5). For example, the hydroxyl group of
dasatinib can be esterified with N-Boc-glycine. Subsequent
removal of the Boc protecting group with trifluoroacetic acid
yields a primary amine suitable for immobilization.
8. Duration of storage depends strongly on the stability of the
bait compound. Although the dasatinib matrix was sufficiently
stable, our experience with other compounds suggests that a
storage of maximum 2 weeks should not be exceeded. If pos-
sible, drug beads should be used immediately.
9. Some primary cells express high levels of protease activity
which may require the addition of additional protease inhibi-
tors to the Lysis buffer, such as the Complete Protease Inhibitor
Cocktail Tablets (Roche).
10. Samples from the first and second elution are stored separately,
but can be combined directly before loading on SDS-PAGE
gel, if desired.
11. Exposure time typically varies.
Acknowledgements
This work was supported by the Austrian Federal Ministry for
Science and Research (BMWF) under the GEN-AU program (GZ
BMWF-70.081/0018-II/1a/2008) and the Austrian Academy of
Sciences (ÖAW).
References
1. Oda, Y., Owa, T., Sato, T., Boucher, B.,
Daniels, S., Yamanaka, H., Shinohara, Y.,
Yokoi, A., Kuromitsu, J., and Nagasu, T.
(2003) Quantitative chemical proteomics for
identifying candidate drug targets, Anal Chem
75, 2159–2165.
2. Rix, U., and Superti-Furga, G. (2009) Target
profiling of small molecules by chemical pro-
teomics, Nat Chem Biol 5, 616–624.
3. Han, S.-Y., and Kim, Y.-A. (2004) Recent
development of peptide coupling reagents in
organic synthesis, Tetrahedron 60, 2447–2467.
4. Anderson, G. W., Zimmerman, J. E., and
Callahan, F. M. (1963) N-Hydroxysuccinimide
esters in peptide synthesis, J. Am. Chem. Soc.
85, 3039.
5. Neises, B., and Steglich, W. (1978) Simple
method for the esterification of carboxylic acids,
Angew. Chem. Int. Ed. Engl. 17, 522–524.
6. Kocienski, P. J. (2005) Protecting Groups, 3rd
ed., Thieme, Stuttgart.
7. Rix, U., Hantschel, O., Durnberger, G.,
Remsing Rix, L. L., Planyavsky, M., Fernbach,
N. V., Kaupe, I., Bennett, K. L., Valent, P.,
Colinge, J., Kocher, T., and Superti-Furga, G.
(2007) Chemical proteomic profiles of the
BCR-ABL inhibitors imatinib, nilotinib, and
dasatinib reveal novel kinase and nonkinase tar-
gets, Blood 110, 4055–4063.
8. Lombardo, L. J., Lee, F. Y., Chen, P., Norris,
D., Barrish, J. C., Behnia, K., Castaneda, S.,
Cornelius, L. A., Das, J., Doweyko, A. M.,
52. wines, wool, and other products are numerous, but unimportant.
The iron ore mines (red and brown hematite) in the Somorrostro range
and district are largely in the hands of English capitalists. These mines,
which began to attract the attention of British iron masters about 1870, occur
chiefly in the mountain limestone, and are worked in open quarries. Short
railways and tramways have been made to San Nicolas on the Nervion; and
a wire tramway has been constructed by the Galdames Mining Company,
who possess a cliff of iron ore about a mile long and 280 feet high. The
tramway carries the ore through a tunnel, 600 feet long, to the quay. The
Landore Siamese Steel Company have important hematite mines connected
with the river by a wire tramway, carrying baskets for loading.
BILBAO—THE ORCONERO IRON ORE COMPANY’S WHARF IN LUCHANA.
Bilbao is largely modern and wholly commercial, and its public buildings
are not notable. But its thoroughfares are full of movement, and the shady
arenal, in the old town—the focus of the life of the whole city—contains the
principal hotels, the chief cafes, and the New Theatre. The land which this
beautiful promenade now occupies was at one time very boggy, and swept
by the tides. Now the two principal avenues are asphalted. The Church of
San Nicolás de Bari, which faces it, is one of the city parish churches. It was
built towards the end of the fifteenth century on the ruins of the sailors’ and
fishermen’s little church. This church has suffered greatly on account of
floods, especially during the year 1553. It was closed in 1740 as ruin
threatened it. When it fell, the present one was begun in 1743. During the
last war it was used as a provisioning station; and, after repairs, was opened
for worship on the 21st of January, 1881.
53. A GALICIAN.
T
In Northern Spain.
HE great bulk of the Spanish people know as little
of Galicia and the neighbouring Principality of the
Asturias as the average Englishman knows of the
Hebrides. Nor can they judge of the inhabitants of these
provinces from the few individual Galicians who
emigrate to Madrid any more than we in England can
form an idea of Italians from the specimens who
perambulate the London streets with a piano organ and
a monkey. The Madrileño comes across a few Galicians
in the capital engaged in menial services, and speaking
a harsh, strange patois, which he finds some difficulty
in understanding; but the Gallegan in exile is a very
different person from the man you meet in his own land
of rain and mist, where the scenery is exquisite, the hotels are famously bad,
and devotion is the chief recreation of the community. At home these people
are poor, but hardy; possessing little intelligence, but great capacity for
work; knowing little comfort, but nursing a passionate attachment for the
country of their birth. Many of the young women are remarkably handsome,
but drudgery and hardship early tell their tale, and very few of them retain
their good looks beyond the age of twenty. The country, for the most part, is
poor to barrenness; the peasantry work day and night for mere subsistance;
the cottages, which do duty for bedroom and nursery, stable, kitchen, rabbit
hutch, pigsty and parlour, are damp and dirty, and destitute of beds or
chimneys. The climate is rainy, the surface is mountainous, and the roads are
generally bad. Small wonder is it that muleteers and commercial travellers
constitute the principal visitors to Galicia—for those who have a soul above
scenery, and an ambition beyond fishing, the country is practically without
attraction.
