49
R.J. Vonk B AS 5897041 Universiteit van Amsterdam 27 mei 2011 Determination of glycosaminoglycans in human serum Development of a method for the sizecharacterization of glycosaminoglycans

Determination of glycosaminoglycans - UvA...R.J. VONK 2 Determination of glycosaminoglycans in human serum Development of a method for the size-characterization of glycosaminoglycans

  • Upload
    others

  • View
    8

  • Download
    0

Embed Size (px)

Citation preview

R.J. Vonk B AS

5897041

Universiteit van Amsterdam

27 mei 2011

Determination of glycosaminoglycans in human

serumDevelopment of a method for the size‐characterization of glycosaminoglycans

R.J. VONK

2

DDeetteerrmmiinnaattiioonn ooff ggllyyccoossaammiinnooggllyyccaannss iinn

hhuummaann sseerruumm

DDeevveellooppmmeenntt ooff aa mmeetthhoodd ffoorr tthhee ssiizzee--cchhaarraacctteerriizzaattiioonn ooff ggllyyccoossaammiinnooggllyyccaannss

RRuuddyy VVoonnkk

Master of Science Analytical Chemistry track Universiteit van Amsterdam Name : R.J. Vonk B AS Student number : 5897041 Research period : October 2010 - April 2011 Supervisors : Dr. W. Th. Kok : R.N. Qureshi MSc

Figure 1: Front page; 4D-structure of polysaccharide hyaluronan. Reprinted from http://www.conformetrix.com

R.J. VONK

3

Tabel of contents

Samenvatting ................................................................................................5

Summary .......................................................................................................6

Abbreviations................................................................................................7

1 Introduction............................................................................................8

2 Glycosaminoglycans...........................................................................10

2.1 The structure of glycosaminoglycans .................................................. 10

2.2 Glycosaminoglycans disturbance in diseases ..................................... 12

3 Analytical methods..............................................................................14

3.1 AsFlFFF-UV-MALS-DRI....................................................................... 15

3.2 RP-HPLC-fluorescence........................................................................ 17

3.3 Pyrolysis-GC/MS.................................................................................. 18

4 Experimental ........................................................................................19

4.1 Chemicals ............................................................................................ 19

4.2 Apparatus............................................................................................. 20

4.3 Method optimization............................................................................. 20

4.3.1 Deproteination of serum samples .................................................. 20 4.3.2 AsFlFFF-MALS-DRI method .......................................................... 21 4.3.3 Enzymatic cleavage ....................................................................... 21 4.3.4 Derivatization with AMAC............................................................... 21 4.3.5 RP-HPLC method........................................................................... 22 4.3.6 Pyrolysis-GC/MS............................................................................ 22

5 Results and discussion.......................................................................23

5.1 Method optimization............................................................................. 23

5.1.1 Deproteination of serum samples .................................................. 23 5.1.2 AsFlFFF-MALS-DRI method .......................................................... 25 5.1.3 Enzymatic cleavage ....................................................................... 29 5.1.4 Derivatization with AMAC............................................................... 31 5.1.5 RP-HPLC method........................................................................... 33 5.1.6 Pyrolysis -GC/MS........................................................................... 35

5.2 Sample preparation overview after optimizing method........................ 35

5.3 Analysis of serum samples .................................................................. 36

R.J. VONK

4

6 Conclusions .........................................................................................37

6.1 Current method .................................................................................... 37

6.2 Recommendation for future work......................................................... 37

7 Literature ..............................................................................................40

Appendix A: AsFlFFF method....................................................................44

Appendix B: RP-HPLC-FLR method..........................................................45

Appendix C: Pyrolysis-GC/MS method .....................................................46

Appendix D: Major project proposal .........................................................47

Acknowledgements / Dankwoord..............................................................49

R.J. VONK

5

Samenvatting

Dit verslag heb ik geschreven naar aanleiding van mijn Research project aan het

einde van de Master of Chemistry met Analytical Science als specialisatie. Deze

Master of Chemistry heb ik gevolgd aan de UvA en de VU in de periode van

september 2008 t/m mei 2011. Dit project is uitgevoerd bij de Analytical

Chemistry groep van de afdeling HIMS aan de UvA onder begeleiding van Dr. W.

Th. Kok.

Tijdens dit project heb ik geprobeerd om een analytische methode op te zetten

voor het bepalen van de grootte en het kwantificeren van polysacchariden in

menselijk bloed. Evenals de grootte van de polysacchariden veranderen ook de

gehalten van deze polysacchariden in het bloed bij patiënten met verschillende

ziekten. De grootte in combinatie met het gehalte kunnen gebruikt worden als

diagnostische tool voor het juist behandelen van patiënten. Om deze

vraagstelling te beantwoorden is er gebruik gemaakt van een Asymmetric Flow-

Flied Flow Fractionation (AsFlFFF) off-line gecombineerd met HPLC-

fluorescentie detectie.

Bij AsFlFFF worden de moleculen geïnjecteerd in een vloeistofstroom. De

AsFlFFF techniek scheidt macromoleculen (>10kDa) door middel van het

toepassen van een kracht loodrecht op de vloeistof stroom (in dit geval een

andere vloeistofstroom). Door deze kracht worden grote moleculen meer

vertraagd dan kleinere moleculen en elueren dus later uit het systeem.

Vervolgens worden, in de opgevangen AsFlFFF fracties, de gehalten van de

polysacchariden bepaald d.m.v. HPLC fluorescentie. De polysacchariden uit de

fracties kunnen niet direct geïnjecteerd worden op de HPLC. Door enzymen

worden de polysacchariden omgezet in disacchariden. Deze disacchariden zelf

geven geen signaal op een fluorescentie detector en daarvoor moeten deze

disacchariden eerst gederivatiseerd worden met een fluorescent actief label.

Tijdens dit project zijn de verschillende enzymatische stappen, de derivatisatie

reactie en de HPLC methode geoptimaliseerd voor een lagere detectielimiet en

een betere omzetting van polysacchariden naar disacchariden.

Al met al een leuk en uitdagend project waarvan ik heb geleerd hoe het is om met

weinig geld en veel tijd een leuk project te kunnen uitvoeren, dit in tegenstelling

tot mijn vorige werk bij Abbott Healthcare BV waar het juist andersom was.

R.J. VONK

6

Summary

In this current research thesis the method development for the size-

characterization and quantification of glycosaminoglycans (GAGs) in human

blood serum will be described.

The separation between serum proteins (BSA) and hyaluronan (HA) on an

asymmetric flow field-flow fractionation (AsFlFFF) has been achieved using

standard solutions. The MW distribution of HA standard was in the range of

120kDa to 3.2MDa. In the collected AsFlFFF-fractions the concentration of HA

was determined with a recovery of 18±9%. Baseline separation between HA and

multiple other GAGs had been achieved on a RP-HPLC-FLR method. The limit of

decision for HA was 20ng/mL. With the achieved limit of decision measurement of

HA in human serum should be possible. Although the limit of decision was low

enough for determination of HA in serum samples, HA could not be quantified in

serum samples so far.

R.J. VONK

7

Abbreviations

ACN acetonitril

AMAC 2-aminoacridone

AsFlFFF asymmetric flow-field flow fractionation

BSA bovine serum albumin

cIMT cardio intima-media thickness

CS chondroitin sulfate

ΔDi disaccharide unit

DRI refractive index detector

DS dermatan sulfate

Fch detector flow

Fcr cross flow

FLR fluorescence detection

GAGs glycosaminoglycans

Gal β-D-galactose

GalNAc N-acetyl-D-galactosamine

GlcA β-D-glucuronic acid

GlcNAc N-acetyl-D-glucosamine

HA hyaluronan

HPLC high performance liquid chromatography

HS heparin sulphate

IdoA α-D-iduronic acid

KS keratan sulfate

MALS multi angle light scattering

MS mass spectrometry

MW molecular weight

NaAce sodium acetate

PB phosphate buffer

PBS phosphate buffer containing saline

Py-GC/MS pyrolysis gas chromatography coupled to mass spectrometry

SEC size exclusion chromatography

TMAH tetramethylammonium hydroxide

TRIS-Cl tris(hydroxymethyl)aminomethane hydrochloride

UV ultra violet absorbance detector

R.J. VONK

8

1 Introduction

The blood vessel wall is covered with glycocalyx which forms a gel-like

endothelial lining [01]. Glycoproteins and proteoglycans are the building blocks

for these glycocalyx. Some glycoproteins and proteoglycans are membrane-

bound and some are soluble molecules [02]. Proteoglycans consist of a core

protein (such as syndecans and glypicans) which in turn is the backbone for

unbranched polysaccharide chains called glycosaminoglycans (GAGs) [01-02].

One GAG, named hyaluronic acid / hyaluronan [03] (HA), is not connected to the

proteoglycan backbone but is synthesized in the glycocalyx matrix [04]. More

than one GAG can bind to the core protein [02]. The endothelial glycocalyx is

important in the vascular integrity and permeability [01-02] and repels red blood

cells from the endothelium [02]. More details about the endothelial glycogalyx can

be found in review articles from Broekhuizen et al. [01] and Reitsma et al. [02].

Figure 2 visualizes the complete network of the glycocalyx, proteoglycans and

GAG.

Figure 2: Schematic representation of the endothelial glycocalyx, showing its main components. Left: The endothelial glycocalyx located on the luminal side of the

vascular endothelium. Right: Components of the endothelial glycocalyx. Incorporated in and on top of this grid are plasma and endothelium-derived soluble components, including HA and other soluble proteoglycans. Note that this figure is not drawn to scale; its purpose is to illustrate glycocalyx composition. This figure and legend are

reprinted from Reitsma et al. [02]

hyaluronan

R.J. VONK

9

GAGs are linear, unbranched, polysaccharides of a repeating disaccharide [05].

Based on the structure GAGs can be subdivided into four groups, namely

hyaluronan (HA), chondroitin sulfate (CS) / dermatan sulfate (DS), keratan sulfate

(KS) and heparin / heparan sulfate (HS) [06].

