You are on page 1of 15

937154

review-article2020
JHCXXX10.1369/0022155420937154Analysis of the GlycosaminoglycansSong et al.

Review
Journal of Histochemistry & Cytochemistry 1­–15
© The Author(s) 2020
Article reuse guidelines:
sagepub.com/journals-permissions
DOI: 10.1369/0022155420937154
https://doi.org/10.1369/0022155420937154
journals.sagepub.com/home/jhc

Analysis of the Glycosaminoglycan Chains of Proteoglycans

Yuefan Song, Fuming Zhang, and Robert J. Linhardt


National R & D Branch Center for Seaweed Processing, College of Food Science and Engineering, Dalian Ocean University, Dalian, P.R. China (YS), and
Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York (YS, FZ, RJL)

Journal of Histochemistry & Cytochemistry


Summary
Glycosaminoglycans (GAGs) are heterogeneous, negatively charged, macromolecules that are found in animal tissues.
Based on the form of component sugar, GAGs have been categorized into four different families: heparin/heparan sulfate,
chondroitin/dermatan sulfate, keratan sulfate, and hyaluronan. GAGs engage in biological pathway regulation through their
interaction with protein ligands. Detailed structural information on GAG chains is required to further understanding of
GAG–ligand interactions. However, polysaccharide sequencing has lagged behind protein and DNA sequencing due to the
non-template-driven biosynthesis of glycans. In this review, we summarize recent progress in the analysis of GAG chains,
specifically focusing on techniques related to mass spectroscopy (MS), including separation techniques coupled to MS,
tandem MS, and bioinformatics software for MS spectrum interpretation. Progress in the use of other structural analysis
tools, such as nuclear magnetic resonance (NMR) and hyphenated techniques, is included to provide a comprehensive
perspective.  (J Histochem Cytochem XX:XXX–XXX, XXXX)

Keywords
glycosaminoglycans, mass spectroscopy, NMR, sequencing, structural analysis

Introduction heparan sulfate (HP/HS), chondroitin/dermatan sulfate


(CS/DS), keratan sulfate (KS), hyaluronan (HA). Among
Glycosaminoglycans (GAGs) are biological macro­ the four families of GAGs, HP/HS, CS/DS, and KS are
molecules ubiquitously present in animal tissues. GAG all conjugated to core proteins as PGs and are synthe­
chains bound to a core protein constitute a proteogly­ sized principally in the Golgi apparatus of cells,
can (PG), which is considered among the most struc­ whereas the HA is synthesized in the cellular plasma
turally complex glycoconjugates. The core protein of a membrane unlinked to a protein core.2
PG is synthesized by a template-driven process, after The HP/HS are bioactive GAGs O-glycosidically
which GAG chains are added in a non-template-driven linked to a serine residue of the core protein through a
synthesis, resulting in the remarkable structural and tetrasaccharide linkage region. The disaccharide
functional diversity of PGs. The heterogeneity of the building block of this family is →4) α-L-IdoA /β-D-GlcA
GAG chains greatly contributes to PG complexity.
GAGs are unbranched, linear anionic polysaccharides
composed of 10 to 200 repeating disaccharide units.1 Received for publication March 18, 2020; accepted May 29, 2020.
Each disaccharide building block consists of an
amino sugar (i.e., N-acetyl-glucosamine [GlcNAc], or Corresponding Authors:
Yuefan Song, National R & D Branch Center for Seaweed Processing,
N-acetyl-galactosamine [GalNAc]) and a uronic acid College of Food Science and Engineering, Dalian Ocean University,
(i.e., glucuronic acid [GlcA], or iduronic acid [IdoA]) or Dalian 116023, P.R. China.
D-galactose. These disaccharide units can be O- or Email: syf@dlou.edu.cn
N- substituted with sulfo groups at different positions. Robert J. Linhardt, Center for Biotechnology and Interdisciplinary
Based on the form of component sugar, GAGs have Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA.
been classified into four different families: heparin/ Email: linhar@rpi.edu
2 Song et al.

(1→4) α-D-GlcNS/-D-GlcNAc (1→. In HP, IdoA growing amount of evidence suggested potential roles
accounts for >70% of the hexuronic acid. The IdoA for KS in cell-signaling processes.11
residue in HP is generally 2-sulfated and the glucos­ HA is the only GAG family that is neither sulfated
amine can be N- and/or 6-sulfated. In contrast to HP, nor linked to a protein core. However, HA is typically
HS is majorly composed of GlcA as its hexuronic acid, bound to matrix proteins in the extracellular space.12 It
and generally contains one sulfo group per disaccha­ has a simple repeating disaccharide unit of →4) β-D-
ride repeating unit. HS can be N-sulfated, 6-sulfated, GlcA(1→3)β-D-GlcNAc(1→. HA is a significant bio­
and 3-sulfated on its glucosamine residue and 2-sul­ compatible support material used in wound healing or
fated on its uronic acid residue.3 While HS is widely as growth scaffolds in surgery. HA has also been
distributed on membrane and extracellular PGs, the widely used for providing lubrication and mechanical
HP PG is found primarily localized to intracellular gran­ support for the joints, or serving as a drug delivery
ules of mast cells. HP has multifaceted biological func­ material.13
tions, including its well-known anticoagulant activity The numerous physiological functions of GAGs are
and other bioactivities such as anti-inflammatory and carried out by their interaction with protein ligand.14
anticancer activities.2,4,5 The complex biological activ­ These interactions are often specific between GAGs
ity of HS-GAGs includes their roles in developmental and ligands, requiring a defined GAG sequence or
biology and cell signaling.4 HS PGs engage in various domain. Therefore, structural elucidation of GAG chains
physiological and pathological processes, including is a prerequisite for exploring the mechanisms of GAG–
embryonic development, adult tissue homeostasis, ligand interactions. Due to the non-template-driven bio­
aging, and infection.6,7 synthesis of GAGs, the development of GAG sequencing
The CS/DS GAGs also occur as PGs O-glycosidically has lagged behind the sequencing of other biological
linked to the serine residues of their core proteins. The macromolecular counterparts, protein and DNA. Thus
GAGs of this family contain repeating disaccharide far, there have been only two cases reporting GAG
unit represented as →4) β-D-GlcA /α-L-IdoA (1→3)β- sequencing and both relatively simple GAGs, bikunin15
D-GalNAc(1→. The GlcA is the hexuronic acid found in and decorin.16 Currently, the prevailing method for
CS, and IdoA is the hexuronic acid found in DS.8 Unlike GAG chain determination is mass spectrometry (MS).
heparin, the CS and DS have no N-sulfo-group substi­ Nuclear magnetic resonance (NMR) also represents an
tution. The GalNAc can be sulfated at 4-O and/or 6-O option for establishing GAG structure. As summarized
positions. The hexuronic acid residues are sulfated at in Fig. 1, GAG samples are prepared through extensive
the 2-O, and the IdoA residues are rarely sulfated at separation processes and then subjected to MS and/or
3-O positions. CS PGs serve as one of the major bar­ NMR analysis using top-down or bottom-up sequencing
rier-forming molecules in the central nervous system. strategies. This review focuses on recent developments
CS is often found as a hybrid structure with DS and is of GAG analysis, including separation techniques cou­
involved in the formation of the neural network by cap­ pled to MS, tandem MS, MS data interpretation, NMR,
turing and presenting growth factors to stem cells or and other novel techniques applied for the characteriza­
neuronal cells.9 tion of GAGs with the purpose of providing useful infor­
KS has a galactose residue as its major backbone mation for glycomics research.
constitute instead of uronic acid. KS is 6-O sulfated
GAGs with the disaccharide building block, →3) β-D-
Preparation of GAGs for Analysis
Gal(1→4) β-D-GlcNAc (1→. KS is subdivided into
three classes according to the ways it is linked to the The native GAG is generally a component of a PG
core protein.2 KS I (corneal type) is N-glycosidically found in a biological sample. Thus, a procedure is
conjugated through a biantennary branched linkage needed for the extraction and purification to obtain
region to an asparagine residue of its core protein. KS GAG samples of sufficient purity for subsequent struc­
II (skeletal type) is O-glycosidically linked to a serine or ture analysis. Detailed methods for preparing PGs and
threonine residue of its core protein. K III is O-linked GAGs from biological origins are extensively described
through mannose to serine, and is preferentially found in several review articles.1,4,17 PGs that are present in
in PG of brain and nervous tissue.10 Generally, the physiological fluids or secreted into the fermentation
GAG chains of KS I are longer and less sulfated than media can directly be obtain by dialysis, ultrafiltration,
that of KSII. Also, KS I can be distinguished from KS II and/or applying the sample to a fractionation column.
by its capped non-reducing end.1 KS is believed to be However, preparing PGs and GAGs from large amounts
the newest GAG from an evolutionary perspective and of animal tissues can be very time-consuming and
its biological functions are the least understood. A labor-intensive. A particularly complex protocol was
Analysis of the Glycosaminoglycans 3

