recent advances in biotechnology for heparin and …...thrombotic disorders[10].heparin...

15
Contents lists available at ScienceDirect Talanta journal homepage: www.elsevier.com/locate/talanta Recent advances in biotechnology for heparin and heparan sulfate analysis Meng Qiao a,1 , Lei Lin b,1 , Ke Xia c , Jun Li d , Xing Zhang a,∗∗ , Robert J. Linhardt c,e,a School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Wenyuan Road 1, Nanjing, 210023, China b School of Environment, Nanjing Normal University, Wenyuan Road 1, Nanjing, 210023, China c Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, USA d CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301, China e Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, USA ARTICLE INFO Keywords: Heparan sulfate Heparin Structure analysis Ultrasensitive analysis ABSTRACT Heparan sulfate (HS) is a class of linear, sulfated, anionic polysaccharides, called glycosaminoglycans (GAGs), which present on the mammalian cell surfaces and extracellular matrix. HS GAGs display a wide range of critical biological functions, particularly in cell signaling. HS is composed of repeating units of 1 → 4 glucosidically linked uronic acid and glucosamine residues. Heparin, a pharmacologically important version of HS, having higher sulfation and a higher content of iduronic acid than HS, is a widely used clinical anticoagulant. However, due to their heterogeneity and complex structure, HS and heparin are very challenging to analyze, limiting biological studies and even resulting in safety concerns in their therapeutic application. Therefore, reliable methods of structural analysis of HS and heparin are critically needed. In addition to the structural analysis of heparin, its concentration in blood needs to be closely monitored to avoid complications such as thrombocy- topenia or hemorrhage caused by heparin overdose. This review summarizes the progress in biotechnological approaches in the structural characterization of HS and heparin over the past decade and includes the devel- opment of the ultrasensitive approaches for detection and measurement in biological samples. 1. Introduction Heparan sulfate (HS) is a glycosaminoglycan (GAG), a linear, sul- fated, polydisperse, structurally complex hetero-co-polysaccharide [1,2]. HS is comprised of a disaccharide repeating unit consisting of β-D- glucuronic acid (GlcA) or ɑ-L-iduronic acid (IdoA) and β-D-glucosamine (GlcN) linked by (1 → 4) glycoside bonds (Fig. 1). Modifications of disaccharide units, including the presence of N-acetyl of N-sulfo sub- stituents on the GlcN (GlcNAc or GlcNS), O-sulfo groups at the 6-O-and/ or 3-O-positions of GlcN, and 2-O-sulfo substituents on the IdoA posi- tions, making HS diverse structurally (> 32 possible units) [3]. HS is widely distributed on animal cell surfaces and in the extracellular matrix (ECM) [2]. There HS binds to many ECM proteins (i.e., growth factors, chemokines, adhesion proteins, etc.) [4,5], allowing these to act as signaling molecules, playing an important role in cell adhesion, migration, differentiation, signaling and angiogenesis [6,7]. Most im- portantly, HS has physiological and pharmacological impact in blood coagulation, embryonic development, inflammatory response, and bacteria/viral infections [3,8] Heparin is a GAG sharing the same biosynthetic pathway as HS but with a higher level of sulfation and prominent repeating trisulfated disaccharides, GlcNS6S(1 → 4)IdoA2S (Fig. 1)[9]. For more than 100 years, heparin has been clinically used as a drug for the treatment of thrombotic disorders [10]. Heparin binds to antithrombin III (AT), an inhibitor of serine proteases, factor Xa and thrombin, resulting a con- formational change amplifying the inhibitory activity of AT [3,11]. There are three major forms: the intravenous drug, unfractionated he- parin (UFH, MWavg 16,000 Da); several types of subcutaneous low molecular weight heparins (LMWH, MWavg 3500–6000 Da; and sub- cutaneous ultralow molecular weight heparins (ULMWH, MW < 2000 Da) [3,9]. There is an antidote, cationic peptide drug, protamine, which can reverse the effect of UFH. Life-threatening complications, such as heparin-induced thrombocytopenia (HIT) can occur particularly with UFH [12]. For the past 30 years LMWH such as enoxaparin (Figs. 1 and 4), generated from UFH by chemical β-elimination, have been widely used because of their longer half-life and subcutaneous https://doi.org/10.1016/j.talanta.2020.121270 Received 12 April 2020; Received in revised form 29 May 2020; Accepted 30 May 2020 Corresponding author. Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, USA. ∗∗ Corresponding author. E-mail addresses: [email protected] (X. Zhang), [email protected] (R.J. Linhardt). 1 M.Q. and L.L. contributed equally to this work. Talanta 219 (2020) 121270 Available online 14 June 2020 0039-9140/ © 2020 Elsevier B.V. All rights reserved. T

Upload: others

Post on 26-Aug-2020

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Recent advances in biotechnology for heparin and …...thrombotic disorders[10].Heparin bindstoantithrombinIII(AT),an inhibitor of serine proteases, factorXa and thrombin, resulting

Contents lists available at ScienceDirect

Talanta

journal homepage: www.elsevier.com/locate/talanta

Recent advances in biotechnology for heparin and heparan sulfate analysisMeng Qiaoa,1, Lei Linb,1, Ke Xiac, Jun Lid, Xing Zhanga,∗∗, Robert J. Linhardtc,e,∗a School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Wenyuan Road 1, Nanjing, 210023, Chinab School of Environment, Nanjing Normal University, Wenyuan Road 1, Nanjing, 210023, Chinac Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, USAd CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301, Chinae Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, USA

A R T I C L E I N F O

Keywords:Heparan sulfateHeparinStructure analysisUltrasensitive analysis

A B S T R A C T

Heparan sulfate (HS) is a class of linear, sulfated, anionic polysaccharides, called glycosaminoglycans (GAGs),which present on the mammalian cell surfaces and extracellular matrix. HS GAGs display a wide range of criticalbiological functions, particularly in cell signaling. HS is composed of repeating units of 1 → 4 glucosidicallylinked uronic acid and glucosamine residues. Heparin, a pharmacologically important version of HS, havinghigher sulfation and a higher content of iduronic acid than HS, is a widely used clinical anticoagulant. However,due to their heterogeneity and complex structure, HS and heparin are very challenging to analyze, limitingbiological studies and even resulting in safety concerns in their therapeutic application. Therefore, reliablemethods of structural analysis of HS and heparin are critically needed. In addition to the structural analysis ofheparin, its concentration in blood needs to be closely monitored to avoid complications such as thrombocy-topenia or hemorrhage caused by heparin overdose. This review summarizes the progress in biotechnologicalapproaches in the structural characterization of HS and heparin over the past decade and includes the devel-opment of the ultrasensitive approaches for detection and measurement in biological samples.

1. Introduction

Heparan sulfate (HS) is a glycosaminoglycan (GAG), a linear, sul-fated, polydisperse, structurally complex hetero-co-polysaccharide[1,2]. HS is comprised of a disaccharide repeating unit consisting of β-D-glucuronic acid (GlcA) or ɑ-L-iduronic acid (IdoA) and β-D-glucosamine(GlcN) linked by (1 → 4) glycoside bonds (Fig. 1). Modifications ofdisaccharide units, including the presence of N-acetyl of N-sulfo sub-stituents on the GlcN (GlcNAc or GlcNS), O-sulfo groups at the 6-O-and/or 3-O-positions of GlcN, and 2-O-sulfo substituents on the IdoA posi-tions, making HS diverse structurally (> 32 possible units) [3]. HS iswidely distributed on animal cell surfaces and in the extracellularmatrix (ECM) [2]. There HS binds to many ECM proteins (i.e., growthfactors, chemokines, adhesion proteins, etc.) [4,5], allowing these to actas signaling molecules, playing an important role in cell adhesion,migration, differentiation, signaling and angiogenesis [6,7]. Most im-portantly, HS has physiological and pharmacological impact in bloodcoagulation, embryonic development, inflammatory response, and

bacteria/viral infections [3,8]Heparin is a GAG sharing the same biosynthetic pathway as HS but

with a higher level of sulfation and prominent repeating trisulfateddisaccharides, GlcNS6S(1 → 4)IdoA2S (Fig. 1) [9]. For more than 100years, heparin has been clinically used as a drug for the treatment ofthrombotic disorders [10]. Heparin binds to antithrombin III (AT), aninhibitor of serine proteases, factor Xa and thrombin, resulting a con-formational change amplifying the inhibitory activity of AT [3,11].There are three major forms: the intravenous drug, unfractionated he-parin (UFH, MWavg 16,000 Da); several types of subcutaneous lowmolecular weight heparins (LMWH, MWavg 3500–6000 Da; and sub-cutaneous ultralow molecular weight heparins (ULMWH, MW <2000 Da) [3,9]. There is an antidote, cationic peptide drug, protamine,which can reverse the effect of UFH. Life-threatening complications,such as heparin-induced thrombocytopenia (HIT) can occur particularlywith UFH [12]. For the past 30 years LMWH such as enoxaparin (Figs. 1and 4), generated from UFH by chemical β-elimination, have beenwidely used because of their longer half-life and subcutaneous

https://doi.org/10.1016/j.talanta.2020.121270Received 12 April 2020; Received in revised form 29 May 2020; Accepted 30 May 2020

∗ Corresponding author. Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute,Troy, NY, 12180, USA.

∗∗ Corresponding author.E-mail addresses: [email protected] (X. Zhang), [email protected] (R.J. Linhardt).

1 M.Q. and L.L. contributed equally to this work.

Talanta 219 (2020) 121270

Available online 14 June 20200039-9140/ © 2020 Elsevier B.V. All rights reserved.

