Strategies for creating antifouling surfaces using self...

149
Strategies for creating antifouling surfaces using self assembled poly(ethylene glycol) thiol molecules LOKANATHAN ARCOT RAGHUPATHI Interdisciplinary Nanoscience Center (iNANO) Aarhus University, Denmark PhD thesis September 2011

Transcript of Strategies for creating antifouling surfaces using self...

Page 1: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

Strategies for creating antifouling surfaces using self­assembled poly(ethylene glycol) thiol molecules

LOKANATHAN ARCOT RAGHUPATHI

Interdisciplinary Nanoscience Center (iNANO)

Aarhus University, Denmark

PhD thesis September 2011

Page 2: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

ii | P a g e

THESIS FOR OBTAINING PHD DEGREE

This thesis has been submitted to the Faculty of Science at Aarhus University,

Denmark, in order to fulfill the requirements for obtaining a PhD degree in

Nanoscience. The work was carried out under the supervision of Prof. Peter

Kingshott and co‐supervisor Rikke Louise Meyer at Interdisciplinary Nanoscience

Center (iNANO).

The PhD project was part of the project "Nano‐ and bio‐functionalised

surfaces for biofilm prevention" funded by the Danish Council for Strategic

Research under the NABIIT program. The project partners were Danish

Technological Institute and Alfa Laval A/S.

Page 3: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

iii | P a g e

ABSTRACT

Microorganisms are one of the most important parts of our ecosystem influencing the

sustenance of human society. The beneficial microbes are of high relevance to food industry,

development of antibiotics and processing of many raw materials. Mankind has indeed

benefitted a lot from large number of microbial species, but then the environment is also

teeming with pathogenic microbes that pose serious threat to human health. Hence the

success of human survival not only depends on exploiting the useful microbes but also on our

ability to defend ourselves against the pathogenic ones. Microbes such as bacteria can exist

either in free floating planktonic form or as biofilm which can defined as adherent microbial

colonies embedded in slime or extracellular polymeric substance. Microbial biofilms are

responsible for a large number of problems posing a serious safety threat within our society.

Biofilms have substantial impact on human health, as many bacterial infections are caused by

or involve biofilms. Biofilm infections are for example often associated with medical implants,

as artificial surfaces in the human body provide a safe haven where biofilms can form. The

food industry daily combats biofilms forming on the surfaces of equipment to avoid

contamination of their products. In addition to posing a health problem, biofilms also pose

technical problems, such as corrosion and reduced water flow in technical water systems.

Removal of a biofilm is very tedious and sometimes even impossible because bacteria in

biofilms are resilient towards antibiotic and biocides. It is therefore desirable to be able to

prevent biofilm formation, rather than attempting to eliminate biofilms after they form. The

formation of a microbial biofilm on a surface can be prevented by creating unfavourable

conditions for the reversible, initial attachment of microbial cells. This effect can be obtained

by grafting hydrophilic polymeric chains onto surfaces and thereby provide a steric barrier

between the substrate surface and the microbial cell. Poly (ethylene glycol) (PEG) is one of

the most widely used polymers for making non-adhesive coatings. The work presented in this

thesis involves grafting PEG chains onto surfaces using different modifications of the ‘grafting

to’ technique.

The main aim of studies presented in this thesis was to develop surfaces which would

prevent bacteria from forming biofilm. The work focuses on novel strategies to self assemble

PEG thiol monolayers with high graft density. One of the strategies investigated involved

Page 4: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

iv | P a g e

backfilling a self assembled layer of 2000 Da PEG thiol with shorter oligo (ethylene glycol)

(OEG) thiol molecules to form a mixed monolayer. Detailed quantitative characterization of

the backfilling process was carried out using x-ray photoelectron spectroscopy (XPS), contact

angle measurements, and atomic force microscopy (AFM). These studies helped in

understanding the arrangement of backfilled molecules and the extent of desorption of PEG

molecules during the backfilling process. The PEG SAM backfilled with OEG molecules was

tested for resistance towards serum adsorption and bacterial attachment, and was found to

better resist fouling compared to PEG SAM that had not been backfilled.

The second strategy involved PEG grafting using supercritical carbon dioxide (SFC),

which is known to possess robust solvation properties. The idea was to demonstrate ‘cloud

point’ type grafting during self assembly using SFC, and produce PEG SAM of high graft

density. The SFC based PEG grafting reported here is the first of its kind, and the results

demonstrated that SFC is a good solvent for polymer grafting. The fouling properties of such

layers were ascertained by quantitative protein adsorption studies and bacterial attachment

studies. The detailed surface characterization of grafted polymeric layer and antifouling

studies helped in development of novel ways to create antifouling surfaces which have the

potential to counter problems caused by biofilms.

Page 5: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

v | P a g e

Table of Contents

List of publications ...................................................................................................... vii-viii

List of abbreviations ....................................................................................................... ix-x

INTRODUCTION ................................................................................................................ 1-2

CHAPTER 1: BACKGROUND ......................................................................................... 3-15 I. Mechanism of biofouling ......................................................................................... 3 II. Antifouling strategies ............................................................................................... 4 III. The physical forces involved in interfacial phenomena, physicochemical

interactions .............................................................................................................. 9 IV. Self assembly and grafting of non-fouling polymeric chains.................................. 10

CHAPTER 2: EXPERIMENTAL TECHNIQUES ............................................................. 16-29 I. Introduction to surface characterization in biointerfaces ....................................... 16 II. X-ray photoelectron spectroscopy ......................................................................... 18 III. Quartz crystal microbalance with dissipation ........................................................ 23 IV. Contact angle based surface energy measurements ............................................ 26 V. Atomic force microscopy ....................................................................................... 27 VI. Ellipsometry .......................................................................................................... 28

CHAPTER 3: Mixed poly(ethylene glycol) and oligo(ethylene glycol) layers created by backfilling ...................................................................................................................... 30-51

I. Introduction ........................................................................................................... 30 II. Materials and methods .......................................................................................... 33 III. Results and discussion ......................................................................................... 37 IV. Summary and conclusions .................................................................................... 50

CHAPTER 4: Immobilization of active subtilisin to mixed poly(ethylene glycol) and oligo(ethylene glycol) layers .......................................................................................... 52-77

I. Introduction .......................................................................................................... 52 II. Materials and methods .......................................................................................... 58

Page 6: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

vi | P a g e

III. Results and discussion ........................................................................................ 62 IV. Conclusion and discussion .................................................................................... 76

CHAPTER 5: Grafting of poly(ethylene glycol) layers using supercritical CO2 ..... 78-96 I. Introduction ................................................................................................................................... 78 II. Materials and methods .......................................................................................... 81 III. Results ................................................................................................................. 83 IV. Discussion and conclusion .................................................................................... 94 V. Future directions ................................................................................................... 96

CHAPTER 6: Summary of thesis ................................................................................. 97-99

Acknowledgments .................................................................................................... 100-101

Bibliography ............................................................................................................. 102-113

APPENDIX Arcot R. Lokanathan, Shuai Zhang, Viduthalai R. Regina, Martin A. Cole, Ryosuke

Ogaki, Mingdong Dong, Flemming Besenbacher, Rikke L. Meyer and Peter Kingshott.

Mixed monolayer formation by backfilling of thiolated poly (ethylene glycol) monolayer with

oligo (ethylene) glycol terminated alkane thiol. Biointerphases, Submitted.

Page 7: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

vii | P a g e

List of Publications

Publications that form the basis of this PhD thesis:

1. Arcot R. Lokanathan, Shuai Zhang, Viduthalai R. Regina, Martin A. Cole, Ryosuke Ogaki,

Mingdong Dong, Flemming Besenbacher, Rikke L. Meyer and Peter Kingshott. Mixed

monolayer formation by backfilling of thiolated poly(ethylene glycol) monolayer with oligo

(ethylene) glycol terminated alkane thiol Biointerphases, submitted

2. Arcot R. Lokanathan, Viduthalai R. Regina, Martin A. Cole, Ryosuke Ogaki, Flemming

Besenbacher, Rikke L. Meyer and Peter Kingshott. Cloud point grafting of thiol

functionalized poly(ethylene glycol) chains using supercritical carbon dioxide. In preparation,

to be submitted to Journal of supercritical fluids

3. Arcot R. Lokanathan, Viduthalai R. Regina, Martin A. Cole, Ryosuke Ogaki, Flemming

Besenbacher, Rikke L. Meyer and Peter Kingshott. Antifouling enzyme immobilized on

poly(ethylene glycol) monolayer with oligo(ethylene glycol) monolayers. In preparation, to be

submitted to Journal of Biointerphases.

Publications to which I have contributed as part of the project "Nano- and bio-

functionalised surfaces for biofilm prevention". These publications are not included in the PhD

thesis and will not be discussed in the thesis.

1. Arcot R. Lokanathan, Ryosuke Ogaki, Flemming Besenbacher, Rikke L. Meyer and

Peter Kingshott. Orbital shaking induced self assembly of colloidal domes. In

preparation.

2. Viduthalai R. Regina, Arcot R. Lokanathan, Peter Kingshott and Rikke L. Meyer.

Development of sol-gel coatings towards functionalization of antibacterial enzymes for

antifouling applications. In preparation.

3. Viduthalai R. Regina, Arcot R. Lokanathan, Peter Kingshott and Rikke L. Meyer.

Influence of surface physico-chemistry on the extracellular DNA mediated initial adhesion

of Staphylococcus xylosus. In preparation.

Page 8: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

viii | P a g e

Conference publications

1. Arcot R. Lokanathan, Viduthalai R. Regina, Rikke L. Meyer and Peter Kingshott

(2011). ‘Formation of underbrushes on thiolated Poly(ethylene glycol) PEG monolayers

by Oligoethylene glycol (OEG) terminated alkane thiols on Gold’, Australian colloid and

interface symposium, OC126.

2. Arcot R. Lokanathan, Viduthalai R. Regina, Rikke L. Meyer and Peter Kingshott

(2011). ‘Grafting poly(ethylene glycol) chains for antifouling purposes using

supercritical CO2’, 32nd Australian polymer symposium, 0021.

Page 9: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

ix | P a g e

List of Abbreviations

AFM – atomic force microscopy AHL – N-acyl homoserine lactones (AHL)

BSA – bovine serum albumin DNA – deoxyribonucleic acid DNase – deoxyribonuclease DOPA – dihydroxyphenylalanine ESCA – electron spectroscopy for chemical analysis EPS – extracellular polymeric substance FBS – fetal bovine serum HAS – hemispherical analyzer LCST – lower critical solution temperature Lyz – lysozyme MEMS – microelectromechanical devices MW – molecular weight OEG – oligo(ethylene glycol) PAA – poly(acrylic acid) PB – polybutylene PBS – phosphate buffer saline PDMS – poly(dimethylsiloxane) PE – polyethylene PEG – poly(ethylene glycol) PEI – poly(ethylenimine) PEO – poly(ethylene oxide) PHEA – poly(hydroxy ethylacrylate) PP – polypropylene PPO – poly(propylene oxide) PLA – poly(lactic acid) PLL – poly-L-lysine PLL-g-PEG – poly(l-lysine)-g-poly(ethylene glycol) PMMA – poly(methyl methacrylate) pNa – para-Nitroaniline PP – polypropylene

Page 10: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

x | P a g e

PU – polyurethane PVC – polyvinyl chloride QCM-D – quartz crystal microbalance with dissipation QNM – quantitative nanomechanical mapping SAM – self assembled monolayer SPR – surface plasmon resonance TSB – tryptic soy broth UV – ultraviolet XPS – x-ray photoelectron spectroscopy

Page 11: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

1 | P a g e

INTRODUCTION

Biofouling processes on surfaces initially involve formation of an adsorbed layer (also

known as a conditioning layer) of biomolecules, such as peptides, proteins and

polysaccharides. This layer is speculated to facilitate attachment of microorganisms and

multicellular organisms, eventually leading to biofilm formation [3, 4]. Biofilms are involved in

many infections, such as urinary tract infection, respiratory diseases, dental plaques, cystic

fibrosis, and failure of medical devices such as catheters, contact lenses, prosthetic joints and

other implants [5, 6]. Biofilm formation is a serious problem in water distribution systems [7],

reverse osmosis membranes of salt water treatment plants [8], food processing, and

transportation equipment [9, 10]. Macrofouling on ship hulls increases drag, and problems

caused by biofilms increase the maintenance costs in the oil industry [4]. Understanding the

fundamental aspects of complex biological processes leading to biofouling would help in

developing novel strategies to prevent biofouling.

The use of strategies to prevent fouling in marine applications dates back to 300 BC

where sheets of heavy metal were used on wooden boats [11]. Medical applications,

however, were first recorded in the early 20th century when attempts to use dental implants,

artificial valves, and prosthetic bone parts started [12]. The first biocompatible polymeric

material was discovered accidentally during the second world war when Ridley, an English

eye surgeon, noticed that splinters from canopies of fighter planes made of PMMA inside

eyes of pilots did not cause irritation [13, 14]. Systematic research on in vivo applications of

natural and synthetic polymeric coatings in the field of biomedical engineering started during

the latter half of the 20th century. Although marine and medical antifouling research fields

developed independently until the latter half of the 20th century, introduction of strict

regulations to preserve ocean ecosystems forced the marine antifouling research groups to

opt for environmentally safe and non-toxic technologies similar to those used in medical

research. Prevention of microbial growth by using antifouling coatings is becoming

increasingly popular in food processing and packaging. The antifouling technologies used in

the food industry obviously need to be non-toxic. These complex requirements for designing

antifouling surfaces need an in-depth understanding of interfacial phenomena involving

Page 12: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

2 | P a g e

interactions of biologically relevant systems with commercially relevant surfaces. During the

20th century, there were many scientific breakthroughs, such as establishment of laws and

principles governing behavior of light, electrons, atoms, molecules and polymers.

Consequently the last few decades of the 20th century witnessed a paradigm shift in the field

of surface science (characterization, design and fabrication) that enabled the understanding

of bio-interfacial phenomena at the molecular level, allowing the design of novel materials by

manipulation of the chemical and physical properties at micro and nano dimensions [3, 15-

17]. The advent of the field of Nanoscience, aided by advances in biotechnology and

advanced analytical instrumentation made the challenging field of biomedical engineering all

the more appreciable and approachable.

This thesis titled ‘Strategies for creating antifouling surfaces using self assembled

poly(ethylene glycol) thiol molecules’ is a collection of studies performed as a part of my three

year PhD project with an aim to develop novel ways to counter protein adsorption, bacterial

attachment and eventually biofilm formation. The scientific principles behind design of

strategies used in development of antifouling surfaces are discussed in Chapter 1. The

project involves developing ‘grafting to’ techniques to maximize the grafting density of

thiolated poly(ethylene glycol) (PEG) self assembled monolayers (SAMs) on gold substrate,

the details of which are discussed in Chapters 3 and 5. Apart from using just the PEG

monolayer, the possibility of using immobilized antifouling protease enzyme was investigated.

The results of enzyme immobilization studies using subtilisin are discussed in Chapter 4. The

thiol based SAM is one of the most popular model systems when it comes to understanding

fundamental aspects of biointerfacial processes. The PEG SAMs were characterized using

surface sensitive techniques; x-ray photoelectron spectroscopy (XPS), ellipsometry, and

surface energy measurements. The antifouling properties of surfaces were tested using single

protein or serum adsorption quantified by quartz crystal microbalance with dissipation (QCM-

D), and bacterial attachment assays. The experimental techniques are described in Chapter

2. The summary of conclusions derived from all the studies performed during my PhD along

with future directions are presented in Chapter 6.

Page 13: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

3 | P a g e

CHAPTER 1: BACKGROUND

I. Mechanisms of biofouling

Microbial biofouling is a multistage process. The first stage is molecular fouling or

conditioning layer formation involving the adsorption of inorganic and organic molecules on

surfaces [4, 18]. The second stage is microfouling, where the biofilm is formed by attachment

of single-cell organisms, such as bacteria and algae. In marine environments, the

microfouling step is followed by macrofouling, which results in attachment of higher

multicellular organisms such as barnacles [4, 18, 19]. The cascade of biophysical processes

in biofilm formation closely resembles the processes that lead to prosthetic implant

encapsulation in mammals. Once an implant is placed inside the body, proteins adsorb to the

surface, followed by attachment of neutrophils, macrophages and finally fibroblasts, that

encapsulate the implant with collagen [3]. The bacterial biofouling process can simultaneously

occur often interfering with the success of prosthetic implants in humans. If bacterial cells

succeed to attach onto an implant before encapsulation, it becomes virtually impossible to

cure the bacterial infection. Although the time scale of bacterial microfouling is different from

that of implant encapsulation, these two processes have very similar onset mechanisms

involving conditioning layer formation followed by interactions of cells with surfaces as shown

in Figure 1.

In spite of detailed studies in the field of ‘cell attachment to surfaces’ spanning over five

decades, our ability to gain absolute control over the fouling processes still remains a major

challenge. A reason for this may lie in the lack of detail in our understanding of how microbes

attach to abiotic surfaces. Initial studies were primarily focused on the role of a few

extracellular proteins or polysaccharides, but new findings in the last decade revealed that

other biomolecules, such as extracellular DNA can also be crucial for bacterial attachment

[20]. The biological control mechanisms involved in switching from planktonic to a sessile

lifestyle is another part of the field that awaits important discoveries. Some mechanisms are

controlled through quorum sensing systems, which were discovered four decades ago [21].

Until the beginning of the 21st century quorum sensing signaling molecules were thought to be

Page 14: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 1 – Mechanisms of Biofouling

4 | P a g e

present only in a few bacterial species [21]. Now microbiologists believe that most, if not all,

bacterial species utilize quorum sensing during biofilm formation, and these findings have

triggered a host of studies trying to relate quorum signaling to initial attachment of bacterial

cells [22]. A complete understanding of the biological and physicochemical processes

involved in fouling processes will help in designing surfaces with the potential to successfully

counteract biofouling. Various strategies employed to counter fouling problems will be

discussed in the next section.

Figure1. A simplified schematic representation of the stages leading to biofouling

caused either by prokaryote (for example bacterial) biofilm formation or encapsulation of

prosthetic implants by phagocytes.

II. Antifouling strategies:

Various strategies have been used to prevent biofouling and target specific stages

during the fouling development. Broadly, the strategies can be put into the categories listed in

Figure 2. Anti-adhesion coatings made by chemical or physical adsorption of hydrophilic

polymer molecules work by presenting a steric and/or hydration barrier between the

underlying substrate and the proteins and/or cells in solution above the surface, thereby

preventing the reversible initial attachment. There are several types of coatings which are

used to prevent protein adsorption and fouling. Anti-adhesive biomaterials could be of natural

origin, such as dextran, BSA, chitosan, alginate, hyaluronic acid and mannitol; important

A

B

C

A – conditioning layer formation

B – reversible physicochemical attachment of cells

C – irreversible adhesion basedcolonization

Page 15: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 1 – Antifouling strategies

5 | P a g e

alternatives to natural polymers are organic molecules such as PEG, PEO and

polyacrylamides which are prepared synthetically [13, 14, 23-30]. Zwitterionic molecules such

as sulfobetaine [31] and phosphorylcholine [32] have been demonstrated to have resistance

towards protein adsorption and cell attachment. The ‘cell adhesion interfering’ surface on the

other hand interferes with the biological process responsible for cell adhesion, but does not

kill the microbe. Post microbial attachment strategies also highly relevant in countering

biofilms. The degradation of biomolecules responsible for inter cellular adhesion which hold

the microbial colony together would result in detachment of cells from biofilm and eventually

the removal of the entire biofilm. The studies presented in this thesis involve proteolytic

enzymes immobilized onto an anti-adhesive hydrophilic coating.

Figure 2. Classification of approaches used to make antifouling surfaces [4, 20, 33-

35].

Various physical and chemical properties of polymers need to taken into consideration

in order to be chosen as candidates for a specific application. Biocompatibility, structural and

chemical stability, biodegradability, glass transition temperature, charge, isoelectric pH are a

few of the important properties of polymers that influence their interactions with biological

environments. All the above mentioned naturally occurring polymers are biodegradable, while

most synthetic polymers are often non-biodegradable. Synthetic polymers can be tailored to

be biodegradable. For instance PMMA is non-biodegradable while PHEA is biodegradable,

and other important examples are the development of biodegradable polyurethane [36],

polylactic acid and polyglycolic acid, and their copolymers. Biodegradability of a polymer

would prevent its accumulation in biological systems thereby making it environmentally safe

and also degradation in response to specific stimuli such as presence enzymes capable

Antifouling

Anti-adhesion: Hydrophilic, hydrogel coatings

Cell detachment: Protease, DNase, Self cleaning

Cytocidal: Silver ion, Nitrous oxide or Reactive oxygen species releasing, Polycation, Antibiotic, Antimicrobial peptides

Cytostatic: Quorum sensing interfering

Cell adhesion prevention: Protease, Dnase, Glycosidases

Page 16: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 1 – Antifouling strategies

6 | P a g e

cleaving bonds binding the monomers could used in site specific drug delivery. It should also

be noted that biodegradability could compromise the stability of antifouling coatings thereby

rendering the surface incapable of resisting fouling. All of the above mentioned polymers have

the disadvantage that they are not completely successful at preventing biofouling. Duracher

et al. demonstrated that PNIPAM brush coated surfaces were protein resistant below its lower

critical solution temperature (LCST) of 32 °C and non-resistive above the LCST, thus making

it unusable at in vivo temperatures [37]. Futamura et al and Yamasaki et al. studied coatings

made of methacrylate polymeric chains terminating with phosphorylcholine groups and found

that the phosphorylcholine group got embedded in the hydrophobic interior of the SAM upon

drying whereas in wet conditions it was located the solvent-SAM interface [38, 39]. They

observed that these surfaces once dried, needed long wetting pretreatment or prehydration

(10h) times in order for the phosphorylcholine groups to reorient after reaching equilibrium,

otherwise the surfaces were less efficient in resisting protein adsorption when compared to

prehydrated surfaces. Therefore, the use of surfaces coated with methacrylate functionalized

phosphorylcholine is not suitable for applications that cannot afford long pretreatment

procedure. The ethylene glycol based polymers are prone to auto-oxidation in the presence of

oxygen and transition metal ions [40]. Also hydroxyl end-groups, if present, are converted

onto aldehydes by alcohol dehydrogenase, initiating further reactions that make these

coatings inefficient at resisting protein adsorption [40]. Among the various types of surface

coatings mentioned above, ethylene glycol based coatings such as PEO, PEG, OEG coated

surfaces are one of the most widely studied, well characterized non-fouling systems which

have been shown to be effective in reducing protein adsorption [41], bacterial attachment

[42], and in vivo leukocyte attachment [43].

The exact mechanism by which PEG layers resist non-specific protein adsorption still

remains unclear, but there are few speculations based on detailed experimental and

theoretical studies. The earliest attempt to explain the nonfouling properties of PEG was

made by De Gennes and Andrade [44, 45]. They argued that when a protein molecule

approaches a surface coated with PEG chains, the water molecules bound to the PEG

through hydrogen bonding have to be expelled due to compression. Since expulsion of bound

water molecules is energetically unfavorable, the PEG molecules do not allow the protein

Page 17: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 1 – Antifouling strategies

7 | P a g e

molecules to reach the substrate. Although this model was partly successful in explaining

systems with high molecular weight PEG, it could not explain the results of studies using

oligo(ethylene glycol) (OEG) (n<6) based self assembled monolayers (SAM) [46]. Whitesides

et al. studied OEG systems, alkane thiol SAMs with various hydrophobic and hydrophilic end

groups. Based on their studies they concluded that inertness of a surface is not only the

property of hydrated polymeric layers but this could also be due to the structured water layer

[47]. The structured water layer could be formed due to orientation of water molecules by

dipole moments (extending over 3-4 layers of water molecules) from hydration layer at

interface as suggested by Grunze et.al. based on computer simulations [48]. They also

experimentally showed that the conformation of OEG was a very important structural aspect

responsible for resistance towards protein adsorption. The OEG SAMs formed on Au were

protein resistant, while on Ag surface the SAM was not resistant to protein adsorption. The

constrained OEG molecules in SAMs formed on Ag, due to higher packing density could not

attain a helical conformation, whereas the OEG molecules on Au could attain helical

conformation and hence protein resistance [46]. The helical conformation enables the

hydrogen bonding of water molecule with oxygen atom of polyether chain. Based on

calculations using single chain mean field theory, Szleifer proposed that loss of

conformational entropy of grafted polymeric chains during approach of protein molecules is

responsible for the repulsive protein-polymeric chain interaction [49, 50]. Despite all the

efforts to ascertain the precise mechanisms of such coatings, a universal explanation of the

antifouling properties of nonionic coatings is still the subject of much conjecture and research

effort. In addition, translating the theoretical findings into experimental coatings, including

stability issues, has proved challenging to surface science.

Biofilms in general consist of 2-5% by volume of cells with the rest being extracellular

polymeric substances (EPS) [51]. Major components of EPS are polysaccharides,

extracellular DNA and glycoproteins [52, 53]. The EPS is a major obstacle in biofilm removal

efforts, and it is one of the reasons for the failure of most antimicrobial strategies aimed at

killing biofilms. EPS acts as a physical barrier thereby minimizing the diffusion of antimicrobial

molecules into the biofilm. Efficient biofilm removal there requires much higher concentrations

of toxic biocides than what is needed to kill planktonic cells [35]. Thus simple and safe

Page 18: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 1 – Antifouling strategies

8 | P a g e

strategies to prevent biofilm formation rather than using harsh treatments to remove biofilm

are highly desirable. Non-adhesive coatings made of hydrogels and hydrophilic coatings have

been demonstrated to minimize microbial biofilm formation in short term experiments.

Nejadnik et al. reported a coverage of approximately 80% (surface coverage) after 20h

incubation with Staphylococcus aureus on a PEO-PPO block copolymer coating on a silicone

substrate [42], and found that uncoated surfaces attained 80% coverage in just 6h, thus

demonstrating that the polymeric coating delayed the colonization. Saldarriaga et al.

conducted attachment studies using five species of bacteria incubated for 168h (isolated from

implants) on surfaces coated with a crosslinked PEG polymer [54]. They compared the effects

of exposure of these surfaces after exposure to bio fluids such as serum, saliva, urine and

tears on the bacterial adherence. Their best surface could reduce bacterial attachment by

80% when compared to the glass control surface. A perfectly successful antifouling surface

with long term ability to prevent microbial colonization remains a challenge to this day.

The success of bacterial attachment onto surfaces depends mainly on the integrity of

EPS as discussed earlier. Damaging the EPS is also an important strategy in preventing

bacterial attachment and biofilm removal [34, 51, 55]. This can be achieved by using enzymes

such as lysozyme, trypsin, DNase, α-amylase and proteins such as lactoferrin [20, 55-57]. All

the enzymes work by cleaving bonds of the polymeric chains of EPS. A detailed introduction

on the specific mechanisms involved with various classes of antifouling enzymes will be

discussed in Chapter 4. Apart from using anti-adhesive PEG coating, I also used a covalently

immobilized protease. Proteases damage the membrane proteins thereby interfering with the

initial bacterial attachment. Among the various proteases, subtilisin was chosen as a model

enzyme to perform immobilization studies on the PEG and OEG mixed surfaces created by

backfilling. The details of enzyme based studies are presented in Chapter 4. Protein

adsorption, bacterial attachment and enzyme immobilization involve interactions of

biomolecular systems with surfaces. In order to control these processes one has to

understand the factors governing the interactions, and this aspect is discussed in the next

section.

