+ All Categories
Home > Documents > Dissertation Marion Fischer - Qucosa

Dissertation Marion Fischer - Qucosa

Date post: 01-Jan-2017
Category:
Upload: hatu
View: 219 times
Download: 1 times
Share this document with a friend
111
I Initiation of blood coagulation – Evaluating the relevance of specific surface functionalities using self assembled monolayers D I S S E R T A T I O N zur Erlangung des akademischen Grades Doctor rerum naturalium (Dr. rer. nat.) vorgelegt der Fakultät Mathematik und Naturwissenschaften der Technischen Universität Dresden von Dipl. Ernährungswissenschaftlerin Marion Fischer geboren am 16.10.1979 in Dresden Eingereicht am 05.06.2010 in Dresden. Die Dissertation wurde in der Zeit von 03/2007 bis 05/2010 im Leibniz Institut für Polymerforschung Dresden angefertigt
Transcript
Page 1: Dissertation Marion Fischer - Qucosa

I

Initiation of blood coagulation – Evaluating the relevance of specific surface functionalities using self assembled

monolayers

D I S S E R T A T I O N

zur Erlangung des akademischen Grades

Doctor rerum naturalium (Dr. rer. nat.)

vorgelegt

der Fakultät Mathematik und Naturwissenschaften

der Technischen Universität Dresden

von

Dipl. Ernährungswissenschaftlerin Marion Fischer

geboren am 16.10.1979 in Dresden

Eingereicht am 05.06.2010 in Dresden.

Die Dissertation wurde in der Zeit von 03/2007 bis 05/2010 im Leibniz Institut für Polymerforschung Dresden angefertigt

Page 2: Dissertation Marion Fischer - Qucosa

II

Gutachter: Prof. Dr. Carsten Werner Prof. Dr. Brigitte Voit

Page 3: Dissertation Marion Fischer - Qucosa

Table of contents

I

Table of contents

Acknowledgements ...................................................................................................... III Preface ............................................................................................................................ V 1. Theoretical background ......................................................................................... 1

1.1. Hemocompatibility of medical devices 1

1.2. Self assembled monolayers as model surfaces 1

1.3. Initial processes of coagulation 3 1.3.1 Protein adsorption 4 1.3.2 Activation of coagulation via contact activation (intrinsic pathway) 7 1.3.3 Activation of coagulation via tissue factor (extrinsic pathway) 9 1.3.4 Sources and activity of TF 9 1.3.5 Cellular responses upon material-blood contact focussing on platelet adhesion 11

2. Experimental part ................................................................................................ 13

2.1. Preparation of gold substrates 13

2.2. Preparation and characterisation of self assembled monolayers 14 2.2.1 Preparation and characterisation of C15-COOH/ C15-CH3 14 2.2.2 Preparation and characterisation of C10-COOH/ C10-CH3 15 2.2.3 Preparation and characterisation of C10-COOH/ C11-(O-CH2CH2)3-O-CH3) 17

2.3. Characterisation of protein adsorption and enzyme activation 17

2.3.1 Human fibrinogen/ fibrin 17 2.3.2 Adsorption of complement fragments C3b and release of C5a 19 2.3.3 Contact activation: factor XIIa and kallikrein activity 20 2.3.4 Thrombin 21

2.4. Surface incubation with human blood plasma or platelet rich plasma

(PRP) 22 2.4.1 LDH assay on COOH/CH3 and COOH/CH3/OH 22 2.4.2 Detection of platelets after immunostaining using fluorescence scanner 22

2.5. Whole blood incubation assay 22

2.6. Western blot of leukocyte isolates 26

2.6.1 Optimisation of leukocyte lysis 26 2.6.2 Optimisation of gel-loading conditions 27 2.6.3 Optimisation of leukocyte isolation using Polymorphprep® 27

Page 4: Dissertation Marion Fischer - Qucosa

Table of contents

II

3. Results .................................................................................................................... 28

3.1. Preparation of gold substrates 28

3.2. Preparation and characterisation of self assembled monolayers 30 3.2.1 Preparation and characterisation of C15-COOH/ C15-CH3 30 3.2.2 Preparation and characterisation of C10-COOH/ C10-CH3 33 3.2.3 Preparation and characterisation of C10-COOH; C11-OH 40 3.2.4 Preparation and characterisation of C10-COOH/ C11-(O-CH2CH2)3-O-CH3) 42

3.3. Characterisation of protein adsorption and enzyme activation 46

3.3.1 Human fibrinogen/ fibrin 46 3.3.2 Adsorption of complement fragment C3b 51 3.3.3 Contact activation: factor XIIa and kallikrein activity 52 3.3.4 Thrombin 58

3.4. Surface adhesion of platelets 61

3.4.1 LDH assay on SAMs after incubation with PRP 61 3.4.2 Detection of platelets after immunostaining using fluorescence scanning 62

3.5. Analysis of TF in leukocyte lysates 64

3.5.1 Isolation of leukocytes using ERL-kit 64 3.5.2 Optimisation: using standard-TF and different gel-loading conditions 66 3.5.3 Isolation of leukocytes using Polymorphprep® 67

3.6. Whole blood incubation 68

3.6.1 Whole blood incubation of -CH3/-COOH terminated SAMs 68 3.6.2 Whole blood incubation of -CH3/-COOH and -COOH/-OH terminated SAMs 75

4. Discussion .............................................................................................................. 84 5. Summary and conclusion ..................................................................................... 91 List of abbreviations ..................................................................................................... 93 References...................................................................................................................... 95

Page 5: Dissertation Marion Fischer - Qucosa

Acknowledgements

III

Acknowledgements

I am deeply grateful to my adviser Dr. Claudia Sperling - not only for giving me the

opportunity to perform this thesis work in her lab but also for many, many hours of

great supervision and discussion…also in hard times. Thanks for letting me participate

in the project that you brought into being and that presented an excellent scientific base

for the present thesis. Thanks for your experience both scientifically and personally, for

critical interpretation of results and for finding time to solve problems. Thank you for

everything.

Special thanks to Prof. Dr. Carsten Werner for the opportunity of joining his group. I

appreciate his essential project supervision, scientific discussions and indispensable

motivation for the projects process. Thanks also to Prof. Dr. Brigitte Voit for her

scientific support and for reviewing the thesis.

Many thanks also to Dr. Manfred Maitz, who acted like a co-adviser for my work.

Manfred contributed significantly to this thesis and to my knowledge about lab work in

general. Thanks for your uncounted proof-readings and all the fruitful discussions.

I also want to thank the group of Prof. Pentti Tengvall, which quickly integrated me and

supported me with help, guidance and suggestions. I thank Pentti for his kindness at any

time and for having created a nice working atmosphere as well as for scientific

discussions and proof-reading of our publication.

Lots of thanks also to Grit Eberth and Martina Franke for their considerable support in

lab works, preparing blood incubation assay and numerous gold surfaces.

Thanks to Babette Lanfer, Marina Prewitz, Andrea Zieris, all from the MBC, for lab

support, advice with several techniques, and for being great colleagues and friends.

Thanks for valuable support in difficult moments and for helpful comments and

discussions.

All this work would not have been possible without the constant support of my family,

especially my parents and my friends. Most of all, I would I like to thank my mother for

Page 6: Dissertation Marion Fischer - Qucosa

Acknowledgements

IV

the uncounted hours of childcare and Tim and Paula for helping me through this time

and for providing encouragement.

Thanks also to the DFG for funding my project under grant SP 966 2.

Marion Fischer

April 30. 2010

Page 7: Dissertation Marion Fischer - Qucosa

Preface

V

Preface

The surface of biomaterials can induce contacting blood to coagulate, similar to the

response initiated by injured blood vessels to control blood loss. This poses a challenge

to the use of biomaterials as the resulting coagulation can impair the performance of

hemocompatible devices such as catheters, vascular stents and various extracorporeal

tubings [1], what can moreover cause severe host reactions like embolism and

infarction.

Biomaterial induced coagulation processes limit the therapeutic use of medical

products, what motivates the need for a better understanding of the basic mechanisms

leading to this bio-incompatibility [2] in order to define modification strategies towards

improved biomaterials [3]. Several approaches for the enhancement of hemocompatible

surfaces include passive and active strategies for surface modifications. The materials’

chemical-physical properties like surface chemistry, wettability and polarity are

parameters of passive modification approaches for improved hemocompatibility and are

the focus of the present work.

In the present study self assembled monolayers with different surface functionalities

(-COOH, -OH, -CH3) were applied as well as two-component-layers with varying

fractions of these, as they allow a defined graduation of surface wettability and charge.

The ease of control over these parameters given by these model surfaces enables the

evaluation of the influence of specific surface-properties on biological responses.

To evaluate the effects of different surface chemistry on initial mechanisms of

biomaterial induced coagulation, the surfaces were incubated with protein solution,

human plasma, blood cell fractions or fresh heparinised human whole blood. Indicative

hemocompatibility parameters were subsequently analysed focusing on protein

adsorption, coagulation activation, contact activation (intrinsic/ enhancer pathway),

impact of tissue factor (extrinsic/ activator pathway) and cellular systems (blood

platelets and leukocytes).

Page 8: Dissertation Marion Fischer - Qucosa

1 Theoretical background

1

1. Theoretical background

Fundamental insight into the experimental set up as well as reactions occurring at

blood-material interfaces are described in this chapter. Regarding the experimental set

up the focus is here on the description of the model surfaces that were used as they

represent an important part of the results presented in this work, whereas more detailed

information on the in vitro whole blood incubation set up is given elsewhere [4].

1.1. Hemocompatibility of medical devices

Biomedical devices that implement blood contacting materials include catheters, blood

vessel grafts, vascular stents, artificial heart valves, circulatory support devices, various

extracorporeal tubings, hemodialysis, hemapheresis and oxygenator membranes. The

performance of these medical products is significantly impaired in the case of

coagulation processes [1]. Thus it is necessary to understand processes occurring at

blood-biomaterial interfaces to improve the surface biocompatibility. Current

approaches to assess the hemocompatibility of biomaterials are mainly based on

analytical methods focusing either on cellular or plasmatic events disregarding the

complex interplay of blood components. To meet the requirements of analysing whole

blood as a complex system, whole blood incubation assays are carried out in this study

for testing the degree to which distinct material surface characteristics promote blood

cell adhesion and initiate the coagulation cascade after surface contact.

1.2. Self assembled monolayers as model surfaces

Former research projects often compared materials varying on a wide range of

parameters [1]. Also the experimental set-up often is far away from an in vivo situation.

The use of self-assembled monolayers (SAMs) of alkyl thiols on gold (see Figure 1) [5]

are advantageous to study material induced coagulation since it allows a variety of

model surfaces to be produced with precisely defined characteristics [6, 7].

Page 9: Dissertation Marion Fischer - Qucosa

1 Theoretical background

2

Figure 1 Schematic presentation of self assembled monolayers of alkanethiols on gold. Layers are glass,

chromium (Cr) and gold (Au) coupled to alkanethiols with their specific head groups (marked in red).

Due to their unique structural integrity combined with the unparalleled uniform surface

chemistry they are especially successful in mimicking biosurfaces. Moreover, the ease

of control over surface chemistry, polarity and charge enables the evaluation of the

influence of specific surface-properties on biological reactions. SAMs of thiol

compounds have become an extensively used model system for surface related research

in the recent years [5, 8, 9] and their use in applied science has increased considerably

[10-15]. Previous work using SAM surface chemistry focused on plasma protein

adsorption [11, 16, 17] and cell-surface interactions including leukocyte [12, 18] and

platelet adhesion [16, 19, 20].

For experimental work a set of self-assembled monolayers exposing a variety of

different functional groups and combinations of the latter was applied to explore the

initial coagulation events brought about by blood-material interaction. For one set of

samples methyl- and carboxyl-terminated self assembled monolayers as well as binary

mixtures were used to investigate the pro-coagulant effect of extreme material

properties compared to a binary surface that displays both characteristics, differing thus

from the extremes. In particular, it was tested which surface characteristics promote

platelet adhesion and/or coagulation initiation, and surface characteristics that result in

the strongest activation of coagulation were determined.

For the second set of samples hydroxyl- and carboxyl-terminated SAMs were combined

to study the influence of surface hydroxylation on cell adhesion and on the initiation of

the extrinsic pathway.

Concerning preparation of SAMs for biomaterial research there is still no general

consensus on preparation conditions despite their frequent utilization. Especially SAMs

that consist of COOH-terminated thiols are observed to cause weak ordered layers with

tendencies to form double layers or cyclic structures because of electrostatic repellence

that inhibits the self assembly process [21]. Arnold et al. only found homogeneously

Page 10: Dissertation Marion Fischer - Qucosa

1 Theoretical background

3

well ordered monolayers when using ethanol +2% acetic acid as immersion solvent

[22]. Other authors suggested the use of triethylamine being essential for the formation

of -NH2 terminated SAMs but not for –COOH modified surfaces [20]. Even standard

protocols have been questioned recently e.g. by Chen et al. [23] who investigated the

effect of temperature and thiol concentration on the kinetics of decanethiol SAM

formation. Further on new methods like ultrasonic assisted rapid formation of SAMs

within 15 minutes are being applied [24]. Kim et al. found different diffusion rates of

alkanethiols with different alkyl chain length and showed that full monolayers of 1-

dodecanethiol (-C12-CH3) are formed after 30 minutes [25], which is in contrast to the

standard protocol indicating an immersion time of >16 hours. The establishment and

optimisation of SAM surface preparation is part of the present work, as well as the

characterisation of the model substrates.

1.3. Initial processes of coagulation

Despite the growing need of materials used in medicine as blood contacting devices,

only few fundamental mechanisms of blood material interactions are elucidated by now.

As known, physicochemical properties of biomaterials determine the fate of blood

components like proteins, enzymes and cells and are relevant for biological responses

(see Figure 2). In the following, protein adsorption and cellular reactions but also

plasmatic coagulation events like initiation of the intrinsic or extrinsic pathways are

elucidated.

Page 11: Dissertation Marion Fischer - Qucosa

1 Theoretical background

4

Figure 2 Overview of blood–material interactions showing the components relevant to thrombosis. Taken

from [2].

1.3.1 Protein adsorption

Initial protein interaction with a surface is the first step of a complex biochemical

cascade that occurs upon blood-material contact. The adsorption of plasma proteins is of

great importance for the surfaces biocompatibility as subsequent reactions like cell

adhesion or inflammation processes are influenced. It was in 1988 when Leo Vroman

first described protein adsorption and displacement processes on foreign surfaces after

incubation with complex liquids. He found surface protein concentrations, which are

different from the concentrations in the liquid and change over time [26]. Protein

adsorption during the initial response to material-blood contact depends on surface

characteristics, including hydrophilicity / hydrophobicity and charge [27]. Elevated

protein adsorption to hydrophobic surfaces was explained as a phenomenon were the

protein is expelled from aqueous solution in order to maximize self-association at the

expense of less-favourable water-protein interactions [28].

Protein adsorption to foreign surfaces affects platelet and leukocyte adhesion and

modulates the response because of the quick formation of a protein film on the

biomaterial surface The most important proteins that promote cell adhesion on

biomaterials include fibrinogen, von-Willebrand-factor, collagen and vitronectin, with

fibrinogen being the most abundant in plasma, considered to be a significant factor for

hemocompatibility. Not only the amount but also the specific properties of individual

proteins as well as their conformation influence subsequent cellular responses and thus

the materials biocompatibility. For example more important for platelet adhesion than

the amount of adsorbed fibrinogen is the orientation, conformation and functionality of

Page 12: Dissertation Marion Fischer - Qucosa

1 Theoretical background

5

the adsorbed protein [29, 30]. The same amount of adsorbed fibrinogen was found on

CH3- and COOH- modified SAMs upon incubation with fibrinogen solution, but fibrin

formation was significantly less on the negatively charged COOH surface [31].

Obviously there is an influence of electrostatic environment and conformational

changes of fibrinogen on thrombin-fibrinogen interactions.

Protein adsorption is not only important concerning cell adhesion but also for the

initiation of blood coagulation, which is initiated by an autoactivation process of the

clotting factor FXII to FXIIa on negatively charged surfaces (see section 1.3.2) [32-34].

Apart from coagulation enzymes and platelet related processes, immunologic reactions

also influence blood coagulation on biomaterial surfaces. As primary immune reactions

are governed by unspecific defence systems the activation and reaction of the

complement system plays a prominent part in determining the immunologic reactions.

Immunologic reactions of blood in contact with biomaterials are realized through

immunocompetent cells and proteins. The complement system is a plasmatic immune

defence system made up of over 20 different proteins usually being activated either by

immunoglobulins or by specific surface structures found on bacteria and other

pathogens. Certain biomaterial surface structures, such as hydroxyl groups, that

resemble natural initiators are known to elicit activation of the complement system [35].

Cellular immune reactions are linked to leukocytes – mainly neutrophilic granulocytes

and monocytes. Upon activation they expose CD11b, FX, fibrinogen, phosphatidylserin,

and tissue factor, mostly being procoagulant mediators [36, 37].

Complement initiation is achieved via 3 different pathways (termed classical, lectin or

alternative). The alternative pathway usually is thought to be responsible for the high

activation noted on surfaces containing hydroxyl-(-OH) or amino-(-NH2) groups

through a direct covalent thioester linkage with complement fragment C3. Some new

experimental data indicate that other effects like a graded protein adsorption, the

mobility of the surface or yet unknown chemical influences might be responsible too

[38, 39]. Also the classical pathway may have consequences for biomaterials [40]

especially if high amounts of IgG adsorb to a surface [41-43]. Especially the extended

blood-material contact during extracorporeal circulation causes a strong activation of

complement factors, which then partly stay on the surface, partly are released to

circulation. They have chemotactic and activating impact on leukocytes, which –in

addition to releasing tissue factor- release matrix degrading enzymes and reactive

oxygen species. Clinically this can manifest as lung fibrosis and susceptibility to

infections. Complement activation was found to correlate with the chemical

composition of the surface [19, 44-46]. Leukocyte adhesion was found to also be

Page 13: Dissertation Marion Fischer - Qucosa

1 Theoretical background

6

affected through adsorbed proteins with cell adhesive characteristics like e.g. fibronectin

[47, 48] and through physical parameters like shear force [49]. Yet it was also shown

that a major elicitor for the leukocyte adhesion is the adsorption of complement

fragments to the surface [46].

The present work addresses the question of protein adsorption considering that the

characterisation and measurement of surface-adsorbed proteins is important for

assessing the materials biocompatibility.

Page 14: Dissertation Marion Fischer - Qucosa

1 Theoretical background

7

1.3.2 Activation of coagulation via contact activation (intrinsic pathway)

Figure 3 Simplified scheme represents plasmatic coagulation by activation of factor XII (intrinsic

pathway) and factor VII / tissue factor (extrinsic pathway). The initial steps of the extrinsic pathways are

marked blue. Adapted from Spatnekar and Anderson [50]. FSAP: Factor VII-activating protease,

HMWK: High-molecular-weight-kininogen

As already mentioned above, material induced thrombotic responses further depend on

humoral events. Blood coagulation is directly influenced through modified protein

functions upon adsorption in the case of specific plasmatic reactions. Most prominent in

this context is the so-called contact activation pathway encompassing the coagulation

factors FXII, FXI, high-molecular-weight-kininogen (HMWK) and pre-kallikrein. This

group of proteins belongs to an initiation pathway of the coagulation cascade that

eventually leads to thrombin formation. The intrinsic pathway of the coagulation

cascade can be described as a cascade of zymogen activation processes starting with the

Page 15: Dissertation Marion Fischer - Qucosa

1 Theoretical background

8

adsorption of FXII to surfaces (see Figure 3). According to the leading concept,

thrombin formation on biomaterials is initiated through an auto-activation process of the

clotting factor FXII to FXIIa on negatively charged surfaces [51]. It is proposed that the

adsorption of coagulation factor FXII to negatively charged surfaces, such as connective

tissues and a number of biomaterials, leads to subtle conformational changes in the

enzyme that provoke auto-activation [32, 52, 53]. Possibly the density of negative

charges on a biomaterial influences the intensity of FXII activation [52] via positively

charged amino acids in its heavy chain [32]. Through complexation with HMWK,

which binds to surfaces in a similar way, the contact factors FXI and prekallikrein are

brought into close vicinity of FXII, triggering their reciprocal activation and leading to a

strong amplification of contact activation [52]. Extending this knowledge a new theory

states that it is not the presence of negative charges but differences in protein

displacement of competing proteins, partly described as an adsorption-dilution effect,

that is responsible for different degrees of contact activation on hydrophobic versus

hydrophilic materials [51, 54, 55].

Thrombin is the final enzyme in the coagulation cascade and acts to cleave the plasma

protein fibrinogen into fibrin monomers, which then polymerise and cross-link under

the action of factor XIII to form a fibrous mesh [32, 52, 53] allowing vessel wall repair

in natural environment but leading to a functional impairment of medical devices if

formed on material surfaces. Fibrin formation on biomaterials has been shown to be

directly related to FXII activation. Therefore contact activation is regarded as an

essential factor for coagulation activation on biomaterials [51, 53, 56, 57]. Numerous

studies on the initiation of the contact pathway of coagulation on biomaterial surfaces

lifted the activation of FXII to a powerful process in biomaterial related coagulation.

Yet the actual clinical relevance of this pathway for coagulation in vivo has been

strongly questioned given that individuals without FXII do not suffer from severe

bleeding. However, contact activation has been shown to play a significant role in

thrombus stability [58, 59]. One possible explanation for the controversial in vivo

effects of FXIIa might be that it inhibits thrombin-induced platelet aggregation by

binding the glycoprotein Ib (GPIb)-IX-V complex [60]. The correlation of surface

chemistry and FXII activation is, however, unquestioned and its impact in concert with

blood platelet response is part of the present work.

Page 16: Dissertation Marion Fischer - Qucosa

1 Theoretical background

9

1.3.3 Activation of coagulation via tissue factor (extrinsic pathway)

The extrinsic pathway of coagulation is triggered by tissue factor (TF). TF is a

transmembraneous protein on many subendothelial cells being also described as a

“hemostatic envelope” limiting bleeding after vessel injury. However, this protein

shows ambivalent action being an essential cofactor belonging to the clotting cascade as

well as a membrane bound molecule. The structural similarity of TF with cytokine

receptors explains its receptor function mediating intracellular signaling capacities

(tumor metastasis, angiogenesis and embryogenesis). On the other hand, TF triggers the

extrinsic pathway of coagulation (see Figure 3, pathway marked in blue). After vascular

injury this factor is exposed to blood, where it forms a complex with FVIIa that enables

the cleavage of factor X [61]. The following assembly of tenase (FIXa+ FVIIIa+ FX as

substrate) and prothrombinase complexes (FXa+ FVa+ prothrombin as substrate) on the

membrane of activated platelets transmits initially formed traces of thrombin to a

downwelling formation of fibrin via the so called thrombin burst.

The elucidation of TF contribution to blood clotting has come a long way and launched

diverse discussions. For many years it has been thought that TF activity is restricted to

the activation of an auxiliary pathway with little biological relevance. Nowadays

numerous publications reflect its prominence to be one of the most important regulators

of arterial thrombosis [62-65]. The role of TF being responsible for the onset of blood

coagulation finally gained acceptance but the mechanism of clot initiation is still a

controversial issue.

1.3.4 Sources and activity of TF

As TF was believed to be derived solely from endothelial cells, the resulting FXa was

supposed to diffuse from the vessel wall to the allured platelets through the formed

thrombus. However, the growing thrombus acts as a barrier restricting the connective

and diffuse exchange of substrates and coagulation products between the blood and the

reactive vessel wall. Hathcock et al. found that 4 hours are needed for diffusion of FXa

through a 1 mm fibrin clot with a diffusion rate of 2.3*10-7cm²/sec, whereas the

presence of platelets in the clot decreased the diffusion rate to 5.3*10-10cm²/sec (3.6

months). As thrombus formation was seen after a period of 10 minutes, an alternative

source of procoagulant activity being localized outside the thrombus seemed to be

essential for clotting [66]. It was in 1999 when Giesen et al. found an additional source

of TF in circulating blood besides the intravascular TF [62]. This so called blood borne

TF was further shown to affect thrombus growth independent of vessel wall derived TF

Page 17: Dissertation Marion Fischer - Qucosa

1 Theoretical background

10

[67]. Several studies followed focusing on the contribution of blood borne TF to

thrombosis, what launched a new concept of a cell-based coagulation model. The

initiation of coagulation by one single coagulation factor in the vessel wall alone was

questioned. Instead it was assumed that vessel wall TF represents a starter of

coagulation, whereas the blood borne TF propagates coagulation within the growing

thrombus.

The form of blood borne TF, however, still leaves open questions concerning its sources

and activity. So far, two different forms of circulating TF have been described: either as

a component of blood cells or as microparticle associated TF. Elicitors for a release of

cellular TF are multifold: activation of immunologic reactions like the formation of

C5a, certain leukocyte release products or the presence of lipopolysaccharides may

represent the more important examples [68]. De novo TF synthesis takes app. 60 min,

substantially longer than the clotting process. Microparticle TF on the other hand

emanates from residing TF in vesicles being formed during transcellular transfer (e.g. to

the membrane of activated platelets). The existence of TF associated with cell derived

microparticles [69, 70] and monocytes [71, 72] is generally accepted and the model of

blood clotting was revised. Following vascular injury P-selectin becomes exposed on

endothelial cells and activated platelets. The following interaction with monocyte CD15

(an epitope on PSGL-1) enables rolling and tethering between these cells [73, 74], being

considered as a possible way of TF transfer. Monocytes are capable of shedding

microparticles exposing TF and phosphatidylserine together with adhesion molecules

CD14, CD11a and CD18 after LPS stimulation [75]. Recruiting of those microparticles

to thrombi is again mediated via an interaction between PSGL-1 on microparticles and

P-selectin on activated platelets allowing growth of the forming thrombus. Several

authors suggest additional expression of TF by granulocytes [76] and platelets [77, 78].