The single province of Oviedo, which constitutes the principality of the
Asturias, harbours a people who have remained unconquered alike by
Roman and Moor. There is protection, if not complete safety, in a country of
mountain and valley, of damp and cold; and the Asturians have ever been
able to spread themselves over the land and farm their straggling holdings in
comparative security. They have cultivated maize for their staple food,
54. A GALICIAN. A GALICIAN.
poached the hills and rivers for
game and fish, cultivated the art
of dancing, and lived in terror of
the evil eye from the most
ancient times; and despite damp,
hard fare, and harder toil, they
have learnt
REDONDELA (PROVINCE OF PONTEVEDRA)—GENERAL VIEW.
the secret of longevity and the charm of a gracious civility of manner.
Minerals in abundance are common to both Asturias and Galicia; and while
the former is the richer in coal and iron, the latter has been worked for gold,
silver, and tin from the time of the Roman occupation. It is on their mineral
resources that these provinces will have to depend for their future prosperity.
55. IN GALICIA.
IN GALICIA.
After the
cities of the
South—
Barcelona,
Toledo,
Granada, or
even modern
Madrid—the
Northern towns
are small,
shabby, and unimportant. Coruña, the chief seaport of Galicia, though
interesting to Englishmen as being the landing place in Spain of John of
Gaunt, and the harbour from which the invincible Armada sailed to conquer
and Romanise Great Britain, is a place of only secondary importance. The
city was founded by the Phœnicians; its name is probably derived from
Columna, the Phœnician Pharos, or lighthouse; and its famous lighthouse,
the Tower of Hercules, has had its counterpart from the earliest days. The
Phœnicians, who made gain rather than discovery the aim of all their
expeditions, were attracted to Galicia and to the province of Orense
particularly by reason of its rich deposits of tin. Coruña in ancient days was
the principal port of the North-west Coast, and the most westerly town in
Europe. It is still the chief military station in Northern Spain, and ranks as a
commercial city of the first importance.
CORUÑA—GENERAL VIEW TAKEN FROM THE OLD TOWN.
The hill-girt city of Santiago, though knowing nothing of commercial
prestige, and having no part in the military system of the country, is to the
traveller of far more interest than the capital of the province. For dead as it
56. now appears to be, with the hand of death on its crooked, branching streets,
and its crazy, deformed squares, which echo the pilgrims’ footfalls to the
deaf ears of the dead, it was at one time the most celebrated religious centre
in Spain—the goal of fanatics from every corner of Europe, the Mecca of
countless thousands of theologians, and the tomb of one of the personal
companions of Christ. Although the ancient glory of Santiago has departed,
although
PONTEVEDRA—GENERAL VIEW.
its broad-flagged pavements are no longer thronged by the feet of the
devout, and it has been much shorn of its former civil and religious dignities,
the city is still the See of an Archbishop with a cathedral, two allegiate
churches, and fifteen parishes. The cathedral is erected on the site of the
chapel which was erected by Alonso II. to mark the spot where Theodomer,
Bishop of Iria Flavia, is said to have discovered the body of St. James the
Apostle; and the city, which sprang up around the memorial, bears the
Spanish name for St. James the Elder. The original cathedral, which was
finished in 879, consecrated in 899, and destroyed by the Moors in 997, was
replaced by the present edifice in 1078. Whether one believes or not the
tradition of the foundation of the cathedral—which, by the way, is no mere
57. tradition in the mind of the Galician—one cannot but regard this mighty pile
of stone with awe, and recognise in it the expression of an influence which
was once felt throughout the Christian world. Even to-day it is one of the
most frequented pilgrim-resorts in Europe.
One passes through Pontevedra, a picturesque granite town, with arcaded
streets and ancient houses bearing armorial shields, on the journey to Vigo.
Here, as everywhere on the Galician coast line, the parish priest goes down
to the shore one day in every year and blesses the sea; here also the oysters
are excellent and abundant, and here the watchman’s night chant is heard in
the streets. The call of the sereno, or watchman, who dates from the building
of the ancient walls of Pontevedra, and the chapel of Alonso II. of Santiago,
seems to catch the imagination of the traveller, and hurl him back into the
mediæval ages, when life was a state that men fought to retain, and religion
was a power for which they laid it down. The sereno, with his theatrical
cloak wrapped about him, his axe-headed staff, his lantern, his majestic
stalking walk, and his thrilling chant, “Ave Maria Purissima. Son las diez y
sereno,” seemed to me impressive, unreal, almost fantastic. At ten o’clock he
passed me in the deserted square, at eleven he was offering up his quavering
invocation beneath my window. Galicia has little in common with the towns
of the South—it retires to rest early in order to be up betimes.
At Vigo a small fragment of the ancient sea walls yet remain, but the
ruins that Lord Cobham made of the town in 1719 have been obliterated, and
in place of the fortified port, which Drake visited in 1585 and 1589, we have
a thriving, modernised town. Vigo is an important place of call for
Mediterranean steamers, it is one of the chief centres of the cattle trade
export to London, and the port of the mineral provinces of Pontevedra and
Orense.
The town of Orense, the capital of its province, is reached by the
magnificent old bridge that spans the river Miño. Though now deprived of
three of its arches, which were removed to give the road more width, and
also of the ancient castle which defended the entrance, it continues to attract
the attention of the traveller on account of its elegant and bold construction,
its ample proportions and majestic appearance. Tradition says it is Roman,
but many learned writers find nothing to confirm this assertion. It is quite
likely that a bridge existed there previously; but the present one, it would
appear, was built by order of Bishop Lorenzo during the first half of the
thirteenth century, and has since undergone many alterations, including those
58. to the largest arch, which is more than forty-three metres in width, and the
reconstruction of which was completed about the middle of the fifteenth
century. In the Roman days Orense was celebrated for its warm baths. These
three springs, which are still in existence, flow copiously from fountains one
above another, but the waters have lost their medicinal virtues—it is
VIGO—VIEW FROM THE CASTLE.
only a supposition that they ever possessed any—and are now used for
domestic purposes. The present cathedral, which is an obvious imitation of
the cathedral at Santiago, was raised in 1220. The cathedral, the warm
springs, and the bridge over the Miño, comprise the three marvels of the city.
59. GIJON—THE WHARF.
Equally ancient, but in many ways more interesting, is the capital town of
Lugo. It boasts a cathedral which shares with San Isidoro of León the
immemorial right to have the consecrated Host always exposed; Roman
walls in an excellent state of preservation that entirely surround the city, and
an establishment of baths. The bath-house contains 200 beds; and the
springs, which contain nitre and antimony, are good for cutaneous diseases
and rheumatism. The river Miño, which is the glory not only of Lugo but of
Galicia, rises in the mountains, some nineteen miles from the city.