Page 19: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 1 – The physical forces involved in interfacial phenomena, physiological interactions

9 | P a g e

III. The physical forces involved in interfacial phenomena, physicochemical interactions

The interactions of all biomolecular systems with their environments are driven by the

fundamental forces of nature that can be broadly classified into long and short range forces.

The main non-covalent interaction based forces relevant to biomolecular systems are

electrostatic or (acid-base) interactions, van der Waals forces, hydrogen bonding, dispersive

forces and hydration forces [58, 59]. In the case of microscopic systems gravitational,

Brownian and hydrodynamic forces also play major roles in their behavior [60]. The forces

dominating the behavior of a system are determined by the length scales they influence. For

example when considering intermolecular interactions in the case of proteins having

nanometer scale dimensions (<10 nm), hydration forces play a significant role, but in the

case of microscopic systems, van der Waals and double layer forces dominate [61]. The

simple laws of thermodynamics (minimization of energy, maximization of entropy) that apply

to chemical systems also govern interfacial biophysical phenomena, but it is difficult to gain a

comprehensive theoretical and experimental understanding of these phenomena due to the

complex interplay of fundamental forces involved [62]. Apart from the system itself, the

solvent properties also change the way a biomolecule interacts with a surface. Some of the

main solvent properties relevant to interfacial adsorption processes are listed below:

pH- The state of ionization of functional groups such as carboxyl, amine, phosphate,

hydroxyl, thiol and other groups at a given pH depends on their pKa values. Thus the surface

charge and also charge of biomolecular systems vary depending on the pH. The efficiency of

enzyme immobilization and grafting of polymers is also greatly influenced by pH.

Ionic strength- The Debye length of a charged surface is inversely proportional to the

ionic strength of the solution. Whether or not the electrostatic interactions dominate the

intermolecular and molecule-surface interactions depends on the Debye length, and hence

the ionic strength [63, 64].

Temperature- Changes in temperature can influence stability of weak bonds, solubility,

and also alter the kinetic vs thermodynamics competition in chemical reactions, and is also

relevant in more complex systems such as protein adsorption [65, 66].

The generally accepted mechanism of biomolecular adsorption starts with attraction to

Page 20: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 1 – The physical forces involved in interfacial phenomena, physiological interactions

10 | P a g e

the surface due to long range attractive forces such as van der Waals forces, which could be

acting against or together with electrostatic forces [59, 62]. Reversible attachment is formed if

the energetics of interactions is favorable. This reversible attachment paves the way for

dispersive hydrophobic interactions based irreversible adsorption. An important implication for

immobilization of bioactive proteins such as enzymes on bare substrates are hydrophobic

interactions that can cause conformational changes that could render the molecule inactive

[67]. The denaturation of immobilized molecules can be minimized by immobilization to

hydrogels or hydrophilic polymer layers thereby shielding the hydrophobic patches of

substrate. In real environments such as those encountered in biomedical applications or in

the food and marine industries is the complex mixture of biomolecules present that result in

competitive adsorption phenomena. To begin with the most abundant proteins adsorb to a

maximum extent, but as system reaches equilibrium, the proteins with higher affinity tend to

replace the loosely adsorbed proteins, this is known as the Vroman effect for blood contacting

devices [68, 69]. Once we understand the physicochemical nature of surface-biomolecular

interactions, the next step would be to design surfaces that can control adsorption. The forces

relevant to adsorption of molecules are also highly relevant to polymeric self assembly

processes that are used in numerous applications such as making antifouling surfaces, which

is the main theme of this thesis. The forces relevant to polymeric self-assembly process are

discussed in the next section.

IV. Self assembly and grafting of non fouling polymeric chains

Surfaces grafted with polymer layers have a wide range of applications such as self

cleaning coatings, entanglement of surfaces, antifouling, anti-corrosive, high temperature

resistance, cell patterning, tissue engineering, biomolecular immobilization, sensing devices

involving protein or DNA microarrays and NEMS/MEMS devices [4, 70-76]. The performance

of a surface coated with polymer molecules in a specific application indeed depends on the

chemical and physical properties of the polymer used. Apart from the choice of polymer there

are several properties of a coating that make it suitable for a specific application, such as the

nature of polymer-substrate interactions (covalent, non-covalent), molecular weight of the

polymer chains (linear or branched), conformation of the polymer on the surface, and the

Page 21: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 1 – Self assembly and grafting non fouling polymeric chains

11 | P a g e

pinning or graft density (chains and end groups per unit area). There are numerous types of

substrate-head group chemistries that could be used for grafting polymers onto surfaces

depending on the availability of reactive chemical groups on the polymer. The dependence of

functional stability of SAMs as a function of physicochemical properties of the polymeric

molecule was discussed in the ‘Antifouling strategies’ section. The nature of substrate-head

group interactions in SAMs is also a very important aspect in regard to the successful

application of SAMs. Various kinds of substrates, modification procedures and substrate-

polymer interactions are listed below.

a. Substrate

i. Metal/metal oxide: Au, Ag, Pt, Pd, Ta; oxides of Ti, Fe, Sn, Zr, V, Hf [77-79]

ii. Organic polymers: PS, PE, PP, PB, PMMA, PDMS, PU, PLA, PVC [80-84]

iii. Ceramics: alumina, zirconia, silicate glass, calcium phosphate, calcium

carbonate [85]

b. Substrate modification

i. Treatments: Concentrated acid or base, plasma, corona discharge, UV, ozone,

flame [84]

ii. Chemical modification using bifunctional linkers, SAMs with desired tail or end

groups, terminating polymerization using required end groups in ‘grafting from’

techniques [80, 81, 84, 86]

c. Substrate-polymer interactions

i. Covalent: Amide (amine-carboxyl, amine-maleic anhydride, azide- aromatic

ester with phosphine group at ortho position), reductive amination (amine-

aldehyde), epoxy-amine or thiol or hydroxyl, thioether (thiol-benzyl or acetyl or

acetyl halide, click chemistry (alkyne-azide, thiol-ene), siloxane (silanol-

hydroxyl), thiol-maleimide/Au/Ag/Pt/Pd, Acid-base (metal oxide-carboxylic or

phosphonic acid) [41, 81, 87, 88]

ii. Electrostatic: Sulfonate on PU, PLL and PEI on oxides [89, 90]

iii. Biomimetic: Catechol and titanium binding aptamer peptides [91-93]

Surfaces of metal or semiconductor oxide generally have hydroxyl groups that are

crucial for a number of chemical modifications and their surface density can be increased by

Page 22: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 1 – Self assembly and grafting non fouling polymeric chains

12 | P a g e

oxidation procedures such as wet chemical treatments, plasma and UV-ozone [94]. Although

organic polymers can be tailored during synthesis to yield surfaces with suitable reactive

groups, post synthesis surface modifications are very popular in polymer functionalization. For

example, PE surfaces upon UV ozone treatment produce carbon species corresponded to

ether, aldehyde, carboxylic carbon [95] and silanol groups are produced on surface of PDMS

using the same treatment, as determined by XPS [96]. Plasma based surface modification

and polymerization is a versatile tool for biomedical applications [97]. For example plasma

polymerization on silica surfaces using allylamine, heptylamine, acetaldehyde produced thin

films with amine and aldehyde groups, respectively, which could be further functionalized

using reductive amination or carbodiimide chemistry as reported by Griesser et al. [98]. In

addition to grafting non-fouling polymers, as demonstrated by Cole et al. [99], the plasma

polymerization technique when combined with masked lithography type procedures is

capable of creating two dimensional chemical patterns which can help in creating arrays of

cells or biomolecules as reported by Thissen et al [100]. Functionalization of surfaces using

SAMs is also a popular approach to tailor the surface chemical groups. For example Seifert et

al. used biotin terminated alkane thiol SAMs to immobilize streptavidin [101], and in a similar

fashion alkyl silanes are mostly used for the functionalization of oxide surfaces [86].

The strength of physical or chemical interaction between the substrate and head group

dictates the stability of SAMs in different environments. Covalent based coupling in general is

more stable in comparison with electrostatic, hydrophobic, van der Waals interactions.

Kingshott et al. demonstrated that PEG chains coupled to covalently immobilized PEI

surfaces were more efficient in resisting bacteria when compared with a physisorbed PEI

surface [102]. The instability of SAMs is mainly due to desorption due to competitive

adsorption or degradation of head group. Thiol based functionalization of noble metals has

limited stability in ambient or physiological conditions due the oxidation of thiol to sulfate,

which has lower affinity towards noble metals. Cerruti et al. showed that PEG thiol SAMs on

Au were prone to thiol oxidation driven desorption upon exposure to air, whereas PEG silane

based SAMs on silicon nitride were stable for over two months [103]. Kanta et al. studied the

stability of octadecyl phosphonic acid SAMs on titanium surfaces in various solvents and

found that the SAMs were not stable in water, ethanol and methanol [104]. Electrostatic

Page 23: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 1 – Self assembly and grafting non fouling polymeric chains

13 | P a g e

interactions are screened in the presence of high salt concentrations such as situations in

marine or physiological environments. Mani et al. showed that there was considerable

desorption when SAMs of dodecyl phosphonic acid was exposed to Tris buffered saline at 37

°C while the deodecyl trichloro silane based SAM was stable [105]. The poly(l-lysine)-g-

poly(ethylene glycol) (PLL-g-PEG) SAMs are used for the functionalization of a range of

negatively charged surfaces based on electrostatic attraction and are very efficient in resisting

protein adsorption. These electrostatic attraction based interactions were demonstrated to be

unstable under high salt conditions [106]. Blattler et al. showed that covalent based coupling

of PLL-g-PEG onto aldehyde groups created by plasma polymerization make the SAM stable

at high salt concentrations [106]. A few research groups are also involved in development of

stable, universal binding functional groups that are capable of binding to all surfaces. This

endeavor gets its inspiration from naturally occurring 3,4-dihydroxyphenylalanine (DOPA) rich

mussel adhesive proteins, which are efficient in binding to most surfaces [107]. Catechol

functionalized PEG molecules were developed trying to mimic the mussel adhesive protein.

Malisova et al. reported detailed studies on surface coverage, serum resistance ability of

various types of catechol (different acidity) and nitrodopamine functionalized PEG SAMs on

oxide surfaces [108].

SAMs of alkane thiols on gold are one of most widely studied self assembly systems.

The typical structure of an alkane thiol SAM is shown in Figure 3a with the labels of the most

important parameters of the SAM. The intermolecular van der Waals interactions make SAMs

of alkane thiols crystalline and rigid when compared to SAMs of PEG, which are flexible.

Though intermolecular van der Waals interactions are absent in PEG monolayers, most

authors refer to the PEG functionalization process as molecular self assembly as long as the

PEG chain is bound to a moiety that anchors it to the substrate. A molecule capable of self

assembly must have a head group to interact with the substrate; a chain group; the end or tail

group which mainly controls the interfacial properties such as being helpful in immobilization

of biomolecules as shown in Figure 3b. The PEG graft density is the most important factor

determining the quantitative ability of PEG surfaces to resist fouling [109], and it has been

observed that there is an inverse relationship between the MW of PEG (also related to Flory

radius) and graft density [110] due to the ‘excluded volume effect’ [111]. The graft density of

Page 24: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 1 – Self assembly and grafting non fouling polymeric chains

14 | P a g e

PEG SAMs determines the conformation of polymeric chains, which could either be in brush,

mushroom or pancake conformations [44, 109, 111], with brush being the most effective

against fouling [110]. The relationship between the MW of PEG, Flory radius, and graft density

of a SAM are given by Equations 1, 2 and 3:

RF – Flory radius, a – monomer size in lattice model, N – number of monomers, L –

interchain distance, M – molecular weight of PEG chain, ρ – density, d – dry thickness, NA –

Avogadro number, S – graft density defined as number of chains per unit area.

Figure 3: (a) SAM of alkane thiol on a gold substrate with labels showing area per

molecule, d - intermolecular distance, tilt angle θt. (b) A SAM of PEG with enzyme molecules

immobilized on the end groups.

The PEG functionalization techniques can be broadly classified into ‘grafting to’ and

‘grafting from’ approaches (Figure 4). The ‘grafting from’ technique involves growth of

polymeric chains from initiators immobilized on a substrate (Figure 4b) and it generally gives

higher graft density since the limiting factor is diffusion of monomer molecules onto the

reactive ends of growing chains. This is unlike the case of the ‘grafting to’ technique where

the limiting factor is the diffusion of the entire polymer molecule to the reactive substrate

(Figure 4a) [112]. The limitation of the grafting-to technique of low graft density can be

overcome using various strategies such as ‘cloud point’ grafting [41], grafting in homopolymer

solutions [113], grafting from polymeric melts [114] and underbrush formation by backfilling

------ Eq (1); ------ Eqn (3)------ Eq (2);

Y Y Y Y Y Y YSubstrate

End or tailgroup

Chain orbackbone

Head group

Substrate

(a) (b)

Page 25: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 1 – Self assembly and grafting non fouling polymeric chains

15 | P a g e

[115]. In the ‘cloud point’ grafting technique solvents with high ionic strengths along with high

temperatures are used. As discussed in section (1c) under these conditions the Debye length

decreases and also the solubility of PEG is greatly diminished in solutions saturated with

salts. The high temperature used (higher than LCST of PEG) disrupts the hydrogen bonding

between water molecules and PEG, thereby further decreasing the solubility [116]. The

presence of ions has been shown to alter the solubility, also lowering the critical solution

temperature [117]. The extent of decrease in solubility of a non-electrolyte due to the

presence of an ion is directly related to its polarizability. Anions in general have higher

polarizability and thus are more important in applications such as ‘cloud point grafting’ where

the solubility of polymer is decreased [117]. Under these conditions the PEG chains exist in a

collapsed state rather than an extended conformation. Grafting under these conditions thus

results in higher graft density. The backfilling based technique unlike the other three

strategies that depend on minimization of excluded volume interactions, is a simple method

wherein the interchain spaces present in layers of higher MW PEG SAMs are backfilled with

shorter OEG chains that can diffuse to the surface. In this thesis two strategies were used for

maximization of the graft density; firstly backfilling using OEG which is discussed in Chapter 3

and secondly grafting under reduced solubility conditions using supercritical carbon dioxide,

as discussed in chapter 5.

Figure 4: (a) Schematic of the ‘grafting-to’ polymer grafting technique and, (b) Various

steps involved in the grafting-from technique.

(a) (b)

Initiatorimmobilization Polymerization Growth Termination

Page 26: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

16 | P a g e

CHAPTER 2: EXPERIMENTAL TECHNIQUES

I. Introduction to surface characterization in biointerfaces

Understanding the behavior of surfaces in biological environments requires thorough

physical and chemical characterization to correlate the observed biointerfacial events to the

properties of the material surface [118]. Some of the main properties of surface coatings that

influence biointerface phenomena are film thickness or coverage, conformation or orientation

of the constituents of the film, chemical composition, wettability or surface energy, charge,

optical properties, refractive index, surface topography and mechanical stiffness [118-120].

The quantification of the content of chemical groups on surfaces can be done directly or by

derivatization using suitable probe molecules [118-120]. Once the material surface has been

characterized, the next challenge would be to gain knowledge about the changes that occur

at the interface when the surface is exposed to a biological environment. Upon exposure to a

biological environment, the physical and chemical properties of the surface may change due

to a lack of stability of the interface; apart from the intrinsic changes at the interface, the

interface itself might get buried under a layer of adsorbing molecules, thereby creating a new

interface which then decides the fate of material surface in a given environment [12]. Thus,

the knowledge of quantitative and qualitative aspects of adsorption events occurring at

material interfaces exposed to complex biological environments is absolutely essential to

comprehend the behavior of materials in biological environments that can be used in order to

optimize the surface for improved performance [118-120]. Real-time characterization of

various changes occurring at an interface would be ideal, but many surface characterization

techniques are unable to analyze samples at ambient or under aqueous conditions. Table 1

gives an overview of some of the most widely used surface characterization techniques used

in biointerface science. The choice of technique is based on its ability to provide the required

information, optimal resolution and required sensitivity.

Page 27: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 2 – Introduction to biointerface related surface characterization

17 | P a g e

Table 1. Overview of commonly used surface characterization techniques [1, 2, 118,

120, 121]

Technique Vertical resolution

Lateral resolution

Sensitivity Remarks

AFM Surface < 1nm high Mainly visualization; ambient conditions, liquid conditions

Contact angle 3-20Å 1mm Low or high Swelling, extraction by solvent STM 5Å 1Å atoms Only conducting samples,

ambient conditions possible, no liquid studies

SIMS, ISS 10Å-1μm 500Å Very high Operates in vacuum, Sample damage (except static mode),

semi quantitative Ellipsometry ≅1nm 1mm High Operates in ambient condition

and liquid. Affected by surface roughness, scratches

SEM 15Å 40 Å High not quantitative

Operates in vacuum. Sample damage, sample preparation artifacts

XPS/ESCA 10-250 Å 10-150μm 0.1 atom% Operates in vacuum, capable of destructive and non destructive depth profiling

AES 50-100 Å 100 Å 0.1 atom% Severe sample damage, quantitation

Vibrational spectroscopies

HREELS

IRAS

ATR-IR Raman

10 Å 100Å 1-5μm >1μm

1μm 1μm

1mol%

High

Water signal subtraction

Limited for depths <1μm

The surface chemistry of materials can be determined using XPS, SIMS, ATR,

HREELS, while information about the distribution of groups require imaging techniques. The

knowledge of molecular orientation can be obtained from polarized IR, HREELS, NEXAFS,

Page 28: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 2 – Introduction to biointerface related surface characterization

18 | P a g e

and imaging techniques like SEM, AFM and STM provide information on the texture of

surface. The modulus information of surface can be obtained from AFM and the surface

energy deduced from contact angle measurements. There are several ways of classifying the

above mentioned techniques. One of the most important classifications is based on whether a

technique is capable of performing real-time, in situ studies or not. Among the above

mentioned techniques, ellipsometry, SPR, AFM and ATR-IR are capable of in situ studies

under physiological conditions involving biomolecular adsorption processes [122, 123].

Optical waveguide based techniques such as OWLS have also become very popular in

biointerfacial studies due to their high sensitivity and good resolution (range 1nm-5μm,

resolution 1Å)[124]. In this thesis QCM was used to perform real-time serum adsorption

studies on PEG coated surfaces. Also XPS was used to estimate the dry thickness of PEG

layer, adsorbed or immobilized protein films. The thickness estimation was also done using

ellipsometry to cross check the XPS based measurements. The thickness of thin films can be

estimated using other sensitive techniques such as stylus profilometry which has a resolution

of 5Å; interferometry which has a resolution of 30Å; using surface plasmon resonance (SPR),

resolution <1nm; [125-127]. In the following section of this chapter, the working principles

behind the techniques used in this thesis are presented.

II. X­ray photoelectron spectroscopy (XPS)

XPS is a quantitative surface spectroscopic technique, providing information about the

presence and quantity of the elements (except H and He), the chemical states of each of the

elements, and the lateral and vertical distributions. The photoelectric effect was discovered by

H.R. Hertz in 1887, and the theoretical basis was provided by Einstein in 1905. Though

several scientists studied the photoelectric effect, it was Kai Seigbahn who first demonstrated

that XPS can be used for quantitative chemical state analysis and coined the term ‘ESCA’

(electron spectroscopy for chemical analysis). This technique works by analysing the

photoelectrons that are ejected from the core level atomic shells of surface atoms during

exposure to incident x-rays as shown in Figure 1. The surface sensitivity of this technique is

due to the low attenuation lengths of photoelectrons, thus only the top few atomic layers of a

Page 29: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 2 – X ray photoelectron spectroscopy

19 | P a g e

surface contribute to the signal intensity. The relation between the incident photon energy and

the photoelectron is given by the photoelectric law given in Equation 4 [1, 2].

A x-ray photoelectron spectrometer typically consists of several components as shown

in Figure 5 [1]. The x-ray source consists of either a thin film of Al or Mg on a water cooled

copper anode, which is bombarded with electrons from a filament, producing x-ray lines with

1486.7 eV or 1254 eV, respectively, and a background due to Bremstrahlung radiation.

Monochromatic x-ray sources use a diffracting crystal to remove the background. For

Photoelectric law in XPS: EKE= hν - EBE - ΦSp ------ Eqn (4)

Where, EKE – kinetic energy of photoelectron,

hν– energy of incident photon, EBE – binding energy

of core electron, ΦSp – Work function of

spectrometer.

Figure 1. The photoionization process

Figure 2: Block diagram of components of a XP spectrometer [1, 2]

Page 30: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 2 – X ray photoelectron spectroscopy

20 | P a g e

example, quartz crystals are used to obtain the monochromatic Alkα line. In the case of

conducting samples, the positive charge developed during the photoemission process gets

discharged due to its electrical contact with the holder. The neutralization of positive charge

developed on an insulating material would require an external source of low energy electrons,

which is provided by a flood gun. An alternate solution uses thin grid of conducting material

deposited over the insulating surface. The ejected photoelectrons enter the analyser where,

depending on the mode of operation, they are either retarded to constant pass energy (fixed

analyser transmission or constant resolution mode), or analysed with varying detector pass

energies (fixed retard ratio mode). The most commonly used analyser is a hemispherical

analyser (HAS) [1]. The photoelectrons are counted by an electron detector that is generally a

channel electron multiplier (CEM) producing a cascade of electrons for each photoelectron

generated from the sample.

Generally two kinds of spectra are recorded, namely a wide energy scan or a survey

spectrum and high resolution spectra, which are shown in Figure 3. A survey spectrum has

several primary features, such as the background with step like appearance due to

inelastically scattered electrons, which are also the primary source of noise. Auger peaks also

appear, which can be differentiated from XPS peaks by altering the energy of incident x-rays.

Non-monochromated sources produce satellite peaks, and x-ray sources with eroded anode

films can produce ghost peaks from the underlying copper substrate. Photoemission peaks

from any orbital other than S orbitals always result in a doublet due to LS coupling; the

relative ratio of the doublet depends on the orbital, and difference in their binding energies

depends on the splitting constant. Aromatic molecules produce shake-up satellite peaks;

metallic surfaces produce plasmon loss peaks and also peak asymmetry is commonly

observed on the higher binding energy side of peaks due to loss of photoejected electron

energy. X-ray fluorescence is a process that competes with photoelectric emission. The

measured intensities from different elements obtained from a survey spectrum can be used to

calculate the relative atomic percentages of elements from the Equation 5 [1].

Where CA is the relative atomic concentration, IA is intensity due to element A and In is

------ Eq (5)

Page 31: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 2 – X ray photoelectron spectroscopy

21 | P a g e

the sum total of intensities from all the elements present on the surface, S is the relative

sensitivity factor.

Figure 3: Example of a survey spectrum and a high resolution C 1s spectrum recorded

from a SAM of COOH-PEG thiol on gold [data from Chapter 3].

The thickness of overlayer on substrates can be calculated by knowing the extent of

attenuation of the substrate signal using Equation (7), which can be integrated to give

Equation (8) [1, 2].

C 1

sO 1

s

Au

S 2p

x 105

2

4

6

8

10

12

CPS

1000 800 600 400 200 0Binding Energy (eV)

C 1x 103

55

60

65

70

75

80

85

CPS

296 294 292 290 288 286 284 282 280Binding Energy (eV)

Surveyspectrum

C 1s Highresolution spectrum

Au 4f C 1s

d = 3 λAL Sinθ ------ Eqn (6) Where, d is escape depth, λAL is PE attenuation length

and θ is take off angle wrt the normal to the surface.

The intensity emanating from atoms at

depth Z is given by:

Where IZ is intensity at Z, I0 intensity

from surface atoms of bulk material, λ and θ

are the same as in the Equation (6).

----- Eqn (7)

Figure 4. Schematic of a substrate being analysed in angle resolved XPS (modified from [1]).

Page 32: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 2 – X ray photoelectron spectroscopy

22 | P a g e

where Z - overlayer thickness, λ – attenuation length, θ is the take-off angle, I is the

substrate signal with thin film coating, I0 bare substrate intensity.

Alternately the overlayer thickness can be determined using signal from any of the

elements present in the overlayer (I), if the elemental signal from an infinitely thick overlayer

(IO) is known, given by Equation (9)

High resolution spectra allow the quantification of various chemical species of an

element with different oxidation states due to variations in chemical bonding environment

surrounding each atom, which can be explained by charge potential model [1]. An example of

a high resolution C 1s spectrum of a SAM of carboxyl capped PEG thiol is shown in Figure 3.

The surface sensitivity of XPS is determined by the inelastic mean free path (IMFP) of the

photoelectron (PE) or attenuation length (λAL),where the analysis depth is approximately three

times the escape depth, which is given by Equation (6) [1, 2]. By changing the take off angle

with respect to the analyser, the sampling depth can be varied thereby enabling compositional

depth profiles. Alternatively depth profiling can be achieved using high energy x ray sources

or using layer by layer sputtering.

In this thesis the thickness of grafted PEG and/or OEG layers on gold substrates was

calculated using Equation (8), using the gold signal. There is always a possibility that a

fraction of PEG molecules are physisorbed instead of being covalently bound. Castner et. al.

reported a way to estimate the proportion of physisorbed alkane thiol molecules in SAM on

gold using high resolution S 2p spectra [128]. The chemisorbed thiolated species on gold can

be distinguished from the physisorbed molecules based on the difference in core electron

binding energies of covalently bound and unbound sulphur atoms. An example of a peak-

fitted high resolution S2p spectrum is shown in Figure 5.

----- Eqn (8)

----- Eqn (9)

Page 33: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 2 – X ray photoelectron spectroscopy

23 | P a g e

Figure 5. An example of a high resolution S 2p spectrum recorded from a SAM of

octadecane thiol on gold, showing components corresponding to bound and unbound thiol

[128].

II. Quartz crystal microbalance (QCM)

QCM is a sensitive surface analytical technique capable of giving quantitative

information on interfacial processes resulting from changes in mass loading on a quartz

crystal. The theoretical and experimental basis of QCM was was proposed and demonstrated

in 1959 by Sauerbrey [129]. The use of QCM in the field of biointerfaces started in the 1980s

and has evolved into a valuable technique in understanding interfacial phenomena such as

protein adsorption, and is particularly useful for studying fouling processes. A typical QCM

sensor is shown in Figure 9a, consisting of a quartz crystal sandwiched between an active

and a counter electrode. The piezoelectric quartz crystal is actuated by applying a potential

across the electrodes. The frequency of vibration is sensitive to the mass of the interfacial

overlayer on the electrodes (Figure 6b). It enables quantification of changes in mass due to

adsorption or desorption occurring on the crystal using the Sauerbrey equation given in

Equation (9a) [43]. This equation is an approximation and assumes that the viscoelastic

Page 34: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 2 – Quartz crystal microbalance

24 | P a g e

properties of the interface remain unchanged while the process is being monitored. The use

of the Sauerbrey equation for systems with varying viscoelastic properties would result in

underestimations of mass. Performing studies using multiple frequencies and knowledge of

dissipation given by Equation 9b enables the accurate estimation of mass, and viscoelastic

properties of films using the Voigt model [130, 131].