Disturbance of the molecular weight (MW) and plasma levels of the GAGs have

been related to diseases such as Rheumatoid arthritis [07-08]. In this disease the

change in MW distribution in combination with the plasma levels of the GAGs can

be used as a biomarker or diagnostic tool [09]. Therefore, the focus of this project

will be the development of a sensitive method for size-characterization and

quantification of GAGs in human blood serum samples. In order to develop this

method a novel approach for off-line combination of Asymmetric Flow-Field Flow

Fractionation combined with Ultra Violet absorbance detector, Multi Angle Light

Scattering, and Refractive Index detection (AsFlFFF-UV-MALS-DRI) and High

Performance Liquid Chromatography combined with Fluorescence detection

(HPLC-FLR) was investigated. Furthermore, Pyrolysis Gas Chromatography

coupled to a mass spectrometry (Py-GC/MS) will be investigated as alternative

for the HPLC-FLR method with the purpose of simplifying the sample preparation.

To measure GAGs in serum samples the following sample preparation steps

have been investigated:

deproteination of serum samples

size-based separation of the GAGs on the AsFlFFF

enzymatic cleavage of polysaccharides into disaccharide units

derivatization of disaccharides with a fluorescent label

the conditions of the RP-HPLC method

the interpretation of the Py-GC/MS data of the GAGs

The next chapter will give more information about the individual GAGs and their

structural properties. The analytical tools that were used are explained in chapter

3. In chapter 4 the details of the experiments performed are described and in

chapter 5 the results and discussion are given. Chapter 6 gives the conclusions

and recommendations for future work.

R.J. VONK

10

2 Glycosaminoglycans

Synthesis routes and location of the different GAGs in the human body are

different. CS, DS and HS are synthesized in the Golgi apparatus and are bound

via a tetrasaccharide to the proteoglycan backbone [04,10]. HA on the other hand

is synthesized in the extracellular glycocalyx matrix and is not bound to the

proteoglycan backbone [04]. The structural differences and the role of the

different GAGs will be explained briefly.

2.1 The structure of glycosaminoglycans

The main structure of all the GAGs is basically the same. The building blocks are

disaccharide units which consist of a hexosamine unit and a hexuronic acid

[05,10]. This second sugar unit is a galactose unit for KS [05].

Hyaluronan (HA) polysaccharide is the only non-sulfated GAG which consists out

of a repeat disaccharide unit of a β-D-glucuronic acid (GlcA) and N-acetyl-D-

glucosamine (GlcNAc). These two monosaccharide units are linked through the β

1,3 bond and the disaccharides are bound through the β 1,4 bond [10]. The

structure of the repeating disaccharide unit of HA (GlcA-β1,3-GlcNAc) is shown in

Figure 3.

Compared to the size of HA (~0.1-10 MDa) other GAG polysaccharides are

relatively short, commonly 15-20 kDa [05,07]. Unlike HA one or more sulfate

groups can be present in the other GAGs.

Chondroitin sulphate (CS) occurs in different sulfated states. The repeating

disaccharide unit, a GlcA linked through the β 1,3 bond to a N-acetyl-D-

galactosamine (GalNAc), is unchanged in the different sulfated states. The

position and the number of sulfated groups can change. Sulphated groups can

occur on three different positions, the 2-carbon position on the GlcA, the 4-carbon

and 6-carbon position in the GalNAc [10]. The repeating disaccharide structure of

CS (GlcA-β1,3-GalNAc) is shown in Figure 3. CS is linked by a tertrasaccharide

to the proteoglycan backbone [04].

.

R.J. VONK

11

O

O

OO

OR

OHOR

OR

O*

*

NH

O CH3

OOH

ß-D-glucuronic acid (GlcA) N -acetyl-D-galactosamine (GalNAc)

CS disaccharide

R = H or SO3H

O

O

OO

OR

OHOR

OR

O*

*

NH

O CH3

OOH

DS disaccharide

N -acetyl-D-galactosamine (GalNAc)a-D-iduronic acid (IdoA)

R = H or SO3H

Figure 3: Structure of the repeating disaccharide unit of HA, CS and DS. R = -H or -SO3H

The disaccharide backbone of dermatan sulphate (DS) is almost similar to that of

CS. Instead of GlcA DS contains an α-D-iduronatic acid (IdoA) monosaccharide

unit connected via α 1,3 bond to a GalNAc unit. DS can be sulfated on the same

carbon positions as CS. The disaccharides are connected via the β 1,4 bond,

similar to HA [10]. Figure 3 shows the structure of the repeating disaccharide unit

of DS (IdoA-α1,3-GlcNAc). DS is linked by a tertrasaccharide to the proteoglycan

backbone [04].

Keratan sulphate (KS) disaccharide backbone consist of a β-D-galactose (Gal)

linked via β 1,4 bond to a GlcA unit. The disaccharide units are linked via a the β

1,3 to each other. KS can be sulphated on the 6-position on both the Gal and the

GlcA unit [10].

The disaccharide backbone of heparan sulphate (HS) and heparin consist of a

GlcA or IdoA connected to a GlcNAc unit. HS can be sulphated in many different

ways, a total of 21 different HS disaccharide backbone units have been identified

[11]. The scope of the project was to determine HA, CS and DS in human blood

serum. Details on the biological and structural properties of HS and KS can be

found in papers by Broekhuizen et al. [01], Sasisekharan et al. [10] and Esko et

al. [11].

O

O

OO OH

OHOH

OH

O*

*

NH

O CH3

OOH

ß-D-glucuronic acid (GlcA) N-acetyl-D-glucosamine (GlcNAc)

HA disaccharide

123

45

6

123

45

6

R.J. VONK

12

2.2 Glycosaminoglycans disturbance in diseases

The disturbance of the glycocalyx structure and changes in the level of GAGs

have been observed in patients with different diseases. GAGs have different

biological functions in the human body. Disturbance of the glyocalyx is related to

inflammation [01], cancer [09], hyperglycemia [01], atherosclerosis [09], and

shear stress [12]. When the endothelial glycocalyx is damaged the adhesion of

circulating cells, influx of lipoproteins and leakage of macromolecules to the

endothelium have been described [01].

HA is found in nearly all tissues and fluids of the body: plasma [13], the

connective tissue [14], lymph [15], synovial fluid [08], eye [16], urine [13], skin

[13], brain [05], and umbilical cord [13]. HA plays a role in the water regulation in

tissue by transporting water through extracellular matrix [05,07] Furthermore it

plays a role as lubricant in the joints [05,17].

The role of HA has been investigated for the understanding and diagnosis of

various diseases, such as malignant mesothelioma [18], exfoliation syndrome

[16], abdominal aortic aneurysm [19] and Rheumatoid arthritis [07-08].

The relation between the increased plasma levels of HA and the increase of the

cardio intima-media thickness (cIMT) in type 1 diabetes patients has been

reported [12]. Also an increase in both the MW and plasma levels of HA were

reported in patients with Primary biliary cirrhosis and Rheumatoid arthritis [07]. In

contrast to the increased MW of HA in plasma, a decreased MW of HA in

synovial fluid has been reported in patients who suffer from Rheumatoid arthritis

[08]. In exfoliation syndrome HA is overproduced in the anterior chamber and the

cornea [16].

The role of CS and DS side chains of the proteoglycan is described in the review

article of Sasisekharan et al. [10]. CS and DS provide electrostatic forces to the

proteoglycan which enables joints to function despite the shear, tensile and

dynamic compressive forces. Furthermore, the mechanical stiffness of cartilage

tissue is provided by CS and DS [10]. Analysis of CS and DS has been used for

diagnosis and understanding of exfoliation syndrome and abdominal aortic

aneurysm [16,19].

In these diseases the levels of some of the GAGs is changed while the other

levels of the GAGs remain constant. In patients with e.g. exfoliation syndrome the

R.J. VONK

13

levels of non-sulphated CS/DS were not increased in the aqueous humor of the

eye, in contrast to HA [16]. Another example is in abdominal aortic aneurysm in

which the aorta tissue levels of both CS and HA were decreased compared to

healthy persons while the aorta tissue levels of DS remained unchanged [19].

The scope of the project was to measure GAGs in plasma. The serum samples

for this study were related to type 1 diabetes patients. The changes in MW

distribution and plasma levels of HA related to the increase of the cIMT have to

be determined [12].

R.J. VONK

14

3 Analytical methods

GAGs can be separated and quantified by different separation techniques such

as HPLC [06,20-21], polyacrylamide gel electrophoresis [22] and capillary

electrophoresis [21,23-24]. These separation methods can be hyphenated with

different detection techniques such as fluorescence detection (FLR) [20-21,24]

and (tandem)mass spectrometry (MS(/MS)) [06,23].

HPLC-FLR coupled to a MS detector has been used for determination of GAGs in

human serum by Volpi [06]. The FLR detector was used for quantification of the

compounds and the MS detector was used for selectivity. The linear range of the

method, using MS as detection, was between 0.1 and 200 ng. The reported

levels of HA in human blood vary between different studies and between healthy

persons and diseased patients. The HA levels in blood serum in healthy persons

ranges from 10 to 300 ng/mL [07,15,25].

For the current project a HPLC-FLR apparatus was applied. In order to measure

HA in human blood serum, with an injection volume around 10µL, the HPLC-FLR

method described by Volpi [06] needed to be adapted to gain enough sensitivity

and selectivity for quantification.

Additional to quantification of the GAGs, determination of the MW distribution of

GAGs in human blood serum was required. The combination of the MW and the

change in serum/plasma levels of e.g. HA can be used as a biomarker approach

in cardio vascular diseases such as cIMT, Rheumatoid arthritis and Primary

biliary cirrhosis [01,07,12].

The MW distribution of the aqueous soluble GAGs polysaccharide can be

determined by AsFlFFF [26-28] and size exclusion chromatography (SEC) [29-

31]. For this project AsFlFFF was used for separation. The advantage of AsFlFFF

is that no column is used for separation. Therefore, this technique has no high-

MW cut-off caused by the pore size of the particles, compared to SEC. High MW

polymers are separated without any major structural breakdown since no

shearing forces are being applied [32].