As the centre of a beautiful and variegated country, which affords good
sport for the angler, and scenery of enchanting loveliness to attract the artist,
Oriedo, the capital of the Astionas, has its charms; but the seaport of Gijon,
with its tobacco manufactory, its railway workshops, its iron foundry, and
glass and pottery works, is a much more thriving and important town. Gijon,
like Santander, is a flourishing port; and both have gained immensely in
importance of late years. While the latter, with its handsome modern houses,
makes a more splendid show, its drainage and sanitary arrangements leave
much to be desired, and the harbour at low water is sometimes most
offensive. Both towns are of Roman origin, but Gijon is the most pleasantly
situated on a projecting headland beneath the shelter of the hill of Santa
Catalina, and the harbour is the safest on the North Coast. It exports apples
and nuts in enormous quantities, coal, and iron, and jet; while its shores are
much frequented by bathers during the summer months.
60. SANTANDER—THE PORT.
SANTANDER—GENERAL VIEW.
It is currently believed, and I have no reason to doubt the accuracy of the
statement, that if a visitor in any town in England stops the first native he
meets and inquires as to the objects of interest that the place possesses, he
will be referred immediately to the principal hostelry of the town. If you
wander in London, and ask your way about, you will be directed right across
the city by references to public-houses, which are the only landmarks that
the Cockney ever dreams of studying. In Spain, cathedrals are as ubiquitous
as inns are in England. You may be sure of finding comfortable
accommodation for man and beast in most English towns, and in the
Peninsula you can be quite as confident of “bringing up” against a cathedral
—if nothing else. In León, the capital of the province of the same name, and
in Salamanca, the second city in the province, we find the same state of
things existing—the cathedral first and the rest nowhere. Yet these two cities
boast of a noble history of ancient splendour and old-time greatness, and
with this—and their cathedrals—they appear to be content. León, in the time
61. LEÓN—THE CATHEDRAL.
LEÓN—CLOISTER IN
CATHEDRAL.
of Augustus, was the headquarters of the legion that defended the plains
from the Asturian marauders; and when the Romans withdrew, it continued
as an independent city to withstand the continued attacks of the Goths until
586. The city yielded to the Moor, was rescued by Ordoño I., and retaken by
the Arabs with every accompaniment of inhuman atrocity. Its defences were
rebuilt by Alonso V. nearly 400 years later, its houses were repeopled, and it
continued to be the capital of the Kings of León until the court was removed
to Seville by Don Pedro. Its present miserable condition is a lamentable
appendix to such a history. Its streets are mean, its shops are miserable, and
its inns are worse. Nothing is left to it but its cathedral.
This temple is truly an architectural
wonder, combining the delicacy of the
purest Gothic style with a solidity which
has stood for centuries; the manner in
which the problem of
stability was solved
is wonderful, the
immense weights
seeming to have no
solid bases. The
finest and most
beautiful chiselled
work is visible
everywhere, and
careful study is
necessary in order to
understand how the
weight and strain of
the arches were made to rest on their
elegant buttresses. The origin of this magnificent temple is not quite clear,
but many archæologists believe that it was founded in the time of King
Ordoño II. It is of irregular form, but the cathedral or nave, transept, and
presbytery are in the form of a perfect Latin cross.
62. LEÓN—THE CATHEDRAL CHOIR STALLS.
The windows are of colossal dimensions, and the ratablos and sculptures
are notable. Among its many famous works the cloister must not be
forgotten. It is an example of the transition style from ogive to renaissance,
with large galleries, interesting groups of sculpture, and a beautiful door
leading into the temple.
LEÓN—VIEW TAKEN FROM THE CEMETERY.
Among all the choral stalls treasured in Spanish churches those in the
cathedral at León stand out prominently. Unfortunately, the names of the
master who designed them, and of the artists who assisted him to carry that
marvel of ogive art into effect, are not known; but it must have been
executed during the last thirty years of the fifteenth century, for it is known
that in 1468 the necessary bulls were obtained from his holiness through
Archbishop Antonio de Veneris in order to arrange means for meeting the
cost of the stalls, and in 1481 the work was still proceeding.
63. SALAMANCA—GENERAL VIEW.
Salamanca has a great name, a florid Gothic cathedral, and a square of
handsome proportions and pleasant prospects. In other respects, it is quite
without attractions. The streets are badly paved and dull, the climate is
shrewd, and fuel, I was told, is scarce and expensive. Even the cathedral,
though grand, is bare; and when one has visited the cathedral and lingered
awhile in the pleasant garden of the Plaza Mayor—one of the largest and
handsomest squares in Spain—and tested the accommodation of “La
Comercio,” one can find little else to entrance one in the disappointing old
city which was once a world-famed seat of learning. In the fifteenth century,
when its university gave precedence to Oxford alone, it boasted of 10,000
students. In the following century its scholars had declined to one half that
number, and to-day only some few hundred students are on its books. The
sun of Salamanca commenced to set at a period of the world’s history that to
all the rest of Europe was one of awakening and advancement. Decline and
decay are writ large on the face of the city. From a distance its noble
situation and fine buildings, built of beautiful creamy stone, gives the place
an imposing and picturesque appearance. But though the shell of Salamanca
remains, its spirit has departed. The ravages of the Romans, the Goths, the
Moors, the Spaniards, and the ruin which the neighbourly French inflicted
less than a hundred years ago, have left their cruel marks upon its historic
walls. Salamanca is but a broken hulk spent by the storms that, from time to
time, have devastated her. Her narrow, tortuous, ill-paved streets, which skirt
its multitude of grandiose buildings, her squalor and poverty, her inferior art
work, but even more the uncorrupted art of the grand old cathedral, all
remind us of what Salamanca was, and turn our eyes backwards from what it
is.
64. SALAMANCA—VIEW OF THE COLLEGE FROM THE IRLANDESES.
ZARAGOZA—“INDEPENDENCIA” PROMENADE.