T

Page 2: Recent advances in biotechnology for heparin and …...thrombotic disorders[10].Heparin bindstoantithrombinIII(AT),an inhibitor of serine proteases, factorXa and thrombin, resulting

bioavailability [13] LMWH binds to AT and acts only on FXa withoutinhibiting thrombin, facilitating a more subtle regulation of coagulationand an improved therapeutic index [14]. A synthetic ULMWH, fonda-parinux, which is the first chemically synthetic antithrombotic penta-saccharide [15], is infrequently used due to its high cost (Figs. 1 and 3).

Recently, biotechnological versions of UFH, LMWH and ULMWHhave reached the stage of commercial development [16–18]. Theseproducts rely on chemoenzymatic synthesis making their preparationmore cost competitive than products like fondaparinux, which relies onchemical synthesis. New analytical approaches that are informationrich, sensitive, robust and reliable are required in the development ofthis new generation of biotechnological heparin and HS products. Thisreview focuses on the analytical methods developed over the pastdecade.

2. Why is analysis of heparin and heparan sulfate important?

The UFH and LMWH currently in clinical use are extracted fromanimal tissues, such as porcine, bovine intestines and bovine lungs [19].As a result, these products have purity and homogeneity issues and avariable based on animal species, age, sex, health, animal feed typesand other environmental factors [20]. More importantly, there is a riskof viral or prion infection, which poses a serious threat to the safety ofthese drugs. A global contamination of heparin broke out in 2007,causing over 200 deaths in the US [21]. This crisis resulted from theadulteration of heparin with a toxic and inexpensive oversulfatedchondroitin sulfate. Therefore, reliable structural analysis techniquesfor animal sourced heparin are highly desirable. Biotechnologicallyprepared heparins and HS offer different but equally challenging ana-lytical objectives. Subtle differences in the structures or process arti-facts require new information rich in highly sensitive analyticalmethods. For instance, the detection of low level process impurities orrelatively subtle structural features, such as sugar epimers or sulfogroup position or differences in linkage position and anomeric

configuration, require more advanced analytical methods. In addition,sensitive, reliable and robust methods to determine heparin levelswithin biological fluids and tissues are needed to avoid complicationssuch as hemorrhage induced by heparin overdose. Thus, this reviewwill summarize the progress of the structure characterization of he-parin/HS in the past decade, and discuss the development of new ul-trasensitive approaches to monitor heparin and HS-based therapeutics.

3. Techniques for structure characterization/quantification ofheparin/HS

Characterization strategies for heparin/HS, particularly LMWHs,can be generally summarized into two approaches, top-down andbottom-up analysis, and these are based on the Food and DrugAdministration (FDA) requirements (Fig. 2) [22]. Similar to proteomicstrategies, top-down analysis analyzes samples without any chemical orenzymatic pretreatment and can rely on simple methods such as sizeexclusion chromatography (SEC) to measure the molecular weight, ormore complicated methods such as liquid chromatography-mass spec-trometry (LC-MS) to obtain fingerprint mapping of intact chains, MS/MS to sequence chains or nuclear magnetic resonance (NMR) to analyzeintact chains [23]. Bottom-up strategies rely on the controlled partialdepolymerization of polysaccharide chains to afford oligosaccharidesfor analysis by high-performance liquid chromatography (HPLC), ca-pillary electrophoresis (CE) and LC-MS disaccharide compositionalanalysis or oligosaccharide sequence analysis [24]. The controlled de-polymerization of a heparin/HS can rely on either random or semi-random chemical methods or somewhat more controlled enzymaticmethods. Heparinases, either lyases [25] or hydrolases [26] provide amild method for the controlled enzymatic depolymerization of heparin/HS. The individual methods applied in both top-down and bottom-upwill be discussed in detail.

Fig. 1. Structures of HS, heparin andheparin derivatives. The major dis-accharide repeating units of HS (pri-marily 1) and heparin (primarily 2) areshown. The structure of an ULMWH,Arixtra® (fondaparinux) 3, containing apentasaccharide sequence corre-sponding to an AT-binding site isshown. The LMWH, Lovenox® (en-oxaparin) 4 contains a diverse mixtureof chain types but only one type ofchain present in enoxaparin, containinga 1,6-anhydro group unique to en-oxaparin, is shown.

M. Qiao, et al. Talanta 219 (2020) 121270

2

Page 3: Recent advances in biotechnology for heparin and …...thrombotic disorders[10].Heparin bindstoantithrombinIII(AT),an inhibitor of serine proteases, factorXa and thrombin, resulting

3.1. Liquid chromatography-MS

Liquid chromatography (LC) is a separation and analysis tool relyingon a liquid as mobile phase and a stationary solid phase. High-perfor-mance liquid chromatography (HPLC) provides faster separation andhigher resolution method than LC and has been widely used in theanalysis of GAG disaccharides and oligosaccharides [27,28]. Usingdifferent mobile and solid phases, based on different separation prin-ciples, HPLC can facilitate the separation and analysis of HS/heparinmixtures and provide diverse structural information. HPLC separationsof HS/heparin include size exclusion chromatography (SEC) stronganion exchange (SAX)-HPLC, reversed-phase (RP)-HPLC, reversed-phase ion-pairing (RPIP)-HPLC, and hydrophilic interaction chromato-graphy (HILIC) [25,29]. Ultraperformance liquid chromatography(UPLC), microflow, nanoflow, capillary HPLC and UPLC has also beenapplied [26]. While LC can be used with a variety of detections such asultraviolet fluorescence and refractive index, it has most recently beenused in conjunction with mass spectrometry (MS), providing high-re-solution spectra in both top-down and bottom-up analysis affording astable and fast continuous analysis platform [22,30].

3.1.1. Top-down analysisTop-down NMR and LC-MS & MS/MS analysis carefully examines

the intact HS or heparin chains and can provide excellent results in theanalysis of the number and types of sulfo groups, and the types ofuronic acid and glucosamine residues present in HS/heparin oligo-saccharides and LMWHs [30]. Top-down analysis LC-MS & MS/MS hasstill not yet been successfully applied to intact chains of UFH because ofits relatively high molecular mass (> 10,000) and net charge (> -40).There are challenges for using MS analysis with a top-down strategy, inparticular, the loss of sulfo groups, and production of multiple adducts.While Fourier transform (FT)-ion cyclotron resonance (ICR) MS offers ahigh-resolution platform, but the availability of such instrumentationhas been problematic. Matrix assisted laser induced ionization (MALDI)MS has limitations in the analysis of highly charged heparin oligo-saccharides of large size [31]. The widespread availability of Orbitrapinstruments has alleviated this limitation to some extent [32]. RPIP-LC,the first LC-MS method for the analysis of LMWH had problems of in-strument contamination with non-volatile ion-pairing reagents, andrelatively low resolution and sensitivity [33]. HILIC is a separation

methods based on the disparity between the overall polarity of poly-saccharides or oligosaccharides, leading to differences in their inter-action with the solid phase support [34]. A relatively high-throughputonline analyical platform, HILIC LC-FT-MS with Orbitrap detection re-sulted in relatively high-resolution, excellent separation efficiency, withgood reproducibility and stability (Fig. 3). The separation relied on aLuna HILIC column, which replaced the standard amine with a cross-linked diol solid-phase support, to improve the resolution of complexLMWHmixtures for MS detection. Bioinformatics software (GlycReSoft)was then used to quickly extract analysis data. This rapid, robustmethod requires no labeling and can be used to compare differentcommercial LMWHs, analyzing ~90% of the chains, up to dp18 in-cluding minor components [35]. Moreover, small amounts of processbyproducts, such as chains with 1,6-anhydro reducing ends could bedetected and use to compare generic versions to the innovator drug,Lovenox®. However, HILIC-LC affords lower resolution than RPIP-HPLC, resulting in challenges for this technology to achieve 100%coverage of chains within LMWH. These challenges might one day beovercome to allow the separation of all of the minor components inLMWH through the use of improved HILIC-LC, and higher resolutionMS instruments [35].

3.1.2. Bottom-up analysisA top-down strategy is not currently applicable to the analysis of full

chain length HS or UFH polysaccharides. Thus, a bottom-up strategy,with high reproducibility and accuracy, is widely used to infer poly-saccharide structure by analyzing disaccharides or oligosaccharidesobtained through controlled depolymerization of the intact poly-saccharides using enzymatic or chemical methods (Table 1) [30].

3.1.2.1. Hydrophilic interaction chromatography-MS. A HILIC-HPLC-electrospray ionization (ESI)-Fourier transform (FT)-MS platform wasestablished for the sensitive analysis of all chemically sulfonated GAG-based contaminants such as the oversulfated chondroitin sulfate thatplayed a role in the 2007 heparin contamination crisis (Fig. 4A) [36].This semi-synthetic GAG could not be enzymatically depolymerized, sothat reactive oxygen species (ROS) were used to prepareoligosaccharides in a highly reproducible and relatively nonselectiveway, while retaining their primary molecular structure. This methodcould successfully detect oversulfated chondroitin sulfate and

Fig. 2. Representative strategies for heparin/HS structural characterization.

M. Qiao, et al. Talanta 219 (2020) 121270

3

Page 4: Recent advances in biotechnology for heparin and …...thrombotic disorders[10].Heparin bindstoantithrombinIII(AT),an inhibitor of serine proteases, factorXa and thrombin, resulting

oversulfated heparan sulfate in the presence of heparin. These, andother potential adulterants, such as N-deacetylated/N-sulfonatedoversulfated chondroitin sulfate, can be detected even at low levels inheparin active pharmaceutical ingredient (API).

In other studies, HILIC-MS was utilized as a high resolution and fastmethod to analyze oligosaccharides prepared from depolymerization ofheparin using heparin lyase II (Fig. 4B). Following heparin lyase IIdepolymerization of enoxaparin it was possible to quantitatively detectfour major 3-O-sulfo group-containing tetrasaccharides (remnants ofthe AT-binding site), oligosaccharides with 1,6-anhydro-amino residues(process artifacts in enoxaparin preparation) and three linkage regionoligosaccharides (where heparin attached to its core protein). Thisanalysis method allows the comparison of the reducing end groups ofdifferent commercial LMWHs that cannot easily be achieved using atop-down strategy. This platform has the advantages of fast separationspeed (about 7 min), wide dynamic range (> 5000) and high sensitivity(detecting 0.01% components), and provides a relatively highthroughput and reliable method to control commercial LMWH products[37].

