Vous êtes sur la page 1sur 14

J Struct Funct Genomics (2013) 14:7790

DOI 10.1007/s10969-013-9160-z

Cross-linking and mass spectrometry methodologies to facilitate


structural biology: finding a path through the maze
Eric D. Merkley John R. Cort Joshua N. Adkins

Received: 15 March 2013 / Accepted: 26 July 2013 / Published online: 7 August 2013
Springer Science+Business Media Dordrecht (outside the USA) 2013

Abstract Multiprotein complexes, rather than individual IGBP1 Immunoglobulin binding protein 1
proteins, make up a large part of the biological macro- IMS Ion mobility spectrometry
molecular machinery of a cell. Understanding the structure LC Liquid chromatography
and organization of these complexes is critical to under- MIX Mixed-isotope cross-linking
standing cellular function. Chemical cross-linking coupled MS Mass spectrometry
with mass spectrometry is emerging as a complementary MS1 Precursor scan in a mass spectrometry
technique to traditional structural biology methods and can experiment
provide low-resolution structural information for a multi- MS2 Tandem mass spectrometry (MS/MS) scan
tude of purposes, such as distance constraints in compu- in a mass spectrometry experiment that
tational modeling of protein complexes. In this review, we yields a fragmentation spectrum of a
discuss the experimental considerations for successful precursor selected in MS1
application of chemical cross-linking-mass spectrometry in MS3 Third-order tandem mass spectrometry (MS/
biological studies and highlight three examples of such MS/MS) scan that yields a fragmentation
studies from the recent literature. These examples (as well spectrum of an MS2-derived fragment ion
as many others) illustrate the utility of a chemical cross- NHS N-hydroxysuccinimide
linking-mass spectrometry approach in facilitating struc- NMR Nuclear magnetic resonance spectroscopy
tural analysis of large and challenging complexes. PLRV Potato leaf roll virus
PP2A Protein phosphatase 2A
Keywords Chemical cross-linking  Mass PP2AA The A subunit of protein phosphatase 2A
spectrometry  Structural biology  Protein complexes  ReACT Real-time analysis for cross-linked peptide
Computational modeling  Integrative structural technology
biology RTP PLRV read-through protein
SCX Strong cation exchange chromatography
Abbreviations SDS-PAGE Sodium dodecylsulfate polyacrylamide gel
AP-MS Affinity purification-mass spectrometry electrophoresis
BS3 Bis[sulfosuccinimidyl]suberate SEC Size exclusion exchange chromatography
CID Collision-induced dissociation XLMS Chemical cross-linking-mass spectrometry
CP PLRV coat protein
DSS Disuccinimidyl suberate
FDR False discovery rate
Introduction

E. D. Merkley (&)  J. R. Cort  J. N. Adkins


From simple homodimeric enzymes to massive multipro-
MS K8-98, Biological Sciences Division, Pacific Northwest
National Laboratory, Richland, WA 99352, USA tein/nucleic acid complexes such as ribosomes or viruses,
e-mail: Eric.Merkley@pnnl.gov biological processes are carried out by dynamic protein

123
78 E. D. Merkley et al.

complexes. Understanding the quaternary structure and The purpose of this review is to provide sufficient
dynamics of these protein complexes is critical to under- background for structural biologists to better understand
standing cellular function. To date, NMR spectroscopy and the range of XLMS methods available, including their
X-ray crystallography have been the analytical workhorses respective capabilities and limitations, and their utility in
for determining protein structures, as evidenced by the structural studies. In the following sections, we identify the
*80,000 protein structures presently stored in the Protein challenges associated with XLMS, considerations and
Data Bank. However, the larger, more heterogeneous, and/or choices at each step of a basic XLMS workflow (and
more dynamic protein complexes, including membrane some selected variations upon it), and highlight three
proteins and complexes whose composition and conforma- examples that illustrate different combinations of methods
tion change as an integral part of their function, are often not that can lead to new structural insights. Note that we do not
amenable to traditional NMR or crystallographic analysis. include a comprehensive survey of the cross-linking liter-
Studying multiprotein complexes requires a toolbox of ature in this review as it already has been ably reported by
alternative structural biology methods that complement the several leaders in the field [1216].
traditional methods [1]. Alternative methods include well-
known techniques such as small-angle X-ray scattering [2],
single-particle cryoelectron microscopy [3, 4] and analyt- Recognizing the challenges
ical ultracentrifugation [5], as well as several mass spec-
trometric techniques [6] that may be less familiar to the By chemical-cross linking of proteins, we refer to the
structural biology community, such as covalent modifica- introduction of a covalent linkage (the cross-link) between
tion/chemical footprinting [7], ion mobility spectrometry two distinct protein sites, either intramolecular or inter-
[8], native mass spectrometry (MS) of protein complexes molecular, by reaction with a bifunctional reagent (the
[9], and chemical cross-linking-MS (XLMS), the focus of cross-linker). The distance between cross-linked sites is
this review. Used in various combinations with each other constrained according to the chemical and physical prop-
and with atomic-resolution structures of domains and erties of the protein(s) and cross-linker. It is important at
subunits from crystallography and NMR spectroscopy, this the outset to understand what XLMS can offer and what it
suite of techniques allows for detailed characterization of cannot. XLMS can complement and extend NMR and
large multiprotein complexes that are inaccessible to crystallographic structural studies, and it is often feasible
crystallography or NMR alone. Increasingly, these tech- when these other methods are not. In addition, XLMS
niques are being combined with computational modeling of data can be used as distance restraints in structural mod-
protein structures to provide accurate structural models, an eling, and as a method for evaluating and filtering com-
approach that is coming to be known as integrative struc- putational models of protein structure. However, XLMS is
tural biology [1, 10, 11]. not in itself a technique for determining atomic-resolution
While XLMS can complement NMR and crystallo- structures.
graphic structural studies, the present capabilities and Although XLMS relies on the same fundamental
limitations of XLMS are generally appreciated by only a technology as MS-based proteomics, the complexity of a
small subset of the proteomics/MS community. As with digest of a cross-linked protein along with the scarcity of
many developing methods, a consensus approach to con- cross-linked peptides in the mixture pose formidable ana-
ducting XLMS experiments and data analysis has yet to lytical and informatics challenges. First, the analytical
emerge. This lack of a clear consensus has been an obstacle platform must have sufficient sensitivity and dynamic
to wider application of XLMS technologies as a non- range to deal with the complexity of a digest of a cross-
expert is faced with many interdependent choices of linked protein sample. Such a sample contains far more
reagents, protocols, and software. Still, recent develop- unique chemical species than a digest of the non-cross-
ments have led to increasingly robust XLMS methods that linked protein, because the number of potential cross-
can generate useful structural insights. For example, vari- linked peptides increases quadratically with the sequence
ations on the basic XLMS approach have been used to length. In addition, cross-linking produces many peptides
study a number of multiprotein complexes, including the modified by a hydrolyzed linker (that is, one end of the
Salmonella type 3 secretion system needle complex [53, linker reacts with the protein and the other with water; this
57], archaeal [24] and eukaryotic proteosomes [3, 27, 67], product is known as a dead-end), as well as the unmodified
yeast transcription initiation complexes [6, 38], and even peptides, creating a highly complex mixture [17].
the prokaryotic ribosome [28]. XLMS also has been used Most often the linker/protein ratio is kept low to avoid
to study proteinprotein interactions in live cells [37], and artifacts such as cross-link-induced polymerization (which
to study the structure of affinity-purified [19] and immu- can lead to the detection of cross-linked peptides that can
noprecipitated [30] complexes. only form in the artifactual polymer) or other structural

