You are on page 1of 13

Analyst

View Article Online


TUTORIAL REVIEW View Journal | View Issue

Bottom-up hydrogen deuterium exchange mass


Cite this: Analyst, 2017, 142, 2874
spectrometry: data analysis and interpretation
Published on 13 July 2017. Downloaded by University of Toronto on 05/11/2017 03:27:58.

a,b
Kerene A. Brown and Derek J. Wilson *a,b

Hydrogen Deuterium Exchange (HDX) Mass Spectrometry (MS) is a sensitive analytical technique that pro-
vides information on protein conformation and dynamics in solution. It is commonly used in the study of
protein–ligand and protein–protein interactions and more recently in the pharmaceutical industry for
epitope mapping, screening drug candidates and in the comparison of biopharmaceuticals to biosimilars.
HDX-MS monitors the exchange of protein backbone hydrogen atoms with deuterium in solution. Recent
advancements in HDX automation and data analysis, have taken the emphasis off developing a funda-
mental understanding of HDX, which is still lacking. This tutorial review will cover the different
mechanisms of exchange and how the exchange reaction is affected by various factors. We also explore
the basis of data analysis and the difficulties that often arise in the interpretation of site-specific and
segment-averaged HDX data, such as overlapping isotopic distributions and correct identification of
peptides. Initial data analysis generates a list of peptides and the deuterium incorporation of each peptide
Received 25th April 2017, at each labeling time point, i.e., a set of deuterium uptake profiles. Data interpretation and error analysis is
Accepted 7th July 2017
subsequently required to ensure that deuterium uptake profiles accurately reflect conformational
DOI: 10.1039/c7an00662d dynamics in solution. Finally, this review will also discuss the different ways in which HDX data can be
rsc.li/analyst represented and how the data can be interpreted.

1. Introduction
a
Hydrogen Deuterium Exchange (HDX) is becoming an increas-
Department of Chemistry, York University, Toronto, ON, M3J 1P3, Canada
b
Center for Research in Mass Spectrometry, York University, Toronto, ON, M3J 1P3,
ingly commonly used technique in the biological sciences. It
Canada. E-mail: dkwilson@yorku.ca allows for the study of solution phase protein conformations

Kerene Brown graduated from Derek Wilson completed his PhD


Carleton University (Ottawa, at Western University (Ontario,
ON) with a BSc in Biochemistry Canada) where he trained in
and Biotechnology and she is bioanalytical mass spectrometry.
currently a PhD student in This was followed by a post-doc
Dr Derek Wilson’s lab at York in 2005 at Cambridge
University (Toronto, ON). Her (Cambridgeshire, UK) in bioana-
research is focused on studying lytical NMR with a focus on
the dynamics of RNA chaperones protein folding and interactions.
and biosimilar characterization Shortly thereafter, he was hired
using Hydrogen Deuterium as an assistant professor at York
Exchange Mass Spectrometry University (Ontario, Canada),
Kerene A. Brown and Ion Mobility. Derek J. Wilson taking up the position in 2007.
He is currently director of the
Center for Research in Mass Spectrometry (CRMS), manager of the
Mass Spectrometry Enabled Science and Engineering (MS-ESE)
training program, and lead investigator in the Technology-
enhanced Biopharmaceuticals Development and Manufacturing
(TBio-DM) initiative.

2874 | Analyst, 2017, 142, 2874–2886 This journal is © The Royal Society of Chemistry 2017
View Article Online

Analyst Tutorial Review

by monitoring the exchange of backbone amide hydrogens 2. Factors affecting intrinsic


with deuterium.1,2 The coupling of HDX to mass spectrometry
(MS) is beneficial as MS allows for the analysis of large and exchange
complex samples and eliminates the need for the incorpor- HX (hydrogen exchange) rates of backbone amide hydrogens
ation of NMR active nuclei (15N and 13C).3 More recently, the vary substantially depending on the presence of hydrogen
development of time-resolved HDX-MS has allowed for the bonding, solvent properties ( pH, temperature), primary
structural analysis of intrinsically disordered proteins which is sequence and most importantly protein structure and
difficult to accomplish using other structural techniques.4,5 dynamics.1,19–21 In order to quantitatively asses HX in back-
The growth of HDX-MS can be attributed to its many bone amides, the observed HX rate should be referenced to
different applications. One of the more recent applications is the rate of the unprotected amide to determine its protection
in the development of biopharmaceuticals, which comprises
Published on 13 July 2017. Downloaded by University of Toronto on 05/11/2017 03:27:58.

factor PF = kint/kobs, where kobs is the experimentally deter-


mostly monoclonal antibodies. HDX-MS has been used in the belonging naturally
mined rate constant and kint is the intrinsic rate constant for a
development of monoclonal antibodies, biosimilar antibodies, given backbone amide hydrogen.21–24
antibody drug conjugates and also in epitope mapping.3,6–8
Another growing area of application for HDX-MS is the study 2.1 pH
of protein–membrane interactions. MS has the ability to differ-
The intrinsic exchange rate of backbone amide hydrogens is
entiate between proteins and lipids and the areas of the
highly sensitive to pH. The mechanism by which the backbone
protein that interacts with the membrane can be deter-
amide exchanges hydrogen with deuterium is primarily cata-
mined.3,9 More common areas of application include, protein–
lyzed by acid (H3O+) or base (OH−) and in some cases there
ligand binding, homology-guided structural characterization
could be a small contribution by water (H2O) catalysis. In base
of proteins, protein folding and protein–protein interactions.
catalysis, which dominates in the pH range above 2.5, the
There are three approaches to local HDX-MS experiments:
amide group is deprotonated by the OH− ion producing an
top-down, middle-down and the most common approach:
amidate anion which in turn is reprotonated by H2O (or D2O)
bottom-up. Top-down analyses, first introduced by Anderegg
to regenerate OH−. Acid catalysis can occur via N- or
et al. 1994, involved deuteration of the intact protein followed
O-protonation. In the case of N-protonation, the mechanism is
by gas phase fragmentation through collision induced dis-
the reverse of base catalysis. The amide N is protonated by
sociation (CID).10 However, CID results in a high degree of
H3O+ (or D3O+), after which H2O (or D2O) extracts a H+ from
hydrogen/deuterium scrambling prior to fragmentation which
the protonated intermediate.25–27 On the other hand,
can significantly alter the results.11–13 ‘Non-ergodic’ fragmen-
O-protonation involves the protonation of the peptide bond
tation techniques such as electron capture dissociation
oxygen which is the most basic site of the peptide bond
(ECD)14,15 and electron transfer dissociation (ETD)16 result in
exchange leading to acidification of the NH proton. The NH proton is
less or no hydrogen scrambling and are now widely used for
then removed by water producing an imidic acid which is later
top-down HDX-MS. Bottom-up HDX involves deuteration of
protonated by D3O+.28 Eqn (1) below, represents the pseudo
the intact protein followed by acid quenching and acid pro-
first order chemical exchange rate constant for unprotected
tease digestion prior to ionization (usually by electrospray).
amides (kch):
The deuterium level of each peptide is determined by monitor-
ing the change in the mass.17 There is no issue with hydrogen kch ¼ kint;A ½Hþ  þ kint;B ½OH  þ kint;W ½H2 O ð1Þ
scrambling as there is no gas phase fragmentation involved,
however, the spatial resolution is low as peptides are typically in terms of the intrinsic second order rate constants for the
4–10 amino acids long. On the other hand, unlike top-down acid catalyzed (A), base catalyzed (B) and water catalyzed (W)
analysis there is no size barrier and large proteins and protein reactions. The contribution by water catalysis is exceptionally
complexes can be studied. To overcome the size barrier of small in comparison to the base and acid catalysis and it is
top-down HDX, a middle-down HDX workflow has been devel- often omitted from eqn (1).29
oped, which involves a combination of the top-down and A plot of amino-acid-averaged log(kch) versus pH results in a
bottom-up approach, in which the intact protein is labelled, V-shaped curve (Fig. 1) in which the minimum pH is 2.5–3.0.
quenched and partially digested followed by gas phase At the pH minimum, the rate of acid and base catalysis is
ETD/ECD fragmentation of the resulting peptides.18 equal and the rate of exchange is at its lowest.20,30 Deuterium
With the growth of HDX-MS there have been many develop- labelling is often carried out under ‘native-like’ conditions at
ments in automated data analysis. However, in using HDX-MS ∼pH 7, where the reaction is primarily base catalyzed (kch =
as a bioanalytical technique, it is important to understand the kint,OH[OH−]), followed by quenching by lowering the pH to
physical basis upon which the method relies. The growth of ∼2.5, to provide sufficient time for Liquid Chromatography
automated data analysis systems has greatly reduced the (LC) separation and MS analysis.20
perceived importance of understanding the fundamentals of
HDX. This tutorial review will cover the physical basis of the 2.2 Temperature
HDX reaction, data analysis, data interpretation and represen- For the base-catalyzed reaction, HX has an activation energy of
tation of results. approximately 17 kcal mol−1, resulting in an increase in the