However, HILIC separation still has major limitations in separatingisomeric compositions and disaccharides with similar overall polarity

having different sulfation patterns. New high-resolution HPLC separa-tion methods compatible with MS detection are critically needed tofully analyze HS/heparin polysaccharides and oligosaccharides [27].

3.1.2.2. RPIP-MS. RPIP involves the interaction between ahydrophobic column (e.g., a C18 column) and analyte ion-paired withlipophilic ion-pairing reagents, such as hexylamine (HXA),tributylamine (TrBA), trimethylamine (TEA), and tetrabutylamine(TBA) [38,39]. A RPIP microflow HPLC-ESI-MS approach has beenused to analyze most HS/heparin disaccharides with reliability andhigh separation efficiency and speed [40]. However, when analyzingbiological samples, this method cannot fully resolve the disaccharidesbecause of the impact of impurities such as salts and proteins in thesamples. UPLC separation, using the 1.7-μm column under highpressure (up to 108 Pa), retains the principle of HPLC [29] butimproves separation speed, resolution, sensitivity and peak capacity(Fig. 5A) [38]. This RPIP-UPLC-ESI-MS approach was used with abottom-up strategy relying on enzymatic depolymerization to analyzesmall amounts of HS/heparin in urine, cells and tissues samples and forthe structural profiling of the pharmaceutical heparins. This approachallows the qualitative and quantitative detection of eight HS/heparin

Fig. 3. Top-down approach for the direct characterization of low molecular weight heparins using HILIC-LC-FT-MS analysis. Copyright 2012 American ChemicalSociety.

Table 1Structures of heparin/HS disaccharide standards are shown where ΔUA corresponds to deoxy-α-L-threo-hex-4-enopyranosyluronic acid.

HS Disaccharides Structure R1 R2 R3 Molecular Weight

0S △UA (1,4)GlcNAc H Ac H 379.11152S △UA2S (1,4)GlcNAc SO3

− Ac H 459.06836S △UA (1,4)GlcNAc6S H Ac SO3

− 459.0683NS △UA (1,4)GlcNS H SO3

− H 417.05772S6S △UA2S (1,4)GlcNAc6S SO3

− Ac SO3− 539.0251

NS2S △UA2S (1,4)GlcNS SO3− SO3

− H 497.0145NS6S △UA (1,4)GlcNS6S H SO3

− SO3− 497.0145

TriS △UA2S (1,4)GlcNS6S SO3− SO3

− SO3− 576.9713

M. Qiao, et al. Talanta 219 (2020) 121270

4

Page 5: Recent advances in biotechnology for heparin and …...thrombotic disorders[10].Heparin bindstoantithrombinIII(AT),an inhibitor of serine proteases, factorXa and thrombin, resulting

disaccharides, with short equilibration time and high sensitivity [38].However, RPIP-HPLC with MS detection still has the disadvantages ofcomplicated preparation, difficult removal of ion-pairing reagent fromthe mobile phase and low compatibility with on-line MS detection[39,41,42].

3.1.2.3. SEC & RPIP-Q/TOF-MSRPIP. Different types of HPLC can beused in combination to achieve better separation results [14]. Forexample, LMWHs contain diverse oligosaccharide chains that cannot beseparated using HILIC, IPRP or SAX. By using multiple heart-cutting(MHC) to connect SEC-HPLC and RPIP-HPLC and detecting withquadrupole time-of-flight (Q/TOF) MS it was possible to provide adefinitive and systematic analysis of LMWH to obtain detailedstructural information (Fig. 4C) [43]. The first dimension, SEC,separated the oligosaccharides by molecular size, while the seconddimension, RPIP, separated the oligosaccharides on the basis ofdifferences in their charge and polarity. This approach facilitated theanalysis of many components having different sizes of similar charge orof the same size but different charge. This relatively high-resolution,stable, easy-to-optimize, high-efficiency, MHC-2D LC-Q/TOF MSapproach could detect 84 oligosaccharides in nadroparin and 122oligosaccharide peaks in enoxaparin. This three-dimensional approachhas the potential to control commercial LMWH production and todetect, analyze and sequence large oligosaccharide chains.

3.1.2.4. SEC & x SAX-MS/MS. SAX-HPLC is a means of separationrelying on the interaction between negatively charged HS/heparin and

the positively charged solid state support of an SAX column [44]. Thismethod affords higher resolution than HILIC and can easily separatedisaccharides with different sulfate levels, especially gratifyingly in theanalysis of disaccharides obtained by the cleavage of heparins andLMWH with lyase [45]. Unfortunately, samples bound to SAX columnstypically need to be eluted with MS-unfriendly high-concentrationsodium salts. Therefore, when analyzing enoxaparin, this method hasoften been used in combination with UV detection after separating thedisaccharide depolymerization using enzymes. This method is highlyaccurate and does not require labeling. However, when envisaging theanalysis of intact LMWH, this method can only be used with enoxaparincontaining double bond as a chromophore, and is not feasible for theLMWH dalteparin sodium due to the lack of chromophore and thehigher molecular weight. However, the higher molecular weight andlack of chromophore in dalteparin sodium limits its application. Anoffline method, which collects, combines, and desalts analytesseparated by HPLC allows their analysis by MS, SAX-HPLC-ESI-MS/MS and is useful for sequencing the short-chain oligosaccharides fromthe LMWH, dalteparin sodium (Fig. 5B and C) [27]. The shortoligosaccharides obtained after preliminary separation by SEC couldbe further separated by SAX-HPLC using a high resolution ProPac PA1SAX column. ESI-MS/MS could then provide a sensitive and reliableapproach for the direct sequencing of low-purity heparin drug samples.This indispensable and sensitive approach was used for the sequencingof 18 oligosaccharides (from tetrasaccharide to octasaccharide)providing a basis for exploring the precise composition of LMWH.

Fig. 4. (A) Oxidative depolymerization and HILIC-MS of heparin and oversulfated semi-synthetic GAGs. Copyright 2014 American Chemical Society. (B) Liquidchromatography-Fourier transform mass spectrometry for extensive characterization of low molecular weight heparin. Copyright 2014 American Chemical Society.(C) Profiling analysis of the LMWHs nadroparin and enoxaparin using MHC-2D-LC-Q time-of-flight (TOF)-MS. Copyright 2015 American Chemical Society.

M. Qiao, et al. Talanta 219 (2020) 121270

5

Page 6: Recent advances in biotechnology for heparin and …...thrombotic disorders[10].Heparin bindstoantithrombinIII(AT),an inhibitor of serine proteases, factorXa and thrombin, resulting

3.2. CE-MS

CE is an effective technique for separating molecules based on theirshape, size, and charge [46,47]. MS can accurately and quickly identifyand analyze molecules by measuring the mass-to-charge ratio of ions[48]. CE-MS is becoming one of the most powerful techniques for theseparation and identification of HS/heparin oligosaccharide due to anumber of advantages including high sensitivity, high resolution,simple operation, high separation efficiency, and a flexible mode ofseparation [49,50]. However, in the process, an effective interface isessential for combining CE and MS. Insufficient research and develop-ment of CE-MS interfaces over the past decade has hindered the ap-plication of CE-MS in heparin/HS analysis. A groundbreaking neotericelectrokinetic pump-based CE-MS interface was used to combine anormal polarity CE separation with a positive-ion electrospray ioniza-tion MS platform. This interface resulted in great sensitivity, resolution

and repeatability allowing the online analysis of heparin disaccharidemixture and LMWH chains (Fig. 6A) [51]. This method creatively in-troduced an uncoated capillary sheath emitter tip as an interface. Thenegatively charged sugar molecules separated by CE were positivelycharged with NH4

+ in ammonium bicarbonate buffer solution and thensprayed into the mass spectrometer for analysis. Reasonable combina-tion of capillary, emitter, MS, rational constitution and ratio betweenbackground electrolyte (BGE) and sheath liquid (SL) reduce the gen-eration of bubbles and ensure stable and steady electrospray at ex-tremely low flow rate (nL/min). This gentle detection environmentgreatly improves the analysis sensitivity and makes the high-throughput detection of HS oligosaccharides possible. Using CE-MSrequires no glycan labeling affording a high signal-to-noise ratio, rapidanalysis (< 10 min), on a small amount of sample (picograms) andutilizes little buffer (< 10 μL).

However, HS is negatively charged and highly sulfated HS chains

Fig. 5. (A) RPIP–UPLC chromatograms of eight heparin/HS-derived disaccharide standards with total ion chromatogram (TIC), UV232 nm, and extracted ionchromatogram (EIC) detection. Copyright 2011 Elsevier. (B) SEC separation of dalteparin; (C) HILIC-ESI–MS of the short dalteparin oligosaccharides collected fromSEC and the short oligosaccharide compositions. Copyright 2018 Elsevier.

M. Qiao, et al. Talanta 219 (2020) 121270

6

Page 7: Recent advances in biotechnology for heparin and …...thrombotic disorders[10].Heparin bindstoantithrombinIII(AT),an inhibitor of serine proteases, factorXa and thrombin, resulting

may undergo desulfation when separated by normal polarity mode CEand analyzing by positive-ion MS. Such decomposition can result re-ducing analysis accuracy. As an alternative approach, using a protein-coated emitter with positively charged as the interface as well as ad-justing the BGE, SL and electrodes allows a reverse polarity CE se-paration and negative-mode electrospray ionization MS (Fig. 6B) [52].This approach is useful in rapid bottom-up and top-down analysis ofLMWH without analyte decomposition. CE-MS has the merits of lowfalse positive results, simple data processing and is capable of detectingand identifying LMWH produced by different manufacturers.