123
Cross-linking mass spectrometry and structural biology 79

perturbation. Therefore, cross-linked peptides are a very to overcome the inherent complexities of XLMS samples
minor component of this complex mixture, sometimes two and data. To be successful, a researcher must be able to
orders of magnitude less abundant than other components. navigate this maze by selecting an appropriate combination
Ionization suppression, a well-known process by which of techniques for each step of the workflow. The following
easily-ionized peptides decrease the observed signal of sections highlight the considerations and choices at each
other peptides, and data-dependent analysis, in which step.
peptide ions observed during the LCMS analysis are
typically selected for fragmentation in order of abundance, Cross-linker reagents
can both increase the difficulty of characterizing low-
abundance cross-linked peptides. A large number of cross-linking reagents have been descri-
The high complexity of XLMS samples also creates bed in the literature (reviews of cross-linker chemistries are
informatics challenges. Database search algorithms work given in references [13] and [20]), and many are commer-
by comparing the observed tandem mass spectrum to the cially available. The most important considerations in
theoretical spectra of all possible peptides within a given selecting a cross-linker are chemical specificity, which
mass range. Increased sample complexity means increased determines the functional groups targeted by the linker, and
degeneracy of peptide masses (within the mass accuracy of linker-arm length. The most commonly used cross-linkers
the instrument) and thus an increased chance of an incor- are the amine-reactive N-hydroxysuccinimide (NHS) esters,
rect identification. Further, the complex fragmentation such as disuccinimidyl suberate (DSS) and its more soluble
patterns present in MS/MS spectra of cross-linked peptides sulfonate analogue bis[sulfosuccinimidyl]suberate (BS3).
necessitate specialized informatics approaches [18]. The NHS esters have the advantage of high specificity and of
informatics issue has until recently been the greatest hurdle targeting a relatively common residue in proteins (lysine).
to widespread application of XLMS. The disadvantages of NHS esters include the tendency to
Successful XLMS workflows must address these chal- hydrolyze in water, and the need for slightly basic reaction
lenges through a combination of sample preparation, conditions which may not be appropriate for all proteins, and
instrumental analysis, and informatics techniques; for the secondary reactivity toward hydroxyl groups. Other
example, using chromatographic enrichment of cross-linked commonly used chemistries include imidoester (which also
peptides to reduce sample complexity, excluding low charge targets amines) and maleimide and thiol (both of which
state peptide ions from fragmentation to increase the sensi- target cysteine residues). The length of the linker determines
tivity for detecting low-abundance cross-linked peptides, the precision of the distance restraint implied by an observed
etc. The diversity of approaches at each stage of the XLMS cross-link, but the main effect of decreasing linker length is
process has led to a profusion of methods described in the to decrease the number of cross-links observed [21]. The
literature. However, without some understanding of the linker length of the DSS and BS3 is *11.4 A, corresponding
interdependent methods, the XLMS field as a whole can be to an eight-carbon aliphatic chain. Lengths of other common
difficult to navigate, especially for the non-expert. reagents range from 8 to 18 A. In addition, many cross-
linking reagents have special features, such as biotin moie-
ties for avidin affinity enrichment, stable isotope labels
Navigating the XLMS methodological maze (mixed heavy and light reagents provide characteristic mass
spectrometric doublets), or labile bonds that are cleavable by
The basic XLMS approach has changed little since its collision-induced dissociation (CID) in the mass spectrom-
introduction by Young and coworkers 13 years ago eter. Use of these CID-cleavable reagents can greatly
(Fig. 1a) [19]. A protein or protein complex is treated with simplify data analysis (see below).
a bifunctional cross-linking reagent that introduces cova-
lent linkages between specific functional groups in the Optimizing the cross-linking reaction
protein. The cross-linked protein is then digested with a
protease, and the resulting mixture is analyzed by liquid Cross-linking reaction conditions must preserve the native
chromatography (LC)-MS/MS to identify cross-linked structure of the protein complex. Excessive cross-linking can
peptides and determine their sequences. The locations of lead to structural distortion, artifactual oligomerization or
cross-links provide structural information about the system aggregation, and precipitation. To avoid these effects, opti-
under study. mal conditions (such as protein and linker concentration,
Despite this conceptual simplicity, the field to date has cross-linker/protein ratio, reaction time, and temperature)
resembled a methodological maze, with a multitude of should be determined empirically. Ideal conditions are spe-
options available for every aspect of sample preparation, cific to the protein or protein complex being studied, but for
data acquisition, and data analysis (Fig. 1b), each intended NHS ester cross-linkers, typical final conditions are low

123
80 E. D. Merkley et al.

Fig. 1 The generalized cross-linking mass spectrometry (XLMS) search method similar to those used in proteomics. The resulting
workflow. a the essential elements of the XLMS approach, cross-links provide distance constraints for use in structural analysis,
indicating the issues to be considered at each step. The protein or including docking and modeling. b there are many options available
protein complex of interest is cross-linked and digested with a at each step of the XLMS workflow. Sometimes these options can be
protease. Cross-linked peptides, which under optimal conditions are freely interchanged in a modular fashion, but in other cases, methods
minor products of the cross-linking reaction, may be chromatograph- used in one step of the workflow require or enable methods in another
ically enriched. The cross-linked peptides are then analyzed by liquid- step. For instance, mass-spectrometry cleavable cross-linkers are
chromatography/tandem mass spectrometry. Specialized software necessary in order to use the multistage MS/MS data acquisition and
identifies MS/MS spectra of cross-linked peptides by a database analysis strategy (see text and Fig. 2 for details)

micromolar protein concentration, 20- to 1,000-fold molar before digestion (for example, see [22, 23]). In some cases, it
excess cross-linker, and reaction times of *30 min at room may be desirable to complete the entire cross-linking
temperature, or 2 h on ice. The reaction buffer must be workflow for several different sets of reaction conditions.
devoid of amines, as these will react with the cross-linker, Agreement between cross-links observed in different con-
thereby excluding many common biological buffers such as ditions is evidence that they are structurally relevant and not
Tris. A commonly-used compatible buffer is phosphate- caused by cross-linker-induced structural perturbations [14].
buffered saline. Excess cross-linker is quenched with an Circular dichroism spectroscopy can be used to assess
amine-containing reagent (e.g., Tris, ammonium bicarbon- whether the cross-linking reaction has introduced unac-
ate, or glycine) and/or removed by buffer exchange. (See the ceptable perturbations of the secondary structure. If
suggested protocols from one supplier of BS3/DSS at http:// unwanted oligomerization occurs, the target quaternary
www.piercenet.com/instructions/2160418.pdf). structure can sometimes be purified by SDS-PAGE (either by
The reaction is optimized by varying these conditions and excising bands from an analytical gel or by preparative SDS-
by analyzing the reaction mixtures using SDS-PAGE or MS PAGE) or size-exclusion chromatography [19].