This journal is © The Royal Society of Chemistry 2017 Analyst, 2017, 142, 2874–2886 | 2875
View Article Online

Tutorial Review Analyst

rate constant of an amide H can be calculated based on


eqn (3) in which Aleft, Aright, and Bleft, Bright are side chain acid
and base factors respectively.21

kch ¼ kHþ ðAleft  Aright Þ½Hþ  þ kOH ðBleft  Bright Þ½OH 


ð3Þ
þ kH2 O ðBleft  Bright Þ:

3. Exchange regimes
Published on 13 July 2017. Downloaded by University of Toronto on 05/11/2017 03:27:58.

Although pH, temperature and primary sequence has a high


impact on the HX rate in random coil polypeptides, in a folded
protein the higher order structure has a greater impact on the
Fig. 1 The dependence of intrinsic rate of exchange (kch) on the solu- HX rate. In order for a labile H to undergo exchange with D2O,
tion pH in which the rate of exchange reaches a minimum at pH 2.5–3. it needs to be exposed to the solvent (i.e. not buried in the
Acid and base catalysis is dominant below and above the pH minimum core of the protein) and not be involved in any H bonding
respectively. network.1,32 The basic model for HX in folded proteins is
depicted as:

As the temperature is increased, the HX rate is increased. kop kch kcl


NHcl Ð NHop ! NDop Ð NDcl : ð4Þ
29,31 kcl kop
HX rate by a factor of 3 for each 10 °C increment. In order
to predict HX rate constants at other temperatures, each rate Due to the dynamic nature of proteins, buried or H-bonded
constant in eqn (1) should be modified according to eqn (2). backbone amide hydrogens do not always remain buried or
The theoretical HX rate constant (kch) as a function of tempera- H-bonded and can often come into an open state in which
ture can be determined by: exchange can occur. In the mechanism depicted above, kop
   and kcl represents the rate constant of the opening and closing
Ea 1 1
kch ðTÞ ¼ k293 exp   : ð2Þ motions respectively. When in the open state, HDX occurs
R T 293
with the rate constant kch. As a result, the observed rate con-
In which k293 is the rate constant kint, H, kint, OH, or kint, H2O stant of exchange in folded proteins is a product of the rate
at 293 K; Ea is the activation energy corresponding to 14, 17 constants of unfolding and refolding (i.e., ‘opening’ and
and 19 kcal mol−1 for the acid, base and water-catalyzed ‘closing’) and the intrinsic rate constant.32–34 The observed
exchange respectively; and R is the gas constant 0.001986 rate constant for exchange kHX, in a folded protein under
kcal mol−1 K−1. During the quench step in a bottom up native-like conditions can then be expressed as:35
HDX-MS experiment, along with decreasing the pH, the temp-
erature is also typically decreased to 0 °C to minimize back- kop  kch
kHX ¼ : ð5Þ
exchange.27 kcl þ kch

2.3 Primary sequence


3.1 EX1 kinetics
Another factor affecting amide HX rates is the primary
sequence of the protein in which neighboring side chains can The conventional framework for analyzing HDX data is orga-
produce an inductive or steric effect. Inductive effects are nized into two ‘regimes’ based on the relative magnitudes of
caused by polar side chains, such as serine, which withdraw kch and kcl. In the EX1 regime, during the opening event, all
electron density from neighboring peptide groups resulting in exposed labile hydrogens undergo exchange prior to the
an increase in their acidity. This increases the rate of deproto- exchangeable sites returning to their ‘closed’ state (i.e., kcl ≪
nation of the amide H by (OH− or OD−) in the base catalyzed kch) and the observed HX rate constant is equivalent to the rate
reaction whereas the overall rate of the acid-catalyzed reaction constant of the opening reaction. Therefore eqn (5) is simpli-
decreases. Therefore, at pH > pHmin, an amide H alongside fied to: since kcl is very small, it's ignored

serine residues will exchange 2.3 times faster than an amide H


kHX ¼ kop : ð6Þ
alongside a non-polar residue (e.g. alanine).21,24 Bulky side
chains such as that of isoleucine decreases the amide HX rate This type of exchange is not typical of proteins under
by blocking solvent accessibility. Depending on pH this can physiological conditions (though it does occur), and can be
decrease the rate of exchange by a factor of 10 to 20 if an induced through the addition of denaturants or a change in
amide is flanked by 2 isoleucines.21 Both steric and inductive pH (usually an increase; pH > 9.0). Although this is not com-
effects are additive and depend on the side chains of the monly representative of physiological protein dynamics, it can
nearest amino acid. In an unstructured peptide, the exchange provide insights into protein folding.34

2876 | Analyst, 2017, 142, 2874–2886 This journal is © The Royal Society of Chemistry 2017
View Article Online

Analyst Tutorial Review

Although somewhat rare, cases of EX1 kinetics have been


observed in proteins under physiological conditions such
as the Hck, Lyn and α-spectrin SH3 domains which undergo
a slow unfolding event under physiological conditions.36,37
One important benefit of coupling HDX with MS is the fact
that EX1 kinetics can be easily identified in a mass
spectrum through its characteristic bimodal distribution
(Fig. 2) representing 2 populations. The low mass popu-
lation represent protein that has never undergone the EX1
conformational transition whereas the high mass popu-
lation has undergone this conformational transition at least
Published on 13 July 2017. Downloaded by University of Toronto on 05/11/2017 03:27:58.

once.34
EX1 hydrogen deuterium exchange kinetics can be useful
in the determination of slow unfolding processes in proteins.
The effectiveness of this method was demonstrated by Engen
et al. in 1997 when they published an account of intact HcK
SH3 demonstrating EX1 kinetics.37 This work was later
continued by Wales and Engen in 2006 in which they studied
11 other SH3 domains using HcK SH3 as a reference.36 Their
results revealed that HcK, Lyn and α-spectrin SH3 domains
Fig. 2 Cooperative unfolding representative of EX1 kinetics in selected
exhibited clear EX1 kinetics that were evident from the
SH3 domains. ESI spectra of peptic peptides of (a) Lyn SH3 99–116 and
(b) α-spectrin 996–1014. Reproduced with permission from ref. 36.
characteristic bimodal distributions, in 3 out of the 12
Copyright 2006, Elsevier. domains (Fig. 3).36

Fig. 3 Deuterium uptake of 12 intact SH3 domains from the centroid(s) of HDX isotopic distributions, in which the dotted line at the top represent
the maximum possible deuterium level. Bimodal distributions corresponding to EX1 exchange are observed in panels a, b and j in which multiple
centroids are identified. Traces with a sigmoidal shape exhibit distortions in their isotopic distributions that may correspond to EX1 exchange.
Reproduced with permission from ref. 36. Copyright, 2006 Elsevier.