65. ZARAGOZA—PILAR CHURCH.
One must approach Zaragoza with one’s mind full of memories of heroes,
queens, poets, and bandits that have been associated with this once mighty
city, and one’s heart filled with sympathy and respect for the old, proud
Aragon that flourished, and was illustrious in history while the Englanders
still decorated themselves with blue paint, and were domiciled in caves. For
Zaragoza is not altogether a gay or an exhilarating city. Many of the streets
have a gloomy aspect, and the old houses are high, dark, and repellant. But
the city is not only important as the seat of a university, an Audiencia, an
archbishop, the captain-general of Aragón, and other officials; it is also the
junction of four railways, and its commercial progress has been steadily
increasing of recent years. For Zaragoza is in reality two cities—the old part
with ancient fortified houses, converted now into stables and wood stores,
and the new part traversed by broad, well-paved, and excellently-lighted
streets, and lined with modern buildings. Until the railway connected the city
with Madrid and Barcelona, Zaragoza was as dead as Salamanca, and as
dilapidated as León. But it has always held the advantage of those places in
having two cathedrals to their one. The principal cathedral, that of La Seo, is
a venerable Gothic pile occupying the site of a Moorish mosque, and its high
arches have echoed many councils, and looked down on the solemn
coronations of the kings of Aragon. More modern is the Cathedral El Pilar,
so called from the identical pillar on which the Virgin descended from
heaven. It was commenced on St. James’s Day, 1686, the work being
designed and carried out by the famous Don Francisco Herrera, the architect.
In the year 1753 King Ferdinand VI. instructed Ventura Rodriguex, the
architect, to design and build a new church, as luxurious as possible, in
which to instal the image without taking it out of its temple. This was done
by erecting a small Corinthian temple under the magnificent cupola, which
was ornamented with the richest marble and jasper that could be procured.
On one of the altars of this temple, which is crowned with a magnificent
silver canopy, reposes the venerated effigy, the jewels on which are of
incalculable value.
66. A FLEMISH DANCE.
AT NUEVALOS.
The Stone Monastery at
Nuevalos, on the right bank of
the river from which it takes its
name, is one of the places most
worthy of a visit in the province
of Zaragoza, not only on account
of the building itself, which is of
great historical interest, having
been built in 1195, but for the
delicious picturesqueness of the
place. Surrounded by rocks,
winding amidst thick woods and
dashing into deep abysses, this
river runs its erratic course, imparting life to a landscape which is, according
to the noted poet, Don Ramon Campoamor, “an improved dream of Virgil.”
Among its many picturesque waterfalls, the one called “La Caprichosa” is
perhaps the most beautiful.
The dress of the Aragonese peasantry is peculiar and picturesque. The
men, as a rule, wear no hats, but have instead a coloured handkerchief
wound round the head, leaving the top bare. Their knee-breeches are slashed
down the sides and tied by strings below the knee. The waistcoats are worn
open. Round the waist they wind a wide sash, in the folds of which pipes,
tobacco, money, and provisions are carried as safely as in a pocket. Their
feet are shod with sandals, and they universally carry a blanket, which is
thrown in a graceful manner over their shoulders.
67. A
Bull-fighting.
BULL-FIGHT is underlined for an early visit in the note-book of every
visitor to Spain. He goes prepared to be disgusted, and he comes away
to denounce it as a revolting and demoralising exhibition. He even
plumes himself upon his moral and human superiority over the Spaniard,
because the spectacle proves too strong for his untutored stomach. The
inference is as gratuitous as it is illogical. In point of fact, the effect of the
spectacle upon the spectator is not so much a matter of sensibility as custom.
The Spaniard grows up to the sport as our Elizabethan ancestors grew to
bull-baiting—even as the present generation of Englishman grows to
pugilism. To the Spaniard, the cruelty of the craft of tauromachy does not
appeal; the spectacle inflames his blood, and stirs not a chord of compassion
in his nature. Yet he can be intensely sympathetic, gentle, and tender-
hearted; but these softer qualities of character are not touched by the sight of
animal suffering. In the first place, the bull is his enemy by heredited
tendency. He cannot forbear to hurl insulting epithets at him when he
chances to pass him on a journey. He witnesses his end with the thrill of
satisfaction which a soldier feels in the death of a treacherous and
implacable foe. The Englishman cannot share, or even realise this sentiment
—it would be strange if he could. His leading feeling is curiosity, and a
nervous apprehensive tension which only magnifies the horror and repulsion
of the sport. With the Spaniard it is entirely different. Long habit has
familiarised him with the bloody details, and his experienced eyes follow
each trick and turn of the contest with the enthusiasm of an athlete watching
an athletic display. Every detail of skill and dexterity and nerve exhibited by
the fighters, and every clever move made by the bull is greeted with critical
applause. Cruelty there must be, but courage in a high degree is a factor in
the contest—danger gives to the contest a dignity which is absent from
pheasant shooting, and which formed no excuse for the vogue to which bear-
baiting and cock-fighting once attained in this country.
68. THE PROCESSION.
It may be thought that I am trying to champion an institution which is
regarded with aversion by all classes of English people, but such is not my
intention. My object is to look at it from the Spanish point of view, and
endeavour to see if there is not some plausible explanation of its popularity
as a national amusement. But when all is said and done, there still exist two
objections to the sport which cannot be explained away. The first is the
almost inexplicable indifference which a Spanish audience shows for the
torture that is inflicted upon the horses that take part in the corrida: the other
is the attendance of the gentler sex. It must, however, be noted that a large
proportion—certainly the majority of Spanish ladies—are opposed to the
sport, and with the rest it is the manly courage and address of the performers
that fascinates them. But the fact remains that women are seen in large
numbers in the amphitheatre, as 300 years ago good Queen Bess was not
ashamed to be a spectator at many an exhibition of bear-baiting. English
sentiments in matters of sport have undergone a great change since the
Elizabethan era, but Spain is notoriously the most conservative country in
Europe.
However, enough has been said of the theoretical side of bull-fighting; let
us accompany the seething populace to the Plaza de Toros, and witness the
sport for ourselves. The streets of Madrid are crowded with people who are
all moving in the same direction. April to October is the regular bull-fighting
season, but the Spaniard finds the lightest excuse a sufficient one for
indulgence in his favourite pastime during the “close” season. And so,
although it is February when I am in Madrid, I am not to forego an
experience of a promising corrida.