123
Cross-linking mass spectrometry and structural biology 81

Digestions of cross-linked protein mixtures variety of systems. While effective, this method has two
with proteases main drawbacks: First, dead-ends are also enriched, since
they are also biotinylated. Second, these cross-linkers are
Denaturation and protease digestion is the next step in the sometimes very long ([30 A instead of the 818 A of more
XLMS workflow. It is possible that the presence of cross- conventional reagents), and the increased bulk of the linker
links hinders protease digestion, but typical protocols in the reagent may prevent the close approach of the linker
literature are similar to protocols used for digesting non- molecule to the reactive lysine residues, preventing cross-
cross-linked proteins or proteomes (e.g., trypsin: protein link formation by steric hindrance. The long linker also
mass ratio of 3050 and digestion times of 316 h at reduces the spatial resolution of the structural information
37 C). Trypsin is the most commonly used enzyme for obtained. To overcome the steric bulk issue, our laboratory
cross-linking studies, as it is for proteomics in general. has developed the CLIP cross-linker [36]. In this approach,
Other endopeptidases have been used, either alone or in the cross-linker contains an alkyne function that can be
combination with trypsin, including chymotrypsin, GluC, ligated to a biotin-containing azide reagent after the cross-
AspN and LysC [22, 24], and the chemical agent cyanogen linking step is complete. Since the alkyne-containing linker
bromide [25]. The use of multiple proteases was shown to is much smaller than many biotinylated cross-linkers, the
improve detection of cross-linked peptides, probably by potential for steric hindrance of the cross-linker/protein
increasing the number of protease cleavage sites in lysine- interaction is minimized. The drawback of this approach is
and arginine-poor regions of the sequence [24]. that the additional chemical modification and purification
steps can result in significant sample loss.
Enrichment of cross-linked peptides
LCMS analysis
The most useful structural information from an XLMS
experiment comes from interpeptide cross-links (i.e. those The liquid chromatography separations for cross-linked
between peptides as opposed to a cross-link between two peptides are similar to those used in typical proteomics
sites within the same peptide). However, interpeptide cross- experimentsmainly C18 or other reversed-phase separa-
links are the least abundant product of a XLMS experiment, tions. The separation can occur concurrently with the MS
being far outnumbered by dead-ends and unmodified pep- measurement (online) or before the MS measurement (off-
tides. To improve detection of these cross-linked peptides, line). A wide variety of mass spectrometric instrumentation
some form of enrichment is desirable. When dealing with and methods have been used for XLMS. Recent successful
complex samples (i.e., a large number of proteins), enrich- XLMS studies use in combination: (1) selection of high-
ment becomes essential. (Complexity is a function of total charge state precursor ions for fragmentation; (2) high res-
sequence length of all proteins in the sample.) The three main olution mass measurements of precursors; and (3) either high
enrichment methods are strong cation exchange chroma- resolution measurements of fragmentation spectra or the use
tography (SCX), size exclusion chromatography (SEC), and of CID-cleavable cross-linkers combined with an appropri-
biotin/avidin affinity chromatography. SCX, in which sep- ate multistage tandem MS data acquisition scheme, such as
aration is based on electrostatic charge [26, 27], is the most the real-time analysis for cross-linked peptide technology
frequently used enrichment strategy. SCX enriches tryptic (ReACT) method of Weisbrod et al. [37] (Fig. 2a). In MS
cross-linked peptides because they carry a greater positive parlance, the initial mass measurement or precursor scan is
charge than non-cross-linked peptides: a positively charged called MS1, selection and fragmentation of a single precursor
peptide N-terminus and a positively charged lysine or argi- ion scan is called tandem MS, MS/MS or MS2, and sub-
nine residue for each peptide in the cross-link. (In addition, sequent rounds of selection and fragmentation are called
some linkers preserve the positive charge on the cross-linked multistage tandem MS or MS3, etc. High resolution MS1
lysine residues [28] or themselves carry a positive charge scans are essential for XLMS, because the high-resolution
[29]). Since cross-linked peptides are also larger than regular mass spectrum allows for determination of the charge state of
peptides, peptide-level SEC has also been used [24, 30]. SEC the peptide, allowing low-charge species to be ignored, and
may be more effective with proteases that cleave less fre- because the accompanying mass accuracy dramatically
quently and create longer peptides, such as lysyl endopep- limits the number of potential precursors that must be con-
tidase (which cleaves after lysine residues) rather than sidered in database searching. The large size and high charge
trypsin (which cleaves after lysine and arginine residues). states of cross-linked peptides mean that a very large number
A much different strategy uses immobilized avidin to of fragments could potentially be observed in MS2 spectra
capture peptides that have been modified with a biotin- (fragments may occur in every charge state up to the charge
containing cross-linker. The research groups of Bruce [31, state of the precursor). High resolution MS2 data allow
32] and others [3335] have used this technique on a determination of fragment ion charge states and accurate

123
82 E. D. Merkley et al.

Fig. 2 Cross-linking the tomato leaf roll virus capsid with protein energy to fragment the peptide backbones (4). Sequences of the
interaction reporter (PIR)/real-time analysis for cross-linked peptide component peptides are determined by database searching using
technology (ReACT) technology. a The PIR multistage data acqui- standard proteomics software tools such as Sequest or Mascot. If the
sition strategy. (1) A potential cross-linked peptide (p) is identified in mass relationship is not satisfied within a tight mass tolerance, the
a high-resolution MS1 scan by its high charge state. (2) MS/MS at low next precursor ion is selected or the next MS1 scan is acquired (1).
energy causes the labile bonds of the PIR reagent (shown in red in b) b The biotinylated Rink (BRink) PIR reagent. Blue, NHS-ester
to break, yielding a reporter ion (r) and the two separated component leaving groups; green, biotin affinity label; orange, mass spectrom-
peptides (a and b), each modified by a linker-derived stump etry cleavable bonds; green and black; reporter ion fragment;
(magenta in b). (3) If the masses of the a and b and r fragments add magenta, the stump atoms that remain attached to the peptides
up to the mass of p observed in (2), then the instrument control after MS2. c Network of cross-linking sites observed in the virus
software triggers two MS3 scans (one each for a and b) at higher capsid. Orange, read-through protein; blue, coat protein. See Ref. [32]

masses, greatly increasing confidence in the identifications. opposed to a proteome) that can be effectively searched. The
For methods that use CID-cleavable linkers and a multistage programs that are suited to large sequence databases include
data acquisition scheme (Fig. 2a), high resolution MS2 xQuest [38, 40], pLinks [41], and the updated ReACT/X-
spectra, arising from fragmentation of the linker only, are links program [32, 37, 42] for cleavable cross-linkers.
critical for confirming the mass relationships. However, in Historically, many of these programs have been used
this strategy, MS3 spectra are simply fragmentation spectra primarily by the research groups in which they were
of individual peptides and do not require high resolution developed. However, some recent studies have published
measurements. direct comparisons of the analysis of the same XLMS
datasets with several different programs [43, 44]. These
Data analysis: identifying cross-linked peptides studies, as well as our own evaluations (unpublished), show
that for small datasets (i.e., one or a few proteins) each
Numerous software applications have been developed to software package generally identifies the same cross-linked
assist in identifying cross-linked peptides from mass spec- peptides, with differences mostly due to differing default
trometric data (reviewed in [18, 21, 38]). All are elaborations parameters. However, as mentioned, not all software
of the basic approach that is well established in the proteo- packages can deal with large sequence databases.
mics field: matching the spectrum of a candidate cross-linked One of the greatest informatics challenges in XLMS is
peptide to a theoretical spectrum derived from a sequence judging the confidence of a cross-linked peptide-spectrum
database and providing a score for the quality of the match match [12]. Traditional proteomics accomplishes this by
[39]. The differences among the various algorithms are estimating the false discovery rate (FDR) [45], which is
mainly in the way that the match is scored and in the size of simply the proportion of matches expected to be false at a
the sequence database (i.e., one or few small proteins as given score threshold. FDR is estimated by the target-