Panes et al. demonstrated the ability of activated platelets to synthesise TF although

being anucleated. The underlying mechanism might be the splicing of platelet pre-

mRNA in mature mRNA upon cell activation [79]. Contrarily Butenas et al. found no

detectable TF activity in quiescent or ionophore-stimulated platelets [80]. Difficulties

concerning the analysis of TF in platelets decelerate the elucidation of that debate.

Besides the easy activation of platelets during isolation, investigations on that topic face

the problem that platelet preparations may contain small amounts of leukocytes.

Additionally TF present on platelets may be derived from TF positive microparticles

released from monocytes. The idea that there may be significant amounts of functional

TF on granulocytes was disproved by Osterud in several publications [81, 82]. The

authors postulate that the reported TF activity of granulocytes does not prove the ability

of those cells to express TF but is a consequence of TF acquired from plasma. In his

Page 18: Dissertation Marion Fischer - Qucosa

1 Theoretical background

11

recent study no TF associated with freshly isolated granulocytes was found using

western blot analysis, whereas TF was detected when cells where incubated with

platelet poor plasma.

Another contentious issue to solve was the question of how microparticle TF is

prevented from initiating blood coagulation in the absence of an injury. Like in the case

of factor VIIa in flowing blood, that is separated from cellular sources of TF by the

endothelium, an existing protection mechanism was assumed. Recent work on that

subject discovered the existence of a latent (encrypted) and an active form of TF

explaining its possible inactive or procoagulant activity [83, 84]. The detailed molecular

processes underlying encryption are uncertain, but protein dimerisation [85], lipid

reorganization [84, 86] and cellular secretion of TF-rich granules [87] are proposed

mechanisms. Two recent studies using purified full length TF incorporated into

phospholipid vesicles proposed that the activation of encrypted TF by protein disulfide

isomerase (PDI) initiates coagulation. On activation, platelets and endothelial cells

secrete PDI, which converts TF on cells or microparticles to its active form by

disrupting a critical disulfide bond [88, 89]. Pendurthi et al. questioned that hypothesis

reporting no effect on either TF coagulant or cell signalling functions after reduction of

PDI with gene silencing [90].

A few reports did show that the TF pathway can be relevant for biomaterials like

extracorporeal devices, catheters and heart valves [62, 91, 92]. Investigations on the

induction of TF by foreign materials deserve much more attention, especially in the

context of designing new drugs targeted to act at the beginning of the coagulation

cascade bearing less side effects and a stronger anticoagulation potential. For instance

inhibiting TF interactions or the transfer of TF-positive microparticles to platelets may

prove to be a novel strategy for the prevention of thrombosis.

1.3.5 Cellular responses upon material-blood contact focussing on

platelet adhesion

Apart from humoral reactions the activation of platelets definitely also plays a role in

the development of thrombi and immunologic reactions on biomaterial surfaces [68].

Platelets are activated in vivo when they adhere to subendothelial layers and/or are

exposed to soluble activators [93, 94].

The contribution of platelets to coagulation processes encompasses the release of

mediators, the change of their morphology from spherical to riffled, the expression of

certain surface receptors as well as the translocation of negatively-charged

phospholipids to the platelet surface mediated by cellular flippases. Consequently the

Page 19: Dissertation Marion Fischer - Qucosa

1 Theoretical background

12

composition of the platelet cell membrane changes such that more negatively charged

phosphatidylserine become exposed and microparticles may be shed [95]. The release of

intracellular granules includes coagulation factors VII and XI, the adhesion molecules

P-selectin and vWF, serotonin and platelet factor 4 (PF4), all of which impact further

the activation of other platelets, coagulation and inflammation [94, 96].

In case of a biomaterial the activation of platelets is surface-initiated and primarily

depends on the composition of the adsorbed protein layer (as detailed further above).

Adsorption of fibronectin, von-Willebrand-factor or fibrinogen is known to support cell

adhesion and/or activation [97-101] via adhesion receptors [102, 103] if the binding

sites of the proteins are exposed towards the cells [104]. The adhesion of blood platelets

to biomaterial associated fibrinogen and to other proteins with cell-adhesive capacity is

mainly mediated via receptors like GPIIb/IIIa that only becomes exposed after a change

of conformation upon adhesion [105].

Yet also activated coagulation factors themselves like thrombin in solution can activate

blood platelets via proteinase activated receptors (PAR). Thrombin activates at least

three different thrombin-receptors on human platelets. Apart from the well-known

activation through G-protein-coupled protease activated receptors (PAR1, PAR4) [106],

it has recently been found that the membrane glycoprotein GPIb also serves as a

thrombin-binding site. This receptor can even promote platelet activation at low

thrombin concentrations [107].

Since most if not all of the above specified reactions depend on the materials surface

and its impact on the protein adsorption from blood (e.g. FXIIa, fibrinogen, complement

fragment C3b) numerous attempts were undertaken to attain reliable parameters

determining the blood reaction with materials [108]. However, recent research in this

field as summarized by Ratner [109] and Sefton [110] confirmed serious difficulties to

establish a simple as well as instructive picture of the impact of the surface chemistry of

materials on the variation of hemocompatibility parameters. Considering these

restrictions the present work attempts to evaluate the influence of surface parameters on

biocompatibility by the interpretation of indicative hemocompatibility parameters using

various and reliable test systems.

Page 20: Dissertation Marion Fischer - Qucosa

2 Experimental part

13

2. Experimental part

The preparation of model substrates for studying initial processes of whole blood upon

material contact is described in the following section. This includes the formation of

gold surfaces as well as the production of self assembled monolayers, followed by the

methods for the characterisation of these surfaces.

2.1. Preparation of gold substrates

Gold substrates used for the preparation of self assembled monolayers should fulfil

special requirements. Important prerequisites are a complete gold coverage, a high

stability of the gold coating and a very low surface roughness. To meet these

requirements, different techniques for the cleaning of glass substrates prior to gold

deposition and different gold coating techniques were compared. Roughness parameters

expressed as root mean square (rms) were obtained by AFM measurements (AFM,

Nanoscope; Digital Instruments, Santa Barbara, USA).

The roughness of glass substrates (float glass disks (25 x 1.1 mm; Berliner Glas KG,

Berlin, Germany)) after application of two cleaning procedures was analysed. Cleaning

by oxidizing and etching agents (ammonium hydroxide/hydrogen peroxide/water: 1/1/5)

was compared to a detergent-based cleaning procedure (hellmanex glass cleaner;

Hellma GmbH, Müllheim, Germany). Subsequently, the mean roughness of gold

coating processes carried out by ion beam sputtering (via magnetron sputter coater) and

by chemical vapor deposition (Leybold univex 300, Köln, Germany) were compared on

the basis of AFM measurements. In both cases, float glass disks were cleaned using

hellmanex glass cleaner (10 min), purified water and ethanol in an ultrasonic water bath.

After immediate priming with a 3 nm chromium layer, an 80 nm gold layer (99.9%;

Saxonia Edelmetalle, Halsbrücke, Germany) was evaporated on the substrates at 5 x

10-2 mbar for the magnetron sputter and at 5 x 10-5 mbar for the chemical vapor

deposition.

Page 21: Dissertation Marion Fischer - Qucosa

2 Experimental part

14

2.2. Preparation and characterisation of self assembled

monolayers

Self assembled monolayers of different alkanethiols were prepared. Besides single

component SAMs also binary mixtures of C15-COOH and C15-CH3, as well as

monolayers consisting of C10-COOH, C10-CH3 and C10-OH were used.

2.2.1 Preparation and characterisation of C15-COOH/ C15-CH3

Self assembled monolayers of 16-Mercaptohexadecanoic acid (MHA; C15-COOH) and

Hexadecanethiol (HDT; C15-CH3) where prepared on gold substrates.

Different solvents used for immersion solutions of thiols as well as different deposition

times were applied in regard to optimal quality and reproducibility of the self assembled

monolayers. Ethanol being the most prominent solvent for SAM preparation was

compared to tetrahydrofurane (THF) and ethanol +5% acetic acid as proposed by

several authors for the formation of COOH terminated SAMs [22, 111]. Ethanol with

5% acetic acid was used as solution for preparation of binary layers as proposed by

Arnold et al. [22], who described the problem of possible dimer formation during self

assembly (formation of another monolayer on top of the existing SAM) between the

carboxylic groups of COOH-terminated alkanethiols.

Gold substrates were flushed with a vigorous stream of CO2/dry ice (“snow jet”). This

technique, used to remove any adhering particles, was found to be optimal to remove

contaminants (see Table 1). After this cleaning procedure samples were immediately

immersed into the thiol solutions. All single-component SAMs were prepared by

chemisorption from 1 mM solutions in immersion solvent (ethanol or THF) for 20 min

at room temperature. All mixed monolayers were prepared by freshly mixing 1 mM

stock alkanethiol solutions in the desired ratio and diluted to obtain a total alkanethiol

concentration of 0.2 mM. After assembly, the substrates were rinsed thoroughly with

immersion solvent and absolute ethanol and dried in a stream of nitrogen. Surfaces for

all experiments were used right after preparation.

SAMs where analysed by several surface sensitive techniques (characterisation methods

are presented below).

Page 22: Dissertation Marion Fischer - Qucosa

2 Experimental part

15

Surface characterisation using physico-chemical methods

Ellipsometry (SE 400adv, Sentech instruments GmbH, Berlin, Germany) was used to

analyse the thickness of the monolayer. Three measurements on each surface were

carried out. For in situ measurements the wafers were left on the site of measurement

during the whole deposition process.

Surface hydrophilicity was determined by water contact angle measurement using the

sessile drop technique with an OCA 30 system (Dataphysics, Filderstadt, Germany). A

volume of 3 µl degassed deionized water (MilliQ quality) was added to an initial 4 µl

drop to measure advancing contact angles on at least 3 spots of each surface.

Surface analysis with X-ray-photoelectron spectroscopy (XPS) was performed with an

AXIS ULTRA (Kratos Analytical, Manchester, UK) spectrometer using

monochromated Al-KR radiation at take off angles of 0°. IRRAS (infrared reflection

adsorption spectroscopy) -grazing incidence- Fourier transform infrared spectroscopy,

internal reflection mode (FTIR-IRRAS) spectra were taken with a Bruker Equinox 55

(Bruker Optics, Ettlingen, Germany) using p-polarized light at a grazing incidence of

80° and a resolution of 4 cm-1 with 3000 scans per measurement.

AFM measurements were performed with a Bioscope to determine surface roughness of

SAM surfaces (AFM, Nanoscope; Digital Instruments, Santa Barbara, USA).

Charging of surface was characterised by streaming current measurements using a

microslit electrokinetic setup that was previously described in detail [112]. Zeta

potentials were derived from the streaming current ratio using the Smoluchowski

equation (for more details see also [112]). Channel height was 30 µm and electrolyte

solutions with different ionic concentrations (0.1, 1.0, 10 mM KCl) were applied, pH

sweeps were performed from pH 9 to pH 3.

2.2.2 Preparation and characterisation of C10-COOH/ C10-CH3

Alternatively to C15-COOH and C15-CH3, alkanethiols with shorter chain length were

used for SAM preparation. 11- Mercaptoundecanoic-acid (HS-(CH2)10-COOH, MUA)

and Undecanethiol (HS-(CH2)10-CH3, UDT) were obtained from Sigma Aldrich

(Deisenhofen, Germany) and used as received. Ethanol for single component SAMs and

THF for binary SAMs was used as previously reported by Ulman et al. [5], who found a

good agreement between the composition of the liquid phase and the monolayer

obtained for mixtures of SAMs with polar and nonpolar endgroups (MHA/HDT) when

THF is used as solvent.

All single-component SAMs were prepared by chemisorption from 1 mM solutions in

absolute ethanol. All mixed monolayers were prepared by freshly mixing 1 mM stock

alkanethiol solutions in the desired ratio and diluted to obtain a total alkanethiol

Page 23: Dissertation Marion Fischer - Qucosa

2 Experimental part

16

concentration of 0.2 mM using dry (molecular sieve) THF. After assembly for 20 min at

room temperature, the substrates were rinsed thoroughly with immersion solvent and

absolute ethanol and dried in a stream of nitrogen. Just before self assembly surfaces

were cleaned with CO2/snow jet. Single component SAMs named 100% CH3 (100%

C10-CH3, also named 0% -COOH) and 100%COOH (100% C10-COOH), as well as

binary mixtures of C10-CH3 and C10-COOH were prepared and tested for their physico-

chemical properties.

Binary mixtures were: 17% -COOH (17% C10-COOH /83% C10-CH3)

34% -COOH (34% C10-COOH /66% C10-CH3)

50% -COOH (50% C10-COOH /50% C10-CH3)

66% -COOH (66% C10-COOH /34% C10-CH3)

83% -COOH (83% C10-COOH /17% C10-CH3)

To control monolayer quality, the samples were examined by wetting measurements,

ellipsometry and reflection-adsorption spectroscopy (IRRAS-FTIR), X-ray-

photoelectron spectroscopy (XPS), and atomic force microscopy (AFM).

Influence of cleaning procedures of gold substrates

Different cleaning procedures of gold substrates like snowjet or plasma cleaning and

heating to 190°C, as well as a combination of those procedures and uncleaned substrates

were compared. Residual contaminants on the surface were evaluated by XPS

measuring the O(1s) signal. SAMs and gold substrates do not contain any oxygen

containing thiols. The detection of oxygen therefore is an indicator of contamination.

Preparation and characterisation of MUOH/MUS (C10-COOH; C11-OH)

SAM were prepared analogue to protocol described in chapter 2.2.2, with the only

difference of using ethanol for single component SAMs as well as for binary SAMs.

Single component SAMs named 100% -COOH (100% C10-COOH, also named 0%

-COOH) and 100%OH (100% C10-OH), as well as the following binary mixtures were

prepared: 50% -OH (50% C10-OH /50% C10- COOH)

90% -OH (90% C10-OH /10% C10- COOH)

A general characterisation of the different SAMs was performed using water contact

angle and XPS measurements.

Page 24: Dissertation Marion Fischer - Qucosa

2 Experimental part

17

2.2.3 Preparation and characterisation of C10-COOH/ C11-

(O-CH2CH2)3-O-CH3)

EG3Ome/MUS SAMs were prepared as described in chapter 2.2.2, with the only

difference of using ethanol for single component SAMs as well as for binary SAMs.

Single component SAMs named 100% -COOH (100% C10-COOH, also named 0%

EG3Ome) and EG3Ome (100% EG3Ome), as well as binary mixtures were prepared:

17% - EG3Ome (17% C10-COOH /83% EG3Ome)

34% - EG3Ome (34% C10-COOH /66% EG3Ome)

50% - EG3Ome (50% C10-COOH /50% EG3Ome)

66% - EG3Ome (66% C10-COOH /34% EG3Ome)

A general characterisation of the different SAMs was performed using water contact

angle and XPS measurements.

This set of surfaces is supposed to have the same gradual alteration of surface charge

like MUA/UDT surfaces but it has different wettability properties. With the help of

EG3Ome/MUA surfaces, the influence of surface parameters like wettability and charge

on initiation of coagulation can be scrutinized.

2.3. Characterisation of protein adsorption and enzyme

activation

The adsorption of proteins relevant for the materials hemocompatibility like fibrinogen

and complement fragment C3b as well as the activation of FXII and thrombin were

assessed.

2.3.1 Human fibrinogen/ fibrin

QCM measurement of fibrinogen adsorption and transformation

Protein adsorption to SAM modified crystals (QSX301) was analysed using the QCM-D

model 300 (Q sense, Västra Frölunda, Sweden) at constant 37°C. Quartz crystals with a

natural frequency of 5 MHz were precleaned with a mixture of hydrogen peroxide,

ammonium hydroxide and water (1/1/5) at 70°C for 10 min before SAM formation. The

crystals were stabilized in HEPES buffer (20 mM HEPES, 150 mM NaCl, 1 mM CaCl2,

pH 7.4) to achieve a stable baseline and subsequently incubated with fibrinogen solution

Page 25: Dissertation Marion Fischer - Qucosa

2 Experimental part

18

(HFG; human fibrinogen (fraction1) Sigma-Aldrich, Deisenhofen, Germany; 1 mg/ml in

HEPES) for 20 min followed by three rinsing steps.

The detection of conformationally changed fibrinogen was carried out by subsequent

incubation with anti-FPA (affinity purified rabbit antibodies specific for human

fibrinopeptide A (FPA); Haemochrom diagnostica, Essen, Germany) for 1 hour was

carried out ensuing again 3 rinsing steps. Frequency and dissipation shifts induced by

adsorbed proteins were monitored in real time at the third, fifth and seventh overtone

(15, 25 and 35 MHz, respectively). All experiments were performed in HEPES buffer

under non-flow conditions.

Immunostaining

SAM surfaces were incubated with fibrinogen solution (human fibrinogen (fraction1)

Sigma-Aldrich, Deisenhofen, Germany; 1 mg/ml in HEPES) for 20 minutes at 37°C.

Surfaces were rinsed with PBS and subsequently stained with FITC-anti-HFG (FITC-

rabbit anti-Human Fibrinogen, Cederlane laboratories, Ontario, Canada; 1/100).

For the detection of conformationally changed fibrinogen, surfaces were incubated with

NYB B4-2 mouse anti-fibrinogen γ chain (Accurate chemical & scientific cooperation,

New York, USA; 1/100) and Alexa488-anti IgG (Alexa Fluor 488 goat anti-mouse IgG,

Invitrogen, Oregon, USA; 1/200) for 1 hour at RT or overnight at 4°C. All antibody

solutions were prepared in PBS with 1%BSA. After three rinsing steps surfaces were

scanned by fluorescence scanning (FLA 5100, FujiFilm, Japan).

Adsorption of 125I-HFG

Radio labelling of fibrinogen (HFG, Sigma-Aldrich, Germany) was carried out with a

modified chloramine-T iodination method (IODO-Beads®, Pierce Protein Research

Products, Thermo Fisher Scientific, Rockford, USA). The remaining non-conjugated 125I was determined to be <3% of the total radioactivity. Freshly obtained human plasma

(blood heparinised with 2 IU/ml centrifuged at 2000g for 10 min at RT) was spiked with 125I-HFG at about 4% of the total HFG concentration. Surfaces were incubated (5, 30, or

120 min) with the spiked plasma and washed several times with distilled water.

Phosphorimaging (FLA 5100, FujiFilm, Japan) was applied to quantify surface-

adsorbed radioactivity. A standard curve was determined by preparing a dilution series

of 125I-HFG-spiked plasma.

Page 26: Dissertation Marion Fischer - Qucosa

2 Experimental part

19

Adsorption of fibrinogen/ fibrin measured by immnochemistry using an anti-HFG

antibody

Surfaces were incubated with heparinised fresh whole blood (1.7 IU/ml) for the period

of 2 hours. Subsequent addition of OPD substrate to these SAMs after incubation with

HRP coupled anti-HFG antibody allowed detection of surface bound HFG by

photometric measurements.

2.3.2 Adsorption of complement fragments C3b and release of C5a

Initiation of the complement cascade on SAM surfaces

The activation of the complement system was evaluated by detecting complement

fragment C3c bound to the surfaces. The antibody for C3c detects this domain also in

C3b. As only this fragment has special affinity to a surface, whereas the C3c fragment is

soluble, surface-adsorbed C3c is regarded as C3b only.

Detection of surface bound complement by ellipsometry

Detection of surface bound C3b upon surface incubation with plasma: The amount of

surface bound blood plasma- and concomitant anti-C3c binding were determined in a

two zone measurement set-up by null ellipsometry (Rudolph Research AutoEL III,

Rudolph technologies, New Jersey, USA) in a glass cuvette at a wavelength of 632.8

nm at a 70º angle of incidence. Single component SAMs were incubated in veronal

buffer (VB, Lonza Group Ltd., Basel, Switzerland; 0.145 mM NaCl; 1.84 mM Na-5,5,

diethyl-barbital; 3.15 mM 5,5-diethylbarbituric acid; 0.2 mM CaCl2; 1.07 mM MgCl2)

for the initial measuring of psi and delta values. Ellipsometric angles were again

recorded after exchanging buffer for 66% plasma (obtained from Linköping hospital,

stored at –80°C until use). Plasma incubation was stopped after 10, 60 and 120 min by

gently washing the surfaces 3 times with VB. Finally a solution of anti-C3c (P0213,

DakoCytomation, Hamburg, Germany; 1/50 in VB) was added for another 20 min and

Ψ and ∆ were determined again after rinsing the surface 3 times with VB. All

experiments were carried out at 37°C by heating all solutions and cuvettes during

measurements. Protein layer thickness was calculated using the McCracking algorithm

to obtain the thickness of the adsorbed protein film, and the de Feijters et al. (de Feijter)

formula was used to calculate the amount of adsorbed proteins [113, 114]. The assumed

protein refractive index was N=1.465+i0.

Page 27: Dissertation Marion Fischer - Qucosa

2 Experimental part

20

Detection of surface bound complement by ELISA

For the analysis of surface complement sample surfaces were washed blood-free with

Veronal buffered saline (Institute Virion, Zurich, Switzerland). Subsequently samples

were rinsed with deionised water, air dried and stored at -70 °C up to analysis. The

surface-adherent protein C3b was quantified by direct immunochemistry. A surface area

of 2 cm2 was incubated in blocking buffer (PBS, 1% bovine serum albumin (BSA),

0.1% Tween-20) for 1 hour. Horseradish peroxidase labelled antibodies were diluted in

blocking buffer (polyclonal anti C3c, 1:500, from DakoCytomation, Glostrup,

Denmark) and incubated on the surfaces for one hour. Free antibody was removed by

washing with a Tween-20 containing buffer. o-phenylenediamine (OPD) (0.4 mg/ml)

with 0.4 mg/ml urea hydrogen peroxide in 50 mM phosphate-citrate buffer pH 5.0

(Sigma Aldrich, Crailsheim, Germany) was used as chromogenic substrate. The reaction

was stopped with 3 M HCl, and the optical absorption was detected at 450 nm. A

calibration curve for excluding a non-linear behaviour of the colour reaction was

obtained from defined concentrations of the antibody.

Detection of complement fragment C5a

Commercial enzyme-linked immunosorbent assays (ELISAs) were used to characterize

complement activation following whole blood incubation (C5a, Enzygnost C5a micro,

DRG Diagnostica, Marburg, Germany) and is described more detailed in section 2.5.

2.3.3 Contact activation: factor XIIa and kallikrein acti vity

Activity assay of factor XIIa/kallikrein

FXIIa and kallikrein activity was assayed after incubation of the samples (2 cm²) for

1 min with 300 µl of citrated plasma (1 part 0.147 mM tri-sodiumcitrate-5,5-hydrate

(Riedel de Haen, Seelze, Germany) in NaCl to 9 parts blood, centrifuged for 10 min at

2,000 g). 30 µl plasma was mixed with 200 µl chromogenic substrate solution (0.3 mM

S 2302TM (Chromogenix, Lexington, MA) in 50 mM HEPES, 120 mM NaCl, pH 7.4)

and the absorbance kinetics were recorded immediately for a 30 min period at 405 nm

using a microplate reader (Tecan GENios, Tecan GmbH, Crailsheim, Germany). The

chromogenic substrate used also detects kallikrein additionally to FXIIa, yet no

inhibitor was used as contact activation on the whole was of interest. Vmax (OD/min)

based on the maximum slope of the kinetic curves was compared between samples.

Plasma incubated on float glass (positive reference surface) and non-activated plasma

were used as references.

Page 28: Dissertation Marion Fischer - Qucosa

2 Experimental part

21

For the determination of the FXIIa and kallikrein activity adsorbed to the surface,

300 µl chromogenic substrate solution were incubated on the surfaces for 30 min and

stopped with 75 µl acetic acid (20%). Data were normalized to 100% -COOH.

Inhibition of factor XIIa by CTI

Activity of FXIIa / kallikrein was evaluated after incubation of citrated plasma with

dextran sulfate (DS) as activator. An amount of 30 µl plasma was mixed with 25 µl corn

trypsin inhibitor (CTI) in various concentrations (10, 20, 40, 60, 120, 160 and 240

µg/ml). After 10 minutes preincubation with CTI, 50 µl DS was added followed by

addition of 100 µl chromogenic substrate solution.

Inhibition of factor XIIa by infestin

An activity assay of FXIIa / kallikrein was performed before and after incubation of

citrated plasma with DS. In the first set up 30 µl plasma was mixed with 25 µl infestin

in various concentrations (0.3, 0.6, 3 and 6 µM). After 10 minutes preincubation with

infestin (kindly provided by CSL Behring, Hattersheim, Germany), 50 µl DS was added

followed by addition of 100 µl chromogenic substrate solution. In the second set up all

the previously reported steps are equal, yet the addition of infestin was done after

plasma preincubation with DS.

2.3.4 Thrombin

Activity of thrombin on SAMs

Thrombin activity was assessed after incubation of the samples (2 cm²) at 37°C for

3 min with 300 µl citrated plasma (centrifuged at 2000 g for 10 min at RT) or citrated

platelet rich plasma (centrifuged at 100 g, 10 min, RT). Thrombin formation was started

by adding CaCl2 (final c=17 mM). After an incubation time of 3, 6, 8 and 10 min the

reaction was stopped again by addition of sodium citrate. Thrombin activity was

determined kinetically using a fluorogenic substrate (ZGGR-AMC 1 mM with 15 mM

CaCl2 Technothrombin, Vienna, Austria) at λex = 360 nm and λem = 465 nm. The

thrombin formation is expressed in the first derivative of the emission curve; time until

peak thrombin formation was evaluated using software by Technothrombin (thrombin

peak time).