Figure 1.  A scheme for GAGs chain analysis. Abbreviations: GAGs, glycosaminoglycans; AEC, anion-exchange chromatography; SEC,
size-exclusion chromatography; GPC, gel permeation chromatography; LC, liquid chromatography (other than AEC and SEC, including
reverse phase liquid chromatography, reverse phase ion-pairing liquid chromatography, and hydrophilic interaction chromatography);
PAGE, polyacrylamide gel electrophoresis; CE, capillary electrophoresis; MS, mass spectroscopy; IMS, ion mobility spectrometry; NMR,
nuclear magnetic resonance; COSY, correlation spectroscopy; TOCSY, total correlation spectroscopy; NOESY, nuclear Overhauser
effect spectroscopy; HSQC, heteronuclear single-quantum coherence; HMBC, heteronuclear multiple bond coherence.

described to obtain decorin PGs and GAGs from por­ obtained from PAGE can often be directly applied to
cine skin for use in sequencing studies.18 In brief, tissues MS analysis for sequencing.16,19
or cells were defatted, sliced, ground, or homogenized,
and a buffered solution of chaotropic agent was used to
extract the PGs. The extracts were dialyzed or applied GAG Chain Analysis
to an anion-exchange column (AEC), the PGs were MS-based Analysis
treated with nonspecific proteases, and the GAGs were
released from core peptides using controlled β- General Introduction.  Mass spectrometry plays a leading
elimination. The resulting crude GAGs can be collected role in glycomics. Although other tools have been used
by ultrafiltration or through solvent precipitation.1,17 in the early stages of glycan analysis, such as NMR
The crude GAG extract, containing impurities and or the gel electrophoresis–based sequencing, mass
chains of many sizes, needs to be properly enriched spectroscopy has become preeminent due to its high
and purified before it can be successfully interrogated resolution, efficiency in providing detailed structural
by MS or NMR. There are a variety of separation tech­ information, adaptability for coupling separation tech­
niques that can be used to purify and fractionate GAGs, niques, and a reduced demand for sample quantity.
including AEC, size-exclusion chromatography (SEC), The invention of soft-ionization techniques has
reverse phase liquid chromatography (RP-LC), reverse tremendously facilitated the application of MS for the
phase ion-pairing LC (RPIP-LC), and hydrophilic inter­ analysis of biological macromolecules. The two major
action chromatography (HILIC). RP-LC, RPIP-LC, and soft-ionization techniques are matrix-assisted laser
HILIC methods for GAG separations are often applied desorption/ionization (MALDI) and electrospray ioniza­
as a hyphenated technique using an HPLC-MS plat­ tion (ESI). The application of MALDI in GAG analysis
form. These methods will be introduced later in MS has, however, been limited due to the propensity for
section. In addition to the above-mentioned chro­ labile sulfo groups to undergo decomposition. Thus, ESI
matographic techniques, preparative polyacrylamide is most frequently applied to GAG analysis. ESI in the
gel electrophoresis (PAGE) represents another good negative-ion mode has been shown to be suitable for
option for the purification of GAGs. The GAG fractions analyzing negatively charged GAGs as well as
4 Song et al.

preserving their sulfo groups.20 ESI is quite adaptable chemical or enzymatic methods. Such an approach is
and can be coupled to a variety of separation tech­ essential for analyzing polysaccharides of very high
niques, including LC, and capillary electrophoresis (CE). molecular weight. The resulting oligosaccharides
Another major MS breakthrough facilitating glycom­ obtained on controlled depolymerization are usually
ics research is the widespread availability of Fourier then subjected to a separation procedure such as LC,
transform mass spectrometry (FTMS), including both CE, or ion mobility (IM) for structural characteriza­
Fourier transform ion cyclotron resonance (FT-ICR) tion.24,26–28 Controlled chemical and enzymatic meth­
and Orbitrap mass spectrometers.21 FTMS provides ods for GAG depolymerization have been described in
high resolution and mass accuracy, allowing unam­ detail.1 Compared with a top-down strategy, the bot­
biguous peak assignments that are essential in ana­ tom-up requires more steps to prepare samples, and
lyzing heterogeneous glycans. the depolymerization procedure may result in the loss
A mass spectrum can provide general information of substantial amounts of structural information.
about the GAG composition, such as degree of polym­ Even when performing top-down sequencing, it is
erization and number of sulfo-group substitutions. quite common to use a bottom-up strategy to provide
However, such information is insufficient to fully char­ additional supporting information on disaccharide units
acterize a GAG chain. Tandem mass spectrometry is or domain structures. In this case, the GAGs are digested
required to gain a deeper insight of the precise struc­ with a combination of different enzymes called GAG
ture of GAG chains. Electron-based ion activation lyases.16 The determination of building block disaccha­
techniques, such as electron detachment dissociation rides can help narrow down peak assignments in the
(EDD) and negative electron transfer dissociation MS spectra, and enables more confident interpretation
(NETD), are capable of generating extensive cross- of MS data.
ring cleavages, allowing identification of epimers, dis­
tinguishing between IdoA and GlcA, and determining Separation Techniques. A sample needs to be properly
the position of sulfo-group substitutions.22 Also, elec­ purified before being examined by MS to undertake
tron-based ion activation largely prevents the loss of the sequencing of a GAG chain. A hyphenated purifi­
sulfo group.23 Although the loss of sulfo groups can cation method with MS detection can be achieved by
also be prevented by deprotonation of sulfo groups or using different separation techniques coupled with
addition of metal adducts.21 Moreover, the application MS. The major separation techniques adapted to MS
of EDD or NETD can provide information of the same coupling are LC methods, including RP-LC, RPIP-LC,
and different ions to aid the deconvolution of the oligo­ HILIC, SEC, porous graphitized carbon (PGC), and
saccharide structure for GAG sequencing. other separation techniques such as CE and IM. There
State-of-the-art MS and tandem MS techniques are a considerable number of review articles on com­
have been major technical achievements responsible bining separations with MS in GAG analysis.28–37 Thus,
for much of the success in GAG analysis. It is expected the following section focuses on only work published in
that the future will bring more progress in glycomic the past few years.
analysis as new analytical instruments and methods
are developed. LC-based Separation.  RP-LC is the most wildly used chro­
matography method. However, due to the polyanionic
Top-down and Bottom-up Strategies.  The structural charac­ features of GAGs, this method is not directly suitable
terization of GAGs can be achieved through different for GAG separations. This limitation can be addressed
types of instrumental analysis, but such characteriza­ by modifying GAG oligosaccharides to make them
tion basically follows two major strategies, referred to amenable to RP-LC. A rapid separation and detection
as top-down (Fig. 2) and bottom-up (Fig. 3). Applying a method for HP/HS disaccharide analysis, relying on
top-down strategy means a purified intact GAG chain is 2-aminoacridone (AMAC) derivatization, was built to
directly subjected to structural analysis without a pre- examine samples by an optimized selected ion record­
depolymerization step.15,16,18,24 The development of MS ing (SIR) RP-ultraperformance liquid chromatography
techniques, especially the emergence of FTMS that (UPLC)-MS38 without requiring the removal of excess
provides high spectral resolution and high mass accu­ unreacted reagent. Liu and coworkers39 also used
racy, has facilitated the application of top-down MS AMAC to label disaccharides derived from CS and HS
strategies. A top-down strategy affords detailed infor­ of human intervertebral disc samples. An RP-LC cou­
mation on the sequence of an intact GAG chain with pled with triple quadruple mass was used for online MS
reduced requirements for sample preparation steps.25 analysis done by multiple reaction monitoring (MRM).
In contrast, in a bottom-up approach (Fig. 3), intact Principal components analysis identified clear separa­
GAG chains are depolymerized using controlled tion of GAG profiles between nucleus pulposus and
Analysis of the Glycosaminoglycans 5

Figure 2.  A mainframe of a typical top-down GAG analysis. Decorin GAGs were purified and subjected to FTMS and MS/MS analysis
for sequencing. Abbreviations: GAGs, glycosaminoglycans; FTMS, Fourier transform mass spectrometry; MS, mass spectroscopy; PAGE,
polyacrylamide gel electrophoresis; FT-ICR, Fourier transform ion cyclotron resonance; CID, collision-induced dissociation. (Adapted
from: Yu Y, Duan J, Leach IIIFE, Toida T, Higashi K, Zhang H, Zhang F, Amster IJ, Linhardt RJ. Sequencing the dermatan sulfate chain of
decorin. J Am Chem Soc. 2017;139(46):16986–16995).
6 Song et al.