Although a CE-MS interface offers a rapid analytical method and hasbeen commercialized, the sensitivity and reproducibility of this methodneeds further improvement. Online reverse-polarity negative-ion modecapillary isoelectric focusing (cIEF)-MS based on an electrokineticallypumped sheath liquid nanospray CE-MS for the separation and detec-tion of HS disaccharides (Fig. 6C) [53]. Using a high voltage a pHgradient with low pH at the anode and high pH at the cathode is formedwithin the capillary. Therefore, an analyte with a low isoelectric point(pI) will flow first out of the anode side of the capillary and spray intothe mass spectrometer for detection. This method enables separation ofpositional isomers of disaccharides with the same number of O-sulfogroups and also discriminates between N-sulfo groups and O-sulfogroups. Unfortunately, this method has some limitations in separatingGAG analytes with very similar pI values.

By combing reverse-polarity capillary zone electrophoresis withnegative-ion mode mass spectrometry (CZE-MS) an effective and

sensitive tool has been developed for the analysis of GAGs mixtures(Fig. 6D) [49]. Using a cation coated capillary, structurally similarsulfated GAG oligosaccharides and complex mixtures of the same weresuccessfully separated and analyzed with CZE-MS in a fast and re-producible manner. The analysis method was applied to a mixture ofheparin/HS oligosaccharides, varying chain length from trisaccharideto dodecasaccharides resulting in more than 80 molecular compositionsbeing identified by accurate mass measurement.

Since the diameter capillary is very small, it is prone to blockage andthe capillary length has not been optimized or standardized so that thereproducibility of CE-MS analysis still has certain limitations. In addition,as with most liquid phase analyses, standards are required [49].

3.3. Nuclear magnetic resonance

Nuclear magnetic resonance (NMR) spectroscopy is based on theprinciple that an NMR signal changes base on the position of the de-tected nucleus within a molecule. NMR widely used in analyzing themolecular structure of HS/heparin (including conformation) and de-tecting sample content and purity [54–56]. Heparin contaminants andimpurities are difficult to detect as they differ only in sulfo group po-sition, degree of sulfation, and sugar epimers. Although a database ofpure HS/heparin oligosaccharides is currently lacking, NMR is con-sidered an advantageous means of determining the fine structure ofheparin. A library consisting of 66 types of HS/heparin oligosaccharidesof different sizes, sulfation levels and positions were by a

Fig. 6. Schematic diagram of CE−MS system under positive-ion mode (A) (Copyright 2016 American Chemical Society.) and negative-ion mode (B) (Copyright 2016Springer.), respectively. (C) Charge-based separation of acidic oligosaccharides using negative-ion mode capillary isoelectric focusing (cIEF) mass spectrometry.Copyright 2019 American Chemical Society. (D) Diagram depicting the forces of electroosmotic flow and electrophoretic forces that act on analytes during a capillaryzone electrophorasis (CZE)-MS experiment with three different inner capillary coatings, BFS (Bare fused silica), DMS (dichlorodimethylsilane) and AHS (N-(6-aminohexyl) aminomethyltriethoxysilane). Copyright 2018 Elsevier.

M. Qiao, et al. Talanta 219 (2020) 121270

7

Page 8: Recent advances in biotechnology for heparin and …...thrombotic disorders[10].Heparin bindstoantithrombinIII(AT),an inhibitor of serine proteases, factorXa and thrombin, resulting

Fig. 7. (A–C) Example of full NMR characterization of a heparin hexasaccharide prepared through chemoenzymatic synthesis. Copyright 2017 The Royal Society ofChemistry. (D) Heparin samples extracted from different species/organ. Signals 1, 2, and 3, corresponding to H5 and H6 of 6-O-desulfated glucosamine, H2 of N-sulfated glucosamine, and acetyl group of N-acetylated glucosamine, respectively, were used as diagnostic peaks for the determination of their origin. Copyright 2019Frontiers.

M. Qiao, et al. Talanta 219 (2020) 121270

8

Page 9: Recent advances in biotechnology for heparin and …...thrombotic disorders[10].Heparin bindstoantithrombinIII(AT),an inhibitor of serine proteases, factorXa and thrombin, resulting

biotechnological chemoenzymatic synthesis (Fig. 7A–C) [57]. The li-brary contained diverse structures, including rare GlcNS3S6S, Glc2Sand IdoA residues, which are found in low abundance in HS/heparinnatural products but are important for a variety of biological activities.In the NMR analysis of this oligosaccharide library prepared usingbiotechnology, the unique anomeric protons showed broad peaks in the1H NMR spectra and low signal intensity in 2D the spectra. The che-mical shifts anomeric protons and carbons of the various uronic acidand glucosamine residues could be assembled in a table representing avaluable database. Signal strength and chemical shifts of residues suchas, IdoA2S, could be significantly changed by adding ethylenediami-netetraacetic acid (EDTA) to remove trace amounts of divalent cationsgreatly improving the accuracy of NMR detection of HS/heparin oli-gosaccharides. This systematic NMR study paves the way for thecharacterization of more complicated HS/heparin polysaccharides(UFH) helping to safeguard the quality of UFH and ensuring the simi-larity of bioengineered heparin products with the current used animal-derived heparins.

In recent years, 2D NMR has shown unique advantages in the ana-lysis of disaccharides and the quantitative detection of heparin andLMWH [58–60], and is capable of the detection of contaminants, suchas oversulfated chondroitin sulfate (OSCS) in heparin [61,62], as wellas the analysis of danaparoid sodium salt, an anticoagulant drug con-taining HS, DS and CS [63]. Furthermore, 2D NMR can be combinedwith SAX-HPLC to expand the range of detectable structural features,and become a powerful tool for distinguishing sources, monitoringmanufacture and ensuring safety and quality of commercial preparedheparin [45]. In response to the call of FDA, which suggesting in-troducing bovine heparins to address the shortage of heparin drugs, acombination of 1D and 2D-HSQC NMR methods have been used todetect the origin or blending of heparins from different animals andtissues (Fig. 7D) [64]. In addition to testing the purity and safety of HS/heparin oligosaccharide products, NMR can also be used to control thesynthesis of heparin in standard and biotechnological production pro-cesses. 1H NMR was used to quantify heparosan, a precursor of bioen-gineered heparin, in a fermentation supernatant during its productionfrom E. coli K5 and to optimize fermentation parameters and calculatethe yield [65]. However the sensitivity of NMR analysis was in-sufficiently high to directly monitor heparosan, thus, there are stillsome limitations for the detection of GAGs.

3.4. Chemometric analysis

There are inevitably limitations when using a single analyticalmethod such as NMR, HPLC-MS to analyze HS/heparin. Multiple assayscan be used with chemometric analysis to build a more complete ana-lytical method for HS/heparin analysis. 1H NMR analysis of intact he-parin was used to determine structural differences among heparinproducts, and combined HPLC-MS analysis on disaccharides treatedwith heparin lyases to analyze disaccharide concentrations and bottom-up HPLC-MS analysis heparin lyase 2- resistant tetrasaccharides to de-termine differences in the anticoagulant activity on 30 heparin samplesfrom different sources (Fig. 8) [66]. Comprehensive analysis non-in-tersecting clusters obtained by principal component analysis (PCA) andthe K-means, obtained from the visual data mining software, estab-lished a detailed database for meticulous analysis of heparin structure.This database was useful in analyzing the biological source of heparinand could also detect mixtures of these heparins prohibited by US FDAregulations. It is worth noting that the NMR spectrum, which providesan array of data of great significance as a reference, is particularlyimportant in PCA and has important applications in many aspects, suchas the identification of animal and tissue source, the purity and safety ofbiologically sourced heparins [67,68], the structural characterization ofheparin derivatives [69], the identification of contaminants [70], theanalysis of anticoagulant activity [71], and the detection of commercialheparin sources [72].

3.5. SPR techniques

Surface plasmon resonance (SPR), analyzes the interaction betweenputative ligands and receptor molecules (i.e., enzymes, peptides, anti-bodies, and DNA) immobilized on a thin metal layer (i.e., silver, gold, oraluminum films) by detecting refractive index changes of the mediumnear the nanometal film based on optical biosensor technology. SPR is alabel-free, high sensitivity, high repeatability, real time detectionmethod [73,74]. With over three decades of use, SPR is considered to beone of the most efficient techniques for identifying affinity, specificity,and kinetic parameters between macromolecules (i.e., protein-poly-saccharide, receptor-drug, enzyme-substrate, etc.) [75]. SPR can beused to detect the interaction of viruses with GAGs to understand viralpathogenesis and develop treatments. SPR technology for the study ofZika virus (ZIKV), recognized in 2016 by the World Health Organiza-tion (WHO) to cause severe congenital defects, was used to explain therole of GAGs in placenta and brain host cells for ZIKV entry (Fig. 9A)[76]. ZIKV envelope protein (ZIKV E) has a strong affinity for long-chain heparin oligosaccharides (octasaccharide to octadecasaccharides)and heparins with higher sulfate levels. Human placenta chondroitinsulfate (CS) also binds more tightly to ZIKV E than does porcine brainHS, suggesting that placenta CS may be the most likely receptor med-iating ZIKV invasion through the placenta. The porcine brain HS wasenriched in trisulfated disaccharide presumably resulting in tighterbinding ability ZIKV E. The exploration of the interaction of HS and CSwith ZIKV E, using SPR, not only lays a foundation for further under-standing ZIKV pathogenesis, but also provides a new approach studyingthe mechanism of viral infection. SPR is also useful for assessing theanticoagulant activity of heparin. Competitive SPR to test the ability ofsoluable heparin to compete with AT preventing its binding to heparinimmobilized on a chip (Fig. 9B–E) [77]. This competitive bindingmethod correlates well with standard bio assay method, and it reducesthe required steps, shortens analysis time, and reduces assay costs whilemaintaining repeatability. However, the current instrument costs forperforming SPR are extremely high limiting this method in points-of-care (PoC) assay until miniaturized and inexpensive SPR sensors be-come available [78].