Page 29: Dissertation Marion Fischer - Qucosa

2 Experimental part

22

2.4. Surface incubation with human blood plasma or platelet rich

plasma (PRP)

2.4.1 LDH assay on COOH/CH3 and COOH/CH3/OH

Freshly drawn whole blood, citrated, was centrifuged at 100g for 20 min in order to

obtain platelet rich plasma (PRP). 300 µl PRP were incubated (45 min, 37°C) on

surfaces (2 cm²). Subsequent rinsing of incubated surfaces with phosphate buffered

saline (PBS) was followed by addition of 100 µl Triton-X (1%) for 5 min to induce cell

lysis and the release of lactate dehydrogenase (LDH). 200 µl of a reagent solution of

0.28 mg/ml NADH and 0.17 mg/ml sodium pyruvate in 80 mM Tris, 200 mM NaCl, pH

7.2 was added to 20 µl lysate [115]. The NADH consumption during the LDH catalysed

conversion of pyruvat to lactate was followed photometrically at 340 nm for 20 min

using a microplate reader (see above). The maximum slope of the kinetics is

proportional to the LDH activity and to the number of adherent platelets [116] and was

used for further evaluation. Standards were obtained from lysates of defined numbers of

platelets.

2.4.2 Detection of platelets after immunostaining using fluorescence

scanner

Surfaces were incubated with 300 µl PRP (adjusted to 300*109 Plt/l) for 45 minutes at

37°C. After fixation of adherent cells with formaldehyde (1% PFA, 1h, RT) and

subsequent blocking with 1% BSA in PBS, surfaces were stained with FITC-anti-

CD41a (FITC conjugate, Becton Dickinson, New Jersey, USA; 1/7 in PBS with 1%

BSA) and measured by fluorescence scanning (FLA 5100, FujiFilm, Japan).

2.5. Whole blood incubation assay

Hemocompatibility was assessed using fresh, heparinised (1.7 IU/ml, Sigma Aldrich,

Deisenhofen, Germany) whole human blood that was incubated in in-house designed

incubation chambers under non-flowing conditions as published in [4]. A sample

surface of 6.3 cm2 was exposed to 1.95 ml blood. The incubation was carried out for 2 h

at 37°C excluding air contact. Results were obtained in at least two separate

experiments with a triplicate set of samples (total n per sample type: 6). The

experiments implementing corn trypsin inhibitor (CTI, 160 µg/ml blood; CellSystems

Page 30: Dissertation Marion Fischer - Qucosa

2 Experimental part

23

Biotechnologie Vertrieb GmbH, St. Katharinen, Germany) and phospholipids (PL,

100 µl/ml blood; Technothrombin TGA reagent RA, Technoclone GmbH, Austria,

Vienna) were performed twice.

Blood was obtained from healthy donors who were unmedicated over the previous 10

days. Either whole blood or blood plasma was used. This study was approved by the

Ethics Committee of the Dresden University Hospital, Germany. Informed consent was

obtained from the donors prior to blood donation.

Blood plasma was analysed using ELISAs, whereas cellular reactions were determined

by measuring cell depletion in blood, cell adhesion on the surface with microscopic

techniques and by flow cytometric analysis.

Blood and plasma analysis

As described previously [4], commercial enzyme-linked immunosorbent assays

(ELISAs) were used to characterize blood reactions like coagulation activation

(thrombin-antithrombin-complex (TAT), Enzygnost TAT micro, Dade Behring,

Marburg, Germany), complement activation (C5a, Enzygnost C5a micro, DRG

Diagnostica, Marburg, Germany) and platelet activation (platelet factor 4 (PF4),

Haemochrom, South Bend, USA). Plasma was frozen and stored at -70 °C until

analysis.

Cell numbers (leukocytes, platelets, erythrocytes) were analysed by means of a whole

blood counter (Coulter AcTdiff. Krefeld, Germany). For all experiments described in this

section FITC-anti-TF from American diagnostica, Pfungstadt, Germany (clone VIC7)

and FITC-anti-CD41a from Becton Dickinson, New Jersey, USA were used.

Expression of TF RNA was analysed following isolation of leukocytes from whole

blood (500 µl) after surface incubation. RNA was extracted using peqGOLD blood

RNA kit (Qiagen, Hilden, Germany) as indicated in the manual and subsequently TF-

RNA was transcribed by means of RT- PCR using QuantiTectPrimerAssay (Qiagen,

Hilden, Germany). The cycle time was analysed relative to TF expression of the initial

value.

Expression of TF associated with granulocytes and monocytes, as well as the activation

marker CD11b on leukocytes and the formation of leukocyte-thrombocyte conjugates

was analysed by flow cytometry. 20 µl blood were stained with 4 µl FITC-anti-TF or

alternatively with 4 µl FITC-anti-CD41a and 4 µl anti-CD11b (phycoerythrine labelled)

(clone ICRF44, Biozol, Eching, Germany) for 30 min at room temperature in

polypropylene tubes; cell activation during this time was inhibited by 0.1 % NaN3. Then

red blood cells were lysed with FacsLyse (Becton Dickinson) and samples were

Page 31: Dissertation Marion Fischer - Qucosa

2 Experimental part

24

analysed immediately in a flow cytometer (FacsCalibur, Becton Dickinson). Positive

controls were obtained by incubation of blood samples with 100 EU/ml

lipopolysaccharide for 2 hours at 37 °C. Granulocytes and monocytes were gated by

their characteristic forward and side scatter pattern. Their CD11b and TF expression

was quantified as relative fluorescent units. Leukocytes positive for the thrombocyte

marker CD41a were regarded as thrombocyte-granulocyte or thrombocyte-monocyte

conjugate, respectively, and rated relative to the total number of the respective

leukocytes.

Western blot was carried out to quantify formed tissue factor after lysis of isolated

white blood cells. Following surface incubation leukocytes were isolated from whole

blood by Polymorphprep® (Axis shield, Oslo, Norway). For that purpose 840 µl whole

blood was centrifuged to remove platelets (1,500 g for 15 minutes at room temperature)

and the pellet was resuspended and added to a Polymorphprep® solution in proportion

of 1/1. Leukocytes were withdrawn from the mixture and adjusted to 2*109/l using a

blood cell counter (Coulter AcTdiff., Krefeld, Germany). Lysis of obtained leukocytes

was induced by 4 cycles of freeze thawing with subsequent addition of RIPA buffer

(Radio immuno precipitation assay; 50 mM Tris NaCl, 150 mM NaCl, 1% Nonident P-

40, 0.5% sodium-desoxychelat, 0.1% SDS, 10% glycerol (87%), 1% protease inhibitor

(HaltTM protease inhibitor single use cocktail, Thermo scientific, Rockford, USA)).

RIPA buffer was used to enable protein extraction from cytoplasmic, membrane and

nuclear proteins. After lysis samples were centrifuged to remove cell debris and strictly

stored at 4°C during all further steps. Protein content was adjusted to 667 µg/ml using

BCA protein kit (Thermo scientific, Rockford, USA). Cell lysate was diluted 1/4 in

4*SDS loading buffer, heated to 95°C for 5 min, resolved by SDS-PAGE (12%

polyacrylamide), and electroblotted onto polyvinylidene difluoride membranefilters

(Bio-Rad, Munich, Germany). Membranes were blocked overnight with 5% bovine

serum albumin / 0.05% TWEEN 20 / PBS followed by an incubation at 4°C for 1h with

monoclonal antibody anti-human TF (clone VIC7, 1/250, American Diagnostica Inc.

Pfungstadt, Germany). After a thorough washing in PBS-TWEEN, membranes were

incubated with polyclonal HRP-linked anti-mouse antibody (P0447; DakoCytomation,

Glostrup, Denmark) at 1/1000 for 1 h at room temperature. Immunoreactive proteins

were detected using the ECL detection system (Lumi-imager F1, Roche, Mannheim,

Germany). A second antibody (GAP-DH labelled with HRP (1/500; Santa Cruz

Biotechnologies, Heidelberg, Germany) was used as a loading control. Band intensities

were analysed using ImageJ relating obtained lane plots to loading control and finally to

the initial value.

Page 32: Dissertation Marion Fischer - Qucosa

2 Experimental part

25

Sample surface analysis

After blood incubation sample surfaces were prepared either for fluorescence or

scanning electron microscopy or for the analysis of complement fragments using

immunologic methods.

For analysis of cell adhesion and changes in morphology, sample surfaces were

prepared for scanning electron microscopy. After washing with phosphate saline buffer

(PBS; pH 7.4) to remove loosely bound cells, adherent cells were fixed with

glutaraldehyde (Serva Electrophoresis GmbH, Heidelberg, Germany) (2% in PBS),

subsequently rinsed with PBS, dehydrated with graded ethanol series and gold coated

with a sputter coater (SCD 050; BAL-TEC; Schalksmühle, Germany). Samples were

analysed by means of scanning electron microscopy (XL 30 ESEM FEG, FEI-Phillips,

Eindhoven, Netherlands) in high vacuum mode.

Fluorescence microscopy was used to detect the number of adhering leukocytes, as well

as for imaging TF expression. The number of thrombocytes was determined by SEM

counting cells at 10 predefined positions. The samples were washed blood-free, fixed

with 2% para-formaldehyde in PBS for at least one hour. Blood platelets were stained

with FITC conjugated anti-CD41a or alternatively TF was stained with FITC

conjugated anti-TF over night at 4 °C, followed by a staining of the nuclei of adherent

leukocytes with 0.5 µg/ml 4',6-Diamidine-2'-phenylindole dihydrochloride (DAPI),

0.2% Triton-X in PBS. Samples were analysed at a fluorescent microscope with

appropriate filter settings (Leica, DMIRE2). Microphotographs were taken of 20

predefined positions per sample. The adherent cell number was determined by computer

aided analysis (Image Tool, UTHSCSA, San Antonio, Texas). Density of adherent

platelets and surface TF was semiquantitavely measured via the surface fluorescence

intensity (FLA 5100, FujiFilm, Japan).

Adsorption of fibrinogen/ fibrin measured by immunochemistry using an anti-HFG

antibody

Surfaces were incubated with heparinised fresh whole blood (1.7 IU/ml) for the period

of 2 hours. Subsequent addition of OPD substrate to these SAMs after incubation with

HRP coupled anti-HFG antibody allowed detection of surface bound HFG by

photometric measurements.

Tissue factor RNA expression of blood cells on CH3/COOH/OH

Expression of tissue factor RNA was analysed following isolation of leukocytes from

whole blood (500 µl) after surface incubation. RNA was extracted using peqGOLD

blood RNA kit as indicated in the instruction manual and TF-RNA was subsequently

Page 33: Dissertation Marion Fischer - Qucosa

2 Experimental part

26

transcribed by RT-PCR using QuantiTectPrimerAssay (Qiagen, Hilden, Germany). The

cycle time was analysed relative to initial TF expression.

2.6. Western blot of leukocyte isolates

The following section describes the optimisation of western blot of leukocyte lysates

after whole blood incubation assay. Different isolation and lysis techniques are

compared.

2.6.1 Optimisation of leukocyte lysis

Cell lysis using SDS: Western blots were used to quantify tissue factor after lysis of

isolated white blood cells. Following surface incubation, leukocytes were isolated from

whole blood by ERL kit (peqGOLD blood RNA kit, peqlab Biotechnologie GmbH,

Erlangen, Germany). Cell lysis was induced by mixing cell pellet with 100 µl non-

reducing loading buffer. The mixture was heated to 95°C for 5 min, resolved by SDS-

PAGE (12% polyacrylamide), and electroblotted onto polyvinylidene difluoride

membrane filters (Bio-Rad, Munich, Germany). Membranes were blocked overnight

with 5% bovine serum albumin in 0.05% TWEEN / PBS followed by an incubation at

4°C for 1 h with monoclonal anti-human TF (clone VIC7, 1:250, American Diagnostica

Inc, Pfungstadt, Germany). After a thorough washing in PBS-TWEEN, membranes

were incubated with polyclonal HRP-linked anti-mouse antibody (P0447;

DakoCytomation, Glostrup, Denmark) at 1:1,000 for 1 h at room temperature.

Immunoreactive proteins were detected using the ECL detection system (Lumi-imager

F1, Roche, Mannheim, Germany). A second antibody, GAP-DH labelled with HRP

(1:500; Santa Cruz Biotechnologies, Heidelberg, Germany), was used as a loading

control. Band intensities were analysed using ImageJ and lane plots compared to the

loading control to determine the initial value.

Cell lysis using RIPA-buffer: Following surface incubation, leukocytes were isolated

from whole blood by ERL kit (peqGOLD blood RNA kit, peqlab Biotechnologie

GmbH, Erlangen, Germany). Cell lysis was induced by addition of 100 µl RIPA-Puffer

(50 mM Tris NaCl, 150 mM NaCl, 1% Nonident P-40, 0.5% sodium-desoxychelat,

0.1% SDS, 10% glycerol (87%)) after addition of 1µl Protease-inhibitor/100µl lysis

buffer. After agitation cell were incubated with lysis buffer for 20min during agitation at

4°C. Cell debris was removed from the samples by centrifugation (14.000 rpm, 5min).

Page 34: Dissertation Marion Fischer - Qucosa

2 Experimental part

27

30 µl cell lysate was mixed with 10 µl non-reducing loading buffer, heated to 95°C for

5 min and resolved by SDS-page as described above.

Cell lysis using RIPA buffer as described above with strictly keeping samples at 4°C:

Leukocyte isolates from whole blood after surface incubation (CH3/COOH and OH)

were analysed by western blot using the protocol as described above keeping the

samples at 4°C during the whole procedure.

2.6.2 Optimisation of gel-loading conditions

Recombinant TF was used to optimise gel loading conditions. TF standards were used

as followed: RD from Technothrombin, AD is an ELISA-kit standard from American

diagnostica.

Pure TF was mixed with loading buffer (either reducing or non-reducing), heated to

95°C for 5 min, resolved by SDS-PAGE (12% polyacrylamide), and electroblotted as

described above.

2.6.3 Optimisation of leukocyte isolation using Polymorphprep®

Western blots were used to quantify tissue factor after lysis of isolated white blood

cells. Following surface incubation, protein extracts from leukocyte isolates were

resolved by SDS-PAGE and electroblotted onto polyvinylidene difluoride membrane

filters as described in section 2.5 in the paragraph called “western blot”. Also here, band

intensities of TF were analysed using ImageJ and lane plots compared to the loading

control to determine the initial value.

The experiment was carried out once again as described above with the only difference

of using a polyacrylamide gel with lower acrylamide concentration (10% acryl amid

instead of 12%) for SDS page.

Statistics

Statistical evaluation was performed by one-way ANOVA and a subsequent pair-wise

multiple comparison procedure according to Tukey Test or Dunn’s method. Results

were regarded as significantly different if P ≤ 0.05.

Page 35: Dissertation Marion Fischer - Qucosa

3 Results

28

3. Results

The presented study aims at investigating the influence of surface properties on the initial

processes of blood coagulation. For that purpose self assembled monolayers of alkanethiols

on gold were prepared as model substrates (Figure 1). In the first results section the

preparation of these substrates is presented, focussing on the characterisation of the

underlying gold surfaces as well as of the alkanethiol monolayers. Following are the results

implementing those surfaces for the incubation with whole blood.

3.1. Preparation of gold substrates

Aiming to produce self assembled monolayers of optimal quality, protocols for gold

substrates with minimum surface roughness were optimised. The critical steps in the

preparation of flat gold substrates are the cleaning of the glass substrate and the technique

used for gold coating.

The cleaning of glass substrates using the detergent based technique (hellmanex) was found to

lead to lower rms values (0.89 nm) compared to using the oxidizing agent (2.52 nm) and was

further on used prior to gold coating. The high surface roughness of glass obtained after the

oxidation treatment (ammonium hydroxide/hydrogen peroxide) point to an induced etching of

the surface.

Gold coatings based on chemical vapor deposition showed lower rms values (0.88 nm)

compared to a roughness of 6.82 nm for coating by magnetron sputtering (see also Figure 4).

In both cases glass substrates were precleaned with hellmanex. For further SAM preparation

gold substrates were thus prepared by chemical vapor deposition of glass slides precleaned

with hellmanex glass cleaner.

Page 36: Dissertation Marion Fischer - Qucosa

3 Results

29

Figure 4 AFM tapping mode of gold surfaces prepared by chemical vapor deposition (A) and magnetron

sputtering (B)

Influence of cleaning procedures of gold substrates

Gold substrates prepared as described above were not always used immediately after

preparation, but stored according to experimental needs. Immediately before alkanethiol

deposition the gold surfaces were thus cleaned again to guarantee contaminant free surfaces.

XPS was used to evaluate the efficiency of surface cleaning as the presence of oxygen on gold

substrates can be interpreted as contaminant derived oxygen. O(1s) signals of gold surfaces

after the application of different cleaning techniques (CO2/dry ice, plasma cleaning and

heating to 190°C) were compared and are presented in Table 1. Snowjet cleaning was found

to be the only method that does remove all contaminants (0.5 % at O(1s)) compared to

uncleaned substrates (29% at O(1s)) and does not increase the O1s signal.

A B

Page 37: Dissertation Marion Fischer - Qucosa

3 Results

30

Table 1 Relative atomic composition of gold substrates after applying different cleaning techniques determined by XPS (values in parentheses correspond to the theoretical values based on stochiometry of thiol compounds)

Atom%

cleaning of gold substrates C(1s) (91.7) O(1s) (0) S(2p) (8.3)

snowjet (SJ) 94.0 0.5 5.5

SJ+plasma cleaner (PC) 84.5 12.5 3.0

SJ+PC+190°C 70.1 22.5 7.5

lab atmosphere/uncleaned 68.6 28.8 2.6

3.2. Preparation and characterisation of self assembled monolayers

The quality of self assembled monolayers was determined using several physico-chemical

characterisation methods: ellisometry, X-ray photoelectron spectroscopy (XPS) and

measurements of contact angle and the zeta potential.

3.2.1 Preparation and characterisation of C15-COOH/ C15-CH3

Ellipsometric analysis of self assembled monolayers provide information on their quality

regarding the layer thickness, the existence of a mono- or multiplayer and the tilt angle. Porter

et al. postulated a thickness of 2.5 nm for n-alkyl thiols adsorbed on gold with n=15 [117].

The results of our ellipsometric thickness measurements of SAMs with different ratios of –

COOH showed the expected value of 2.5 nm only for one sample (83% -COOH). All other

samples reached a thickness of 29-70% of the expected thickness (Table 2). This points to

incomplete or non-ordered alkanethiol assembly on the surface leading to reduced tilt angles

of the thiol chains .

Table 2 Ellipsometric measurement of thickness of MHS/HDT-SAMs (deposition solvent: ethanol)

XCOOH Thickness

solution (nm)

0 1.49

0.17 0.74

0.34 0.72

0.50 0.77

0.66 1.78

0.83 2.66

1.00 1.49

Page 38: Dissertation Marion Fischer - Qucosa

3 Results

31

XPS measurements of SAMs reflect the chemical composition of the monolayers and the

degree of thiol coverage. Measurements of O(1s) signals were done to determine the degree of

surface carboxylation and are presented in Table 3. O(1s) values should increase

proportionally to –COOH ratios of the surface. This tendency was not seen for any of the

immersion solvents used. Instead the O(1s) measurements for ethanol based immersion

solvents used for binary thiol mixtures showed no correlation at all. When using THF for

binary mixtures there is a weak correlation between –COOH rates of immersion solution and

O(1s) signals, but the O(1s) signals are too high for all measured surfaces.

Table 3 Relative atomic composition of SAMs prepared from mixtures of C15-CH3 and C15-COOH determined

by XPS using different solvents for immersion solution. EtOH: Ethanol for all SAMs; EtOH+AA: Ethanol for

single component SAMs and :Ethanol+5% acetic acid for binary SAMs and THF: Ethanol for single component

SAMs and THF for binary SAMs

O (1s) Atom%

XCOOH,soln calculated EtOH EtOH+AA THF

0 0 2.3 7.4 6.5

0.17 1.8 16.9 18.5 6.0

0.34 3.6 9.9 12.2 5.9

0.5 5.3 14.9 12.5 9.1

0.66 6.9 3.2 6.5 16.3

0.83 8.7 2.1 12.3 37.2

1.00 10.5 19.2 7.5 21.6

Water contact angle measurements are a simple and reliable technique to control the

monolayer homogeneity and give information on the presence of certain functional groups.

Values on surface wettability are presented in Figure 5. Surface hydrophilicity should scale

with the amount of –COOH groups on the surface. This expected trend, however, was not

found for binary MHA/HDT-SAMs: static water contact angles were 80-90° when using THF

or ethanol + acetic acid as immersion solvents. Incomplete or non-ordered self assembly can

again be mentioned as a reason.

Page 39: Dissertation Marion Fischer - Qucosa

3 Results

32

17 34 50 66 830

20

40

60

80

100

Sta

tic w

ater

con

tact

ang

le

in d

egre

es

% COOH of SAM (balance to CH3)

in EtOH/AA in THF

Figure 5 Water static contact angle of MHA/HDT-SAMs; SAM deposition solvent for 100% CH3 and 100%

COOH was ethanol with static contact angles of 94° and 73° respectively

Zeta potential measurements were carried out to determine the surface charge and potential in

electrolyte solutions. For surfaces displaying acidic groups the IEP should be at pH <4, for

alkaline surfaces at pH >4 [112]. The IEP of C15-COOH and C15-CH3 equalled 3.5 in both

cases (Figure 6), what further points to problems in surface preparation as the influence of

acidic and alkaline surface groups is supposed to alter the IEP for carboxylated and

methylated surfaces.

2 3 4 5 6 7 8 9 10-250

-200

-150

-100

-50

0

50

100% MHS, ζa(U

S), 1st measurement

100% MHS, ζ(IS), 1st measurement

100% MHS, ζa(U

S), 2nd measurement

100% MHS, ζ(IS), 2nd measurement

ζ [m

V]

pH2 3 4 5 6 7 8 9 10

-250

-200

-150

-100

-50

0

50

100% HDT, ζa(U

S), 1st measurement

100% HDT, ζ(IS), 1st measurement

100% HDT, ζa(U

S), 2nd measurement

100% HDT, ζ(IS), 2nd measurement

ζ [m

V]

pH

Figure 6 Zeta potential of C15-COOH (left) and C15-CH3 (right) at electrolyte concentration of 0.1mM KCl

All surface analytical tools used to characterise SAMs reflected ongoing problems with the

intended preparation of a dense monolayer with the same ratio of surface carboxylation/

methylation as found in the immersion solution. Varying values of layer thickness measured

by ellipsometry revealed that no uniform layer was formed and contact angle measurements

did not show the expected alteration in surface wettability dependant on surface

Page 40: Dissertation Marion Fischer - Qucosa

3 Results

33

carboxylation. Detection of sulfur compounds by XPS analysis showed some thiol formation

on the gold surfaces, but absence of increasing oxygen ratio confirmed no well ordered and

defined assembly of binary -CH3 and -COOH mixed thiols on the surface.

A possible explanation for the unsatisfying physico-chemical properties was given by Kind et

al. [118]. The authors described highly disordered MHA SAMs because of relatively strong

hydrogen bonds between the carboxylic acid functions and the interaction of –COOH groups

with the substrate as illustrated in Figure 7. This phenomena is more probable to occur in the

case of long chain alkanethiols like C15-COOH. Shorter alkanethiols were further on tried for

the preparation of well ordered self assembled monolayers.

Figure 7 Schematic illustration of a highly disordered C15-COOH SAM, taken from [118]

3.2.2 Preparation and characterisation of C10-COOH/ C10-CH3

Another type of alkanethiols was used for the preparation of self assembled monolayers

displaying the same functional groups as MHA/HDT but consisting of shorter chain length. In

the following section C10-COOH and C10-CH3 (and C11-OH) were applied for SAM

preparation.

Surface hydrophilicity as a function of deposition time was analysed. Contact angle

measurements of C15-COOH showed a strong variation depending on deposition time

displaying lowest values of 30° after a period of 20 minutes while after one hour constant

values of 60° where measured (Figure 8). Formation of SAMs with C10-COOH showed

similar results with low contact angle values at 20 minutes deposition time. Contrarily,

Page 41: Dissertation Marion Fischer - Qucosa

3 Results

34

constant contact angles where observed for C10-CH3 and C11-OH after 0, 3, 16 and 24 hours.

Best surface properties were found after an immersion time of 20 minutes for C10-COOH.

10 100 1200 1400

30

40

50

60

70

80

stat

ic c

onta

ct a

ngle

in d

egre

es

time (min)

CH3 COOH OH0

20

40

60

80

100

120

stat

ic c

onta

ct a

ngle

in d

egre

esterminal group of SAM

0,3h deposition time 16h deposition time 24h deposition time

Figure 8 Water static contact angle depending on different deposition times Figure left: 100% C15-COOH Figure

right: 100% C10-CH3; 100% C10-COOH and 100% C11-OH. For preparation of 100% OH (100% C11-OH) see

section 0

A possible explanation for the time-dependent increase of contact angles of COOH terminated

SAMs could be the formation of hydrogen bonds between two terminal COOH-groups

leading to the formation of a second layer on top of the SAM as already discussed by Arnold

et al. [22]. Wettability values of C10-SAMs where found to be similar to those described in

literature with 105-110° for C10-CH3 and values <20° for C11-OH and C10-COOH. Therefore

single component as well as binary SAMs were prepared based on self assembly time of 20

minutes and their physico-chemical properties are further characterised below. Despite the

proposed deposition time of >4 hours [119], also short deposition times being sufficient for

formation of self assembled monolayers were already described by Kim et al. The authors

found different diffusion rates of alkanethiols with different alkyl chain length and showed

that a full monolayers of 1-dodecanthiol (CH3−(CH2)11−SH) is formed already after a short

incubation time of 30 minutes [25, 120].

The characterisation of binary mixtures of C10-CH3/C10-COOH is described in the following

section.