Where, Δm – change in overlayer mass, C – 17.7 ng Hz-1 cm-2 for 5 MHz quartz

crystal, n – overtone number, Δf – change in frequency, D – dissipation, EDissipated – energy

lost, EStored – energy stored.

QCM has been used in a number studies in biomedical sciences, such as

biorecognition, cell-surface interaction studies, lipid bilayer formation and fouling processes

[91, 131, 132]. QCM is a sensitive and versatile tool to study the qualitative and quantitative

aspects of protein adsorption process on a variety of antifouling surfaces such as

oligo(ethylene glycol) [133, 134] and poly(ethylene glycol) [132, 135] SAMS. In this thesis,

QCM was used to ascertain the ability of PEG functionalized surfaces to resist protein

adsorption. Surfaces were tested with solutions of single proteins and complex bovine serum.

An example of a QCM plot involving fetal bovine serum (FBS) adsorption on various

substrates is given in Figure 7. The advantage of QCM is that it allows the studies to be

conducted without the need for any probe labels, at variable temperature, ambient pressure,

and importantly in aqueous conditions, which more closely mimic ‘real’ biological

environments. The results of protein adsorption studies using QCM are discussed in Chapters

3 and 5

----- Eqn (9a)

----- Eqn (9b)

Page 35: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 2 – Quartz crystal microbalance

25 | P a g e

Figure 6. (a) Shows the two sides of typical QCM crystal with the various electrodes

labeled. (b) Schematic of actuation of quartz layer sandwiched between electrodes [136].

Figure 7. An example of a QCM graph showing the change in frequency during the

adsorption of 10% FBS on SAMs of oligo(ethylene glycol) (OEG), poly(ethylene glycol)

(PEG), alkane (R) and gold, 750 μl of 10 % FBS solution, flow rate 100 μl/ min, 37° C, followed

by PBS rinsing. The time points of serum exposure and PBS rinse are indicated with arrows

[data from Chapter 3].

Activeelectrode

Counterelectrode

Contactelectrodes

Upperside

Lowerside

Δf massΔD Viscoelasticity

(a) (b)

-60-50-40-30-20-10

0

0 500 1000 1500 2000 2500Freq

uenc

y sh

ift (H

z)

X axis (Seconds) Gold

Serum adsorptionPBS rinse

PEG-COOHSAM

RSAM

OEG3-SAM

Page 36: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 2 – Contact angle based surface energy measurements

26 | P a g e

III. Contact angle based surface energy measurements

Contact angle measurements have a very high surface sensitivity (<1nm) [137] and

allow the determination of the surface energy of materials [138]. In 1988 Van Oss and R.J.

Good provided the theoretical background to correlate the contact angle of liquids on surfaces

with the surface energy components. The components of surface tension can be calculated

by measuring contact angles of three different liquids and solving the three simultaneous

equations given in Equation (10) [138]. The surface tension components of three liquids used

in this thesis to measure the surface energy components of various surfaces are given in

Table 2. Surface hydrophobicity is determined by the γsd value; the lower the value, the more

hydrophobic a surface is. The γs− values give a quantitative idea about the electron donor

groups; the greater the value of γs− , the higher the number of electron donor groups on a

surface; and the γs+ values represent the number of electron acceptor groups. Surface energy

measurements were used to ascertain the homogeneity of mixed monolayers of PEG and

OEG created by backfilling. There is a possibility that backfilling might result in islands of PEG

and OEG similar to phase separated type arrangements [139, 140]. Since the surface

wettability of PEG and OEG would be very similar, we used octane thiol for backfilling

experiments instead of OEG in order to follow the mechanism of addition. Alkane molecule

being more hydrophobic than PEG molecule, it would be possible to check for the presence of

islands if at all formed during backfilling by octane thiol using surface energy studies. The

results of these studies are presented and discussed in Chapter 3.

Table 2: Lifshitz van der Waals component (γSd), electron acceptor component (γS

+),

electron donor component (γS−) of water, formamide and α-bromonapthalene [138].

Liquid γs d mJ/m2 γs+ mJ/m2 γs

− mJ/m2

Water 21.8 25.5 25.5

Formamide 39 2.28 39.6

α-Bromonapthalene 44 0 0

Page 37: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 2 – Contact angle based surface energy measurements

27 | P a g e

θ – Measured contact angle. γL, γL

d, γL+ and γL

− represent the total surface tension, the

Lifshitz van der Waals component, the electron acceptor component, and the electron donor

component of liquid, respectively. γSd, γS

+ and γS− represent the Lifshitz van der Waals

component, the electron acceptor component, and the electron donor component of a

surface, respectively.

IV. Atomic force microscopy (AFM)

AFM is a scanning probe microscopy technique, which has the advantage that the

samples analyzed do not need to be conducting, and the analysis does not have to occur in

vacuum. Biological specimens can thus be analyzed under ambient conditions or even in

liquid [141] . The first AFM was built by Quate and Gerber in 1986. The basic working setup of

an AFM is given in Figure 8a. It consists of a sharp tip mounted on an optical lever facing the

sample while a laser beam is reflected from the opposite side of the tip. The reflected laser

beam is incident on a position sensitive photodiode (PSPD), thereby enabling the conversion

of tip-sample interactions into an electrical signal, which is used to construct topographic

images of the area being scanned. Apart from imaging, AFM can record force-distance curves

(Figure 8b). Using these curves, it is possible to study forces in the pico Newton range that

are involved in molecular interactions with surfaces. These studies can give valuable

information such as graft density [142], molecular conformation and frictional properties [143]

of interfaces grafted with polymers [141], helping to understand complex interfacial

phenomena relevant to fouling processes. The traditional way of acquiring force-distance

curves involves an approach-retraction procedure from a large number of positions on a

sample followed by analysis all the recorded curves to provide statistically meaningful results.

In this thesis, the topography and elastic modulus properties of PEG functionalized surfaces

were mapped using Quantitative Nano-Mechanical Mapping (QNM)-AFM under Peakforce

Tapping mode conditions. The force-distance curves were fitted by the Derjaguin-Müller-

Toporov (DMT) model [144, 145]. QNM-AFM enables one to map mechanical properties of

----- Eqn (10)

Page 38: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 2 – Atomic force microscopy

28 | P a g e

samples quantitatively, while simultaneously capturing topographic images at resolutions as

good as traditional tapping mode AFM [146, 147]. The modulus values were used to confirm

the absence of phase separated islands in mixed SAMs, and the results of these studies are

discussed in Chapter 3.

Figure 8. (a) A typical experimental setup used in AFM. (b) A typical force-distance

curve consisting of approach and retraction curves.

V. Ellipsometry

Ellipsometry is a surface sensitive technique that determines the thickness of surface

layers ranging from angstroms to tens of micrometers. It works by measuring the complex

refractive index given by Equation 10a or alternatively the complex dielectric function of any

material given by Equation (10b) [148, 149]. Drude in 1888 was the first to realize the

sensitivity of ellipsometry towards measuring the thickness of monolayer films. This was

followed by development of a matrix formalism by Abeles in 1947, who applied ellipsometry to

multilayered thin films. A typical ellipsometer setup is shown in Figure 9. The probe is a

linearly polarized laser beam with field components Ep and Es in the directions parallel and

perpendicular to the plane of incidence of the light. It works by analyzing the extent of

attenuation and phase shift in the s- and p- components of incident linearly polarized light that

Page 39: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 2 - Ellipsometry

29 | P a g e

becomes elliptically polarized upon reflection. Ellipsometry was used in this thesis as a

complementary tool to measure overlayer thicknesses of PEG SAMs on gold. The

calculations were performed similar to Unsworth et.al.[150]. The PEG modified surfaces were

analyzed using a three-layer air–PEO–gold model.

ň = n + i k ------ Eqn (10a) ; ε = ε1 + i ε2 ------ Eqn (10b)

Where, ň – complex refractive index, n – refractive index, k – extinction coefficient, ε1 –

real dielectric constant, ε2 – complex dielectric constant.

Figure 9. A typical experimental setup used in ellipsometry showing a beam of plane

polarized light becoming elliptically polarized after refection accompanied by attenuation and

phase shift shown on the ellipse [56][149].

Page 40: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

30 | P a g e

CHAPTER 3 Mixed poly(ethylene glycol) (PEG) and oligo(ethylene glycol) (OEG) layers on

gold as non­fouling surfaces

I. INTRODUCTION

The relevance of non-fouling coatings in a variety industrially and clinically applications

was discussed in Chapter 1 [3, 4]. Biofilm formation is proposed to be facilitated by a

conditioning layer, which is followed by reversible physiochemical attachment of microbes

[151]. Among the various strategies used in making non-fouling coatings, preventing initial

microbial attachment is a popular approach that generally involves the use of a hydrophilic

polymeric coating. Polyethylene glycol (PEG) is widely used to functionalize surfaces in order

to render them non-fouling. The osmotic and elastic properties of PEG along with its neutral

charge and non-toxic nature make it an effective and safe choice to minimize fouling in a

number of biologically relevant applications [45]. The ability for a given PEG functionalized

surface to resist fouling is thought to be mainly dependent on the graft density and molecular

weight (MW) of PEG used, both of which are key properties that decide the conformation of

PEG chains on surfaces [109, 110, 152]. For example, in the case of high or low graft density,

PEG chains are arranged in either a ‘brush’ or ‘mushroom’ conformation respectively [150].

Self-assembled monolayers (SAMs) of PEG chains are known to resist non specific

adsorption best when present in the brush conformation at high graft densities [109]. Studies

have shown that covalently bound PEG coatings also provide superior antifouling properties,

because they cannot be easily displaced by conditioning layer molecules [102]. PEG

functionalization techniques can be broadly classified into ‘grafting to’ and ‘grafting from’

approaches. The advantage of ‘grafting from’ over ‘grafting to’ technique was discussed in

chapter 1. Strategies such as ‘cloud point’ grafting [41], grafting in homopolymer solutions

[113], grafting from polymeric melts [114] and underbrush formation by backfilling with shorter

molecules [115] could be used to increase the graft density of polymer layers grafted using

’grafting to’ technique. The backfilling approach, unlike the other three strategies that depend

on minimization of excluded volume interactions, is a simple method wherein the interchain

Page 41: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 3 - Introduction

31 | P a g e

spaces present in layers of higher MW PEG chains are filled with shorter PEG chains that can

diffuse to the surface. Reducing non-specific adsorption by backfilling strategy is well known

strategy. For instance Uchida et al. showed that backfilling self assembled layer of 5 kDa

PEG thiol with shorter PEG thiol (MW 2 kDa) improved the ability of 5 kDa PEG SAM to resist

non specific protein adsorption [115, 153]. Lee et al. studied the effects of backfilling a self

assembled layer of thiol functionalized single strand DNA on gold with OEG capped-11-

mercapto-1-udecanol molecules and found that backfilling improved the specific binding

based hybridization of complementary DNA strands [154]. Also Lee et al. observed that

prolonged exposure of SAM of thiol functionalized single strand DNA to OEG alkyl thiol

molecules resulted in the replacement of DNA strands through desorption. The aspect of

desorption was not dealt with in reports by Uchida et al., whose backfilling studies are highly

relevant to the studies reported in this chapter. Also there is a possibility of islands of short

PEG thiol and long PEG thiol SAMs instead of homogeneous incorporation of short PEG thiol

molecules into the vacancies found in the SAM of long chain PEG thiol. The arrangement of

protein repelling molecules in a mixed layer formed by backfilling greatly influences the

antifouling ability of mixed SAMs. For instance if islands are formed, the protein repelling

ability of long chain islands would not have improved while the islands of short chain

molecules would have higher resistance towards protein adsorption. Mixed monolayers can

consist of either islands or homogeneous mixed layer. Examples of both types of mixed

monolayer formed by backfilling or co-adsorption can found in literature. For instance Herne

et al. obtained backfilling by exposing SAMs of thiolated DNA strands to mercaptohexanol,

and found that physically adsorbed DNA strands get replaced upon backfilling [155]. Brunner

et al. observed that phase separated islands were formed by replacement when SAM of

poly(p-phenylene) was exposed to octadecane thiol [156]. Tosatti et al. studied the SAMs

formed by co-adsorption of dodecyl phosphate and hydroxyl dodecyl phosphate on titanium

oxide [157]. They showed that homogenous mixed monolayers with no phase separation,

thus enabling the systematic variation of wettability properties of a surface by changing the

mole fractions of two compounds in solution. Bozzini et al. performed co-adsorption

experiments using PEG functionalized alkane phosphate and hydroxy alkane phosphate, and

demonstrated that the graft density of PEG chains could be varied systematically by varying

Page 42: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 3 - Introduction

32 | P a g e

the relative mole fractions in solution, also the fouling properties of surfaces changed

accordingly [158]. Amstad et al. stabilized iron oxide nanoparticles using a mixed monolayer

formed by co-adsorption hydroxy dopamine-PEG and biotin tagged hydroxy dopamine-PEG,

and demonstrated that the surface density of biotin could be varied by changing the relative

mole fraction [159]. Apart from the above mentioned studies, one can find reports on alkane

thiol co-adsorption and backfilling experiments, which have been shown to produce phase

separated islands [139, 140, 160].

The mechanism of formation of mixed layers by backfilling is not yet fully understood for

SAMs of PEG thiol exposed to shorter thiol molecules, similar to the study be Uchida et al.

[115, 153]. The possibility of the formation of domains exists (Figure 1a), where the

mechanism of addition is similar to that of the replacement phenomena observed for short

alkane thiol SAMs when they are exposed to longer alkane thiol molecules [139]. Since

alkane thiol self-assembly is driven by intermolecular van der Waals interactions and are

kinetically more reactive than high MW PEG thiols, the aim this study was to thoroughly

explore whether phase separated domains are formed (Figure 1a), and also the extent of

PEG thiol desorption. The structures of the thiolated PEG, OEG and alkane molecules used

are presented in Figure 1c. Although SAMs of OEG have been shown to be very efficient at

resisting protein adsorption [46] for short durations, it has been identified that applications

involving immobilization of bioactive molecules such as enzymes would benefit from longer

PEG chains due to higher mobility and substrate accessibility during biorecognition [161].

Thus it is expected that the combination of PEG-COOHSAM backfilled with OEG3 will be an

ideal support for immobilizing bioactive molecules. The use of antifouling enzyme as one of

the strategies to prevent biofilm formation was discussed in chapter 1. Thus the backfilled

PEG-COOHSAM would be ideal support for immobilization of antifouling enzymes such as

proteases and the results of this study are presented in chapter 4.

Page 43: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 3 - Introduction

33 | P a g e

Figure 1. The schematic diagrams of backfilled PEG SAMs in island type (a) and

underbrush type arrangements (b). The chemical formula and assigned abbreviations for

various molecules used in self assembly and backfilling experiments (c).

The following notations for SAM of PEG and for the backfilled surfaces are used:

PEG-COOHSAM, OEG3-SAM, and RSAM correspond to SAMs formed with PEG-COOH,

OEG3, and R molecules

PEG-COOHSAM+OEG3: PEG-COOHSAM backfilled with OEG3

PEG-COOHSAM+R: PEG-COOHSAM backfilled with R

II. MATERIALS AND METHODS

A. Substrate preparation and functionalization

Chemicals α-carboxyl-ω-thiol poly(ethylene glycol) (PEG-COOH, 99% purity, Mw-

2000Da) was purchased from Laysan Bio Inc. (Alabama, USA), and hydroxy-terminated

Underbrush formationIsland formation

(b)

(c)

(a)

-CH2 -NCO(OCH2CH2)45 – COOH HS

HS

(R)

(OEG3)

(PEG-COOH)

(OCH2CH2)3 –OH HS

carboxyl capped 2 kDa thiolated poly (ethylene glycol)

oligo (ethylene glycol)3 terminated alkane thiol

alkane thiol

Page 44: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 3 – Materials and methods

34 | P a g e

tri(ethylene glycol) undecanethiol (OEG3, 99% purity) was purchased from Assemblon

(Washington, USA). Fetal bovine serum (FBS), octanethiol (R) (99% purity), SYBR Green II,

phosphate buffered saline (PBS) buffer tablets, 25% ammonium hydroxide (NH4OH), 30%

hydrogen peroxide (H2O2) and absolute ethanol were purchased from Sigma Aldrich (Aarhus,

Denmark). Ultrapure MilliQ (MQ) water with resistivity of 18.2 MΩ was used for making buffers

and aqueous ethanolic solutions.

B. Substrate preparation and functionalization

Gold substrates were prepared by sputtering 50 nm gold layer onto a 3 nm titanium

attachment layer on silicon wafers. Sputtering was done using a RF sputtering system with Ti

and Au targets of 10 cm diameter (2.54 W cm−2) inside a standard chamber maintained at an

Ar pressure of 2 × 10−3 mbar. The substrate-target separation distance was 7 cm, while the

deposition rates for Ti and Au were 0.4 nm s−1 and 1 nm s−1, respectively. The gold surfaces

were cleaned by UV/ozone treatment for 30 minutes, followed by treatment with basic piranha

solution (H2O: NH4OH: H2O2 in ratio 4:1:1) at 70˚-80˚C for 5 minutes and then rinsed with MQ

water. (CAUTION: Piranha solution reacts violently upon contact with organic solutions).

Cleaned gold slides were immersed for 3 h at room temperature in 0.2 mM PEG-COOH

solution (75% ethanol), followed by rinsing with MQ water to remove any physically adsorbed

molecules. Backfilling involved immersion of PEG-COOHSAM for 3 h at room temperature in

1mM absolute ethanolic solution of OEG3 or R, followed by rinsing with ethanol and drying

with a jet of nitrogen. The SAMs of OEG3 and R for control experiments were made by

immersing cleaned gold slides in 1mM ethanolic solution of OEG3 or R for a period of 3 h

followed by rinse with ethanol and drying with nitrogen gas.

C. X­ray photoelectron spectroscopy

XPS spectra were recorded using a Kratos Axis UltraDLD instrument (Kratos Ltd, Telford,

UK) equipped with a monochromated aluminum source (Alkα 1486 eV) operating at a power of

150 W (15 kV and 10 mA) with pass energies of 80 eV and 160 eV for high resolution and

survey spectra, respectively. A hybrid lens mode was employed during analysis (electrostatic

and magnetic). The XPS spectra were measured at three areas on each sample and the take-

Page 45: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 3 – Materials and methods

35 | P a g e

off angle for all measurements was 0°. The measured binding energy positions were charge

corrected with reference to 285.0 eV, corresponding to the C-C/C-H species. Quantification

and curve fitting was conducted using CasaXPS software. A linear background with a

Gaussian to Lorentzian ratio of 30 was used to fit all spectra. The S2p high resolution spectra

were peak fit into two doublets corresponding to bound and unbound thiol each with full width

half maximum of 1.2 eV. The doublet corresponding to photoemission from 2p orbital has two

peaks in the ratio 1:2 which can be assigned to 2p1/2 and 2p3/2 respectively. The high

resolution S2p spectra with peak fitting of various surfaces analyzed using XPS are shown in

Figure 2. The lower binding energy peak of the doublet with its 2p3/2 at 161.9 eV was

assigned to bound thiol while the higher binding energy doublet with its 2p3/2 peak at 163.5 eV

corresponded to unbound thiol as described by Castner et al. [128]. Overlayer thickness and

graft density were calculated as described in Chapter 2.

D. Contact angle measurements

Static contact angles were measured on all surfaces using water, formamide and α-

bromonapthalene. All images of liquid drops on surfaces were recorded using a KRUSS

DSA100 (KRUSS GmbH, Hamburg, Germany), followed by drop shape analysis using

ImageJ software. Reported contact angles are the average of at least 6 measurements at

different surface positions. The values of γLd, γL

+ , γL− for the three liquids used are given in

Table 1 in Chapter 2. The interfacial Lifshitz van der Waals (γSd) and polar components (γS

+

, γS−) of surface tension of a surface can be determined by measuring contact angles with

three different liquids followed by solving the corresponding equations whose general form is

given by equation 10 in Chapter 2 [138].

E. Atomic force microscopy

All of the AFM images were recorded using a commercial Nanoscope VIII MultiMode

SPM system (Bruker AXS, Santa Barbara, CA) in MQ water. Topography images were

recorded using Quantitative Nano-Mechanical Mapping (QNM) under Peakforce Tapping

mode [147]. Ultrasharp silicon nitride cantilevers (triangular, Mpp-12120-10, Bruker AXS)

were used with a typical resonance frequency of 150 KHz in air, a spring constant of 5 N/m

Page 46: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 3 – Materials and methods

36 | P a g e

and a normal tip radius of 8 nm. All AFM images were recorded with 512 × 512 pixels

resolution per image, and they were flattened and analyzed with Scanning Probe Image

Processor software (SPIPTM, Image Metrology ApS, version 5.1.3, Lyngby, Denmark).

F. Quartz crystal microbalance with dissipation

QCM-D studies were performed using a Q-Sense E4 system (Gothenburg, Sweden). The

PEG-COOH functionalization followed by backfilling with OEG3 or R was performed on QSX

301 crystals (Q-Sense) coated with 100 nm gold which had approximate resonance frequency

of 5 MHz. Serum adsorption experiments were performed in triplicate. Functionalized gold

coated crystal surfaces were equilibrated at 37 °C in PBS (100 mM pH 7.4) prior to exposure

to serum solution. All serum adsorptions studies were carried out using 10% fetal bovine

serum (FBS) at 37 °C. Once a stable baseline was attained, the surfaces were exposed to

750 μL of protein solution delivered at a flow rate of 100 μL/minute followed by rinsing with

PBS buffer. The shift in resonance frequency corresponding to the 7th overtone was used to

monitor the adsorption. After the QCM crystals were exposed to serum they were rinsed with

MQ water to remove any salts before drying under a jet of nitrogen for XPS analysis.

G. Bacterial attachment assay

A Staphylococcus aureus (ATCC 12598) starter culture was inoculated from agar

plates and grown in 3 ml of 1% tryptic soy broth (TSB) medium in 50 ml conical bottom tube

by incubating overnight in a shaker at 37°C. An inoculum prepared by adding 1 ml of starter

culture to 100 ml 1% TSB medium was incubated in a shaker at 37°C until cultures were in

the late-exponential growth phase (OD600 = 1.0). Cells were then harvested by centrifugation

(5 minutes at 3000 x g), washed twice and resuspended in PBS (pH 7.4) and diluted to obtain

an OD600 of 0.5. Surfaces were incubated with the bacterial suspensions in 24 well plates for

5 h at 37°C and gently washed three times with sterile PBS to remove non-adherent cells.

The adherent cells on the surfaces were stained with ~10 µl 20x SYBR Green II RNA (2 µl ml-

1 of 10.000x SYBR Green II stock), covered with glass cover slips and sealed with nail polish

to avoid evaporation. Slides were stored in the dark at 4 °C until analyzed. Adherent cells

were counted by fluorescence microscopy using a Zeiss Axiovert 200M epifluorescence

Page 47: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 3 – Materials and methods

37 | P a g e

microscope (Carl Zeiss GmbH, Jena, Germany) equipped with Zeiss filter set 10 and 63x oil

immersion objective. Cells were counted manually in 109 µm2 grids on triplicates of sample

on a total of fifteen random positions on each of the surface type. A student t-test was used

to assess the significance of difference in bacterial attachment count on different surfaces.

III. RESULTS AND DISCUSSION

A. Extent of addition and surface coverage in backfilled layers

XPS was used to monitor the chemical changes occurring on Au substrates

functionalized with PEG-COOH upon exposure to OEG3 or R molecules. The respective

relative elemental and chemical species calculated from survey and high resolution XPS

scans are summarized in Table 1. The substrate photoelectrons are attenuated by the

overlayer and hence the substrate signal has an inverse relationship with overlayer coverage.

The Au 4f signal was used to obtain quantitative data on the addition of molecules to the

PEG-COOHSAM. The Au 4f signal of PEG-COOHSAM+OEG3 is 3.6% lower than that of PEG-

COOHSAM, indicating that OEG3 molecules have added to the PEG-COOHSAM. From the

chemical structure of OEG3 and R molecules it can be observed that they have lower O/C and

higher alkane (285.0 eV)/PEG (286.7 eV) [46] ratios when compared to PEG-COOH and

hence any addition during backfilling results in a corresponding change in the detected O/C

and alkane/PEG ratios.

Page 48: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 3 – Results and discussion

38 | P a g e

Figure 2. XPS high resolution spectra for the SH-PEG-COOHSAM, SH-PEG-

COOHSAM+SH-R and SH-PEG-COOHSAM+SH-R-OEG3 surfaces. (a) shows C 1s spectra, (b)

shows S 2p spectra.

285 eV

286.7 eV

288 eV

289.4 eV

Binding energy (eV)

Inte

nsity

CPS

PEG-COOHSAM+R

PEG-COOHSAM+OEG3

PEG-COOHSAM

C 1s(a)

161.9 eV163.1 eV163.4 eV164.6 eV

Binding energy (eV)

Inte

nsity

CPS

(b) S 2p

PEG-COOHSAM+R

PEG-COOHSAM+OEG3

PEG-COOHSAM

Page 49: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 3 – Results and discussion

39 | P a g e

Table 1. Summary of XPS results. Au % - relative atomic percentage of Au. O/C - ratio

of relative atomic percentage oxygen and carbon. Alkane (285.0 eV)/PEG (286.7 eV) - ratio of

alkane and PEG components in C 1s high resolution spectra. % S - relative atomic

percentage of S. % bound S - percentage of bound sulfur from components in S 2p high

resolution spectra.

Sample PEG-COOHSAM

OEG3-SAM RSAM PEG- COOHSAM +R

PEG-COOHSAM + OEG3

Au % 39.0±0.4 32.6±0.4 55.3±2.4 41.6±0.5 36.2±0.5

O/C 0.49±0.03 0.25±0.01 0.0±0.0 0.40±0.01 0.42±0.01

Alkane/PEG 0.1±0.0 0.8±0.0 21.0±0.0 0.4±0.0 0.3±0.0

S % 2.1±0.2 1.5±0.2 3.2±0.4 2.1±0.2 1.9±0.6

% bound S 78.3±0.7 80.7±1.0 88.1±1.0 81.8±1.0 79.9±0.7

The alkane/PEG ratio was calculated from the peak areas of components

corresponding to binding energies of 285.0 eV (alkane) and 286.7 eV (PEG) obtained from

the high resolution C 1s spectra shown in Figure 2a. Addition of OEG3 is further supported by

a decrease in O/C ratio from 0.49 to 0.42 and an increase in alkane/PEG ratio from 0.1 to 0.3

as shown in Table 1. The PEG-COOHSAM+R system exhibits a decrease in O/C ratio from

0.49 to 0.40 and an increase in alkane/PEG ratio from 0.1 to 0.4 similar to PEG-

COOHSAM+OEG3 except for the Au signal which increased by 2.6 %. The increase in

substrate signal indicates a decrease in overlayer thickness that could be caused by

desorption of some PEG-COOH molecules during backfilling by R molecules. Based on the

O/C ratios obtained from XPS for PEG-COOHSAM, OEG3-SAM, and RSAM, a change in O/C ratio

as a function of relative coverage of PEG-COOH and OEG3 or R was plotted assuming a

linear relationship (Figure 3). By correlating the experimentally obtained values of the O/C

ratios for PEG-COOHSAM+OEG3 and PEG-COOHSAM+R, we estimate the coverage of OEG3

or R on the backfilled PEG-COOHSAM surface to be 29% and 18%, respectively. It must be

noted that this estimation assumes that the intermolecular spaces in PEG-COOHSAM ideally

are free from any contamination, which in real laboratory conditions is not the case and thus

we overestimate the number of added molecules. Here, however, we mainly focus on the

relative differences rather than the absolute values.