69. Although I have seen bull-fights in some of the best rings in Spain,
including those of San Sebastian, Valencia, Barcelona, and Madrid, it is
more particularly of my experiences at the latter place that I shall write.
During the fashionable months, a boletin de Sombra, or “ticket for the
shade,” is a luxury to be prized; but in February, in Madrid, we need all the
warmth and glare that the sun can give us. The present Bull Ring, which was
built at a cost of £80,000, and opened in 1874, seats 15,000 persons. It
stands on a gentle elevation in a broad stretch of bare yellow land, where it
raises its brick-coloured walls—the only land-mark in the barren, treeless,
desolate expanse between the city and the solemn distant mountains. Around
the various entrances countless human beings cluster like bees, and the Plaza
is alive with men and horses, mules with tinkling bells, soldiers, police,
picadors, and fruit-sellers. What strikes one most curiously about this
concourse of human beings, both outside the bull-ring and within the huge
amphitheatre, which rises tier above tier from the brown sand till it is almost
lost in the vast expanse of blue above, is its single-mindedness, its patience,
and the entire absence of horseplay. To a Spaniard this is not curious, but to
the English spectator some familiar characteristic of a crowd appears to be
absent.
ENTRANCE OF THE BULL.
Punctuality is not a strong trait in the Spanish character, but punctuality
will be observed to-day. At the hour and the minute appointed, the President
enters his palco, the signal is given, and the proceedings commence. The
procession, headed by two caballeros, habited in black velvet, moves slowly
across the ring to the front of the President’s seat. The two espadas in yellow
and violet, and gold and green costumes respectively, follow the caballeros.
After them come half-a-dozen stoutly-protected picadores, then eight
70. banderilleros, gay with a profusion of silk sashes, short breeches, and
variously-coloured hose, and the rear is brought up by a posse of attendants,
leading the mules, all bedecked in plumes and rich trappings, which are to
drag off the carcases from the arena. The entrance of the glittering cavalcade
is announced by a trumpet sound, and the President tosses the key of the
toril into the ring.
To the “new chum,” all this preliminary detail, commonplace and
“circusy” as it is, is sufficient to strain the nerves, and expectancy changes to
apprehension. The creak emitted by the opening of the heavy door of the
toril intensifies the feeling. The clutch of curiosity with which the entire
concourse awaits the entrance of the first bull is contagious. Instinctively one
strains forward and catches one’s breath. Toro does not keep us long in
suspense. There is a momentary lull, and then the bull dashes from his dark
cell into the glint of the Spring sunshine. The novelty of the environment
staggers him for a moment. He hesitates in the centre of the ring, and looks
wildly around him. The arena is empty, with the exception of three
picadores, who sit rigidly in a row on their sorry hacks, waiting for the bull
to recognise their presence.
Our first victim is a doughty warrior. He is as ignorant as the blindfold
knackers—that would be dear at a pound a leg—of the fate in store for him.
He may make a brave fight, kill horses, upset men, and leap the barriers with
a heroic rush, but in twenty minutes his corpse will be coupled up to the
mules, and fresh sand will be strewn on the red trail that will mark his last
passage across the arena. The inevitableness of the outcome of the
encounter, so far as the principal actor is concerned, is the least pleasing
feature of the sport. The fox and the stag are
71. ANTONI
O
FUENTE
S.
LUIS MAZZANTINI AND
CUADRILLA.
U
E
R
R
I
T
A
.
B
a
n
d
i
l
l
e
r
o
.
given a gambling chance, the grouse is not without hope, and the gladiator of
the cock-pit may live to fight another day, but the bull is a doomed animal.
Happily he is not capable of calculating the uselessness of his efforts. The
horses stand but little better chance, and the picadores, despite their iron and
leather greaves and spears, are paid to take risks.
The art of the picador is displayed in the skill with which he avoids the
charge of the bull, and turns him on to the next picador, who, in turn, will
pass him on to the third. In this instance the manœuvre does not come off.
The bull’s rush is met by the first picador with the point, but the horse he
strides is too ancient to obey with sufficient celerity the rider’s injunction to
swerve, and horse and man are rolled over with the force of the impact. The
72. wretched equine is lacerated on his opposing flank, but the spearman appears
to be uninjured, and before the bull has completed his circuit of the ring, the
horse is on his feet again, and the picador is waiting for the next attack. The
toreros, with their red capa, are immediately on the spot to draw the bull
from his victim, but the bull is too eager to waste time on a fallen foe. The
second and third horseman avoid his rush; and the bull, smarting from spear
thrusts, and confused by the cheers, is inclined, in racing parlance, to “turn it
up.” The first horse who crosses the line of sight is caught on the brute’s
horns, and is so deeply impaled that the bull has to swerve at right angles to
rid himself of his enemy. The second horse is impaled before the combatant
can plant his spear in the bull’s neck. Steed and rider are lurched in the air,
and fall heavily to the ground, and the momentary victor lowers his head
again to the prostrate man, and rolls him over and over. Toreros hasten to the
spot to get him away, the people rise in their places, ladies lift their fans and
avert their faces, while the air is filled with the usual murmur of lamentation
which accompanies an accident. Both the other picadores are unhorsed
before the President gives the signal for them to retire. Act one of this most
realistic of sporting melodramas is over.
The banderilleros now come forward. They are costumed like Figaro, in
the opera of “Il Barbiere de Sevilla,” and their hair is tied into a knot behind.
To the English spectator, this part of the performance is the most fascinating
and least abhorrent of the entire piece. The banderillero inflicts no more pain
on the bull than the humane angler deals out to the wily trout, and the agility
and daring with which he addresses himself to his task is superb. His aim is
to plant small barbed darts, or banderillas, on each side of the neck of the
bull. The chulos, or apprentices, here open the ball by tantalising the animal,
and working him up to a proper pitch of fury. Then the banderilleros circle
round him, and one, standing full in his line of flight, “defies” him with the
arms raised high over his head. If the bull stops, as he is doing now, the man
walks composedly towards him. Then the bull lowers his head and makes his
rush, and the athlete, swerving nimbly to one side, pins in his banderillas
simultaneously. Again and again the maddened animal, frantic more from
impotence than pain, makes his rushes from one tormentor to another. At
each rush he receives further instalments of his hated decorations. Then one
man bungles. He loses his nerve, or, failing to time the animal’s charge,
shirks the onslaught. A howl of execration greets the exhibition, and the
unfortunate baiter is tempted to more rash efforts. He seats himself in a
73. chair, and waits with suicidal calmness the rush of the bull. Just as the
animal’s horns are thrust beneath him he jumps lightly up, manipulating his
darts with miraculous precision, while the chair is tossed high in the air.