Wettability measurements were carried out to get information on the presence of certain

functional groups of the monolayer. Contact angles of MUA and UDT, as well as of binary

mixtures of these thiols scaled with the amount of –COOH groups on the surface as presented

in Figure 9. Advancing water contact angles were plotted against the ratio of –COOH groups

on the surface and an inverse linear dependency was found (Figure 10). This confirms that the

Page 42: Dissertation Marion Fischer - Qucosa

3 Results

35

composition of the monolayer resembles the ratio of MUA/UDT of the immersion solution.

The results further evidence the accessibility of the functional groups on the surface, as only

freely accessible groups that are not buried inside the monolayer can lead to graded change in

surface wettability.

Figure 9 Images of water drops on MUA/UDT SAMs (from left to right: 100% -CH3, 17% -COOH, 34% -

COOH, 50% -COOH, 66% -COOH, 83% -COOH, 100% -COOH)

0 20 40 60 80 1000

20

40

60

80

100

120 experimental data theoretical values

Adv

anci

ng c

onta

ct a

ngle

in d

egre

es

%COOH of SAM (balance to CH3)

Figure 10 Advancing contact angle measurements of MHA/UDT-SAMs

XPS analysis done to determine the chemical composition of the monolayer are shown in

Table 4. XPS measurements were used to determine the degree of surface carboxylation as the

O(1s) signals reflect –COOH derived oxygen in the monolayer. Increasing O(1s) signals were

found for SAMs with higher –COOH ratios of the immersion solution. Again here, results

indicate that the ratio of –COOH terminated thiols in immersion solution scales with surface

carboxylation grades.

Page 43: Dissertation Marion Fischer - Qucosa

3 Results

36

Table 4 Relative atomic composition of SAMs of C15-CH3 and C15-COOH determined by XPS (values in

parentheses correspond to the theoretical values based on stochiometry of thiol compounds)

Atom%

XCOOH,soln C(1s) O(1s) S(2p)

0 91.5 (91.7) 2.5 (0) 6.0 (8.3)

0.17 91.7 (89.5) 3.4 (2.4) 5.0 (8.1)

0.34 87.3 (87.2) 5.9 (4.9) 6.9 (7.9)

0.50 86.0 (85.2) 8.8 (7.2) 5.2 (7.7)

0.83 82.6 (80.8) 12.4 (11.9) 5.0 (7.3)

1.00 81.5 (78.6) 14.5 (14.3) 4.0 (7.1)

The ratio of –COOH groups on the surface is calculated based on XPS measurements and

plotted against the ratio of MUA in immersion solution. A good correlation between

composition of the SAM and the MUA/UDT ratio of the immersion solution can be seen

(Figure 11). Theoretical values were calculated based on stochiometry of thiol compounds.

0.0 0.2 0.4 0.6 0.8 1.00.0

0.2

0.4

0.6

0.8

1.0

1.2 theoretical values experimental data

XC

OO

H o

f SA

M

XCOOH

in immersion solution

Figure 11 Ratio of –COOH groups on the surface as a function of MUA in immersion solution

Page 44: Dissertation Marion Fischer - Qucosa

3 Results

37

FTIR-IRRAS of SAMs

In order to guarantee a dense package of alkanethiols with well ordered polymethylene chains

after SAM formation of 20 minutes IRRAS spectra C10-CH3 and C10-COOH where analysed.

IR data obtained from SAMs not only allow for a reliable non-destructive determination of

the organic layer chemical composition, but also provide information on the molecular

orientation within the layer. IRRAS FTIR spectra showed peak positions for symmetrical

(2850 cm-1) and asymmetrical CH2-bands (2919 cm-1) on 100% -COOH and 100% -CH3 that

correspond to well-ordered crystalline layers with stretching vibrations at 2851 cm-1 and

2919 cm-1 respectively [119] (see Figure 12 left). IR data of single component SAMs of C10-

CH3 and C10-COOH as well as of binary mixtures thereof were also determined and are

presented in Figure 12 right. Peak intensities for methylene bands (2966 cm-1) were

proportional to the ratio of -CH3 of the monolayers with the highest peak for 100% -CH3.

3000 2950 2900 2850 2800-0.0001

0.0000

0.0001

0.0002

0.0003

0.0004

0.0005

0.0006

0.0007

wavenumber [cm-1]

abso

rban

ce u

nits

2965

.6

2919

.4

2878

.4

2849

.8

CH2 symCH

3 symCH

2 asCH

3 as

3000 2950 2900 2850 2800

0% COOH /

100% COOH

17% COOH

50% COOH

CH2 symCH

3 symCH

2 as

wavenumber [cm-1]

2966

2920

2880

2851

CH3 as

100% CH3

83% COOH

Figure 12 left: IRRAS FTIR spectra of 100% -COOH (red) and 100% -CH3 (blue), right: IRRAS FTIR spectra

of 100% -COOH and 100% -CH3 and binary mixtures

Zeta-potential of SAMs

Zeta potential measurements were carried out to investigate the charging of self assembled

monolayers in certain electrolyte solutions. The IEP of C10-COOH was determined at pH 3.4,

whereas the IEP of C10-CH3 was found to be at pH 3.9 (Figure 13). This is in accordance with

Werner et al. who found an IEP of 4 for non charged planar surfaces with neither acidic nor

alkaline groups. In this case the charging of the interface can be attributed to the preferential

adsorption of -OH ions. For surfaces displaying acidic groups the IEP is at pH <4, for alkaline

surfaces at pH>4 [112].

Page 45: Dissertation Marion Fischer - Qucosa

3 Results

38

1 2 3 4 5 6 7 8 9 10-180

-150

-120

-90

-60

-30

0

30

60 C

10-COOH

C10

-CH3

zeta

pot

entia

l [m

V]

pH

Figure 13 Zeta potential of C10-COOH and C10-CH3 at the electrolyte concentration of 0.1 mM KCl

Schweiß et al. discussed that also monolayers of alkanethiols on gold without dissociable sites

accumulate negative surface charges upon contact with aqueous electrolyte solution [7].

Electrokinetic measurements on hydrophobic HDT monolayers revealed a negative charge at

pH > 4 [121]. The reversal of the surface forces and the sign of the zeta potential takes place

near pH 4 being independent of the background electrolyte concentration. It is discussed that

the negative surface charge over a wide pH range is therefore acquired by preferential

adsorption of -OH. Recent studies and theoretical efforts on hydrophilic interfacial systems

confirm the dominating role of hydroxide ions in the surface charge formation of

nondissociating organic surfaces regardless of their hydrophilicity [122]. Therefore,

isoelectric points of about 4 were found for insoluble monolayers of long-chain alcohols [123]

and poly(ethylene glycol) ethers [124] at the air-water interface as well.

C10-COOH shows a strongly attenuated acidity with an IEP of about pH 3.4 in KCl solutions,

which is significantly higher than the IEP of 2-2.8 found for polymer brushes with -COOH

moieties such as spin-coated maleic anhydride-copolymer films [125]. The IEP shift can be

ascribed to reduced dissociation of –COOH groups in the densely packed self-assembled

monolayers.

The charging of C10-COOH was evaluated using KCl solutions in various concentrations. An

IEP of 3.7 was found for C10-COOH in low ion concentrations (0.1 mM KCl) (Figure 14).

This result is similar to the IEP of 3.4 as determined in the above mentioned experiment for

the same electrolyte concentration.

Page 46: Dissertation Marion Fischer - Qucosa

3 Results

39

2 3 4 5 6 7 8 9 10-320

-280

-240

-200

-160

-120

-80

-40

0

40

80

0.1 mM KCl 1 mM KCl 10 mM KCl

stre

amin

g cu

rren

t vs.

pre

ssur

e gr

adie

nt [p

A/m

bar]

pH

MUA (C10

-COOH)

-280

-245

-210

-175

-140

-105

-70

-35

0

35

70

zeta

pot

entia

l [m

V]

Figure 14 Zeta potential of C10-COOH using different background electrolyte concentrations of KCl

Decreasing values for the zeta potential of C10-COOH were observed with increasing KCl

concentration (Figure 14), a phenomenon that was also described for the alkane (docosane)-

water interface [126]. The zeta potential for C10-COOH at higher pH values varied compared

to results of the previous experiment Figure 13. This phenomenon can be explained by

differences in layer thickness of the underlying gold surface (80 compared to 50 nm for the

two experiments), that do influence the zeta potential in solutions of high pH ranges. Reduced

gold thicknesses provide an increased specific resistance possibly leading to the observed

polarisation effects.

Cleaning of gold substrates for re-usage of gold surfaces /QCM-crystals

For QCM measurements it was necessary to remove alkanethiols from gold substrates to

enable the crystal reuse. The loss of atomic % of S (determined by XPS as the S2p-Signal)

reflects the efficiency of thiol desorption. Elevated temperature and UV treatment were used

as well as combinations of both to remove thiols from gold surfaces. UV treatment for 1 hour

with subsequent rinsing of the surface was found to be the most efficient method with a loss

of S of 54%.

Page 47: Dissertation Marion Fischer - Qucosa

3 Results

40

Table 5 Loss of sulfuric compounds of gold surfaces after removal of self assembled monolayers using different

cleaning procedures. Values are based on XPS measurements, where the S(2p) signal before and after cleaning

was determined. Ref: reference, T(260°C): heating of samples to 260°C in vacuum oven, UV(60 mW): UV

treatment of samples at 60 mW

Cleaning procedure Loss of S (%) SD Ref 2 27

T(260°C) 34 2 UV(60mW) 54 6

UV/60mW+T(260°C) 28 4

A general protocol was established for the preparation of SAMs that display reproducible

high quality.

3.2.3 Preparation and characterisation of C10-COOH; C11-OH

Water contact angle measurements are carried out to determine the presence of certain

functional groups in the monolayer. Surface modification with –COOH or –OH terminated

thiols should lead to highly hydrophilic monolayers with static water contact angles of 10-20

degrees. According to these expectations, static contact angles <20° were found for SAMs

consisting of C10-COOH; C11-OH and for SAMs of binary mixtures of these thiols (Table 6).

Table 6 Water static contact angle (stat CA) of C10-COOH and C11-OH-SAMs (Ethanol as deposition solvent; deposition time of 20 min)

stat CA SD 100% COOH 15.9 6.0

50% OH 18.7 3.2 90% OH 18.3 5.6 100% OH 12.8 1.6

To determine the monolayer composition in the case of MUA/MUOH mixtures, the hydroxy

groups were derivatised by esterification with trifluoroacetic anhydride (TFAA) as proposed

in literature [127, 128] (Table 7). TFAA exclusively reacts with the OH groups of MUOH in

the frame of the reaction time applied in this work (30 min). Based on this procedure, the

surface fraction of OH-terminated alkanethiol can be estimated by analysing the F (1s)

signals.

Page 48: Dissertation Marion Fischer - Qucosa

3 Results

41

Table 7 Relative atomic composition of SAMs prepared from mixtures of C10-CH3 and C10-COOH after

esterification of OH groups with the anhydride of trifluoroacetic acid was determined by XPS (values in

parentheses correspond to the theoretical values based on stochiometry of thiol compounds)

Atom%

XOH,soln C(1s) O(1s) S(2p) F(1s)

0 74.2 (78.6) 23.7 (14.3) 1.8 (7.1) 0.3 (0)

0.5 68.3 (72.7) 17.3 (12.6) 3.3 (6.4) 9.3 (8.3)

0.9 73.0 (67.9) 8.1 (11.4) 5.0 (5.8) 12.1 (14.9)

1.0 80.0 (66.7) 5.7 (11.1) 3.4 (5.6) 12.6 (16.6)

Increasing F(1s) signals revealed increasing ratio of hydroxylic functionalities on the surface

according to higher amounts of OH- terminated thiols in immersion solution (Table 7).

Figure 15 shows roughness parameters of binary monolayers for 50% -OH (1.17 nm rms) and

90% -OH (1.40 nm rms). Both values are in an acceptable range for self assembled

monolayers and the existence of an unfavourable multiplayer formation can be ruled out.

Page 49: Dissertation Marion Fischer - Qucosa

3 Results

42

Figure 15 Roughness and section analysis of A: Mercaptoundecanol (100% -OH); B: binary monolayer (90% -

OH) measured by AFM in tapping mode

3.2.4 Preparation and characterisation of C10-COOH/ C11-

(O-CH2CH2)3-O-CH3)

The preparation of tri-methoxy terminated ethylene glycol (EG3Ome) was performed

according to the standard protocol used for the preparation of -CH3/-COOH terminated SAMs

(see section 2.2.2). Water contact angle measurements were done to control the monolayer

homogeneity and give information on the presence of carboxylic functional groups. With

decreasing surface carboxylation the values for static water contact angles should increase.

Wettability measurements done after a deposition time of 20 minutes and after 24 hours both

did not show the expected correlation for all the samples. Repetitive measurements revealed

the same trend with at least one sample being out of the expected range. This indicates a

disturbed accessibility of functional groups of the monolayers.

A

B

Page 50: Dissertation Marion Fischer - Qucosa

3 Results

43

0 17 34 50 1000

10

20

30

40

50

60

70C

onta

ct a

ngle

s in

deg

rees

static advancing receding

% EG3Ome

0 17 34 50 1000

10

20

30

40

50

60

70

Con

tact

ang

les

in d

egre

es

static advancing receding

% EG3Ome

Figure 16 Static (dark grey), advancing (black) and receding (grey) water contact angles of mixtures of C15-

COOH and EG3Ome after 20 min deposition time (left) and 24h deposition time (right )

The chemical composition of the monolayer was studied by XPS analysis. Here the ratio of

carboxylic and carbonyl compounds (deriving from the glycol functionality) was determined

to evaluate the degree of surface carboxylation and the ratio of ethylene groups in the

monolayer. A good correlation between the carboxylic and carbonyl compounds in the SAM

surface and the ratio of ethylene glycol in solution was found (see Table 8).

Table 8 Relative atomic composition of different C-compounds of SAMs (aliphatic, carboxylic and carbonyl

group) prepared from mixtures of C15-COOH and EG3Ome determined by XPS (values in parentheses

correspond to the theoretical values based on stochiometry of C-compounds)

Atom%

XCOOH,soln CH CO COOH

1.00 93 (94) 0 (0) 7 (6)

0.83 80 (87) 13 (7.5) 7 (5)

0.66 80 (81) 14 (15) 6 (4)

0.50 76 (75) 18 (22) 6 (3)

0 66 (56) 34 (44) 0 (0)

The chemical composition of surfaces was further analysed by XPS after derivatisation of

-COOH groups using diaminobutane. By determination of the N(1s) signal, the degree of

surface carboxylation can be estimated. Increasing N(1s) signals were found in accordance

with higher –COOH ratios of the immersion solution (Table 9).

Page 51: Dissertation Marion Fischer - Qucosa

3 Results

44

Table 9 Relative atomic composition of SAMs prepared from mixtures of C15-COOH and EG3Ome determined

by XPS after derivatisation of activated COOH groups with diaminobutane (values in parentheses correspond to

the theoretical values based on stochiometry of thiol compounds)

Atom%

XEG3Ome,soln C(1s) O(1s) N(1s) S(2p)

0 85.6 (83.3) 6.2 (4.2) 5.9 (8.3) 2.3 (4.2)

0.17 83.5 (82.5) 7.0 (6.4) 5.8 (6.9) 3.7 (4.2)

0.34 81.3 (81.6) 11.0 (8.7) 4.7 (5.5) 3.0 (4.2)

0.50 77.3 (80.8) 15.7 (10.8) 3.4 (4.2) 3.7 (4.2)

1.00 78.1 (78.3) 17.0 (17.4) 1.2 (0) 3.6 (4.3)

Alkanethiols are present on the surface in the desired ratio. Wettability measurements,

however, were unsatisfying pointing to an unordered structure of the assembled thiols.

Different solvents for SAM preparation were thus tested to achieve the expected physico-

chemical properties. Besides using ethanol absolute either 5% acetic acid was added to

ethanol as described in Arnold et al. [22] or 5%H2O was added as proposed by Tengvall

(personal communication). Only the ethanol water mixture as immersion solvent gave

expected static water contact angles of <30° for MUA (Figure 17) and was thus used to

prepare binary EG3Ome/MUA monolayers.

EtOH EtOH + H20 EtOH +AA

0

20

40

60

80

Con

tact

ang

les

in d

egre

es

static advancing receding

Immersion solvent

Figure 17 Water contact angles of C15-COOH after assembly using different immersion solvents: EtOH: ethanol

absolute, EtOH + H2O: ethanol absolute + 5% H2O, EtOH+AA: ethanol absolute + 5% acetic acid (20 min

deposition time)

Page 52: Dissertation Marion Fischer - Qucosa

3 Results

45

Single component monolayers as well as binary EG3Ome/MUA were tested for their static

water contact angle (Figure 18). A good correlation of surface hydrophobicity to the ratio of

ethylene glycol in the immersion solution was found proving the preparation of well ordered

self assembled monolayers with a defined degree of certain functional groups.

0 17 34 50 1000

10

20

30

40

50

60

70

Con

tact

ang

les

in d

egre

es

static advancing receding

% EG3Ome

Figure 18 Water contact angles of mixtures of C15-COOH and EG3Ome after assembly using ethanol absolute +

5% H2O as immersion solvent (20 min deposition time)

Page 53: Dissertation Marion Fischer - Qucosa

3 Results

46

3.3. Characterisation of protein adsorption and enzyme activation

Protein adsorption phenomena reflect the initial material induced reactions upon blood

contact and is thus studied in the following chapter. Fibrinogen, complement fragments C3b

and C5a as well as factor XII and thrombin are among the proteins being relevant for

hemocompatibility. The adsorption of these proteins was investigated as well as their activity

upon surface adsorption. Protein adsorption to SAM surfaces was studied well during the last

decades [6, 129, 130] revealing differences in adsorption patterns depending on surface

wettability. Adsorption processes additionally strongly depend mostly on experimental

parameters like temperature, deposition time, protein concentration and the presence of

competitive proteins or other cells explaining the necessity for the application of more than

one unique test for these investigations.

3.3.1 Human fibrinogen/ fibrin

The following chapter describes different results on fibrinogen adsorption to SAM surfaces.

Several authors found out that hydrophilic surfaces adsorb lowest amount of fibrinogen, while

its adsorption is elevated on hydrophobic surfaces [16, 131]. To get a realistic picture of

protein deposition processes on SAM surfaces, different tests were conducted applying

piezoelectric analysis using a microbalance as well as immunochemical and radiolabeling

techniques. For the following experiments a fibrinogen solution of 1mg/ml in HEPES buffer

was used, whereas surface incubation time was varied.

Quartz crystal microbalance (QCM)

The adsorption of fibrinogen using fibrinogen solution was determined by QCM analysis

(Figure 19). This method enables to study the thickness of a protein layer after its incubation

with the surface. Protein adsorption was determined using single protein solution after a

period of 20 minutes and significantly higher values for 100% -CH3 (13 nm ± 0.7) were found

compared to 100% -COOH (10.9 nm ± 1.3) (P<0.05).

Page 54: Dissertation Marion Fischer - Qucosa

3 Results

47

100% CH3

100% COOH0

2

4

6

8

10

12

14 *T

hick

ness

of p

rote

in la

yer

[nm

]

Figure 19 Detection of fibrinogen adsorption after 20 min from fibrinogen solution measured by QCM

(Statistical evaluation by ANOVA with subsequent Tukey test: ´*´100% -CH3 is significantly different to 100% -

COOH (P<0.05)

Immunostaining

Immunochemical methods are used to detect surface adsorbed proteins by antibody mediated

binding to specific proteins. By using antibodies labelled with a fluorescent dye, the amount

of the adsorbed protein can be estimated via fluorescence intensity measurements. Fibrinogen

adsorption of SAMs determined by immunostaining after 20 minutes scaled with surface

hydrophobicity showing minimal adsorption to hydrophilic surfaces either terminated with -

OH or -COOH surface groups (Figure 20). Fibrinogen adsorption to the methylated surface

was significantly higher than on 100% -COOH and 100% -OH, whereas adsorption values for

83% -COOH are significantly different from 100% -OH.

Page 55: Dissertation Marion Fischer - Qucosa

3 Results

48

CH3

83% COOH COOH OH0

20,000

40,000

60,000

80,000

100,000

120,000

140,000

**

FIT

C-A

nti-f

ibrin

ogen

[LA

U]

*

Figure 20 Detection of adsorbed HFG after immunostaining measured by fluorescence scanning after 20 min,

LAU are luminescence absorbance units (Statistical evaluation by ANOVA with subsequent Tukey test:

´*´100% -CH3 is significantly different to 100% -COOH and 100% -OH; ´**´83% -COOH is significantly

different to 100% -OH (P<0.05)

Adsorption of 125I-HFG

Protein adsorption was further studied by radiolabeling techniques, where the specific protein

is labelled with a radioactive marker. Surface adsorbed radioactivity can be detected by

phosphorimaging techniques. Fibrinogen adsorption from whole plasma spiked with

radiolabelled fibrinogen (was measured by spiking with radiolabelled fibrinogenFigure 21)

and was found to be low (< 0.6 µg/cm²) after short incubation times (5 min) on all tested

surfaces (-CH3, -COOH and 83%-COOH SAMs). 100% -CH3 terminated SAM surfaces

adsorbed slightly more fibrinogen than the 100% and 83% -COOH surfaces. A significant

increase in fibrinogen adsorption was seen for incubation times longer than 30 min on 100% -

CH3 surfaces (4.3 µg/cm² at 120 min), whereas no differences in adsorption were seen

between 100% -COOH and 83% -COOH surfaces at all time-points.

Page 56: Dissertation Marion Fischer - Qucosa

3 Results

49

0 20 40 60 80 100 120 140

0.4

0.6

456

Sur

face

fibr

inog

en [µ

g/cm

²]

incubation time [min]

CH3

83% COOH COOH

*

Figure 21 Fibrinogen adsorption to surfaces from plasma spiked with 125I, incubated for 5, 30 or 120 min. SAM

surfaces 100% -CH3, 100% -COOH and 83% -COOH were compared. (Statistical evaluation by Student

Newman Keuls: ´*´100% -CH3 is significantly different to 100% -COOH and 83% -COOH at 120 min

incubation time (P<0.005))

The results presented above showed protein adsorption studies of surfaces with varying

wettability. All the different methods that were applied (QCM, immunochemistry and

radiolabeling) uncovered significantly higher fibrinogen adsorption to the very hydrophobic

surface (100% - CH3) compared to the hydrophilic 100% -COOH being in accordance with

recent studies.

Detection of fibrinogen denaturation

Besides the amount of adsorbed fibrinogen also its orientation, conformation and functionality

upon adsorption is of great importance for the materials hemocompatibility [29, 30].

To determine differences in the conformation or orientation of the adsorbed fibrinogen and to

evaluate the possibility of thrombin-fibrinogen interactions, the binding of the site-specific

anti-fibrinopeptide A (anti-FPA) was assayed [132, 133]. Fibrinopeptides represent two small

peptide chains removed from the N-terminal segment of the alpha chains of fibrinogen by the

action of thrombin during the blood coagulation process. Each peptide chain contains 18

amino acid residues. Fibrinopeptide A is used as a marker to determine the rate of conversion

of fibrinogen to fibrin by thrombin. The accessibility of thrombin to FPA is required for the

conversion of adsorbed fibrinogen to fibrin.

Page 57: Dissertation Marion Fischer - Qucosa

3 Results

50

QCM-measurements of FPA on surface adsorbed Fibrinogen

QCM measurements was employed to characterize adsorption on hydrophilic and

hydrophobic SAM surfaces incubated using with a pure fibrinogen solution (Figure 22)

detected . We layer thicknesses to be 18.9 ± 0.3 nm for -CH3 surfaces, and 17.4 ± 0.5 nm for

-COOH surfaces. Subsequent binding of anti-FPA antibodies to preadsorbed fibrinogen

showed clear differences between the materials: the antibody binding, seen as increase in

layer thickness was elevated on -CH3 surfaces (21.5 ± 0.3 nm) compared to the -COOH

surfaces (18.1 ± 0.3 nm). Rates of conformationally changed fibrinogen are substantially

different for carboxylated and methylated surfaces. The layer thickness of anti-FPA (after

subtraction of the layer of preadsorbed fibrinogen) was 1.8 nm for 100% -CH3 and 0.6 nm for

100% –COOH.

CH3

COOH14

16

18

20

22

Laye

r th

ickn

ess

[nm

]

HFG Anti-FPA

*

Figure 22 Detection of fibrinogen adsorption from fibrinogen solution (1mg/ml in HEPES) and subsequent anti-

FPA incubation measured by QCM (Statistical evaluation by ANOVA with subsequent Tukey test: ´*´HFG

adsorption to 100% -CH3 is significantly different to 100% -COOH (P<0.005))

Detection of HFG and denatured HFG by immunostaining

The rate of conformationally changed fibrinogen on different SAM surfaces was further

detected by immunostaining using an antibody that detects conformationally changed

fibrinogen. Results revealed higher amounts of conformationally changed fibrinogen on 100%

-CH3 compared to carboxylated surfaces, but no significant differences.

Detection of fibrin formation on carboxylated and methylated surfaces showed elevated

values for the latter surface by QCM as well as immunochemical methods, but values did not

Page 58: Dissertation Marion Fischer - Qucosa

3 Results

51

reach statistical significances. The trend, however, seen in our experimental set ups is in line

with recent studies on fibrin formation, where significantly lower values for the negatively

charged -COOH surface were found [31]. The authors claim an influence of electrostatic

environment and conformational changes of fibrinogen on thrombin-fibrinogen interactions.

Evans-Nguyen et al. further found that hydrophobic surfaces most efficiently promote fibrin

proliferation, while the conversion of fibrinogen to fibrin at negatively charged surfaces was

surprisingly reduced [30]. The adsorption of fibrinogen at the negatively charged surface in a

conformation that is inert with respect to fibrin proliferation is discussed by the authors. It is

suggested that the conversion of surface-bound fibrinogen to fibrin is influenced considerably

by the surface properties of the underlying substrate.