This journal is © The Royal Society of Chemistry 2017 Analyst, 2017, 142, 2874–2886 | 2877
View Article Online

Tutorial Review Analyst

3.2 EX2 kinetics solvent.17 Separation is not typically required for global ana-
When the rate constant of the closing reaction is faster that lysis so a trap column is often used in the LC step to desalt the
the intrinsic rate of exchange (kcl ≫ kch) EX2 kinetics are protein sample prior to MS analysis. The LC step also elimi-
observed, and eqn (5) becomes:32 nates deuterium that was incorporated into side chains (all
ultra-fast exchangers) through complete back-exchange. As a
kop result, by the time the sample is subjected to ESI, deuterium
kHX ¼  kch : ð7Þ
kcl will have equilibrated with solvent for all but the backbone
EX2 HDX results in a gradual increase in the mass over amides, which significantly simplifies data analysis and
time in a single (binomial) distribution. As most proteins are interpretation.17,48
structurally stable at physiological pH, transient unfolding As there is no protease digestion or gas phase fragment-
ation involved in global HDX analysis, it provides an overall
Published on 13 July 2017. Downloaded by University of Toronto on 05/11/2017 03:27:58.

events in structured regions occur faster than kch, so EX2 is the


most common exchange regime of a protein under native con- picture of the conformational dynamics of a protein of inter-
ditions.34,36 EX2 is also the more informative of the two domi- est. To determine the amount of deuterium incorporated in
nant exchange regimes, as it allows for the determination of the the protein, the mass of the non-deuterated protein is sub-
‘open’/‘closed’ equilibrium, which, when measured specific tracted from the mass of the deuterated protein at each time
sites or segments, can be viewed as a local measurement of point being studied. This can be done for individual charge
local thermodynamic structural stability.38 It is important to be states of the protein or by using deconvoluted masses. The
able to distinguish between EX1 and EX2 because there are data are then plotted as a change in mass or relative deuterium
some cases in which EX1 is observed under native conditions as level (y axis) vs. time (x axis).48 The information that is gained
discussed previously, and this can confound automated data from global HDX is limited and so local HDX is more common.
analysis software. Experimentally, HDX-MS provides the advan- However, global HDX can be useful to determine if the protein
tage of easily differentiating between EX1 and EX2, which is of interest is stable under HDX-MS conditions and serve as an
more difficult to do with NMR experiments.37 initial guide as to how dynamic the protein is. The relative
extent of disordered regions in a protein can be estimated from
3.3 EXX kinetics the number of hydrogens that exchanged at early time points
(10 s or less). Likewise, the number of hydrogens involved in
EXX kinetics refer to an intermediate state where kcl is neither more rigid or buried residues can be determined by subtracting
faster nor slower than the kch (kcl ≈ kch). This phenomenon the maximum observed exchange (after several hours of label-
has been explored by Xiao et al., in which they observed a ling) from the total number of exchangeable backbone hydro-
bimodal distribution similar to that in the EX1 regime gens.49 To ensure the accuracy of these estimates, an internal
however each population drifted as observed in EX2 kinetics.39 calibrant (such as an unstructured peptide) should be used to
EXX kinetics should not be confused with mixed EX1 and EX2 account for backbone amide back-exchange. An example of the
exchange, which is what is commonly observed in whole pro- utility of global HDX is provided in the comparison of biosimi-
teins that undergo an EX1 conformational transition (e.g., lar antibodies published by Pan and coworkers in 2016,7 in
some of the SH3 domains in Fig. 3). which the deuterium uptake was calculated for the intact heavy
and light chains for each antibody being studied (Fig. 4). Their
results showed that the antibodies had the same solution
4. Data analysis dynamics which was later verified by local HDX.7

HDX-MS data analysis involves the correct identification of 4.2 Local HDX
peptides (in the case of local HDX), back exchange correction, The experimental workflow for local HDX is essentially
determination of deuterium uptake and rates of exchange. identical to global HDX with one additional step to digest the
Many software programs (e.g. MS Studio,40 DynamX, HDX deuterated protein sample. The term “local” refers to the type
Workbench,41 HX-Express v2,42 Hexicon 2,43 HDXFinder,44 of information one gets from the experiment, in which it is
HDX-Analyzer,45 ExMS,46 MassAnalyzer,47 and more) are avail- possible to measure the conformational or dynamic changes
able for automated data analysis but it remains crucial to in localized regions of the protein, allowing for instance, for
understand the process, as this can aid in the interpretation of the determination of ligand binding sites.7,50 There are two
the results. In some cases, there may be a need to double- main types of local HDX workflows: bottom up and top down.
check results obtained from automated software packages. Bottom-up local HDX is the more common of the two,
which involves digestion of the deuterated sample with an acid
4.1 Global HDX resistant protease prior to ESI. This approach is in some ways
A typical workflow for global HDX involves the protein of inter- more straightforward than top-down HDX and does not require
est incubated in an excess of D2O for a period of time, after specialized equipment, but provides peptide-level resolution
which the reaction is quenched by decreasing the pH to 2.5 (typically 4–10 residues). The top down workflow involves gas
and the temperature to 0 °C in order to minimize the exchange phase fragmentation of the deuterated protein. Although top-
rate in what is (from quenching onwards) mainly protic down analysis can provide amino acid level resolution, there is

2878 | Analyst, 2017, 142, 2874–2886 This journal is © The Royal Society of Chemistry 2017
View Article Online

Analyst Tutorial Review

measure relative rather than absolute deuterium uptake and


studies have shown that back exchange correction does not
affect the final results in relative measurement experiments
(i.e., where one state is being compared to another state of the
same protein).50,51 In addition, back exchange correction is
more of an estimate, as it relies heavily on preparing a fully
deuterated form of the sample of interest, which can be
difficult to prepare and maintain.51
The 1st step in data analysis is the determination of the
peptide sequences based on their m/z and LC retention time.
Accurate mass is often not sufficient to determine the peptide
Published on 13 July 2017. Downloaded by University of Toronto on 05/11/2017 03:27:58.