Figure 3.  A mainframe of a typical bottom-up GAG analysis. Low-molecular-weight heparins were depolymerized and subjected to
LC-MS and MS/MS analysis. Abbreviations: GAGs, glycosaminoglycans; LC, liquid chromatography; MS, mass spectroscopy; HILIC,
hydrophilic interaction chromatography; LC, liquid chromatography; FTMS, Fourier transform mass spectrometry (Adapted from: Li
G, Steppich J, Wang Z, Sun Y, Xue C, Linhardt RJ, Li L. Bottom-up low molecular weight heparin analysis using liquid chromatography-
Fourier transform mass spectrometry for extensive characterization. Anal Chem. 2014;86(13):6626–6632).

annulus fibrosus in specimens from young and old solely by the order of their N-sulfation and N-acetyla­
patients. In the research carried out by the Sharp group,40 tion. A stable isotope-labeled hydrazide tag (INLIGHT)
a method using trideuteroacetyl derivatization and RP- was used for labeling oligosaccharides derived from
LC-MS/MS was developed to sequence a complex digested heparin and enabled identification of a total
mixture of HS tetrasaccharides. Liang and coworkers41 amount of 116 unique oligosaccharides.42 Derivatiza­
introduced a propionylation method to replace sulfo tion and labeling are frequently used in glycomics to
groups from oligosaccharide and used LC-MS/MS with enhance RP-LC separation for oligosaccharides
coupled C18 columns to separate and fully sequence and for a variety of other purposes, including stabiliz­
two mixed tetrasaccharide isomers, which differed ing residues, serving as a linker for oligosaccharide
Analysis of the Glycosaminoglycans 7

attachment, facilitating ultraviolet/fluorescent/MS detec­ with common stationary phase materials. In a PGC-LC
tion, and supporting detailed structural characterization experiment, longer equilibration time is required to pro­
by tandem mass.37 Recently published works using duce repeatable results.25 However, PGC-LC-ESI-MS
double labeling43 and sequential labeling44 both have analysis is characterized by a high isomer separation
demonstrated the virtue of labeling for the subsequent power enabling a specific glycan compound analysis
detection. on the level of individual structures, and is well
GAG oligosaccharides can be separated and ana­ described in Stavenhagen et  al.’s56 review. Turnbull
lyzed in an RP-LC-MS platform without derivatization by and coworkers recently applied a commercial PGC
adding ion-pairing reagent (IPR) to the mobile phase. column to an LC-MS system to investigate digested
An alkylamine ion-pairing reagent can essentially neu­ HS oligosaccharide mixtures. The method was effec­
tralize the ionic features of GAG oligomers. This offers tive and allowed the separation of oligosaccharide
the advantage of enhanced volatility, especially through from tetrasaccharide to octasaccharide, enabling
a pH change above the pKa after the separation, more confident interpretation of the MS/MS data.57
facilitating coupled MS detection.45 Reagents such as
tributylamine,24,45 n-pentylamine,46 n-hexylamine,47 CE.  In addition to LC-based methods, CE is another
and n-butylamine48 are often used as anion-pairing attractive option for GAG analysis amenable to MS
reagent to increase separation efficiency in RPIR-LC coupling. CE has been shown to be superior in multi­
system for GAG analysis. Persson et al.48 developed a ple aspects such as a high separation efficiency, short
novel approach based on dibutylamine RPIP-LC-MS/ analysis time, low consumption of sample, and repro­
MS platform and used in disaccharide fingerprinting, ducibility.34,58 Linhardt and coworkers used a reversed-
revealing differences in xyloside-primed CD/DC as well polarity CE coupled with LTQ Orbitrap MS method to
as sialylation of the linkage region from two different analyze HP/HS disaccharides, Arixtra, and low-molec­
cell lines. The N-unsubstituted glucosamine residues in ular-weight heparin (LMWH) building blocks. The
HP/HS were examined using RPIP-LC (reversed phase results obtained were comparable or better than those
ion-pairing liquid chromatography)-MS method. By add­ obtained when these analyses were performed using
ing hexylamine47 and pentylamine,46 N-unsubstituted LC-MS.59 This method could also separate oligosac­
disaccharides/oligosaccharides were well separated charides from tetrasaccharides to dodecasaccharides,
and characterized. elucidating sulfation position and epimeric structural
HILIC is a preferential method for polar compound differences, and could also determine more than 80
separations and it is also easily coupled with MS. molecular compositions from complex GAG mixture.60
Therefore, it has become a popular approach in GAG Recent work published by Stickney et al.61 combined,
analysis. HILIC is efficiently coupled with LC-Orbitrap for the first time, negative electron transfer dissociation
MS for oligosaccharide characterization.24,27 HILIC has (NETD) and CE for assigning the structures of GAG
also been coupled with triple quadrupole M/M with an oligomers. This approach allowed enoxaparin (a com­
MRM approach to analyze and quantify heparin build­ plex mixture of LMWHs) to be separated within 30 min,
ing blocks.49,50 HILIC can be combined with weak anion 37 unique molecular compositions be identified, and
exchange (WAX) method. Turiák and coworkers51 cou­ nine structures were assigned with tandem MS.
pled self-packed HILIC-WAX capillary columns to a
UPLC Q-Tof MS system, revealing that HS disaccha­ Ion Mobility Spectrometry (IMS).  IMS is a new technique,
ride composition varies in different grades of prostate which emerged as a promising choice for GAG sepa­
cancer tissues. Zhang and coworkers used UPLC- ration. Different from LC and CE, IMS is a postioniza­
HILIC/WAX MS/MS equipped with a commercial HILIC/ tion, gas-phase method, providing fast separation, and
WAX column to evaluate both the epimerization and easy to be coupled to MS.23 IMS defines how an ion
composition of heparin and dalteparin.52 HILIC can drifts through a gas buffer under the influence of a
also be used to separate derivatized oligosaccharides. varying or fixed electric field. Different ions are sepa­
Antia and coworkers used a HILIC-ESI-MS in positive- rated according to their motion, which are affected by
ion mode to investigate procainamide-derivatized HS both the acceleration caused by the electric field and
disaccharides, completing separation of eight HS the deceleration caused by collisions with gas mole­
disaccharides together with an internal standard.53 A cules. The ion’s motion is associated with molecular
HILIC method was used in conjunction with a microflu­ mass, charge, size, and shape. Thus, IMS is consid­
idic chip LC-MS and provided an efficient separation of ered to be a powerful tool for separating isomers. A
GAGs.54,55 variety of IMS methods have been developed, such as
Porous graphitized carbon (PGC) can be used as a field asymmetric IMS (FAIMS), trapped IMS (TIMS),
stationary phase in LC. PGC is less durable compared traveling wave ion mobility (TWIMS), and drift Tube
8 Song et al.