Page 50: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 3 – Results and discussion

40 | P a g e

Figure 3. Relative coverage of PEG-COOH and R or OEG3 on backfilled surfaces

determined by correlating the XPS O/C ratios (X axis) to a plot obtained by extrapolating

values of O/C ratio for 0% and 100% relative coverage (Y axis). The data points and +

represent the experimentally obtained O/C ratios for PEG-COOHSAM+OEG3 and PEG-

COOHSAM+R, respectively.

Despite the greater number of OEG3 molecules being added to the PEG-COOHSAM,

the decrease in carboxyl peak area (Table 1) in the C 1s spectra of PEG-COOHSAM+OEG3

and PEG-COOHSAM+R are comparable. This suggests that desorption of PEG-COOH is lower

when OEG3 is added in comparison to when R is added. It is known that the relative atomic

percentage of S from SAMs of PEG thiol depends not only on the stoichiometric ratio but also

on the extent of dehydration and collapse of brushes during grafting of chains as described by

Unsworth et al. [150]. The amount of S in PEG-COOHSAM was 2.1 %, which is higher than the

corresponding stoichiometric ratio of <1%, very similar to what was reported by Unsworth et

al. [150] with hydroxyl terminated 2 kDa PEG thiol molecules grafted under ‘cloud point’

conditions. Though R and OEG3 molecules have S/C ratios of 0.11 and 0.045, respectively,

which is higher than the ratio for PEG-COOHSAM (0.008), the backfilling of PEG-COOHSAM

with either R or OEG3 did not significantly change the relative atomic percentage of S (Table

1). Hence the S content was not helpful in providing additional information about the

backfilling process. The percentages of bound thiol on functionalized surfaces were

determined as described above in the methods section chapter 2. The amount of bound

0

20

40

60

80

100

0.00 0.10 0.20 0.30 0.40 0.50

Perc

enta

ge

O/C ratio

% coverage and O/C ratio

100% PEG-COOHSAM

(18%, 0.4)

(29%, 0.42)

100% RSAM 100% OEG3-SAM

PEG-COOHSAM+R

PEG-COOHSAM+OEG3 ++

Page 51: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 3 – Results and discussion

41 | P a g e

sulfur was found to increase by a small extent (increase of 3.5%) upon backfilling PEG-

COOHSAM with R, and for OEG3 it increased by 1.6%. This small increase in bound sulfur

could be due to Au-thiol bond formation of backfilled molecules, also indicating that a

significant proportion of added molecules have chemisorbed to the gold surface and not

physically adsorbed. The curve-fitted high resolution S 2p spectra of various samples are

shown in Figure 2b.

B. Arrangement of backfilled molecules

As discussed earlier, the backfilling process could result in a mixed monolayer with two

possible arrangements namely homogeneously mixed layers or islands. Contact angle based

surface energy measurements were used to study the arrangement of backfilled molecules.

Contact angle measurements have a very high surface sensitivity (<1nm) [137] and allow the

determination of the surface energy of material surfaces [138]. The components of surface

tension can be calculated using the contact angle values of three different liquids. The values

of contact angles for various surfaces are shown in Table 2, and their respective surface

tension components; Lifshitz van der Waals (γSd), electron acceptor (γS

+), electron donor (γS−)

components are shown in Table 3.

Table 2. Contact angles for water, formamide and α-bromonapthalene measured on the

SH-PEG-COOHSAM, SH-PEG-COOHSAM+SH-R, SH-PEG-COOHSAM+ SH-R-OEG3, SH-R-

OEG3-SAM, SH-R surfaces.

Sample Water Formamide Bromonapthalene

OEG3-SAM 24.3˚ ± 1.6 13.6˚ ± 0.5 11.9˚ ± 0.4

RSAM 100.3˚ ± 1 85.3˚ ± 2.6 48.5˚ ± 2.6

PEG-COOHSAM 25˚ ± 1.7 9.6˚ ± 1.1 4.8˚ ± 0.7

PEG-COOHSAM+R 34.3˚ ± 0.7 16.8˚ ± 1 7.2˚ ± 0.9

PEG-COOHSAM+OEG3 26.9˚ ± 1.6 12.3˚ ± 1.1 7˚ ± 0.9

The hydrophobicity of a surface is determined by its γSd value, with more hydrophobic

surfaces having lower values. Since alkane thiol molecules are hydrophobic a SAM of alkane

thiol would have a lower γSd value than PEG or OEG based SAMs and this is apparent for the

Page 52: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 3 – Results and discussion

42 | P a g e

values determined for RSAM and OEG3-SAM (Table 3). If alkane thiol molecules form islands

instead of being homogeneously mixed during the backfilling of the PEG-COOHSAM, then the

γSd value of PEG-COOHSAM+R would significantly decrease. The γS

d value for PEG-

COOHSAM+R is 44.0 mJ/m2 and is virtually the same as that of PEG-COOHSAM indicating that

hydrophobic islands are not present. The γS− value of a surface gives a quantitative idea

about the presence of electron donor groups, with the greater the value of γS− the higher the

number of surface electron donor groups. PEG and OEG have oxygen atoms and thus have

high γS− values when compared to the alkane thiol (Table 2). Backfilling PEG-COOHSAM with R

results in a 7 mJ/m2 decrease in γS− indicating that a significant number of PEG-COOH

molecules have desorbed from the surface. The values for γSd and γS

− for PEG-COOHSAM after

addition of OEG3 do not change significantly since both molecules have very similar values

and could not easily be followed by contact angle measurements.

Table 3. Surface tension components: the Lifshitz van der Waals component (γSd),

electron acceptor component (γS+), and electron donor component (γS

−) determined from the

surfaces of OEG3-SAM, RSAM, PEG-COOHSAM, PEG-COOHSAM+R and PEG-COOHSAM+OEG3.

Sample γSd mJ/m2 γS

+ mJ/m2 γS− mJ/m2

OEG3-SAM 43.45±0.06 0.85±0.05 45.66±1.49

RSAM 30.67±1.27 0.01±0.01 0.69±0.26

PEG-COOHSAM 44.24±0.04 0.92±0.05 43.97±1.56

PEG-COOHSAM+R 44.05±0.08 0.96±0.03 36.58±0.80

PEG-COOHSAM+OEG3 44.07±0.1 0.90±0.05 42.90±1.59

C. Arrangement of molecules in mixed layer studied by AFM

QNM-AFM has a very high lateral resolution and has been used to characterize the

surface morphology of the different SAM layers with the aim to check the presence of phase

separated domains. With the use of the peak forces less than 1 nN, QNM AFM is able to map

mechanical properties of surfaces quantitatively while simultaneously capturing topographic

images with the same resolutions as traditional tapping mode AFM [146][147]. All QNM-AFM

measurements were performed in MQ. The topography and modulus images of Au surfaces

Page 53: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 3 – Results and discussion

43 | P a g e

and various SAMs are shown in Figure 4, and the corresponding surface roughnesses are

summarized in Table 4.

Table 4. Normalized surface roughness (to Au) values for OEG3-SAM, RSAM, PEG-

COOHSAM, PEG-COOHSAM+R and PEG-COOHSAM+ OEG3. SqSAM/SqAu stands for ratio of the

means of the two surface roughness; SqAu= 0.48±0.02nm

Sample Roughness (Sq) (nm) SqSAM/SqAu

PEG-COOHSAM 0.69±0.04 1.44

OEG3-SAM 0.63±0.04 1.31

RSAM 0.21±0.02 0.44

PEG-COOHSAM+OEG3 0.81±0.05 1.69

PEG-COOHSAM+R 0.66±0.06 1.38

Figure 4. Topography and Young’s modulus images recorded by QNM-AFM

measurements in MQ water. (a, a’) Au substrate, (b, b’) PEG-COOHSAM, (c, c’) OEG3-SAM, (d,

Page 54: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 3 – Results and discussion

44 | P a g e

d’) RSAM, (e, e’) PEG-COOHSAM+OEG3, and (f, f’) PEG-COOHSAM+R. The scan area of each

image is 1µm×1µm.

The topography of the bare Au surface (Figure 4a) shows Au grains without any

additional features on top of the grains. The recorded nanomechanical map of a Au surface

(Figure 4a’) demonstrates the homogeneous Young’s modulus distribution as the whole

image has nearly the same range of stiffness showing the same color contrast. Comparing

the topography images of modified surfaces with the bare Au, one can clearly see the

additional particle-like features on all the modified surfaces (Figure 4b-f). In addition, the

modulus images of the modified surfaces show that many of the grain boundaries are slightly

invisible due to the SAMs. Furthermore, the surface roughness has been changed

dramatically after modification. Hence these results prove the successful formation of the

SAMs. Interestingly, the modulus of the modified surfaces also represents the surface

modulus variations, because of the slightly different color contrast (Figure 4b’-4f’). The small

variations in material properties observed in elastic modulus measurements are particularly

useful in the analysis of heterogeneous surfaces. However most importantly, phase

separations were not observed in any of images of the backfilled surfaces, indicating that they

are homogeneous. Hence, it is concluded that the SH-R-OEG3 or SH-R molecules form

homogeneous mixed monolayers upon addition to SH-PEG-COOHSAM rather than separate

domains. The surface roughness of PEG-COOHSAM increases by 0.12 nm upon exposure to

OEG3 molecules. Lee et al. [154] showed that the proportion of DNA molecules in upright

orientation in a SAM of thiolated single strand DNA on gold increased upon exposure to OEG

terminated alkane thiol molecules. An increase in proportion of PEG molecules in upright

orientation could explain the increase in surface roughness of PEG-COOHSAM upon exposure

to OEG3. Lee et al. [162] showed that addition of alkylthiol molecules onto SAM of thiolated

single strand DNA on gold increased the fraction of DNA molecules in upright orientation.

Once should expect a similar effect during the addition of R onto PEG-COOHSAM, but the

surface roughness of PEG-COOHSAM+R in fact decreases by 0.03 nm (decrease not

significant due to high deviation). It must be noted that the addition of R caused more

desorption of PEG-COOH molecules during backfilling when compared to that observed

during the addition of OEG3. Thus desorption of PEG-COOH molecules during the addition of

Page 55: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 3 – Results and discussion

45 | P a g e

R could be the reason behind the surface roughness of PEG-COOHSAM +R not being higher

than PEG-COOHSAM.

D. Resistance of backfilled PEG­COOH surface towards protein adsorption and bacterial attachment

Resistance towards non-specific adsorption can be tested using simple model systems

such as single protein solutions. Examples have included collagen [163] and bovine serum

albumin [132]. More rigorous tests are conducted with serum [132] or microbial cell

suspensions [42]. Once the surfaces have been assessed by model testing techniques, their

performance in real environments such as a food processing plant, as coatings on clinical

implants or marine waters could be studied. The non-fouling properties of the PEG-COOHSAM

and mixed surfaces were evaluated by serum adsorption and bacterial attachment studies.

Addition of smaller OEG chains in theory increases the graft density of protein resistance

molecules within the layer of diffuse PEG-COOHSAM molecules and hence is expected to

improve its resistance towards non-specific adsorption. QCM and XPS have been employed

to quantitatively compare the serum adsorption resistance properties of the SAMs. The

frequency shift after serum exposure in QCM is directly proportional to the amount of protein

adsorbed [132], and the relative atomic percentage of nitrogen from XPS also enables a

quantitative estimation of the amount of adsorbed protein [163]. Representative QCM kinetic

plots are shown in Figure 5. In Figure 6 we show the data points corresponding to various

samples studied, where the X-axis shows the percentage of nitrogen (% N) obtained from

XPS and the Y-axis shows the frequency shift from the QCM data. Using the N atomic

percentage and the frequency shift in QCM, the amount of protein adsorbed was determined

as shown in Table 5. Upon serum exposure the resonance frequency decreases by 7 Hz for

the PEG-COOHSAM+OEG3 surface, compared to both PEG-COOHSAM and PEG-COOHSAM+R

surfaces, which exhibit a frequency shift of 15 Hz. The lower frequency shift for PEG-

COOHSAM+OEG3 proves that OEG3 addition improves the non-fouling abilities of PEG-

COOHSAM surfaces. It is also speculated that protein adsorption would be lower if the PEG

thiol molecules used did not have a carboxyl end-group, since carboxyl groups under

physiological pH would interact electrostatically with serum proteins, thus facilitating higher

Page 56: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 3 – Results and discussion

46 | P a g e

adsorption. Figure 6 shows nitrogen contents on the X-axis, where the PEG-COOHSAM+OEG3

surface has 2% N compared to PEG-COOHSAM and PEG-COOHSAM+R surfaces both showing

approximately 5% N, which follows the same trend as seen with shift in resonance

frequencies obtained using QCM. The OEG3-SAM had the highest ability to resist serum

adsorption which agrees with previous studies with OEG based SAMs [46]. The hydration

factor was calculated by taking the ratio of wet mass and dry mass from QCM and XPS

respectively [164]. The adsorption of proteins onto hydrophobic surfaces would increase the

chances of denaturation of the molecule thereby making it more rigid and less hydrated [46].

The hydration factor values calculated are in the following order PEG-COOHSAM+OEG3 >

PEG-COOHSAM ≅ PEG-COOHSAM +R. The higher hydration factor of OEG backfilled SAM

could be due to the higher hydration and lower rigidity of adsorbed serum protein molecules

during the exposure to serum or a different type(s) of proteins adsorbing to each surface from

the complex serum. This further supports the speculation the PEG SAM backfilled OEG would

be ideal surface for immobilization of bioactive molecules. Gold was used as reference

sample which had a hydration factor of 1.8, least among all samples, indicating that the major

proportion of proteins adsorbed onto gold are rigid due to higher surface hydrophobicity.

Table 5. Hydration factor for PEG-COOHSAM, PEG-COOHSAM+R and PEG-COOHSAM+

OEG3 calculated from the ratio of QCM based wet thickness and XPS based dry thickness as

described by Ayyoob et al.[164].

QCM ng/cm2 XPS ng/cm2 Hydration factor

PEGSAM 36.7 12.7 2.9

PEGSAM +OEG 19.5 5.0 3.9

PEGSAM +R 36.4 13.5 2.7

Gold 126 67.7 1.8

Page 57: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 3 – Results and discussion

47 | P a g e

Figure 5. Representative QCM plots showing the change in frequency upon exposure to

10 % FBS, 37° C 750 μl, 100μl/ min on: Au, PEG-COOHSAM, PEG-COOHSAM+R, PEG-

COOHSAM+OEG3, RSAM and OEG3-SAM. The time points of serum exposure and PBS rinse are

indicated with arrows.

Figure 6. Quantification of N% (by XPS) and resonance frequency change (by QCM)

after exposure of PEG-COOHSAM, PEG-COOHSAM+R and PEG-COOHSAM+OEG3, OEG3-SAM

and RSAM surfaces to 10% FBS at 37°C.

-60-50-40-30-20-10

0

0 500 1000 1500 2000 2500

Freq

uenc

y sh

ift (H

z)

X axis (Seconds)

QCM studies of FBS adsorption

Gold

PEG-COOHSAM+OEG3

Serum adsorptionPBS rinse

PEG-COOHSAM

PEG-COOHSAM+R

RSAM

OEG3-SAM

-35

-30

-25

-20

-15

-10

-5

00 2 4 6 8

Freq

uenc

y sh

ift (H

z)

Nitrogen atomic %

Adsorption studies with FBS

OEG3-SAM

PEG-COOHSAM+OEG3

PEG-COOHSAM

PEG-COOHSAM+R

RSAM

Page 58: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 3 – Results and discussion

48 | P a g e

The surfaces were tested for bacterial attachment using a Staphylococcus aureus strain.

The results of bacterial attachment studies are shown in Figure 7. Clearly, the PEG-

COOHSAM+OEG3 (4.2 × 107 cells/cm2) surface is better at resisting bacterial attachment when

compared to PEG-COOHSAM (7.2 × 107 cells/cm2) and PEG-COOHSAM+R (9.2 × 107

cells/cm2). Hence it can be concluded that adding OEG3 molecules improve the non-fouling

properties of the PEG-COOHSAM. Though the OEG3-SAM has the least bacterial attachment

and serum adsorption, we propose that PEG-COOHSAM+OEG3 system would be a better

system for applications involving immobilization of bioactive molecules such as enzymes and

further advantages of PEG-COOHSAM+OEG3 will be discussed in the next section. The

bacterial attachment studies performed here are short term studies and a comprehensive

study would involve the study of the time biofilm formation starting from the initial adhesion,

followed by microcolonies formation and then the maturation phases I, II [151]. Thus the

preliminary bacterial initial attachment studies give an idea about the effect of backfilling on

the resistance of surfaces towards initial attachment of bacterial cells.

Figure 7. Attachment studies using Staphylococcus aureus to PEG-COOHSAM, PEG-

COOHSAM+R and PEG-COOHSAM+OEG3, OEG3-SAM, RSAM.

0 40 80 120Log (number of adhered cells/cm2

Bacterial adhesion study

PEG-COOHSAM +OEG3

PEG-COOHSAM

PEG-COOHSAM+ R

OEG3-SAM

RSAM

*P < 0.01

300

Page 59: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 3 – Results and discussion

49 | P a g e

E. Discussion

We have studied mixed layers of OEG3 and PEG-COOHSAM using XPS, contact angles

and AFM. The contact angle results support the notion that phase separated surface

arrangement of the two molecules does not occur proving that homogeneous mixed layers

are formed. There have been some reports on grafting 2 kDa PEG thiol chains. For example

Unsworth et al. [150] reported a maximum graft density of 0.58 chains/nm2 using methoxy

capped 2 kDa PEG thiol chains under ‘cloud point’ conditions, and Tokumitsu et al. [165]

reported a maximum graft density of 3.6 chains/nm2 using 2 kDa PEG conjugated to

undecane thiols. From our XPS data we have calculated a graft density of PEG-COOHSAM to

be 1.0 chain/nm2. Thus, the PEG-COOH chains in PEG-COOHSAM have a density that is not

close-packed and there exists sufficient space for the smaller OEG molecules to fill the gaps,

although in a good solvent the PEG is likely to extend and form highly mobile chains [150].

The graft density of close packed SAM of PEG brush is approximately 4.5 chains//nm2

assuming a cross-sectional area of PEG molecule in helical conformation to be 22 Å2 as

reported by Harder et al. [46]. There are several reports on protein resistant PEG SAMs in the

literature. Dalsin et al. measured the protein resistance of DOPA functionalized 5 kDa PEG

SAMs on titanium dioxide surface [166]. They reported serum asdorption <1ng/cm2 for PEG

surface with an approximate graft density of 3.2 chains/nm2, and that graft density

corresponded to brush conformation. A similar study was reported by Pasche et al. who used

PLL-g-PEG SAMs to create surfaces with PEG brushes that showed serum adsorption as low

as <1ng/cm2 on nobium oxide surface [167]. The size of protein molecules is one of the most

important parameters determining the qualitative and quantitative aspects of adsorption on

polymer coated surfaces. If the SAM of PEG on a surface is exposed to protein molecules

that are smaller than the interchain distances, then the PEG SAMs fail to effectively resist

protein adsorption. Uchida et al. [115] performed protein adsorption studies using a range of

proteins with varying molecular weight on mixed monolayers of 5 and 2 kDa PEG thiol on gold

using backfilling method. They demonstrated these backfilled monolayer could resist the

adsorption of protein in the MW range 400Da – 340 kDa (surface protein <1ng/cm2). The

carboxyl terminated PEG chains chosen in experimental studies performed in this chapter

Page 60: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 3 – Results and discussion

50 | P a g e

would possess a negative charge at physiological pH. Carboxyl capped PEG molecules were

used since it is intended to use them for immobilization of antibacterial enzymes in future

studies. These molecules are expected to contribute to the surface negative charge. It is

suspected that the non-fouling properties would be better with hydroxyl or methoxy capped

PEG molecules, which are neutrally charged and thus minimize electrostatic interactions. The

mixed PEG-COOHSAM+OEG3 system is anticipated to be an ideal platform for immobilization

of bioactive molecules, since the OEG maximizes the non-fouling potential while the longer

PEG chains would confer higher mobility and thus improved bioactivity capabilities to the

immobilized molecule [161]. There have been reports on bacterial attachment studies on

OEG [168] and PEG [42] surfaces. The attachment studies with Staphylococcus aureus

involve different incubation times compared to that used by Nejadnik et al. [42], and Ostuni et

al. [168], whoused a counting technique involving removal of attached cells by sonication

followed by counting the colony forming units on culture plates that introduces possibility of 10

fold error in estimation as mentioned by authors. Hence the absolute values the bacterial

attachment studies found here could not be compared with these earlier reported studies.

Based on the studies of backfilled PEG-COOHSAM surfaces, it was found that OEG3-SAM are

better at resisting fouling when compared to all other surfaces studied in the project. Otsuka

et al. demonstrated that the biorecognition efficiency benefit from longer PEG chains is due to

higher mobility [161]. Thus the combination of PEG-COOHSAM backfilled with OEG3 will be an

ideal support for immobilizing bioactive molecules because the longer PEG molecule would

confer higher mobility and backfilled OEG molecules would improve ability to resist non

specific adsorption. In Chapter 4 preliminary results are presented of relating to immobilizing

antifouling enzyme molecules (subtilisin) on backfilled PEG-COOHSAM+OEG3 surfaces with

further understandings of the effect of backfilled OEG molecules on the activity and

performance of enzyme molecules.

IV. SUMMARY AND CONCLUSION

Backfilling of a PEG-COOHSAM with alkane and oligo ethylene glycol terminated thiol

molecules was investigated. From XPS and contact angle studies it was shown that R

molecules form mixed layers and the addition process is accompanied by some desorption of

Page 61: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 3 – Summary and conclusion

51 | P a g e

the PEG-COOH molecules. OEG3 molecules, upon addition, also formed mixed layers but

less desorption of the PEG occurs, when compared to addition of R molecules. The non-

fouling properties of various surfaces were compared, and it was shown that the PEG-

COOHSAM+OEG3 surface was found to be better than the PEG-COOHSAM surface with respect

to resisting serum adsorption and bacterial attachment. Thus, the strategy involving backfilling

using OEG terminating alkane thiols is indeed capable of improving the non-fouling properties

of layer of PEG chains of significant initial graft density.

Page 62: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 4 - Introduction

52 | P a g e

CHAPTER 4

Immobilization of active subtilisin to mixed poly(ethylene glycol) and

oligo(ethylene glycol) layers

A. Introduction

Antifouling enzymes are naturally occurring biodegradable biocatalysts that have the

potential to be an efficient and environmentally friendly strategy for preventing biofouling [4,

34, 169]. The usefulness of an enzyme in a specific application depends on the reaction it

catalyzes. The choice of appropriate enzyme for prevention of microbial biofilm formation

processes requires that the enzyme using its catalytic reaction capability interferes with

biofilm formation process. The steps involved in microbial biofilm formation processes were

discussed in Chapter 1. The extracellular polymeric substance (EPS) has been shown to be a

very important in microbial adhesion, colonization and biofilm formation [20, 51-53]. The EPS

mainly consists of polysaccharides [51], extracellular DNA [20] and glycoproteins. Disturbing

the integrity of EPS by damaging one of the important constituents of EPS renders the

microbe incapable of surface adhesion and/or biofilm formation [20, 51]. According to

Nomenclature Committee of the International Union of Biochemistry and Molecular Biology

(NC-IUBMB), enzymes can be classified based on the types of reactions they catalyze [171].

Various classes of enzymes are given in Table 1. Among the classes of enzymes mentioned

in Table 1, hydrolases and lyases are most relevant when it comes to non-toxic antifouling

applications. Since the enzymes do not kill organisms and get degraded easily technologies

based on enzymes are environmentally benign [170]. Many examples of enzyme based

prevention or treatment of biofilms can be found in literature. Lysozyme, trypsin, pepsin,

subtilisin, α-amylase, DNase I and alginate lyase have been demonstrated to interfere with

either the biofilm formation process or damage an existing biofilm [20, 34, 51, 57, 169, 172,

173]. The modes of action of different groups of enzymes are listed in Table 2. Apart from the

EPS damaging enzymes listed, there are a few other protein molecules which are capable of

interfering with biofilm formation. Intercellular communication in biofilm occurs through release

of quorum sensing molecules such as the N-acyl homoserine lactones (AHL) [174]. Enzymes

Page 63: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 4 - Introduction

53 | P a g e

such as AHL acylase interrupt the quorum sensing mechanism thereby stopping further

development of biofilm [175]. Lactoferrin is a mammalian protein relevant to immune system

which limits bacterial infections by sequestering iron from the immediate surrounding

environment, and use of this protein has been demonstrated to slow down the biofilm

formation process [56]. The work presented in this chapter focuses on investigating the

antifouling properties of backfilled PEG-COOH SAMs immobilized with subtilisin, which is a

widely used serine protease.

Table 1. Various classes of enzymes and their corresponding function. The classification is in accordance with NC-IUBMB.

Enzyme class Function

EC 1

Oxidoreductases

catalyze red-ox reactions

EC 2 Transferases catalyze the transfer of a chemical group

from donor to acceptor

EC 3 Hydrolases catalyze the hydrolysis of various bonds

EC 4 Lyases catalyze cleaving bonds by means other

than by hydrolysis or oxidation

EC 5 Isomerases catalyze isomerization

EC 6 Ligases catalyze ligation of molecules

Table 2. Mode of action for different groups of enzymes used in biofilm prevention or

treatment [20, 34, 51, 57, 169, 172, 173].

Enzyme Mode of action

Trypsin, pepsin,

subtilisin

Peptide bond cleavage

Protein damage

Lysozyme, α-amylase,

alginate lyase

Glycosidic cleavage

Polysaccharide damage

Dnase I Phosphodiester cleavage

Extracellular DNA damage

Page 64: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 4 - Introduction

54 | P a g e

Proteases constitute the largest volume of enzymes used in industrial applications.

Subtilisin enzyme accounts for 35 percent of the total enzyme market, with an approximate

annual production of 500 tones [176, 177]. Subtilisin is one of the most well studied serine

proteases used in a number of applications such as food processing, leather treatment,

detergents, waste management, pharmaceuticals, silver recovery and organic synthesis [178-

180]. Subtilisin has been shown to have antifouling properties against several species of

bacteria [169] and it works by cleaving bacterial cell membrane proteins consequently

inhibiting adhesion to surfaces. Microbes use special adhesion organelles such as pili [181]

and proteolytic or glycosidase enzymes capable of damaging these adhesion organelles and

thus prevent the microbes from adhering to surfaces. Due to the wide spread usage in

industrial applications and antifouling capabilities of serine proteases, subtilisin was chosen

as a model enzyme to perform immobilization studies on mixed SAM of PEG and OEG

created by backfilling as described in Chapter 3. Preliminary experiments with BSA and

lysozyme were also performed to compare the results of immobilization studies with those

obtained using subtilisin.