sequences but it can be used to determine the possibilities.55


To ensure accuracy in peptide peak assignments, tandem MS
can be used to conduct classical MS/MS peptide sequencing.56
This step is crucial as miss-identifying the peptides can lead to
fundamental misinterpretation of the results. There are
2 main methods for peptide sequencing by LC MS/MS: Data
Fig. 4 Global HDX of the intact light chain and heavy chain. (A) BEV vs. Dependent Acquisition (DDA) and Data Independent
BEV1; (B) BEV vs. BEV2. Each data point represents an average of two
Acquisition (DIA). DDA performs the MS/MS analysis on pre-
experiments. Reproduced with permission from ref. 7. Copyright 2016,
Elsevier.
cursor m/z selected from the MS survey scans. There is a selec-
tion criteria and specific m/z values can be included or
excluded from the MS/MS analysis.57 Conversely, DIA acquires
the MS/MS scans using a wide isolation window with no
an analyte size barrier and specialized equipment is required to specific precursor ion selected.58
prevent deuterium scrambling in the gas phase.7,50,51 After the peptide list is generated, deuterium incorporation
In the bottom-up workflow, because the reaction is can be quantified. There are 2 main ways in which this is
quenched with acid, an acid resistant protease must be used. done: through the change in the centroid mass or through the
Pepsin is the most commonly used protease in HDX experi- change in the isotopic distribution of the peptide. Through
ments,52 but others can be employed as well such as: aspergil- the former method, the centroid mass of the non-deuterated
lopepsin ( protease type XIII),53 rhizopuspepsin ( protease type peptide (MUND) and deuterated peptide (Mt) is calculated and
XVIII)53 and plasmepsin.54 Non-specific proteases often result the relative deuterium level (Dt) of each peptide is determined
in multiple overlapping peptides and with careful analysis by the equation:
information on single residue amide hydrogens can theoreti-
cally be obtained but this should be done with caution as this Dt ¼ M t  M UND : ð9Þ
could lead to over-interpretation of the results. A major dis-
This method is not a direct measure of uptake but it can be
advantage of protease digestion is incomplete sequence cover-
sensitive enough in detecting changes in conformation.
age or peptides that are too large in some cases where the
However, this method is not ideal for detecting subtle changes
protein does not get digested very well. This can sometimes be
in deuterated peptide isotopic distributions associated with
overcome with the use of a mix of different proteases or by
small levels of uptake and it may miss exchanges in the EX1 or
adding a denaturant in the quench buffer to improve the
EXX regime.59,60
digestion.53 After the protein is digested, the peptides are
Another method used to quantify deuterium uptake levels
separated using reversed-phase LC prior to MS analysis.
is to model and then fit the isotopic distribution of the deute-
With the addition of lengthy digestion and LC separation
rated peptide, which is often more accurate than using the cen-
steps, back exchange may occur, but can usually be accounted
troid. This is particularly true at low deuteration levels or where
for in data analysis.17 To account for back exchange, a comple-
there is overlap for some peaks in the distribution. In a non-
tely deuterated sample (M100%) is used as a control and the
deuterated peptide, the isotopic distribution observed is mostly
total deuterium uptake level in each peptide, D, is calculated
as a result of 12C and 13C. However, as deuterium is incorpor-
at each time point using the equation:
ated the distribution becomes a convolution of (mainly) the
Mt  MUND 12 13
C/ C distribution and 1H/2H distribution.55 The isotopic dis-
D¼ N ð8Þ
M100%  MUND tribution after labelling can be modelled as a convolution of the
natural isotope distribution given by the equation:
where Mt and MUND is the mass of the deuterated peptide and
mass of the un-deuterated peptide respectively and N is the D¼NB ð10Þ
maximum number of deuterium atoms that can be incorpo-
rated.17 There has been debate over the years as to whether a where D is the isotope distribution after deuterium
back-exchange correction is useful as most HDX experiments labeling, N is the natural isotope distribution and B is the

This journal is © The Royal Society of Chemistry 2017 Analyst, 2017, 142, 2874–2886 | 2879
View Article Online

Tutorial Review Analyst

binomial distribution. This binomial distribution is rep- 4.3 Automated data analysis
resented by: The introduction of automated data analysis has made
HDX-MS more widely used, as analyzing HDX-MS data can
n! become extremely time-consuming if it is done manually.
Bðp; n; kÞ ¼ pk ð1  pÞnk : ð11Þ
k!ðn  kÞ! Recent software developed by Zhang et al. 2012 can be used to
automatically derive protection factors for each peptide,
making data analysis and interpretation more straight-
In which n is the total number of exchangeable hydrogens
forward.47 The most common approach in quantifying deuter-
in the peptide of interest and k is the number of hydrogens
ium levels is the use of centroid masses, which is the basis of
that have undergone exchange (0,1,…n). The segment averaged
many automated HDX analysis software packages. As men-
level of deuteration (0–100%) is represented by p. The actual
Published on 13 July 2017. Downloaded by University of Toronto on 05/11/2017 03:27:58.

tioned earlier, if only the centroid masses are used and isotopic
deuteration level of each peptide can then be determined by
distribution is ignored, a degree of accuracy could be lost and it
fitting a simulated distribution B, to the experimental data.61
is not possible to directly identify EX1 exchange.60 To correct
It is important to emphasize that the best-fit value of p rep-
this, some recently developed software packages automatically
resents the average uptake for the peptide and does not
identify bimodal distributions characteristic of EX1 kinetics
provide information on the distribution of uptakes at individ-
using alternative methods.46,65 Another downside with many
ual sites on the peptide. For instance, in an 8 amino acid
automated systems is the inability to analyze overlapping pep-
peptide, a highly skewed distribution (e.g., 4 sites exchanging
tides and peptides with low signal intensity. Recently developed,
at 100% and 4 sites at 0%) would produce the same value for p
is a semi-automated platform (HX-Express v2) which has tools
as a flat distribution (e.g., 8 sites exchanging at 50%), both of
for bimodal deconvolution and binomial fitting, and the ability
which are correct (i.e., in both cases, a perfect fit to the data
to analyze overlapping peptides.42 The use of automated
would yield p = 50% and the peptide is in fact 50%
systems is becoming more common as it significantly reduces
exchanged). There are several methods to fit theoretical distri-
the time spent on data analysis and increases the amount of
butions to the data. The most common and straightforward of
results being processed. With that being said, it is important to
these is the least-squares approach which minimizes the
understand the data analysis process as this will aid in the
square of the difference between the normalized peak intensi-
interpretation of the results obtained and often times manual
ties of the theoretical and observed distributions (Fig. 5).59,62
checks are required to ensure the data was analyzed correctly.
A major hurdle in the quantitation of deuterium levels, is
the presence of over lapping peptides. Although the LC step
after digestion separates the peptides, overlap still occurs often
in samples from large proteins. One way in which this 5. Data interpretation
problem could be overcome is using a 2nd dimension of separ-
ation, such as ion mobility, that separates species based on After the data is analyzed, a list of peptides with the amount of
deuterium incorporated into each peptide at each time point
their charge and conformation. This could essentially be used
to separate peptides with the same LC retention time.63 It is studied is generated. However, after this step, there is still more
also noted that the ‘distribution fitting’ method described work to be done. There are several post data analysis steps
which include but are not limited to: extracting kinetic infor-
above requires only three overlap-free peaks within a distri-
bution to accurately determine the uptake level. In reporting mation, determination of exchange amplitudes, determining
the relative deuterium level, it is also important to provide an the difference between binding interactions and allosteric
effects and finding the best way in which to represent the data.
error in these measurements. Errors are commonly estimated
through standard deviations which are usually obtained from
at least three technical replicates of the same labelling time 5.1 Exchange kinetics vs. exchange amplitudes
point. It is equally important to use biological replicates where HDX experiments are often carried out at multiple labelling
possible to test the reproducibility of the results.55,64 times, most commonly from 10 s to 5 h or more. As a result,

Fig. 5 Representation of how the theoretical distribution D (red dots) is fitted to the spectrum of a deuterated peptide to determine the percent
deuterium incorporation.