3.3.2 Adsorption of complement fragment C3b

The adsorption of complement fragments onto SAM surfaces was studied by ellipsometric

measurements using single protein solutions. The deposition of complement fragments is an

important parameter for material induced host responses as it triggers complement activation

and subsequent induction of inflammation processes. Complement deposition was analysed

using polyclonal antibodies to detect surface bound C3b and its degradation fragments.

Complement C3b fragment was detected on surfaces following incubation with plasma for up

to 2 h. C3b binding to 100%-OH was detected after 10 min (78.7 ± 5.1 nm) and leveled off

after 60 min to reach a final thickness of 0 nm after 2 h (Figure 3). The initial C3b deposition

onto 100%-OH was significantly higher than on the other surfaces. No C3b was observed on

methylated surfaces at any time during incubation, whereas 100%-COOH showed a slow

increase after 60 min to 3 nm, where it remained for the rest of the 2 h incubation period (30.8

± 6.2 nm). To look at complement activation in the presence of competitive plasma proteins

as well as cell-derived mediators that could influence adhesion dynamics. C3b deposition on

SAMs was studied following 2 h of surface incubation in whole blood. C3b levels were

significantly higher on SAMs with high OH-ratios (100 and 90%-OH) compared to 50%-OH

and OH- free surfaces (Figure 4). Together our data indicate that complement activation is

elevated on OH containing surfaces.

Page 59: Dissertation Marion Fischer - Qucosa

3 Results

52

0 20 40 60 80 100 120

0

20

40

60

80

100

*

Thi

ckne

ss [A

]

time [min]

CH3

OH COOH

**

Figure 23 Adsorption of complement fragment C3b upon surface incubation with 66% plasma. Ellipsometric

detection by measuring thickness of protein layer after incubation with anti-C3c at 37°C (Statistical evaluation

by ANOVA with subsequent Tukey test: ´**´100% -OH at 10 min is significantly different to 100% -OH at 0. 60

and 120 min (P<0.05); ´*´ COOH at 120 min is significantly different to OH at 10 and 0 min (P<0.05), n=6 from

6 individual experiments)

High adsorption of C3b to hydroxylated surfaces as a reason for subsequent elevated

leukocyte adhesion is discussed in chapter 3.6.2.

3.3.3 Contact activation: factor XIIa and kallikrein acti vity

Kallikrein/ FXII activation are part of plasmatic events that initiate the intrinsic pathway of

coagulation. The induction of this pathway was studied in plasma after incubation of SAMs

consisting of C10-COOH/ C10-CH3, C10-COOH/ C11-OH as well as monolayers of EG3Ome/-

COOH terminated SAMs. FXII adsorption to surfaces induces its activation to FXIIa, what is

detected by a fluorogenic substrate for kallikrein and FXIIa. FXIIa activity can be measured

directly on the surface as well as in the liquid phase, where lower concentrations are found

because of dilution effects.

FXIIa on C10-COOH/ C10-CH3: Activation of FXII and kallikrein was enhanced with

increasing COOH ratio of the surface (Figure 24) indicating an impact of negatively charged

functional groups on contact activation as postulated by several authors [51, 53]. In the liquid

phase, the 100% -COOH surface showed a strong activation of FXII and kallikrein (Vmax =

0.0116 ± 0.0023 OD/min) that was significantly different to all other surfaces and almost as

high as the reference material glass (Vmax = 0.012 OD/min), that is known for its strong FXII

activation / kallikrein generation. This activity was reduced as the -COOH ratio of the surface

decreased to 83% (this value also being significantly different to all others), indicating that

Page 60: Dissertation Marion Fischer - Qucosa

3 Results

53

surface chemistry does impact contact activation in plasma. Surfaces with less than 50% -

COOH did not show contact system activity within the plasmatic phase (because of activity

dilution effects) but significantly higher activity of adsorbed FXII was measured on the

surface. This surface bound activity significantly decreased further as the surfaces approached

100% -CH3. On that surface activation levels were in the range of PTFE, which served as a

negative control with low influence on contact activation (Vmax= 0.0008 ± 0.0003 OD/min).

0 20 40 60 80 1000

2

4

6

8

10

++

++

plasmasurface

% COOH

Act

ivity

FX

IIa in

pla

sma

Vm

ax[O

D/m

in*1

00]

*+

40

60

80

100

120

**

Act

ivity

FX

IIa o

n su

rfac

e [r

el to

100

% C

OO

H]

Figure 24: Activity of FXIIa/kallikrein in plasma and on the surface after incubation on -CH3/-COOH and -

COOH/-OH-terminated SAMs (incubation time: 1 min). Vmax FXII in plasma for glass and PTFE was 0.012

and 0.0019 respectively. (Statistical evaluation by ANOVA with subsequent Tukey test: activity in plasma:

´**´COOH is significantly different to all other surfaces including binary SAMs of COOH/OH (P<0.001);

´*´83COOH is significantly different to 100% -COOH, 100% -CH3 and 50% -COOH/-CH3(P<0.05); activity on

the surface: ‘+’COOH is significantly different to 0%-COOH, 17%-COOH and 34%-COOH; ‘++’ 83%-COOH

and 50%-COOH are both significantly different to 0%-COOH and 17%-COOH (P<0.05).

FXIIa on C10-COOH/ C10-CH3 and C10-COOH/ C11-OH: Incubation of surfaces with blood

uncovered significant differences in FXII activation / kallikrein generation between surfaces

with high COOH ratios (83%-COOH and 100%-COOH) and all the other surfaces tested

(Figure 25). As already described above, contact activation in plasma was elevated on 83%-

COOH and showed the highest values on 100%-COOH (Vmax= 0.0083 ± 0.0005 OD/min).

Kallikrein detection dropped on 100%-OH surfaces to 0.0007 ± 0.0002 OD/min. No contact

activation was thus observed for 100%-OH and on our tested binary COOH/OH mixtures

(Vmax <0.0026 OD/min). The activation of the intrinsic pathway was low on surfaces

displaying only -OH or -CH3 groups as well as on 90%-OH and 50%-OH.

Page 61: Dissertation Marion Fischer - Qucosa

3 Results

54

0 20 40 60 80 1000.000

0.002

0.004

0.006

0.008

0.010

** OH CH

3V

max

kal

likre

in/F

XIIa

[OD

/min

]

% COOH of SAM

*

Figure 25 Kallikrein/Factor XIIa activity in plasma after 1 min plasma incubation of SAMs. Statistical evaluation by

ANOVA with subsequent Tukey test. ´**´COOH is significantly higher than all other surfaces including binary SAMs of

COOH/OH (P<0.001); ´*´83COOH is significantly higher than 100% -CH3 and 50% -COOH/-CH3 (P<0.05), n=9 from 3

individual experiments)

FXIIa on EG3Ome/-COOH terminated SAMs: The COOH-dependent change in FXII

activation was also found for EG3me0/-COOH terminated SAMs. Contact activation was

highest on 100% -COOH, activation levels decreased with lower surface –COOH ratios

having the lowest level on non-activating 100% EG3me0. Surfaces with less than 50% -

COOH did not show contact system activity in the liquid phase. Again here results indicate an

impact of surface chemistry on contact activation in plasma.

Page 62: Dissertation Marion Fischer - Qucosa

3 Results

55

0 17 34 50 100

0.01

0.02

0.03

0.04V

max

kal

likre

in/ F

XIIa

[OD

/min

]

% EG3meO (balance to COOH)

Figure 26 Activity of FXIIa and kallikrein in plasma (1 min) after incubation on -EG3me0/-COOH terminated

SAMs. Vmax of glass was 0.39 ± 5.8*10-5

Inhibition of contact activation by corn trypsin inhibitor

Inhibition of FXIIa activity was assessed to prove that material induced clotting processes are

solely initiated by contact activation. To verify the observed initiation of the intrinsic pathway

on carboxylated SAM surfaces, assays in standardised systems were used to prove the

specifity of FXII activation excluding thus the possibility of a parallel activation of other

coagulation factors (e.g. FX). For that purpose FXII activation in plasma was studied after

activation with known activators of the contact system (dextrane sulfate). The specifity of the

FXII inhibitor named corn trypsin inhibitor (CTI) in DS activated plasma was studied as well.

Inhibition of dextrane sulfate (DS) induced factor XIIa activity by different concentrations of

CTI:

DS is known for its activation of the contact system through its negatively charged sulfate

groups and is widely used to induce FXII activation in plasma. Our results showed that

activation of plasma by DS was possible yielding reaction kinetics of FXIIa (Vmax) of 0.002

OD/min being 100 times higher than initial Vmax of 1.9*10-5 OD/min for non-activated

plasma.

Page 63: Dissertation Marion Fischer - Qucosa

3 Results

56

0 10 20 40 60 120 160 240

0.0000

0.0005

0.0010

0.0015

0.0020V

max

kal

likre

in/F

XIIa

[OD

/min

])

Concentration of CTI [µg/ml]

Figure 27 FXIIa/ kallikrein activity assay of DS activated plasma and subsequent inhibition with different

concentration of CTI. Vmax of non-activated plasma was 1.9*10-5 OD/min

A concentration of 160 µg/ml of CTI was found sufficient to fully knock down FXII

activation and concentrations of at least 160 µg/ml were used for further studies using DS

activated plasma. At this inhibitor concentration Vmax of FXIIa was reduced to levels of 1/17

of activated, non-inhibited plasma (Vmax of 1.2*10-4 OD/min) and 10 fold higher than non-

activated plasma.

Inhibition of COOH induced FXIIa activity by CTI (240µg/ml):

Based on the established method for FXII inhibition that are described above. FXII inhibition

in plasma after surface incubation was studied (Figure 28). Negatively charged 100% -COOH

induced FXII activation of 0.021 OD/min being 23 fold higher compared to non-activated

plasma (0.001 OD/min). CTI addition of 240µg/ml to the activating 100% COOH lead to

decreased FXII activation of 0.001 OD/min being in the same range as non-activated plasma

and also below poorly activating 100% -CH3 (0.001 OD/min).

Page 64: Dissertation Marion Fischer - Qucosa

3 Results

57

Plasma COOH+ CTI

COOH CH3

0.000

0.005

0.010

0.015

0.020

0.025

Vm

ax k

allik

rein

/FX

IIa

[OD

/min

]

Figure 28 CTI (240 µg/ml) mediated inhibition of FXIIa in plasma after incubation with 100% -COOH and

100% -CH3

Inhibition of contact activation by infestin

A second specific inhibitor for FXII inhibition named infestin was tested. Infestin belongs to

the family of kazal inhibitors acting through irreversible binding of proteinases. Considering

the fact that CTI only inhibits FXIIa when being already incubated with non-activated FXII,

infestin was tried being possibly able to inhibit also the activated state of FXII.

A dose-dependent inhibition of FXIIa by infestin is seen in Figure 29. By activation of plasma

using dextrane sulfate FXII activation was induced (Vmax >0.08 OD/min vs. initial value of

0.001 OD/min). Subsequent activation of plasma with dextrane sulfate after addition of 3 µM

infestin showed sufficient FXIIa inhibition (Vmax < 0.002 OD/min). When adding infestin to

FXIIa activated by dextran sulfate, inhibition was not sufficient with still 50% Vmax compared

to initial values (OD/min >0.04). Incubation of FXII with infestin prior to activation was

shown to be essential to knock down contact activation (OD/min <0.005) indicating that

infestin is not able to sufficiently inhibit FXIIa in the activated state. Preincubation of blood

with infestin before activation by foreign surfaces is thus indispensable for suppression of

contact activation.

Page 65: Dissertation Marion Fischer - Qucosa

3 Results

58

PlasmaPlasma+DS

0.3 0.6 3 60.00

0.02

0.04

0.06

0.08V

max

kal

likre

in/F

XIIa

[OD

/min

]

CInfestin

[µM]

inhibition/activation activation/inhibition

Figure 29 Inhibition of FXIIa in plasma; inhibition/activation: addition of infestin before activation with DS

(dextrane sulfate); activation /inhibition: addition of infestin followed activation with DS

3.3.4 Thrombin

Thrombin formation was investigated on SAM surfaces to determine the plasmatic

coagulation activation and to see whether induced intrinsic pathway already seen by elevated

FXII/kallikrein activation on certain surfaces finally leads to the formation of the key enzyme

thrombin. Thrombin formation is measured using a fluorogenic substrate evaluating the onset

time. The onset time expresses the period of time upon surface incubation needed to

overcome a predefined level of plasmatic thrombin. Shorter thrombin onset times indicate

higher potential of plasmatic coagulation processes and can be related to surface properties.

Investigations on thrombin activation upon surface contact are described in the following

section as well as inhibition assays using specific FXII inhibitor CTI.

Activity of thrombin on SAMs

Thrombin formation, measured as the thrombin onset time was fastest for the 83% -COOH

surfaces after an incubation time of 3 min using PRP (36 ± 2.8 min). followed by PRP

incubated with 100% -COOH (47 ± 8.5 min). Incubation with -CH3 caused substantially

slower thrombin formation (58 ± 18 min). Thus, thrombin formation was fastest for the two

surfaces with the strongest contact activation. The faster thrombin formation in PRP on the

83% -COOH compared to the 100% -COOH surfaces can be attributed to a supporting effect

of platelet adhesion on thrombin formation. This conclusion is supported by the fact, that PPP

incubated with the different surfaces showed considerably slower peak thrombin formation

(>100 min), almost equal to the thrombin peak time for non-activated plasma.

Page 66: Dissertation Marion Fischer - Qucosa

3 Results

59

4 6 8 1020

40

60

80100120140160

t ons

et [m

in]

Incubation time on material surface [min] )

COOH PRP 83% PRP CH3 PRP COOH PPP 83% PPP CH3 PPP

Figure 30 Thrombin formation on surfaces after short time incubation with PPP or PRP. detection using

fluorogenic substrate

Inhibition of thrombin formation by CTI addition

Inhibition of FXII by CTI was shown in section 2.3.3. The effect of CTI addition to thrombin

generation was tested to elicit whether the observed FXII inhibition leads to a complete

reduction of the plasmatic coagulation. A thrombin activity assay of DS- activated plasma

inhibited with different concentrations of CTI was carried out. Results are presented in Figure

31 and reveal a dose dependent inhibition of thrombin formation by this specific inhibitor.

The addition of up to 40 µg/ml CTI did not prolong the onset time compared to CTI-free

activated plasma. CTI concentrations above 160 µg/ml clearly show a thrombin lag time of

>100 minutes.

Page 67: Dissertation Marion Fischer - Qucosa

3 Results

60

0 50 100 150 200 2500

20

40

60

80

100

120

t ons

et 1

000

cps

[min

]

Concentration of CTI [µg/ml]

Figure 31 Thrombin activity assay measured photo-spectroscopically using a fluorogenic substrate dependent on

addition of different CTI concentrations. Interpretation of the t onset time of 1000 cps.

Verification of non-initiation of thrombin formation after its specific activation by viper

venom (RVV):

Previous test on CTI inhibition confirmed its effect on FXII and on thrombin. To exclude the

possibility of unspecific inhibitory effects on FX, a specific activator of FX (RVV) was used

for plasma activation in a thrombin activity assay. Thrombin formation was clearly induced

by RVV (114 µg/ml), whereas non-activated plasma showed no to minor thrombin activation

during the whole incubation time. A strong activation was further found for RVV activated

plasma after addition of 240 µg/ml CTI. No difference between both RVV samples point to

the absent inhibition of FX by CTI. The specific inhibition of FXIIa and not of FX by CTI can

thus be concluded.

Page 68: Dissertation Marion Fischer - Qucosa

3 Results

61

3.4. Surface adhesion of platelets

Considering the impact of cellular responses on blood compatibility, this work aims at

investigating the adhesion of platelets to foreign surfaces. The adhesion of blood cells to

surfaces was studied after surface incubation with whole blood (see section 3.6) and with

platelet rich plasma (PRP). Platelet adhesion upon surface incubation using whole blood

uncovers a different picture of cellular behaviour than using PRP because in the latter case

platelets are able to react in the absence of other blood cells. Experiments described here

focus on adhesion studies using PRP, as these assays enable to specifically dissect platelet

adhesion and function.

3.4.1 LDH assay on SAMs after incubation with PRP

Platelet adhesion evaluated by measuring the level of platelet derived lactate dehydrogenase

after surface incubation with PRP correlated well to surface hydrophobicity (Figure 32).

following thus the same trend as fibrinogen adsorption. The most hydrophobic surface (100%

-CH3) showed a significantly higher coverage (8.4*105 platelets/ cm²) than all other tested

surfaces (P<0.5) and cell surface densities dropped with increasing COOH ratios. Basically no

platelets were found on the 100% -COOH surfaces (0 platelets/ cm²; value below detection

limit) and only 15% of the maximum cell density was seen on the 83%-COOH surface. Both

of the surfaces significantly differed from the surfaces with a 50% or less percentage of –

COOH surface groups. Maximum adhesion was found for 0 and 17% -COOH (83 and 100% -

CH3, respectively) with platelet adhesion being even higher than on the strongly hydrophobic

reference PTFE.

Page 69: Dissertation Marion Fischer - Qucosa

3 Results

62

0 20 40 60 80 1000.0

0.2

0.4

0.6

0.8

1.0

1.2

*

**+

Adh

eren

t pla

tele

tsre

lativ

e to

0%

-CO

OH

%-COOH

Figure 32 Adherent platelets on surfaces after incubation with citrated PRP for 45 min determined by

photometrical measurement of platelet derived LDH. Additionally to the SAM surfaces platelet and glass were

measured as references. (Statistical evaluation: ‘*’: 83 and 100% C10-COOH significantly different to 0, 17, 34

and 50%-COOH; ‘+’:0%-COOH significantly different to 34 and 50%-COOH; ‘**’:17% C10-COOH

significantly different to 50%-COOH, n=7 from 3 individual experiments)

3.4.2 Detection of platelets after immunostaining using fluorescence scanning

The scaling of platelet adhesion with surface hydrophobicity was also found by

immunofluorescent staining of the platelet specific CD 41 antigen (FITC conjugated); the

intensity of platelet derived CD41a was measured by a fluorescence scanner. Surface

incubation with platelet rich plasma showed increased cell density on 100% -CH3 and

minimal adhesion on the hydrophilic 100% -COOH and 100% -OH.

Page 70: Dissertation Marion Fischer - Qucosa

3 Results

63

CH3

COOH OH30000

60000

90000

120000F

ITC

-CD

41a

[LA

U]

Figure 33 Adherent platelets after incubation of surfaces with PRP determined after immunostaining with

CD41a measured by fluorescence scanning, LAU: luminescence absorbance units

In sum, our results using isolated plasma proteins and platelets uncovered that the highest

fibrinogen adsorption and fibrin formation occurs on hydrophobic -CH3 SAMs and correlates

with maximum platelet adhesion.

Page 71: Dissertation Marion Fischer - Qucosa

3 Results

64

3.5. Analysis of TF in leukocyte lysates

For the investigation of intracellular and cell bound leukocyte TF a western blot analysis of

leukocytes after surface incubation was established and optimised according to the

experimental requirements. This chapter contains results on optimisation of leukocyte

isolation and leukocyte lysis as well as the comparison of different treatments of the cell

lysates before loading the SDS page.

3.5.1 Optimisation of leukocyte lysis

The first step in western blot analysis of leukocytes is the isolation of the cells after surface

incubation. For that purpose leukocytes were withdrawn from blood using the commercially

available ERL-kit. Subsequent cell lysis was either done by addition of sodium dodecyl

sulfate (SDS) or by using RIPA-buffer.

Cell lysis using SDS: Loading buffer used for cell lysis contains SDS (0.01%) that was added

at very high concentrations to the leukocyte pellet for the induction of cell lysis (100 µl to the

leukocyte pellet). The appearance of a clearly defined band on the western blot membrane in

the range of the molecular weight of TF (at 47 kDa) was expected. Cell lysates ,however,

showed too many cleavage products of TF, what points to damage of the proteins during

handling of protein lysates (lane A in Figure 34). Probably the formation of cleavage products

of tissue factor was induced by the addition of the high amount of loading buffer/ SDS used in

the experiment. Probably also the absence of protease inhibitors lead to this cleavage effect.

RIPA buffer containing 1% protease inhibitor will be used in the next experiment.

Page 72: Dissertation Marion Fischer - Qucosa

3 Results

65

47 kDa

A B C

TF

Figure 34 Western blot of leukocyte lysates after leukocyte isolation by ERL-kit and application of different cell

lysis methods: A: cell lysis induced by SDS; B: cell lysis induced by RIPA-buffer; C: cell lysis induced by

RIPA-buffer, samples were kept at 4°C after cell lysis

Cell lysis using RIPA-buffer: The usage of RIPA buffer with protease inhibitor for cell lysis

was tried to avoid protein cleavage. Protein samples on the gel show smeary lanes but no

clear bands indicating a loss of proteins probably due to manipulation of cell lysates at room

temperature (lane B in Figure 34). The protocol including cell lysis by RIPA buffer and

adjustment of protein content before gel loading prolongs the time of cell manipulation

extremely. Keeping of samples strictly at 4°C has to be considered in that case and will thus

be guaranteed for the next assays, that are presented below.

Cell lysis using RIPA buffer as described above with strictly keeping samples at 4°C: Protein

samples of leukocyte isolates are finally visible in the upper region of the gel (lane C in

Figure 34). Cleavage as well as loss of proteins was successfully avoided but separation of

proteins by migration into the gel needs to be improved. Tertiary structure of proteins might

hinder protein migration into the gel, reducing loading buffer containing mercaptoethanol will

be used to cleave disulfide bonds leading to monomer formation, what might improve

separation of cellular proteins.

Page 73: Dissertation Marion Fischer - Qucosa

3 Results

66

3.5.2 Optimisation of gel-loading conditions

To clarify optimal gel loading conditions used for TF analysis by western blot, different

sources of recombinant TF (American diagnostica, Technothrombin) were used and separated

by SDS page. Processes carried out before loading of gels like heating and usage of reducing/

non-reducing loading buffers were tested (Figure 35). TF from American diagnostica (Figure

35, lane 6-9 and lane 15-18) showed various bands at different molecular weights, all bigger

than non-lipidated TF. This sample seems to include different forms of lipidated TF, where

the fatty acid residue increases the molecular weight of TF. The sample from technothrombin

shows bands characteristic for TF (at 47 kDa) and no other proteins with different molecular

weights in the sample (Figure 35, lane 2-5 and 11-14). Here conclusions for the optimisation

of gel loading conditions can be draw. Usage of non-reducing loading buffer shows proteins

with higher molecular weight indicating the presence of TF dimers, whereas reducing loading

buffer leads to cleavage of disulfidic bonds giving protein bands at 47 kDa, being

characteristic for monomeric TF.

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18

TF

Figure 35 Western blot of recombinant tissue factor at different gel loading conditions (see legend in Table 10)

Table 10 Legend for different gel loading condition used in experiment 3.5.2; M: marker, TT: TF from

Technothrombin, AD: TF from american diagnostica, Red LB: reducing loading buffer, heat: heating of samples

to 95°C before gel loading

Slot 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18

sample M TT TT TT TT AD AD AD AD M TT TT TT TT AD AD AD AD

Red

LB

+ + + + + + + + + - - - - - - - - -

heat + + - - + + - - + + - - + + - -

Page 74: Dissertation Marion Fischer - Qucosa

3 Results

67

3.5.3 Isolation of leukocyte isolation using Polymorphprep®

Optimisation of cell lysis and gel loading conditions that were described in the above sections

were considered in the following experiment using samples from whole blood incubation

experiments. Here Polymorphprep® was used for cell isolation. Protein samples of leukocyte

isolates stayed in the upper region of the gel (lane A in Figure 36). Reducing loading buffer

containing mercaptoethanol has enabled protein unfolding and cannot be considered as reason

for bad separation in that case. Pore sizes of the gel influence protein migration as well and

will be varied for the next experiment.

50 kDa

30 kDa

20 kDa

A M

Figure 36 Western blot of leukocytes isolated by Polymorphprep and cell lysis induced by RIPA-buffer.

polyacrylamide gel with 12% acrylamide

Gels with lower concentration of acrylamide (10% instead of 12%) were used to enable

protein migration into the gel because of bigger gel pore sizes. Determination of TF in cell

lysates of isolated leukocytes after whole blood incubation assay was carried out using

protocol optimisation described above showing bands characteristic for TF (molecular weight

at 47 kDa). Second band represents GAP-DH (36 kDa) used as loading control. The

experiment was done twice, results from the repetition experiment are presented in Figure 37.

More precise interpretation of western blot experiments focusing on blood surface

interactions are explained in section 3.6.2.

Page 75: Dissertation Marion Fischer - Qucosa

3 Results

68

TF

GAP-DH

20 kDa

30 kDa

40 kDa

50 kDa

M I 1 2 3 4 5 6 7

Figure 37 Western blot of leukocytes isolated by Polymorphprep and cell lysis induced by RIPA-buffer,

polyacrylamide gel with 10% acrylamide, samples: M: marker, I: initial, 1-3: C10-COOH, 4-6: C10-CH3, 7: C10-

OH

3.6. Whole blood incubation

Blood contact with foreign surfaces induces host responses that mostly impair the function of

the materials. Whole blood incubation assays are used to relate surface properties to their

hemocompatibility. Using this test a wide set of parameters including cellular and plasmatic

events can be determined. Blood incubation assays of SAM surfaces containing C10-COOH

and C10-CH3 are described here as well as of surfaces containing C10-COOH and C11-OH.

3.6.1 Whole blood incubation of -CH3/-COOH terminated SAMs

Alkanethiol based surface modification with -CH3 and -COOH terminated molecules enables

tailoring of surface hydrophobicity and charge. The influence of these parameters on whole

blood was studied in the following section.