3.6. Enzyme-linked immunosorbent assay

The enzyme-linked immunosorbent assay (ELISA) is a highly sen-sitive bioassay method based on antigen-antibody binding that can beused in both qualitative and quantitative determinations [79,80]. ELISAwith enzyme signal amplification has been used as a high-throughputplatform for the detection of 3-OST sulfation of a bioengineered heparinprecursor (a non-anticoagulant heparin missing its 3-O-sulfo groupmade in a biotechnological chemoenzymatic process (Fig. 10A) [81].Biotin-labeled UFH is immobilized on the inner surface of a 96-wellplate coated with streptavidin. After the addition of AT and bioengi-neered heparin precursor for incubation and AT antibody was added, asecondary antibody horseradish peroxidase (HRP)-conjugated IgG, and3,3′,5,5′-tetramethylbenzidine (TMB) substrate were then added forenzymatic signal amplification. The anticoagulant activity of thebioengineered heparin could then be determined by UV measurementto assess the progress of 3-OST catalyzed reaction. After improvingdetection performances such as shortening time and reducing costs bycontinuously optimizing the main factors affecting signal response. Thissensitive and accurate detection method provides a new approach forensuring the consistent biotechnology process for the industrial pro-duction of bioengineered heparin.

3.7. Ultrasensitive analysis of heparin and heparan sulfate

During clinical procedures such as cardiovascular surgery andkidney dialysis, heparin levels need to be closely monitored to avoidcomplications such as hemorrhage induced by heparin overdose and

M. Qiao, et al. Talanta 219 (2020) 121270

9

Page 10: Recent advances in biotechnology for heparin and …...thrombotic disorders[10].Heparin bindstoantithrombinIII(AT),an inhibitor of serine proteases, factorXa and thrombin, resulting

HIT. The therapeutic dosing level of heparin is 2–8 U mL−1

(17–67 mM) during cardiovascular surgery and 0.2–1.2 U mL−1

(1.7–10 mM) in postoperative and long-term care [82]. Thus, simple,real-time continuous measurements of heparin levels in serum duringcardiovascular surgery and the postoperative therapy period, is desir-able. There is also a need for detection methods to monitor heparin ininfusion solutions to avoid human dosing errors, particularly in pedia-tric patients. Traditional clinical methods for heparin detection rely onbioassays such as activated clotting time (ACT) or activated partialthromboplastin time (aPPT) [83]. These methods are often not unreli-able and inaccurate are subject to interference and are difficult toperform in clinical settings. Thus, developing new methods for heparindetection, especially the ultrasensitive determination is desirable, notonly facilitate clinic studies, but also improve our understanding of thebiological roles of heparin involved in the critical biological processesthat it regulates. However, ultrasensitive analysis of heparin is verychallenging due to the low concentrations present in complex biologicalfluids (i.e., plasma or serum) as well as the chemical complexity ofglycan chains present in UFH and LMWH drugs. Heparin has no naturalchromophores or fluorophores, and because of its highly negativecharge often shows poor ionization efficiency in MS analysis. Heparinscannot be detected by amplification methods such as those used todetect nucleic acids, and in contrast to proteins, there are few availablecarbohydrate-specific antibodies.

Polymerase chain reaction (PCR) is a technique capable of ampli-fying DNA fragments exponentially for ultrasensitive detection [84,85].HS/heparin detection cannot be achieved signal amplification used to

detect DNA. A biotin-labeled oligosaccharide conjugated to DNA “barcode” constructed and glyco-qPCR was explored as an ultrasensitiveonline detection platform giving higher sensitivity than MS and fluor-escence detection (Fig. 10B) [86]. Biotinylated CS disaccharide (as amodel system) obtained by reductive amination with a DNA “bar code”,and unreacted DNA completely removed. The CS-DNA conjugate wasthen used as a template for Qpcr. The results showed that the platformcould detect CS oligosaccharides at concentrations< 1 zmol. However,this platform can only analyze to detect the total content of a particularGAG. If complex detection, such as composition, is required, glycol-qPCR requires a micro-separation step. When coupled with CE, glyco-qPCR could detect as few as 500 molecules of CS present in Chinesehamster ovary (CHO) cells. These results suggest the possibility of ul-trasensitive detection and the single-cell analysis of GAGs. This ap-proach can also be used for the sensitive analysis of GAG-protein in-teractions providing a reliable basis for exploring the mechanism ofGAGs actions and in drug development. Although these experimentsonly applied glyco-qPCR to the CS GAG it should be possible to applythis ultrasensitive method to the detection of HS/heparin.

This method is not yet sufficiently mature to apply to the analysis ofmore complex oligosaccharide and polysaccharide analytes. The auto-mation and integration of glyco-qPCR for the ultrasensitive analysis ofheparin/HS oligosaccharides and polysaccharides should one day bepossible. One major challenge is the complete removal of the unreactedDNA as residual DNA can adversely impact on the accuracy and sen-sitivity of the analysis.

Fluorescence resonance energy transfer (FRET), a technique that

Fig. 8. Statistical analysis of bovine, ovine, and porcine UFHs based on 1H NMR information (A,B) and LC-MS disaccharide analysis (C,D). PCA score plot of two firstprinciple components and K-means clustering analysis of the NMR data and disaccharide data are presented. Copyright 2019 Elsevier.

M. Qiao, et al. Talanta 219 (2020) 121270

10

Page 11: Recent advances in biotechnology for heparin and …...thrombotic disorders[10].Heparin bindstoantithrombinIII(AT),an inhibitor of serine proteases, factorXa and thrombin, resulting

utilizes a phenomenon of fluorescence energy transfer that occurs at asuitable distance between the donor and acceptor fluorophores[87–90], has been applied to the ultrasensitive analysis of HS/heparin[91,92]. An approach for the detection of a chondroitin sulfate dis-accharide as a model GAG oligosaccharide was developed based onFRET using a CdSe–ZnS core-shell nanocrystal quantum dot (QD) donorand a Cy5 acceptor [93]. This method allows the ultrasensitive detec-tion of GAGs disaccharides with a sensitivity more than 1000 times

higher than CE-laser-induced fluorescence (LIF) (Fig. 11A-C). In addi-tion, there are currently studies using fluorescent probes to detect he-parin in plasma. Warttinger et al. using Heparin Red, a commercialfluorescent probe that can oligomerize polyanionic polysaccharides toquench fluorescence, can result in the quantitative detection of non-anticoagulant heparin in human plasma, such as N-desulfated-N-acetylated heparin and LMWH derivative, tafoxiparin (Fig. 11D) [94].

Fig. 9. (A) Sensorgrams of interactions between ZIKV E protein and UFH. Copyright 2017 American Chemical Society. (B) Diagram of SPR solution competitionexperiment for anticoagulant activity measurement of heparin samples; (C) to (E): SPR sensorgrams of AT binding to heparin surface competing with differentheparin samples, where (C): USP UFH standard; (D): LMWH (E): bovine lung derived UFH. Copyright 2017 Elsevier.

M. Qiao, et al. Talanta 219 (2020) 121270

11

Page 12: Recent advances in biotechnology for heparin and …...thrombotic disorders[10].Heparin bindstoantithrombinIII(AT),an inhibitor of serine proteases, factorXa and thrombin, resulting

4. Conclusions and perspectives

HS and heparins are important GAGs that are responsible for a rangeof physiological and pharmacological functions. However, due to thehigh heterogeneity and complex structures, their analysis is very chal-lenging, limiting their biotechnological synthesis, biological studies,and preventing the adequate addressing of safety concerns. After theheparin contamination crisis of 2007, the US FDA has promoted thediversification of heparin sources such as the reintroduction of bovineheparin or the introduction of bioengineered heparin into the USmarket. These urgings have stimulated the study of heparin analysisresulting in the steady improvement in this research field. MS is still themost widely used analytical tool particularly when coupled to separa-tion methods, providing high sensitivity, high resolution, and highspecificity. In particular, the development of MS and MS/MS in recentyears has provided more comprehensive information on the finestructures and domain structures of HS/heparin and has greatly im-proved detection sensitivity. In addition, combined with stable isotopelabeling, LC-MS/MS represents one of the most promising methods toinvestigate the roles of heparin/HS in metabolic pathways to distin-guish exogenously administered stable isotope labeled heparin/HS fromtheir endogenously biosynthesized counterparts [95]. CE-MS has

recently gained popularity with the introduction of new CE-MS inter-faces due to its short separation time, high efficiency, and the smallamounts of sample required. NMR is able to analysis disaccharide,characterize heparin and LMWH and distinguish contaminants presentin UFH or LMWH drugs. Despite its relatively low sensitivity, NMRaffords high resolution capable of assessing glycosidic bonds, uronicacid epimers and sulfo group substitution. Chemometric analysis can beused to combine a variety of analytical methods and apply statistics andmachine learning to provide more comprehensive structural informa-tion. SPR uniquely and rapidly analyzes HS/heparin interactions withproteins in real time. The various analytical techniques introduced inthis review can be applied to specific analytical problems or coupled toone another to facilitate the heparin/HS analysis. Reliable ultra-sensitive analysis techniques for heparin are also needed clinically toavoid complications such as hemorrhage and HIT.