R.J. VONK

15

3.1 AsFlFFF-UV-MALS-DRI

Separation of polymers by AsFlFFF is without a stationary phase that may

interact, degrade, or alter the sample and therefore is gentle, rapid, and non-

destructive [32]. A schematic view of AsFlFFF is depicted in Figure 4. A channel

flow runs from the inflow (left) to the outflow (right), the sample is introduced at

the sample inflow (middle). Before the sample is injected into the channel, the

outflow is reversed and the net flow is zero at the sample inflow point, which

causes focusing of the analytes to the channel after injection. Once the analytes

are focused the flow runs again from inflow to outlflow and the analytes are

transported over the channel. A side view of the channel is given in the “Exploded

view” in Figure 4. The upper channel plate is impermeable. The bottom channel

plate, on the other hand, is made of a porous frit material. To prevent the lost of

analytes through the bottom plate an ultra filtration membrane covers the bottom

plate [32].

When the analytes are transported over the channel a parabolic flow profile is

created due to the laminar flow of the liquid. This profile results in slower moving

stream closer to the walls than it does at the center of the channel flow [32-33].

Figure 4: Schematic drawing of AsFlFFF. Reprinted and adjusted from website of Wyatt [32]

R.J. VONK

16

When the perpendicular force field (in the case of AsFlFFF a liquid flow) is

applied to the flowing, laminar stream, the analytes are driven towards the

boundary layer the so-called "accumulation wall" of the channel [33].

Diffusion, due to concentration difference in the channel [33], creates a

counteracting motion. Smaller particles, or particles with a higher diffusion rate,

tend to diffuse higher in the channel, where the longitudinal flow is faster. The

smaller particles are transported much more rapidly along the AsFlFFF-channel

than the larger particles. This is the opposite of a SEC separation in which the

large molecules elute first [32].

The elution time in AsFlFFF is related to the diffusion rate of the molecules and

can be calculated via equation 3.1 [34]:

sch

crc

0in2

ch

cr2

r dF

FA

AF

ln2kT

πηw

F

F

6D

wt

3.1

tr = elution time

w = channel thickness

D = diffusion coefficient

Fcr = cross flow

Fch = detector / channel flow

Fin = total flow through the channel (Fcr + Fch)

A0 = area to focus point from inlet

Ac = area of accumulation wall

ds = hydrodynamic diameter

For quantifying and MW-calibration an UV detector, MALS detector, and a DRI

detector were used. Response of the UV detector is a combination of absorption

of light and light scattering. The light scattering occurs by bigger particles and is a

function of particle size [34]. In the MALS laser light is scattered by passing

macro-molecules. All the scattered light is detected by 18 different detectors

under different angles (θ). When the composition of the sample cell changes the

.

Figure 5: Schematics of DRI detector, light passing the reference and sample cell can be deflected by molecules present in the sample cell

R.J. VONK

17

deflections of the light in the DRI is changed when passing the reference cell and

sample cell. Deflection of light is depended on the concentration of the passing

solution. A schematic drawing of a DRI is given in Figure 5. By using equation 3.2

and equation 3.3 the MW of the passing molecule could be determined.

I(sc) = intensity of the scattered light in the MALS detector

k(θ) = instrumental constant of the MALS, based on angle

measured, and used laser wavelength

c = the concentration of the solute molecules

MW = the weight-average molar mass

RI = refractive index deflection

k’ = instrumental constant of the DRI detector, based

on dn/dc, and refractive index of the solvent

3.2 RP-HPLC-fluorescence

For this project RP-HPLC-FLR was chosen for separation and quantification of

the different GAGs. To separate and determine the different GAGs with HPLC-

FLR two additional steps needed to be performed in order to measure the

polysaccharides which were present in the AsFlFFF fractions. First, enzymes

have to digest the polysaccharides into their disaccharide units (ΔDi) because

polysaccharides are too large for separation with RP-HPLC [35]. Secondly, a

fluorescent active label needs to be bound to the compounds. Once the

disaccharides were labeled separation with a RP system was possible and at the

same time this label provides possibilities for sensitive and selective detection.

AMAC is a hydrophobic compound which is retained on a RP column [20] and

have been used for separation and detection of disaccharides [06,20,22,35]. If

AMAC is derivatized to a disaccharide the combination is more polar and will

elute earlier from RP column than AMAC which is non-derivatized. The

derivatization reaction of HA disaccharide (ΔDiHA), so after enzymatic cleavage

of polysaccharide, with AMAC is depicted in Figure 6. This scheme can be used

for other GAGs as well. AMAC can also react with the present tetra- or

hexasaccharide. So a loss in signal might be observed if AMAC reacts with a lot

of other compounds present in the sample.

MW *c * θ)k(I(sc)

c * k'RI

3.2

3.3

R.J. VONK

18

Figure 6: Reaction scheme of unsaturated disaccharide (ΔDiHA) with fluorescent label AMAC [06]

3.3 Pyrolysis-GC/MS

A method based on GC/MS was investigated as an alternative for the HPLC-FLR

method. When using Py-GC/MS instead of the HPLC-FLR method less sample

preparation will be needed since the polysaccharide can be injected directly to

the Py-GC/MS and no fluorescent label has to be connected for measurement.

Prior to analysis an online reaction was performed using tetramethylammonium

hydroxide (TMAH) for methylating present hydroxyl groups in the

polysaccharides. The formed products were pyrolyzed and the fragments were

separated by the GC column and electron impact (EI)-MS was used for

identifying products and fragments.

R.J. VONK

19

4 Experimental

4.1 Chemicals

Dissacharide standards of GAGs were purchased from Iduron, Manchester, UK

and are listed in Table 1. The water for the mobile phase was of MQ standard

quality (>18.2MΩ). The ACN was obtained from BioSolve, Valkenswaard, The

Netherlands. Coomassie Blue dye was purchased from Bio-Rad Laboratories,

Veenendaal, The Netherlands. The purchased materials from Sigma Aldrich,

St. Louis, USA were;

RP-HPLC column; Ascentis Express, C18, 2.1x150mm, 2.7μm

Proteinase K from Tritirachium album

Chondroitinase ABC from Proteus vulgaris

Hyaluronidase from bovine testes

HA polysaccharide potassium and sodium salt from human umbilical cord

Other salts of the highest quality

Table 1: ΔDiGAGs names, backbone description, and used abbreviations

name Disaccharide backbone Abbreviation 1

hyaluronan GlcA- β1,3-GlcNAc ΔDiHA

chondroitin GlcA-β1,3-GalNAc ΔDi0s

chondroitin 4-disulfate GlcA-β1,3-GalNAc(4s) ΔDi4s

chondroitin 6-disulfate GlcA-β1,3-GalNAc(6s) ΔDi6s

chondroitin 4,6-disulfate GlcA-β1,3-GalNAc(4s,6s) ΔDi4,6dis

chondroitin 2,4,6-trisulfate GlcA(2s)-β1,3-GalNAc(4s,6s) ΔDi2,4,6tris

1 Abbreviations start with ΔDi next number(s) represents the position of the sulphated group(s), the “s”, “dis” and “tris” were respectively used for monosulphated, disulphated and trisulphated variant of the GAG.

R.J. VONK

20

4.2 Apparatus

An overview of the instruments used is given in the scheme below. AsFlFFF-UV-MALS-DRI

apparatus type supplier

Pump HP1200 isocratic apparatus Agilent Technologies, Santa Clara, USA

Flow control Eclipse 2 MALS DAWN DSP laser photometer Software Eclipse and Astra v4 and v5

Wyatt Technology Europe GmbH, Dernbach, Germany

UV UV757 Applied Biosystems, Foster City, USA DRI RID-10A Shimadzu, Colombia, USA

HPLC-FLR

apparatus type supplier HPLC Alliance W2690 FLR Acquity FLR UPF Software Empower build 2.154

Waters, Milford, USA

Py-GC/MS

apparatus type supplier GC GC-2010 MS GCMS-QP2010

Shimadzu, Colombia, USA

Injector Optic 3 PTV Software GCMS analysis

ATAS GL International B.V., Eindhoven, The Netherlands

4.3 Method optimization

The previous method for quantification of GAGs in human blood serum [06] was

optimized for the size-characterization of GAGs in human blood serum. The

following steps were optimized: deproteination of serum samples, enzymatic

cleavage, derivatization of disaccharide with fluorescent label and the conditions

of the RP-HPLC method. Size-characterization of the GAGs on AsFlFFF was an

extra step added to the sample preparation process.

4.3.1 Deproteination of serum samples

Optimization of the deproteination step with proteinase K was performed with 60

mg/mL Bovine Serum Albumin (BSA) in water. The added amounts of proteinase

K and incubation time were optimized. Proteinase K was added at 600, 1800 and

3600mU, samples were incubated up to 48h at 37°C.

R.J. VONK

21

4.3.2 AsFlFFF-MALS-DRI method

An AsFlFFF method was setup for separation of GAGs from other larger

molecules, such as enzymes, which were present in serum samples. Calibration

of the HA polysaccharide and separation from BSA was investigated on AsFlFFF

with mobile phases of water, NaNO3 and PBS. Channel space was made by

cutting a 350-μm-thick Mylar spacer with a tip-to-tip length of 13.5cm. The

channel had a trapezoidal shape (the initial breadth decreased from 2.15cm to

0.3cm), and the volume of the channel was 0.56cm3.

4.3.3 Enzymatic cleavage

In previous studies either chondroitinase ABC [05,36] or hyaluronidase [21,36]

were used for the enzymatic cleavage of the polysaccharides into their ΔDi. In

this study the enzymatic conditions for a combination of both chondroitinase ABC

and hyaluronidase have been investigated. The absolute amount of

hyaluronidase added was kept constant (10U) while the amount of chondroitinase

ABC added varied between 1 and 10mU. Sample volumes were 100μL of HA

polysaccharide concentrations of 0.5, 5 and 100μg/mL. Incubations were

performed at 37°C up to 48h. Different solutions for enzymatic cleavage were

used namely, NaAce at pH 6.82, PBS and PB buffer at pH 7.43.