123
Cross-linking mass spectrometry and structural biology 83

decoy approach (reviewed in [46]). In this procedure, the 53], including the cross-linker known as BRink ([53],
target sequences are the sequences expected to be present Fig. 2b), which contains two NHS esters as the lysine-reac-
in the sample. The decoy sequences are sequences known tive groups, a biotin moiety for enrichment, and two CID
not to be present in the sample, often reversed or ran- cleavable bonds. Tryptic cross-linked peptides enriched by
domized target protein sequences. The FDR is estimated as SCX fractionation and immobilized avidin affinity chroma-
the percentage of sequences meeting the score criteria that tography were analyzed by LCMS using the CID-cleavable
are decoys. Several XLMS studies have reported an FDR multistage strategy (Fig. 2a). In this approach, the identifi-
estimate [27, 42, 4749]; however, this has not been done cation of a cross-linked peptide depends on the mass rela-
consistently. In an important recent advance, Walzthoeni tionship between the precursor, the cross-linker-derived
et al. [38] and Yang et al. [41] have shown how to calculate reporter ion fragment, and the two released cross-link pep-
a modified FDR estimate which accounts for hybrid target- tides. Using high-resolution instrumentation (Fourier trans-
decoy matches, in which one arm of the cross-linked form-ion cyclotron resonance MS) allows the mass
peptide is a target sequence and one is a decoy. We relationships to be determined with high accuracy, increasing
anticipate that an FDR estimate that accounts for hybrid the confidence of the identification. Another great strength of
target-decoy matches will become as much of a strong this approach is that the sequences of the individual peptides
requirement for XLMS studies as standard FDR calcula- in the cross-link can be identified using straightforward and
tions are for proteomics. well established database search methods (e.g., Sequest [54]
or Mascot [55]), avoiding the complexities associated with
cross-linked peptide database searches. The programs
Pathways to successful XLMS applications X-links [42] and BLinks [47] were developed to detect these
mass relationships in the LCMS data. Importantly, the
Here we present several examples of XLMS relevant to recently reported ReACT method [37], implements the
current problems in structural biology, each of which algorithm of X-links in real-time by integrating analysis of
illustrates a unique combination of methods (i.e., a path the PIR mass relationships and instrument control during the
through the methodological maze) that can lead to suc- LCMS experiment. If no PIR mass relationship is detected in
cessful identification of cross-linked peptides. These an MS2 spectrum as evaluated in real time during the mea-
examples are by no means exhaustive; rather, they were surement, no MS3 is performed and a new MS1 or MS2
selected to illustrate the range of methods available, the spectrum is acquired instead. This method focuses the anal-
types of problems that can be addressed, and the utility of ysis on probable cross-linked peptides, thus improving the
XLMS in structural biology. instrument duty cycle, increasing the number of cross-linked
peptides that can be detected, and partially overcoming the
Example 1: cross-linking virus capsids with the protein lower relative abundance of cross-linked species.
interaction reporter approach In a single LCMS experiment, Bruce and coworkers
collected ninety-two sets of MS1 ? MS2 spectra that sat-
Our first example illustrates XLMS analysis of the potato isfied the PIR mass relationship. For some of these mass
leaf roll virus (PLRV) capsid by the Bruce laboratory [32], relationships, satisfactory identifications from MS3 could
using the Protein Interaction Reporter approach. PLRV only be obtained for one of the released peptides. The sets
belongs to the luteovirus family, and is transmitted to of MS1 ? MS2 spectra that could be completely identified
plants via an aphid vector. Prior to this study, essentially no all belonged to four unique cross-linked peptides, each
experimental structural information was available for detected multiple times. MS3 sequencing revealed that
PLRV, except for knowledge of the icosahedral symmetry these four cross-linked peptides arose from modifications at
and T = 3 quasi-equivalence of the capsid. The capsid is five different sites: K188, K230, K351, K405, and K470,
thought to contain 180 copies of the coat protein (CP), with according to the network shown in Fig. 2c. A symmetrical
a small fraction of the CP molecules replaces by the read- cross-linked peptide containing K188 was derived from an
through protein (RTP) which consist of the CP plus a long intermolecular interaction, indicating that K188 is a point
C-terminal region predicted to be disordered. A homology of contact between CP monomers. Cross-links from K405
model had previously been generated using the CP crystal to both K430 and K188 were observed, suggesting that
structure from another virus [50], but the cross-linking both K188 and K430 are in proximity to K405 in the
results were the first experimental molecular-level struc- structure, or that two conformations of the RTP structure
tural information obtained for PLRV. were observed.
Custom cross-linking reagents called protein interaction Bruce and coworkers built a structural model of the
reporters (PIRs) were introduced by the Bruce laboratory in PLRV CP monomer [32] using Phyre2 [56]. Since the
2005 [51]. Several variant linkers have been described [52, expected symmetry of the icosahedral capsid includes a

123
84 E. D. Merkley et al.