Adsorption of fibrinogen/ fibrin measured by immnochemistry using an anti-HFG antibody

The correlation of fibrinogen adsorption to surface hydrophobicity as seen in adsorption

studies using fibrinogen solution (see section 3.3.1) was also found after whole blood

incubation assays. The amount of surface adsorbed fibrinogen measured by immunochemistry

on 100% -CH3 was substantially different to 100% –COOH (3.29±1.5 vs. 3.16±0.5).

adsorption to 83% -COOH ranked between both extreme surfaces (2.3±0.5).

Page 76: Dissertation Marion Fischer - Qucosa

3 Results

69

In vitro blood incubation assay

Coagulation activation as determined by TAT formation (thrombin-antithrombin complex,

Figure 39) were neither found on the surface with the highest contact activation (100% -

COOH) nor on the SAMs showing maximum platelet adhesion (100% - CH3) but levels were

significantly elevated on the binary 83% -COOH. TAT levels were high on 50% to 83% -

COOH with a maximum at 83% -COOH, which was more than 10 times the activation on 0%

-COOH (-CH3) surfaces. Surfaces with 100% -COOH and –COOH ratios <83% were

associated with baseline TAT levels. Since it was found that 100% and 0% -COOH surfaces

have distinctively different qualities concerning platelet adhesion and contact activation, this

was unexpected.

0 20 40 60 80 100

1

10�

*

TA

T [r

el to

0 %

CO

OH

]

% COOH

Figure 38 TAT formation in plasma after whole blood incubation of SAMs detected by ELISA assay * P<0.001

different to all other surfaces � P<0.05 different to all other surfaces

Platelet factor 4 (PF4) is a very sensitive marker for platelet degranulation. Our results for

PF4 strongly correlated to those for TAT (Figure 38). PF4 release was significantly elevated

on the 83% -COOH surface, indicating high platelet activity. PF4 formation peaked on 83% -

COOH as well (1,516 IU/ml vs. levels below 480 IU/ml on 100% -CH3 and 100% -COOH).

Surfaces with 0 to 50% -COOH induced a moderate release of PF4, while the 100% -COOH

surface had a tendency toward lower values (app. half the value of 0% -COOH). Blood cell

counts also confirmed a large reduction in platelets (27.4%) after incubation with 83% -

COOH surfaces (Figure 39 right).

Page 77: Dissertation Marion Fischer - Qucosa

3 Results

70

0 20 40 60 80 100

0

2

4

6 PF4 Platelets on surface

% COOH

PF

4 [r

el to

0%

CO

OH

]

0.02.0x10

54.0x10

5

Platelets on surface/m

m2

0 20 40 60 80 100

0

10

20

30

40

Dec

reas

e of

pla

tele

t num

ber

(rel

ativ

e to

initi

al)

% COOH

Figure 39 left: Platelet factor 4 (PF4) in plasma after whole blood incubation assay measured by ELISA and

surface platelet adhesion determined by SEM; right: Decrease of platelet number in plasma in percent relative to

initial platelet count after whole blood incubation determined by blood cell counter

Fluorescence microscopy and SEM analysis allowed the identification of cells adhering to the

surfaces as platelets (FITC conjugated anti-CD41a, green fluorescence; Figure 40). Surface

analysis uncovered clot formation on 83% -COOH, whereas almost no platelets and only

small amounts of leukocytes adhered to 100% -COOH. The hydrophobic 100%-CH3 was

confluently covered by platelets and only minor leukocyte adhesion. Adherent platelets were

counted following SEM analysis (Figure 39 left) revealing higher values on -CH3-terminated

SAM surfaces (151*103 cells/mm²) than on -COOH terminated SAMs (29x103 cells/mm²).

This was similar to the behaviour in PRP where platelet adhesion was shown to be a direct

function of the -CH3:-COOH ratio. Contrasting to this direct correlation platelet presence after

whole blood incubation was highest on the 83% -COOH surface (app. 190x103 cells/mm²).

Given that the platelets on 83% -COOH were embedded in a blood clot, this difference in

results can likely be attributed to an interaction between the platelets and the coagulation

process occurring in addition to that with the surface.

Page 78: Dissertation Marion Fischer - Qucosa

3 Results

71

83% -COOH50% -COOH 100% -COOH

100% - CH3 17% -COOH 34% -COOH

83% -COOH50% -COOH 100% -COOH

100% - CH3 17% -COOH 34% -COOH

Figure 40 Fluorescence microscopic analysis of SAM surfaces with rising content of COOH terminated thiols

(diminishing content of CH3 terminated thiols) after incubation with whole blood (FITC-labelled anti-CD 41a)

The adhesion of leukocytes was also evaluated showing an absence of white blood cells on

100% -COOH surfaces (DAPI-staining of leukocyte DNA, blue fluorescence; Figure 41 left).

In comparison to binary layers of -COOH and -CH3, less leukocytes adhered on 100%-CH3,

but no significant differences were seen. The markedly enhanced adhesion seen on 83% -

COOH surfaces can be attributed to physical entrapment during clot formation. Leukocyte

activation as marked by CD11b expression did not differ significantly for the tested SAM

surfaces, although the amount of conjugates of granulocytes and platelets was highest for the

83% -COOH surface (2.4±1.5 on 83% -COOH vs. 1.0±0.3 on 100% -CH3 and 0.5±0.5 on

100% -COOH) (Figure 41 right). Complement activation was not significantly different

between the tested surfaces when looking for C5a in the plasma or for C3b on the surfaces

(data not shown).

Page 79: Dissertation Marion Fischer - Qucosa

3 Results

72

0 20 40 60 80 1000

5

10

15

20

25Le

ukoc

ytes

[rel

to 0

% C

OO

H]

% COOH0 20 40 60 80 100

0

1

2

3

4 CD11b conjugates

CD

11b

[rel

to 0

% C

OO

H]

% COOH

0

2

4

6

8

10

%

Con

juga

tes

of

gran

uloc

ytes

[rel

to 0

%]

Figure 41 left: Surface adherent leukocytes following whole blood incubation determined by fluorescence

microscopy after DAPI staining. right: Activation of leukocytes and formation of conjugates after whole blood

incubation assay determined by FACS analysis using FITC-anti-CD11b and PE-anti-CD41a respectively

Mimicking and knockout studies: the impact of corn trypsin inhibitor (CTI) and

phospholipids (PL) on blood reactions

An additional blood incubation set-up using either CTI to selectively inhibit FXIIa or PL to

mimic the surface of activated platelets was employed to dissect the synergistic effects seen

with our surfaces containing both carboxylic surface groups and adherent blood platelets. CTI

addition significantly lowered the formation of thrombin (TAT) on 83%-COOH to levels

below the negative control being 100%-CH3 (TAT on 83%-COOH: 213 µg/l with CTI

addition versus 1,400 µg/l without CTI) (Figure 42). Additionally, preincubation of blood

with CTI before incubation with binary 83%-COOH surfaces significantly reduced PF4

formation (0.2 IU/ml with CTI addition versus 1,500 IU/ml) (Figure 42, right). This data was

confirmed by microscopic analysis which showed an absence of clotting on 83%-COOH

(Figure 43). The presence of CTI did not change PF4 levels on 100% –COOH surfaces,

indicating that the residual platelet activation that was detected did not depend on contact

activation. CTI addition to 100% -CH3 was not analysed because of absent FXII activation

already in plasma without additives (see section 3.3.3).

The addition of PL to whole blood before incubation did not influence TAT levels for the 0%

and 83%-COOH surface. On the 100% -COOH-terminated surfaces the addition of PL

induced significantly elevated thrombin formation (TAT: 1,152 µg/l vs. 827 µg/l in whole

blood without additive) as well as higher PF4 levels (PF4: 829 IU/ml vs. 467 IU/ml

respectively). PF4 release after Pl addition was decreased on the 83% -COOH surface while

no effect was seen for the 0% -COOH surface. The presence of platelets was enhanced more

than 3 times while the decrease of platelets in blood was 6±7 % for -COOH compared to

25±4 % for -COOH+PL (P=0.003, t-test). Aggregation of cells was further found on 100%-

COOH after phospholipid addition. The detection of complement fragment C5a in plasma

using ELISA after whole blood incubation unravelled no differences between additive free

Page 80: Dissertation Marion Fischer - Qucosa

3 Results

73

blood incubation and whole blood preincubated with PL, whereas there is a slight increase in

complements activation when preincubating blood with CTI (data not shown).

100% CH3

83% COOH 100% COOH0

500

1000

1500

2000

** ***

no addition + PL + CTI

TA

T [µ

g/l]

* *

100% CH3

83% COOH 100% COOH0

500

1000

1500

2000

2500

3000

**

* *

no addition + PL + CTI

PF

4 [IU

/ml]

Figure 42 left: TAT levels after whole blood incubation of SAMs; no addition: without any additives,

+PL/+CTI: addition of phospholipids/corn trypsin inhibitor to whole blood just before whole blood incubation

assay; * 83% -COOH is significantly different from 100% -CH3 and 100% –COOH in the case of whole blood

incubation without additives, ** 83% -COOH is significantly different from 83% -COOH pre-incubated with

CTI, *** 100% -COOH is significantly different from 100% -COOH pre-incubated with PL; (P<0.05 for all

statistics), right: PF4 levels after whole blood incubation of SAMs; normal: without any additives, +PL/+CTI:

addition of phospholipids/corn trypsin inhibitor to whole blood just before whole blood incubation assay; * 83%

-COOH is significantly different from 100% -CH3 and 100% –COOH in the case of whole blood incubation

without additives, ** 83% -COOH is significantly different from 83% -COOH pre-incubated with CTI; (P<0.05)

for all statistics

Whole blood +CTI +PL

CH3

83COOH

COOH

Figure 43 SEM and fluorescence microscopy of SAM surfaces after whole blood incubation. Fluorescence

microscopy: Staining of platelets with FITC-anti CD 41a und DAPI staining of leukocyte nuclei

Page 81: Dissertation Marion Fischer - Qucosa

3 Results

74

Summing up the addition of CTI to whole blood just before surface incubation showed similar

cell adhesion patterns as seen for 100%-CH3 incubated with pure whole blood. In contrast,

addition of phospholipids to 100% -COOH equals the picture of additive-free whole blood on

83% -COOH.

Page 82: Dissertation Marion Fischer - Qucosa

3 Results

75

3.6.2 Whole blood incubation of -CH3/-COOH and -COOH/-OH terminated

SAMs

A second type of surface modification was applied to address the question of influences of

surface properties on the induction of the extrinsic pathway. By using mixtures of -CH3/-

COOH and -COOH/-OH terminated SAMs, the possible correlation of cell adhesion/

activation and TF release was studied. In the following section the influence of surface

hydroxylation is inter alia compared to influences by surface wettability and charge, so some

results from the previous chapter are partly presented again for comparison reasons.

To test whether an induction of the intrinsic pathway occurs in parallel to TF activation,

kallikrein/ FXIIa activation in plasma was assayed. The data is presented in section 3.3.3. It

was found that the activation of the contact system declined as ratios of -CH3 and –OH

terminated thiols on the surfaces increased. No contact activation was thus observed for

100%-OH and on our tested binary COOH/OH mixtures. The activation of the intrinsic

pathway was low on surfaces displaying only -OH or -CH3 groups as well as on 90%-OH and

50%-OH. It is thus unlikely that procoagulant activities on those surfaces are due to FXIIa.

In addition to analysing the adsorption of FXII, also the adsorption of complement fragments

onto SAM surfaces was studied The deposition of complement fragments reflect the degree of

immunological reactions against foreign materials, that are governed by non-specific,

lymphocyte independent reactions. Surface bound C3 molecules were determined by an

immunosorbent method, where anti-C3c also detects surface bound C3b and its degradation

fragments. Detection of complement fragment C3b following plasma incubation showed

significantly increased initial C3b deposition onto 100%-OH compared to 100% -COOH and

100% -CH3 (see section 3.3.2).

To assay the complex interplay of cellular and plasmatic responses, SAM surfaces after whole

blood incubation were analysed. C3b deposition on SAMs was studied following 2 h of

surface incubation with whole blood to evaluate complement activation in the presence of

competitive plasma proteins and cell-derived mediators that could influence adhesion

dynamics. C3b levels were significantly higher on SAMs with high OH-ratios (100 and 90%-

OH) compared to 50%-OH and OH- free surfaces (Figure 44). Together our data indicate that

complement activation is elevated on OH containing surfaces.

Page 83: Dissertation Marion Fischer - Qucosa

3 Results

76

0 20 40 60 80 1001.0

1.2

1.4

1.6

1.8

2.0

*

% COOH of SAM

OH CH

3C

ompl

emen

t fra

gmen

t C3b

[a.u

.]

Figure 44 Complement activation: Detection of complement fragment C3b on SAM surfaces after whole blood

incubation (2 hours) (Statistical evaluation by ANOVA with subsequent Tukey test: ´*´100% -OH is

significantly different to 100% -COOH and 50% -OH (P<0.05), n=8 from 2 individual experiments)

Platelet adhesion and activation (PF4) were low on 100%-COOH, 100%-OH and mixtures of

COOH and OH, whereas both were elevated on 50%-COOH and 83%-COOH (Figure 45).

0 20 40 60 80 1000.1

1

10

100

1000

10000

% COOH of SAM

OH CH

3

Pla

tele

ts o

n su

rfac

e(r

elat

ive

to 1

00%

CO

OH

)

*

0 20 40 60 80 1000

2

4

6

8

10

% COOH of SAM

OH CH

3

PF

4 [IU

/ml]

*

Figure 45 left: Platelet adhesion on surface determined by SEM (Statistical evaluation by Dunn’s method:

´*´100% -COOH is significantly different to all other surfaces including binary SAMs of COOH/OH (P<0.05).

n=6 from two individual experiments) right: PF4 release into plasma (right) after whole blood incubation assay

(2 hours) (Statistical evaluation by Dunn’s method: ´*´83% -COOH is significantly different to initial value

(P<0.05), n=5 from two individual experiments)

Leukocyte adhesion on mixed COOH-/OH- terminated SAMs increased with the level of OH-

terminated thiols, peaking at 100%-OH (1,174 leukocytes/mm²; significantly different to

100%-COOH and 50%-OH). The lowest adhesion was detected on 100%-COOH (36

leukocytes/mm²; significantly different to all other surfaces) (Figure 46 left). The high

Page 84: Dissertation Marion Fischer - Qucosa

3 Results

77

number of leukocytes on 50%-COOH and 83%-COOH surfaces in a binary CH3 mixture

(1,461 and 2,940 leukocytes/mm² respectively) can be explained by clot formation, as

described in our recent publication [134]. Cells on such surfaces adhere largely as the result of

following chemoattractants secreted by clot forming cells and by passive incorporation into

the forming fibrin mesh, rather than by attraction to the surface.

Leukocyte adhesion to binary COOH/OH-terminated surfaces correlated well with leukocyte

activation measured by CD11b expression (Figure 46 right). Significantly higher activation on

100%-OH and 90%-OH SAMs was found in comparison to acidic 100%-COOH. Further,

initiation of the complement cascade showed a significant increase of complement fragments

C3b/C5a on 100% -OH in comparison to 50%-OH and 100%-COOH (C5a in plasma and C3b

on the surface; Figure 47 and Figure 44 respectively).

0 20 40 60 80 1001

10

100+

**

% COOH of SAM

OH CH

3

Leuk

ocyt

es o

n su

rfac

e(r

elat

ive

to 1

00%

CO

OH

)

*

0 20 40 60 80 1000.5

1.0

1.5

2.0

2.5

3.0

*

% COOH of SAM

OH CH

3

Gra

nulo

cyte

CD

11b

(rel

ativ

e to

100

% C

OO

H)

**

Figure 46 left: Number of leukocytes adherent on surfaces determined by fluorescence microscopy after DAPI

staining of cell nuclei; (Statistical evaluation by Dunn’s method: ´*´100% -COOH is significantly different to all

other surfaces including binary SAMs of COOH/OH; ´**´100% -OH is significantly different to 100% -COOH

and 50% -OH; ´+´83% -COOH is significantly different to 100% -COOH, 50% -COOH/CH3 and 100% -CH3

(P<0.05), n=6 from 4 individual experiments Right: Activation of leukocytes (expression of CD11b) evaluated

by flow cytometric analysis, both after whole blood incubation assay (Statistical evaluation by Dunn’s method:

´*´100% -OH and ´**´90% -OH are significantly different to 100% -COOH (P<0.05). n=12 from 4 individual

experiments

Page 85: Dissertation Marion Fischer - Qucosa

3 Results

78

0 20 40 60 80 100

0

4

8

12

16

**

*

% COOH of SAM

OH CH

3C

5a(r

elat

ive

to 1

00%

CO

OH

)

Figure 47 Detection of complement fragment C5a in plasma using ELISA after whole blood incubation (2

hours) (Statistical evaluation by Dunn’s method: : ´*´100% -OH is significantly different to 100% -COOH and

50% -OH (P<0.05);´**´90% -OH is significantly different to 100% -COOH (P<0.05), n=10 from 4 individual

experiments).

Almost no leukocytes where found on 100%COOH whereas the number of adherent

leukocytes increased with higher OH ratio in SAMs. Mixture of CH3 and COOH showed

highest platelet adhesion and leukocyte attachment only to moderate extend. Existence of

leukocyte-granulocyte aggregates in blood incubated on CH3/COOH surface as well as on

100% CH3 was confirmed by FACS analysis.

RT-PCR of mRNA was done to study de novo synthesis of TF by leukocytes upon material

contact. As the duration of blood incubation increased, also increasing amounts of TF-mRNA

in leukocytes were found after incubation with all tested surfaces (Figure 48). Thus, material-

induced synthesis of cellular TF appears to continue at late time points. At all times, TF

expression on 100%-OH was 2-3 fold higher than on 100%-CH3, 100%-COOH and 83%-

COOH. After 3 h of incubation, the relative expression on OH-free surface was less than 200

times the initial value, whereas on OH-terminated SAMs a significant increase in TF

expression was observed, being 590-times the initial value (Figure 1). Surfaces with

uncharged hydrophilic OH- surface groups showed a significant impact on TF expression

compared to hydrophobic and negatively charged surfaces. Levels of mRNA thus scaled with

the amount of surface OH groups.

Page 86: Dissertation Marion Fischer - Qucosa

3 Results

79

0

100

200

300

400

500

600

700

C H 3 83C OOH C OOH OH

Rel

ativ

e T

F e

xpre

ssio

n 1h2h

3h

0 20 40 60 80 100

0

100

200

300

400

500 OH CH

3

Rel

ativ

e m

RN

A e

xpre

ssio

n

% COOH of SAM

*

Figure 48 Expression of tissue factor on SAMs after whole blood incubation assay relative to initial value; left:

expression after 1, 2 and 3 hours of blood incubation; right: expression after 2 hours of blood incubation,

endotoxin induced expression was 2629 (SD: 234) (Statistical evaluation by ANOVA with subsequent Tukey

test: ´*´100% -OH is significantly different to all other surfaces including binary SAMs of COOH/OH (P<0.001)

Next the location and quantity of TF was elucidated after blood incubation of SAMs using

fluorescence imaging (Figure 49). TF was clearly associated with monocytes as well as

granulocytes. Granulocyte-platelet aggregate formation was observed by SEM and flow

cytometry, with no significant differences found between the materials (data not shown).

Figure 49A shows CD14 positive cell fragments that possibly represent monocyte derived

microparticles. TF formation, as detected by staining with FITC-labelled anti-TF antibody,

increased with OH/COOH-ratios (Figure 50). The pronounced TF presentation on OH-

containing surfaces is consistent with the high TF mRNA expression that was observed on

surfaces with high OH-ratios (90%-OH or 100%-OH). Based on the data on surface TF

obtained by fluorescence imaging TF/leukocyte ratios were calculated to compare TF levels

to the number of adherent cells. The highest ratio was found on 100%-COOH, where the

lowest cell adhesion was observed, followed by 100%-CH3 and SAMs containing OH groups

Figure 51).

Page 87: Dissertation Marion Fischer - Qucosa

3 Results

80

Figure 49 left: Fluorescence imaging of surface adherent leukocytes/ cell fragments and tissue factor; A: DAPI

staining of leukocyte nuclei (blue), FITC-anti TF staining (green) and PerCP-Cy5-anti CD14a (red). B Left:

DAPI staining of leukocyte nuclei (blue). Right: additional FITC-anti TF staining of cells (green) adherent on

surface taken at the same spot on 50%-OH. Inset: Granulocyte-platelet-formation on–100%-OH detected by

SEM.

Figure 50 FITC-anti TF (green) and DAPI staining of leukocyte nuclei (blue) on 100% -COOH, 50%-OH/-

COOH; 90%-OH/-COOH and 100% -OH (left to right)

100% COOH 50% OH 90% OH 100% OH

A

Page 88: Dissertation Marion Fischer - Qucosa

3 Results

81

0 20 40 60 80 1000

2

4

6

8

10

12

14

16

18 - OH - CH

3

+

**

Sur

face

TF

[LA

U]

(rel

ativ

e to

100

% C

OO

H)

% COOH of SAM

*

CH3

83COOH COOH 50OH 90OH OH0.0

0.2

0.4

0.6

0.8

1.0

TF

/Leu

kocy

te r

atio

Figure 51 left: TF on cell surface. Intensity of fluorescence measured by fluorescence scanning after FITC-anti

TF staining on SAM of -COOH/-OH (Statistical evaluation by Dunn’s method: : ´*´100% -OH is significantly

different to 100% -COOH and 50% -OH (P<0.05); ´*´90% -OH is significantly different to 100% -COOH

(P<0.05);´+´83% -COOH/-CH3 is significantly different to 100% -COOH (P<0.05), n=7 from 4 individual

experiments) right: TF/leukocyte ratio showing the relation of surface TF to the number of adherent leukocytes.

To analyse cellular TF of non-adhering cells after material contact, TF positive granulocytes

and monocytes were detected by flow cytometry (Figure 52). A substantially higher TF

content was found on monocytes that had been exposed to 100%-OH surfaces, while mixtures

of CH3-/COOH- resulted in less TF on both cell types.

The presence of TAT reflects the degree of activation of coagulation. TAT-levels were not

increased on surfaces with high OH-ratios (Figure 53). This was unexpected given the

elevated TF expression on 90%-OH/COOH and 100%-OH surfaces. Western blot analysis of

protein extracts obtained from isolated leukocytes after whole blood incubation revealed no

significant difference in intercellular TF between COOH-/OH- or CH3-containing SAMs

(Figure 54). Significant differences, however, were seen between intracellular TF levels in

blood after incubation of 100%-OH surfaces compared to the initial TF amount of leukocyte

protein extracts.

Page 89: Dissertation Marion Fischer - Qucosa

3 Results

82

0 20 40 60 80 100

0.6

0.8

1.0

1.2

% COOH of SAM

OH CH

3

Gra

nulo

cyte

TF

[RF

U]

0 20 40 60 80 100

0.8

1.0

1.2

1.4

% COOH of SAM

OH CH

3

Mon

ocyt

e T

F [R

FU

]

Figure 52 Flow cytometry analysis of tissue factor presence associated with granulocytes (left) or monocytes

(right) on mixtures of CH3/COOH and COOH/OH-terminated SAMs after whole blood incubation assay (n=9

from 3 individual experiments for mixtures of 100%, 90% and 50% -OH and 100% -COOH and n=3 from 1

experiment for the rest)

0 20 40 60 80 100

1

10

% COOH of SAM

OH CH

3

TA

T(r

elat

ive

to 1

00%

CO

OH

)

*

Figure 53 TAT formation in plasma after whole blood incubation assay (Statistical evaluation by Dunn’s

method: ´*´83% -COOH is significantly different to 100% -COOH and 100% -CH3 (P<0.05), n=at least 6 from 2

individual experiments)

Page 90: Dissertation Marion Fischer - Qucosa

3 Results

83

-20 0 20 40 60 80 1000

1

2

3

4

5

6

TF

ban

d in

tens

ities

re

lativ

e to

GA

PD

H

initial OH CH

3

*

% COOH of SAM

Figure 54 Western blot of leukocyte derived protein lysate after 2 h whole blood incubation assay, primary

antibody: anti TF, detection by chemiluminescence assay left: lane plot of samples where upper lane is TF and

lane below represents loading control GAPDH; I=initial, 1=100%-CH3, 2=83%-COOH, 3=100%-COOH,

4=500%-OH, 5=90%-OH, 6=100%-OH; right: Evaluation of lane intensities done with ImageG software

(Statistical evaluation by Dunn’s method: ´*´100% -OH is significantly different to initial value (P<0.05), n=at

least 5 from 2 individual experiments)

In summary, our data indicate that different surface functionalities induce different plasmatic

and cellular events. Surfaces displaying 100% -CH3 or 100% -COOH groups either showed

strong platelet adhesion or high contact activation, but both monolayers stimulated neither TF

expression nor plasmatic coagulation after whole blood incubation. In contrast, hydroxylated

surfaces induced high TF expression and cell surface TF levels. Leukocyte-derived TF

mRNA scaled with the fraction of surface –OH/-COOH groups, and the amount of surface TF

on SAMs was significantly higher with greater –OH/-COOH ratios, i.e. with increasing

hydroxylation. These results correlated with leukocyte adhesion to the SAM surfaces. The

number of leukocytes on the surface scaled with -OH/-COOH ratios and was highest on

SAMs composed only of hydroxyl groups (100%-OH). Surfaces containing OH groups

showed not only higher leukocyte attachment but also positive expression of activation

marker CD11b. These SAMs where also found to activate the complement system by

induction of C5a formation. TF transcription further correlated with TF presence on

leukocytes and granulocyte activation.

Page 91: Dissertation Marion Fischer - Qucosa

4 Discussion

84

4. Discussion

The discussion is composed of two parts focussing either on the results of surfaces with

graded hydrophobicity and charge (SAMs containing C10-COOH and C10-CH3) or with

graded degree of surface hydroxylation and charge (surfaces of C10-COOH and

C11-OH).