Thunders of applause greet this venturesome feat, and the other
banderilleros, warmed to their work by the plaudits of the public, vie with
one another in deeds of coolness and “derring do.” One waits, alert but
motionless, for the attacks of the charging bull, and as the galloping brute
lowers his head to toss him, places his foot between the terrible horns, and is
lifted clear over his onrushing enemy. Another, seizing hold of the lashing
tail, swings himself along the bull’s side, and plants himself for one thrilling
moment right between the horns.
THE PICADOR.
I once saw a banderillero, in response to the jeers of the crowd, take the
darts, which are about two feet long, break them across his knee, and plant
the stumpy weapons, with unerring precision, on each side of the neck of the
bull.
These feats appear to be fraught with infinite danger, and the agility with
which the performers acquit themselves cannot be witnessed without a
tremour of amazement and admiration. Several times the venturesome
chulos escape death as by a miracle: they sometimes seem so close to their
end when they vault over the barriers to avoid the pursuing bull, that they
appear to be helped over the fence by the bull’s horns. One bull exhibits at
this stage of the proceedings an emphatic disinclination to continue the fight.
He paws the ground when the darts are driven home, but makes no show of
retaliation, and the hoots and opprobrious epithets that are hurled at him by
the populace fail to inspire him to renewed efforts. Then the banderillas de
74. fuego are called for. These are arrows, provided with fire crackers, which
explode the moment they are affixed in the neck. In a moment the spectacle,
which had worked me up to a high pitch of excitement, becomes intensely
distasteful. The tortured animal, driven mad with fright and pain, bounds
across the ring in a series of leaps like a kid. The people scream with delight,
and I mentally wonder what kind of “steadier” the Spaniard resorts to when
his stomachic nerve is affected by a detail of the exhibition. The firework
display had not lasted long when the last trumpet sounded, and the espada
walks forward to a storm of rapturous applause.
The finale of the spectacle is approaching. The executioner comes alone:
the bull, who has hitherto been tormented by a crowd of enemies, is now
able to concentrate his whole attention on one object. Toro has become
exhausted with his previous exertions, and he moves without his old dash.
The espada studies his foe carefully, to judge the temper of the animal with
which he has to deal. With his left hand he waves the muleta—the red cloak
—to lure the beast into a few characteristic rushes and disclose his
disposition. If he is a dull, heavy bull, he will be despatched with the
beautiful half-volley; but if he proves himself a sly, dangerous customer, that
is cunning enough to run at the man, instead of at the muleta, a less
picturesque, but safer thrust must be employed. But our bull is neither sly
nor leaden. He has recovered from his fright, and is quick to seize his
opportunity to make a final effort before the stinging banderilleros return to
distract him. Once or twice he thrusts his horns into the unresisting cloak,
then gathers himself together for a final rush. The swordsman raises the
point of his glimmering Toledo blade; while every nerve of his sinuous,
graceful body quivers with the absolute constraint and concentrated effort
that hold him. The duellists are both of the same mind. The espada has
summed up his antagonist—he is levantados, the bold bull, a fit subject for
la suerte de frente. The bull’s next rush is his last. The fencer receives the
charge on his sword, which enters just between the left shoulder and the
blade. The bull staggers, lurches heavily on to his knees, and rolls over, at
the feet of his conqueror, vomiting blood.
The assembled multitude rend the air with their cheers, the men yell
applause, and every face is distorted with excitement and enthusiasm. The
only indifferent person in the building is the espada. With a graceful and
unassertive turn of his wrist, he waves the sword over his fallen foe, wipes
the hot blood from the blade, and turning on his heel, approaches the
75. President’s box, and bows with admirable sang-froid. The team of jingling
mules enter, and the dead bull is carried off at a rapid gallop. The espada
walks composedly away, without another glance at the result of his
handiwork.
The superb imperturbability of these espadas always fills me with
admiration. They accept the plaudits of the spectators with the same
unconcern with which they hear the execrations that fill the air if they do not
at the first attempt inflict the coup de grace. During the first corrida I
attended, an espada failed to aim at the precise spot, and the bull tore up the
sand in agony. The populace insulted the swordsman with jeers and
howlings, but he remained perfectly cool and collected, and nerved himself
with as much composure to his second and successful thrust as if he had
been practising with a sack of potatoes in an empty arena. When I had been
witness to the death of two bulls, I remarked to my Spanish friend that I had
seen as much as I desired, and was quite ready to quit the spot. But my
companion was a friend of long standing: he could be firm without seeming
discourteous. “No! no!” he said, “you kept me in the theatre last night until
‘Don Juan’ was played to the bitter end: you shall remain to-day to reward
me for my exemplary patience and respect for your wishes.” I saw five other
bulls done to death during the afternoon.
AT CLOSE QUARTERS.
Although not to be compared with an ordinary corrida as a display of
skill, and capacity, and artistic finish, a Royal bull-fight, such as Madrid saw
on the occasion of the coronation of King Alfonso XIII., is more interesting
as being a revival of the sport as it was originally practised. Bull-fighting to-
day is a purely professional business, but in the knightly days of ancient
Spain it was employed as a means to teach the chivalrous youth the use of
76. arms. In those days, mounted caballeros encountered the bulls in the ring
with lances alone—a more dangerous pastime than is bull-fighting in its
modern sufficiently hazardous form. Then the combatants were mounted on
good horses, and their business was to save them and turn the bull, to kill the
bull if possible, but, at the risk of their own lives, to protect their steeds from
injury. It is recorded that in one Fiesta de Toros at the beginning of the
sixteenth century, no less than ten young knights lost their lives. The corrida,
Real con Caballeros en plaza—a Royal bull-fight with gentlemen in the
arena—on the olden lines, that was held on May 21st, 1902, in Madrid, was
fought by young officers and scions of noble families, who were attired in
the gorgeous costumes of Spanish knights of the reign of Philip IV., and
attended by their pages and grooms wearing the dress of the same period,
and displaying the colours of the noble house which they served. On that
occasion, the Paseo de las Cuadrillas, or preliminary procession of the bull-
fighters across the arena to the strains of military music, was a most
imposing sight. The Padrinos, the grandees who acted as supporters or
godfathers of the knights, accompanied the fighters, followed by their
mediævally-clad retinues, to the foot of the Royal box, and presented them
to the King. The spectacle was strikingly brilliant, but the display was not to
be compared with a professional bout. The horses of the cavaliers had
evidently not been sufficiently trained for their work, and the best riding in
the world could not bring them off scathless. Let me condense an account of
the scene to convey an impression of what the present-day bull-fight has
been derived from.