In the future, the analysis of HS/heparin will continue to developresulting in even higher sensitivity, higher resolution, shorter analysistimes, lower cost, more user-friendliness, higher throughput and im-proved automation. The development of analytical technology shouldpromote a better understanding of the functions, pharmacodynamiccharacteristics, and pharmacokinetics of HS/heparin, the analysis ofcommercial drugs, and the characterization of biotechnological

Fig. 10. (A) Schematic representation of high-throughput method for in-process monitoring 3-OST sulfation in bioengineered heparin synthesis. Copyright 2019Elsevier. (B) Schematic representation of glyco-qPCR. Target carbohydrates (GAGs) from biological samples can be conjugated with different DNA “bar code”,followed by removing unreacted DNA and detecting the corresponding GAG–DNA conjugates with amplified signals (qPCR).

M. Qiao, et al. Talanta 219 (2020) 121270

12

Page 13: Recent advances in biotechnology for heparin and …...thrombotic disorders[10].Heparin bindstoantithrombinIII(AT),an inhibitor of serine proteases, factorXa and thrombin, resulting

products such as bioengineered heparins and chemoenzymaticallysynthesized HS/heparin oligosaccharides.

Disclosure statement

The authors report no declarations of interest. The authors weresupported in part by grants from Open Project Program of the KeyLaboratory of Tropical Marine Bio-resources and Ecology, ChineseAcademy of Sciences (LMB20201006 to XZ), Natural Science ResearchProject of Jiangsu Higher Education Institutions (19KJB150012 to LL)and the National Institutes of Health (CA231074 to RJL).

Declaration of competing interest

The authors declare that they have no known competing financialinterests or personal relationships that could have appeared to influ-ence the work reported in this paper.

References

[1] X. Zhang, L. Lin, H. Huang, R.J. Linhardt, Chemoenzymatic synthesis of glycosa-minoglycans, Acc. Chem. Res. 53 (2020) 335–346.

[2] J.P. Li, M. Kusche-Gullberg, Heparan sulfate: biosynthesis, structure, and function,in: K.W. Jeon (Ed.), International Review of Cell and Molecular Biology, ElsevierAcademic Press Inc, San Diego, 2016, pp. 215–273.

[3] L. Fu, M. Suflita, R.J. Linhardt, Bioengineered heparins and heparan sulfates, Adv.Drug Deliv. Rev. 97 (2016) 237–249.

[4] I. Capila, R.J. Linhardt, Heparin - protein interactions, Angew. Chem. Int. Ed. 41(2002) 391–412.

[5] D. Xu, J.D. Esko, Demystifying heparan sulfate-protein interactions, in:R.D. Kornberg (Ed.), Annu. Rev. Biochem., Annual Reviews, Palo Alto, 2014, pp.129–157.

[6] X.M.R. van Wijk, T.H. van Kuppevelt, Heparan sulfate in angiogenesis: a target fortherapy, Angiogenesis 17 (2014) 443–462.

[7] X. Zhang, R.J. Linhardt, Chemoenzymatic synthesis of low-molecular-weight he-parin and heparan sulfate, in: Z. Tan, L.X. Wang (Eds.), Chemical Biology ofGlycoproteins, The Royal Society of Chemistry Cambridge, 2017, pp. 233–252.

[8] E. Kamhi, E.J. Joo, J.S. Dordick, R.J. Linhardt, Glycosaminoglycans in infectiousdisease, Biol. Rev. 88 (2013) 928–943.

[9] J. Liu, R.J. Linhardt, Chemoenzymatic synthesis of heparan sulfate and heparin,

Fig. 11. Scheme of the FRET system construction in this study. (A) Conjugation scheme of the FRET system. (B) The FRET donor, QD, is excited with a laser at488 nm; because Cy5 dye, located on the same disaccharide, is close, FRET occurs and Cy5 is excited by emission from the QD, and the emission of Cy5 is thendetected. (C) CE-LIF instrumental setup for FRET detection. A 488 nm (filter I, donor-acceptor channel) and 650 nm (filter II, acceptor channel) band-pass filter wasused for FRET detection. Copyright 2013 American Chemical Society. (D) Schematic representation of fluorescence quenching of the molecular probe Heparin Red inthe presence of polyanionic polysaccharides. Copyright 2016 Springer. (For interpretation of the references to colour in this figure legend, the reader is referred to theWeb version of this article.)

M. Qiao, et al. Talanta 219 (2020) 121270

13

Page 14: Recent advances in biotechnology for heparin and …...thrombotic disorders[10].Heparin bindstoantithrombinIII(AT),an inhibitor of serine proteases, factorXa and thrombin, resulting

Nat. Prod. Rep. 31 (2014) 1676–1685.[10] H.C. Hemker, A century of heparin: past, present and future, J. Thromb.

Haemostasis 14 (2016) 2329–2338.[11] G. Spadarella, A. Di Minno, M.B. Donati, M. Mormile, I. Ventre, G. Di Minno, From

unfractionated heparin to pentasaccharide: paradigm of rigorous science growing inthe understanding of the in vivo thrombin generation, Blood Rev. 39 (2020) 14.

[12] G.M. Lee, G.M. Arepally, Heparin-induced thrombocytopenia, Hematol.-AM. Soc.Hemat. (2013) 668–674.

[13] J.L. Babin, K.L. Traylor, D.M. Witt, Laboratory monitoring of low-molecular-weightheparin and fondaparinux, Semin. Thromb. Hemost. 43 (2017) 261–269.

[14] B. Mulloy, J. Hogwood, E. Gray, R. Lever, C.P. Page, Pharmacology of heparin andrelated drugs, Pharmacol. Rev. 68 (2016) 76–141.

[15] M. Petitou, C.A.A. van Boeckel, A synthetic antithrombin iii binding penta-saccharide is now a drug! what comes next? Angew. Chem. Int. Ed. 43 (2004)3118–3133.

[16] B.F. Cress, U. Bhaskar, D. Vaidyanathan, A. Williams, C. Cai, X.Y. Liu, L. Fu, V. M-Chari, F.M. Zhang, S.A. Mousa, J.S. Dordick, M.A.G. Koffas, R.J. Linhardt, Heavyheparin: a stable isotope-enriched, chemoenzymatically-synthesized, poly-compo-nent drug, Angew. Chem. Int. Ed. 58 (2019) 5962–5966.

[17] Y.M. Xu, K. Chandarajoti, X. Zhang, V. Pagadala, W.F. Dou, D.M. Hoppensteadt,E.M. Sparkenbaugh, B. Cooley, S. Daily, N.S. Key, D. Severynse-Stevens, J. Fareed,R.J. Linhardt, R. Pawlinski, J. Liu, Synthetic oligosaccharides can replace animal-sourced low-molecular weight heparins, Sci. Transl. Med. 9 (2017) 10.

[18] Y.M. Xu, S. Masuko, M. Takieddin, H.M. Xu, R.P. Liu, J. Jing, S.A. Mousa,R.J. Linhardt, J. Liu, Chemoenzymatic synthesis of homogeneous ultralow mole-cular weight heparins, Science 334 (2011) 498–501.

[19] D. Keire, Manufacturing heparin with equivalent chemical composition from dif-ferent animal sources, Thromb. Haemostasis 119 (2019) 688-688.

[20] Y.L. Yu, Y. Chen, P. Mikael, F.M. Zhang, A.M. Stalcup, R. German, F. Gould,J. Ohlemacher, H. Zhang, R.J. Linhardt, Surprising absence of heparin in the in-testinal mucosa of baby pigs, Glycobiology 27 (2017) 57–63.

[21] A.Y. Szajek, E. Chess, K. Johansen, G. Gratzl, E. Gray, D. Keire, R.J. Linhardt, J. Liu,T. Morris, B. Mulloy, M. Nasr, Z. Shriver, P. Torralba, C. Viskov, R. Williams,J. Woodcock, W. Workman, A. Al-Hakim, The us regulatory and pharmacopeiaresponse to the global heparin contamination crisis, Nat. Biotechnol. 34 (2016)625–630.

[22] X.Y. Liu, K. St Ange, L. Lin, F.M. Zhang, L.L. Chi, R.J. Linhardt, Top-down andbottom-up analysis of commercial enoxaparins, J. Chromatogr., A 1480 (2017)32–40.

[23] S. Beni, J.F. Limtiaco, C.K. Larive, Analysis and characterization of heparin im-purities, Anal. Bioanal. Chem. 399 (2011) 527–539.

[24] N. Volpi, F. Maccari, J. Suwan, R.J. Linhardt, Electrophoresis for the analysis ofheparin purity and quality, Electrophoresis 33 (2012) 1531–1537.

[25] C.K.M. Tripathi, J. Banga, V. Mishra, Microbial heparin/heparan sulphate lyases:potential and applications, Appl. Microbiol. Biotechnol. 94 (2012) 307–321.

[26] E.A. Yates, T.R. Rudd, Recent innovations in the structural analysis of heparin, Int.J. Cardiol. 212 (2016) S5–S9.

[27] Z.J. Wang, T.J. Zhang, S.S. Xie, X.Y. Liu, H.M. Li, R.J. Linhardt, L.L. Chi, Sequencingthe oligosaccharide pool in the low molecular weight heparin dalteparin with off-line hplc and esi-ms/ms, Carbohydr. Polym. 183 (2018) 81–90.

[28] F.M. Zhang, B. Yang, M. Ly, K. Solakyildirim, Z.P. Xiao, Z.Y. Wang, J.M. Beaudet,A.Y. Torelli, J.S. Dordick, R.J. Linhardt, Structural characterization of heparinsfrom different commercial sources, Anal. Bioanal. Chem. 401 (2011) 2793–2803.

[29] B. Yang, K. Solakyildirim, Y.Q. Chang, R.J. Linhardt, Hyphenated techniques for theanalysis of heparin and heparan sulfate, Anal. Bioanal. Chem. 399 (2011) 541–557.