2880 | Analyst, 2017, 142, 2874–2886 This journal is © The Royal Society of Chemistry 2017
View Article Online

Analyst Tutorial Review

the data can be presented as a graph of deuterium uptake vs. ommended to group the backbone amides into fast, intermedi-
HDX reaction time. Exchange amplitude refers to the ate and slow exchangers and fit the data using the equation:
maximum amount of deuterium incorporated into a peptide
or whole protein at infinite time, represented by level at which X
N
D¼N expðki tÞ: ð12Þ
uptake reaches an apparent steady state in the kinetic data. i¼1
When the HDX profiles of a native protein are compared to a
ligand-bound protein a difference in the exchange amplitude In which N is the number of exchangeable backbone
(usually a decrease) is often observed in peptides that are amides, ki is the exchange rate and t is the time allowed for
within the binding region due to the formation of new strong exchange. Depending on the complexity of the data collected,
H-bonds.55 Changes in solvent accessibility may affect uptake the kinetic curves can be fit to between one and three exponen-
kinetics, but are unlikely to change the exchange amplitude tials.17 Another approach is to use a ‘stretching’ term β, which
Published on 13 July 2017. Downloaded by University of Toronto on 05/11/2017 03:27:58.

since that would require a subset of exchangeable sites to takes into account the multimodal nature of the distribution
become completely solvent inaccessible upon complexation. of intrinsic rates.66
If enough time points are obtained in a bottom-up HDX Here we will briefly discuss how kinetics can be used to
experiment, the uptake rate can be determined for each interpret HDX data using an ATPase packaging motor,
peptide in the protein sample. These rates can then be com- P4 hexamer as an example. HDX kinetics of the free hexamer
pared between the native protein and protein bound state to was compared to a PC associated P4. In any given peptide,
determine any changes in rate of the HDX reaction. In order to each amide hydrogen has different exchange rates which leads
determine rates, the absolute number of deuterium should be to the kinetics becoming rather complex. Therefore, the kine-
known and therefore a back-exchange correction must be per- tics obtained can be used to generate exchange rate distri-
formed. While each backbone amide will have a different rate butions shown in Fig. 6.67,68 A maximum entropy method
of exchange, the observed rate is effectively the average of (MEM) algorithm developed by Zhang et al. 1997 can be used
those rates, with the possibility of multi-phasic behavior due to derive hydrogen exchange rate distributions from kinetic
to a multi-modal distribution of rates. It is therefore rec- curves obtained from HDX-MS experiments.67 With the rate

Fig. 6 HDX-MS kinetic curves of selected peptides of the P4 hexamer (red) and P4–PC complex (blue): residues 69–83 (A), 131–152 (B), 215–224
(C), 230–244 (D), 294–320 (E), 316–324 (F). Panels from left to right: Localization of the region within the structure of the P4 subunit, HDX kinetic
curves plotted as deuterium content with respect to labelling time, rate distribution, number of sites exchanging with slow (<0.1 h−1, blue), inter-
mediate (0.1–100 h−1, green) and fast (>100 h−1, red) rates.68 Reproduced with permission from ref. 68. Copyright 2006, Elsevier.

This journal is © The Royal Society of Chemistry 2017 Analyst, 2017, 142, 2874–2886 | 2881
View Article Online

Tutorial Review Analyst

distributions obtained, the number of protected sites, sites mational change. However, if ligand binding results in
with intermediate exchange and sites with fast exchange rates decrease deuterium uptake in an allosteric region, it becomes
can be determined.68,69 difficult to differentiate between the binding interactions and
Peptides on the surface of the isolated hexamer were allosteric conformational changes.70,71 If a crystal structure of
revealed to have fast or intermediate exchange rates (Fig. 6A, E the protein being studied is available that shows the protein in
and F) in which 2 of these peptides (Fig. 6E and F) became pro- complex with its ligand, then HDX can be used to monitor
tected upon binding to PC which was evident by the decrease in conformational changes upon ligand binding to determine the
the number of fast and intermediate exchange sites and an allosteric hotspots.72 A recent publication by Chandramohan
increase in the number of slow exchange sites. However, the et al. 2016, used HDX-MS to distinguish allosteric effects from
peptide 69–83 revealed no evidence of protection in the P4–PC ligand binding in Hsp90. The ligand interactions are known
complex as depicted in Fig. 6A. Peptides that are located within based on published high resolution structures of Hsp90 with
Published on 13 July 2017. Downloaded by University of Toronto on 05/11/2017 03:27:58.

the core of the hexamer (Fig. 6B and C) showed significantly 2 high affinity ligands (PDB ID:4EGK and 1YET). HDX-MS
slower kinetics than the other peptides and when bound to PC revealed significant decreases in deuterium uptake in the
there was an even greater decrease in the HDX kinetics. This is ligand binding regions. In addition to testing high affinity
likely as a result of global stabilization as it is not expected that ligands, low affinity ligands (fragment 1 and 2) were tested
these buried residues would come into contact with PC. In the and found to have more significant allosteric responses to
isolated hexamer, the C-terminal peptides (Fig. 6E and F) have ligand binding. The same 4 ligand binding sites were observed
almost no protected sites but upon binding to PC both peptides when using the high or low affinity ligands but the high
gained a significant amount of protected sites.68 affinity ligands had a much more significant decrease in deu-
terium uptake. The fragments are smaller and are expected to
5.2 Binding vs. allosteric effects have fewer interactions in the binding pocket.70
A major limitation of HDX-MS is the inability to distinguish Both fragments had similar changes in the binding pocket
between the binding interactions and allosteric conformational but had significantly different allosteric responses. This was
changes. If ligand binding results in an increase in deuterium unexpected as both fragments have similar molecular weight
uptake in a certain region of the protein, this change can and affinities and they belong to the same phenolic class. It is
be often (but not always) be attributed to an allosteric confor- proposed that changes due to protection from HX as a result

Fig. 7 Conformational changes due to ligand binding and allosteric effects. (A) a difference plot showing the absolute difference in deuterium
uptake between the free and ligand bound state for each peptic peptide of Hsp90 at (t = 0.5, 2, 5, 10 min). Positive shifts represent decreases in deu-
terium uptake and negative shifts represents increases in deuterium uptake in comparison to the apo-Hsp90. Orthosteric sites are highlighted in the
blue boxes while allosteric sites are highlighted in the red boxes. (B, C) Orthosteric and allosteric regions are mapped onto the 3D structure of
Hsp90 (PDB ID:4EGK). Reproduced with permission from ref. 70. Copyright 2016.