4.3.4 Derivatization with AMAC

All consulted studies used the method of Jackson [22], optimized by Kitawaga

and coworkers [21]. In this method AMAC, sodium-cyanoborohydride (NaCNBH3)

and DSMO/water were added to the samples. In this current study the optimal

derivatization conditions were investigated. First, the concentration of the AMAC

solution was varied between 0.05M and 0.1M. Secondly, the derivatization

reaction was monitored at 20°C up to 100h after incubation at 37°C for 2h and

16h. Thirdly, a lyophilization step was investigated to check if the samples could

be concentrated after derivatization. Water and DMSO/water (50/50 (v/v%)) were

used for dissolving the samples after lyophilization.

2 50mM NaAce solution for enzymatic cleavage contained 0.02% BSA for activation of chondroitinase ABC. 3 PBS & PB buffers for enzymatic cleavage contained 50mM NaAce and 0.02% BSA for activation of chondroitinase ABC.

R.J. VONK

22

4.3.5 RP-HPLC method

The separation of multiple derivatized standard ΔDi GAGs solutions has been

verified on a C18, 2.1x150mm, 2.7µm column. The mobile phases used were

60mM NH4Ace at pH 5.6 and ACN. The gradient used in previous studies was

optimized for a faster sample run time and the new column dimension. Column

temperature was 30°C with a constant flow at 0.2mL/min and an injection volume

of 5μL. The initial percentage ACN was increased from 2% up to 20% in 32

minutes, next increased to 90% in 3 minutes and kept constant for 2 minutes,

finally back to 2% in 1 minute and kept constant for 7 minutes before starting a

new injection. For the fluorescence detector a λexcitation= 428nm and λemission =

525nm were used [06]. The linearity of the ∆DiGAG standards mentioned in

Table 1 was checked for injected levels of 0, 5, 10, 20, 50, 100, and 1000 ng/mL.

4.3.6 Pyrolysis-GC/MS

For the alternative GC analysis of high MW polar polysaccharides online

methylation and pyrolysis were used for formation of products. Fragment ions

were formed by an EI source and the mass spectrometry was used for

identification of the formed products and fragments. Both EI and pyrolysis are

high energetic processes which cause lot of products and fragments to be formed

and data interpretation will be difficult. To get some idea about the formed

products and fragments the both monosaccharide units of HA, GlcA and GlcNAc,

were injected separately from HA. The concentration of the three compounds was

500µg/mL. All injections were performed at different incubation temperatures in

the range of 50°C to 125°C and at different pyrolysis temperatures in the range of

225°C to 500°C for optimizing the conditions. The used TMAH concentration was

2.5% in water.

R.J. VONK

23

5 Results and discussion

HA was taken as a model compound for a size-characterization of GAGs in

serum samples. The separation on AsFlFFF, enzymatic cleavage and

derivatization process, as described in chapter 4 sessions 3.2, 3.3 and 3.4, were

only optimized for HA. The baseline separation on RP-HPLC-FLR of different

AMAC-labeled ΔDiGAGs was verified.

5.1 Method optimization

5.1.1 Deproteination of serum samples

Albumin concentration in human blood ranges from 35 to 55mg/mL [37-38].

Injecting this amount of proteins to the AsFlFFF-channel will ruin the AMAC

derivatization in the individual collected fractions. Furthermore, possible complex

forming of HA with proteins might disturb the MW determination. Deproteination

was required before injecting the samples in the AsFlFFF. Proteinase K

hydrolyses the proteins (serum albumin) into small peptide fragment [39] and has

been used in previous studies [06]. The reaction time had to be optimized and

therefore the proteinase K activity was monitored by using a BSA solution. After

incubation with proteinase K the total protein concentration was monitored using

Coomassie Blue dye [40]. Intact protein binds to the dye and hereby the

absorbance maximum of the dye shifts from 465 to 595nm [40]. The decrease in

protein concentration was monitored at 595nm.

BSA samples were 10 times diluted in 50mM Tris(hydroxymethyl)-aminomethane

hydrochloride (TRIS-Cl) buffer (~pH 7) containing ~0.1% SDS for an optimal

enzymatic environment [41]. Three different absolute amounts of proteinase K

added were 600, 1800, and 3600mU the diamond, cube, and triangular marks,

respectively in Figure 8. Incubation was performed at 37°C for maximum of 48h.

An incubation temperature of 60°C, used in a previous study [06], caused

proteins to denaturate and no HA could be extracted.

After incubated with proteinase K the samples were 4 times diluted in water

because both TRIS and SDS influence the absorbance maximum of Coomassie

Blue [40]. Dilution of BSA in TRIS buffer was performed with a ratio of

respectively 1:9. Before adding the dye the BSA concentration was 1.5mg/mL. A

BSA calibration curve was prepared via the same scheme as the samples.

R.J. VONK

24

Figure 7 shows the BSA calibration curve and the proteinase K activity is shown

in Figure 8. The activity is displayed as percentage of BSA measured, compared

to the control level (0h incubation).

Figure 7: Calibration curve of BSA using Coomassie Blue dye for visualizing proteinase K activity. Highest BSA concentration used was 60mg/mL in water, 10 times diluted in TRIS-Cl

Figure 8: Proteinase K activity displayed as % BSA measured compared to control (0h incubation). BSA concentration used was 60mg/mL in water, before incubation 10 times

diluted in TRIS-Cl

R.J. VONK

25

Linear curve (R2=0994) was observed for the BSA concentration (spheres). All

the different concentrations of proteinase K show similar activity in 48h. The

signal of ~15% proteins after 48h incubation was not caused by proteinase K

since the enzyme did not gave a shift in the absorbance maximum of the dye

(data not shown). This 15% signal might be caused by BSA left in solution.

Additionally, an experiment was performed in which the dilution ratio of the BSA

in TRIS buffer was changed to 1:46 and 1:1 with the same final BSA

concentration of 1.5mg/mL before adding the dye (data not shown). The high

concentrations of SDS and TRIS in the 1:46 experiment influenced the

absorbance maximum of the Coomassie Blue too much so no BSA concentration

could be determined in these samples. For the other dilution ratio (1:1) the

percentage BSA after adding 1600mU proteinase K and incubation for 48h at

37°C was 25% compared to the 0h incubation. This low activity of the proteinase

K might be caused by the low TRIS concentration / low buffer capacity during

incubation.

5.1.2 AsFlFFF-MALS-DRI method

After enzymatic deproteination of serum samples, still peptides and proteins

might be present in the sample. When the sample is injected to an AsFlFFF-

channel, the peptides will be lost due to the 10kDa cut-off of the used channel

membrane. The proteins on the other hand has to be separated from the HA by

the AsFlFFF method. HA polysaccharide is an aqueous soluble molecule and

therefore the use of water as mobile phase in AsFlFFF-MALS-DRI was preferred.

Normalization of the MALS signals was performed with a BSA solution

(MW=66kDa). The RIcal constant of the used DRI detector was determined using KCl

solution. Separation of BSA and HA was investigated using PBS4, 10mM NaNO3

and water.

First water was used a mobile phase but BSA and HA were not retained in the

channel using a cross flow / detector flow (Fcr/Fch) value of three. AsFlFFF

program details are given in Appendix A. Due to the low ionic strength of water,

HA and BSA cannot approach the channel wall due to electrostatic repulsion

4 PBS solution was prepared by weighing 1.03 gr NaH2PO4.H2O, 4.25 gr Na2HPO4.2H2O, 8.06 gr NaCl and 0.4 gr KCl dissolved in 1L water

R.J. VONK

26

between particles and the wall [42]. Due to this repulsion BSA and HA were not

sufficiently driven towards the accumulation wall during focusing and therefore

the equilibrium inside the channel was not reached and the molecules were

eluted at an elevated position [42-43]. When using a Fcr/Fch–value of five HA

could be slightly retained in the AsFlFFF-channel, as shown in Figure 9.

Figure 9: Separation of HA polysaccharide (Na-salt) on AsFlFFF with a high cross flow using water as mobile phase. Concentration HA was 1mg/mL, 50µL injection volume and detector

flow 0.6mL/min, secondary Y-axis showed the cross flow program used. No baseline subtraction was applied

No separation between BSA and HA could be obtained while using water as

mobile phase and applying this high cross flow. Next, 10mM NaNO3 solution was

used as mobile phase for separation of BSA and HA. The ionic strength of this

10mM NaNO3 solution was high enough to overcome the electrostatic repulsion

and a separation between BSA and HA was obtained, even at Fcr/Fch–value of

three. BSA and HA eluted 6.0 and 18 minutes after injection started, respectively.

The DRI signal showed a third peak between BSA and HA. Since this third peak

influenced the MW calibrations another mobile phase was tried. Finally, PBS was

used and it showed a nice separation between BSA and HA. Solution containing

1mg/mL of both BSA and HA was injected. Elution times of BSA and HA were 5.7

and 20 minutes, respectively. The DRI signal of the AsFlFFF separation between

BSA and HA using PBS was shown in Figure 10a with the used cross flow

program on the secondary y-axis. Figure 10b showed the MALS signal (θ = 90°)

of HA and BSA with the MW-distribution calibrations on the secondary y-axis.

Additionally, fractions per minute were collected from 10 to 25 minutes,

R.J. VONK

27

lyophilized, and reconstituted in 50mM sodium acetate (NaAce). In each fraction

the concentration of ΔDiHA was determined after lyophilization and enzymatic

cleavage (24h). The concentration (μg/mL) of the ΔDiHA in the separate fractions

and the observed DRI signal are given in Figure 10c. For calculation of the

AsFlFFF yield the weighted amount of HA (1mg/mL) was corrected using the

65% purity provided by the supplier and corrected for the 48±17% enzymatic

yield (see PBS-6, 10mU chondroitinase ABC in chapter 5.1.3).