IMS (DTIMS). A combined FAIMS-FTICR-MS/MS was EDD also minimizes sulfo-group loss, which is a major
used in GAG analysis and was shown to be capable of complication when using the CID method. Research
resolving isomeric and isobaric GAG negative ions carried out by Amster and coworkers has confirmed
having the same mass-to-charge ratio, and it also facili­ the benefit of using EDD for identification of C5 uronic
tated MS/MS analysis.62 Wei and coworkers23 used acid epimers.22,62,70 According to these reports, EDD
gated-trapped ion mobility spectrometry (gated-TIMS), can generate different ion fragment patterns for
which allowed the ions of a given mobility selected by 2-O-sulfo uronic acid epimeric tetrasaccharides.70
an electrical gate and accumulated in a low-pressure Further research confirmed the efficacy of using these
collision cell. Highly sulfated HP/HS isomers were ions for assigning the C-5 stereochemistry of the
separated by gated-TIMS and examined by coupling reducing end uronic acid in 33 HS tetrasaccharides.22
NETD MS/MS. The method was useful for both quali­ The combination with high-field asymmetric waveform
tative and quantitative GAG analysis. A TWIMS ion mobility spectrometry (FAIMS) was described pre­
method was used to investigate changes in the gas- viously. EDD was able to distinguish the two sets of
phase conformation of antithrombin III upon binding epimeric GAG tetramers separated in FAIMS method.62
of Arixtra, validating the utility of IMS to measure pro­ Although EDD can provide rich informative frag­
tein conformational changes induced by the binding ments, it is a relatively slow technique intended for
of GAG ligands.63 Miller and coworkers64 used ion FT-ICR because it requires long acquisition times and
mobility mass spectrometry and tandem MS to inves­ is not conducive to a high-throughput platform. Another
tigate heparin/HS-like isomers. Six synthetically pro­ electron activation method NETD shows its promise in
duced octasaccharides, which are isomeric with both generating sequence-informative fragmentation
regard to either glucuronic acid (GlcA) or iduronic acid spectra and improving throughput rate (i.e., 1 s for EDD
(IdoA) residues at various positions, were analyzed vs. 0.1 s for NETD).21 NETD is as excellent as EDD
and differentiated. Changing from IdoA to GlcA in spe­ in generating extensive, structurally informative frag­
cific locations resulted in strong conformational distor­ ments and minimizing the occurrence of sulfate loss
tions, showing the importance of sequence to overall peaks.71 NETD can enable structural characterization
conformation. of synthetic HS isomers containing 3-O-sulfation.72 In
In addition, hyphenated separations have also been recent years, NETD has been applied in GAG analysis
used in research, such as combination of RPIP to when coupled to new separation techniques such as
HILIC,65 and RPIP44 or SEC66 coupled to AEC. These CE61 and gated-TIMS.23 NETD was found well adapted
separation methods play significant roles in the analy­ for these separation approaches, providing efficient
sis of glycan and GAG structures. information for stereoisomer identification, and negligi­
ble sulfo losses.
Tandem MS.  Tandem MS plays a crucial role in char­ Besides the electron-based ion activation, a photo­
acterization of GAG structure. It generates glycosidic dissociation method, referring to ultraviolet photodis­
and cross-ring cleavage, leading to fragmentation sociation (UVPD), has been suggested for the analysis
of precursor ions. Fragments, particularly cross-ring of GAG chains.21 UVPD has also been applied in pro­
ones, can provide detailed information about the con­ teomics and some preliminary studies targeting oligo­
figuration and modification of each monosaccharide saccharides.73,74 Further exploration is needed for its
residue, allowing the GAG structures to be fully application in glycomics.
characterized.
There have been different kinds of ion activation Structural Interpretation.  Tandem MS is a powerful tool to
methods applied in tandem MS, including collision- reveal the complex structure of GAG chains. Fragments
induced dissociation (CID),67 infrared multi-photon generated by tandem MS are crucial for establishing a
dissociation (IRMPD),68 electron-induced association precursor’s structure. The fragmentation of oligosac­
(EID),69 EDD,22,62,68,70 and NETD.23,61,71,72 CID, an ion­ charide follows a certain pattern, and a nomenclature
ization method based on collision-activation, has been has been established by Domon and Costello.21,75,76
widely applied for characterizing GAG chains. Intact The processing of an MS spectrum into meaningful
GAG chains have been sequenced using CID by information is a time-consuming task. Manual interpre­
Linhardt and coworkers.15,16 tation of tandem data sets required for GAG oligosac­
In recent years, the electron-based ionization meth­ charide analysis may take many hours or even days.17
ods such as EDD and NETD have become new trends Hence, suitable bioinformatics software is essential to
in glycomics research. EDD can produce more infor­ assist data interpretation. Many labs have developed
mative spectra of both cross-ring and glycosidic cleav­ tools for automated or semi-automated interpretation of
age product ions, while CID and IRMPD are not as MS data for GAG analysis. The following section is an
efficient as EDD in causing cross-ring cleavages.68 introduction to these programs.
Analysis of the Glycosaminoglycans 9

HOST is a computational tool early developed to spectra of glycan standards.85 A tandem mass spec­
help HP/HS oligosaccharide sequencing using enzy­ trum peak finding program was recently developed by
matic digestion and ESI-MSn. Based on disaccharide Hogan and coworkers specifically for GAGs. The pro­
composition analysis, the program scores and returns gram being called GAGfinder uses precursor composi­
the most likely sequence.21,77 The widely used soft­ tion information to generate all theoretical fragments,
ware, GlycoWorkbench is a package designed to which is a targeted, brute force approach to carry out
semi-automatically annotate glycomics data. It can be spectrum interpretation.86
used to annotate MS and MS/MS spectra of free oligo­ There have been major achievements in MS spec­
saccharides, N- and O-linked glycans, GAGs and gly­ trum interpretation in the last few years. However,
colipids, as well as MS spectra of glycoproteins.78 some of the tools are more suitable for specific types
GlycoWorkbench allows the user to draw a glycan of GAGs and certain workflows. A more rapid, high-
structure and calculate a list of theoretical fragment throughput, universal and user-friendly software will
ions.21 Another popular bioinformatics software called need to be developed for future GAG analysis.
GlycReSoft was developed for automated recognition
of glycans from LC/MS data.79 It is able to automati­
NMR
cally assign structures within 5-ppm mass accuracy.80
The software GAD-ID is the first to automate the inter­ In addition to MS-based techniques, NMR spectros­
pretation of mixtures when coupled to LC-MS/MS, but copy has become another powerful technique for
it requires derivatization to replace sulfo group with determining GAG structures. NMR permits structural
acetyl groups. This tool uses a scoring system based analysis directly on unmodified GAGs using isotopes
1
on peak intensities and could properly assign 21 syn­ H, 13C, and 15N.1,87 The most basic NMR approaches
thetic tetrasaccharides in a defined mixture from a used for GAG analysis are 1H and 13C mono-dimen­
single LC-MS/MS run.81 Recently, a statistical model, sional NMR (1D NMR). A 1H-NMR spectrum is quite
which using a multivariate expectation maximization often used in distinguishing IdoA and GlcA residues,
algorithm, was build to estimate the accuracy of deriva­ revealing monosaccharide compositions, and assign­
tized HP/HS assignments to tandem mass (MS/MS) ing anomeric configurations. 13C-NMR is less sensitive
spectra made by GAG-ID. This analysis makes it pos­ than 1H-NMR but offers more dispersion of chemical
sible to filter large MS/MS database search results shift resulting in less signal overlap.1 13C-NMR spec­
with predictable false identification error rates.82 For troscopy is particularly useful in determining sulfation
HP/HS analysis, GlycCompSoft is designed to enable pattern and positional linkages of polysaccharides.
the comparison of top-down analytical glycomics data However, due to the structural complexity or impurity
on two or more LWMHs. In a test based on three lots of some GAG samples, the area of signals in 1D
of Lovenox, Clexane, and three generic enoxaparin NMR spectra is often very complicated and affected
samples, the program proved its low error rate and by severe signal overlap. In this case, multidimen­
good time efficiency when processing large data sets. sional NMR spectroscopy is required to help assign
Hu and coworkers developed an algorithm to convert signals, complete structural characterization, or even
the raw data from electron-based dissociation (ExD) quantify the composition of sugar residues.88 The
tandem mass spectra into HS saccharide sequences.83 commonly used multidimensional NMR in examining
A genetic algorithm has been used by Duan and glycans is two-dimensional (2D) NMR both homonu­
Amster to design a de novo approach for rapid, high- clear and heteronuclear, including 1H–1H correlation
throughput GAG analysis. The program is coded in a spectroscopy (COSY), total correlation spectroscopy
software package using the MATLAB environment. By (TOCSY), nuclear Overhauser effect spectroscopy
using a genetic algorithm, the search space of iso­ (NOESY), rotating frame Overhauser effect spectros­
meric structures that are considered is greatly reduced, copy (ROESY), 1H–13C heteronuclear single-quan­
which enables the analysis time to be reduced to a tum coherence (HSQC), and heteronuclear multiple
maximum of a few minutes. This approach was tested bond coherence (HMBC).
and properly identified structures from MS2 data of NMR is an attractive technique for GAG analysis
GAG chain from bikunin.84 Machine learning strategy because it directly interrogates a sample requiring no
was introduced by Hong and coworkers to develop derivatization procedure and results in no sulfo loss.
GlycoDeNovo. This is a novel algorithm that can auto­ As NMR is nondestructive, it allows a sample to be
matically learn fragmentation patterns from real-world further used for other analytical methods. Moreover,
tandem MS data. It contains a data-driven IonClassifier, NMR is sensitive to conformational changes and can
to which machine learning is applied to distinguish B provide information on the secondary structure of
and C ions from other ion types using experimental GAGs and their complexes with proteins.1 However,
10 Song et al.