[30] X.J. Sun, A.R. Sheng, X.Y. Liu, F. Shi, L. Jin, S.S. Xie, F.M. Zhang, R.J. Linhardt,L.L. Chi, Comprehensive identification and quantitation of basic building blocks forlow-molecular weight heparin, Anal. Chem. 88 (2016) 7738–7744.

[31] T.N. Laremore, S. Murugesan, T.J. Park, F.Y. Avci, D.V. Zagorevski, R.J. Linhardt,Matrix-assisted laser desorption/ionization mass spectrometric analysis of un-complexed highly sulfated oligosaccharides using ionic liquid matrices, Anal. Chem.78 (2006) 1774–1779.

[32] S. Eliuk, A. Makarov, Evolution of orbitrap mass spectrometry instrumentation, in:R.G. Cooks, J.E. Pemberton (Eds.), Annual Review of Analytical Chemistry, AnnualReviews, Palo Alto, 2015, pp. 61–80.

[33] C. Thanawiroon, K.G. Rice, T. Toida, R.J. Linhardt, Liquid chromatography/massspectrometry sequencing approach for highly sulfated heparin-derived oligo-saccharides, J. Biol. Chem. 279 (2004) 2608–2615.

[34] A.M. Hitchcock, K.E. Yates, C.E. Costello, J. Zaia, Comparative glycomics of con-nective tissue glycosaminoglycans, Proteomics 8 (2008) 1384–1397.

[35] L. Li, F. Zhang, J. Zaia, R.J. Linhardt, Top-down approach for the direct char-acterization of low molecular weight heparins using lc-ft-ms, Anal. Chem. 84 (2012)8822–8829.

[36] G.Y. Li, C. Cai, L.Y. Li, L. Fu, Y.Q. Chang, F.M. Zhang, T. Toida, C.H. Xue,R.J. Linhardt, Method to detect contaminants in heparin using radical depolymer-ization and liquid chromatography-mass spectrometry, Anal. Chem. 86 (2014)326–330.

[37] G.Y. Li, J. Steppich, Z.Y. Wang, Y. Sun, C.H. Xue, R.J. Linhardt, L.Y. Li, Bottom-uplow molecular weight heparin analysis using liquid chromatography-fourier trans-form mass spectrometry for extensive characterization, Anal. Chem. 86 (2014)6626–6632.

[38] B. Yang, A. Weyers, J.Y. Baik, E. Sterner, S. Sharfstein, S.A. Mousa, F.M. Zhang,J.S. Dordick, R.J. Linhardt, Ultra-performance ion-pairing liquid chromatographywith on-line electrospray ion trap mass spectrometry for heparin disaccharideanalysis, Anal. Biochem. 415 (2011) 59–66.

[39] C.E. Doneanu, W.B. Chen, J.C. Gebler, Analysis of oligosaccharides derived from

heparin by ion-pair reversed-phase chromatography/mass spectrometry, Anal.Chem. 81 (2009) 3485–3499.

[40] Z.Q. Zhang, J. Xie, H.Y. Liu, J. Liu, R.J. Linhardt, Quantification of heparan sulfatedisaccharides using ion-pairing reversed-phase microflow high-performance liquidchromatography with electrospray ionization trap mass spectrometry, Anal. Chem.81 (2009) 4349–4355.

[41] G.M. Campo, S. Campo, A.M. Ferlazzo, R. Vinci, A. Calatroni, Improved high-per-formance liquid chromatographic method to estimate aminosugars and its appli-cation to glycosaminoglycan determination in plasma and serum, J. Chromatogr. B:Analyt. Technol. Biomed. Life Sci. 765 (2001) 151–160.

[42] C. Thanawiroon, K.G. Rice, T. Toida, R.J. Linhardt, Liquid chromatography/massspectrometry sequencing approach for highly sulfated heparin-derived oligo-saccharides, J. Biol. Chem. 279 (2004) 2608–2615.

[43] Y.L. Ouyang, Y.Y. Zeng, Y.X. Rong, Y. Song, L. Shi, B. Chen, X.L. Yang, N.Y. Xu,R.J. Linhardt, Z.Q. Zhang, Profiling analysis of low molecular weight heparins bymultiple heart-cutting two dimensional chromatography with quadruple time-of-flight mass spectrometry, Anal. Chem. 87 (2015) 8957–8963.

[44] L. Yi, Q.H. Zhang, Y. Meng, J. Hao, B.Y. Xie, H. Gan, D.X. Li, K. Dong, Z.Q. Zhang,Qualitative and quantitative analysis of 2, 5-anhydro-d-mannitol in low molecularweight heparins with high performance anion exchange chromatography hyphe-nated quadrupole time of flight mass spectrometry, J. Chromatogr., A 1569 (2018)160–167.

[45] F. Spelta, L. Liverani, A. Peluso, M. Marinozzi, E. Ursa, M. Guerrini, A. Naggi, Sax-hplc and hsqc nmr spectroscopy: orthogonal methods for characterizing heparinbatches composition, Front. Med. 6 (2019) 11.

[46] M. Stickney, P. Sanderson, F.E. Leach, F. Zhang, R.J. Linhardt, I.J. Amster, Onlinecapillary zone electrophoresis negative electron transfer dissociation tandem massspectrometry of glycosaminoglycan mixtures, Int. J. Mass Spectrom. 445 (2019)116209.

[47] E. Ucakturk, C. Cai, L. Li, G. Li, F. Zhang, R.J. Linhardt, Capillary electrophoresis fortotal glycosaminoglycan analysis, Anal. Bioanal. Chem. 406 (2014) 4617–4626.

[48] K. Solakyildirim, Recent advances in glycosaminoglycan analysis by various massspectrometry techniques, Anal. Bioanal. Chem. 411 (2019) 3731–3741.

[49] P. Sanderson, M. Stickney, F.E. Leach, Q.W. Xia, Y.L. Yu, F.M. Zhang, R. Linhardt,I.J. Amster, Heparin/heparan sulfate analysis by covalently modified reverse po-larity capillary zone electrophoresis-mass spectrometry, J. Chromatogr., A 1545(2018) 75–83.

[50] B. Yang, K. Solakyildirim, Y. Chang, R.J. Linhardt, Hyphenated techniques for theanalysis of heparin and heparan sulfate, Anal. Bioanal. Chem. 399 (2011) 541–557.

[51] X. Sun, L. Lin, X. Liu, F. Zhang, L. Chi, Q. Xia, R.J. Linhardt, Capillary electro-phoresis-mass spectrometry for the analysis of heparin oligosaccharides and lowmolecular weight heparin, Anal. Chem. 88 (2016) 1937–1943.

[52] L. Lin, X. Liu, F. Zhang, L. Chi, I.J. Amster, F.E. Leach, Q. Xia, R.J. Linhardt, Analysisof heparin oligosaccharides by capillary electrophoresis–negative-ion electrosprayionization mass spectrometry, Anal. Bioanal. Chem. 409 (2016) 411–420.

[53] Y.L. Ouyang, X.R. Han, Q.W. Xia, J.L. Chen, S. Velagapudi, K. Xia, Z.Q. Zhang,R.J. Linhardt, Negative-ion mode capillary isoelectric focusing mass spectrometryfor charge-based separation of acidic oligosaccharides, Anal. Chem. 91 (2019)846–853.

[54] M. Hricovini, M. Guerrini, A. Bisio, G. Torri, M. Petitou, B. Casu, Conformation ofheparin pentasaccharide bound to antithrombin iii, Biochem. J. 359 (2001)265–272.

[55] M. Guerrini, A. Naggi, S. Guglieri, R. Santarsiero, G. Torri, Complex glycosami-noglycans: profiling substitution patterns by two-dimensional nuclear magneticresonance spectroscopy, Anal. Biochem. 337 (2005) 35–47.

[56] L. Mauri, G. Boccardi, G. Torri, M. Karfunkle, E. Macchi, L. Muzi, D. Keire,M. Guerrini, Qualification of hsqc methods for quantitative composition of heparinand low molecular weight heparins, J. Pharmaceut. Biomed. 136 (2017) 92–105.

[57] X. Zhang, V. Pagadala, H.M. Jester, A.M. Lim, T.Q. Pham, A.M.P. Goulas, J. Liu,R.J. Linhardt, Chemoenzymatic synthesis of heparan sulfate and heparin oligo-saccharides and nmr analysis: paving the way to a diverse library for glycobiolo-gists, Chem. Sci. 8 (2017) 7932–7940.

[58] L. Mauri, G. Boccardi, G. Torri, M. Karfunkle, E. Macchi, L. Muzi, D. Keire,M. Guerrini, Qualification of HSQC methods for quantitative composition of he-parin and low molecular weight heparins, J. Pharmaceut. Biomed. Anal. 136 (2017)92–105.

[59] D.J. Langeslay, C.N. Beecher, A. Naggi, M. Guerrini, G. Torri, C.K. Larive,Characterizing the microstructure of heparin and heparan sulfate using N-sulfo-glucosamine H-1 and H-5 NMR chemical shift analysis, Anal. Chem. 85 (2013)1247–1255.

[60] M. Guerrini, T.R. Rudd, L. Mauri, E. Macchi, J. Fareed, E.A. Yates, A. Naggi,G. Torri, Differentiation of generic enoxaparins marketed in the United States byemploying nmr and multivariate analysis, Anal. Chem. 87 (2015) 8275–8283.

[61] M. Guerrini, Z.Q. Zhang, Z. Shriver, A. Naggi, S. Masuko, R. Langer, B. Casu,R.J. Linhardt, G. Torri, R. Sasisekharan, Orthogonal analytical approaches to detectpotential contaminants in heparin, Proc. Natl. Acad. Sci. U.S.A. 106 (2009)16956–16961.

[62] G.L. Sassaki, D.S. Riter, A.P. Santana, M. Guerrini, M.A. Lima, C. Cosentino,L.M. Souza, T.R. Cipriani, T.R. Rudd, H.B. Nader, E.A. Yates, P.A.J. Gorin, G. Torri,M. Iacomini, A robust method to quantify low molecular weight contaminants inheparin: detection of tris(2-n-butoxyethyl) phosphate, Analyst 136 (2011)2330–2338.