The MW of the HA potassium salt eluting between 17 and 26 minutes was in the

range of 0.2 to 3.4MDa. Figure 10c shows that the fractions of 10 to 17 minutes

contained HA. The MW of HA eluting between 10 and 17 minutes was in the

range of 120 to 200kDa. The total amount ΔDiHA in the fractions collected was

6±3μg this was 18±9% (ave±stdev, n=2) of the injected absolute amount HA.

During AsFlFFF ~80% of the injected HA was lost. This might be low molecular

HA (>10kDA <120kDa) which elutes before the fractions were collected or even

smaller material (<10kDa) that disappears through the membrane during

focusing. Another explanation might be that the HA sticks to the membrane and

does not elute with the current AsFlFFF method.

R.J. VONK

28

.

Figure 10: For a), b) and c) PBS was used as mobile phase, 50μL injection of 1mg/mL BSA and HA with a detector flow 0.6mL/min; a) Elution time of BSA and HA in relation to the crossflow; b) calculation of MW of BSA and HA, based on MALS signal 90°; c) ΔDiHA

levels in collected AsFlFFF-fractions using HPLC-FLR. Concentration ΔDiHA (μg/mL) (ave±stdev, n=2) on the left Y-axis, the DRI signal (mV) was plotted on the right Y-axis

B

BSA

HA

C

A

R.J. VONK

29

5.1.3 Enzymatic cleavage

Enzymatic cleavage of HA polysaccharide was done by chondroitinase ABC and

hyaluronidase. These enzymes cleave the β 1-4 bond between the disaccharide

units of HA [06,44]. The conditions, for enzymatic cleavage, vary in previous

performed studies and have to optimized for the best result [06,09,35-36,44].

Chondroitinase ABC is activated after addition of NaAce. Chondroitinase ABC

degraded the most HA in a 50mM NaAce solution at pH 6.8 [44]. In a primary

experiment only chondroitinase ABC was used. In this experiment a cleavage

yield less than 50% was obtained. Because the expected levels of HA in serum

were in the range of 10 to 300ng/mL [07,15,25] a 50% loss of the HA

polysaccharide due to low enzymatic cleavage yield was not desirable. Therefore,

a combination of both chondroitinase ABC and hyaluronidase was investigated.

The solutions used as matrix for enzymatic cleavage were NaAce at pH 6.8, PBS

and PB at pH 7.4. PBS and PB were used as buffer for enzymatic cleavage

because the AsFlFFF fractions contained either PBS or PB. Lyophilizing the

AsFlFFF fractions will increase the salt concentrations. Therefore higher

concentrations of PB(S) buffers were used for observation of the enzymatic yield.

A 6 times and 15 times higher concentrated PB(S) were used, named PB(S)-63

and PB(S)-153, respectively. PBS-6 matrix was used for quantification of HA in

the collected AsFlFFF fractions as described in chapter 5.1.2.

For determination of the optimal enzymatic conditions a 0.5, 5, and 100μg/mL HA

polysaccharide solutions (weighted, not corrected for purity) were incubated. To

each sample (100μL) either 1mU or 10mU chondroitinase ABC and 10U of

hyaluronidase were added. Next the samples were incubated at 37°C up to 48h.

In Figure 11, Figure 12, and Figure 13 an overview is given of the different

enzymatic conditions. In Figure 11, the enzymatic yield of 1mU chondroitinase

ABC at a concentration of 5µg/mL HA was investigated in relation to increasing

salt concentrations of PB(S)-1, -6, and -15. The comparison of adding 1mU or

10mU chondroitinase ABC to both PBS-6 and NaAce is shown in Figure 12. In

Figure 13, the enzymatic yield in PBS-6 matrix with addition of 1mU

chondroitinase is compared at three different HA concentrations. All yields

obtained were corrected for 65% purity of HA.

R.J. VONK

30

When increasing the salt concentration, as shown in Figure 11, a decrease in

enzymatic activity was observed. The high salt concentrations of PB(S)-15,

triangular marks in Figure 11, caused that no enzymatic activity was observed.

The addition of salt to the matrix, shown in Figure 12, caused a decrease in

enzymatic activity ~50% when 1mU of chondroitinase ABC was added. After

adding 10mU of chondroitinase ABC a decrease in enzymatic yield of ~40% was

observed when comparing NaAce to PBS-6 matrix.

Adding 10 times more chondroitinase ABC gained ~20% extra yield in both PBS-

6 and NaAce matrix. Drawback of the high chondroitinase ABC concentration

was that ∆DiHA signal was more decreased after 48h incubation compared to

Figure 11: Optimization enzymatic cleavage in relation to salt concentration, expressed as % ΔDiHA levels measured

compared to levels expected in the samples. A) 100µg/mL HA incubated with 1mU chondroitinase ABC in (concentrated) PBS buffers; B) 100µg/mL HA incubated with 1mU chondroitinase ABC in (concentrated) PB buffers. % ΔDiHA formed

was shown as ave±stdev (n=2)

Figure 12: Optimization enzymatic cleavage in relation to addition of 10 times higher chondroitinase ABC

concentration to 5µg/mL HA in both PBS-6 and NaAce matrix. % ΔDiHA formed was shown as ave±stdev (n=2)

Figure 13: Optimization enzymatic cleavage, comparison of obtained yield at different HA concentrations. PBS-6

matrix was used and 1mU chondroitinase ABC was added. %ΔDiHA formed was shown as ave±stdev (n=2)

R.J. VONK

31

24h incubation. The decrease in ∆DiHA might have been caused by excessive

cleavage of HA polysaccacharide into e.g. monosaccharide units. When

comparing the enzymatic yield between the different concentrations of HA no

difference could be observed after 24h incubation, as shown in Figure 13.

Furture AsFlFFF-fractions will be lyophilized and reconstituted in 6 times less

volume of 50mM NaAce containing 10mU of chondroitinase ABC and 10U

hyaluronidase. The enzymatic yield for this condition was about 47% (PBS-6 –

10mU).

5.1.4 Derivatization with AMAC

Parameters optimized for fluorescent labeling of the ΔDi were the used AMAC

concentration, derivatization time, and concentrating the samples by

lyophilization.

The reaction of AMAC with ΔDiHA is described by Jackson [22]. The fluorescent

label AMAC was dissolved in 85% DMSO/15% acetic acid. NaCNBH3 was added

to the samples to make the reaction irreversible. The volumes of DMSO/water

(50/50 (v/v%)) vary between the articles [06,21,35]. Non-derivatized AMAC

disturbed the chromatography (data not shown). Since excess of AMAC was not

used for derivatization the concentration of AMAC was reduced for a better peak

shape of the ΔDiGAGs. In Figure 14 the response and linearity of a 0.1M and

0.05M AMAC solution are shown, spheres and cubes are used respectively.

Figure 14: ΔDiHA calibration curve on HPLC-FLR for optimization AMAC concentration used for fluorescent labeling of ΔDi

R.J. VONK

32

The described derivatization time and temperature of the AMAC with the ΔDi

ranges from 2h to 4h at 45°C [21,35] to 16h at 37°C [21]. For the enzymatic

reactions a temperature of 37°C was required, in order to be most flexible the

derivatization time at 37°C was investigated. Two experimental conditions were

investigated. First, the samples were left at 37°C for 2h and then repeatedly

analyzed for 100h at 20°C. Secondly, the derivatization was performed at 37°C

for 16h followed by analyzed over 80h at 20°C. The two experiments: “37°C for

2h” and “37°C for 16h” are the diamond and triangular marks shown in Figure 15.

All data points shown in Figure 15 were averaged values of three individual

measurements and concentration of ΔDiHA used was 100ng/mL. The calculated

levels were normalized to the ΔDiHA level after 43h from the ”37°C for 2h”.

For concentrating the derivatized samples a lyophilization step was added after

the derivatization. After incubation at 37°C for 16h the samples were lyophilized

and reconstituted in 3 times less volumes of water or DSMO/water (50/50 (v/v%)).

Figure 15: Optimization of derivatization time of AMAC fluorescent label with ΔDiHA. Control level was 43h of the ”37°C for 2h”. Each point were averaged of three individual runs

Figure 14 shows that the response of the 0.1M AMAC (spheres) was twice as

high compared to the 0.05M AMAC (cubes). Despite the higher response the

0.05M AMAC concentration gained a better peak shape and less background

signal which resulted in lower ΔDiHA concentrations observable. Derivatization of

R.J. VONK

33

the ΔDi was a flexible and time consuming process as visualized in Figure 15.

For this study 10% fluctuation in the observed signal was allowed and therefore

the derivatization should be performed either at 37°C and left there for 2h before

adding DSMO/water and additionally wait for 48h before analyzing, or left at 37°C

for 16h before adding DMSO/water and wait for 24h before measurement.

Higher DMSO concentration in concentrated samples caused peak broadening.

The peak shape was not influenced when the samples were reconstituted in

water (data not shown). Increase in limits of decision was not observed since the

background signal increased as well. Since lyophilization after derivatization did

not gained a better limit of decision this step was not incorporated to the sample

preparation.

5.1.5 RP-HPLC method

To achieve lower detection limit the C18 column with a 4.6mm I.D. used in

previous studies [06,35] was replaced for an Ascentis Express C18, 2.1x150mm,

2.7μm column. The used Fused-Core™ particle technology of this column, could

provide both high speed and high efficiencies of sub-2 μm particles while

maintaining lower backpressures [45]. The gradient and column flow needed to

be adapted because the I.D. of the column was decreased compared to previous

studies [06,35]. The HPLC method details were given in Appendix B.

Baseline separation on the used HPLC method has been confirmed for ΔDiHA,

ΔDi0s, ΔDi4s, ΔDi4,6dis and ΔDi2,4,6tris. Separation of the different GAGs is

shown in Figure 16, concentration of all GAGs was 1000ng/mL dissolved in

water. The obtained calibration curves are visualized in Figure 17. The injected

levels were 0, 5, 10, 20, 50, 100, and 1000ng/mL. The observed peak at 19.5

minutes in Figure 16 was related to AMAC, as described by Deakin et al. [20].