the NMR spectroscopy is limited by its relatively low ratio. Beccati and coworkers93 reported an integrated
sensitivity, and usually requires milligram amounts of approach using IPRP-HPLC, LC-MS, NMR, SEC-MS,
pure samples for glycan analysis. In a protocol pub­ and ESI-Q-TOF-MS to characterize bovine kidney HS,
lished by Carnachan and Hinkley,89 a concentration of providing overlapping and confirmatory information
17 mg/mL of GAG sample was the minimum for highly from different perspectives for the determination of the
reproducible HS analysis using 2D NMR. Increasing GAG structure. Recently, some research carried out
the strength of the static magnetic field can help using hyphenated or individual spectroscopic tech­
improve NMR sensitivity. However, it is hard to detect a niques was also quite inspiring. Renois-Predelus and
minor monosaccharide residue in a long glycan chain. coworkers94 reported a novel approach coupling mass
For a precise de novo structural characterization of spectrometry with ion spectroscopy for the structural
unknown oligosaccharide using NMR, the size of GAG analysis of GAGs. Distinctive spectroscopic finger­
chains is limited to approximately an octadecasaccha­ prints of the monosaccharide standards GalNAc4S
ride.1 Therefore, the NMR is often used alone in a bot­ and GalNAc6S were found in the 3 μm range. Khanal
tom-up approach or as assistance for top-down MS and coworkers95 used IMS combined with cryogenic,
analysis. messenger-tagging, infrared (IR) spectroscopy, and
One-dimensional 1H-NMR has been used to help MS to identify different isomeric disaccharides of CS
to determine the ratio of IdoA/IdoA2S: GlcA in DS and HS origin. Disaccharide isomers can be uniquely
chain, in support with MS analysis.16 One- and two- distinguished by their vibrational spectrum between
dimensional NMR were used to compare relative –3200 and 3700 cm–1 due to their different hydroxyl
quantities of seven types of monosaccharide between group hydrogen-bonding patterns. Another spectro­
parent and daughter heparins.24 NMR was also used scopic associated research method used a combina­
to characterize structures of pure oligosaccharides tion of gas-phase IRMPD (infrared multiple photon
derived in a bottom-up approach. A series of GAG oli­ dissociation) spectroscopy with MS to address the dif­
gosaccharides up to octasaccharides were separated ferentiation of positional isomers of sulfated carbohy­
from different kinds of sea cucumbers and character­ drates on monosaccharide standards and disaccharides
ized by 1D and 2D (COSY, TOCSY, ROESY, HSQC of HP and CS. Mass spectrometric fingerprints and
and HMBC) NMR.90 A bottom-up approach was used gas-phase vibrational spectra in the near and mid-IR
to analyze the central core of fucosylated CS from two regions were obtained, which validate the potential of
species of sea cucumbers using COSY, TOCSY, and the spectroscopic approach to resolve isomeric disac­
HSQC, and only slight differences were found between charides.96 In a review published by Mohamed and
the two species.91 Guerrini and coworkers92 used coworkers,97 the application of vibrational spectros­
HSQC for the structural characterization of three copy for GAG analysis was introduced. By comparing
commercially available LMWHs of enoxaparin, dalte­ the GAG IR and Raman spectral signatures of the
parin, and tinzaparin. In this research, the minor resi­ media and live cells, Chinese hamster ovary (CHO)
dues generated by a depolymerization procedure, as wild type cell line and its mutant counterpart, CHO-
well as the relevant residues belonging to the parent 745, which lacks xylosyltransferase associated with
heparin, have been characterized and quantified. low GAG synthesis, were discriminated. In recently
HSQC has recently been considered a quantitative published research, surface plasmon resonance imag­
tool for GAG analysis. This can be achieved by proper ing (SPRi) was coupled to an on-chip mass spectrom­
selection of analytical signals among those with sim­ etry. The thermodynamic parameters of the interactions
ilar magnetic relaxation and one bond proton-carbon between cytokines and GAGs were determined using
J-coupling. The method was elaborated by Torri and SPRi monitoring, and the captured carbohydrates
Guerrini88 that can be confidently used to compare were directly examined on the biochip surface using
different GAGs, by quantification of their various sub­ MALDI-TOF MS.98
stituted monosaccharide components.
Perspectives
Hyphenated and Other Techniques The future development of GAG analysis and sequenc­
Although both MS and NMR show their specific advan­ ing will be facilitated by the emerging innovative tech­
tages in characterizing GAG chain, in most cases, the niques, materials, and methods. A “shotgun ion
combined application of both methods provides more mobility mass spectrometry sequencing” (SIMMS2)
informative details and allows a more thorough struc­ method was recently be developed by Miller and
tural characterization. NMR can assist MS in identify­ coworkers.99 Intact HS saccharides are subjected to
ing monosaccharides and quantifying IdoA/GlcA ion mobility mass spectrometry and MS2 analysis.
Analysis of the Glycosaminoglycans 11

Matching of data for intact and fragment ions against Competing Interests
known values for defined HS oligosaccharide struc­ The author(s) declared no potential conflicts of interest with
tures permits unambiguous sequence determination respect to the research, authorship, and/or publication of
of validated standards as well as unknown natural sac­ this article.
charides, including variants with 3-O-sulfo groups. It is
envisioned that by limited digestion with multiple site- Author Contributions
specific enzymes, sets of partially overlapping frag­ YS contributed to the data searching and organizing, and
ments can be separated and sequenced by ion mobility draft the article. FZ assisted in writing and revising the man­
mass spectrometry and MS2. Overlaps of such frag­ uscript. RJL designed the topic and assisted in writing and
ments may enable assembly in a similar manner to the revising the manuscript.
shotgun sequencing approaches used in genomics
and proteomics. A newly emerging technique, based Funding
on nanopore sensing, seems to represent a promising The author(s) received no financial support for the research,
breakthrough. The nanopore can be made of either authorship, and/or publication of this article.
biological (such as aerolysin and α-hemolysin)100 or
inorganic materials.101–103 The nanopore filled with Literature Cited
electrolyte solution allows ions to pass through unless 1. Li L, Ly M, Linhardt RJ. Proteoglycan sequence. Mol
a charged polymer in the passage and disrupts ion Biosyst. 2012;8(6):1613–25.
flow, generating a blocked-pore current. Karawdeniya 2. Köwitsch A, Zhou G, Groth T. Medical application of
and coworkers102 reported this technique allowed easy glycosaminoglycans: a review. J Tissue Eng Regen
differentiation between a clinical heparin sample and Med. 2018;12(1):e23–41.
one contaminated with oversulfated chondroitin sulfate 3. Zhao J, Zhu Y, Song X, Xiao Y, Su G, Liu X, Wang Z,
(OSCS). In other research carried out by Im and Xu Y, Liu J, Eliezer D. 3-O-sulfation of heparan sulfate
coworkers, solid-state nanopore device with a support enhances tau interaction and cellular uptake. Angew
Chem Int Ed Engl. 2020;59(5):1818–27.
vector machine (SVM) learning algorithm was used for
4. Fu L, Suflita M, Linhardt RJ. Bioengineered heparins and
GAG analysis. The nanopore/SVM technique was able heparan sulfates. Adv Drug Deliv Rev. 2016;97:237–49.
to distinguish between monodisperse fragments of HP 5. Yip GW, Smollich M, Götte M. Therapeutic value of gly­
and CS with high accuracy (>90%), allowing the detec­ cosaminoglycans in cancer. Mol Cancer Ther. 2006;5
tion of as little as 0.8% (w/w) CS impurity in a heparin (9):2139–48.
sample.103 6. De Pasquale V, Pavone LM. Heparan sulfate proteo­
The development of new materials will also promote glycans: the sweet side of development turns sour in
progress in GAG-related research. Mothéré and mucopolysaccharidoses. Biochim Biophys Acta Mol
coworkers used molecular imprinting technologies to Basis Dis. 2019;1865:165539.
prepare a library of polyethylene glycol acrylate func­ 7. Kamhi E, Joo EJ, Dordick JS, Linhardt RJ. Glyco-
tionalized hydrogels, which showed certain recognition saminoglycans in infectious disease. Biol Rev Camb
Philos Soc. 2013;88(4):928–43.
specificity and selectivity to sulfated oligosaccharide.104
8. Volpi N. Chondroitin sulfate safety and quality.
Also, GAGs are natural negatively charged biopoly­ Molecules. 2019;24(8):1447.
mers, which suggest they have great potential to be 9. Sugahara K, Mikami T. Chondroitin/dermatan sulfate
engaged in new material development.2,105 in the central nervous system. Curr Opin Struct Biol.
New methods that are fast, high throughput, and 2007;17(5):536–45.
user-friendly are still needed for GAG analysis, such 10. Uchimura K. Keratan sulfate: biosynthesis, structures,
as a fast and sensitive detection of HP based on che­ and biological functions. Methods Mol Biol. 2015;1229:
miluminescence sensing,106 high-throughput microar­ 389–400.
rays that enable in situ structural characterization of 11. Caterson B, Melrose J. Keratan sulfate, a complex
GAGs,98,107 and more easy-to-use structural interpre­ glycosaminoglycan with unique functional capability.
tation software for MS will be important in advancing Glycobiology. 2018;28(4):182–206.
12. Toole B. Hyaluronan and its binding proteins, the
the field. In addition, due to the highly complex and
hyaladherins. Curr Opin Cell Biol. 1990;2(5):839–44.
heterogeneous nature of GAGs, it is quite common 13. Tamer TM. Hyaluronan and synovial joint: function,
that multiple analytical techniques are used to com­ distribution and healing. Interdiscip Toxicol. 2013;6(3):
plete GAG analysis. Therefore, a bioinformatics plat­ 111–25.
form on which integrated information can be stored 14. Yang J, Chi L. Characterization of structural motifs for
and crosschecked will no doubt be meaningful for gly­ interactions between glycosaminoglycans and pro­
comics scientists.25 teins. Carbohydr Res. 2017;452:54–63.
12 Song et al.