When the procession had withdrawn, leaving only the chulos and the
gallant caballeros in the arena, the door of the toril swung on its heavy
hinges, and a splendid specimen of a bull, dungeoned for several hours
previously in utter darkness, darted into the light of day, tearing up the
ground with its hoofs, and ploughing the air with its horns. Suddenly, a
horseman and his prancing steed vaulted into the centre of the ring—the
charger, with flowing mane, full-veined ears and shapely head slanted
forward—to meet the onrush of the goaded bull. The second picador seeing
the bull worried and dazed by the tantalising assistants, scudded past on a
swift, white racer, sitting gracefully in his saddle, and then turning deftly as
he passed the great brute, plunged his lance into his neck, and whirled aside
to avoid possible pursuit. But by sheer accident, the bleeding steer dashed
77. off in the same direction, caught the horse in the hindquarters, raising it on
its forelegs and endangering the equilibrium of the rider.
Before the scampering bull had time to recover from the compact, the
second caballero, dashing up, had planted his lance deep into its neck. The
white horse, stung with pain, made a wild rush, but was brought to hand by
splendid horsemanship, and his rider urged him along, to inflict another
wound in the animal’s head. Then two toreros advanced, beguiling and
wearying the bull. By the time the bull had received the fifth lance in his
neck, and the white steed had been twice wounded, the edge was taken off
the keen thirst for violent emotions, and another torero unfolded his red
capa, waved it to and fro until the bull swooped down upon him, and a
moment later he was sprawling in the sand seemingly gored by the infuriated
animal. The next minute the wounded steer tottered, dropped on its forelegs,
and turned over on the sand, and a knife put a speedy end to its sufferings.
The second bull, a black massive creature, appeared listless and faint, and
made little effort to defend itself. It made one successful attack on the white
charger; and, then, at the signal from the King, an amateur espada stepped
forward. The attempt was a miserable failure. The young swordsman
dedicated, in a few well-chosen words, the death of the bull to his sovereign,
and after a dozen passes with the red capa, plunged the gleaming blade of
Toledo steel into the animal’s neck, but so ineffectually that a storm of hisses
resounded through the ring. The second attempt was still more awkward, the
sword entering but a few inches. The sword was pulled out, and another
effort, made amid groans and hisses, proved equally unsuccessful.
A TURN WITH HIS BACK TO THE BULL.
Although the madness had died out of the expiring brute’s eyes, and his
forelegs were bending under him, the inexperienced torero seemed unable to
78. put him out of pain. However, he grasped the short, sharp knife, and
unsteadily taking aim, plunged it into the neck. Another failure. Yells,
groans, shrieks, whistling, and hissing marked the anger of the crowd. The
espada may be a paid professional, or the greatest noble in Spain, but in the
ring he is judged by the rules of the ring, and his bungling is recognised with
the most poignant scorn to which failure could be subjected. He again
grasped the sword; and, spurred by the vitriolic exclamations of the public,
sheathed it in the bull’s neck. The animal stood still and tottered, his forelegs
bent, his head sank upon the moist, red sand, his hind feet quivered, and a
flourish of trumpets announced that life was extinct.
It is curious to find, in talking with learned enthusiasts on the relative
merits of the bull-fighters, what diversity of opinion exists; but all parties are
agreed upon the unrivalled skill and daring of the mighty Frascuelo. In his
day, for death’s whistle summoned him from the arena in the height of his
fame, Frascuelo was regarded as the greatest matador that Spain had ever
seen; and Spaniards, in debating the subject of the bull-ring, never indulge
the hope that his equal will ever arise to shed a new glory on the National
sport. Frascuelo is dead, and his famous rival, Guerra, or Guerrita—to give
him his professional name—has long since cut off his coleta, and lives in
well-earned retirement at Córdova. But the school of fighters, who claim
Frascuelo as their master—the fearless, dare-devil toreros, who scorn to
concede a yard of ground to the bull, and do all their fighting at close
quarters—is widely popular; and if their terribly dangerous methods are
attended by frequent casualties, the intoxicating applause that rewards the
accomplishment of a brilliant coup is, apparently, ample compensation for
the risks that it entails. But the wildest appreciation of a successful feat does
not exempt the most popular performer from the furious condemnation of
the multitude when his scheme miscarries. The allowances made by a
Spanish audience at the ring-side are of the most grudging nature. I once
travelled from Barcelona to Madrid in the company of Bombita-Chico—the
boy Bombita—who, although he was barely recovered from an unfortunate
encounter with a tricky bull eight days before, was on his way to take part in
a grand corrida that was to be held in the capital. He was—as his name
denotes—no more than a lad, with large, strong hands that sparkled with
jewels, while a formidable anchor about five inches long, set with
magnificent diamonds, dangled from his watch-chain. I saw him again in the
arena a few days later. He seemed nervous, and was, it appeared to me, a
79. little perturbed by the demonstration that welcomed his reappearance in the
ring after his accident. Ill fortune allotted him a troublesome animal, and his
kill, while creditable enough to untutored eyes, lacked the grace and finish
that the critical spectator requires. Bombita was their own Boy of Madrid,
and because of his recent misfortune they forgave him, but they did not
cheer him; and the lad walked out of the arena amid a silence that could be
felt.
FIXING THE BANDERILLAS.
Mazantini, now grown old and heavy, was in his day an undoubtedly fine
matador. There are some that still regard him as the head of his profession.
But the majority, remembering what he was, regret that he has not gone into
honourable retirement. But Mazantini cannot tear himself away from the
fascination of the arena, although his appearances grow less frequent every
year. Conejito, who was wounded in Barcelona in the spring of 1903, is
generally regarded as the most accomplished matador now before the public;
but Fuentes is, par excellence, the best all-round man. For, with the
exception of the picador business, Fuentes plays every part in the piece.