The major AMAC peak of the non derivatized AMAC elutes at >25 min.

Limit of decision5 of ΔDiHA with the current method was 20ng/mL.

5 Limit of decision was the concentration which gives signal that was 3x stdev of blank run (n=4) higher than the average signal of blank run (n=4), assuming the blank peak was present in all observed signals (personal communication with Dr. W.Th. Kok).

R.J. VONK

34

Figure 16: Separation of different AMAC labeled ΔDiGAGs with the used HPLC method.

Concentration of all compounds was 1000ng/mL

Figure 17: Calibration curves of the AMAC labeled ΔDiGAGs injected on HPLC-FLR, injection

volume was 5µL from the concentrations of 0, 5, 10, 20, 50, 100, and 1000ng/mL

ΔDi6s

ΔDi2,4,6tris ΔDi4,6dis

ΔDi4s ΔDiHA

ΔDi0s

AMAC

R.J. VONK

35

5.1.6 Pyrolysis -GC/MS

Optimizing the incubation temperature with TMAH and optimizing the pyrolysis

temperature was examined. Incubation temperatures used were from 50 to

125°C, with a step size of 25°C. Pyrolysis temperatures examined were from 225

to 300 (step size of 25°C) and from 300 to 500°C (step size 50°C). More details

of the GC method are depicted in Appendix C.

The expected trend in the formation of products with neither the

monosaccharides nor the disaccharide was obtained. Many products and

fragments were formed and no data interpretation could be performed because

no reaction mechanism could be made for any of the formed products or

fragments. During pyrolysis no unique fragments of the products of HA compared

to other GAGs were formed. The fragments that were formed did not have a

unique part of the structure present in the GlcNAc part of HA.

5.2 Sample preparation overview after optimizing method

To quantify HA in human blood serum the following sample preparation steps

were applied:

Deproteination of serum samples by 10 times dilution in 50mM TRIS

buffer (~pH7), adding 600mU proteinase K and incubate for 48h at 37°C

Inject 50μL sample in the AsFlFFF for size-based separation, using PBS4

as mobile phase and collect the fractions

Lyophilize the AsFlFFF fractions and reconstitute in 100μL of 50mM

NaAce solution (pH 6.8) containing 0.02% BSA

Enzymatic cleavage of HA polysaccharide to disaccharides using

chondroitinase ABC (10mU) and hyaluronidase (10U) for 24h at 37°C

Derivatization of disaccharide with 10μL AMAC (0.05M) and 10μL

NaCNBH3 (1M) and after 16h at 37°C add 60μL DMSO/water (50/50 v/v%)

Centrifuge 24h after adding DMSO/water at 11.000rpm for 5 min

Analyze supernatant on RP-HPLC method with 5µL injection volume using

λexcitation = 428nm and λemission = 525nm

R.J. VONK

36

5.3 Analysis of serum samples

This method has been used for size-characterization of HA in serum samples. HA

has been spiked to serum samples and compared to the levels in non-spiked

serum samples. The serum samples were prepared via the sample preparation

scheme given in chapter 5.2 but no HA could be measured in any of the collected

serum samples. Possibly due to cleavage of HA by proteinase K which causes

HA to be lost when injected at AsFlFFF. Also the low AsFlFFF recovery might

contribute to the lack of HA signal observed.

R.J. VONK

37

6 Conclusions

6.1 Current method

The method described in this thesis can be used for size-characterization of HA

in human serum samples. The separation of BSA and HA on the AsFlFFF

channel by has been achieved. The MW of the used HA standard was in the

range of 0.12 to 3.2MDa and was separated from BSA (MW=66kDA). The

obtained separation between BSA and HA was sufficient for the expected MW of

HA in human serum samples of (~0.1-10MDa) [05].

Separation of different AMAC labeled ΔDi of the GAGs was performed on the

HPLC-FLR. The observed limit of decision for ΔDiHA of 20ng/mL was expected

to be low enough for quantification of HA in human blood serum.

6.2 Recommendation for future work

For the size-characterization and quantification of HA in human serum, and later

all the GAGs, some additional experiments have to be performed. Some

recommendations are listed below.

After deproteination of the serum samples with proteinase K still 15% of the

proteins were left. Other enzymes such as trypsin [46] or pronase [47] might be

more sufficient for deproteination of serum samples. Ambrosius et al. [35] added

10% TCA for deproteination of serum samples which causes proteins to

precipitate. When the 15% signal was caused by BSA left, addition of TCA might

be the solution after 48h incubation with the enzymes.

The low AsFlFFF-recovery can be investigated by two additional experiments.

First, proteinase K might cleave HA polysaccharides. An experiment to check this

possible HA cleavages is recommended. Two serum sample could be injected

into AsFlFFF without deproteination by proteinase K. The first one will be spiked

with HA polysaccharide, the second will be blank serum. The AsFlFFF-fractions

which do not contain proteins, so no UV response at 280nm, will be prepared for

measurement on HPLC-FLR. When proteins are present in the collected fractions

derivatization of HA will be difficult. The spiked HA serum samples, corrected for

HA levels present in the blank serum samples, should give a similar size-

R.J. VONK

38

distribution as have been shown in Figure 10. If proteinase K cleaves HA

polysaccharides a higher HA recovery will be obtained with the described

experiment. Second explanation for the low AsFlFFF recovery was that low MW

material (<120kDa) might be present in the HA standard. This material can elute

with BSA (>10kDa<120kDa) or will be lost during focusing (<10kDa). Quantifying

the HA levels in all fractions and in the cross flow liquid can give an idea about

the distribution of HA standard during AsFlFFF.

The enzymatic cleavage yield of HA polysaccharide in the lyophilized AsFlFFF-

fractions (PBS-6) was only ~20% for 1mU of chondroitinase. This yield was ~3

times lower compared to non-lyophilized AsFlFFF-fractions (PBS-1), shown in

Figure 11. A higher HA conversion could be obtained by enzymatic cleavage in

non-lyophilized AsFlFFF fractions and lyophilize after enzymatic cleavage but

before adding AMAC to the samples.

Enzymatic cleavage of the polysaccharides was optimized for HA. The optimal

enzymatic conditions for other GAGs might be different. For CS polysaccharide

cleavage by chondroitinase ABC in a buffer at pH 8.0 is recommended while for

HA pH 6.8 gives the highest yield [44]. In previous performed studies

hyaluronidase was used for cleavage of GAGs at pH 5-6 [36, 48].

Different enzymes have different primary substrates. The chondroitinase ABC

“cocktail” used was not specific for cleavage of HA or the other GAGs. ΔDi of the

GAGs cleaved by chondroitinase ABC and hyaluronidase, were mentioned in

Table 2. More specific enzymatic cleavage could performed by chondroitinase

AC, chondroitinase B, or by chondroitinase C [10,36]. Apart from more specific

cleavage these enzymes might also give higher cleavage yields for the

polysaccharide. Other enzymes, such as heparinase, keratanase and keratanase

II can be used for cleavage of HS [04,10,36] and KS [49] polysaccharides.

With the AsFlFFF method separation of BSA (MW=66kDa) and HA standard

(MW=0.12 to 3.2MDa) was possible. Other GAGs, with common MW of 15-

20kDa [05,07], will be separated from HA but not from BSA. Present proteins

after deprotienation could be separated from the other GAGs by changing the

cross flow program. With a higher Fcr/Fch-value BSA will be retained and other

GAGs will elute before BSA.

R.J. VONK

39

Table 2: ΔDiGAGs names, backbone description, and used abbreviations

name Disaccharide backbone Abbreviation

hyaluronan GlcA- β1,3-GlcNAc ΔDiHA

chondroitin GlcA-β1,3-GalNAc ΔDi0s

chondroitin 2-sulfate GlcA(2s)-β1,3-GalNAc ΔDi2s

dermatan 4-sulfate IdoA-α1,3-GalNAc(4s) 6 ΔDi4s

chondroitin 4-disulfate GlcA-β1,3-GalNAc(4s) 6 ΔDi4s

chondroitin 6-disulfate GlcA-β1,3-GalNAc(6s) ΔDi6s

chondroitin 2,4-disulfate GlcA(2s)-β1,3-GalNAc(4s) ΔDi2,4dis

chondroitin 2,6-disulfate GlcA(2s)-β1,3-GalNAc(6s) ΔDi2,6dis

chondroitin 4,6-disulfate GlcA-β1,3-GalNAc(4s,6s) ΔDi4,6dis

chondroitin 2,4,6-trisulfate GlcA(2s)-β1,3-GalNAc(4s,6s) ΔDi2,4,6tris

The used RP-HPLC method obtained baseline separation between ΔDiHA,

ΔDi0s, ΔDi4s, ΔDi4,6dis and ΔDi2,4,6tris. The elution times observed with the

current standard compounds suggest that this method was also suitable for other

sulphated ΔDi from CS/DS. Although, note that AMAC related peak at 19,5 min in

Figure 17 and ΔDi2s might elute at the same time using the current method. No

ΔDi2s standard was available so separation between the AMAC related peak and

ΔDi2s has not been verified. Deakin et al. [20] described that they clean up the

AMAC before derivatization reactions with ΔDi. Cleaning AMAC before

derivatization of disaccharides might be the solution to get rid of the observed

AMAC related peak eluting in the same time as the ΔDiGAGs. For obtaining

lower limits of detection the peak shape of ΔDi2,4,6tris which is non symmetric as

can be seen in Figure 16, might be improved. Structural conformations could be

performed by other detectors such as MS.

The Py-GC/MS based method was tried as an alternative for the HPLC-FLR

method to simplify the sample preparation. The problem at this point was that

nothing can be said from formed products and fragments. Additional time needs

to be spend on data interpretation and elucidating the pyrolysis reactions.