Enzymes can be used either as suspensions or in immobilized form [182]. Using

immobilized form of enzyme has several advantages over a suspended enzyme such as easy

recovery and reusability, but on the down side the immobilization step can lead to a loss of

enzymatic activity due to conformational changes and/or denaturation [182, 183]. Protein

molecules adsorb onto surfaces through van der Waals, electrostatic, covalent and dispersion

interactions, with dispersive or hydrophobic-hydrophobic interactions being

thermodynamically most significant when it comes to protein denaturation [184-186].

Strategies used to improve or retain the activity of immobilized subtilisin can be broadly

classified into two categories. The most popular one being enzyme structure modification

using protein engineering techniques such as addition of affinity tags, sulfhydryl and site

directed mutagenesis [183, 187, 188]. The second category includes strategies which involve

substrate (surface) modification using polymers that prevent protein denaturation [188-190].

The changes introduced by protein engineering techniques improve the overall performance

of an immobilized enzyme by making it thermally more stable towards denaturation or

providing specific conformational orientation to the enzyme molecule. The strategy involving

Page 65: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 4 - Introduction

55 | P a g e

hydrophilic polymer coatings on substrates minimizes the loss of enzyme activity upon

immobilization by presenting a steric barrier between the enzyme molecule and the substrate

surface thus preventing the unfavorable interactions such as hydrophobic-hydrophobic

interactions that denature the enzyme or protein molecules [188-190]. PEG is one of the most

widely used hydrophilic nonfouling polymers for reasons discussed in Chapter 1. There are

several strategies to maximize the graft density of PEG coatings via ‘grafting to’ techniques

such as ‘cloud point grafting’ [41] and backfilling [115]. In Chapter 3 we presented results of

detailed surface characterization of SAMs formed by backfilling PEG thiol layers with OEG

thiol molecules. These backfilled surfaces are an ideal support for immobilizing bioactive

molecules such as enzymes as they would benefit from longer PEG chains allowing higher

mobility and potentially enhanced biorecognition [161] while avoiding non-specific adsorption

of proteins [153]. In this chapter, the focus was to immobilize subtilisin on the surfaces of

carboxyl terminated PEG-COOH and OEG-COOH thiols assembled on gold with comparisons

being made to PEG-COOH backfilled with OEG thiols on gold. The hypothesis is that

immobilization of enzyme on PEG-COOH SAM backfilled with OEG thiols would result in

minimum non-specific adsorption and higher enzyme activity when compared to PEG-COOH

SAMs.

The covalent immobilization of biomolecules onto surfaces requires that they have

appropriate functional groups present in order for the coupling reactions to take place. Non-

covalent immobilization is also used but covalent coupling often provides greater stability

[191]. Some of the widely used surfaces such as titanium, steel, and polymeric materials do

not have the appropriate reactive groups required to react with biomolecules, and this

limitation can be overcome by functionalizing the surfaces, for example, by oxidation to

introduce hydroxyl or carboxyl groups; coating with SAMs with appropriate tail groups [84,

86]. There are several well known reactions used for coupling purposes such as carboxyl-

amine, aldehyde-amine, thiol-thiol, amine-thiol, hydroxyl-amine, amine-amine, thiol-carbonyl,

and thiol-maleimide. A host of other reactions are possible depending on the availability of

functional groups, reaction efficiencies and conditions required for reactions [84, 87]. The

amino acids relevant to immobilization of proteins are listed in Table 3. Aspartic acid and

glutamic acid residues have a carboxylic acid group that can be used to couple the protein by

Page 66: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 4 - Introduction

56 | P a g e

forming linkages such as ester (with hydroxyl), thioester (with thiol), amide (amine). Often

these reactions require activation of the carboxyl to form a active ester intermediate, which

acts as a better electrophile when compared to the carboxyl anion [87]. Lysine residues have

a primary amine that can react either directly with alkyl halides to give alkyl substituted

amines or activated carboxyl or acyl group resulting in an amide bond formation [87]. In this

project we choose the amine based amide formation to couple the proteins onto PEG SAM

whose tail groups were an active ester intermediate. The yield of a reaction between a

nucleophile (amine) and an electrophile (carboxylic ester intermediate) depends on reaction

conditions such as medium temperature, state of protonation of amine (depending on pI, pH)

[87].

Table 3. Properties of subtilisin, lysozyme and BSA relevant to the immobilization

experiments. The MW, the number of important residues, theoretical relative atomic

percentages of C, N, O, and S were found using ExPASy ProtParam tool [192]. The amino

acid sequences were obtained from NCBI protein database [193]. The pI values of proteins

were provided by supplier Sigma Aldrich, Denmark.

Subtilisin Lysozyme BSA

MW (Kda) 27.2 14.2 69.2

Carbon (%) 62.1 61.4 63.3

Nitrogen (%) 17.2 19.2 16.8

Oxygen (%) 20.5 18.4 19.1

Sulphur (%) 0.3 1.0 0.8

pI (isoelectric pH) 9.4 11.3 5 Functional group

Arg (R) no of AA 4 11 26 Guanidinium

Glu (E) no of AA 5 2 59 Carboxyl

Asp (D) no of AA 9 7 40 Carboxyl

Cys (C) no of AA 0 8 35 Thiol

His (H) no of AA 5 1 17 Imidazole

Lys (K) no of AA 9 7 60 primary amine

Tyr (Y) no of AA 13 3 21 Phenol

Page 67: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 4 - Introduction

57 | P a g e

Water soluble carbodiimide chemistry is often used to catalyze carboxyl reactions with

primary amine groups when immobilizing proteins. One example of such amide bond

formation is the use of both N-hydroxy succinimide (NHS) and ethylenediamine carbodiimide

(EDC), which forms an active ester intermediate as shown in Figure 1 [87, 194, 195]. The

next step involves nucleophilic attack on the carbonyl of the active ester adjacent to the amine

is also shown in Figure 1. This second step is highly sensitive to the state of ionization of the

amine group, since deprotonated amines (neutral) are nucleophilic while protonated amines

(positively charged) are not nucleophilic. The state of protonation depends on the pH of

reaction medium relative to the isoelectric pH of the protein being immobilized [87]. The pI

values of the proteins used in the immobilization experiments in this project are listed in Table

3.

Figure 1. The schematic of the mechanism of sulfo-NHS-EDC mediated coupling of

enzyme molecules onto carboxyl coating surfaces such as end-capped PEG SAMs [196,

197].

The determination of the extent of activity retained by immobilized enzyme can be

Page 68: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 4 - Introduction

58 | P a g e

measured using colorimetric assays. In this study the amount of enzyme on the surfaces was

quantified using XPS by monitoring changes in the N 1s signal from the nitrogen containing

amino acids of enzyme molecules. The activity of enzyme immobilized on the PEG surface

was quantified by a colorimetric assay using a chromogenic substrate, which was an enzyme

cleavable oligopeptide coupled to para-nitroaniline (pNa) [183]. The intact unhydrolyzed pNa

molecule (bound to oligopeptide) has an absorption maximum at 316 nm, whereas the free p-

Nitroaniline (pNa) has an absorption maximum at 380 nm. Thus the rate of change of

absorbance at 385 nm allows a measure of the hydrolysis of oligopeptides [198]. In general

practice absorption is measured at 405 nm instead of 385 nm to minimize the interference

due to the absorption peak at 310 nm due to intact unhydrolyzed pNa molecules [198]. The

enzyme functionalized surfaces were tested for their ability to resist bacterial adhesion using

Staphylococcus xylosus (S. xylosus) and Staphylococcus aureus (S. aureus). The bacteria S.

xylosus is a strain of bacteria highly relevant to dairy industry, where there is interest in

preventing fouling by our industrial collaborator Arla Foods A/S. S. aureus is a clinically

relevant bacterium that is responsible for a number of infections and also responsible for the

failure of implants.

II. Materials and Methods

A. Chemicals

Gold surfaces were cleaned the same way as described in Chapter 3. 2 kDa α-

carboxyl-ω-thiol poly(ethylene glycol) (PEG-COOH, >98% purity) was purchased from Laysan

Bio Inc. (Alabama, USA); hydroxyl-terminated tri (ethylene glycol) undecane thiol (OEG, 99%

purity) and carboxy-terminated tetra (ethylene glycol) undecane thiol (OEG-COOH, 99%

purity) were purchased from Assemblon (Washington, USA). Subtilisin A (from Bacillus

licheniformis), chicken lysozyme (Lyz), bovine serum albumin (BSA), octane thiol (R) (99%

purity), SYBR Green II, pNa, phosphate buffered saline (PBS) buffer tablets, 25% ammonium

hydroxide (NH4OH), 30% hydrogen peroxide (H2O2) and absolute ethanol were purchased

from Sigma Aldrich (Aarhus, Denmark). Chromogenic substrate for subtilisin suc-ala-ala-phe-

pNa was purchased from Bachem, Switzerland. Ultrapure MilliQ (MQ) water with the

Page 69: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 4 – Materials and methods

59 | P a g e

resistivity of 18.2 MΩ was used for buffers and aqueous ethanolic solutions.

B. Gold substrate functionalization

The SAMs of PEG-COOH thiols backfilled with OEG thiols were prepared as described

in Chapter 3. Mixed SAMs of OEG and OEG-COOH thiol were prepared by co-adsorption

from solutions. The gold slides were incubated in 1mM thiol solutions (appropriate

proportions) in ethanolic solutions (99.8%) for 24h. The functionalized slides were rinsed with

ethanol to remove any physically adsorbed OEG or OEG-COOH thiol molecules.

Figure 2. The chemical structures of thiolated molecules used for functionalizing gold.

The following notations for SAM of PEG and for the backfilled surface:

OEG + OEG-COOH : mixed OEG and OEG-COOH SAM created by coadsorption

PEG-COOH+OEG: PEG-COOH SAM backfilled with OEG

PEG-COOH+R: PEG-COOH SAM backfilled with R

C. Enzyme immobilization

The NHS-EDC based coupling of proteins is a well studied reaction. For example

-CH2 -NCO(OCH2CH2)45 – COOH HS

(PEG-COOH)carboxyl capped 2 kDa thiolated poly (ethylene glycol)

(OEG)

(OCH2CH2)3 –OH HS oligo (ethylene glycol)3 terminated alkane thiol

HS

(R)alkane thiol

(OEG-COOH)

(OCH2CH2)3 –COOH HS Carboxyl capped oligo (ethylene glycol)4 terminated alkane thiol

Page 70: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 4 – Materials and methods

60 | P a g e

Grainger et al reported the need for a 15 minutes carboxyl activation step using 0.5M NHS-

EDC in MQ followed by 30 minutes incubation with an amine containing molecule to perform

amide bond based biomolecular immobilization [194, 199]. Optimal NHS-EDC concentrations

required for immobilization of subtilisin was determined experimentally by systematically

varying the NHS-EDC concentration from 1 mg-10 mg/ml, 10 mg-100 mg/ml, 100 mg-1 g/ml

and 500 mg-5 mg/ml in MQ water for 30 minutes. After the NHS-EDC incubation, the

activated slides were incubated in 1 mg/ml subtilisin solution in PBS (pH 7.4) for 3h. Based on

these findings the OEG/OEG-COOH functionalized surfaces were activated for 30 minutes

with a mixed solution of EDC and NHS (50 and 5 mg/ml respectively) at pH 6.0 in MQ water,

followed by incubation with 1 mg/ml subtilisin solution for 3h. The enzyme immobilized

surfaces were rinsed with 100mM PBS (pH 7.4) to remove loosely bound enzyme molecules

prior to characterization and testing. During the NHS-EDC coupling experiments it was

observed that air bubbles were formed on PEG-COOH functionalized surfaces. Thus the

NHS-EDC concentration had to be lowered. It is known that sulfo-NHS has higher solubility

than NHS, thus a lower concentration of sulfo-NHS was used in order to minimize bubble

formation. The enzyme immobilization on SAMs of PEG-COOH backfilled with OEG thiols

was done the same way as mentioned above, except for lower EDC/Sulfo-NHS (2/0.2 mg/ml)

concentration used. Since the carboxyl activation conditions used were different for

OEG/OEG-COOH and PEG-COOH systems, we could not make direct comparison between

these sets of samples. Control experiments were also performed using denatured subtilisin

enzyme. Enzyme denaturation was performed using a PCR machine for heating active

enzyme solutions in PBS (pH 7.4) at 90° C for 3h. The heated solution of enzyme was tested

for activity, which confirmed that all activity was lost. Since the immobilization condition

required for subtilisin immobilization was different from those reported in the literature using

other proteins, it was checked if the surfaces used in the project had something to do with the

low NHS-EDC concentration required for protein immobilization. Lysozyme and BSA was

used to perform immobilization experiments on PEG-COOH SAMs backfilled with OEG thiols

using the NHS-EDC concentrations similar to that reported by Grainger et al. [194, 199]. The

PEG-COOH slides were activated using EDC-NHS (0.1/0.1 M) for 20 minutes followed by

incubation of 1 mg/ml solutions of either lysozyme (PBS pH 7.4) or BSA (acetate buffer pH

Page 71: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 4 – Materials and methods

61 | P a g e

4.0) for 30 min. This incubation step was followed by rinsing with PBS and then with MQ. The

slides were then dried using a jet of nitrogen gas, followed by XPS analysis.

D. Enzyme quantification by XPS and activity measurement

The quantification of enzymes on the surfaces was done using XPS by monitoring

changes in the N 1s signal from the survey spectra. The activity of the immobilized subtilisin

on the PEG surfaces was quantified by a colorimetric assay. The colorimetric assay was

performed by immersing 1cm×1cm substrates in 12 well microtiter plates containing 2 ml,

5mM suc-ala-ala-phe-pNa solution (HEPES buffer pH 8.0) and measuring the absorbance (at

405 nm) of 2 μl reaction mixture which was pipetted out of the reaction mixture once every 3

minutes. The absorbance was monitored using a nanodrop instrument (NanoDrop 1000,

Nanodrop products, Deleware, USA) capable of measuring absorbance of 1-10 μl volumes.

The small sampling volume was important in order to ensure that the volume of chromogenic

substrate solution did not decrease significantly during the measurement. Based on the

measured rate of change of absorbance, the rate of change of pNa concentration was

calculated using a calibration curve obtained from absorbance measurements using various

concentrations of pNa. The unit of enzyme activity is the enzyme unit (U- μM/min) and is

defined as the number of micromoles of substrate modified in one minute [200].

E. Bacterial adhesion assay

Bacterial adhesion assays were performed using two gram positive bacteria, namely,

Staphylococcus xylosus (S. xylosus DSM 20266) and Staphylococcus aureus (S. aureus

ATCC 12598). A starter culture was inoculated from agar plates and grown in 3 ml of 1%

tryptic soy broth (TSB) medium in 50 ml conical bottom tube by incubating overnight in a

shaker at 37°C. An inoculum prepared by adding 1 ml of starter culture to 100 ml 1% TSB

medium was incubated in a shaker at 37°C until cultures were in the late-exponential growth

phase (OD600 = 1.0). Cells were then harvested by centrifugation (5 minutes at 3000 x g),

washed twice and resuspended in PBS (pH 7.4) and diluted to obtain the required optical

density (OD). The slides to be tested for bacterial adhesion were exposed to bacterial

suspensions of S. xylosus: OD-0.01, incubation time 2h; and S. aureus: OD-0.1, incubation

Page 72: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 4 – Materials and methods

62 | P a g e

time of 5h, on an orbital shaker at 37˚C. The slides were rinsed with PBS and stained with

SYBR green dye. Bacterial counting was done by manual counting using a fluorescence

microscope. Cells were counted in 109 µm2 grids in triplicate with a total of fifteen random

positions on each type of surface. Adherent cells were counted manually by fluorescence

microscopy using a Zeiss Axiovert 200M epifluorescence microscope (Carl Zeiss GmbH,

Jena, Germany) equipped with Zeiss filter set and 10 and 63x oil immersion objective.

Student T test was used to assess the statistical significance results during comparison.

III. Results

A. Subtilisin immobilization OEG­COOH/OEG mixed monolayer

i. XPS characterization of the OEG­COOH/OEG mixed surfaces The optimum NHS-EDC concentration required for immobilization of subtilisin was

found experimentally by performing immobilization at varying concentrations of NHS-EDC.

Except for the concentration of NHS-EDC, the immobilization of subtilisin on 100% OEG-

COOH SAM, and characterization using XPS was done the same way as described in

methods section. The results of this study are presented in Table 4. The amount enzyme

immobilized was followed using the relative atomic percentage of nitrogen. The maximum

enzyme immobilization was achieved using a NHS-EDC concentration of 100 mg-1 g/ml, and

achieved a %N of 4.6%. Surprisingly the %N decreased at higher NHS-EDC concentrations

(500 mg-5 g/ml). On the other hand the decrease in %N at lower NHS-EDC concentrations is

expected since at lower concentrations of NHS-EDC, there would be a decreased number of

activated carboxyl groups. Thus the increase in potential sites for immobilization of subtilisin

molecules beyond a certain threshold leads to a decrease in the total number of molecules

immobilized. To understand the relationship between surface coverage of subtilisin and

specific activity, we performed immobilization experiments on surfaces with varying

proportions of COOH, created by co-adsorption of mixtures of OEG-COOH and OEG thiols.

Table 4. Results of XPS studies on 100% OEG-COOH SAM immobilized with subtilisin

using different NHS-EDC concentrations of 1 mg-10 mg/ml, 10 mg-100 mg/ml, 100 mg-1 g/ml

Page 73: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 4 - Results

63 | P a g e

and 500 mg-5 mg/ml. The relative atomic percentages were obtained from survey spectra.

NHS-EDC Concentration

C rel at%

O rel at%

S rel at%

Au rel at%

N rel at%

1 mg-10 mg/ml 48.3±1.2 11.7±0.5 2.4±0.4 37.6±0.4 0±0

10 mg-100 mg/ml 51.3±1.9 12.8±1.5 2.1±0.6 32.4±2.6 1.5±0.2

100 mg-1 g/ml 57.8±0.7 15.5±0.6 1.1±0.2 20.9±0,8 4.6±0.8

500 mg-5 mg/ml 50.4±1.1 12.3±1.6 2.1±0.3 32.1±2.0 2.8±0.5

Mixed monolayer of OEG-COOH/OEG thiols was created by

coadsorption. The relative concentrations of OEG-COOH/OEG in solution may not be the

same as their concentration on the surface, since the kinetic parameter of SAM formation

depends on the molecular weight and also on their chemical properties such as charge, and

hydrophobicity. The results of XPS studies on mixed monolayer surfaces are presented in

Table 5. The SAM of OEG thiol is a well studied and characterized system. Harder et al

reported a cross section area of 21.3 A2/molecule [46]. The density of OEG-COOH SAM was

calculated from the thickness of the SAMs as described in Chapter 2 based on the

attenuation of substrate signal. The cross-sectional area of OEG-COOH molecules in the

SAM was found to be 28.8 A2/molecule. The higher cross-sectional area for OEG-COOH

when compared to OEG could be due to the presence of the terminal carboxyl group that

might sterically hinder the packing compared to the OEG SAM. Using the cross-sectional area

for 100% OEG-COOH SAM, the COOH groups/cm2 was calculated to be 3.5×1014/ cm2, this

assumption is valid since each OEG-COOH molecule terminates with a carboxyl group. Since

the OEG SAM does not possess any COOH groups we assumed the carboxyl surface

concentration for OEG SAM to be zero. Figure 3 shows a plot of ratio of peak areas

COOH/alkane against the surface concentration of carboxyls. The peak areas of COOH and

alkane were obtained from high resolution C1s spectra shown in Figure 4. The variation of

surface COOH concentration was assumed to vary linearly with change in relative proportions

of OEG and OEG-COOH thiols on the surface. An estimate of the surface concentration of

COOH groups present in mixed monolayers created using 25% and 75% OEG-COOH thiol

Page 74: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 4 - Results

64 | P a g e

(in solution) was made by correlating the COOH/alkane ratios to the surface COOH

concentration in the plot shown in Figure 3

Table 5. Results of XPS studies on mixed monolayers of OEG/OEG-COOH thiols

created by co-adsorption. The relative peak area percentages were calculated from the

individual peaks in C 1s high resolution spectra.

OEG-COOH % Alkane PEG COOH COOH/Alkane COOH/cm2 (in solution) 285 eV % 286.8 eV % 289.7 eV % ratio (on surface)

CM-OEG 100% 50.5±0.1 41.1±0.1 4.7±0.1 0.092±0.002 3.5E+15

CM-OEG 75% 50.5±0.1 41.5±0.1 4.1±0.1 0.080±0.001 2.70E+15

CM-OEG 25% 50.8±0.1 42.4±0.1 3.6±0.1 0.070±0.001 2.00E+15

CM-OEG 0% 48.5±0.3 46.5±0.7 1.9±0.1 0.038±0.002 0

Figure 3. A plot of COOH/alkane peak percentage ratio (y axis) against surface

COOH concentration (x axis). The data points denoted by ♦ and Δ correspond to OEG SAM

and OEG-COOH SAM, respectively; while × and ∗ represent the mixed monolayer created

using 75% and 25% OEG-COOH, respectively.

0.03

0.04

0.05

0.06

0.07

0.08

0.09

0.1

0 6E+14 1.2E+15 1.8E+15 2.4E+15 3E+15 3.6E+15

Rat

io o

f CO

OH

% /

Alka

ne%

COOH groups / cm2

OEG‐COOH/OEG mixturesCOOH surface concentration

×∗

♦(0% OEG-COOH)

25% OEG-COOH 75%

OEG-COOH

100% OEG-COOH

Page 75: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 4 - Results

65 | P a g e

Figure 4. XPS high resolution C1s spectra of mixed layers created using 100%, 75%,

25% and 0% solution concentration of OEG-COOH thiol by coadsorption with OEG thiols.

ii. Determination of the enzyme content and activity The quantification of surface subtilisin concentration was done using XPS based on the

relative atomic percentage of nitrogen. Based on the calculations described in Chapter 2, the

thickness of the immobilized subtilisin layer was determined. Using the value of molecular

weight and assuming dry protein density of 1.27 g/cm3 [201], number of moles of subtilisin

and thus the number of molecules per unit area was calculated. The results of these

calculations are summarized in Figure 5. The surface concentration of subtilisin was found to

increase with an increase in the surface concentration of surface COOH groups, although the

increase was not linear. The enzyme concentration on surfaces with 75% and 100% OEG-

COOH did not differ significantly even though their surface COOH concentration differed by 8

×1014/cm2. This suggests that there is limitation in the number of molecules of subtilisin that

35

40

45

50

55

CPS

294 292 290 288 286 284 282 280Binding Energy (eV)

285.0 eV

286.8 eV

289.6 eV OEG-COOH %100%

OEG-COOH %75%

OEG-COOH %25%

OEG-COOH %0%

High resolution C 1sspectra

Page 76: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 4 - Results

66 | P a g e

can be immobilized per unit area irrespective of the number of binding sites available for

covalent coupling. The surface with 0% OEG-COOH did not have any detectable amount of

enzyme, indicating that there is minimum amount of non-specific adsorption. The possibility of

non-specific adsorption on surfaces with significant surface concentrations of carboxyl cannot

be ruled out, since at pH 7.4 (reaction buffer PBS), the carboxylic acid group carries negative

charge while the subtilisin itself would be positively charged (pI 9.4), thus creating room for

electrostatic interactions. The washing step should ideally remove weakly bound enzyme

molecules, thus ensuring minimum non-specific adsorption.

Figure 5. Concentration of subtilisin on mixed monolayers of OEG/OEG-COOH,

determined based on the relative atomic percentage measured using XPS. The percentage of

OEG-COOH indicated on the x axis is the concentration in solution during coadsorption.

The activity of subtilisin immobilized surfaces was measured using the colorimetric

assay as described in methods section. The results of colorimetric studies are summarized in

Figure 6a. Since the number of enzyme molecules per unit area was already determined, it

was possible to calculate the activity per molecule of subtilisin and the results of this analysis

is shown in Figure 6b. The mixed OEG-COOH/OEG thiol surface with 50% OEG-COOH had

an enzyme activity of approximately 455 mU/cm2 with 4.4 × 1011 molecules/cm2, while the

surface with 100% OEG COOH had much lower activity 121 mU/cm2 in spite of higher

subtilisin 8.8 ×1011 molecules/ cm2. The surfaces with 0% and 25% OEG-COOH showed no

0

2E+11

4E+11

6E+11

8E+11

1E+12

1.2E+12

1.4E+12

0% 25% 50% 75% 100%

Enzy

me

mol

ecul

es/c

m2

OEG-COOH % (Solution)

Surface concenration of subtilisin on OEG/OEG-COOH mixed SAM

Page 77: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 4 - Results

67 | P a g e

enzyme activity and also had lower enzyme concentrations when compared to higher OEG-

COOH surface contents. This absence of direct correlation between enzyme concentration

and surface activity could be due to autolysis of enzyme molecules, which is a well observed

process with subtilisin [183] and its variants.

Figure 6. Enzyme activity results of mixed monolayers of OEG/OEG-COOH

immobilized with subtilisin. (a) Shows the activity per unit area of surfaces expressed in

mU/cm2. (b) Summarizes the activity per enzyme molecule values of various surfaces. The

percentage of OEG-COOH indicated on the x axis is the concentration in solution during

coadsorption.

One would expect higher autolysis when the enzyme molecules are present at higher

concentrations on the surface, and the nitrogen quantification indicated that 100% and 75%

02E-104E-106E-108E-101E-09

1.2E-09

0% 25% 50% 75% 100%

mU

/sub

tilis

in

OEG-COOH %(solution)

Activity/enzyme molecule(mU/subtilsin)

0

100

200

300

400

500

0% 25% 50% 75% 100%

Act

ivity

(mU

/cm

2 )

OEG-COOH (solution) %

Enzyme activity (mU/cm2) (a)

(b)

Page 78: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 4 - Results

68 | P a g e

OEG-COOH have higher enzyme contents when compared to 50% OEG-COOH surfaces. An

alternate explanation of lower activity at high surface concentrations could be due to

restrictions on activity due to higher packing, which might hinder the accessibility of the active

site of the enzyme towards the substrate. Apart from autolysis, denaturation is also a

significant process that hinders the success of creating subtilisin immobilized surfaces with

optimum activity. Several groups working with subtilisin solved the denaturation issue by

protein engineering techniques to enhance the stability of naturally occurring enzyme

molecules [178, 179].

iii. Bacterial adhesion studies

The results of bacterial adhesion studies on subtilisin immobilized mixed SAMs of

OEG-COOH/OEG thiols are presented in Figure 7. The surfaces with 50% and 100% OEG-

COOH thiol had approximately the same number of cells, but the 75% OEG-COOH surface

had a cell count approximately higher by a factor of 5. The order of increasing enzyme activity

of surfaces is 0% = 25% <100%<75% <50%. There was no correlation between the results of

enzyme activity studies and bacterial adhesion assays. Also the amount of enzyme on the

surfaces did not correspond either to the enzyme activity on the surfaces or to the number of

bacteria adhering to the surfaces. This could be due to the fact that subtilisin is incapable of

minimizing the adhesion of S. xylosus. This absence of direct correlation between enzyme

concentration and surface activity could be due to autolysis of enzyme molecules which is

well known process observed with subtilisin [183]. The subtilisin immobilization studies on

mixed monolayers of OEG/OEG-COOH thiols helped in understanding the relationship

between the surface coverage and enzyme activity. With this understanding further

investigations were done by performing enzyme immobilization studies on mixed layer of

PEG-COOH and OEG thiol. The bacterial inhibition ability of subtilisin was tested using active

and denatured forms of the enzyme immobilized on PEG-COOH surfaces backfilled with OEG

thiol. The preparation of these surfaces was discussed in Chapter 3. The denatured subtilisin

molecules do not possess proteolytic activity and thus if proteolytic activity is incapable of

preventing bacterial adhesion, then there should not be any significant difference in bacterial

adhesion on surfaces functionalized with active and denatured enzymes. Also, S. xylosus

Page 79: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 4 - Results

69 | P a g e

could be outside the list of bacterial strains that can be prevented from forming biofilms by

subtilisin. S. xylosus was initially chosen since it is highly relevant to the dairy industry. It has

been reported that S. aureus can be prevented from forming biofilm by subtilisin like

proteases [202] and hence the subtilisin based surfaces were tested against S. aureus. The

motive behind this study is to understand the effect of backfilling on the performance of

immobilized enzyme.