2882 | Analyst, 2017, 142, 2874–2886 This journal is © The Royal Society of Chemistry 2017
View Article Online

Analyst Tutorial Review

of H-bonding would show up in earlier reaction times whereas needed in order to thoroughly test this hypothesis.
changes due to allosteric conformational changes would Nonetheless, when used together, HDX-MS and X-ray crystallo-
appear at longer times. Additional experiments would be graphy can be used to distinguish between binding inter-
actions and long range allosteric changes.
Another important application of HDX-MS is in the study of
allosteric regulation in enzymes. This has been recently impli-
cated by Sowole et al., 2016 using Dihydrodipicolinate Synthase.
X-ray crystallography reveals only very subtle changes in the struc-
ture of inhibitor free and the bis Lys-inhibited enzyme which
does not explain the allosteric inhibition of the enzyme. HDX-MS
revealed increased dynamics in the inhibitor free enzyme which
Published on 13 July 2017. Downloaded by University of Toronto on 05/11/2017 03:27:58.

allows binding of the substrate. On the other hand, there is


decreased dynamics in the bis Lys-inhibited state which prevents
the substrate from binding. This study shows how valuable
HDX-MS can be in the study of allosteric regulation.73

5.3 Data representation


There are many ways in which HDX-MS data can be presented
and it sometimes challenging to choose a method that best
represents the data. After the initial data analysis, the result is
a list of peptides with the amount of deuterium incorporated
at each reaction time studied. Arguably the most common way
of representing HDX data is by using a butterfly plot.74 This
type of plot allows for easy comparison, as multiple time
Fig. 8 Difference in deuterium uptake mapped to CjDHDPS crystal points can be plotted on the same graph which allows for the
structures. Blue represents the regions with lower deuteration levels observations of trends that may develop over the HDX reaction
after (a) Changes in deuterium uptake upon Lys binding. (b) Same as
times being studied. Different protein states are also plotted
panel a, viewed from the central cavity for subunit C in space-filling
representation. (c and d) Corresponding deuteration changes after together for easy comparisons. An example of this can be seen
bisLys binding. Reproduced with permission from ref. 73. Copyright in Fig. 7A. To compare the uptake rates of individual peptides,
2016, ACS publications. deuterium uptake curves are the most direct method (Fig. 3, 4

Fig. 9 HDX exchange on Cdc42. (A) Deuterium level of Cdc42 measured at 6 time points (10 s to 3000 s) shown per the color scheme. The HDX
results at (B) 10 s and (C) 3000 s are mapped onto the crystal structure of Cdc42 (PDB ID: 2QRZ). Reproduced with permission from ref. 75.
Copyright 2016, Elsevier.

This journal is © The Royal Society of Chemistry 2017 Analyst, 2017, 142, 2874–2886 | 2883
View Article Online

Tutorial Review Analyst

and 6). If the data are fitted to eqn (12), the HX rate can be 2 J. R. Engen, Analysis of Protein Conformation and
determined for each peptide. This is an easy way to compare Dynamics by Hydrogen/Deuterium Exchange MS, Anal.
peptides within the same protein or the same peptide in Chem., 2009, 81, 7870–7875.
different protein states (e.g. free vs. bound protein). 3 G. F. Pirrone, R. E. Iacob and J. R. Engen, Applications of
When two or more states of a protein are studied, the differ- Hydrogen/Deuterium Exchange MS from 2012 to 2014,
ence in uptake can be mapped onto the ground-state 3D struc- Anal. Chem., 2015, 87, 99–118.
ture if one is available from X-ray crystallography or NMR data. 4 C. Lento, G. F. Audette and D. J. Wilson, Time-resolved
In the simplest case, decreases in deuterium uptake are often electrospray mass spectrometry — a brief history,
represented on the structure with blue while increases are Can. J. Chem., 2014, 93, 7–12.
represented as red (Fig. 8). This provides an easy way to diplay 5 S. Zhu, et al., Hyperphosphorylation of intrinsically dis-
HDX-MS results in the context of localized changes in ordered tau protein induces an amyloidogenic shift in its
Published on 13 July 2017. Downloaded by University of Toronto on 05/11/2017 03:27:58.

dynamics. For a more nuanced view of the dynamics, a color conformational ensemble, PLoS One, 2015, 10, e0120416.
scheme that typically ranges from blue to red with the lowest 6 B. Deng, C. Lento and D. J. Wilson, Hydrogen deuterium
deuterium uptake represented as blue and the highest uptake exchange mass spectrometry in biopharmaceutical discov-
represented as red can be used. An example is shown in Fig. 9, ery and development – A review, Anal. Chim. Acta, 2016,
in which HDX experiments were carried out on Cdc42 at 940, 8–20.
6 HDX reaction time points from 10 s to 3000 s. The results 7 J. Pan, S. Zhang and C. H. Borchers, Comparative higher-
of 1st and last time points were mapped onto the crystal struc- order structure analysis of antibody biosimilars using com-
ture and a very high deuterium uptake can be observed at the bined bottom-up and top-down hydrogen-deuterium
3000 s vs. 10 s that show very little uptake in comparison.75 exchange mass spectrometry, Biochim. Biophys. Acta,
Proteins Proteomics, 2016, 1864, 1801–1808.
8 M. Prądzińska, et al., Application of amide hydrogen/deu-
6. Conclusions and outlook terium exchange mass spectrometry for epitope mapping
in human cystatin C, Amino Acids, 2016, 48, 2809–2820.
HDX-MS is a fast-growing technique that continues to evolve 9 O. Vadas, M. L. Jenkins, G. L. Dornan and J. E. Burke,
with new applications but the core process remains the same. Chapter Seven - Using Hydrogen–Deuterium Exchange
This review has outlined some of the basic concepts of Mass Spectrometry to Examine Protein–Membrane
HDX-MS and the process of data analysis and interpretation. Interactions, in Methods in Enzymology, ed. M. H. Gelb,
With understanding the basic concepts of HDX it becomes Academic Press, 2017, vol. 583, pp. 143–172.
easier to interpret the data correctly. HDX can be used to deter- 10 R. J. Anderegg, D. S. Wagner and C. L. Stevenson, The mass
mine exchange kinetics of peptides and intact proteins and spectrometry of helical unfolding in peptides, J. Am. Soc.
the effect the ligand binding has on the kinetics.34,36,68 Mass Spectrom., 1994, 5, 425–433.
Although most applications of HDX-MS involve the determi- 11 A. G. Harrison and T. Yalcin, Proton mobility in protonated
nation of binding sites, it can be used to determine allosteric amino acids and peptides, Int. J. Mass Spectrom. Ion
changes under varying conditions although it remains difficult Processes, 1997, 165, 339–347.
to distinguish binding sites from allosteric sites if the binding 12 R. S. Johnson, D. Krylov and K. A. Walsh, Proton mobility
sites are not previously known.70,73 There may be a time within electrosprayed peptide ions, J. Mass Spectrom., 1995,
dependent observable change that is affected differently by 30, 386–387.
ligand binding and allosteric effects.70 This would be an inter- 13 P. T. M. Kenny, K. Nomoto and R. Orlando, Fragmentation
esting development if HDX-MS could be used to differentiate studies of peptides: The formation of Y ions, Rapid
allostery from binding without the use of other techniques but Commun. Mass Spectrom., 1992, 6, 95–97.
this is an area that needs to be explored further. The use of 14 K. Breuker, H. Oh, C. Lin, B. K. Carpenter and
computational modelling, is another fast growing field and F. W. McLafferty, Nonergodic and conformational control
when used along with HDX-MS, it could potentially aid in the of the electron capture dissociation of protein cations, Proc.
interpretation of results to distinguish binding and allosteric Natl. Acad. Sci. U. S. A., 2004, 101, 14011–14016.
responses.76 HDX-MS has come a long way but continues to 15 R. A. Zubarev, N. L. Kelleher and F. W. McLafferty, Electron
improve rapidly, so a firm understanding of the process is Capture Dissociation of Multiply Charged Protein Cations.
necessary for anyone who wishes to develop (rather than A Nonergodic Process, J. Am. Chem. Soc., 1998, 120,
simply use) this technique. 3265–3266.
16 J. E. P. Syka, J. J. Coon, M. J. Schroeder, J. Shabanowitz and
D. F. Hunt, Peptide and protein sequence analysis by elec-
References tron transfer dissociation mass spectrometry, Proc. Natl.
Acad. Sci. U. S. A., 2004, 101, 9528–9533.
1 C. Woodward, I. Simon and E. Tüchsen, Hydrogen 17 Z. Zhang and D. L. Smith, Determination of amide hydro-
exchange and the dynamic structure of proteins, Mol. Cell. gen exchange by mass spectrometry: a new tool for protein
Biochem., 1982, 48, 135–160. structure elucidation, Protein Sci., 1993, 2, 522–531.