6 Enzymatic cleavage of dermatan 4-sulfate and chondroitin 4-sulfate causes the loss of stereo chemical difference present in the polysaccharide, as been shown in Figure 3. They both form the same disaccharide unit (ΔDi4s) [06]

R.J. VONK

40

7 Literature

[01] Broekhuizen L.N., et al., Endothelial glycocalyx as potential diagnostic and therapeutic

target in cardiovascular disease, Current Opinion in Lipidology, 20 (2009) 57-62

[02] Reitsma S., et al., Pflügers Archiv - European Journal of Physiology, 454 (2007) 345-

359

[03] Balazs E.A., Laurent T.C., and Jeanloz R.W., Nomenclature of hyaluronic acid,

Biochemical Journal, 235 (1986) 903

[04] Taylor K.R., and Gallo R.L., Glycosaminoglycans and their proteoglycans: host-

associated molecular patterns for initiation and modulation of inflammation, The FASEB

Journal, 20 (2006) 9-22

[05] Fraser J.R.E., Laurent T.C., and Laurent U.B.G., Hyaluronan: its nature, distribution,

functions and turnover, Journal of Internal Medicine, 242 (1997) 27-33

[06] Volpi N., High-performance liquid chromatography and on-line mass spectrometry

detection for the analysis of chondroitin sulfates/hyaluronan disaccharides derivatized

with 2-aminoacridone, Analytical Biochemistry, 397 (2010) 12-23

[07] Laurent T. C., and Fraser J. R. E., Catabolism of hyaluronan, In Degradation of Bio-

active Substances: Physiology and Pathophysiology (Henriksen, J. H., ed) (1991) 249-

265, CRC Press, Boca Raton, Florida

[08] Dahl L.B., et al., Concentration and molecular weight of sodium hyaluronate in synovial

fluid from patients with rheumatoid arthritis and other arthropathies, Annals of the

Rheumatic Diseases, 44 (1985) 817-822

[09] Lamari F.N., et al., Analysis of glycosaminoglycan-derived disaccharides in illogic

samples by capillary electrophoresis and protocol for sequencing glycosaminoglycans,

Biomedical Chromatography, 16 (2002) 95-102

[10] Sasisekharan R., Raman R., and Prabhakar V., Glycomisa approach to structure-

function relationships of glycosaminoglycans, Annual Review of Biomedical Engineering,

8 (2006) 181-231

[11] Esko J.D., and Selleck S.B., Order out of chaos: Assembly of ligand binding sites in

Heparan Sulfate, Annual Review of Biomedical Engineering, 718 (2002) 435-471

[12] Nieuwdorp M., et al., Perturbation of hyaluronan metabolism predisposes patient with

type 1 diabets mellitus to artherosclerosis, Diabetologia, 50 (2007) 1288-1293

[13] Faser J.R.E., and Laurent T.C., Turnover and metabolism of hyaluronan, In The biology

of Hyaluronan, Ciba Foundation Symposium, 143 (1989) 41-59, Wiley, Chichester,

England.

[14] Nurminen M., et al., Clinical Utility of liquid-chormatographic analysis of effusions for

hyaluronate content, General Clinical Chemistry, 40(5) (1994) 777-780

[15] Tengblad A., et al., Concentraion and relative molecular mass of hyaluronate in lymph

and blood, Biochemical Journal, 236 (1986) 521-525

R.J. VONK

41

[16] Lamari F., et al., Profiling of the eye aqueous humor in exfoliation syndrome by high-

performance liquid chromatographic analysis of hyaluronan and galactosaminoglycans,

Journal of Chromatography B, 709 (1998) 173–178

[17] Laurent U.B.G., and Reed R.K., Turnover of hyaluronan in the tissues, Advanced Drug

Delivery Reviews, 7 (1991) 237-256

[18] Karamanos N.K., and Hjerpe A., High-performance capillary electrophoretic analysis of

hyaluronan in effusions from human malignant mesothelioma, Journal of

Chromatography B, 697 (1997) 277–281

[19] Theocharis A.D., et al., human abdominal aortic aneurysm is closely associated with

compositional and specific structural modifications at the glycosaminoglycan level,

Atherosclerosis, 145(2) (1999) 359–368

[20] Deakin J.A., and Lyon M., A simplified and sensitive fluorescent method for disaccharide

analysis of both heparin sulfate and chondroitin/dermatan sulfates from biological

samples, Glycobiology, 18(6) (2008) 483-491

[21] Kitagawa H., Kinoshita A., and Sugahara K., Microanalysis of glycosaminoglycan-

derived disaccharides labeled with the fluorophore 2-aminoacridone by capillary

electrophoresis and high-performance liquid chromatography, Analytical Biochemistry,

232 (1995) 114-121

[22] Jackson P., Polyacrylamide gel electrophoresis of reducing saccharides labeled with the

fluorophore 2-aminoacridone: subpicomolar detection using an imaging system based

on a cooled charge-coupled device, Analytical Biochemistry, 196 (1991) 238-244

[23] Zamfir A.D., et al., On-line sheathless capillary electrophoresis/ nanoelectrospray

ionization-tandem mass spectrometry for the analysis of glyocsaminoglycan

oligosaccharides, Electrophoresis, 25 (2004) 2010-2016

[24] Lamari F.N., et al., Ultrasensitive capillary electrophoresis of sulfated disaccharides in

chondroitin/dermatan sulfates by laser-induced fluorescence after derivatization with 2-

aminoacridone, Journal of Chromatography B, 730 (1999) 129-133

[25] Laurent U.B.G., and Laurent T.C., On the origin of hyaluronate in blood, Biochemistry

International, 2(2) (1981) 195-199

[26] Moon M.H., et al., Flow field-flow fractionation/multiangle light scattering of sodium

hyaluronate from various degradation processes, Journal of Chromatography B, 864

(2008) 15-21

[27] Shin D.Y., et al., Molecular weight and structure characterization of sodium hyaluronate

and its gamma radiation degradation products by flow field-flow fractionation and on-line

multiangle light scattering, Journal of Chromatography A, 1160 (2007) 270-275

[28] Lee H., Cho I.H., and Moon M.H., Effect of dissolution temperature on the structures of

sodium hyaluronate by flow field-flow fractionation/multiangle light scattering, Journal of

Chromatography A, 1131 (2006) 185-191

R.J. VONK

42

[29] Orvisky E., et al., The Determination of Hyaluronan Molecular Weight Distribution by

Means of High- Performance Size Exclusion Chromatography, Journal of liquid

Chromatography, 15(18) (1992) 3203-3218

[30] Tømmeraas K., and Wahlund P.O., Poly-acid properties of biosynthetic hyaluronan

studied by titration, Carbohydrate polymers, 77 (2009) 194-200

[31] Chaidedgumjorn A., et al., Conductivity detection for molecular mass estimation of per-

O-sulfonated glycosaminoglycans separated by high-performance size-exclusion

chromatography, Journal of Chromatography A, 959 (2002) 95-102

[32] Technical details of the AsFlFFF apparatus provided by the manufacturer, website visited

on 1st October 2010; http://www.wyatt.eu/index.php?id=field_flow_fractionation

[33] Giddings J.C., Field-flow fractionation: Analysis of macromolecular, colloidal, and

particulate materials, Science, 260 (1993) 1456-1465

[34] Moon M.H., and Williams K.R., Course material from short course SCM-5, Field-Flow

Fractionation, January 2011

[35] Ambrosius M., Kleesiek K., and Götting C., Quantitative determination of the

glycosaminoglycan Δ-disaccharide composition of serum, platelets and granulocytes by

reversed-phase high-performance liquid chromatography, Journal of Chromatography A,

1201 (2008) 54-60

[36] Volpi N., and Linhardt R.J., High-performance liquid chromatography-mass spectrometry

for mapping and sequencing glycosaminoglycan-derived oligosaccharides, Nature

Protocols, 5 (2010) 993-1004

[37] Alvarez-Perez F.J., Castelo-Branco M., and Alvarez-Sabin J., Albumin level and stroke.

Potential association between lower albumin level and cardioembolic aetiology,

International Journal of Neuroscience, 121 (2011) 25-32

[38] Wissenschaftliche Tabellen Geigy, CIBA-GEIGY, Basel Switzerland (1976) 121-123

[39] Hilz H., Wiegers U., and Adamietz P., Stimulation of proteinase K action by denaturing

agents: application to the isolation of nucleic acids and the degradation of ‘masked’

proteins, European Journal of Biochemistry, 56 (1975) 103-108

[40] Bradford M.M., A rapid and sensitive method for the quantification of microgram

quantities of protein utilizing the principle of protein-dye binding, Analytical Biochemistry,

72 (1976) 248-254

[41] Product details at the website of the provider, visited on 10th Januari 2011;

http://www.sigmaaldrich.com/catalog/ProductDetail.do?lang=en&N4=P2308|SIAL&N5=S

EARCH_CONCAT_PNO|BRAND_KEY&F=SPEC

[42] Moon M.H., Effect of carrier solutions on particle retention in flow field-flow fractionation,

Bulletin of the Korean Chemical Society, 16(7) (1995) 613-619

[43] Moon M.H., Park I., and Kim Y., Size characterization of liposomes by flow filed-flow

fractionation and photon correlation spectroscopy effect of ionic strength and pH of

carrier solutions, Journal of Chromatography A, 813 (1998) 91-100

R.J. VONK

43

[44] Yamagata T., et al., Purification and properties of bacterial chondroitinases and

chondrosulfatases, The Journal of Biological Chemistry, 243 (1968) 1523-1535

[45] Product details at the website of the provider, visited on 10th February 2011;

http://www.sigmaaldrich.com/catalog/ProductDetail.do?lang=en&N4=53825U|SUPELCO

&N5=SEARCH_CONCAT_PNO|BRAND_KEY&F=SPEC

[46] Mota M.V.T., et al., Trypsin hydrolysis of whey protein concentrates: Characterization

using multivariate data analysis, Food Chemsitry, 94(2) (2006) 278-286

[47] Sweeney P.J., and Walker J.M., Pronase, Methods in Molecular Biology 16 (1993) 271-