[63] C. Gardini, E. Urso, M. Guerrini, R. van Herpen, P. de Wit, A. Naggi,Characterization of danaparoid complex extractive drug by an orthogonal analyticalapproach, Molecules 22 (2017) 25.

[64] L. Mauri, M. Marinozzi, N. Phatak, M. Karfunkle, K. St Ange, M. Guerrini,

M. Qiao, et al. Talanta 219 (2020) 121270

14

Page 15: Recent advances in biotechnology for heparin and …...thrombotic disorders[10].Heparin bindstoantithrombinIII(AT),an inhibitor of serine proteases, factorXa and thrombin, resulting

D.A. Keire, R.J. Linhardt, 1D and 2D-HSQC NMR: two methods to distinguish andcharacterize heparin from different animal and tissue sources, Front. Med.-Lausanne. 6 (2019) 9.

[65] Z. Wang, Z. Zhang, S.A. McCallum, R.J. Linhardt, Nuclear magnetic resonancequantification for monitoring heparosan k5 capsular polysaccharide production,Anal. Biochem. 398 (2010) 275–277.

[66] Y.L. Ouyang, X.R. Han, Y.L. Yu, J.L. Chen, L. Fu, F.M. Zhang, R.J. Linhardt,J. Fareed, D. Hoppensteadt, W. Jeske, A. Kouta, Z.Q. Zhang, K. Xia, Chemometricanalysis of porcine, bovine and ovine heparins, J. Pharmaceut. Biomed. 164 (2019)345–352.

[67] M.A. Lima, T.R. Rudd, E.H.C. de Farias, L.F. Ebner, T.F. Gesteira, L.M. de Souza,A. Mendes, C.R. Cordula, J.R.M. Martins, D. Hoppensteadt, J. Fareed, G.L. Sassaki,E.A. Yates, I.L.S. Tersariol, H.B. Nader, A new approach for heparin standardization:combination of scanning UV spectroscopy, nuclear magnetic resonance and prin-cipal component analysis, PloS One 6 (2011) 9.

[68] L. Mauri, M. Marinozzi, G. Mazzini, R.E. Kolinski, M. Karfunkle, D.A. Keire,M. Guerrini, Combining nmr spectroscopy and chemometrics to monitor structuralfeatures of crude heparin, Molecules 22 (2017) 14.

[69] T.R. Rudd, M.A. Skidmore, S.E. Guimond, C. Cosentino, G. Torri, D.G. Fernig,R.M. Lauder, M. Guerrini, E.A. Yates, Glycosaminoglycan origin and structure re-vealed by multivariate analysis of NMR and CD spectra, Glycobiology 19 (2009)52–67.

[70] T.R. Rudd, D. Gaudesi, M.A. Skidmore, M. Ferro, M. Guerrini, B. Mulloy, G. Torri,E.A. Yates, Construction and use of a library of bona fide heparins employing H-1NMR and multivariate analysis, Analyst 136 (2011) 1380–1389.

[71] Y.B. Monakhova, J. Fareed, Y.M. Yao, B.W.K. Diehl, Anticoagulant activity ofporcine heparin: structural-property relationship and semi-quantitative estimationby nuclear magnetic resonance (nmr) spectrometry, J. Pharmaceut. Biomed. Anal.174 (2019) 639–643.

[72] Y.B. Monakhova, J. Fareed, Y.M. Yao, B.W.K. Diehl, Improving reliability of che-mometric models for authentication of species origin of heparin by switching from1D to 2D nmr experiments, J. Pharmaceut. Biomed. Anal. 153 (2018) 168–174.

[73] F. Zhang, K.B. Lee, R.J. Linhardt, Spr biosensor probing the interactions betweentimp-3 and heparin/gags, Biosensors 5 (2015) 500–512.

[74] F. Zhang, J.M. Beaudet, D.M. Luedeke, R.G. Walker, T.B. Thompson, R.J. Linhardt,Analysis of the interaction between heparin and follistatin and heparin and follis-tatin-ligand complexes using surface plasmon resonance, Biochemistry 51 (2012)6797–6803.

[75] H.H. Nguyen, J. Park, S. Kang, M. Kim, Surface plasmon resonance: a versatiletechnique for biosensor applications, Sensors (Basel) 15 (2015) 10481–10510.

[76] S.Y. Kim, J. Zhao, X. Liu, K. Fraser, L. Lin, X. Zhang, F. Zhang, J.S. Dordick,R.J. Linhardt, Interaction of zika virus envelope protein with glycosaminoglycans,Biochemistry 56 (2017) 1151–1162.

[77] J. Zhao, X. Liu, A. Malhotra, Q. Li, F. Zhang, R.J. Linhardt, Novel method formeasurement of heparin anticoagulant activity using spr, Anal. Biochem. 526(2017) 39–42.

[78] B.A. Prabowo, A. Purwidyantri, K.C. Liu, Surface plasmon resonance optical sensor:a review on light source technology, Biosensors 8 (2018) 80.

[79] S.D. Gan, K.R. Patel, Enzyme immunoassay and enzyme-linked immunosorbentassay, J. Invest. Dermatol. 133 (2013) e12.

[80] E. Dobrovolskaia, A. Gam, J.E. Slater, Competition enzyme-linked immunosorbant

assay (elisa) can be a sensitive method for the specific detection of small quantitiesof allergen in a complex mixture, Clin. Exp. Allergy 36 (2006) 525–530.

[81] L. Lin, Y. Yu, F. Zhang, X. Zhang, R.J. Linhardt, High-throughput method for inprocess monitoring of 3-o-sulfotransferase catalyzed sulfonation in bioengineeredheparin synthesis, Anal. Biochem. 586 (2019) 113419.

[82] R.Y. Zhan, Z. Fang, B. Liu, Naked-eye detection and quantification of heparin inserum with a cationic polythiophene, Anal. Chem. 82 (2010) 1326–1333.

[83] D.J. Murray, W.J. Brosnahan, B. Pennell, D. Kapalanski, J.M. Weiler, J. Olson,Heparin detection by the activated coagulation time: a comparison of the sensitivityof coagulation tests and heparin assays, J. Cardiothorac. Vasc. Anesth. 11 (1997)24–28.

[84] S. Pecorini, G. Camurri, L. Torrini, R. Ferraresi, Highly sensitive real-time pcrmethod to identify species origin in heparinoids, Anal. Bioanal. Chem. 412 (2020)289–298.

[85] C. Auguste, S. Dereux, C. Martinez, P. Anger, New developments in quantitativepolymerase chain reaction applied to control the quality of heparins, Anal. Bioanal.Chem. 399 (2011) 747–755.

[86] S.J. Kwon, K.B. Lee, K. Solakyildirim, S. Masuko, M. Ly, F. Zhang, L. Li, J.S. Dordick,R.J. Linhardt, Signal amplification by glyco-qpcr for ultrasensitive detection ofcarbohydrates: applications in glycobiology, Angew. Chem. Int. Ed. 51 (2012)11800–11804.

[87] J.N. Dai, C.G. Ma, P. Zhang, Y.Q. Fu, B.X. Shen, Recent progress in the developmentof fluorescent probes for detection of biothiols, Dyes Pigments 177 (2020) 13.

[88] B.X. Shen, Y. Qian, Red emission cysteine probe with high selectivity based onfluorescent protein chromophores and turn-on fluorescence in cell cultures, DyesPigments 166 (2019) 350–356.

[89] B.X. Shen, L.F. Wang, X. Zhi, Y. Qian, Construction of a red emission bodipy-basedprobe for tracing lysosomal viscosity changes in culture cells, Sensor. Actuator. BChem. 304 (2020) 9.

[90] B.X. Shen, W.Y. Zhu, X. Zhi, Y. Qian, A lysosome targeting probe based on fluor-escent protein chromophore for selectively detecting GSH and Cys in living cells,Talanta 208 (2020) 7.

[91] Q. Long, J.N. Zhao, B.D. Yin, H.T. Li, Y.Y. Zhang, S.Z. Yao, A novel label-free up-conversion fluorescence resonance energy transfer-nanosensor for ultrasensitivedetection of protamine and heparin, Anal. Biochem. 477 (2015) 28–34.

[92] J.J. Xu, A. Takai, M. Takeuchi, Red-green-blue trichromophoric nanoparticles withdual fluorescence resonance energy transfer: highly sensitive fluorogenic responsetoward polyanions, Chem.-Eur. J. 22 (2016) 13014–13018.

[93] Y.Q. Chang, C. Cai, L.Y. Li, J.J. Miao, E. Ucakturk, G.Y. Li, M. Ly, R.J. Linhardt,Ultrasensitive detection and quantification of acidic disaccharides using capillaryelectrophoresis and quantum dot-based fluorescence resonance energy transfer,Anal. Biochem. 85 (2013) 9356–9362.

[94] U. Warttinger, C. Giese, J. Harenberg, E. Holmer, R. Krämer, A fluorescent probeassay (heparin red) for direct detection of heparins in human plasma, Anal. Bioanal.Chem. 408 (2016) 8241–8251.

[95] X. Zhang, X.R. Han, K. Xia, Y.M. Xu, Y.M. Yang, K. Oshima, S.M. Haeger, M.J. Perez,S.A. McMurtry, J.A. Hippensteel, J.A. Ford, P.S. Herson, J. Liu, E.P. Schmidt,R.J. Linhardt, Circulating heparin oligosaccharides rapidly target the hippocampusin sepsis, potentially impacting cognitive functions, Proc. Natl. Acad. Sci. U.S.A.116 (2019) 9208–9213.

M. Qiao, et al. Talanta 219 (2020) 121270

15