2884 | Analyst, 2017, 142, 2874–2886 This journal is © The Royal Society of Chemistry 2017
View Article Online

Analyst Tutorial Review

18 J. Pan, S. Zhang, A. H. Chou and C. Borchers, Higher-order 35 S. W. Englander and N. R. Kallenbach, Hydrogen exchange
structural interrogation of antibodies using middle-down and structural dynamics of proteins and nucleic acids,
hydrogen/deuterium exchange mass spectrometry, Q. Rev. Biophys., 1983, 16, 521–655.
Chem. Sci., 2016, 7, 1480–1486. 36 T. E. Wales and J. R. Engen, Partial Unfolding of Diverse
19 P. A. McBride-Warren and D. D. Mueller, Hydrogen- SH3 Domains on a Wide Timescale, J. Mol. Biol., 2006, 357,
Deuterium Exchange of Lysozyme. I. Rate Constants and 1592–1604.
pH Dependence, Biochemistry, 1972, 11, 1785–1792. 37 J. R. Engen, T. E. Smithgall, W. H. Gmeiner and
20 J. B. Matthew and F. M. Richards, The pH Dependence of D. L. Smith, Identification and Localization of Slow,
Hydrogen Exchange in Protein, J. Biol. Chem., 1983, 258, Natural, Cooperative Unfolding in the Hematopoietic Cell
3039–3044. Kinase SH3 Domain by Amide Hydrogen Exchange and
21 Y. Bai, J. S. Milne, L. Mayne and S. W. Englander, Primary Mass Spectrometry, Biochemistry, 1997, 36, 14384–14391.
Published on 13 July 2017. Downloaded by University of Toronto on 05/11/2017 03:27:58.

structure effects on peptide group hydrogen exchange, 38 J. Hollien and S. Marqusee, Structural distribution of stabi-
Proteins: Struct., Funct. Bioinf., 1993, 17, 75–86. lity in a thermophilic enzyme, Proc. Natl. Acad. Sci. U. S. A.,
22 J. J. Englander, D. B. Calhoun and S. W. Englander, 1999, 96, 13674–13678.
Measurement and calibration of peptide group hydrogen- 39 H. Xiao, et al., Mapping protein energy landscapes with
deuterium exchange by ultraviolet spectrophotometry, amide hydrogen exchange and mass spectrometry: I. A gen-
Anal. Biochem., 1979, 92, 517–524. eralized model for a two-state protein and comparison with
23 G. P. Connelly, Y. Bai, M.-F. Jeng and S. W. Englander, experiment, Protein Sci., 2005, 14, 543–557.
Isotope effects in peptide group hydrogen exchange, 40 M. Rey, et al., Mass Spec Studio for Integrative Structural
Proteins: Struct., Funct. Bioinf., 1993, 17, 87–92. Biology, Structure, 2014, 22, 1538–1548.
24 R. S. Molday, S. W. Englander and R. G. Kallen, Primary 41 B. D. Pascal, et al., HDX Workbench: Software for the
structure effects on peptide group hydrogen exchange, Analysis of H/D Exchange MS Data, J. Am. Soc. Mass
Biochemistry, 1972, 11, 150–158. Spectrom., 2012, 23(9), 1512–1521.
25 B. H. Leichtling and I. M. Klotz, Catalysis of Hydrogen- 42 M. Guttman, D. D. Weis, J. R. Engen and K. K. Lee,
Deuterium Exchange in Polypeptides, Biochemistry, 1966, 5, Analysis of Overlapped and Noisy Hydrogen/Deuterium
4026–4037. Exchange Mass Spectra, J. Am. Soc. Mass Spectrom., 2013,
26 A. Berger, A. Loewenstein and S. Meiboom, Nuclear 24, 1906–1912.
Magnetic Resonance Study of the Protolysis and Ionization 43 R. Lindner, et al., Hexicon 2: Automated Processing of
of N-Methylacetamide1, J. Am. Chem. Soc., 1959, 81, Hydrogen-Deuterium Exchange Mass Spectrometry Data
62–67. with Improved Deuteration Distribution Estimation, J. Am.
27 I. M. Klotz and B. H. Frank, Deuterium-Hydrogen Soc. Mass Spectrom., 2014, 25, 1018–1028.
Exchange in Amide N—H Groups1, J. Am. Chem. Soc., 1965, 44 D. E. Miller, C. B. Prasannan, M. T. Villar, A. W. Fenton
87, 2721–2728. and A. Artigues, HDXFinder: Automated analysis and
28 C. L. Perrin, Proton exchange in amides: Surprises from data reporting of deuterium/hydrogen exchange mass
simple systems, Acc. Chem. Res., 1989, 22, 268–275. spectrometry, J. Am. Soc. Mass Spectrom., 2012, 23(2),
29 S. W. Englander, Hydrogen Exchange and Mass 425–429.
Spectrometry: A Historical Perspective, J. Am. Soc. Mass 45 S. Liu, et al., HDX-Analyzer: a novel package for statistical
Spectrom., 2006, 17, 1481–1489. analysis of protein structure dynamics, BMC Bioinf., 2011,
30 S. O. Nielsen, W. P. Bryan and K. Mikkelsen, Hydrogen- 12, S43.
deuterium exchange of small peptides in aqueous solution, 46 Z.-Y. Kan, L. Mayne, P. S. Chetty and S. W. Englander,
Biochim. Biophys. Acta, 1960, 42, 550–552. ExMS: Data Analysis for HX-MS Experiments, J. Am. Soc.
31 S. W. Englander, N. W. Downer and H. Teitelbaum, Mass Spectrom., 2011, 22, 1906.
Hydrogen Exchange, Annu. Rev. Biochem., 1972, 41, 903– 47 Z. Zhang, A. Zhang and G. Xiao, Improved Protein
924. Hydrogen/Deuterium Exchange Mass Spectrometry
32 D. M. Ferraro, N. D. Lazo and A. D. Robertson, EX1 Platform with Fully Automated Data Processing, Anal.
Hydrogen Exchange and Protein Folding, Biochemistry, Chem., 2012, 84, 4942–4949.
2004, 43, 587–594. 48 V. Katta, B. T. Chait and S. Carr, Conformational changes
33 J. Witten, et al., Mapping Protein Conformational in proteins probed by hydrogen-exchange electrospray-
Landscapes under Strongly Native Conditions with ionization mass spectrometry, Rapid Commun. Mass
Hydrogen Exchange Mass Spectrometry, J. Phys. Chem. B, Spectrom., 1991, 5, 214–217.
2015, 119, 10016–10024. 49 C. R. Morgan and J. R. Engen, Investigating solution-phase
34 D. D. Weis, T. E. Wales, J. R. Engen, M. Hotchko and protein structure and dynamics by hydrogen exchange
L. F. Ten Eyck, Identification and Characterization of mass spectrometry, Curr. Protoc. Protein Sci., 2009, ch. 17,
EX1 Kinetics in H/D Exchange Mass Spectrometry by Peak Unit 17.6, pp. 1–17.
Width Analysis, J. Am. Soc. Mass Spectrom., 2006, 17, 1498– 50 I. A. Kaltashov, C. E. Bobst and R. R. Abzalimov, H/D
1509. exchange and mass spectrometry in the studies of protein