15. Ly M, Leach FE III, Laremore TN, Toida T, Amster IJ, 29. Zaia J. On-line separations combined with MS for
Linhardt RJ. The proteoglycan bikunin has a defined analysis of glycosaminoglycans. Mass Spectrom Rev.
sequence. Nat Chem Biol. 2011;7(11):827–33. 2009;28(2):254–72.
16. Yu Y, Duan J, Leach FE III, Toida T, Higashi K, Zhang 30. Yang B, Solakyildirim K, Chang Y, Linhardt RJ. Hyphe­
H, Zhang F, Amster IJ, Linhardt RJ. Sequencing the nated techniques for the analysis of heparin and hepa­
dermatan sulfate chain of decorin. J Am Chem Soc. ran sulfate. Anal Bioanal Chem. 2011;399(2):541–57.
2017;139(46):16986–95. 31. Zaia J. Mass spectrometry and the emerging field of
17. Ly M, Laremore TN, Linhardt RJ. Proteoglycomics: recent glycomics. Chem Biol. 2008;15(9):881–92.
progress and future challenges. OMICS. 2010;14(4): 32. Korir AK, Larive CK. Advances in the separation, sensitive
389–99. detection, and characterization of heparin and heparan
18. Zhao X, Yang B, Solakylidirim K, Joo EJ, Toida T, sulfate. Anal Bioanal Chem. 2009;393(1):155–169.
Higashi K, Linhardt RJ, Li L. Sequence analysis and 33. Fasciano JM, Danielson ND. Ion chromatography
domain motifs in the porcine skin decorin glycosami­ for the separation of heparin and structurally related
noglycan chain. J Biol Chem. 2013;288(13):9226–37. glycoaminoglycans: a review. J Sep Sci. 2016;39(6):
19. Bodet P-E, Salard I, Przybylski C, Gonnet F, Gomila 1118–29.
C, Ausseil J, Daniel R. Efficient recovery of glycos­ 34. Volpi N, Maccari F, Linhardt RJ. Capillary electropho­
aminoglycan oligosaccharides from polyacrylamide resis of complex natural polysaccharides. Electro-
gel electrophoresis combined with mass spectrom­ phoresis. 2008;29(15):3095–106.
etry analysis. Anal Bioanal Chem. 2017;409(5): 35. Sisu E, Flangea C, Serb A, Zamfir AD. Modern develop­
1257–69. ments in mass spectrometry of chondroitin and dermatan
20. Zaia J. Glycosaminoglycan glycomics using mass spec­ sulfate glycosaminoglycans. Amino Acids. 2011;41(2):
trometry. Mol Cell Proteomics. 2013;12(4):885–92. 235–56.
21. Duan J, Amster IJ. Application of FTMS to the analy­ 36. Laremore TN, Leach FE III, Solakyildirim K, Amster
sis of glycosaminoglycans. In: Schmitt-Kopplin P, IJ, Linhardt RJ. Glycosaminoglycan characterization
Kanawati B, editors. Fundamentals and applications by electrospray ionization mass spectrometry includ­
of Fourier transform mass spectrometry. Amsterdam: ing Fourier transform mass spectrometry. Methods
Elsevier; 2019. p. 623–49. Enzymol. 2010;478:79–108.
22. Agyekum I, Zong C, Boons G-J, Amster IJ. Single 37. Ruhaak L, Zauner G, Huhn C, Bruggink C, Deelder A,
stage tandem mass spectrometry assignment of Wuhrer M. Glycan labeling strategies and their use in
the C-5 uronic acid stereochemistry in heparan identification and quantification. Anal Bioanal Chem.
sulfate tetrasaccharides using electron detach­ 2010;397(8):3457–81.
ment dissociation. J Am Soc Mass Spectrom. 38. Antia IU, Yagnik DR, Munoz LP, Shah AJ, Hills FA.
2017;28(9):1741–50. Heparan sulfate disaccharide measurement from biolog­
23. Wei J, Wu J, Tang Y, Ridgeway ME, Park MA, Costello ical samples using pre-column derivatization, UPLC-MS
CE, Zaia J, Lin C. Characterization and quantification and single ion monitoring. Anal Biochem. 2017;530:
of highly sulfated glycosaminoglycan isomers by gated- 17–30.
trapped ion mobility spectrometry negative electron 39. Liu X, Krishnamoorthy D, Lin L, Xue P, Zhang F, Chi
transfer dissociation MS/MS. Anal Chem. 2019;91(4): L, Linhardt R, Iatridis J. A method for characterising
2994–3001. human intervertebral disc glycosaminoglycan disac­
24. Liu X, St Ange K, Wang X, Lin L, Zhang F, Chi L, charides using liquid chromatography-mass spectrom­
Linhardt RJ. Parent heparin and daughter LMW hepa­ etry with multiple reaction monitoring. Eur Cell Mater.
rin correlation analysis using LC-MS and NMR. Anal 2018;35:117–31.
Chim Acta. 2017;961:91–99. 40. Huang R, Zong C, Venot A, Chiu Y, Zhou D, Boons
25. Solakyildirim K. Recent advances in glycosamino­ G-J, Sharp JS. De novo sequencing of complex mix­
glycan analysis by various mass spectrometry tech­ tures of heparan sulfate oligosaccharides. Anal Chem.
niques. Anal Bioanal Chem. 2019;411:3731–41. 2016;88(10):5299–307.
26. Li G, Steppich J, Wang Z, Sun Y, Xue C, Linhardt RJ, Li L. 41. Liang Q, Chopra P, Boons G-J, Sharp JS. Improved
Bottom-up low molecular weight heparin analysis using de novo sequencing of heparin/heparan sulfate oli­
liquid chromatography-Fourier transform mass spec­ gosaccharides by propionylation of sites of sulfation.
trometry for extensive characterization. Anal Chem. Carbohydr Res. 2018;465:16–21.
2014;86(13):6626–32.  42. Mangrum JB, Mehta AY, Alabbas AB, Desai UR,
27. Lin L, Yu Y, Zhang F, Xia K, Zhang X, Linhardt RJ. Hawkridge AM. Comparative analysis of INLIGHT™-
Bottom-up and top-down profiling of pentosan polysul­ labeled enzymatically depolymerized heparin by reverse-
fate. Analyst. 2019;144(16):4781–6. phase chromatography and high-performance mass
28. Volpi N, Linhardt RJ. High-performance liquid spectrometry. Anal Bioanal Chem. 2017;409(2):499–
chromatography-mass spectrometry for mapping and 509.
sequencing glycosaminoglycan-derived oligosaccha­ 43. Przybylski C, Bonnet V, Vivès RR. A microscale dou­
rides. Nat Protoc. 2010;5(6):993–1004. ble labelling of GAG oligosaccharides compatible with
Analysis of the Glycosaminoglycans 13