In the first part of the discussion the focus is set on initial processes of blood

coagulation on a hydrophilic, negatively charged surface (100% -COOH), on a

hydrophobic surface (100% -CH3) and on surfaces combining these two properties. The

experimental focus here was set on protein adsorption and cell adhesion, as well as on

the resulting coagulation processes. Surface specific processes are summarized in a

simple form in Figure 55 and described more detailed in the section below.

Protein adsorption and platelet adhesion: A strong fibrinogen adsorption to

hydrophobic 100% -CH3 demonstrated here has been described also by other authors

[16, 30, 131] and is regarded as an elicitor for the high platelet adhesion on that surface

after incubation with PRP. These results confirm previously described findings for

methyl-terminated SAMs after plasma pre-adsorption [16]. In contrast, the acidic

surface was mainly free of blood platelets possibly resulting from electrostatic

repellence [135-137]. However, the relevance of electrostatic interaction under

physiological conditions is questionable as Coulomb forces are limited under high ionic

strength. The role of competitive protein adsorption first described by Vroman [26]

seems to be more plausible, where surface adsorbed fibrinogen gets replaced by

HMWK.

Contact activation: FXII activation in plasma determined by activity assays was found

to correlate to the amount of negatively charged surface groups as reported previously

[51, 53, 57]. It was proposed that the negative charge density influences FXII

conformational changes upon surface adsorption leading to a switch of function [52].

Initiation of the intrinsic pathway of coagulation is mediated by auto-activation of the

clotting factor FXII to FXIIa upon surface adsorption to negatively charged

(polyanionic) surfaces due to an anion-binding exosite [34, 53]. There is a strong

relevance of this reaction for biomaterials although the responsible triggering

mechanisms are still not clear in detail. Negative surfaces favour the process, however

protein displacement of competing proteins might be responsible, instead of

electrostatic interaction with FXII [56, 138]. The actual relevance of FXII activation

clinically has been strongly questioned yet a strong significance for thrombus stability

Page 92: Dissertation Marion Fischer - Qucosa

4 Discussion

85

has been shown [58]. It is generally considered, that coagulation on foreign surfaces is

initiated by that pathway, however, also here details are not fully understood and

questions arise after work with purified molecules [94].

Blood clotting: The strongest formation of the coagulation factor thrombin and of

cellular coagulation mediators (PF4) did not correlate either with the surface showing

maximum contact activation (100% -COOH) nor with the one with highest platelet

adhesion (100% -CH3) but is considerably increased on binary 83% -COOH, where

both acidic and hydrophobic surface groups are present. Sample surface analysis

confirmed coagulation induction on the latter surface showing the formation of a blood

clot. This was surprising as surfaces with exclusively hydrophobic or acidic groups

showed low coagulation potential regarding hemostatic plasmatic or cellular parameters,

respectively. The presence of both acidic and hydrophobic groups shows a completely

different picture than the pure monolayers (100% -COOH, 100% -CH3), where only

either contact activation or cell adhesion occurs. The resulting combination of cellular

and plasmatic events that are induced on the binary 83% -COOH seem to be

indispensable for a strong coagulation reaction. To uncover the causal relationship for

this additional experiments were conducted using an inhibitor for FXIIa as well as an

imitator of activated platelet membranes. Coagulation on 83% -COOH was knocked

down by the addition of FXII inhibitor CTI confirming contact activation as the

essential trigger for clotting in that case. The essential role of platelets for coagulation

on the other hand was shown by induction of clot formation on 100% -COOH following

phospholipid addition to whole blood before surface incubation. The activated platelet

membrane can thus be speculated as a dominant pro-coagulant mechanism if an initial

activation of coagulation is found. It can be concluded that initial processes of

coagulation on foreign materials require an interplay of plasmatic reactions on the one

hand -leading to the formation of thrombin above a certain level as well -as the presence

and activation of blood platelets on the other hand -providing a platform for thrombin

formation [134]. Although surface properties were found to lead to the maximal

activation of one of the pathways without triggering the other reaction, there is an over

additive effect on coagulation for surfaces with intermediate properties.

Page 93: Dissertation Marion Fischer - Qucosa

4 Discussion

86

XII XIIa PSPS

XIaIXa Xa

IIa

83% COOH

PS PS

100% CH3

XII XIIa

100% COOH

XII XIIa

XII XIIa PSPS

XIaIXa Xa

IIa

83% COOH

XII XIIa PSPSPSPS

XIaIXa Xa

IIa

83% COOH

PS PS

100% CH3

PSPS PSPS

100% CH3

XII XIIa

100% COOH

XII XIIaXII XIIa

100% COOH

XII XIIa

Figure 55 Scheme of reactions on biomaterials surfaces with charged groups (100% -COOH),

hydrophobic groups (100% -CH3) and a binary mixture with a predomination of charged groups (83%

-COOH) Description see text (adapted from [139])

The second part of the discussion deals with questions of the relevance of functional

surface groups of materials for the expression, release and procoagulant activity of TF,

the key activator of the extrinsic pathway. Using -OH, -CH3 and -COOH-terminated

SAMs, as well as binary mixtures of these groups, it was shown that TF varies in

expression and cellular concentration depending on the surface chemistry of the

material. This is the first work showing the relevance of surface characteristics of

biomaterials for the activation of the extrinsic pathway.

TF on expression-, protein- and cellular levels

To investigate the relevance of the extrinsic pathway on coagulation, leukocyte TF on

expression, protein and cellular levels were analysed following whole blood incubation

assays of SAM surfaces. The whole blood incubation assay enables the evaluation of

the specific effect of leukocyte TF due to the absence of tissue/endothelial cells during

surface incubation. Both parameters (leukocyte TF m-RNA and cell bound TF)

correlated to surface hydroxylation. Also, the number of leukocytes on the surface

Page 94: Dissertation Marion Fischer - Qucosa

4 Discussion

87

scaled with -OH/--COOH ratios and was highest on SAMs composed only of hydroxyl

groups (100% -OH), consistent with recent studies [140]. It seems that hydroxylated

surfaces with their high leukocyte adhesion have the potential to induce the TF

pathway. In general, natural hydroxylated structures such as hydroxyapatite,

lipopolysaccharides or cellulose do partly induce inflammation processes upon blood

contact but a general scheme on its role in coagulation processes is missing.

Leukocyte adhesion: Leukocyte adhesion seems to trigger the TF pathway activation

and will be discussed in the following. Among the underlying mechanisms that lead to

distinct cell adhesion processes are protein adsorption processes known to take place

during the initial blood-biomaterial interaction. In the present work the adsorption of

complement fragment C3b and release of C5a upon incubation with SAM surfaces were

studied. Hydroxylated surfaces showed enhanced adsorption of C3b upon blood contact,

providing a possible explanation for elevated leukocyte adhesion. Leukocyte attraction

to –OH groups was recently described as a complement driven process and suggested to

be mediated by C3b bound to OH-surfaces [35]. The observed correlation between

complement activation and leukocyte adhesion are the initial events leading to the

activation of the extrinsic pathway and will be discussed in more detail in the following

section.

Slow spontaneous complement activation in the fluid phase has long been proposed to

occur, leading to hydrolysis of C3 to C3b –a cleavage product that is able to bind to

-OH surface groups [141]. In support of this, Hirata et al. suggested enhanced

spontaneous C3b generation in the fluid phase upon serum contact to OH-terminated

SAMs. In this case, C3b attaches in a nucleophilic way, leading to the formation of the

alternative pathway C3 convertase. They proposed the induction of complement

activation on OH-terminated SAMs via the alternative pathway [140] and conducted

experiments using EGTA as a chelating agent to knock out complement activation via

the classical pathway. According to these findings on elevated binding of complement

fragments to 100% -OH after surface incubation in whole blood and blood plasma, it

was suggested that pronounced leukocyte adhesion occurs via its C3b receptor (CR1)

[35]. Results presented in the present work reveal increased amounts of surface bound

complement fragments on OH surfaces upon incubation in whole blood or blood

plasma, and additionally the soluble plasma anaphylatoxin C5a was also found to be

significantly increased. Also, it has been shown that clustering of C3b around C3

convertases, conferred by the presence of OH groups on the surface, may lead to

formation of C5 convertases that results in the generation of C5a [142]. The C5a

concentration correlated strongly with leukocyte CD11b expression and -OH/-COOH

ratios. These findings are not surprising since leukocyte activation can be induced by

Page 95: Dissertation Marion Fischer - Qucosa

4 Discussion

88

C5a via leukocyte C5a receptors (CD88) [143]. Unexpectedly, the number of adherent

leukocytes was even more pronounced on 83% -COOH surfaces compared to 100%

-OH, although no cell activation (CD11b) was observed. Leukocytes on 83% -COOH

were presumably passively incorporated into the growing clot. The coagulation process,

as confirmed by elevated TAT complexes, appeared independent of leukocyte CD11b

and C5a expression, but likely resulted from joint action between coagulation and

platelets [134]. Enhanced leukocyte adhesion to surfaces with high OH ratios seems to

be in part mediated by C3b adsorption that consequently supports high TF generation on

these surfaces.

Tissue factor on cellular level (microscopic techniques): To uncover the location and

distribution of surface TF immunostaining techniques were assessed. In the following

section I refer to “surface TF” measured by fluorescence scanning that reflects the

detection of TF being present on the surface –either adsorbed to the surface (bound to

microparticles/ cell fragments) or associated with adherent cells. I thus equate “surface

TF” with the amount of TF being associated with adherent cells as microparticles and

cell fragments also represent cellular TF. Surface TF on 83% -COOH yielded the

highest TF signal and the lowest TF/leukocyte ratio compared to the other surfaces. It is

suggested that low amounts of surface TF are compensated by the abundant cell

adhesion due to surface-located clot formation. In contrast to this, on 100% -COOH

exhibiting the lowest leukocyte density, the highest surface TF content was found with a

5-fold TF/leukocyte ratio compared to the 83% -COOH/OH surfaces (Figure 51). Even

though the TF/leukocyte ratio was pronounced, the net TF presence at the surface was

limited due to low cell adhesion potential. On 100% -OH, with the highest mRNA and

extracellular TF, the TF/leukocyte ratio was not higher than on binary COOH/OH and

100% -CH3 even though the high leukocyte adhesion helps to enhance the net surface

TF. High surface TF on OH-rich surfaces could thus be explained by the presence of

large numbers of leukocytes that may express TF upon activation, thus triggering the

induction of the extrinsic pathway.

The TF on cell surfaces was found to be associated with both granulocytes and

monocytes. FACS and SEM analysis showed formation of platelet-granulocyte-

conjugates, a proposed mechanism of TF transfer to granulocytes. This might explain

the high amount of membrane bound TF on granulocytes, whose ability to express TF is

still questioned. These data indicate that conjugate formation should be further

considered as an adhesion phenomenon with possible relevance for hemocompatibility.

By fluorescence imaging CD14 positive cell fragments were found most probably

representing monocyte-derived microparticles that mediate intercellular TF transfer via

PSGL-1 and P-selectin interaction.

Page 96: Dissertation Marion Fischer - Qucosa

4 Discussion

89

Tissue factor on cellular level (flow cytometry): The amount of leukocyte bound TF was

assessed by FACS analysis and showed a slight increase in TF positive monocytes and

granulocytes on 100% -OH, but these levels were not significant. These results are

consistent with Gorbet et al. who found that after incubation of PS and PS-PEG beads

with isolated leukocytes in plasma, TF expression and exposure of phosphatidyl serine

was at background levels whereas CD11b was significantly upregulated [144].

Tissue factor on expression and protein level: Leukocyte TF expression determined as

TF mRNA was significantly elevated on 100% -OH compared to all other surfaces. The

amount of intracellular and membrane bound leukocyte TF (both detected by western

blot after cell lysis) showed no significant differences among the SAMs tested, whereas

values were significantly reduced on 100% -OH in compared to initial values. This

suggests an earlier TF release by leukocytes upon cell activation or leukocyte-derived

shedding of microparticles containing considerable amounts of TF.

Procoagulant activity of tissue factor: Although 100% -OH showed the highest TF

expression levels and elevated TF of adherent cells, no sufficient coagulation activation

was achieved and the TAT levels were below necessary threshold levels. The statement

to be drawn is that despite the initiation of the TF pathway on hydroxylated surfaces no

subsequent coagulation activation is triggered. This probably explains the discrepancy

on the definite relevance of the TF pathway in vivo and its controversial significance in

biomaterials induced thrombotic reactions. With our present work data is provided to

contribute to the ongoing discussion on the relevance of the extrinsic pathway in vivo

inside the blood vessel in case of implanted biomaterials.

The absence of both elevated TAT and clot formation on OH-containing surfaces points

to the inability of expressed/released TF to induce coagulation under the conditions of

our in vitro hemocompatibility set-up. High TF expression does not necessarily lead to

the formation of functionally active TF. One explanation may be the lack of lymphocyte

recruitment in our model, as immune complex-induced procoagulant activity may

require the collaboration of both lymphocytes and monocytes [145]. Alternatively, TF

may be quickly inactivated by effective enzymatic agents after its appearance on the cell

surface. Orfeo et al. found the time needed for TF-FVIIa-assembly to be 10-20 seconds.

and after 120 seconds most of the TF was inactivated by TFPI [146]. Considering the

quick inhibition of active TF together with the fact that TF concentrations required to

initiate coagulation were found to be in the range of less than 100 femtomolar [147,

148], an analysis of TF in plasma is challenging. It would, however, greatly contribute

to our understanding of the impact of TF activation at blood material interfaces. Assays

on the activity of TF have to pursued to answer these questions.

Page 97: Dissertation Marion Fischer - Qucosa

4 Discussion

90

As no coagulation activation was found following elevated TF expression, the relevance

of TF for biomaterial-induced coagulation may be of minor importance under the

conditions in our hemocompatibility assay. However, the observed initiation of the

extrinsic pathway can play a significant role in local host responses and the elevated

immune complex-mediated coagulation in wound sites surrounding implants may be of

great importance for concomitant healing and integration processes.

Page 98: Dissertation Marion Fischer - Qucosa

5 Summary and conclusion

91

5. Summary and conclusion

Binary self assembled monolayers of functionally terminated alkyl thiols were

successfully applied to demonstrate the relevance of materials‘ surface characteristics

for initial processes of blood coagulation. The in vitro whole blood incubation of self

assembled monolayers enabled to study the effects of surface wettability and charge on

hemocompatibility parameters like protein adsorption, coagulation activation, contact

activation (intrinsic/ enhancer pathway), the impact of tissue factor (extrinsic/ activator

pathway) and cellular systems (blood platelets and leukocytes).

After the optimisation self assembled monolayer preparation, these model surfaces were

used for the biological assays: the first part of the work focused on studying the

influence of surface properties on platelet- and contact activation, the two main actors of

blood coagulation, which are often considered as separate mechanisms in biomaterials

research. Results show a dependence of contact activation on acidic surface groups and

a correlation of platelet adhesion to surface hydrophobicity. It was found that neither

platelet adhesion -on hydrophobic surfaces- without concurrent contact activation, nor

contact activation -on negatively charged surfaces- without the presence of activated

platelets lead to strong thrombus formation. Clot formation resulting from the interplay

of blood platelets and contact activation was only found on surfaces combining both

surface groups but not on monolayers displaying extreme hydrophobic/acidic

properties. This indicates an influence of the cellular systems on the plasmatic

coagulation. These findings were confirmed by special experiments where coagulation

was knocked down by specific FXIIa inhibition, whereas PL addition was able to

substitute platelet derived procoagulant activities on surfaces without platelet activation.

Materials with adequate surface properties regarding hemocompatibility possibly inhibit

coagulation in whole blood (100% -COOH or 100% -CH3 in our case). Minor changes

of surface properties due to practical reasons, however, can induce inverse

(procoagulant) effects that were lacking on the surfaces displaying extreme properties

(as seen for 83% -COOH). This synergistic effect may be attributed to the procoagulant

lipid surface of adherent platelets and to traces of activated enzymes including FXa and

thrombin formed through the contact activation pathway. Generation of thrombin

considerably boosts platelet activation and leads to a reciprocal activation loop. The

conclusion can be drawn that the coexistence of different activation processes leads to

an on/off switch for coagulation activation on biomaterials, as opposed to the direct

Page 99: Dissertation Marion Fischer - Qucosa

5 Summary and conclusion

92

scaling of platelet adhesion and contact activation with certain physicochemical surface

properties.

The present results evidence that surface design and testing of materials contacting

blood needs to consider a complete picture of activation processes. The findings further

emphasize the necessity to study biomaterial induced coagulation on each particular

surface in the complex system of whole blood instead of analysis and optimisation of

only one hemostasis related parameter. The work thus provides important insight for the

rational design of blood compatible surfaces and for adequate test systems.

In the second part the relevance of surface properties for material related-induction of

the tissue Factor (TF) pathway was investigated. TF expression in leukocytes and

leukocyte bound TF in whole blood after incubation with surfaces displaying –OH

functionalities showed higher levels than on any CH3– and COOH– exposing surfaces.

In addition, a positive correlation between TF transcription and its presence on

leukocytes, granulocyte activation, and complement activation was found. Cells

displaying the highest TF expression after material contact had significantly lower

intracellular TF, possibly pointing to previous TF release. The TF pathway was shown

to be induced by characteristic patterns of physicochemical surface properties on

artificial materials. However, the observed initiation of the extrinsic pathway did not

trigger blood coagulation in the absence of tissue –a condition that is given in our whole

blood incubation setup. Missing correlation between leukocyte bound TF and

coagulation activation is possibly a result of lacking tissue-derived TF (a precondition

given by the experimental set up) or a consequence of quick TF inhibition by

serological enzymes. It can be concluded that TF does not activate blood coagulation in

short term applications of biomaterials in blood contacting medical devices, such as

blood purification systems. The significance of TF pathway activation at local wound

sites can, however, be of great importance in terms of implant healing and integration.

Page 100: Dissertation Marion Fischer - Qucosa

List of abbreviations

93

List of abbreviations

ADP adenosine diphosphate

AFM atomic force microscopy

BSA bovine serum albumin

CTI corn trypsin inhibitor

DAPI diamidine-phenylindole dihydrochloride

DS dextrane sulfate

EDTA ethylen-diamine tetra acetic acid

EG3Ome tri-methoxy terminated ethylene glycol

ELISA enzyme linked immuno sorbent assay

FITC fluorescein isothiocyanate

FPA fibrinopeptide A

FTIR fourier transformed infrared spectroscopy

GAP-DH glyceraldehyde -phosphate dehydrogenase

GP glycoprotein

HDT hexadecanethiol

HEPES hydroxyethyl-piperazineethanesulfonic acid

HFG human fibrinogen

HMWK high-molecular-weight-kininogen

HRP horse reddish peroxidase

IEP isoelectrical point

Ig immunoglobulin

IRRAS infrared reflection absorption spectroscopy

IU international unit

LDH lactate dehydrogenase

LSM laser scanning microscopy

MHA mercapto-hexadecanoic acid

MUA mercapto-undecanoic acid

OD optical density

OPD o-phenylenediamine

PAR proteinase activated receptors

PBS phosphate buffered saline

PDI protein disulfide isomerase

PE phycoerythrine labelled

PF4 platelet factor 4

Page 101: Dissertation Marion Fischer - Qucosa

List of abbreviations

94

PFA para formaldehyde

PL phospholipids

PRP platelet rich plasma

PSGL-1 P-selectin glycoprotein ligand-1

PTFE poly tetra fluoro ethylene

QCM quartz crystal microbalance

RVV Russell’s viper venom

SAM self assembled monolayer

SD standard deviation

SDS sodium dodecyl sulfate

SEM scanning electron microscopy

TAT thrombin antithrombin complex

TF tissue factor

TFAA trifluoroacetic anhydride

TFPI tissue factor pathway inhibitor

THF tetrahydrofurane

UDT undecanoic acid

XPS X-ray-photoelectron spectroscopy

β-TG β-thromboglobulin

Page 102: Dissertation Marion Fischer - Qucosa

95

References

[1] Ratner BD. The catastrophe revisited: blood compatibility in the 21st Century. Biomaterials 2007;28:5144-7.

[2] Gorbet MB, Sefton MV. Biomaterial-associated thrombosis: roles of coagulation factors, complement, platelets and leukocytes. Biomaterials 2004;25:5681-703.

[3] Werner C, Maitz MF, Sperling C. Current strategies towards hemocompatible coatings. J Mater Chem 2007;17:3376-84.

[4] Streller U, Sperling C, Hübner J, Hanke R, Werner C. Design and evaluation of novel blood incubation systems for in vitro hemocompatibility assessment of planar solid surfaces. J Biomed Mater Res 2003;66B:379-90.

[5] Ulman A. Formation and structure of self-assembled monolayers. Chem Rev 1996;96:1533-54.

[6] Prime KL, Whitesides, G.M. SElf-assembled organic monolayers: Model systems for studying adsorption of proteins at surfaces. Science 1991;252:1164-66.

[7] Schweiss R, Pleul D, Simon F, Janke A, Welzel P, Voit B, et al. Electrokinetic Potentials of Binary Self-Assembled Monolayers on Gold: Acid-Base Reactions and Double Layer Structure. J Phys Chem B 2004:2910-17.

[8] Mrksich M, Whitesides GM. Using self-assembled monolayers to understand the interactions of man-made surfaces with proteins and cells. Annu Rev Biophys Biomol Struct 1996;25:55-78.

[9] Mrksich M, Dike LE, Tien J, Ingber DE, Whitesides GM. Using microcontact printing to pattern the attachment of mammalian cells to self-assembled monolayers of alkanethiolates on transparent films of gold and silver. Exp Cell Res 1997;235:305-13.

[10] Barrias CC, Martins MA, Sa Miranda MA, Barbosa MA. Adsorption of a therapeutic enzyme to self-assembled monolayers: effect of surface chemistry and solution pH on the amount and activity of adsorbed enzyme. Biomaterials 2005/5;26:2695-704.

[11] Benesch J, Svendheim S, Svensson SCT, Valiokas R, Liedberg B, Tengvall P. Protein adsorption to oligo (ethylene glycol) self-assembled monolayers: Experiments with fibrinogen, heparinized plasma, and serum. J Biomater Sci Polym Ed 2001;12:581-97.

[12] Faucheux N, Schweiss R, Lutzow K, Werner C, Groth T. Self-assembled monolayers with different terminating groups as model substrates for cell adhesion studies. Biomaterials 2004;25:2721-30.

[13] Kalltorp M, Oblogina S, Jacobsson S, Karlsson A, Tengvall P, Thomsen P. In vivo cell recruitment, cytokine release and chemiluminescence response at gold, and thiol functionalized surfaces. Biomaterials 1999;20:2123-37.

[14] Zhang Y, Lu XQ, Liao TL, Cheng YN, Liu XH, Zhang LM. Studies on interaction of porphyrin and its complexes with DNA at interface on gold electrode modified by thiol-porphyrin self-assembled monolayer. J Solid State Electrochem 2007;11:1303-12.

Page 103: Dissertation Marion Fischer - Qucosa

References

96

[15] Patel KR, Tang HY, Grever WE, Ng KYS, Xiang JM, Keep RF, et al. Evaluation of polymer and self-assembled monolayer-coated silicone surfaces to reduce neural cell growth. Biomaterials 2006;27:1519-26.

[16] Rodrigues SN, Goncalves IC, Martins MC, Barbosa MA, Ratner BD. Fibrinogen adsorption, platelet adhesion and activation on mixed hydroxyl-/methyl-terminated self-assembled monolayers. Biomaterials 2006;27:5357-67.

[17] Lestelius M, Liedberg B, Tengvall P. In vitro plasma protein adsorption on w-functionalized alkanethiolate self-assembled monolayers. Langmuir 1997;13:5900-08.

[18] Barbosa JN, Barbosa MA, Aguas AP. Inflammatory responses and cell adhesion to self-assembled monolayers of alkanethiolates on gold. Biomaterials 2004;25:2557-63.

[19] Sperling C, Schweiss RB, Streller U, Werner C. In vitro hemocompatibility of self-assembled monolayers displaying various functional groups. Biomaterials 2005;26:6547-57.

[20] Chuang W-H, Lin J-C. Surface characterization and platelet adhesion studies for the mixed self-assembled monolayers with amine and carboxylic acid terminated functionalities. J Biomed Mater Res A 2007;82A:820-30.

[21] Arnold R, Struktur und Ordnung selbstordnender Monolagen aliphatischer und aromatischer Thiole auf Goldoberflächen, in faculty of chemistry. 2001, Ruhr-Universität Bochum: Bochum. p. 140.

[22] Arnold R, Azzam W, Terfort A, Woll C. Preparation, Modification, and Crystallinity of Aliphatic and Aromatic Carboxylic Acid Terminated Self-Assembled Monolayers. Langmuir 2002;18:3980-92.

[23] Chen H, Chen J, Aoki K, Nishiumi T. Electrochemically instantaneous reduction of conducting polyaniline-coated latex particles dispersed in acidic solution. Electrochimica Acta 2008;53:7100-06.

[24] Dai J, Cheng J, Li Z, Jin J, Bi S. Rapid formation of high-quality self-assembled monolayers of dodecanethiol on polycrystalline gold under ultrasonic irradiation. Electrochimica Acta 2008;53:3479-83.

[25] Kim HJ, Kwak S, Kim YS, Seo BI, Kim ER, Lee H. Adsorption kinetics of alkanethiols studied by quartz crystal microbalance. Thin Solid Films 1998;329:191-94.

[26] Vroman L. The life of an artificial device in contact with blood. Bull.N.Y.Acad.Med. 1988;64:352-57.

[27] Wittmer CR, Van Tassel PR. Probing adsorbed fibronectin layer structure by kinetic analysis of monoclonal antibody binding. Colloids Surf B Biointerfaces 2005;41:103-09.