276

[48] Gherezghiher T., et al., Rapid and sensitive method for measurement of hyaluronic acid

and isomeric chondroitin sulfates using high-performance liquid chromatography,

Journal of Chromatography, 413 (1987) 9-15

[49] Plaas A.H.K., West L.A., and Midura R.J., Keratan sulfate disaccharide composition

determination by FACE analysis of keratanase II and endo-β-galactosidase digestion

products, Glycobiology, 11(10) (2001) 779-790

[50] Hokputsa S., et al., A comparison of molecular mass determination of hyaluronic acid

using SEC/MALS and sedimentation equilibrium, European Biophysics Journal, 32

(2003) 450-456

R.J. VONK

44

Appendix A: AsFlFFF method

Instrumental details of AsFlFFF method used for mobile phases of water, 10mM

NaNO3 and PBS.

mobile phase : PBS4 buffer detector flow : 0.6mL/min injection flow : 0.2mL/min channel membrane : 10kDa cut-off channel dimensions : channel space was made by cutting a 350-μm-thick

Mylar spacer with a tip-to-tip length of 13.5cm. The channel had a trapezoidal shape (the initial breadth decreased from 21.5mm to 3mm), and the geometrical volume of the channel was 0.56cm3.

injection volume : 50μL wavelength UV : 250nm sample temperature : 5°C total runtime : 30 minutes RIcalibration constant : 1.35x10-5

DAWNconstant : 2.20x10-5

Refractive OH2index : 1.3364

Refractive PBSindex : 1.3400

dn/dc HA in PBS : 0.167mL/g [49]

Table 3: AsFlFFF flow program for MW calibration of BSA and HA

time (min)

cross flow start (mL/min)

cross flow end (mL/min)

focus flow (mL/min)

mode

0 - - 1,8 Focus + injection 3 1,8 1,8 - Elution 8 1,8 0,66 - Elution

11 0,66 0,24 - Elution 14 0,24 0,08 - Elution 17 0,08 0,03 - Elution 20 0,03 0,02 - Elution 30 0,02 0,02 - Elution

R.J. VONK

45

Appendix B: RP-HPLC-FLR method

Instrumental details of HPLC-FLR method used for quantification ΔDiHA. mobile phase A : 60mM NH4Ace at pH 5.6 mobile phase B : ACN sample temperature : RT injection volume : 5μL 7 column : Ascentis Express, C18, 2.1x150mm, 2.7μm column temperature : 30°C column flow : 0.2mL/min used gradient : time (min) % A % B 0 98 2 32 80 20 35 10 90 37 10 90 38 98 2 45 98 2 λexcitation : 428nm λemission : 525nm run time : 45 minutes

7 Sample was prepared by taking 100μL sample, add 10µL AMAC (0.05M) and 10µL

NaCNBH3 (1M). After incubation at 37°C add 60μL DMSO/H2O.

R.J. VONK

46

Appendix C: Pyrolysis-GC/MS method

Instrumental details of Py-GC/MS method used for quantification HA. carrier gas : He injection volume sample : 15µL TMAH concentration : 2.5% in water injection volume TMAH : 20µL injection temperature : 40°C injection program : time (min) temp (°C) event 0 - 1 100 solvent evaporation 1 - 2 40 TMAH injected 2 – 3,5 vary *1 incubation with TMAH 3,5 - end vary *2 pyrolysis reactions occur *1 incubation temperatures : 50 – 125°C, step size 25°C *2 pyrolysis temperatures : 225 – 300°C, step size 25°C : 300 – 500°C, step size 50°C column : 30m x 0.25mm I.D. TC 5MS column (5% phenyl-

methylpolysiloxane) with a film thickness of 0.25µm (GL science)

column flow : 1mL/min column temperature : time (min) temp (°C) ramp (°C/min) 0 – 8 60 8 – 25,3 320 15 25,3 – 27,3 320 MS detection : full scan mode range 50-600 amu.

R.J. VONK

47

Appendix D: Major project proposal

Major Project/ Research Thesis proposal Rudy Vonk (5897041)

October 2010 to March 2011

University of Amsterdam

Analytical Chemistry department

Supervisor: Dr. W.Th. Kok

Co-supervisor: R.N. Qureshi MSc

Develop a method for the size-characterization of

glycosaminoglycans (GAGs) in human blood serum

The glycocalyx forms a gel-like layer covering the endothelial lining, shielding

endothelial cells from direct contact with circulating blood cells. The glycocalyx-

related functions might be a diagnostic tool or even a therapeutic target for

preventive strategies in clinical practice [01]. Glycocalyx consist of a number of

glycoproteins and proteoglycans. The core proteoglycans are firmly connected to

the endothelial cell membrane and soluble proteoglycans. These proteoglycans

are the backbone for various glycosaminoglycans (GAGs) [01].

GAGs consist of repeating units of a disaccharide unit. Based on structure there

are four groups of GAGs namely (1) hyaluronic acid/hyaluronan [03] (HA), (2)

chondroitin sulfate (CS) and dermatan sulfate (DS), (3) heparin sulfate (HS) and

heparin (H), (4) keratin sulfate (KS) [35]. The differences between the four groups

are primary the building blocks of the disaccharides and the number of sulfate

groups. The property of the GAGs is divers. HA is crucial for structure and the

maintenance of the entangled network providing stability to the glycocalyx [01].

Due to shear stresses GAGs are synthesized in the blood vessel and damage the

artery which might lead to cardiovascular diseases. The size of the synthesized

GAGs ranges from 5 kDa to ~20 MDa.

In disease states HA is one of the first molecules to be shed from the glycocalyx

to exert several biological functions. Damaging the glycocalyx with hyaluronidase

R.J. VONK

48

has been associated with increased vascular vulnerability towards atherogenic

insults.

Looking at the sizes of HA, low-molecular-size polymers appear to function as

endogenous “danger signals”, while even smaller fragments can ameliorate these

effects. In pathologic situations, such as in shock, septicemia, massive trauma,

post surgery, blood loss and in burn patients, circulating high-molecular-mass HA

increases.

In the literature Volpi describes a method for the quantification of GAGs in human

plasma using mass spectrometry as detection method [06]. This method includes

enzymatic degradation of GAGs from plasma samples but does not include the

molecular-size determination of the GAGs in relation to the disease state.

Goal is to develop a method which is suitable for size-characterization of the

GAGs in human serum.

This project consists of, but might be changed based on rolling apprehension, the

following steps;

(1) The deproteination of serum samples, extractions of GAGs,

(2) Asymmetric Flow Field-Flow Fractionation (AsFlFFF) size-based

separation the GAGs into sample fragments

(3) Measure GAGs in the fragments of AsFlFFF by enzymatic breakdown

polysaccharides into disaccharide units, derivatization with fluorescence label

and measure on a HPLC-fluorescence detector.

(4) Measure the GAGs in the fragments of AsFlFFF on a pyrolysis GCMS.

Our aim is to measure GAGs using HPLC combined with fluorescence as a

detection method. Step 4 is opted in order to confirm structural information of the

AsFlFFF fragments and if pyrolysis GCMS is suitable it would be a fast and new

method for quantification of GAGs.

This research project focuses on analyzing the vast range of molecular weight of

HA polymers to show patterns that would provide insight into their production and

regulation of HA in different disease states.

R.J. VONK

49

Acknowledgements / Dankwoord

Een dankwoord schrijven is net als een Sinterklaas gedicht, het begint altijd met dezelfde zin… Dat vind ik niet erg origineel maar ja het hoort erbij. Ik denk niet dat ik aan één A4-tje genoeg heb om iedereen te bedanken die mij geholpen heeft bij het onderzoek de afgelopen 7 maanden, met het schrijven van de thesis en natuurlijk het volgen van de studie. Maar ik ga het toch proberen…. Als eerste wil ik Arnoud, inmiddels mijn voormalig leidinggevende, bedanken voor het mogelijk maken van het volgen van de Master studie naast mijn gewone werk. Ook toen het onweerde in Weesp heb ik van jou de mogelijkheid gehad om mijn studie af te ronden, bedankt voor je inzet daarin! Ook Niels en Sigrid, collega’s van de BM-groep, bedankt dat jullie het werk overgenomen hebben zodat ik mij volledig kon focussen op het research project. Sigrid verder wil ik je ook bedankt voor je kritische blik op mijn thesis en je tips en tricks om het geheel zo overzichtelijk mogelijk op papier te krijgen. Next I would like to thank you Rashid for your supervising during my research project. Your view on the project was a good guidance during the past 7 months. Also your notes and remarks during building this thesis were helpful. Ook wil ik graag Wim bedanken voor de begeleiding van het project en de verhelderende blik toen ik vast zat tijdens het onderzoek. Zonder je hulp bij het uitvoeren van de berekeningen waren die niet gelukt. Verder wil ik graag een paar collega’s bij de universiteit bedanken dat ik gebruik heb mogen maken van hun kennis en apparatuur tijdens het project. Winfried, dhr. L. de Jong en Prof. C. de Koster, zonder jullie bijdrage waren sommige stappen niet zo snel gegaan als ze nu zijn gegaan. In het rijtje van bedankjes hoort mijn dochter Isabel zeker thuis. Isabel hoewel je nog niet veel zegt, je bent per slot van rekening pas 10 maanden jong, en alleen maar veel lacht en af en toe ondeugend bent wil ik je bedanken voor je liefde het afgelopen jaar, nou ja eigenlijk pas 10 maanden. Als ik het niet zag zitten of het typen zat was liet jij mij altijd met je knuffelen zodat ik er daarna weer tegen kon! Bedankt lieve meid! Last but certainly not least wil ik graag mijn lieve vrouw Gerdien bedanken voor de steun tijdens de avonden achter de computer gedurende mijn gehele Master studie. Zonder jouw goede zorg en bemoedigende woorden weet ik niet of ik zover was gekomen. Rudy 20 april 2011