three-fold axis, the CP monomer model was then used to between inter-protein and intra-protein cross-links from a
create a trimer model using Symmdock [57], which uses a homomultimer. The cross-linking strategies described so
shape-complementarity-based rigid-body docking algo- far in this review are not capable of making this distinction
rithm guided by cyclic symmetry arrangements. Inclusion unless the sequences of the two peptides are identical or
of a constraint derived from the K188-K188 homodimer overlapping, which decreases the quaternary structural
cross-linked peptide reduced the number of candidate information inherent in a given cross-link. Thus far, mixed
structures generated by Symmdock from [1,780 to just isotope cross-linking (MIX) is the only reported method
four. The top-scoring model was similar to known viral CP that can distinguish inter-molecular from intra-molecular
structures from T3 viruses. No confident models of the cross-links (Fig. 3a). First described by Taverner et al. [60]
RTP could be produced, which is unsurprising since the and used in a limited number of other published studies
RTP is predicted not to contain regular secondary structure. [17, 48, 61], this method uses isotopically labeled proteins.
Thus, the K230-K351, K405-K450, and K188-K450 cross- Specifically, normal isotopic abundance (light) and
links could not be directly used as distance constraints uniformly heavy-isotope labeled (heavy) multimeric
during modeling. However, the CP trimer model does place proteins are mixed in a 1:1 ratio and allowed to equilibrate
K188 in a surface position, accessible to cross-linking from such that mixed heavy/light complexes are formed. These
K405. Furthermore, all of the cross-linking sites are adja- mixed heavy/light complexes are cross-linked, enyzmati-
cent to the sites of known point mutants with virus trans- cally digested to create peptides, and analyzed by MS.
mission and/or infection phenotypes, which supports the Intramolecular cross-linked peptides appear as doublet
physiological relevance of the observed cross-links. peaks (light and heavy) in the mass spectrum.
Several features of this study are illustrative of the Intermolecular cross-links, however, appear as quadruplets
information available from XLMS. First, only four cross- of peaks (Fig. 3a), because four forms of the intermolec-
links were conclusively identified. This could be due to the ular cross-linked peptide exist (i.e., all light, all heavy, and
difficulty detecting and identifying the cross-linked pep- two mixed light/heavy versions).
tides, or to the structure of the protein complex itself. The This method has been used to identify intermolecular
number of lysine residue pairs (the BRink reagent is a cross-links in dimeric interleukin-6 [60], HIV integrase
lysine-selective NHS ester) closer than the linker length, [61], and Hsp21, a small oligomeric chaperone [17]. The
their solvent accessibility, and variations in their reactivity three intermolecular cross-linked peptides detected in the
(pKa) caused by local electrostatic effects [58] all influence interleukin-6 study revealed that the dimer is formed by
the likelihood of cross-linking occurring at a given site. In three-dimensional domain swapping via exchange of alpha
studies of proteins with known structure, the number of helices D and E. Knowledge of the intermolecular nature of
cross-links observed is typically far lower than the number these cross-links peptides was essential to ruling out other
of lysine residues potentially within the range of the cross- potential modes of domain swapping [60]. The case of
linker [21, 30, 59]. Therefore, no conclusions should be Hsp21 [17], which forms dodecamers consisting of 6
drawn based on the absence of evidence for cross-linking at dimers, is more complicated, since the nine detected
a given site. Second, this study illustrates the power of even intermolecular cross-links could arise from within-dimer
a single experimental restraint in evaluating computational interactions, or from interactions between adjacent dimers
models. While restraints such as cross-links do not guar- in the dodecamer. A cross-link spanning a distance that was
antee the accuracy of a computational model, they do evidently too great for the linker length in the dimer was
dramatically decrease the search space, thus grounding the attributed to an interaction between adjacent dimers in the
model in experimental data and improving outcomes [10]. dodecamer.
Third, cross-links provide the most useful information Two recent improvements to the MIX method [48]
when they can synergize with other types of data, whether include the use of uniform 13C,15N double-labeled proteins
theoretical or experimental. The K188-K188 cross-link as the heavy component of the mixed complexes and the
could be used as a distance restraint because homology addition of ion mobility spectrometry to the LCMS plat-
modeling was successful for the CP protein. form to enhance the analysis of the cross-linked peptides.
Note that other reported studies used only uniform 15N
Example 2: distinguishing intermolecular labeling. The double label has the advantage that the size of
from intramolecular interactions in homomultimeric the mass shift between the peaks of the multiplet is highly
complexes specific with respect to amino acid composition. The mass
shifts are determined by the number of incorporated heavy
Many protein complexes are either homomultimers or atoms. The number of nitrogen atoms per amino acid res-
contain multiple copies of a particular protein or proteins. idue is relatively constant across the 20 common amino
Our second example highlights an approach to distinguish acids. The number of carbon atoms is much less

123
Cross-linking mass spectrometry and structural biology 85

Fig. 3 Mixed isotope cross-linking (MIX) combined with ion and illustrating the advantages of IMS separation. Top, mass spectrum
mobility spectroscopy (IMS). a schematic of the MIX-IMS process averaged across the entire drift time range; middle, mass spectrum
showing the quadruplets in the m/z dimension. b histograms of the averaged across the drift time range indicated by the yellow lines;
predicted MIX mass shift values for tryptic peptides from an in silico bottom, ion-mobility-mass spectrum. Using MS alone, the heaviest
digest of the human proteome labeled with 15N, 13C, or both. Carbon peak of the quadruplet (arising from the cross-linked peptide with
plus nitrogen has the broadest and flattest distribution; thus, the MIX both arms heavy-isotope labeled), is obscured by an unrelated
mass shift from 13C ? 15N labeling provides the most powerful interfering peak. When IMS-MS is used, the interference is separated
constraint on sequence identification. For simple samples (a few from the cross-linked peptide signal in the drift time dimension,
proteins), the mass shift combined with the accurate mass can provide allowing recognition of the quadruplet pattern and identification of
unique or nearly unique identifications. c MIX-IMS-MS data for a the peptide. Data from Ref. [47]
cross-linked peptide, showing the characteristic quadruplet pattern,

degenerate, and the number of carbon plus nitrogen resi- long as it is not too large), one can infer the sequence of
dues is even more unique (Fig. 3b), which means that by cross-linked peptides without MS/MS measurements.
using the accurate mass and mass shift values, and a priori Ion mobility spectrometry is a gas-phase separation
knowledge of the sequence of the protein under study (as technique that separates analyte ions by their mobility (drift

123
86 E. D. Merkley et al.

time) as they migrate through an inert carrier gas under the AP-MS workflow. While bound to the stationary phase of
influence of an electric field [62]. By coupling this sepa- the second affinity purification step (nickel-NTA in this
ration technique with LC-MS, a two-dimensional ion- case), the tagged protein complexes were treated with the
mobility/mass spectrum can be obtained for every time cross-linker DSS as a 1:1 mixture of isotopically heavy
point during the LC elution. Heavy isotope labeling does (deuterated, d12) and light (d0) forms. Lysyl endopeptidase
not affect drift time [76], so the quadruplet signals of (LysC) digestion was also performed directly on the beads.
intermolecular cross-links align in the drift time dimension Peptides were fractionated by SEC to enrich for cross-
(Fig. 3a). This property allows unambiguous assignment of linked peptides. LCMS/MS data were searched with
signals belonging to a true quadruplet, as opposed to a pair xQuest [21, 40] using a database containing only proteins
of doublets, which could have the same appearance in a identified in the initial AP-MS experiment.
conventional mass spectrum. The use of an ion mobility While several proteinprotein interactions were char-
separation has increased the number of cross-links detected acterized in the PP2A study, we will review just one, the
and decreased the FDR for cross-linked peptides. The complex between the catalytic A subunit of PP2A (PP2AA)
greatest challenge of this approach is equilibration of the and the regulatory subunit immunoglobulin binding protein
heavy and light subunits into mixed complexes [63]. Some 1 (IGBP1). This interaction was confirmed by the detection
small dimeric proteins exchange quickly with simple of several cross-linked peptides. The structure of PP2AA
mixing. Others researchers have used organic solvents [60] was already known from crystallography (PDB codes 3fga
or heat [17] to promote mixing of the heavy and light and 3dw8 [67, 68]). There is no structure of human IGBP1,
subunits. but there is a crystal structure of the N-terminal domain of
the mouse homologue (3qc1). Full-length human IGBP1
Example 3: computational modeling of affinity-purified was modeled with ROSETTA [6971] using the mouse
protein phosphatase complex with distance constraints protein as a template and the observed cross-links as dis-
from XLMS tance restraints. The homology modeling procedure failed
to converge to a single fold; however, filtering by agree-
Our final example is an extensive cross-linking and mod- ment with the XLMS cross-links reduced the ensemble of
eling study of human protein phosphatase 2A (PP2A) and models from 150,000 to 190. All 190 IGBP1 models were
several of its binding partners by Aebersold and coworkers then used in ROSETTA docking protocols [6971] with the
[30]. PP2A holoenzymes are heterotrimers containing a known structure of PP2AA, in order to model the PP2AA-
catalytic A subunit, a scaffold C subunit, and one of *18 IGBP1 interface. The ROSETTA docking energy function
regulatory B subunits [64]. The various B subunits direct was supplemented by distance restraints derived from the
the catalytic activity to specific substrates involved in seven observed intermolecular cross-linked peptides.
several important biological pathways. Thus, proteinpro- Resulting models were also filtered by the agreement
tein interactions play a key role in PP2A function. To between the distances between cross-linking sites in the
investigate the structural features of PP2A interactions, models and the theoretical maximum distance. Residues
Aebersold and coworkers combined XLMS with a pow- K163 and E37 were present in the modeled interface in
erful method for identifying protein interaction networks, 95 % of the models. Binding assays with point mutants at
affinity purification-MS (AP-MS, [65]). In AP-MS, an these residues of PP2AA and the predicted neighbor resi-
affinity-tagged protein (known as the bait protein) is puri- due R155 of IGBP1 resulted in decreased or abrogated
fied under conditions that preserve native proteinprotein binding, thus verifying the predictions of cross-linking-
interactions (complexes). Often, tandem affinity tags are guided modeling.
used (as in this example) to provide the requisite specificity This example clearly illustrates both the power and the
under native conditions. Proteins in the tagged complex are limitations of combining experimental distance restraints
then identified by proteomics methods. The interactions and computational modeling. The procedure was unable to
identified can be either direct or indirect [66]. By com- create an atomic-resolution structural model of the com-
bining AP-MS with XLMS, Aebersold and coworkers not plex, because the structure of one of the binding partners
only identified direct interactions, but elucidated structural (IGBP1) could not be modeled successfully, despite
details that could be used to predict binding interfaces and, numerous XLMS distance restraints and the structure of a
in some cases, to create detailed atomic models. close homologue. (Note that complete atomic models for
The experimental approach involved a first round of AP- other PP2A complexes were created using XLMS
MS without cross-linking to generate a map of protein restraints and described in the same study.) Still, the binding
protein interactions. Target bait proteins for cross-linking interface was correctly identified and experimentally vali-
AP-MS studies were chosen from this map. In the second dated by site-directed mutagenesis of predicted interface
set of experiments, cross-linking was introduced into the residues and coimmunoprecipitation/immunoblotting [30].