Figure 7. Results of bacterial adhesion assays performed using S. xylosus, OD-0.01,

incubation time 2h, 37°C (y-axis: cells/μm2). The bacterial count on gold was 1.7±0.2×10-3

B. Subtilisin immobilization on mixed PEG­COOH/OEG surface

i. XPS characterization of backfilled PEG­COOH surface The preparation and characterization of surfaces formed by backfilling PEG-COOH

SAM with OEG or R thiol was discussed in detail in chapter 3. Some of the important results

of the XPS analysis are summarized in Table 6. Since the ratio of the alkane/ether peak area

for OEG SAM is known, it is possible to estimate the relative contribution made to the PEG

peak from OEG thiol molecules present in the backfilled PEG-COOH SAM using the area of

the alkane peak. Once the contribution of OEG thiol is deducted, it is possible to estimate the

thickness of the PEG-COOH overlayer, and using the thickness the graft density can be

calculated, as described in chapter 2. Since each PEG-COOH molecule is end capped with

one carboxyl group, and OEG thiol or R has no carboxyl groups, thus it can be assumed that

the number of PEG-COOH molecules per unit area is same as the number of carboxyl groups

0

0.5

1

1.5

2

0% 25% 50% 75% 100%

Cel

ls /μ

m2

×10

-3

OEG-COOH (solution) %

Bacterial cells/μm2

Page 80: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 4 - Results

70 | P a g e

per unit area. The results for the carboxyl concentration estimations on backfilled PEG-COOH

surfaces are summarized in Figure 8.

Table 6. Summarizes results of XPS analysis of PEG-COOH SAM backfilled with OEG

or R thiol molecules. The Au%, O/C and S% were obtained from survey spectra. The

alkane/PEG ratio and COOH% were obtained from high resolution C1s spectra.

In Chapter 3 it was speculated that backfilling PEG-COOH SAM with R thiol molecules

results in a higher amount of PEG-COOH desorption when compared to that caused by OEG

thiol addition. Desorption of PEG-COOH molecules should cause a decrease in the surface

concentration of carboxyl groups. The results of our estimation of COOH concentration clearly

shows that PEG-COOH surface backfilled with R thiol has approximately 20% lower COOH

concentration than that of the PEG-COOH SAM or the SAM backfilled with OEG thiols as

shown in Figure 8. In Chapter 3 it was demonstrated that backfilling PEG-COOH SAM using

OEG thiols improved its ability to resist serum adsorption. Based on these results we expect

minimum non-specific adsorption of subtilisin on PEG-COOH+OEG when compared to PEG-

COOH SAM or PEG-COOH+R.

PEG-COOHPEG-COOH+

OEG PEG-COOH+

R

Au % 39.0± 0.4 36.2± 0.5 41.6± 0.5

O/C 0.49±0.03 0.42±0.01 0.40±0.01

Alkane/PEG 0.1± 0 0.3± 0 0.4± 0

S % 2.1± 0.2 1.9± 0.6 2.1± 0.2

COOH % 4.5± 0.3 3.5± 0.1 3.6± 0.3

Page 81: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 4 - Results

71 | P a g e

Figure 8. Surface COOH concentration on a PEG-COOH SAM backfilled with OEG

and R thiol. The estimation was based on PEG/alkane peak ratios obtained from high

resolution C 1s XPS spectra.

ii. Subtilisin immobilization on mixed PEG­COOH SAMs Subtilisin enzyme was immobilized on mixed monolayers of PEG-COOH SAM back

filled with OEG or R thiols as described in the methods section. XPS was used to quantify the

amount of enzyme immobilized on PEG-COOH based SAMs. The relative atomic percentage

was used to determine the thickness of immobilized enzyme layer, using this value the

enzyme molecules per unit area was estimated as described in the previous section. The

surface concentrations of enzyme on PEG-COOH SAM, PEG-COOH+OEG and PEG-

COOH+R are summarized in Figure 9.

Figure 9. Surface enzyme concentration on a PEG-COOH SAM backfilled with OEG

and R thiols. The estimation was based on %N obtained from XPS survey spectra.

5E+14

5.5E+14

6E+14

6.5E+14

7E+14

7.5E+14

8E+14

PEG-COOH PEG-COOH+OEG PEG-COOH+R

CO

OH

gro

ups/

cm2

Surface carboxyl concentrationCOOH groups/cm2

1E+12

1.4E+12

1.8E+12

2.2E+12

2.6E+12

PEG-COOH PEG-COOH+OEG PEG-COOH+R

Enzy

me

mol

ecul

es/c

m2

Surface subtilisin concentrationEnzyme/cm2

Page 82: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 4 - Results

72 | P a g e

Surprisingly, there was no significant difference in the amount of enzyme on surface

between the three surfaces analyzed. The enzyme immobilized surfaces did not differ in their

colorimetric activity values and also did not exhibit any significant difference in activity per

enzyme molecule value as shown Figure 10 and Figure 11, respectively

Figure 10. Colorimetric activity of subtilisin immobilized surfaces; PEG-COOH SAM,

PEG-COOH+OEG and PEG-COOH+R. Activity unit : mU/cm2.

Figure 11. Activity per subtilisin molecule immobilized on a PEG-COOH SAM

backfilled with OEG and R thiols. The estimation was based on N% obtained from XPS

survey spectra. Activity unit: mU/molecule.

4E-11

5E-11

6E-11

7E-11

8E-11

PEG-COOH PEG-COOH+OEG PEG-COOH+R

Activ

ity p

er e

nzym

e m

olec

ule

(mU

)

Activity per subtilisin molecule(mU)

Page 83: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 4 - Results

73 | P a g e

iii. Bacterial adhesion studies on subtilisin immobilized mixed PEG­

COOH/OEG surfaces

Bacterial adhesion studies using S. aureus were performed on mixed monolayers

immobilized with active and denatured enzyme using the procedure described in methods

section. The inclusion of denatured enzyme would help to check the capability of subtilisin to

minimize bacterial adhesion. The results of adhesion studies are shown in Figure 12. The

subtilisin immobilized surfaces resist bacterial adhesion better than the surfaces immobilized

with denatured subtilisin. For example, the SAM of PEG-COOH with active subtilisin has a

cell count of 11×10-3 cells /μm2, whereas with denatured subtilisin a much higher cell count of

56 ×10-3 cells /μm2 was observed. This result confirms there is some activity of subtilisin is

capable of decreasing the adhesion of S. aureus. Also, the PEG+OEG surface with no

enzyme is the best among all surfaces in resisting bacterial adhesion. Thus, we conclude that

native subtilisin A immobilized on PEG-COOH or OEG-COOH is not any better than just the

PEG-COOH or OEG thiol SAMs at resisting bacterial adhesion, despite the detection of

significant amount of enzyme activity.

Figure 12: Bacterial adhesion assay results performed using S. aureus, OD-0.1,

incubation time 5h, 37°C (y-axis : cells/μm2) on PEG, PEG+R, PEG+OEG surface; bare

PEG-COOH mixed surfaces (NoEnz), active enzyme (ActEnz) and denatured enzyme

(DeEnz) immobilized surfaces. Coupling was done by using sulfo-NHS-EDC catalyzed

reaction as described in methods section. The count on alkane thiol (R) SAM was 11±0.4

0

20

40

60

80

100

120

NoEnz DeEnz ActEnz NoEnz DeEnz ActEnz NoEnz DeEnz ActEnz

PEG-COOH PEG-COOH+OEG PEG-COOH+R

Cel

ls /μ

m2

×10

-3

Adhesion studies using S. aureusBacterial cells/μm2

Page 84: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 4 - Results

74 | P a g e

cells/μm2.

Therefore, the subtilisin immobilized surface may not be efficient enough to be an

effective antifouling surface in comparison to the PEG-only surfaces. The average N%

obtained using PEG-COOH based surfaces is higher than OEG-COOH based surfaces.

Although the N% of the PEG-COOH or PEG-COOH+OEG is higher, the activity measured

using the colorimetric assay is lower than that for OEG-COOH (50%, 75% and 100%) by at

least a factor of two. This much lower value could be due to a higher proportion of autolysed

subtilisin molecules (as a result of high concentration) or higher proportion of denatured

molecules. An alternate reason could be blocking of the active site of subtilisin enzyme at

high surface concentrations. Further investigation needs to be done to find out the exact

mechanism by which the subtilisin molecules lose activity upon immobilization at high surface

concentrations. It was surprising to find out that the surface concentration of subtilisin reached

a maximum with a NHS-EDC concentration of 100 mg-1 g/ml, and the concentration

decreases at higher concentration of NHS-EDC. At higher NHS-EDC concentrations one

might expect higher surface concentrations due to availability of greater number of activated

carboxyl groups for amide bond formation with amine residues of the enzyme. On the

contrary, the opposite effect is observed with subtilisin. In literature examples can be found of

similar protein immobilization experiments made using NHS-EDC concentrations higher than

100 mg-1 g/ml by a factor of 10 at least [194, 199]. The main difference between those

systems and the here was the surface of immobilization and the protein being immobilized. To

rule out the possibility that the backfilled PEG-COOH surface could be responsible for the

observation of lower protein coupling at high NHS-EDC concentrations, immobilization

experiments were performed on mixed PEG-COOH monolayers using lysozyme and BSA.

Lysozyme was chosen since its pI is closer to that of subtilisin. More importantly lysozyme is

a glycoside hydrolase capable of damaging the integrity of extracellular polysaccharides of

bacterial cells and could potentially prevent them from adhering to surfaces. Also, the enzyme

has been shown to be a robust protein capable of being immobilized on various types of

surfaces such as steel, titania glass and range of organic polymers with significant retention

of activity [172, 203, 204]. BSAis commonly used in most post-immobilization steps to block

the sites of non-specific adsorption.

Page 85: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 4 - Results

75 | P a g e

C. Immobilization of Lysozyme and BSA

Immobilization experiments with lysozyme and BSA were performed in order to find out

if the PEG-COOH mixed surface created by backfilling with OEG thiols is capable of

minimizing non-specific adsorption during covalent coupling of proteins. Immobilization was

identical to those described. The results of nitrogen quantification using XPS are shown in

Figure 13.

Figure 13. Studies on lysozyme and BSA immobilized SAMs of PEG and PEG+OEG

(PEG-COOH thiol SAM backfilled with OEG thiols). Protein molecules immobilized with amd

without the use of EDC/NHS. The y-axis shows the % of N measured using the survey

spectra of XPS.

In order to gain insight into the amount of non-specifically adsorbed protein,

experiments were carried out with and without EDC/NHS. In the absence of EDC/NHS no

covalent coupling of Lysozyme and BSA can occur. It was discussed in Chapter 3 that the

PEG-COOH+OEG surface is better at preventing non-specific adsorption when compared to

PEG-COOH SAMs. From the XPS results, nitrogen signals were absent on PEG-

COOH+OEG surfaces exposed to BSA and lysozyme without EDC/NHS, whereas 3.8% N

and 1.3% N were detected on the surfaces of PEG-COOH SAMs, respectively. It should be

noted that we observed similar results ( 0% N ) during immobilization experiments performed

using subtilisin without NHS-EDC on mixed OEG-COOH/OEG monolayers created by co-

adsorption (data not shown). In the experiments using EDC/NHS activation, the N% on PEG-

-1

1

3

5

7

PEG PEG+OEG PEG PEG+OEG PEG PEG+OEG PEG PEG+OEG

NHS-EDC NHS-EDC

Lysozyme BSA

N

ato

mic

%

N atomic % (XPS)

Page 86: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 4 - Results

76 | P a g e

COOH and PEG-COOH+OEG was 5.3% and 3.5% for BSA, and 3.9% and 2.6% for

lysozyme, respectively. It is speculated that the higher atomic N% with PEG SAMs in

comparison to PEG+OEG is due to a non-specific protein adsorption component in addition to

the contribution from covalent coupling. Thus the mixed PEG-COOH+OEG surface is a

suitable platform for immobilizing active biomolecules using the conventional protocols

available in the literature that use higher NHS-EDC concentrations than the optimal

concentration required for immobilization subtilisin.

IV. Conclusions and discussion

Immobilized subtilisin was investigated on co-adsorbed OEG-COOH/OEG surfaces using an

enzyme activity assay, nitrogen quantification using XPS and bacterial adhesion assays with

S. xylosus. It was found that a large proportion of immobilized subtilisin molecules were

inactive. There was no correlation between the amount of enzyme on surface and the number

of bacterial cells adhering onto it. This could be possible if the enzyme molecules denature or

autolyse upon immobilization at high surface concentrations. Immobilization studies using

active and denatured subtilisin molecules on PEG-COOH surface backfilled with OEG thiol

showed that the active enzyme activity is indeed capable of reducing the adhesion of S

.aureus relative to denatured enzyme. The bare PEG-COOH and PEG-COOH+OEG surface

were better at resisting bacterial adhesion (S. aureus) than subtilisin immobilized surfaces.

Since the carboxyl activation conditions were different for PEG-COOH and OEG-COOH

surfaces, direct comparisons could not be made for results of enzyme activity, N atomic

percentage and bacterial adhesion studies. Immobilization studies using lysozyme and BSA

showed that PEG-COOH+OEG surfaces have minimum non-specifically adsorbed protein.

Control experiments without the NHS-EDC activating agent demonstrated that PEG+OEG

surfaces are capable of efficiently resisting non-specific adsorption and hence most of the

adsorbed protein is covalently coupled. The bacterial attachment studies using S. xylosus and

S. aureus on subtilisin immobilized surface show that the attachment of former bacterial

species is unaffected by the activity of enzyme under the conditions used in this work, while

the attachment of later seem to be minimized by activity of enzyme. This difference in the

effect of enzyme on attachment could be due to the difference in the biological mechanism

Page 87: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 4 – Conclusion and discussion

77 | P a g e

used by them in the initial adhesion or structural differences in the extracellular matrix

surrounding the cell membrane. It should be noted that in the experiments with denatured

enzyme it was assumed that the surface concentration is same as that of obtained by

immobilization of active enzyme. The quantitative difference in the bacterial attachment

between surfaces immobilized with active and denatured enzyme could be due to difference

in amounts of protein immobilized rather than the activity of enzyme. Hence the confirmation

of role of enzyme activity in minimizing attachment of S. aureus further quantification of

surface immobilized with denatured enzyme.

Page 88: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

78 | P a g e

CHAPTER 5

Grafting of poly(ethylene glycol) (PEG) layers using supercritical carbon dioxide

I. Introduction

In our daily lives we commonly encounter matter in three phases; gas, liquid and solid.

Although the needs for most of our applications can be met with these phases of matter,

alternate phases such as plasma and supercritical fluids possess unique characteristics.

When a gas is compressed by increasing pressure or lowering temperature, it undergoes a

phase transition from gas to liquid and finally to solid. But under a certain range of pressure-

temperature conditions this phase transition results in a supercritical fluid (SFC) (Figure 1a).

SFC has a solvation ability close to that of a liquid while its diffusivity is similar to that of a gas

(Table 1). Thus the use of SFC as a solvent combines the advantages of liquids and gases

[205-207]. SFCs have extremely low surface tension, and thus high wettability. These unique

physical properties make the use of SFC very popular in small and large scale industrial

applications such as extraction or purification of natural products, reaction media, polymer

processing and nanoparticle synthesis [205, 207, 208]. Supercritical carbon dioxide (scCO2)

is the most used SFC since it is a ‘green’ solvent and enters the supercritical phase at

relatively moderate temperature-pressure conditions of 31°C/7.3 MPa [206] when compared

to other commonly used SFCs such as water and methanol. Several systematic studies have

been conducted to understand the relationship between the partition coefficient of molecules

in SFC and various pressure-temperature conditions [209]. In general, with an increase in

pressure, the solubility increases because of increases in solvent density [208-212]. The

temperature generally is inversely related to solubility (also with solvent density) [208], but this

relationship can be directly proportional instead of inverse if the vapor pressure of a

compound being dissolved is significantly high [210]. Apart from studying small molecules,

studies have been conducted extensively on the interactions of macromolecules such as

polymers within SFCs. Some of the well studied polymers are polyethylene, polystyrene,

poly(methyl methacrylate) and other industrially relevant polymers [211-214]. The solubility of

Page 89: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 5 - Introduction

79 | P a g e

polymers in SFC depends on the pressure and temperature similar to the solubility of small

molecules [211, 212].

Figure 1: (a) The phase diagram of CO2 at various ranges of pressure and

temperature, including the supercritical range. (b) A typical set-up used for compressing and

heating CO2 gas to make it supercritical.

Table 1: Comparison of the physical properties of solvent in gas, supercritical and

liquid phases [205].

Density

(kg/m3)

Viscosity

(mPa.s)

Diffusivity

(mm2/s)

Gas 1 10 1 - 10

SFC 100 - 1000 50 - 100 0.01 – 0.1

Liquid 1000 500 - 1000 0.001

The addition of co-solvents such as ethanol, water and toluene to scCO2 alters the

solubility of polymers [211]. Co-solvents increase the polarity of scCO2 thus improving its

ability to solubilize polar macromolecules. There are many important parameters that

influence the solubility of polymers in SFC such as molecular weight, polarity, type of end

groups and molecular architecture (e.g. branched/linear). As already discussed in Chapter 1,

for applications involving grafting polymers onto surfaces, the graft density depends on the

solubility of a polymer in a given solvent. The physical properties of SFC can be easily

manipulated thus it can be a versatile solvent in the field of surface science. Applications

Page 90: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 5 - Introduction

80 | P a g e

requiring solid phase chemical modification (reaction of bulk rather than just the surface) of

polymeric materials are efficiently done by scCO2 since it has the ability to diffuse into

polymeric materials (also known as swelling). For example, Liu et al. [213] reported solid state

grafting of maleic anhydride onto polypropylene using scCO2, and Kunita et al. [214] grafted

glycidyl methacrylate onto polypropylene surfaces using scCO2. Both Liu et al. and Kunita et

al. demonstrated that scCO2 is an efficient solvent for solid state transfer of reactants onto

polypropylene. SFCs such as scCO2 have the unique ability to chemically modify buried

interfaces. Jia et al. used scCO2 to perform a silane based surface functionalization of silanol

groups at the silicon/polystyrene interface (silanol groups buried underneath the polystyrene

layer) [215]. They showed that the supercritical CO2 phase was the reason behind the

success of the modification that could not be achieved using a liquid solvent. There have

been a few other reports on silane based functionalization of alumina [216], titania [216] and

silica [217] surfaces using scCO2. Although there are a number of reports on scCO2 based

surface modifications, there are no reports on grafting of polymeric chains onto surfaces. In

this chapter results are presented involving grafting of PEG thiol molecules onto gold

substrates using scCO2.

One of the main strategies used in ‘grafting to’ techniques is the use of ‘cloud point’

grafting, where the solubility of the polymer (e.g. PEG) is reduced using high ionic strength

and high temperature, thereby decreasing the excluded volume effects leading to high graft

densities. Cloud point conditions are easily attained using SFC by systematic variation of the

pressure-temperature conditions. There have been some reports on the effect of molecular

weight, end groups, pressure, temperature and co-solvent on the partition coefficients of PEG

in scCO2 [212, 218, 219]. There are no reports on studies involving effects of pressure-

temperature conditions on the surface PEG graft density. This chapter presents the results of

studies on grafting PEG thiol on gold using scCO2 at different conditions of pressure-

temperature and co-solvents. Comparisons of these results are made using conventional

liquid solvents such as ethanol, toluene and methanol.

Page 91: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 5 – Materials and methods

81 | P a g e

II. Materials and methods

A. Chemicals

α-methoxy-ω-thiol poly(ethylene glycol) ( 2, 5 or 10 KDa PEG thiol , >98% purity) were

purchased from Laysan Bio Inc. (Alabama, USA), Lysozyme (Lyz), BSA, Casein, 25%

ammonium hydroxide, 30% hydrogen peroxide, absolute ethanol (EtOH), methanol (MeOH)

and toluene (Tol) were purchased from Sigma Aldrich (Aarhus, Denmark). Ultrapure MilliQ

(MQ) water with a resistivity of 18.2 MΩ was used for making ethanolic solutions.

B. Substrate preparation and functionalization

Gold substrates were prepared by sputtering 50 nm gold layer onto a 3 nm titanium

adhesion layer on silicon wafers in the same way as mentioned in Chapter 3. The gold

surfaces were cleaned by UV/ozone treatment for 30 minutes, followed by treatment with

basic piranha solution (H2O: NH4OH: H2O2 in ratio 4:1:1) at 70˚-80˚C for 5 minutes and then

rinsed with MQ water. (CAUTION: Piranha solution reacts violently upon contact with organic

solutions). Grafting in liquid unless otherwise mentioned was done by immersing gold slides

for 3h at room temperature and ambient pressure (RTP 20°C/101 kPa) in 0.2 mM PEG thiol

solution, followed by rinsing with MQ water to remove any physically adsorbed molecules.

The SFC based grafting unless otherwise mentioned was done using a set-up shown in

Figure 1b. The gold slides and appropriate amount of PEG thiol were placed in the SFC

chamber (30 ml capacity), followed by filling up with CO2 and adjustment to the required

pressure-temperature. Experiments with scCO2-co-solvent (5% co-solvent v/v) mixtures were

performed by dissolving PEG thiol in 1.5 ml liquid co-solvent, which was placed in the SFC

chamber followed by placing of the gold slides. Precaution was taken to prevent the gold

slides coming into contact with the liquid by placing them on a Teflon support to keep the

substrate holder above the liquid at the bottom of SFC chamber. Unless otherwise mentioned

the SFC based functionalization was performed for 30 minutes at the required pressure-

temperature condition. Once the SFC chamber was depressurized, the slides were rinsed

with MQ to remove any physically bound PEG thiol molecules.

Page 92: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 5 – Materials and methods

82 | P a g e

C. X­ray photoelectron spectroscopy

XPS spectra were recorded the same as mentioned in Chapter 3. Overlayer thicknesses

were calculated based on attenuation of the substrate (Au) signal using Equation 8, as

described in Chapter 2 in the XPS section.

Where Z - overlayer thickness, λ – attenuation length, θ is take-off angle, I is substrate

signal with a thin film coating, I0 bare substrate intensity.

The quantification of protein adsorption was done using the relative atomic percentage of

nitrogen calculated based on the N 1s peak obtained from survey spectra of surfaces

exposed to 1 mg/ml lysozyme (Lyz) or bovine serum albumin (BSA) or casein in PBS at 37°C

for 24h. The protein exposed slides were rinsed with PBS followed by rinsing with MQ and

dried with nitrogen gas.

D. Ellipsometry

The overlayer thickness of PEG thiol monolayers were measured using ellipsometry.

An ELX-02C (DRE, Germany) ellipsometer with a He-Ne laser (632.8 nm) was used with an

incident angle of 70º. The calculations were performed according to the methods of Unsworth

et al. [150]. For the PEG modified surfaces, the polarizer (P) and analyzer (A) angles for the

null condition were measured and thickness values were determined using a three-layer (air–

PEO–gold) model. Complex refractive-index values (ni + ιki) of 0 + ι0 and (nf + ιkf) of 1.47 + ι0

were used for air and the PEG films, respectively. Final thickness values were obtained by

fitting the experimental data to the model using the Ellips 32 software.

E. Quartz crystal microbalance with dissipation (QCM­D)

QCM-D studies were performed using a Q-Sense E4 system (Gothenburg, Sweden). The

PEG thiol functionalization using either scCO2 or liquid solvents was performed directly on

QSX 301 crystals (Q-Sense) coated with 100 nm gold having an approximate resonance

frequency of 5 MHz. Protein adsorption experiments using BSA, casein and Lyz were

----- Eqn (8)

Page 93: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 5 – Materials and methods

83 | P a g e

performed in triplicate. Functionalized gold coated crystal surfaces were equilibrated at 37°C

in PBS (100 mM pH 7.4) prior to exposure to the protein solutions. Protein adsorption studies

were carried out using 1 mg/ml Lyz or BSA or casein in PBS (pH 7.4) at 37°C. Once a stable

baseline was attained, the surfaces were exposed to 750 μL of protein solution delivered at a

flow rate of 100 μL/minute followed by rinsing with PBS buffer. The shift in resonance

frequency, corresponding to the 7th overtone, was used to monitor the protein adsorption.

III. Results

A. Preliminary studies

Kingshott et al. have previously shown that covalent attachment of PEG layers is very

efficient in preventing fouling for a longer period of time when compared to non-covalently

attached PEG molecules [102]. Hence the nature of chemical interaction between PEG and

substrate is very important for determining the non-fouling efficiency of a PEG coating. Since

there have been no studies on the quality of PEG monolayers grafted using scCO2,

preliminary studies involved comparing the surfaces prepared from SFC and ethanol to

confirm that the scCO2 based PEG thiol grafting results in a monolayer that is chemically

similar to that formed from conventional solvents such as methanol. The main parameters

considered are the thickness of overlayer and the relative percentages of bound to unbound

thiols. The thickness was calculated using XPS, as described in Chapter 2. The high

resolution S2p spectra were used to determine the percentage of bound thiol, which

represents the amount of covalently bound PEG thiol molecules, as described by Castner et

al. [128].

The pressure-temperature conditions of SFC grafting used in this work were chosen

based on reports by Daneshwar et al. [219], Drohmann et al. [212] and Mishima et al. [218].

The preliminary SFC experiments were performed at 20 MPa/60°C and under these

conditions we expect a solubility of 0.5-1.5 wt% or approximately 5 mg/ml (assuming the

density of SFC to be 0.5 g/ml). Since there are no reports on the time required for PEG thiol

self assembly using SFC, our preliminary studies were aimed at optimizing the time needed

for monolayer formation. Figure 3 summarizes the results of kinetic studies involving PEG

Page 94: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 5 - Results

84 | P a g e

thiol functionalization carried out for varying periods of time using 75% liquid ethanol (room

temperature/pressure, 25°C/101 kPa) and scCO2 (60°C/20 MPa). The PEG thiol monolayer

formed using liquid ethanol after 30 minutes had approximately 40% bound S and this value

increased with the increase in the incubation time. Thus grafting in liquid ethanol for several

hours is required for the formation of a monolayer with an optimum amount of bound S. The

highest thickness attained in 75% ethanol was measured to be approximately 5.5 nm. On the

other hand, functionalization using scCO2 for 30 min produced a layer that was 6 nm thick

with approximately 63% bound S. Since the experimental set up used in scCO2 based

grafting requires a minimum time of 20 minutes to attain the set pressure/temperature

condition, it was impossible to do grafting experiments with incubation time less than 30

minutes. It is possible that scCO2 based functionalization of PEG thiol may require a much

lower incubation time than 30 minutes. Based on preliminary studies we decided to carry out

all our SFC based functionalization experiments for 30 minutes and liquid based self

assembly for 3 hours. The faster reaction in scCO2 could be due to faster diffusion of PEG

thiol molecules since scCO2 has a lower viscosity than liquids. Thus, it is concluded that

scCO2 requires a much lower time for forming a PEG thiol monolayer with an optimal

proportion of covalently bound molecules. Therefore, faster kinetics and the high wettability of

scCO2 make it an ideal solvent for efficiently grafting polymers such as PEG onto surfaces.