enzymatic treatment and mass spectrometry. Chem 56. Stavenhagen K, Kolarich D, Wuhrer M. Clinical glycom­
Commun (Camb). 2019;55(29):4182–85. ics employing graphitized carbon liquid chromatog­
44. Shioiri T, Tsuchimoto J, Watanabe H, Sugiura N. raphy–mass spectrometry. Chromatographia. 2015;
Sequence determination of synthesized chondroitin 78(5–6):307–320.
sulfate dodecasaccharides. Glycobiology. 2016;26(6): 57. Miller RL, Guimond SE, Prescott M, Turnbull JE,
592–606. Karlsson N. Versatile separation and analysis of heparan
45. Yang B, Weyers A, Baik JY, Sterner E, Sharfstein S, sulfate oligosaccharides using graphitized carbon liq­
Mousa SA, Zhang F, Dordick JS, Linhardt RJ. Ultra- uid chromatography and electrospray mass spectrom­
performance ion-pairing liquid chromatography with etry. Anal Chem. 2017;89(17):8942–50.
on-line electrospray ion trap mass spectrometry for 58. Zamfir AD. Applications of capillary electrophoresis
heparin disaccharide analysis. Anal Biochem. 2011; electrospray ionization mass spectrometry in glycos­
415(1):59–66. aminoglycan analysis. Electrophoresis. 2016;37(7–8):
46. Liang QT, Xiao XM, Lin JH, Wei Z. A new sequencing 973–86.
approach for N-unsubstituted heparin/heparan sulfate 59. Lin L, Liu X, Zhang F, Chi L, Amster IJ, Leach FE, Xia
oligosaccharides. Glycobiology. 2015;25(7):714–25. Q, Linhardt RJ. Analysis of heparin oligosaccharides
47. Du JY, Chen LR, Liu S, Lin JH, Liang QT, Lyon M, by capillary electrophoresis–negative-ion electrospray
Wei Z. Ion-pairing liquid chromatography with on-line ionization mass spectrometry. Anal Bioanal Chem.
electrospray ion trap mass spectrometry for the struc­ 2017;409(2):411–20.
tural analysis of N-unsubstituted heparin/heparan sul­ 60. Sanderson P, Stickney M, Leach FE III, Xia Q, Yu Y,
fate. J Chromatogr B Analyt Technol Biomed Life Sci. Zhang F, Linhardt RJ, Amster IJ. Heparin/heparan sul­
2016;1028:71–76. fate analysis by covalently modified reverse polarity
48. Persson A, Toledo AG, Vorontsov E, Nasir W, Willén capillary zone electrophoresis-mass spectrometry. J
D, Noborn F, Ellervik U, Mani K, Nilsson J, Larson G. Chromatogr A. 2018;1545:75–83.
LC–MS/MS characterization of xyloside-primed gly­ 61. Stickney M, Sanderson P, Leach FE III, Zhang F,
cosaminoglycans with cytotoxic properties reveals Linhardt RJ, Amster IJ. Online capillary zone elec­
structural diversity and novel glycan modifications. J trophoresis negative electron transfer dissociation
Biol Chem. 2018;293(26):10202–19. tandem mass spectrometry of glycosaminoglycan mix­
49. Sun X, Guo Z, Yu M, Lin C, Sheng A, Wang Z, Linhardt tures. Int J Mass Spectrom. 2019;445:116209.
RJ, Chi L. Hydrophilic interaction chromatography-mul­ 62. Kailemia MJ, Park M, Kaplan DA, Venot A, Boons G-
tiple reaction monitoring mass spectrometry method J, Li L, Linhardt RJ, Amster IJ. High-field asymmet­
for basic building block analysis of low molecular ric-waveform ion mobility spectrometry and electron
weight heparins prepared through nitrous acid depoly­ detachment dissociation of isobaric mixtures of glycos­
merization. J Chromatogr A. 2017;1479:121–8. aminoglycans. J Am Soc Mass Spectrom. 2013;25(2):
50. Sun X, Sheng A, Liu X, Shi F, Jin L, Xie S, Zhang F, 258–68.
Linhardt RJ, Chi L. Comprehensive identification and 63. Zhao Y, Singh A, Li L, Linhardt RJ, Xu Y, Liu J, Woods
quantitation of basic building blocks for low-molecular RJ, Amster IJ. Investigating changes in the gas-phase
weight heparin. Anal Chem. 2016;88(15):7738–44. conformation of Antithrombin III upon binding of
51. Turiák L, Tóth G, Ozohanics O, Révész Ács ÁA, Vékey Arixtra using traveling wave ion mobility spectrometry
K, Zaia J, Drahos L. Sensitive method for glycosami­ (TWIMS). Analyst. 2015;140(20):6980–9.
noglycan analysis of tissue sections. J Chromatogr A. 64. Miller RL, Wei W, Schwörer R, Zubkova OV, Tyler PC,
2018;1544:41–48. Turnbull JE, Leary JA. Composition, sequencing and
52. Zhang T, Liu X, Li H, Wang Z, Chi L, Li J-P, Tan T. ion mobility mass spectrometry of heparan sulfate-like
Characterization of epimerization and composition of octasaccharide isomers differing in glucuronic and idu­
heparin and dalteparin using a UHPLC-ESI-MS/MS ronic acid content. Eur J Mass Spectrom (Chichester).
method. Carbohydr Polym. 2019;203:87–94. 2015;21(3):245–54.
53. Antia IU, Mathew K, Yagnik DR, Hills FA, Shah AJ. 65. Liu H, Joshi A, Chopra P, Liu L, Boons G-J, Sharp
Analysis of procainamide-derivatised heparan sulphate JS. Salt-free fractionation of complex isomeric mix­
disaccharides in biological samples using hydrophilic tures of glycosaminoglycan oligosaccharides compat­
interaction liquid chromatography mass spectrometry. ible with ESI-MS and microarray analysis. Sci Rep.
Anal Bioanal Chem. 2018;410(1):131–43. 2019;9(1):1–13.
54. Staples GO, Bowman MJ, Costello CE, Hitchcock 66. Miller RL, Guimond SE, Shivkumar M, Blocksidge
AM, Lau JM, Leymarie N, Miller C, Naimy H, Shi X, J, Austin JA, Leary JA, Turnbull JE. Heparin iso­
Zaia J. A chip-based amide-HILIC LC/MS platform for meric oligosaccharide separation using volatile salt
glycosaminoglycan glycomics profiling. Proteomics. strong anion exchange chromatography. Anal Chem.
2009;9(3):686–95. 2016;88(23):11542–50.
55. Huang Y, Shi X, Yu X, Leymarie N, Staples GO, Yin H, 67. Zaia J. Mass spectrometry of oligosaccharides. Mass
Killeen K, Zaia J. Improved liquid chromatography-MS/ Spectrom Rev. 2004;23(3):161–227.
MS of heparan sulfate oligosaccharides via chip-based 68. Wolff JJ, Amster IJ, Chi L, Linhardt RJ. Electron
pulsed makeup flow. Anal Chem. 2011;83(21):8222–9. detachment dissociation of glycosaminoglycan
14 Song et al.