123
Cross-linking mass spectrometry and structural biology 87

Independent docking calculations without any experimental XLMS is not without limitations, which include the
cross-linking data identified a binding site on the opposite need for access to high-resolution MS instrumentation, and
side of the protein, demonstrating the critical importance of the need for expert data analysis and interpretation. In
the XLMS derived distance restraints. Neither XLMS nor addition, the difficulty of an XLMS analysis increases
hybrid modeling is a structure determination technique, and with the sample complexity, which is a function of the size
it is reasonable that computational models be viewed with a and heterogeneity of the protein complex studied. XLMS
certain degree of circumspection. However, the incorpora- analysis of small homodimeric complexes (B100 residues/
tion of experimental data such as cross-links from XLMS monomer) should be possible without any enrichment
can improve confidence in computational models, as well as procedure. Much larger protein complexes can be studied if
generate hypotheses about protein complex architectures a protocol involving enrichment of cross-linked peptides is
that can be experimentally tested. used. In general, if a protein complex can be isolated with a
degree of purity similar to that needed for other biophysical
methods, XLMS analysis should be feasible, although
Applicability and limitations of XLMS Example 3 above demonstrated that immunoprecipitated
complexes can also be analyzed with state-of-the art
The preceding examples have demonstrated the utility of methodology.
XLMS in identifying interacting proteins, in providing
residue-level structural information on protein complexes,
and in providing structural restraints for molecular mod- Conclusion: which path to take?
eling. Indeed, the major role of XLMS as a method in
structural biology is in the elucidation of the quaternary The XLMS examples above all used unique combinations
structures of protein complexes in, particularly those that of methods to successfully navigate the inherent com-
are refractory to crystallization due to size, heterogeneity, plexities of XLMSeach to good effect. Several common
integral membrane proteins, protein flexibility, or insta- elements in successful approaches are already emerging,
bility. XLMS has also been used to determine the fold such as the need for high-resolution MS, the use of an
family of a protein growth factor in combination with effective enrichment strategy to increase the number of
homology modeling [19], to identify two distinct binding cross-linked peptides detected, and an advanced informat-
poses of the peptide melittin in a melittin-calcium-calmo- ics tool. Another key feature apparent in the examples is
dulin complex [72], to identify a complex of membrane- internal consistency. Figure 1 may give the impression that
associated bacterial c-type cytochromes [73], to identify the XLMS workflow is modular, but in truth the success
the binding interface in a transient, metal dependent of one step depends on the choices made in other steps. For
binary protein complex [74], and to rank binding affinities instance, the ReACT data analysis approach only works
of a proteins and a series of peptide ligands [75], and even with PIR MS-cleavable cross-linkers. Similarly, the effec-
to study proteinprotein interactions in live cells [31]. tiveness of SEC for enrichment of cross-linked peptides is
The benefits of XLMS are maximized when it is enhanced by digesting with LysC instead of trypsin,
combined with one or more additional structural tech- because LysC generates longer peptides, and so forth.
niques. Recently, Schneidman-Duhovny and others [10] Rather than a single gold standard methodology, we
evaluated the contribution of five types of low-resolution anticipate that the rapidly maturing XLMS field will
structural data (small-angle x-ray scattering, two-dimen- converge on a small number of consensus methods with
sional class average images and three-dimensional density demonstrated records of success, becoming less like a maze
maps from cryoelectron microscopy, information about and more like a supermarket, with a small number of off-
residue types at the interface based on NMR spectroscopy, the-shelf protocols that are effective and ready to
and XLMS) ion protein docking with a benchmark data implement.
set. They found that including any of these types of data led One such emerging consensus approach, and our sub-
to higher success rates than standard docking alone, and jective recommendation as a starting point for researchers
that XLMS compared favorably to the other methods seeking to initiate XLMS studies for structural biology,
studied, at least up to complexes as large as 675 total amino combines the following features: the water soluble NHS-
acids. Furthermore, when the different data types were ester cross-linker BS3 (optionally as a mix of heavy/light
used in combination, inclusion of XLMS data was found isotopic versions), trypsin digestion, enrichment of cross-
to improve the success rate of all other data types, showing linked peptides by SCX chromatography, any standard
that XLMS provides a unique type of data that is com- high-resolution peptide reverse-phase LC separation, and
plementary to other structural techniques. high resolution MS for both MS1 and MS2, with MS2

123
88 E. D. Merkley et al.