Figure 2: (a) Overlayer thickness; (b) percentage of bound sulfur of a 2 kDa PEG thiol

SAM grafted using 75% ethanol (EtOH:25°C/101 kPa), and scCO2 (SFC:60°C/20 MPa)

measured using XPS.

4.04.55.05.56.06.57.0

30 min 2h 6h 10h

Thic

knes

s (n

m)

Grafting time

Overlayer thickness (XPS)

EtOH

SFC30

45

60

75

90

30 min 2h 6h 10h

% o

f bou

nd S

ulph

ur

Grafting time

% of bound sulphur

EtOH

SFC

Kinetics of self assembly of 2 kDa PEG in EtOH and SFC(a) (b)

Page 95: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 5 - Results

85 | P a g e

The non-fouling abilities of the 2 kDa PEG thiol SAM coated using scCO2 (SFC:

60°C/20 MPa) and 75% ethanol (3h/25°C/101 kPa) were compared by determining the

nonspecific adsorption of Lyz, BSA and casein. Since this project is in collaboration with a

milk producing company (Arla Foods A/S), the protein casein was included which is known to

be the most abundant milk protein [220]. Lyz is an important part of immune system which

provides protection against gram positive bacteria [221]; BSA is the most abundant plasma

protein [222]. Protein adsorption studies were performed on the PEG thiol monolayer grafted

from SFC and ethanol. These surfaces are exposed to the proteins and the amount of protein

adsorbed on the surfaces was quantified using XPS and QCM. The results of protein

adsorption studies are summarized in Figure 3.

Figure 3: Adsorption studies of BSA, Lyz and casein quantified using XPS and QCM

on gold surfaces grafted with 2 kDa PEG thiol using liquid 75% ethanol (EtOH: 3h/25°C/101

kPa) and scCO2 (SFC: 30min/20 MPa/60°C). Figure 3a shows relative amounts of protein

-4

0

4

8

12

16

Gold EtOH SFC Gold EtOH SFC Gold EtOH SFC

BSA Lyz Casein

Ato

mic

per

cent

age

of

Nitr

ogen

(N %

)

Protein adsorption studied using XPS (N %)(a)

-0.5

0.5

1.5

2.5

Gold EtOH SFC Gold EtOH SFC Gold EtOH SFC

BSA Lyz Casein

Freq

uenc

y sh

ift(H

z -v

e sc

ale)

Protein adsorption studied using QCM(Freqency shift Hz -ve)(30.5)

(22.1) (72.5)

(b)

Page 96: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 5 - Results

86 | P a g e

adsorbed after exposure to 1 mg/ml protein solution for 24 hrs on various PEG surfaces, N%

on the y-axis which is directly related to amount of protein adsorbed, while the y-axis on

Figure 3b shows the change in resonance frequency (-ve) of QCM crystals upon protein

adsorption (1 mg/ml, 750 μl, flow rate 100 μl/minute).

The QCM results show that the surface grafting of PEG thiol using ethanol and SFC

results in overlayers that are equally efficient in resisting protein adsorption as shown in

Figure 3b. It must be noted that the QCM studies were using a total volume of 750 μL of 1

mg/ml protein solution over a period of 7.5 minutes; whereas the XPS quantification studies

were done on gold slides exposed to 1 mg/ml protein solutions over a period of 24h, thus the

results of QCM and XPS cannot be directly correlated. The XPS results show that SFC based

grafting is better at resisting protein adsorption owing to a higher PEG layer thickness and

higher proportion of bound sulfur determined from S 2p spectra. Thus based on chemical

characterization of PEG thiol monolayers via XPS and protein adsorption studies via QCM, it

is preliminary concluded that scCO2 based PEG grafting results in a monolayer that is very

similar to those produced by conventional liquid solvents such as ethanol. The QCM based

short term protein adsorption studies showed no difference between PEG SAMs grafted using

scCO2 and ethanol. The quantification of 24 h protein adsorption studies showed that PEG

SAM grafted using scCO2 has better resistance to protein adsorption than the SAM grafted

using ethanol.

B. Grafting studies in liquids

The effect of polymer molecular weight on the graft density of SAMs was discussed in

section 1d of Chapter 1. There is an inverse relationship between graft density and molecular

weight of polymer molecules. Graft density can also be varied by altering the solubility of

polymer molecules in the grafting solvent. The solubilities of PEG in ethanol, toluene and

water vary in the following order; water > toluene > ethanol. This order was deduced

experimentally by comparing the sonication time required for dissolving 2 kDa PEG thiol in the

three solvents at room temperature. Thus, by mixing ethanol with water or toluene, it is

possible to systematically vary the solubility of PEG and thus the graft density. The objective

was to compare the effect of solubility of PEG thiols on the surface graft density of SAMs

Page 97: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 5 - Results

87 | P a g e

formed from liquid and scCO2 solvents. Grafting studies in liquid were done on gold slides

using various mixing ratios of ethanol and water/toluene. The overlayer thicknesses were

determined using XPS and ellipsometry as described in the methods section. The results of

studies using 2 kDa, 5 kDa and 10 kDa PEG thiol are shown in Figures 4a, 4b and 4c,

respectively. It was observed that increasing the solubility of PEG by adding water to ethanol

results in a decrease in the overlayer thickness, as the proportion of added water increases.

For example the thickness of 2 kDa PEG thiol monolayer grafted using 75 % ethanol is half of

what was obtained from 100 % ethanol as shown in Figure 4a. The reason for difference in

values between XPS and ellipsometry will be dealt with in the discussion section. The XPS

and ellipsometry measurements showed similar trends of changing overlayer thickness with

varying solubility. All the thickness comparisons in this section (III b) are discussed based on

ellipsometry results for the sake of simplicity. For 100% ethanol, it was observed that PEG

thiol did not readily dissolve in the solvent, so the solutions had to be sonicated for 30

minutes.

Figure 4a: Overlayer thicknesses of 2 kDa PEG thiol SAM grafted using mixtures of

ethanol(EtOH)/water(H2O) and ethanol(EtOH)/toluene(Tol) (3h/25°C/101 kPa), measured

using ellipsometry and XPS.

2 3 4 5 6 7 8 9 10 11

Liq-EtOH 75%/25% H2O

Liq-EtOH 95%/5% H2O

Liq-EtOH 100%

Liq-EtOH 97.5%/2.5% Tol

Liq-EtOH 95%/5% Tol

Liq-EtOH 90%/10% Tol

Liq-Tol 100%

Thickness (nm)

2 kDa PEG thiol - Liquid grafting

XPSEllipsometry

(a)

Page 98: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 5 - Results

88 | P a g e

Figure 4b: Overlayer thicknesses of 10 kDa PEG thiol SAM grafted using mixtures of

ethanol(EtOH)/water(H2O) and ethanol(EtOH)/toluene(Tol) (3h/25°C/101 kPa), measured

using ellipsometry and XPS.

Figure 4c: Overlayer thicknesses of 5 kDa PEG thiol SAM grafted using mixtures of

ethanol (EtOH)/water (H2O) and toluene (Tol) (3h/25°C/101 kPa), measured using

ellipsometry and XPS.

1 2 3 4 5 6 7

Liq-Tol 100%

Liq-EtOH 75%/25% Tol

Liq-EtOH 95%/5% Tol

Liq-EtOH 97.5%/2.5% Tol

Liq-EtOH 99%/1% Tol

Liq-EtOH 100%

Liq-H2O 100%

Liq-EtOH 75%/25% H2O

Liq-EtOH 95%/5% H2O

Liq-EtOH 97.5%/2.5% H2O

Liq-EtOH 99%/1% H2O

Liq-EtOH 100%

Thickness (nm)

10 kDa PEG thiol - Liquid grafting

XPSEllipsometry

(b)

2 4 6 8 10 12 14

Liq-EtOH 75%/25% H2O

Liq-EtOH 100%

Liq-Tol 100%

Thickness (nm)

5 kDa PEG thiol - Liquid grafting

XPSEllipsometry

(c)

Page 99: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 5 - Results

89 | P a g e

The 10 kDa PEG thiol solution in 100 % ethanol continued to remain cloudy in spite of

the prolonged sonication, while the 2 kDa and 5 kDa PEG temporarily dissolved during the

sonication but rapidly became cloudy in 10-20 min once the sonication step was stopped. The

10 kDa PEG thiol solutions prepared using ethanol/toluene mixtures with 97.5 % or lower

percentage of ethanol formed clear solutions upon sonication for 30 minutes. This suggests

that grafting in 100 % ethanol occurs at cloud point conditions very similar to that reported by

Kingshott et al. using high ionic strength solutions [41]. This alternative way of cloud point

grafting based PEG thiol functionalization using ethanol can be extended to reactions other

than Au-thiolate formation provided the reactants are soluble in and stable towards ethanol.

Since toluene is also an important solvent in polymer grafting, studies were performed

on the effect of solubility of PEG in ethanol-toluene mixtures on the graft density of PEG thiol

layers. The solubility of PEG thiol in toluene was lowered by the addition of ethanol. The

results of overlayer thickness of 2 kDa and 10 kDa PEG thiol molecules grafted with various

proportions of toluene-ethanol mixtures are given in Figure 4a and 4b, respectively. With

increasing percentage of ethanol the thickness of overlayer increases. The thickness of 10

kDa PEG thiol monolayer grafted using 25% toluene solution results in a overlayer with a

thickness of 1.8 nm, where as the thickness is 4.4 nm when 1% toluene is used, as shown in

Figure 4b. Thus it could be concluded from the liquid based PEG thiol grafting study that

decrease in solubility of PEG results in higher graft densities due to decreased excluded

volume effect. The effect of Mw of PEG thiol on the graft density is clearly evident upon

comparing the thicknesses of SAMs of 2 kDa, 5 kDa and 10 kDa PEG thiols grafted using

100% ethanol, which are 7.6nm, 10.9nm and 5.5nm respectively. Thus the order of the

thicknesses are 5 kDa>2 kDa>10 kDa.

Since the 5 kDa PEG thiol formed SAM of greatest thickness as seen from the results

presented so far in this chapter, comparative studies in scCO2 mainly focused on 5 kDa PEG

apart from the preliminary studies using 2 kDa PEG thiol. It should be noted that in this

section, there was no comparison of percentages of bound sulphur unlike the previous

preliminary studies section. Since the PEG thiol SAM has the sulfur atom below the layer of

PEG, the S photoelectron signal attenuation would increase with increase in PEG thiol SAM

Page 100: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 5 - Results

90 | P a g e

thickness. The S signal became extremely low for samples that had thicknesses >7-8nm,

peak fitting of the high resolution S 2p spectrum into bound and unbound thiol species was

thus difficult due very low signal intensity. To ensure that a significant proportion of PEG thiol

molecules in the SAM are chemisorbed, the samples with PEG thiol SAM thickness of >7-

8nm were sonicated for 30 minutes followed by XPS and ellipsometry measurements.

Sonication is known to cause desorption of loosely physisorbed molecules from SAMs. The

sonication step did not result in any significant decrease in thickness (data not shown), thus

confirming that these thicker SAMs have an optimal percentage of covalently bound PEG.

C. Grafting studies in scCO2

As mentioned the solubility of a polymer can also be manipulated in SFC by altering

the pressure-temperature conditions. Grafting 2 kDa and 5 kDa PEG thiols on gold was done

in scCO2 as described in section II b. The thickness of the 2 kDa PEG thiol SAM determined

from ellipsometry increased from 3 nm to 4.9 nm when the temperature of scCO2 was

changed with constant pressure, from 20 MPa/60°C to 20 MPa/50°C. This increase in the

thickness (Figure 5a) can be attributed to decrease in PEG solubility with decreasing

temperature as described by Kirby et al. [211]. The degree of increase in the thickness was

not significant when both pressure and temperature were decreased from 20 MPa/60°C to 15

MPa/50°C, and this could be due to extremely low solubility. The problem of low solubility

becomes apparent from experiments with 5 kDa PEG thiol (Figure 5b), in which case a lower

thickness of 2.2 nm was obtained at 20 MPa/50°C in comparison to 2 kDa PEG thiol SAM

with a thickness of 4.9 nm. This decrease in thickness with increase in Mw is in agreement

with de Gennes theory discussed in Chapter 1. Furthermore it can be seen that upon a

decrease in temperature or pressure of the scCO2, the thickness of the 5 kDa PEG thiol SAM

does not change significantly, unlike in the case of the 2 kDa PEG thiol where the thickness

increases. The low solubility issue with the 5 kDa PEG thiol or higher Mw PEG thiols could be

overcome by using higher temperature-pressure conditions where the density of scCO2 would

be higher. Future experiments will need to investigate scCO2 based 5 and 10 kDa PEG thiol

grafting using pressure-temperature conditions higher than those reported in this section.

Apart from using higher pressure-temperature conditions, one can also think of using co-

Page 101: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 5 - Results

91 | P a g e

solvents along with scCO2 to help improve the partition coefficients of high MW PEG thiol

molecules. The results of studies using co-solvents are discussed in the next section.

Figure 5a: Overlayer thicknesses of 2 kDa PEG thiol SAM grafted using scCO2, at

varying pressure and temperature, measured using ellipsometry and XPS.

Figure 5b: Overlayer thicknesses of 5 kDa PEG thiol SAM grafted using scCO2, at

varying pressure and temperature, measured using ellipsometry and XPS.

D. Grafting studies in scCO2–co­solvent mixtures

As mentioned in the previous section, the solubility issues with high MW PEG thiol

molecules in scCO2 can be solved using co-solvents. In the literature, ethanol, toluene and

methanol have been shown to increase the partition coefficients of polar molecules in scCO2

[211]. The objective is to achieve optimal solubility and then gradually decrease the pressure

or temperature to achieve grafting close to cloud point conditions. The main advantage of

using co-solvents is that they will enable cloud point grafting at lower pressure-temperature

2 3 4 5 6 7 8

SFC-20 MPa, 60 C

SFC-20 MPa, 50 C

SFC-15 MPa, 50 C

Thickness (nm)

2 kDa PEG thiol - scCO2

XPSEllipsometry

(a)

1 2 3 4 5

SFC-20 MPa, 60 C

SFC-15 MPa, 60 C

SFC-20 MPa, 50 C

Thickness (nm)

5 kDa PEG thiol - scCO2

XPSEllipsometry

(b)

Page 102: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 5 – Results

92 | P a g e

conditions for high MW polymers, which would be a significant cost benefit in large scale

industrial applications. Figure 6b summarizes the results of 5 kDa PEG thiol SAMs grafted

using scCO2–5% co-solvent mixtures as described in section II b. In this section the thickness

comparisons are made using XPS measurements. The effect of addition of ethanol, methanol

and toluene was studied. Ethanol-scCO2 at 20 MPa/60°C produced a SAM thickness (3.9 nm)

that was slightly lower than that obtained using scCO2 at 20 MPa/60°C (4.2 nm), but upon a

decrease in temperature, the ethanol-scCO2 (20 MPa/50°C) produced a thicker SAM (5.0

nm). The results of grafting experiments using 5 kDa PEG thiol with toluene-scCO2 are shown

in Figure 6b. Upon lowering of temperature the toluene-scCO2 resulted in a SAM with lower

thickness of 4.9 nm (20 MPa/50°C) when compared to 6.0 nm obtained using 20 MPa/60°C.

The three solvents selected for this study were an attempt to understand the relationship

between the polarity of solvent and graft density of PEG thiol SAM obtained. Based on the

data obtained from XPS and ellipsometry, there is no straightforward relationship between

solvent polarity and thickness. For example the increasing order of thickness obtained at 20

MPa/60°C for the three co-solvents is ethanol<toluene<methanol, whereas the increasing

order of polarity is toluene<methanol<ethanol. The increasing order of solubility of PEG in

liquids of co-solvents under consideration is ethanol<toluene<methanol [223], which is exactly

the order obtained for thicknesses with scCO2-co-solvents. Thus, the solubility of PEG in the

solvents seems to play a significant role in deciding the overlayer thickness obtained in SFC-

co-solvent mixtures. Based on this co-solvent study, it is conclude that the cloud point

conditions can be lowered for high MW PEG thiol molecules thus enabling us to achieve

higher graft densities under economical conditions. The scCO2-co-solvent studies for 2 and

10 kDa PEG thiol will be made in a future project. A detailed analysis of PEG SAM

thicknesses obtained for PEGs of different Mw under varying conditions of pressure-

temperature and co-solvents would enable better understanding of polymeric self assembly

processes in scCO2.

Page 103: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 5 – Results

93 | P a g e

Figure 6a: Overlayer thicknesses of 2 kDa PEG thiol SAM grafted using scCO2 with 5

% co-solvent ethanol/toluene, at varying temperature, measured using ellipsometry and XPS.

Figure 6b: Overlayer thicknesses of 5 kDa PEG thiol SAM grafted using

scCO2 with 5 % co-solvent ethanol/toluene/methanol, at varying temperature,

measured using ellipsometry and XPS. ’∗’ denotes ’data not available’

2 4 6 8

SFC-20 MPa, 60 C/EtOH 5%

SFC-20 MPa, 50 C/EtOH 5%

SFC-20 MPa, 60 C/Tol 5%

SFC-20 MPa, 50 C/Tol 5%

Thickness (nm)

2 kDa PEG thiol - scCO2 - cosolvent

XPSEllipsometry

(a)

0 1 2 3 4 5 6 7

SFC-20 MPa, 60 C / EtOH 5%

SFC-20 MPa, 50 C / EtOH 5%

SFC-20 MPa, 60 C / Tol 5%

SFC-20 MPa, 50 C / Tol 5%

SFC-20 MPa, 60 C / MeOH 5%

SFC-20 MPa, 50 C / MeOH 5%

Thickness (nm)

5 kDa PEG thiol - scCO2 - cosolvent

XPSEllipsometry

(b)

***

*

Page 104: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 5 – Discussion and conclusion

94 | P a g e

IV. Discussion and Conclusion

The cloud point grafting procedure reported by Kingshott et al. [41] using high

ionic strength is very efficient, but cannot be applied to grafting reactions sensitive to water.

Examples of types of reactions used in polymer grafting procedures were mentioned in

Chapter 1. Thus, development of a simple protocol applicable to all reaction sequences is of a

significant interest. Here it was demonstrated that scCO2 is a very efficient solvent for grafting

PEG thiol molecules, and since CO2 is chemically inert towards most functional groups, it will

not interfere with most industrially relevant reactions such as polymer functionalization and

surface modification. From the studies presented in this chapter it can be concluded that SFC

based grafting under the conditions used here produced PEG thiol SAMs of moderate graft

density. For example with 2 kDa PEG thiols, the highest graft density of 1.9 chains/nm2 was

produced using a SFC-toluene combination. Tokumitsu et al. [165] reported a maximum graft

density of 3.6 chains/nm2 using 2 kDa PEG conjugated to undecane thiols self assembled on

gold substrates. Dalsin et al. [166] and Pasche et al. [167] reported graft densities > 3

chains/nm2 using SAMs of DOPA functionalized PEG and PLL-g-PEG respectively. Cloud

point grafting is thus a very efficient technique to produce SAMs of high graft density. Since

the SFC based grafting technique is in its initial stages, further research needs to done to

appreciate the potentials of this simple versatile technique.

It should be noted that for all the thickness results the values obtained using XPS are

consistently higher than those calculated from ellipsometry. In all the overlayer thickness

estimations using XPS, an attenuation length (λ) of 3.9 nm for Au 4f photoelectrons (1404.6

eV K.E) through the PEG overlayer was used in all our calculations. The attenuation length

value was based on calculations suggested by Cumpson et al. [224]. The value of λ used in

this work is close to those reported by Laibinis et al. [225] and Sofia et al. [110]. An alternate

way of calculating thickness using XPS involves comparing the signal from a infinitely thick

layer of PEG. As an example the thickness of PEG SAM of 5kDa PEG thiol grafted using SFC

(SFC 60°C/20Mpa/5% EtOH) was calculated using a λ value of 2.9 nm for the O1s

photoelectron [224], assuming Oxygen atomic percentage of 33.3% for an infinitely thick layer

of PEG. This method of calculation gave a thickness value of 2.7 nm, while the other method

Page 105: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 5 – Discussion and conclusion

95 | P a g e

based on attenuation of gold signal gave a thickness vale of 3.9 nm. The calculation based on

O% signal gave values much closer to the value estimated by ellipsometry 1.7 nm. Thus

calculation using O signal seems more appropriate way of estimating the thickness instead of

using attenuation of gold substrate signal. All these methods of calculating λ assume a PEG

density of 1 gm/cm3. Unsworth et al. [150] reported a PEG density of 0.8 gm/cm3 and 1.0

gm/cm3 using neutron reflectometry for 5 and 2 kDa PEG thiol SAMs grafted under cloud

point conditions. Hence the PEG density could be different depending on the Mw and grafting

conditions. Piehler et al. used a dry density of 1.2 gm/cm3 [226]. Since the value of λ depends

on PEG density (ρ) [225], a wrong assumption of the ρ value would lead to an erroneous

estimation of thickness. Using the equation given by Laibinis et al. a difference in density of

0.2 gm/cm3 could introduce approximately 5% error [225]. In the ellipsometry calculations, the

refractive index of PEG overlayers was assumed to be 1.47 as reported by Unsworth et al.

based on calculations by Tokumitsu et al. [165] (PEG is assumed to be a Cauchy layer with

the two lowest Cauchy parameters An = 1.45 and Bn = 0.01). The refractive index values used

for ellipsometric estimation of thickness in other reports on PEG brushes are very close the

value used in this Chapter. Ostaci et al. used a refractive index value of 1.45 [227], Piehler et

al. used 1.5 [226] and Mehne et al. performed their calculations using 1.5 [228]. The error in

refractive index values in the range of 0.05 causes a error in the calculated thickness by only

few Å [125]. Although the absolute values obtained using XPS and ellipsometry were

different, the trends in thickness variations under different experimental conditions hold good

for both techniques.

Based on preliminary studies it was concluded that PEG thiol grafting using scCO2

results in SAMs that are similar to those formed using conventional solvents such as ethanol.

It was further demonstrated that it is possible to perform ‘cloud point’ type grafting by

modifying the pressure-temperature conditions of SFC. By comparing results of scCO2 based

studies using 2 and 5 kDa PEG thiol molecules, it could be speculated that cloud point

grafting of higher MW PEG molecules might need higher pressure-temperature conditions

when compared to that required by low MW PEG molecules. This requirement for higher

pressure-temperature can be overcome by using scCO2/co-solvent mixtures as demonstrated

from studies on 5 kDa PEG thiol SAMs grafted using methanol and toluene as co-solvents.

Page 106: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 5 – Future directions

96 | P a g e

V. Future directions

The high wettability of SFC makes it an efficient solvent for the functionalization of

extremely rough surfaces, where it is tough for conventional liquids to perform reactions due

to their high surface tension. Comparisons of the PEGs grafted on rough surfaces (gold

coated sand blasted steel) using SFC and conventional liquids will be made in a future

project. The studies with scCO2-cosolvent mixtures using 10 kDa PEG thiols are also part of a

future project. Comparison of results from SFC based studies of 2 kDa, 5 kDa and 10 kDa

PEG thiol SAMs would enable an understanding of the strengths and advantages of using

SFC as a grafting solvent.

Page 107: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER 6 – Thesis summary

97 | P a g e

CHAPTER 6: Thesis summary

The work presented in this thesis mainly focused on investigating novel ‘grafting to’

strategies for self assembly of poly(ethylene glycol) monolayers intended to serve as

antifouling surfaces. Extensive studies were carried out using surface sensitive techniques

including x-ray photoelectron spectroscopy, ellipsometry, contact angle measurements and

atomic force microscopy. Once the surfaces were characterized, their antifouling abilities

were assessed either by single protein adsorption studies or serum adsorption study and also

bacterial attachment assays.

The graft density of 2kDa PEG SAM can be increased by backfilling the monolayer

using shorter oligo(ethylene glycol) molecules. This strategy was systematically studied using

carboxyl capped PEG-COOH thiols and hydroxy-terminated tri(ethylene glycol) undecanethiol

(OEG). The backfilling process was found to result in the formation of homogeneous mixed

monolayers rather than islands of phase separated PEG-COOH and OEG thiol molecules.

The XPS and contact angle based quantitative assessment of desorption of PEG-COOH thiol

molecules from the PEG-COOH SAM during the backfilling process indicated that there was

minimum desorption during addition of OEG thiol whereas an alkanethiol caused significant

desorption. The QCM and XPS based serum adsorption studies using 10% FBS showed that

backfilled PEG-COOH SAM was better than non backfilled PEG-COOH SAM at resisting non

specific adsorption. Bacterial attachment studies using Staphylococcus aureus demonstrated

the greater ability of backfilled surface to resist initial attachment of cells when compared to

non backfilled surfaces. The OEG SAM was better than backfilled PEG-COOH SAMs in

resisting both serum adsorption and bacterial attachment. The antifouling properties of PEG-

COOH SAM could be even lower in the absence of carboxyl tail group which contributes a

negative charge. The presence of carboxyl groups enables the immobilization of bioactive

molecules such as enzymes and with backfilled OEG thiol molecules, the PEG-COOH SAM

would have maximum activity and minimal non specific adsorption.

The next strategy involved the use of PEG-COOH SAM immobilized with subtilisin a

Page 108: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER – Thesis summary

98 | P a g e

known antifouling enzyme as non-fouling surface. The covalent immobilization was carried

out using an NHS-EDC catalyzed reaction between amine residues of enzyme and the

carboxylic group of PEG SAM resulting in amide bond formation. Subtilisin was immobilized

on mixed OEG and OEG-COOH SAMs created by co-adsorption from solutions containing

varying proportions of the two different thiol molecules. The quantification of immobilized

subtilisin was done based on the N atomic percentage and the activity was measured using a

colorimetric assay. The surface concentration of enzyme increased with an increase in

surface concentration of carboxyl groups. It was found that a large proportion of immobilized

subtilisin molecules were inactive and there was no linear relationship between the surface

enzyme concentration and enzyme activity. The antifouling ability of the enzyme subtilisin

immobilized on co-adsorbed OEG-COOH/OEG surfaces was quantified using bacterial

attachment assays with Staphylococcus Xylosus and it was observed that there was no

correlation between the amount of enzyme on surface and the number of bacterial cells

adhering onto it. Immobilization studies using active and denatured subtilisin molecules on

PEG-COOH surface backfilled with OEG thiol confirmed the ability of active enzymes to

minimize adhesion of Staphylococcus aureus. There was no quantitative difference in the

amount of enzyme immobilized on the backfilled PEG-COOH SAM and the non backfilled

SAM, which did not agree with the serum adsorption studies on the same surfaces. The main

reason behind the discrepancies with regards to subtilisin immobilized surfaces could be due

to the autolysis of enzyme molecules upon immobilization at high coverage. The enzyme

molecules could interfere with the activity of neighboring molecules when located at close

proximity. Immobilization studies using lysozyme and BSA showed that PEG-COOH SAM

after backfilling with OEG had minimum non-specifically adsorbed protein. Control

experiments without the NHS-EDC activating agent demonstrated that OEG backfilled PEH-

COOH SAMs are capable of efficiently resisting non-specific adsorption and hence most of

the attached protein is covalently coupled.