This journal is © The Royal Society of Chemistry 2017 Analyst, 2017, 142, 2874–2886 | 2885
View Article Online

Tutorial Review Analyst

conformation and dynamics: Is there a need for a top-down 64 J. R. Engen and T. E. Wales, Analytical Aspects of Hydrogen
approach?, Anal. Chem., 2009, 81, 7892–7899. Exchange Mass Spectrometry, Annu. Rev. Anal. Chem., 2015,
51 T. E. Wales and J. R. Engen, Hydrogen exchange mass spec- 8, 127–148.
trometry for the analysis of protein dynamics, Mass 65 A. Kreshuk, et al., Automated detection and analysis of
Spectrom. Rev., 2006, 25, 158–170. bimodal isotope peak distributions in H/D exchange mass
52 J. J. Rosa and F. M. Richards, An experimental procedure spectrometry using HeXicon, Int. J. Mass Spectrom., 2011,
for increasing the structural resolution of chemical hydro- 302, 125–131.
gen-exchange measurements on proteins: Application to 66 T. R. Keppel, B. A. Howard and D. D. Weis, Mapping
ribonuclease S peptide, J. Mol. Biol., 1979, 133, 399–416. Unstructured Regions and Synergistic Folding in
53 L. Cravello, D. Lascoux and E. Forest, Use of different pro- Intrinsically Disordered Proteins with Amide H/D
teases working in acidic conditions to improve sequence Exchange Mass Spectrometry, Biochemistry, 2011, 50, 8722–
Published on 13 July 2017. Downloaded by University of Toronto on 05/11/2017 03:27:58.

coverage and resolution in hydrogen/deuterium exchange 8732.


of large proteins, Rapid Commun. Mass Spectrom., 2003, 17, 67 Z. Zhang, W. Li, T. M. Logan, M. Li and A. G. Marshall,
2387–2393. Human recombinant [C22A] FK506-binding protein amide
54 J. Marcoux, et al., Investigating Alternative Acidic Proteases hydrogen exchange rates from mass spectrometry match
for H/D Exchange Coupled to Mass Spectrometry: Plasmepsin and extend those from NMR, Protein Sci., 1997, 6,
2 but not Plasmepsin 4 Is Active Under Quenching 2203–2217.
Conditions, J. Am. Soc. Mass Spectrom., 2010, 21, 76–79. 68 J. Lísal, et al., Interaction of packaging motor with the poly-
55 T. Wales, M. Eggertson and J. Engen, Considerations in the merase complex of dsRNA bacteriophage, Virology, 2006,
Analysis of Hydrogen Exchange Mass Spectrometry Data, in 351, 73–79.
Mass Spectrometry Data Analysis in Proteomics, ed. 69 J. Lísal, et al., Functional visualization of viral molecular
R. Matthiesen, Humana Press, 2013, pp. 263–288. motor by hydrogen-deuterium exchange reveals transient
56 D. F. Hunt, J. R. Yates, J. Shabanowitz, S. Winston and states, Nat. Struct. Mol. Biol., 2005, 12, 460–466.
C. R. Hauer, Protein sequencing by tandem mass spec- 70 A. Chandramohan, et al., Predicting Allosteric Effects from
trometry, Proc. Natl. Acad. Sci. U. S. A., 1986, 83, 6233–6237. Orthosteric Binding in Hsp90-Ligand Interactions:
57 J. D. Venable, M.-Q. Dong, J. Wohlschlegel, A. Dillin and Implications for Fragment-Based Drug Design, PLoS
J. R. Yates, Automated approach for quantitative analysis of Comput. Biol., 2016, 12, e1004840.
complex peptide mixtures from tandem mass spectra, Nat. 71 S. Krishnamurthy, et al., Distinguishing direct binding
Methods, 2004, 1, 39–45. interactions from allosteric effects in the protease–HK97
58 R. S. Plumb, et al., UPLC/MSE; a new approach for generat- prohead I δ domain complex by amide H/D exchange mass
ing molecular fragment information for biomarker struc- spectrometry, Bacteriophage, 2014, 4, e959816.
ture elucidation, Rapid Commun, Mass Spectrom., 2006, 20, 72 T. Rob, P. K. Gill, D. Golemi-Kotra and D. J. Wilson, An
1989–1994. electrospray ms-coupled microfluidic device for sub-second
59 J. K. Chik, J. L. Vande Graaf and D. C. Schriemer, hydrogen/deuterium exchange pulse-labelling reveals allo-
Quantitating the Statistical Distribution of Deuterium steric effects in enzyme inhibition, Lab Chip, 2013, 13,
Incorporation To Extend the Utility of H/D Exchange MS 2528–2532.
Data, Anal. Chem., 2006, 78, 207–214. 73 M. A. Sowole, S. Simpson, Y. V. Skovpen, D. R. J. Palmer and
60 J. Zhang, P. Ramachandran, R. Kumar and M. L. Gross, L. Konermann, Evidence of Allosteric Enzyme Regulation via
H/D Exchange Centroid Monitoring is Insufficient to Show Changes in Conformational Dynamics: A Hydrogen/
Differences in the Behavior of Protein States, J. Am. Soc. Deuterium Exchange Investigation of Dihydrodipicolinate
Mass Spectrom., 2013, 24, 450–453. Synthase, Biochemistry, 2016, 55, 5413–5422.
61 P. L. Ferguson, et al., Hydrogen/deuterium scrambling 74 D. Houde, S. A. Berkowitz and J. R. Engen, The utility of
during quadrupole time-of-flight MS/MS analysis of a zinc- hydrogen/deuterium exchange mass spectrometry in bio-
binding protein domain, Anal. Chem., 2007, 79, 153–160. pharmaceutical comparability studies, J. Pharm. Sci., 2011,
62 Z. Zhang, S. Guan and A. G. Marshall, Enhancement of the 100, 2071–2086.
effective resolution of mass spectra of high-mass bio- 75 S.-W. Yang, et al., Guanine nucleotide induced confor-
molecules by maximum entropy-based deconvolution to mational change of Cdc42 revealed by hydrogen/deuterium
eliminate the isotopic natural abundance distribution, exchange mass spectrometry, Biochim. Biophys. Acta,
J. Am. Soc. Mass Spectrom., 1997, 8, 659–670. Proteins Proteomics, 2016, 1864, 42–51.
63 R. E. Iacob, J. P. Murphy and J. R. Engen, Ion mobility adds 76 R. G. McAllister and L. Konermann, Challenges in the
an additional dimension to mass spectrometric analysis of Interpretation of Protein H/D Exchange Data: A Molecular
solution-phase hydrogen/deuterium exchange, Rapid Dynamics Simulation Perspective, Biochemistry, 2015, 54,
Commun. Mass Spectrom., 2008, 22, 2898–2904. 2683–2692.

2886 | Analyst, 2017, 142, 2874–2886 This journal is © The Royal Society of Chemistry 2017

You might also like