Fig. 4 Integration of affinity-purification mass spectrometry (AP- experiments, isolated protein complexes are cross-linked and proteo-
MS) and cross-linking mass spectrometry (XLMS). Tandem affinity lytically digested while bound to the stationary phase of the second
tagged bait proteins are expressed in cell lines and isolated by tandem affinity chromatography step, cross-linked peptides are enriched by
affinity purification, along with interacting proteins. These protein size exclusion chromatography, and analyzed by LCMS. The data is
complexes are then digested and analyzed by mass spectrometry (the searched with xQuest [40], specialized software for cross-linked
traditional AP-MS approach). These data are used both to generate an peptides, and identified cross-links serve as distance restraints in
interaction map, and to create a list of proteins that will be used as the structural modeling
database in searching for cross-linked peptides. In a parallel set of

analysis limited to peptides of charge greater than ?2. Data realized, significant new structural and biological insights
acquired in this manner can easily be analyzed with xQuest are likely to follow.
[38, 40] or pLinks [41], or, if the total sequence length of
all proteins in the analyzed sample is small, with almost Acknowledgments This research was funded by the National
Institute of General Medical Sciences PSI:Biology project (NIGMS
any dedicated XLMS software [18]. This workflow is grant GM094623). Portions of this work were performed in the
similar that described by Yang et al. [41], and also Environmental Molecular Science Laboratory, a U.S. Department of
resembles the later steps of the workflow described in Energy/BER national scientific user facility at Pacific Northwest
Example 3 above and shown in Fig. 4 [30]. For protein National Laboratory in Richland, WA. The authors would like to
thank Nathan Johnson for assistance in preparing the figures, and
complexes containing several copies of the same subunit, it Penny Colton, Michael Daily, and Gyorgy Babnigg for reviewing the
may be helpful to combine this workflow with some vari- manuscript in advance of publication.
ation of the MIX isotope labeling approach in order to
identify interprotein interactions.
Methods using CID cleavable reagents, such as ReACT
References
(Example 1 above), are probably the best available methods
for studies involving very complex samples, such as live 1. Ward AB, Sali A, Wilson IA (2013) Science 339:913915
cells [31, 37], because breaking the cross-link in the mass 2. Lipfert J, Doniach S (2007) Small-angle X-ray scattering from
spectrometer means that the peptide identification search RNA, proteins, and protein complexes. Annual Review of Bio-
physics and Biomolecular Structure, vol 36. Annual Review of
space is proportional to the sequence length, rather than the
Biophysics. Annual Reviews, Palo Alto, pp 307327
square of the sequence length as for cross-linked peptide 3. Subramaniam S, Milne JLS (2004) Annu Rev Biophys Biomol
fragmentation spectra. However, neither the reagents nor Struct 33:141155
the instrument control and data analysis software tools for 4. Jore MM, Lundgren M, van Duijn E, Bultema JB, Westra ER,
Waghmare SP, Wiedenheft B, Pul U, Wurm R, Wagner R, Beijer
these approaches are widely available at this time.
MR, Barendregt A, Zhou K, Snijders APL, Dickman MJ, Doudna
While the challenges facing XLMS analysis are sig- JA, Boekema EJ, Heck AJR, van der Oost J, Brouns SJJ (2011)
nificant, the examples discussed in this review clearly Nat Struct Mol Biol 18:529536
demonstrate that XLMS can be a useful tool to aid in the 5. Lebowitz J, Lewis MS, Schuck P (2002) Protein Sci
11:20672079
development of structural models for proteinprotein
6. Hyung S-J, Ruotolo BT (2012) Proteomics 12:15471564
interactions. XLMS is poised to become a routine com- 7. Mendoza VL, Vachet RW (2009) Mass Spectrom Rev
ponent of the structural biology toolbox. If this vision is 28:785815