[28] Krishnan A, Cha P, Liu YH, Allara D, Vogler EA. Interfacial energetics of blood plasma and serum adsorption to a hydrophobic self-assembled monolayer surface. Biomaterials 2006/6;27:3187-94.

[29] Hylton DM, Shalaby SW, Latour RA, Jr. Direct correlation between adsorption-induced changes in protein structure and platelet adhesion. J Biomed Mater Res A 2005/6/1;73:349-58.

[30] Evans-Nguyen KM, Schoenfisch MH. Fibrin proliferation at model surfaces: Influence of surface properties. Langmuir 2005;21:1691-94.

[31] Evans-Nguyen KM, Tolles LR, Gorkun OV, Lord ST, Schoenfisch MH. Interactions of thrombin with fibrinogen adsorbed on methyl-, hydroxyl-, amine-

Page 104: Dissertation Marion Fischer - Qucosa

References

97

, and carboxyl-terminated self-assembled monolayers. Biochemistry 2005;44:15561-68.

[32] Colman RW, Schmaier AH. Contact system: A vascular biology modulator with anticoagulant, profibrinolytic, antiadhesive, and proinflammatory attributes. Blood 1997;90:3819-43.

[33] Frank RD, Weber J, Dresbach H, Thelen H, Weiss C, Floege J. Role of contact system activation in hemodialyzer-induced thrombogenicity. Kidn.Int. 2001;60:1972-81.

[34] Yarovaya GA, Blokhina TB, Neshkova EA, Contact system. New concepts on activation mechanisms and bioregulatory functions. MAIK Nauka/Interperiodica ed. Biochemistry (Moscow). Vol. 67. 2002: Springer Science+Business Media LLC. 13-24.

[35] Sperling C, Maitz MF, Talkenberger S, Gouzy M-F, Groth T, Werner C. In vitro blood reactivity to hydroxylated and non-hydroxylated polymer surfaces. Biomaterials 2007;28:3617-25.

[36] Sefton MV. Material-induced tissue factor expression but not CD11b upregulation depends on the presence of platelets. J Biomed Mater Res 2003;67A:792-800.

[37] Gorbet MB, Yeo EL, Sefton MV. Flow cytometric study of in vitro neutrophil activation by biomaterials. J Biomed Mater Res 1999;44:289-97.

[38] Wettero J, Askendal A, Bengtsson T, Tengvall P. On the binding of complement to solid artificial surfaces in vitro. Biomaterials 2002;23:981-91.

[39] Berglin M, Andersson M, Sellborn A, Elwing H. The effect of substrate molecular mobility on surface induced immune complement activation and blood plasma coagulation. Biomaterials 2004/8;25:4581-90.

[40] Wetterö J, Askendal A, Tengvall P, Bengtsson T. Interactions between surface-bound actin and complement, platelets, and neutrophils. J Biomed Mater Res 2003;66A:162-75.

[41] Tengvall P, Askendal A, Lundstrom I. Complement activation by IgG immobilized on methylated silicon. J Biomed Mater Res 1996;31:305-12.

[42] Wettero J, Bengtsson T, Tengvall P. Complement activation on immunoglobulin G-coated hydrophobic surfaces enhances the release of oxygen radicals from neutrophils through an actin-dependent mechanism. J Biomed Mater Res 2000;51:742-51.

[43] Tengvall P, Askendal A, Lundstrom I, I. Ellipsometric in vitro studies on the activation of complement by human immunoglobulins M and G after adsorption to methylated silicon. Colloids Surf B Biointerfaces 2001;20:51-62.

[44] Kao WJ. Evaluation of leukocyte adhesion on polyurethanes: the effects of shear stress and blood proteins. Biomaterials 2000;21:2295-303.

[45] Labarre D, Laurent A, Lautier A, Bouhni S, Kerbellec L, Lewest M, et al. Complement activation by substituted polyacrylamide hydrogels for embolisation and implantation. Biomaterials 2002;23:2319-27.

[46] Sperling C, Maitz MF, Talkenberger S, Gouzy MF, Groth T, Werner C. In vitro blood reactivity to hydroxylated and non-hydroxylated polymer surfaces. Biomaterials 2007;28:3617-25.

[47] Jenney CR, Anderson JM. Adsorbed serum proteins responsible for surface dependent human macrophage behavior. J Biomed Mater Res 2000;49:435-47.

Page 105: Dissertation Marion Fischer - Qucosa

References

98

[48] Shen M, Horbett A. The effects of surface chemistry and adsorbed proteins on monocyte / macrophage adhesion to chemically modified polystyrene surfaces. J Biomed Mater Res 2001;57:336-45.

[49] Simon SI, Goldsmith HL. Leukocyte adhesion dynamics in shear flow. Annals of Biomedical Engineering 2002;30:315-32.

[50] Spatnekar S, Anderson JM, Hemocompatibility: Effects on humoral elements. 2nd edition ed. Handbook of biomaterials evaluation: Scientific, technical and clinical testing of implant materials, ed. von Recum AF. Philadelphia PTF. 1999. 353-65.

[51] Zhuo R, Siedlecki CA, Vogler EA. Autoactivation of blood factor XII at hydrophilic and hydrophobic surfaces. Biomaterials 2006;27:4325-32.

[52] Kozin F, Cochrane CG. The contact activation system of plasma - biochemistry and pathophysiology. In: Gallin JI Goldstein IM, Snyderman R, editors. Inflammation: Basic Principles and Clinical Correlates. 2nd ed. New York: Raven Press 1992:101-20.

[53] Chen X, Wang J, Paszti Z, Wang F, Schrauben JN, Tarabara VV, et al. Ordered adsorption of coagulation factor XII on negatively charged polymer surfaces probed by sum frequency generation vibrational spectroscopy. Anal Bioanal Chem 2007:65-72.

[54] Zhuo R, Siedlecki CA, Vogler EA. Competitive-protein adsorption in contact activation of blood factor XII. Biomaterials 2007;28:4355-69.

[55] Leibner ES, Barnthip N, Chen W, Baumrucker CR, Badding JV, Pishko M, et al. Superhydrophobic effect on the adsorption of human serum albumin. Acta Biomater 2009;5:1389-98.

[56] Schulman G, Hakim R, Arias R, Silverberg M, Kaplan AP, Arbeit L. Bradykinin generation by dialysis membranes: possible role in anaphylactic reaction. J Am Soc Nephrol 1993;3:1563-69.

[57] Sanchez J, Lundquist PB, Elgue G, Larsson R, Olsson P. Measuring the degree of plasma contact activation induced by artificial materials. Thromb Res 2002;105:407-12.

[58] Gailani D, Renné T. Intrinsic pathway of coagulation and arterial thrombosis. Arterioscler Thromb Vasc Biol 2007;27:2507-13.

[59] Schmidbauer S, Nerlich C, Weimer T, Kronthaler U, Metzner H, Schulte S. Prevention of thrombotic events by Factor XIIa inhibitors. Hämostaseologie 2009;29:A22.

[60] Bradford HN, Pixley RA, Colman RW. Human factor XII binding to the glycoprotein Ib-IX-V complex inhibits thrombin-induced platelet aggregation. J Biol Chem 2000;275:22756-63.

[61] Camerer E, Kolsto AB, Prydz H. Cell biology of tissue factor, the principal initiator of blood coagulation. Thromb Res 1996;81:1-41.

[62] Giesen PLA, Rauch, U., Bohrmann B, Kling D, Roqué M, et al. Blood-borne tissue factor: another view of thrombosis. Proc Natl Acad Sci U S A 1999;96:2311-15.

[63] Rauch U, Nemerson Y. Tissue factor, the blood, and the arterial wall. Trends Cardiovasc.Med 2000/5;10:139-43.

[64] Spek CA. Tissue factor: from 'just one of the coagulation factors' to a major player in physiology. Blood Coagul Fibrinolysis 2004;15 Suppl 1:S3-10.

Page 106: Dissertation Marion Fischer - Qucosa

References

99

[65] Maly MA, Tomasov P, Hajek P, Blasko P, Hrachovinova I, Salaj P, et al. The role of tissue factor in thrombosis and hemostasis. Physiol Res 2007;56:685-95.

[66] Hathcock JJ, Nemerson Y. Platelet deposition inhibits tissue factor activity: in vitro clots are impermeable to factor Xa. Blood 2004;104:123-7.

[67] Mallat Z, Benamer H, Hugel B, Benessiano J, Steg PG, Freyssinet JM, et al. Elevated levels of shed membrane microparticles with procoagulant potential in the peripheral circulating blood of patients with acute coronary syndromes. Circulation 2000;101:841-3.

[68] Heemskerk JW, Bevers EM, Lindhout T. Platelet activation and blood coagulation. Thromb Haemost 2002;88:186-93.

[69] Sturk-Maquelin KN, Nieuwland R, Romijn FP, Eijsman L, Hack CE, Sturk A. Pro- and non-coagulant forms of non-cell-bound tissue factor in vivo. J Thromb Haemost 2003;1:1920-6.

[70] Berckmans RJ, Neiuwland R, Boing AN, Romijn FP, Hack CE, Sturk A. Cell-derived microparticles circulate in healthy humans and support low grade thrombin generation. Thromb Haemost 2001;85:639-46.

[71] Kappelmayer J, Bernabei A, Edmunds H, Edgington TS, Colman RW. Tissue factor is expressed on monocytes during extracorporeal circulation. Circ Res 1993;72:1075-81.

[72] Osterud B. Cellular interactions in tissue factor expression by blood monocytes. Blood Coagul Fibrinolysis 1995;6:20-25.

[73] Rauch U, Bonderman D, Bohrmann B, Badimon JJ, Himber J, Riederer MA, et al. Transfer of tissue factor from leukocytes to platelets is mediated by CD15 and tissue factor. Blood 2000/7/1;96:170-75.

[74] Polgar J, Matuskova J, Wagner DD. The P-selectin, tissue factor, coagulation triad. J.Thromb.Haemost. 2005/8;3:1590-96.

[75] Eilertsen KE, Osterud B. The role of blood cells and their microparticles in blood coagulation. Biochem Soc Trans 2005;33:418-22.

[76] Nakamura S, Imamura T, Okamoto K. Tissue factor in neutrophils: yes. J Thromb Haemost 2004;2:214-7.

[77] Camera M, Frigerio M, Toschi V, Brambilla M, Rossi F, Cottell DC, et al. Platelet activation induces cell-surface immunoreactive tissue factor expression, which is modulated differently by antiplatelet drugs. Arterioscler Thromb Vasc Biol 2003;23:1690-6.

[78] Muller I, Klocke A, Alex M, Kotzsch M, Luther T, Morgenstern E, et al. Intravascular tissue factor initiates coagulation via circulating microvesicles and platelets. Faseb J 2003/3;17:476-78.

[79] Panes O, Matus V, Saez CG, Quiroga T, Pereira J, Mezzano D. Human platelets synthesize and express functional tissue factor. Blood 2007;109:5242-50.

[80] Butenas S, Mann KG. Blood coagulation. Biochemistry (Moscow) 2002;67:3-12.

[81] Osterud BJ, Mohan Rao LV, Olsen JO. Induction of tissue factor expression in whole blood: Lack of evidence for the presence of tissue factor expression in granulocytes. Thromb Haemost 2000;83:861-67.

[82] Osterud B. Tissue factor in neutrophils: no. J Thromb Haemost 2004;2:218-20. [83] Giesen PL, Nemerson Y. Tissue factor on the loose. Semin Thromb Hemost

2000;26:379-84.

Page 107: Dissertation Marion Fischer - Qucosa

References

100

[84] Bach RR. Tissue factor encryption. Arterioscler.Thromb.Vasc.Biol. 2006/3;26:456-61.

[85] Stone MD, Harvey SB, Martinez MB, Bach RR, Nelsestuen GL. Large enhancement of functional activity of active site-inhibited factor VIIa due to protein dimerization: insights into mechanism of assembly/disassembly from tissue factor. Biochemistry 2005;44:6321-30.

[86] Dietzen DJ, Page KL, Tetzloff TA. Lipid rafts are necessary for tonic inhibition of cellular tissue factor procoagulant activity. Blood 2004;103:3038-44.

[87] Osterud B. The role of platelets in decrypting monocyte tissue factor. Semin Hematol 2001;38:2-5.

[88] Chen VM, Ahamed J, Versteeg HH, Berndt MC, Ruf W, Hogg PJ. Evidence for activation of tissue factor by an allosteric disulfide bond. Biochemistry 2006;45:12020-8.

[89] Ahamed M, Kumar A, Siddiqui MK. Lipid peroxidation and antioxidant status in the blood of children with aplastic anemia. Clin Chim Acta 2006;374:176-7.

[90] Pendurthi UR, Ghosh S, Mandal SK, Rao LVM. Tissue factor activation: is disulfide bond switching a regulatory mechanism? Blood 2007;110:3900-08.

[91] Balasubramanian V, Grabowski E, Bini A, Nemerson Y. Platelets, circulating tissue factor, and fibrin colocalize in ex vivo thrombi: real-time fluorescence images of thrombus formation and propagation under defined flow conditions. Blood 2002;100:2787-92.

[92] Siddiqui FA, Desai H, Amirkhosravi A, Amaya M, Francis JL. The presence and release of tissue factor from human platelets. Platelets 2002;13:247-53.

[93] Cicala C, Cirino G. Linkage between inflammation and coagulation: an update on the molecular basis of the crosstalk. Life Sci 1998;62:1817-24.

[94] Zarbock A, Polanowska-Grabowska RK, Ley K. Platelet-neutrophil-interactions: linking hemostasis and inflammation. Blood Rev 2007;21:99-111.

[95] Colman RW, Cook JJ, Niewiarowski S. Mechanisms of platelet aggregation. In: Colman RW, Hirsh J, Marder VJ, Salzmann EW, editors. Hemostasis and thrombosis: basic principles and clinical practice. 3rd ed. Philadelphia: Lippincott; 1994. p. 508-23

[96] Furie B, Furie BC. Mechanisms of thrombus formation. N Engl J Med 2008;359:938-49.

[97] Beumer S, Ijsseldijk MJW, de Groot PG, Sixma JJ. Platelet adhesion to fibronectin in flow: dependence on surface concentration and shear rate, role of platelet membrane glycoproteins GP IIb/IIIa and VLA-5, and inhibition by heparin. Blood 1994;84:3724-33.

[98] Grunkemeier JM, Tsai WB, McFarland CD, Horbett TA. The effect of adsorbed fibrinogen, fibronectin, von Willebrand factor and vitronectin on the procoagulant state of adherent platelets. Biomaterials 2000;21:2243-52.

[99] Broberg M, Nygren H. Platelet interactions with surface-adsorbed plasma proteins: exposure of CD62P induced by von Willebrand factor. Colloids Surf B Biointerfaces 1998;11:67-77.

[100] Haimovich B, Lipfert L, Brugge JS, Shattil SJ. Tyrosine phosphorylation and cytoskeletal reorganization in platelets are triggered by interaction of integrin receptors with their immobilized ligands. J Biol Chem 1993;268:15868-77.

Page 108: Dissertation Marion Fischer - Qucosa

References

101

[101] Massa TM, Yang ML, Ho JY, Brash JL, Santerre JP. Fibrinogen surface distribution correlates to platelet adhesion pattern on fluorinated surface-modified polyetherurethane. Biomaterials 2005/12;26:7367-76.

[102] Andrews RK, Berndt MC. Adhesion-dependent signalling and the initiation of haemostasis and thrombosis. Histol.Histopathol. 1998;13:837-44.

[103] Parise LV. Integrin alpha IIb beta 3 signaling in platelet adhesion and aggregation. Current Op.in Cell Biol. 1999;11:597-601.

[104] Cook BC. Reactivity of human platelets with immobilized fibrinogen is dictated by the chemical character of the surface. Thromb Res 2001;104:39-48.

[105] Blombäck B, Bark N. Fibrinopeptides and fibrin gel structure. Biophys Chem 2004;112:147-51.

[106] Davi G, Patrono C. Platelet activation and atherothrombosis. N Engl J Med 2007;357:2482-94.

[107] Adam F, Guillin MC, Jandrot-Perrus M. Glycoprotein Ib-mediated platelet activation. A signalling pathway triggered by thrombin. Eur J Biochem 2003;270:2959-70.

[108] Sharma CP. Blood-compatible materials: A perspective. J Biomater Appl 2001;15:359-80.

[109] Ratner BD. Blood compatibility--a perspective. J Biomater Sci Polym Ed 2000;11:1107-19.

[110] Sefton MV, Sawyer A, Gorbet M, Black JP, Cheng E, Gemmel C, et al. Does surface chemistry affect thrombogenicity of surface modified polymers? J Biomed Mater Res 2001;55:447-59.

[111] Willey TM, Vance AL, vanBuuren T, Bostedt C, Nelson AJ, Terminello LJ, et al. Chemically Transformable Configurations of Mercaptohexadecanoic Acid Self-Assembled Monolayers Adsorbed on Au(111). Langmuir 2004;20:2746-52.

[112] Werner C, Korber H, Zimmermann R, Dukhin S, Jacobasch HJ. Extended Electrokinetic Characterization of Flat Solid Surfaces. J Colloid Interface Sci 1998;208:329-46.

[113] McCrackin FL. A FORTRAN Program for Analysis of Ellipsometer Measurements. In: Technical Note 479, National Bureau of Standards, US Government Printing Office, Washington, DC1969.

[114] Feijter JAD, Benjamins J, Veer FA. Ellipsometry as a tool to study the adsorption behavior of synthetic and biopolymers at the air-water interface. Biopolymers 1978;17:1759-72.

[115] Racher A. LDH Assay. In: Doyle A, Griffiths JB, editors. Cell and tissue culture: Laboratory procedures in biotechnology. Chichester, New York, Weinheim: John Wiley & Sons; 1998. p. 71-5

[116] Tsai W.B. JMGTAH. Human plasma fibrinogen adsorption and platelet adhesion to polystyrene. J Biomed Mater Res 1999;44:130-39.

[117] Porter MD, Bright TB, Allara DL, Chidsey CED. Spontaneously organized molecular assemblies. 4. Structural characterization of n-alkyl thiol monolayers on gold by optical ellipsometry, infrared spectroscopy, and electrochemistry. J Am Chem Soc 1987;109:3559-68.

[118] Kind M, Wöll C. Organic surfaces exposed by self-assembled organothiol monolayers: Preparation, characterization, and application. Prog Surf Sci 2009;84:230-78.

Page 109: Dissertation Marion Fischer - Qucosa

References

102

[119] Chechik V, Stirling CJM, Gold-thiol self-assembled monolayers. 1999: In: Patai S, Rappoport Z, editors. The chemistry of organic derivativesof gold and silver. New York: John Wiley & Sons, Ltd. p. 551-640.

[120] Karpovich DS, Blanchard GJ. Direct measurement of the adsorption-kinetics of alkanethiolate self-assembled monolayers on a microcrystalline gold surface. Langmuir 1994;10:3315-22.

[121] Schweiss R, Welzel PB, Werner C, Knoll W. Interfacial charge of organic thin films characterized by streaming potential and streaming current measurements. Coll Surf A 2001:97-102.

[122] Kreuzer HJ, Wang RL, Grunze M. Hydroxide ion adsorption on self-assembled monolayers. J Am Chem Soc 2003;125:8384-9.

[123] Usui S, Healy TW. Zeta potential of insoluble monolayer of long-chain alcohol at the air-aqueous solution interface. J Colloid Interface Sci 2001;240:127-32.

[124] Karraker KA, Radke CJ. Disjoining pressures, zeta potentials and surface tensions of aqueous non-ionic surfactant/electrolyte solutions: theory and comparison to experiment. Adv Colloid Interface Sci 2002;96:231-64.

[125] Schmidt U, Zschoche S, Werner C. Modification of poly(octadecene-alt-maleic anhydride) films by reaction with functional amines. J Appl Polym Sci 2003;87:1255-66.

[126] Dunstan DE, Saville DA. Electrokinetic potential of the alkane aqueous-electrolyte interface. Journal of the Chemical Society-Faraday Transactions 1993;89:527-29.

[127] Bertilsson L, Liedberg B. Infrared study of thiol monolayer assemblies on gold - preparation, characterization, and functionalization of mixed monolayers. Langmuir 1993;9:141-49.

[128] Hutt DA, Leggett GJ. Functionalization of hydroxyl and carboxylic acid terminated self-assembled monolayers. Langmuir 1997;13:2740-48.

[129] Arima Y, Iwata H. Effect of wettability and surface functional groups on protein adsorption and cell adhesion using well-defined mixed self-assembled monolayers. Biomaterials 2007;28:3074-82.

[130] Ostuni E, Yan L, Whitesides GM. The interaction of proteins and cells with self-assembled monolayers of alkanethiolates on gold and silver. Colloids Surf B Biointerfaces 1999;15:3-30.

[131] Ostuni E, Chapman RG, Holmlin RE, Takayama S, Whitesides GM. A survey of structure-property relationships of surfaces that resist the adsorption of protein. Langmuir 2001;17:5605-20.

[132] Geer CB, Rus IA, Lord ST, Schoenfisch MH. Surface-dependent fibrinopeptide A accessibility to thrombin. Acta Biomater 2007;3:663-8.

[133] Evans-Nguyen KM, Tolles LR, Gorkun OV, Lord ST, Schoenfisch MH. Interactions of thrombin with fibrinogen adsorbed on methyl-, hydroxyl-, amine-, and carboxyl-terminated self-assembled monolayers. Biochemistry 2005/11/29;44:15561-68.

[134] Sperling C, Fischer M, Maitz MF, Werner C. Blood coagulation on biomaterials requires the combination of distinct activation processes. Biomaterials 2009;30:4447-56.

[135] Miyamoto M, Sasakawa S, Ozawa T, Kawaguchi H, Ohtsuka Y. Mechanisms of blood coagulation induced by latex particles and the roles of blood cells. Biomaterials 1990;11:385-88.

Page 110: Dissertation Marion Fischer - Qucosa

References

103

[136] Liebe S. Effect of ampholines on blood coagulation: 1. Activation of factor VIII (antihemophilic globulin A). Folia Haematol Int Mag Klin Morphol Blutforsch 1975;102:454-61.

[137] Sagnella S, Mai-Ngam K. Chitosan based surfactant polymers designed to improve blood compatibility on biomaterials. Colloids Surf B Biointerfaces 2005;42:147-55.

[138] Vogler EA, Graper JC, Harper GR, Sugg HW, Lander LM, Brittain WJ. Contact activation of the plasma coagulation cascade. I. Procoagulant surface chemistry and energy. J Biomed Mater Res 1995;29:1005-16.

[139] Fischer TH, Thatte HS, Nichols TC, Bender-Neal DE, Bellinger AD, Vournakis JN. Synergistic platelet integrin signaling and factor XII activation in poly-N-acetyl glucosamine fiber-mediated hemostasis. Biomaterials 2005/9;26:5433-43.

[140] Hirata I, Hioki Y, Toda M, Kitazawa T, Murakami Y, Kitano E, et al. Deposition of complement protein C3b on mixed self-assembled monolayers carrying surface hydroxyl and methyl groups studied by surface plasmon resonance. J Biomed Mater Res 2003;66A:669-76.

[141] Pangburn MK, Schreiber RD, Muller-Eberhard HJ. Formation of the initial C3 convertase of the alternative complement pathway. Acquisition of C3b-like activities by spontaneous hydrolysis of the putative thioester in native C3. J Exp Med 1981;154:856-67.

[142] Hakim RM, Breillatt J, Lazarus JM, Port FK. Complement activation and hypersensitivity reactions to dialysis membranes. N Engl J Med 1984;311:878-82.

[143] Guo RF, Ward PA. Role of C5a in inflammatory responses. Annu Rev Immunol 2005;23:821-52.

[144] Gorbet MB, Sefton MV. Complement inhibition reduces material-induced leukocyte activation with PEG modified polystyrene beads (Tentagel) but not polystyrene beads. J Biomed Mater Res A 2005/9/15;74:511-22.

[145] Schwartz BS, Edgington TS. Lymphocyte collaboration is required for induction of murine monocyte procoagulant activity by immune complexes. J Immunol 1981;127:438-43.

[146] Orfeo T, Butenas S, Brummel-Ziedins KE, Mann KG. The tissue factor requirement in blood coagulation. J Biol Chem 2005;280:42887-96.

[147] Mann KG, Orfeo T, Butenas S, Undas A, Brummel-Ziedins K. Blood coagulation dynamics in haemostasis. Hämostaseologie 2009;29:7-16.

[148] Butenas S, Bouchard BA, Brummel-Ziedins KE, Parhami-Seren B, Mann KG. Tissue factor activity in whole blood. Blood 2005;105:2764-70.

Page 111: Dissertation Marion Fischer - Qucosa

104

Versicherung § 5 Abs. 1 Nr. 5

a) Hiermit versichere ich, dass ich die vorliegende Arbeit ohne unzulässige Hilfe Dritter und ohne Benutzung anderer als der angegebenen Hilfsmittel angefertigt habe; die aus fremden Quellen direkt oder indirekt übernommenen Gedanken sind als solche kenntlich gemacht. Die Arbeit wurde bisher weder im Inland noch im Ausland in gleicher oder ähnlicher Form einer anderen Prüfungsbehörde vorgelegt. b) Die vorliegende Arbeit mit dem Titel „Initiation of blood coagulation – Evaluating the relevance of specific surface functionalities using self assembled monolayers“ wurde vom März 2007 bis Mai 2010 im Leibniz-Institut für Polymerfoschung Dresden/ Max Bergmann Centre for Biomaterials unter der Betreuung von Prof. Dr. Carsten Werner angefertigt. c) Hiermit versichere ich, dass ich keine früheren erfolglosen Promotionsverfahren bestritten habe. d) Hiermit erkenne ich die Promotionsordnung vom 20.03.2000 der Fakultät Mathematik und Naturwissenschaften an der Technischen Universität Dresden an. Marion Fischer Dresden, den 03.05.2010


Recommended