tetrasaccharides. J Am Soc Mass Spectrom. 2007; spectrometry (MS/MS) and database search. Mol Cell
18(2):234–44. Proteomics. 2017;16(2):255–64.
69. Wolff JJ, Laremore TN, Aslam H, Linhardt RJ, Amster 83. Hu H, Mao Y, Huang Y, Lin C, Zaia J. Bioinformatics of
IJ. Electron-induced dissociation of glycosamino­ glycosaminoglycans. Perspect Sci. 2017;11:40–44.
glycan tetrasaccharides. J Am Soc Mass Spectrom. 84. Duan J, Jonathan Amster I. An automated, high-
2008;19(10):1449–58. throughput method for interpreting the tandem mass
70. Agyekum I, Patel AB, Zong C, Boons G-J, Amster IJ. spectra of glycosaminoglycans. J Am Soc Mass
Assignment of hexuronic acid stereochemistry in syn­ Spectrom. 2018;29(9):1802–11.
thetic heparan sulfate tetrasaccharides with 2-O-sulfo 85. Hong P, Sun H, Sha L, Pu Y, Khatri K, Yu X, Tang Y,
uronic acids using electron detachment dissociation. Lin C. GlycoDeNovo–an efficient algorithm for accurate
Int J Mass Spectrom. 2015;390:163–69. de novo glycan topology reconstruction from tandem
71. Leach FE III, Riley NM, Westphall MS, Coon JJ, Amster mass spectra. J Am Soc Mass Spectrom. 2017;28(11):
IJ. Negative electron transfer dissociation sequencing 2288–301.
of increasingly sulfated glycosaminoglycan oligosac­ 86. Hogan JD, Klein JA, Wu J, Chopra P, Boons G-J,
charides on an Orbitrap mass spectrometer. J Am Soc Carvalho L, Lin C, Zaia J. Software for peak finding
Mass Spectrom. 2017;28(9):1844–54. and elemental composition assignment for glycosami­
72. Wu J, Wei J, Hogan JD, Chopra P, Joshi A, Lu W, noglycan tandem mass spectra. Mol Cell Proteomics.
Klein J, Boons G-J, Lin C, Zaia J. Negative electron 2018;17(7):1448–56.
transfer dissociation sequencing of 3-O-sulfation- 87. Soares PA, Queiroz IN, Pomin VH. NMR structural biol­
containing heparan sulfate oligosaccharides. J Am ogy of sulfated glycans. J Biomol Struct Dyn. 2017;35(5):
Soc Mass Spectrom. 2018;29(6):1262–72. 1069–84.
73. Devakumar A, Thompson MS, Reilly JP. Fragmentation 88. Torri G, Guerrini M. Quantitative 2D NMR analysis
of oligosaccharide ions with 157 nm vacuum ultravio­ of glycosaminoglycans. In: Wawer I, Diehl B, edi­
let light. Rapid Commun Mass Spectrom. 2005;19(16): tors. NMR spectroscopy in pharmaceutical analysis.
2313–20. Amsterdam: Elsevier; 2008. p. 407–28.
74. Ko BJ, Brodbelt JS. 193 nm ultraviolet photodissocia­ 89. Carnachan SM, Hinkley SF. Heparan sulfate identifi­
tion of deprotonated sialylated oligosaccharides. Anal cation and characterisation: method I. Heparan sulfate
Chem. 2011;83(21):8192–200. identification by NMR analysis. Bio Protoc. 2017;7(7)
75. Domon B, Costello CE. A systematic nomenclature for 1–7. doi:10.21769/BioProtoc.2196.
carbohydrate fragmentations in FAB-MS/MS spectra 90. Shang F, Gao N, Yin R, Lin L, Xiao C, Zhou L, Li
of glycoconjugates. Glycoconj J. 1988;5(4):397–409. Z, Purcell SW, Wu M, Zhao J. Precise structures of
76. Zhang Z, Linhardt RJ. Sequence analysis of native oli­ fucosylated glycosaminoglycan and its oligosaccha­
gosaccharides using negative ESI tandem MS. Curr rides as novel intrinsic factor Xase inhibitors. Eur J
Anal Chem. 2009;5(3):225–37. Med Chem. 2018;148:423–35.
77. Saad OM, Leary JA. Heparin sequencing using enzy­ 91. Santos GR, Porto AC, Soares PA, Vilanova E, Mourão
matic digestion and ESI-MS n with HOST: a heparin/ PA. Exploring the structure of fucosylated chondroitin
HS oligosaccharide sequencing tool. Anal Chem. sulfate through bottom-up nuclear magnetic resonance
2005;77(18):5902–11. and electrospray ionization-high-resolution mass
78. Damerell D, Ceroni A, Maass K, Ranzinger R, Dell A, spectrometry approaches. Glycobiology. 2017;27(7):
Haslam SM. The GlycanBuilder and GlycoWorkbench 625–34.
Glycoinformatics tools: updates and new develop­ 92. Guerrini M, Guglieri S, Naggi A, Sasisekharan R, Torri
ments. Biol Chem. 2012;393(11):1357–62. G. Low molecular weight heparins: structural differentia­
79. Maxwell E, Tan Y, Tan Y, Hu H, Benson G, Aizikov tion by bidimensional nuclear magnetic resonance spec­
K, Conley S, Staples GO, Slysz GW, Smith RD. troscopy. Semin Thromb Hemost; 2007;33(5):478–87.
GlycReSoft: a software package for automated recogni­ 93. Beccati D, Lech M, Ozug J, Gunay NS, Wang J, Sun
tion of glycans from LC/MS data. PLOS ONE. 2012;7(9): EY, Pradines JR, Farutin V, Shriver Z, Kaundinya GV.
e45474. An integrated approach using orthogonal analytical
80. Li L, Zhang F, Zaia J, Linhardt RJ. Top-down techniques to characterize heparan sulfate structure.
approach for the direct characterization of low molec­ Glycoconj J. 2017;34(1):107–17.
ular weight heparins using LC-FT-MS. Anal Chem. 94. Renois-Predelus G, Schindler B, Compagnon I.
2012;84(20):8822–29. Analysis of sulfate patterns in glycosaminoglycan oli­
81. Chiu Y, Huang R, Orlando R, Sharp JS. GAG-ID: gosaccharides by MSn coupled to infrared ion spec­
heparan sulfate (HS) and heparin glycosaminogly­ troscopy: the case of GalNAc4S and GalNAc6S. J Am
can high-throughput identification software. Mol Cell Soc Mass Spectrom. 2018;29(6):1242–49.
Proteomics. 2015;14(6):1720–30. 95. Khanal N, Masellis C, Kamrath MZ, Clemmer DE,
82. Chiu Y, Schliekelman P, Orlando R, Sharp JS. A multi­ Rizzo TR. Glycosaminoglycan analysis by cryogenic
variate mixture model to estimate the accuracy of gly­ messenger-tagging IR spectroscopy combined with
cosaminoglycan identifications made by tandem mass IMS-MS. Anal Chem. 2017;89(14):7601–6.
Analysis of the Glycosaminoglycans 15

96. Schindler B, Barnes L, Gray C, Chambert S, Flitsch 102. Karawdeniya BI, Bandara YND, Nichols JW, Chevalier
S, Oomens J, Daniel R, Allouche A, Compagnon I. RB, Dwyer JR. Surveying silicon nitride nanopores for
IRMPD spectroscopy sheds new (infrared) light on the glycomics and heparin quality assurance. Nat Commun.
sulfate pattern of carbohydrates. J Phys Chem A. 2017; 2018;9(1):1–8.
121(10):2114–20. 103. Im J, Lindsay S, Wang X, Zhang P. Single Molecule
97. Mohamed HT, Untereiner V, Sockalingum GD, Brézillon Identification and Quantification of Glycosaminoglycans
S. Implementation of infrared and Raman modalities Using Solid-State Nanopores. ACS Nano. 2019;13(6):
for glycosaminoglycan characterization in complex sys­ 6308–18.
tems. Glycoconj J. 2017;34(3):309–23. 104. Mothéré M, Singabraya D, Driguez PA, Siñeriz F, Papy
98. Przybylski C, Gonnet F, Saesen E, Lortat-Jacob H, Daniel Garcia D. Poly(ethylene glycol acrylate)-functionalized
R. Surface plasmon resonance imaging coupled to on- hydrogels for heparan sulfate oligosaccharide recogni­
chip mass spectrometry: a new tool to probe protein-GAG tion. J Mol Recognit. 2017;30(3):e2584.
interactions. Anal Bioanal Chem. 2020;412(2):507–19. 105. Kemp MM, Kumar A, Clement D, Ajayan P, Mousa
99. Miller RL, Guimond SE, Schwörer R, Zubkova OV, Tyler PC, S, Linhardt RJ. Hyaluronan-and heparin-reduced sil­
Xu Y, Liu J, Chopra P, Boons G-J, Grabarics M. Shotgun ver nanoparticles with antimicrobial properties. Nano-
ion mobility mass spectrometry sequencing of heparan medicine. 2009;4(4):421–29.
sulfate saccharides. Nat Commun. 2020;11(1):1–12. 106. Qi Y, He J, Xiu F-R, Yu X, Li Y, Lu Y, Gao X, Song
100. Fennouri A, Ramiandrisoa J, Bacri L, Mathé J, Daniel Z, Li B. A facile chemiluminescence sensing for ultra­
R. Comparative biosensing of glycosaminoglycan sensitive detection of heparin using charge effect of
hyaluronic acid oligo-and polysaccharides using aero­ positively-charged AuNPs. Spectrochim Acta A Mol
lysin and alpha hemolysin nanopores. Eur Phys J E Biomol Spectrosc. 2019;216:310–18.
Soft Matter. 2018;41(10):127. 107. Kunzke T, Balluff B, Feuchtinger A, Buck A, Langer R,
101. Rivas F, Zahid OK, Reesink HL, Peal BT, Nixon AJ, Luber B, Lordick F, Zitzelsberger H, Aichler M, Walch
DeAngelis PL, Skardal A, Rahbar E, Hall AR. Label- A. Native glycan fragments detected by MALDI-FT-
free analysis of physiological hyaluronan size distribu­ ICR mass spectrometry imaging impact gastric cancer
tion with a solid-state nanopore sensor. Nat Commun. biology and patient outcome. Oncotarget. 2017;8(40):
2018;9(1):1–9. 68012.

You might also like