123
Cross-linking mass spectrometry and structural biology 89

8. Ruotolo BT, Benesch JLP, Sandercock AM, Hyung S-J, Robin- 38. Walzthoeni T, Claassen M, Leitner A, Herzog F, Bohn S, Forster
son CV (2008) Nat Protocols 3:11391152 F, Beck M, Aebersold R (2012) Nat Meth 9:901903
9. Sharon M, Robinson CV (2007) The role of mass Spectrometry in 39. Marcotte EM (2007) Nat Biotech 25:755757
structure elucidation of dynamic protein complexes. Annual 40. Rinner O, Seebacher J, Walzthoeni T, Mueller L, Beck M,
Review of Biochemistry, vol 76. Annual Review of Biochemis- Schmidt A, Mueller M, Aebersold R (2008) Nat Meth 5:315318
try. Annual Reviews, Palo Alto, pp 167193 41. Yang B, Wu Y-J, Zhu M, Fan S-B, Lin J, Zhang K, Li S, Chi H,
10. Schneidman-Duhovny D, Rossi A, Avila-Sakar A, Kim SJ, Li Y-X, Chen H-F, Luo S-K, Ding Y-H, Wang L-H, Hao Z, Xiu
Velazquez-Muriel J, Strop P, Liang H, Krukenberg KA, Liao M, L-Y, Chen S, Ye K, He S-M, Dong M-Q (2012) Nat Meth
Kim HM, Sobhanifar S, Dotsch V, Rajpal A, Pons J, Agard DA, 9:904906
Cheng Y, Sali A (2012) Bioinformatics 28:32823289 42. Anderson GA, Tolic N, Tang XT, Zheng CX, Bruce JE (2007) J
11. Lasker K, Forster F, Bohn S, Walzthoeni T, Villa E, Unverdorben Proteome Res 6:34123421
P, Beck F, Aebersold R, Sali A, Baumeister W (2012) Proc Natl 43. Gotze M, Pettelkau J, Schaks S, Bosse K, Ihling CH, Krauth F,
Acad Sci USA 109:13801387 Fritzsche R, Kuhn U, Sinz A (2012) J Am Soc Mass Spectrom
12. Rappsilber J (2011) J Struct Biol 173:530540 23:7687
13. Sinz A (2006) Mass Spectrom Rev 25:663682 44. Li W, ONeill HA, Wysocki VH (2012) Bioinformatics
14. Fabris D, Yu ET (2010) J Mass Spectrom 45:841860 28:25482550
15. Singh P, Panchaud A, Goodlett DR (2010) Anal Chem 45. Storey JD, Tibshirani R (2003) Proc Natl Acad Sci USA
82:26362642 100:94409445
16. Miteva YV, Budayeva HG, Cristea IM (2012) Anal Chem 46. Nesvizhskii AI (2010) J Proteomics 73:20922123
85:749768 47. Hoopmann MR, Weisbrod CR, Bruce JE (2010) J Proteome Res
17. Soderberg CAG, Lambert W, Kjellstrom S, Wiegandt A, Wulff 9:63236333
RP, Mansson C, Rutsdottir G, Emanuelsson C (2012) PLoS ONE 48. Merkley ED, Baker ES, Crowell KL, Orton DJ, Taverner T,
7:e38927. doi:10.1371/journal.pone.0038927 Ansong C, Ibrahim YM, Burnet MC, Cort JR, Anderson GA,
18. Mayne SLN, Patterton H-G (2011) Brief Bioinform 12:660671 Smith RD, Adkins JN (2013) J Am Soc Mass Spectrom
19. Young MM, Tang N, Hempel JC, Oshiro CM, Taylor EW, Kuntz 24:444449
ID, Gibson BW, Dollinger G (2000) Proc Natl Acad Sci USA 49. Lee YJ, Lackner LL, Nunnari JM, Phinney BS (2007) J Proteome
97:58025806 Res 6:39083917
20. Paramelle D, Miralles G, Subra G, Martinez J (2013) Proteomics 50. Lee L, Kaplan IB, Ripoll DR, Liang D, Palukaitis P, Gray SM
13:438456 (2005) J Virol 79:12071214
21. Leitner A, Walzthoeni T, Kahraman A, Herzog F, Rinner O, Beck 51. Tang XT, Munske GR, Siems WF, Bruce JE (2005) Anal Chem
M, Aebersold R (2010) Mol Cell Proteomics 9:16341649 77:311318
22. Schilling B, Row RH, Gibson BW, Guo X, Young MM (2003) J 52. Chowdhury SM, Munske GR, Tang XT, Bruce JE (2006) Anal
Am Soc Mass Spectrom 14:834850 Chem 78:81838193
23. Pettelkau J, Schroder T, Ihling CH, Olausson BES, Kolbel K, 53. Zhang HZ, Tang XT, Munske GR, Tolic N, Anderson GA, Bruce
Lange C, Sinz A (2012) Biochemistry 51:49324949 JE (2009) Mol Cell Proteomics 8:409420
24. Leitner A, Reischl R, Walzthoeni T, Herzog F, Bohn S, Forster F, 54. Eng JK, McCormack AL, Yates JR (1994) J Am Soc Mass
Aebersold R (2012) Mol Cell, Proteomics 11:M111.014126. Spectrom 5:976989
doi:10.1074/mcp.M111.014126 55. Perkins DN, Pappin DJC, Creasy DM, Cottrell JS (1999) Elec-
25. Novak P, Haskins WE, Ayson MJ, Jacobsen RB, Schoeniger JS, trophoresis 20:35513567
Leavell MD, Young MM, Kruppa GH (2005) Anal Chem 56. Kelley LA, Sternberg MJE (2009) Nat Protocols 4:363371
77:51015106 57. Schneidman-Duhovny D, Inbar Y, Nussinov R, Wolfson HJ
26. Fritzsche R, Ihling CH, Gotze M, Sinz A (2012) Rapid Commun (2005) Nucleic Acids Res 33:W363W367
Mass Spectrom 26:653658 58. Guo X, Bandyopadhyay P, Schilling B, Young MM, Fujii N,
27. Lauber MA, Reilly JP (2011) J Proteome Res 10:36043616 Aynechi T, Guy RK, Kuntz ID, Gibson BW (2008) Anal Chem
28. Clifford-Nunn B, Showalter HDH, Andrews PC (2012) J Am Soc 80:951960
Mass Spectrom 23:201212 59. Kahraman A, Malmstrom L, Aebersold R (2011) Bioinformatics
29. Lu Y, Tanasova M, Borhan B, Reid GE (2008) Anal Chem 27:21632164
80:92799287 60. Taverner T, Hall NE, OHair RAJ, Simpson RJ (2002) J Biol
30. Herzog F, Kahraman A, Boehringer D, Mak R, Bracher A, Chem 277:4648746492
Walzthoeni T, Leitner A, Beck M, Hartl F-U, Ban N, Malmstrom 61. Guo X, Schilling B, Young M, Medzihradsky M, Kuntz ID, Guy
L, Aebersold R (2012) Science 337:13481352 RK, Gibson BW (2002) Using homobifunctional crosslinking
31. Zheng CX, Yang L, Hoopmann MR, Eng JK, Tang XT, Weisbrod reagents with normal and N-15 labeled proteins for the deter-
CR, Bruce JE (2011) Mol Cell Proteomics 10: M110.006841. mination of protein tertiary structure and proteinprotein inter-
doi:10.1074/mcp.M110.006841 actions. Annual Conference of the American Society of Mass
32. Chavez JD, Cilia M, Weisbrod CR, Ju HJ, Eng JK, Gray SM, Spectrometry, Orlando, FL, USA
Bruce JE (2012) J Proteome Res 11:29682981 62. Bohrer BC, Mererbloom SI, Koeniger SL, Hilderbrand AE,
33. Petrotchenko EV, Serpa JJ, Borchers CH (2011) Mol Cell Pro- Clemmer DE (2008) Biomolecule Analysis by Ion Mobility
teomics 10: M110.001420. doi:10.1074/mcp.M110.001420 Spectrometry. Annual Review of Analytical Chemistry, vol 1.
34. Sinz A, Kalkhof S, Ihling C (2005) J Am Soc Mass Spectrom Annual Review of Analytical Chemistry. Annual Reviews, Palo
16:19211931 Alto, pp 293327
35. Luo J, Fishburn J, Hahn S, Ranish J (2012) Mol Cell Proteomics 63. Back JW, de Jong L, Muijsers AO, de Koster CG (2003) J Mol
11: M111.008318. doi:10.1074/mcp.M111.008318 Biol 331:303313
36. Chowdhury SM, Du XX, Tolic N, Wu S, Moore RJ, Mayer MU, 64. Janssens V, Goris J (2001) Biochem J 353:417439
Smith RD, Adkins JN (2009) Anal Chem 81:55245532 65. Gingras A-C, Gstaiger M, Raught B, Aebersold R (2007) Nat Rev
37. Weisbrod CR, Chavez JD, Eng JK, Yang L, Zheng C, Bruce JE Mol Cell Biol 8:645654
(2013) J Proteome Res 12:15691579 66. Gingras AC, Raught B (2012) FEBS Lett 586:27232731

123
90 E. D. Merkley et al.

67. Xu Y, Chen Y, Zhang P, Jeffrey PD, Shi Y (2008) Mol Cell 70. Das R, Baker D (2008) Ann Rev Biochem 77:363382
31:873885 71. Kaufmann KW, Lemmon GH, DeLuca SL, Sheehan JH, Meiler J
68. Xu Z, Cetin B, Anger M, Cho US, Helmhart W, Nasmyth K, Xu (2010) Biochemistry 49:29872998
W (2009) Mol Cell 35:426441 72. Schulz DM, Ihling C, Clore GM, Sinz A (2004) Biochemistry
69. Leaver-Fay A, Tyka M, Lewis SM, Lange OF, Thompson J, Ja- 43:47034715
cak R, Kaufman K, Renfrew PD, Smith CA, Sheffler W, Davis 73. Zhang HZ, Tang XT, Munske GR, Zakharova N, Yang L, Zheng
IW, Cooper S, Treuille A, Mandell DJ, Richter F, Ban YEA, CX, Wolff MA, Tolic N, Anderson GA, Shi L, Marshall MJ,
Fleishman SJ, Corn JE, Kim DE, Lyskov S, Berrondo M, Mentzer Fredrickson JK, Bruce JE (2008) J Proteome Res 7:17121720
S, Popovic Z, Havranek JJ, Karanicolas J, Das R, Meiler J, 74. Mealman TD, Bagai I, Singh P, Goodlett DR, Rensing C, Zhou
Kortemme T, Gray JJ, Kuhlman B, Baker D, Bradley P (2011) H, Wysocki VH, McEvoy MM (2011) Biochemistry
Rosetta3: an object-oriented software suite for the simulation and 50:25592566
design of macromolecules. In: Johnson ML, Brand L (eds) 75. Madler S, Seitz M, Robinson J, Zenobi R (2010) J Am Soc Mass
Methods in Enzymology, vol 487: computer methods, Pt C. Spectrom 21:17751783
methods in enzymology. Elsevier Academic Press Inc, San 76. Kindy JM, Taraszka JA, Regnier FE, Clemmer DE (2002) Anal
Diego, pp 545574 Chem 74:950958

123

Vous aimerez peut-être aussi