Other espadas have their assistants, who play the bull with their capas, and
stand by while the banderilleros ply their infuriating darts. It is only when
the bull has been prepared for the slaughter by the other performers that the
matador comes forward to put the finishing touch to the grim tragedy.
Fuentes, on the other hand, on special occasions—of which the corrida
which I attended in Madrid was one—keeps his assistants entirely in the
background; he takes the stage when the picadores leave it, and keeps it to
the end. So close does he keep to the bull, that during the corrida in Madrid,
of which I am writing, he seldom allowed the animal to be a dart’s length
away from him. On one occasion his capa got caught so tightly on the bull’s
80. horns that he tore it in jerking it away; and at another time the bull stopped
dead, with his forefeet on the hated sash. As a banderillero, Fuentes is
without equal in Spain. He frequently works with darts that have previously
been broken short, and he uses them sparingly. Yet the encounter between
the banderillero and the bull when Fuentes is on the scene is the most
thrilling part of the whole performance. It is a contest between human
intellect and brute intelligence—a duel between mind and matter. Fuentes
does not avoid the bull, but by exerting some magnetic power he repulses the
animal and compels it to halt. When the bull charges, in response to his
“defiance,” he waits with the banderillas suspended above his head until the
animal is within a few yards of him. Then he deliberately, but without haste,
lowers one arm until the arrow is on a level with the brute’s eyes. The bull
wavers in his onslaught, slows up, and stops dead within a foot or two of the
point. Sometimes Fuentes walks backwards, while the bull glares at him
with stupefied impotence, until he escapes the eyes that
THE MATADOR.
hold him, and gallops away. Again and again the banderillero taunts his
enemy to attack him, only to arrest his charge and force him to turn from his
deadly purpose by the irresistible power of his superior mentality. The crowd
follows this superb exhibition with breathless interest, and in a silence that is
more eloquent of admiration than the wildest cheers would be. But the end is
nearly reached. Fuentes grasps his stumpy darts and advances against his
bewildered antagonist, who waits his approach with sulky indifference. The
man’s arms are flung up with a gesture of exasperating defiance, and when
the bull makes his final rush, his opponent, instead of stopping him, steps
lithely on one side, and the brute thunders past him with the two galling
arrows firmly implanted in his huge neck. Fuentes has already moved to the
81. side of the ring. The bull turns and charges back at him. The banderillero
glides gracefully over the sand, but his pace is not equal to that of his
infuriated pursuer. The distance between them decreases rapidly; in half-a-
dozen yards he will be upon him. Fuentes glances over his shoulder and,
without changing his pace, doffs his cap and flings it in the bull’s face. This
stratagem only arrests the rush of the brute for a moment, but it gives the
man time to reach the barrier, where he receives his muleta and sword from
an attendant and returns to complete his task.
All the kings of the bull-ring have their own particular feats or strokes,
which the Spaniards appreciate as Englishmen revel in Ranjitsinhji’s
acrobatic hitting, or Morny Cannon’s inimitable “finishes.” Bombita-Chico’s
speciality in playing his bull is to kneel in the arena and allow the animal to
charge through the capa which is held within three feet of the ground. The
nerve required for this feat fires the audience with enthusiastic approval. The
tale is told of a torero, whose name I have forgotten, who gained distinction
by his exceptional skill in facing the bull with the long vaulting pole, known
as the salto de la garrocha. With this instrument he would goad the bull on
to the attack. When the brute was in full gallop he would, timing his
movements to the instant, run a few yards to meet him, and swing himself
high into the air at the end of his pole. The oncoming bull would charge the
pole, the grounded end would be tossed upwards, and the torero would drop
lightly to the ground and make good his escape. On one occasion the man
performed his risky “turn” at a moment when the attention of a royal lady
was attracted from the arena, and she sent an attendant to the expert to
command him to repeat it. In vain the poor fellow protested that it was
impossible for him to accomplish the same feat again with the same bull.
The lady’s desire had been expressed. “But it is more than my life is worth,”
argued the athlete. “It is the lady’s wish,” responded the attendant. The
torero bowed, and “I dedicate my life to Her Royal Highness,” he said. The
attempt fell out as he foretold. The bull charged and stopped dead. The man
vaulted aloft, his body described a half circle, and fell—on the horns of the
bull. He was dead before the attendants could entice the animal from his
victim.
82. THE FINAL STROKE.
Lagartijo, Lagartijillo, Mazantini, and Montes all have their
distinguishing methods of attacking and despatching the bull, but none of
these are capable of the feat by which Guerrita was wont to throw the bull-
ring into transports of deafening enthusiasm. In the ordinary way, the espada
having taken the measure of his adversary, receives him standing sideways,
and having thrust his sword at arm’s length between the left shoulder and the
blade, leaps aside as the bull blunders forward on to his knees and falls to the
earth. But Guerrita advanced his left arm across his body and waved his
muleta under his right uplifted arm. When the bull lowered his head at the
charge he passed the sword over the animal’s horns and plunged the blade
into the vital spot behind the shoulder. In other words, he stopped the brute
and killed him while his head was under his arm; and so closely were the
duellists locked in that last embrace, that Guerrita’s side was frequently
scratched by the bull’s horns. One may lecture, write, and preach against the
barbarity of bull-fighting; but so long as Spain can breed men of such
amazing nerve, and skill, and dexterity that they can successfully defy death
and mutilation to provide their countrymen with such lurid sport, so long
will bull-fighting continue to flourish in Spain.
83. Welcome to our website – the ideal destination for book lovers and
knowledge seekers. With a mission to inspire endlessly, we offer a
vast collection of books, ranging from classic literary works to
specialized publications, self-development books, and children's
literature. Each book is a new journey of discovery, expanding
knowledge and enriching the soul of the reade
Our website is not just a platform for buying books, but a bridge
connecting readers to the timeless values of culture and wisdom. With
an elegant, user-friendly interface and an intelligent search system,
we are committed to providing a quick and convenient shopping
experience. Additionally, our special promotions and home delivery
services ensure that you save time and fully enjoy the joy of reading.
Let us accompany you on the journey of exploring knowledge and
personal growth!
ebookultra.com