The third strategy investigated involved use of supercritical carbon dioxide to graft PEG

thiol molecules under solubility conditions close to ‘cloud point’, thereby producing SAMs of

high graft density due to minimized exclusion volume effects. The thickness of SAMs formed

in SFC and conventional liquid solvents such as ethanol, ethanol and toluene were compared

Page 109: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

CHAPTER – Thesis summary

99 | P a g e

using XPS and ellipsometry. The kinetics studies of SAM formation by 2 kDa PEG thiol in

SFC and 75% ethanol indicated that the SAMs with maximum thickness and bound sulfur

percentage were formed within 30 minutes in SFC, whereas 75 % ethanol required 3-6 h.

QCM based adsorption studies of BSA, Lyz and casein demonstrated that both SFC and

ethanol based SAMs were equally good at resisting protein adsorption. Longer studies

involving protein exposure for 24h and quantifications using XPS showed that SFC based

SAMs were better at resisting protein adsorption. It was also demonstrated that ‘cloud point’

type grafting can be performed in SFC by modifying the pressure-temperature conditions. By

comparing results of scCO2 based studies using 2 and 5 kDa PEG thiol molecules, it can be

speculated that ‘cloud point’ grafting of higher MW PEG molecules might need higher

pressure-temperature conditions when compared to that required by low MW PEG molecules.

This requirement for higher pressure-temperature can be overcome by using scCO2/co-

solvent mixtures as demonstrated from studies on 5 kDa PEG thiol SAMs grafted using

methanol and toluene as co-solvents. Thus, this work demonstrated that SFC has the

potential to be suitable solvent for grafting non-fouling polymers and when combined with

suitable co-solvents, it is capable of serving as a versatile solvent to carryout surface

modifications relevant to the designing of successful material surfaces that control the

interactions with biological environments.

Page 110: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

100 | P a g e

Acknowledgement

First and foremost, I would like to express my sincere gratitude to my thesis supervisor,

Prof. Peter kingshott, for giving me an opportunity to pursue my PhD in the exciting field of

biointerfaces. It was with his constant support and encouragement my thesis grew up to a

complete one. I owe a huge debt of gratitude to my co-supervisor, Dr. Rikke Louise Meyer,

whose systematic and meticulous approach towards research and science in general helped

me learn a lot and improve myself. I am also eternally grateful to Dr. Ryosuke Ogaki, who

provided invaluable assistance and advice on experiments, writing and planning. Without

Ryo’s timely help I would not be possible for me to have finished the thesis and an accepted

article. Thanks Ryo. I also wish to thank Dr. Martin Cole for the insightful discussions and

advices. Thanks to Viduthalai’s long interesting and thought provoking discussion, my

knowledge on biofilms has improved a lot. I also thank him for the important bacterial

attachment experiments that were conducted as part of the work presented in this thesis.

Constant encouragement and support from Prof. Flemming Besenbacher during his busy

schedule helped in the smoother progress in the scientific and administrative processes

during to my PhD. I thank him for creating such wonderful research environment and useful

interactions during the iNANO Autumn School sessions.

The research activities undertaken by would not be possible without the availability of

financial support. I thank the Faculty of Science, AU, Carlsberg foundation for the financial

support. I also thank all the administrative staff of iNANO and Aarhus graduate school of

science, for their kind help.

I would like to thank the following people for sharing their knowledge, giving useful

feedback, suggestions and sicnetific discussions, during my PhD ; Prof.Dunccan, Dr.

Mingdong, Dr.Peter Hald, Dr. Claus Bischoff, Dr. Helmer Sohoel, Dr. Jing Wang, Dr. Jenny

Malmstrom, Dr. Maria Sund, Dr. Esben Unmack Larsen, Dr. Vladimir, Dr.Gurvinder Singh,

Dr.Manjunath Puttaswami, Yuya Hayashi, Uffe Jensen, Maj Frederiksen, Stine Kristensen.

My sincere thanks are to Jacques Chevallier and Folmer Lyckegaard for sharing his valuable

technical knowledge and for their technical support. The biointerface group of iNANO or the

‘4th floor lab’ was indeed an awesome place to perform experiments and conduct interesting

Page 111: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

Acknowledgement

101 | P a g e

research. I wish to thank all members of this group for supporting, cooperating and

coordinating with me during my experiments.

I thank my parents and brother for their constant support and love. Without their support

and cooperation my dream to do PhD would have remained unfulfilled. I also wish to thank Sri

Sathya Sai Baba for his love and teachings. I am blessed to have met friends who have been

more than just friends; they supported me during the turbulent times. I thank Shyamal,

Ganesh, Sashi kiran, Vikas, Vivek Raj, Manjula, Vini Gautham, Ramesh Subramani, Ramesh

Bhai, Phani kumar, Chandini, Shirin, Vijay, Indu, Umadevi.

Having covered the acknowledgements in the real world, I cannot miss out on the

virtual world that has become an important part of my professional and personal life. I thank

Microsoft, Google, Thomas Reuters for developing software that greatly helped me simplify

the scientific writing, search and bibliography. Also wish to thank Google, Facebook, and

LinkedIn which kept me connected and at times share exciting scientific information with all

my friends, colleagues spread across the world.

Page 112: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

Bibliography

102 | P a g e

Bibliography

1. Briggs D, Grant JT: Surface analysis by Auger and x‐ray photoelectron spectroscopy: IM Publications; 2003.

2. Ratner BD, Castner DG: Electron Spectroscopy for Chemical Analysis. In: Surface Analysis – The Principal Techniques. John Wiley & Sons, Ltd; 2009: 47‐112.

3. Castner DG, Ratner BD: Biomedical surface science: Foundations to frontiers. Surface Science 2002, 500(1‐3):28‐60.

4. Banerjee I, Pangule RC, Kane RS: Antifouling Coatings: Recent Developments in the Design of Surfaces That Prevent Fouling by Proteins, Bacteria, and Marine Organisms. Advanced Materials 2010, 23(6):690‐718.

5. Costerton JW, Stewart PS, Greenberg EP: Bacterial Biofilms: A Common Cause of Persistent Infections. Science 1999, 284(5418):1318‐1322.

6. Parsek MR, Singh PK: BACTERIAL BIOFILMS: An Emerging Link to Disease Pathogenesis. Annu Rev Microbiol 2003, 57(1):677‐701.

7. S. M. September FAE, S. N. Venter and V. S. Bro¨ zel: Prevalence of bacterial pathogens in biofilms of drinking water distribution systems. Journal of Water and Health 2007, 5(2):219‐227.

8. C. WHITTAKER HR, AND B. H. OLSON: Evaluation of Cleaning Strategies for Removal of Biofilms from Reverse‐Osmosis Membranes. Applied and Environmental Microbilogy 1984, 48(3):395‐403.

9. Kumar CG, Anand SK: Significance of microbial biofilms in food industry: a review. International Journal of Food Microbiology 1998, 42(1‐2):9‐27.

10. Van Houdt R, Michiels CW: Biofilm formation and the food industry, a focus on the bacterial outer surface. Journal of Applied Microbiology 2010, 109(4):1117‐1131.

11. Dafforn KA, Lewis JA, Johnston EL: Antifouling strategies: History and regulation, ecological impacts and mitigation. Marine Pollution Bulletin 2011, 62(3):453‐465.

12. Buddy D. Ratner ASH, Frederick J. Schoen, Jack E.Lemon: Biomaterials Science: An introduction to materials in medicine, Elsevier 2nd Edition.

13. Bruin P, Meeuwsen EAJ, van Andel MV, Worst JGF, Pennings AJ: Autoclavable highly cross‐linked polyurethane networks in ophthalmology. Biomaterials 1993, 14(14):1089‐1097.

14. K.Lee DK (ed.): Tissue Engineering I ‐ Scaffold Systems for Tissue Engineering: Springer; 2006. 15. Le Guéhennec L, Soueidan A, Layrolle P, Amouriq Y: Surface treatments of titanium dental implants

for rapid osseointegration. Dental materials : official publication of the Academy of Dental Materials 2007, 23(7):844‐854.

16. Desai TA: Micro‐ and nanoscale structures for tissue engineering constructs. Medical Engineering & Physics 2000, 22(9):595‐606.

17. Scardino AJ, Zhang H, Cookson DJ, Lamb RN, Nys Rd: The role of nano‐roughness in antifouling. In., vol. 25: Taylor & Francis; 2009: 757 ‐ 767.

18. Flemming HC: Biofouling in water systems – cases, causes and countermeasures. Applied Microbiology and Biotechnology 2002, 59(6):629‐640.

19. Rosenhahn A, Schilp S, Kreuzer HJ, Grunze M: The role of "inert" surface chemistry in marine biofouling prevention. Physical Chemistry Chemical Physics 2010, 12(17):4275‐4286.

20. Whitchurch CB, Tolker‐Nielsen T, Ragas PC, Mattick JS: Extracellular DNA Required for Bacterial Biofilm Formation. In., vol. 295: Science; 2002: 1487.

21. Miller MB, Bassler BL: Quorum sensing in bacteria. Annu Rev Microbiol 2001, 55:165‐199.

Page 113: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

Bibliography

103 | P a g e

22. George M, Pierce G, Gabriel M, Morris C, Ahearn D: Effects of Quorum Sensing Molecules of Pseudomonas aeruginosa on Organism Growth, Elastase B Production, and Primary Adhesion to Hydrogel Contact Lenses. Eye & Contact Lens 2005, 31(2):54‐61.

23. Kingshott P, Griesser HJ: Surfaces that resist bioadhesion. Current Opinion in Solid State & Materials Science 1999, 4(4):403‐412.

24. Venkat K. Vendra LW, and Sitaraman Krishnan (ed.): Nanomaterials for the Life Sciences : Nanostructured Thin Films and Surfaces

WILEY‐VCH Verlag GmbH & Co. KGaA, Weinheim; 2010. 25. Bongaerts JHH, Cooper‐White JJ, Stokes JR: Low Biofouling Chitosan‐Hyaluronic Acid Multilayers with

Ultra‐Low Friction Coefficients. Biomacromolecules 2009, 10(5):1287‐1294. 26. Luk Y‐Y, Kato M, Mrksich M: Self‐Assembled Monolayers of Alkanethiolates Presenting Mannitol

Groups Are Inert to Protein Adsorption and Cell Attachment. Langmuir 2000, 16(24):9604‐9608. 27. Buddy Ratner AH, Frederick Schoen, Jack Lemons (ed.): Biomaterials Science. An Introduction to

Materials in Medicine. San Diego, USA: Acamedic press; 1996. 28. Callow JA, Callow ME: Trends in the development of environmentally friendly fouling‐resistant

marine coatings. Nat Commun 2011, 2:244. 29. Nakanishi J, Takarada T, Yamaguchi K, Maeda M: Recent Advances in Cell Micropatterning Techniques

for Bioanalytical and Biomedical Sciences. Analytical Sciences 2008, 24(1):67‐72. 30. Perrino C, Lee S, Choi SW, Maruyama A, Spencer ND: A Biomimetic Alternative to Poly(ethylene

glycol) as an Antifouling Coating: Resistance to Nonspecific Protein Adsorption of Poly(l‐lysine)‐graft‐dextran. Langmuir 2008, 24(16):8850‐8856.

31. Zhang Z, Chao T, Chen S, Jiang S: Superlow Fouling Sulfobetaine and Carboxybetaine Polymers on Glass Slides. Langmuir 2006, 22(24):10072‐10077.

32. Chen S, Zheng J, Li L, Jiang S: Strong Resistance of Phosphorylcholine Self‐Assembled Monolayers to Protein Adsorption: Insights into Nonfouling Properties of Zwitterionic Materials. Journal of the American Chemical Society 2005, 127(41):14473‐14478.

33. Torres C, Lenon G, Craperi D, Wilting R, Blanco Á: Enzymatic treatment for preventing biofilm formation in the paper industry. Applied Microbiology and Biotechnology 2011:1‐9.

34. Olsen SM, Pedersen LT, Laursen MH, Kiil S, Dam‐Johansen K: Enzyme‐based antifouling coatings: a review. In., vol. 23: Taylor & Francis; 2007: 369 ‐ 383.

35. Meyer B: Approaches to prevention, removal and killing of biofilms. International Biodeterioration & Biodegradation 2003, 51(4):249‐253.

36. Grad S, Kupcsik L, Gorna K, Gogolewski S, Alini M: The use of biodegradable polyurethane scaffolds for cartilage tissue engineering: potential and limitations. Biomaterials 2003, 24(28):5163‐5171.

37. Duracher D, Veyret R, Elaïssari A, Pichot C: Adsorption of bovine serum albumin protein onto amino‐containing thermosensitive core‐shell latexes. Polymer International 2004, 53(5):618‐626.

38. Futamura K, Matsuno R, Konno T, Takai M, Ishihara K: Rapid Development of Hydrophilicity and Protein Adsorption Resistance by Polymer Surfaces Bearing Phosphorylcholine and Naphthalene Groups. Langmuir 2008, 24(18):10340‐10344.

39. Yamasaki A, Imamura Y, Kurita K, Iwasaki Y, Nakabayashi N, Ishihara K: Surface mobility of polymers having phosphorylcholine groups connected with various bridging units and their protein adsorption‐resistance properties. Colloids and Surfaces B: Biointerfaces 2003, 28(1):53‐62.

40. Yi Chang Chung YHC: Protein Adsorption and Cell Alignment on Micropatterned Phosphorylcholine Surfaces. Journal of Medical and Biomedical Engineering 2009, 29(6):320‐324.

41. Kingshott P, Thissen H, Griesser HJ: Effects of cloud‐point grafting, chain length, and density of PEG layers on competitive adsorption of ocular proteins. Biomaterials 2002, 23(9):2043‐2056.

Page 114: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

Bibliography

104 | P a g e

42. Nejadnik MR, van der Mei HC, Norde W, Busscher HJ: Bacterial adhesion and growth on a polymer brush‐coating. Biomaterials 2008, 29(30):4117‐4121.

43. Bridges AW, Singh N, Burns KL, Babensee JE, Andrew Lyon L, García AJ: Reduced acute inflammatory responses to microgel conformal coatings. Biomaterials 2008, 29(35):4605‐4615.

44. De Gennes PG: Polymers at an interface; a simplified view. Advances in Colloid and Interface Science 1987, 27(3‐4):189‐209.

45. Jeon SI, Lee JH, Andrade JD, De Gennes PG: Protein‐surface interactions in the presence of polyethylene oxide: I. Simplified theory. Journal of Colloid and Interface Science 1991, 142(1):149‐158.

46. Harder P, Grunze M, Dahint R, Whitesides GM, Laibinis PE: Molecular conformation in oligo(ethylene glycol)‐terminated self‐assembled monolayers on gold and silver surfaces determines their ability to resist protein adsorption. Journal of Physical Chemistry B 1998, 102(2):426‐436.

47. 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(18):5605‐5620.

48. Pertsin AJ, Grunze M: Computer Simulation of Water near the Surface of Oligo(ethylene glycol)‐Terminated Alkanethiol Self‐Assembled Monolayers†Langmuir 2000, 16(23):8829‐8841.

49. Szleifer I: Protein adsorption on tethered polymer layers: effect of polymer chain architecture and composition. Physica A: Statistical and Theoretical Physics 1997, 244(1‐4):370‐388.

50. Szleifer I: Protein Adsorption on Surfaces with Grafted Polymers: A Theoretical Approach. Biophysical Journal 1997, 72(2, Part 1):595‐612.

51. Kristensen JB, Meyer RL, Laursen BS, Shipovskov S, Besenbacher F, Poulsen CH: Antifouling enzymes and the biochemistry of marine settlement. Biotechnology Advances 2008, 26(5):471‐481.

52. Sutherland IW: Biofilm exopolysaccharides: a strong and sticky framework. Microbiology 2001, 147(1):3‐9.

53. Watnick P, Kolter R: Biofilm, City of Microbes. J Bacteriol 2000, 182(10):2675‐2679. 54. Saldarriaga Fernández IC, van der Mei HC, Lochhead MJ, Grainger DW, Busscher HJ: The inhibition of

the adhesion of clinically isolated bacterial strains on multi‐component cross‐linked poly(ethylene glycol)‐based polymer coatings. Biomaterials 2007, 28(28):4105‐4112.

55. Bradford Craigen AD, and Daniel E Kadouri: The Use of Commercially Available Alpha‐Amylase Compounds to Inhibit and Remove Staphylococcus aureus Biofilms. Journal of open Microbiology 2011, 5:10.

56. Singh PK, Parsek MR, Greenberg EP, Welsh MJ: A component of innate immunity prevents bacterial biofilm development. Nature 2002, 417(6888):552‐555.

57. De Morais L, Bernardes‐Filho R, Assis O: Wettability and bacteria attachment evaluation of multilayer proteases films for biosensor application. World Journal of Microbiology and Biotechnology 2009, 25(1):123‐129.

58. Charles A. Haynes WN: Globular proteins at solid/liquid interfaces. Colloids and Surfaces B Biointerfaces 1994, 2:517‐566.

59. Hlady V, Buijs J: Protein adsorption on solid surfaces. Current Opinion in Biotechnology 1996, 7(1):72‐77.

60. Boks NP, Norde W, van der Mei HC, Busscher HJ: Forces involved in bacterial adhesion to hydrophilic and hydrophobic surfaces. In., vol. 154: Microbiology; 2008: 3122‐3133.

61. Leckband D, Sivasankar S: Forces controlling protein interactions: theory and experiment. Colloids and Surfaces B: Biointerfaces 1999, 14(1‐4):83‐97.

62. Ruggiero C, Mantelli M, Curtis A, Rolfe P: Protein‐surface interactions. Cell Biochemistry and Biophysics 2005, 43(3):407‐417.

63. Tadmor R, Hernández‐Zapata E, Chen N, Pincus P, Israelachvili JN: Debye Length and Double‐Layer

Page 115: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

Bibliography

105 | P a g e

Forces in Polyelectrolyte Solutions. Macromolecules 2002, 35(6):2380‐2388. 64. Piret F, Su BL: Effects of pH and ionic strength on the self‐assembly of silica colloids to opaline

photonic structures. Chemical Physics Letters 2008, 457(4‐6):376‐380. 65. Kondo A, Fukuda H: Effects of Adsorption Conditions on Kinetics of Protein Adsorption and

Conformational Changes at Ultrafine Silica Particles. Journal of Colloid and Interface Science 1998, 198(1):34‐41.

66. Santos O, Nylander T, Paulsson M, Trägårdh C: Whey protein adsorption onto steel surfaces‐‐effect of temperature, flow rate, residence time and aggregation. Journal of Food Engineering 2006, 74(4):468‐483.

67. Kasemo B: Biological surface science. Current Opinion in Solid State and Materials Science 1998, 3(5):451‐459.

68. Brash John L, Horbett Thomas A: Proteins at Interfaces. In: Proteins at Interfaces II. vol. 602: American Chemical Society; 1995: 1‐23.

69. Jung S‐Y, Lim S‐M, Albertorio F, Kim G, Gurau MC, Yang RD, Holden MA, Cremer PS: The Vroman Effect: A Molecular Level Description of Fibrinogen Displacement. Journal of the American Chemical Society 2003, 125(42):12782‐12786.

70. Giraud S, Bourbigot S, Rochery M, Vroman I, Tighzert L, Delobel R: Microencapsulation of phosphate: application to flame retarded coated cotton. Polymer Degradation and Stability 2002, 77(2):285‐297.

71. Parkin IP, Palgrave RG: Self‐cleaning coatings. Journal of Materials Chemistry 2005, 15(17):1689‐1695. 72. Kuhr M, Bauer S, Rothhaar U, Wolff D: Coatings on plastics with the PICVD technology. Thin Solid Films

2003, 442(1‐2):107‐116. 73. Kurkuri MD, Driever C, Johnson G, McFarland G, Thissen H, Voelcker NH: Multifunctional Polymer

Coatings for Cell Microarray Applications. Biomacromolecules 2009, 10(5):1163‐1172. 74. Kodnani R, Jaffe D, Jaccodine R: Evaluation of polymeric coatings for MCM applications. Components,

Packaging, and Manufacturing Technology, Part B: Advanced Packaging, IEEE Transactions on 1995, 18(4):658‐665.

75. Hoy RS, Grest GS: Entanglements of an End‐Grafted Polymer Brush in a Polymeric Matrix. Macromolecules 2007, 40(23):8389‐8395.

76. Matrab T, Save M, Charleux B, Pinson J, Cabet‐deliry E, Adenier A, Chehimi MM, Delamar M: Grafting densely‐packed poly(n‐butyl methacrylate) chains from an iron substrate by aryl diazonium surface‐initiated ATRP: XPS monitoring. Surface Science 2007, 601(11):2357‐2366.

77. Hofer R, Textor M, Spencer ND: Alkyl Phosphate Monolayers, Self‐Assembled from Aqueous Solution onto Metal Oxide Surfaces. Langmuir 2001, 17(13):4014‐4020.

78. Love JC, Estroff LA, Kriebel JK, Nuzzo RG, Whitesides GM: Self‐Assembled Monolayers of Thiolates on Metals as a Form of Nanotechnology. Chemical Reviews 2005, 105(4):1103‐1170.

79. Zhang F, Kang ET, Neoh KG, Wang P, Tan KL: Surface modification of stainless steel by grafting of poly(ethylene glycol) for reduction in protein adsorption. Biomaterials 2001, 22(12):1541‐1548.

80. Chen H, Yuan L, Song W, Wu Z, Li D: Biocompatible polymer materials: Role of protein‐surface interactions. Progress in Polymer Science 2008, 33(11):1059‐1087.

81. Bhattacharya A, Misra BN: Grafting: a versatile means to modify polymers: Techniques, factors and applications. Progress in Polymer Science 2004, 29(8):767‐814.

82. Zdyrko B, Luzinov I: Polymer Brushes by the “Grafting to” Method. Macromol Rapid Commun 2011, 32(12):859‐869.

83. Griffith LG: Polymeric biomaterials. Acta Materialia 2000, 48(1):263‐277. 84. Goddard JM, Hotchkiss JH: Polymer surface modification for the attachment of bioactive compounds.

Progress in Polymer Science 2007, 32(7):698‐725.

Page 116: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

Bibliography

106 | P a g e

85. Liu Y, Sheng X, Dan X, Xiang Q: Preparation of mica/apatite glass‐ceramics biomaterials. Materials Science and Engineering: C 2006, 26(8):1390‐1394.

86. Ferretti S, Paynter S, Russell DA, Sapsford KE, Richardson DJ: Self‐assembled monolayers: a versatile tool for the formulation of bio‐surfaces. TrAC Trends in Analytical Chemistry 2000, 19(9):530‐540.

87. Hermanson GT (ed.): Bioconjugate Techniques, 2nd edn: Academic Press, Inc; 2008. 88. Binder WH, Sachsenhofer R: ‘Click’ Chemistry in Polymer and Material Science: An Update. Macromol

Rapid Commun 2008, 29(12‐13):952‐981. 89. Bergstrand A, Rahmani‐Monfared G, Östlund Å, Nydén M, Holmberg K: Comparison of PEI‐PEG and

PLL‐PEG copolymer coatings on the prevention of protein fouling. Journal of Biomedical Materials Research Part A 2009, 88A(3):608‐615.

90. Park KD, Kim YS, Han DK, Kim YH, Lee EHB, Suh H, Choi KS: Bacterial adhesion on PEG modified polyurethane surfaces. Biomaterials 1998, 19(7‐9):851‐859.

91. Yoshinari M, Kato T, Matsuzaka K, Hayakawa T, Shiba K: Prevention of biofilm formation on titanium surfaces modified with conjugated molecules comprised of antimicrobial and titanium‐binding peptides. Biofouling: The Journal of Bioadhesion and Biofilm Research 2010, 26(1):103 ‐ 110.

92. Dalsin JL, Hu B‐H, Lee BP, Messersmith PB: Mussel Adhesive Protein Mimetic Polymers for the Preparation of Nonfouling Surfaces. Journal of the American Chemical Society 2003, 125(14):4253‐4258.

93. Ye Q, Zhou F, Liu W: Bioinspired catecholic chemistry for surface modification. Chemical Society Reviews 2011, 40(7):4244‐4258.

94. Parfitt GD: Surface chemistry of oxides. Pure and applied chemistry 1976, 48:415‐418. 95. Mathieson I, Bradley RH: Improved adhesion to polymers by UV/ozone surface oxidation.

International Journal of Adhesion and Adhesives 1996, 16(1):29‐31. 96. Xiao D, Zhang H, Wirth M: Chemical Modification of the Surface of Poly(dimethylsiloxane) by Atom‐

Transfer Radical Polymerization of Acrylamide. Langmuir 2002, 18(25):9971‐9976. 97. Siow KS, Britcher L, Kumar S, Griesser HJ: Plasma Methods for the Generation of Chemically Reactive

Surfaces for Biomolecule Immobilization and Cell Colonization ‐ A Review. Plasma Processes and Polymers 2006, 3(6‐7):392‐418.

98. Hans JG, et al.: Interfacial properties and protein resistance of nano‐scale polysaccharide coatings. Smart Materials and Structures 2002, 11(5):652.

99. Cole MA, Voelcker NH, Thissen H: Electro‐induced protein deposition on low‐fouling surfaces. Smart Materials and Structures 2007, 16(6):2222‐2228.

100. Thissen H, Johnson G, Hartley PG, Kingshott P, Griesser HJ: Two‐dimensional patterning of thin coatings for the control of tissue outgrowth. Biomaterials 2006, 27(1):35‐43.

101. Seifert M, Rinke MT, Galla H‐J: Characterization of Streptavidin Binding to Biotinylated, Binary Self‐Assembled Thiol Monolayers—Influence of Component Ratio and Solvent. Langmuir 2010, 26(9):6386‐6393.

102. Kingshott P, Wei J, Bagge‐Ravn D, Gadegaard N, Gram L: Covalent attachment of poly(ethylene glycol) to surfaces, critical for reducing bacterial adhesion. Langmuir 2003, 19(17):6912‐6921.

103. Cerruti M, Fissolo S, Carraro C, Ricciardi C, Majumdar A, Maboudian R: Poly(ethylene glycol) Monolayer Formation and Stability on Gold and Silicon Nitride Substrates. Langmuir 2008, 24(19):10646‐10653.

104. Kanta A, Sedev R, Ralston J: The formation and stability of self‐assembled monolayers of octadecylphosphonic acid on titania. Colloids and Surfaces A: Physicochemical and Engineering Aspects 2006, 291(1‐3):51‐58.

105. Mani G, Johnson DM, Marton D, Dougherty VL, Feldman MD, Patel D, Ayon AA, Agrawal CM: Stability

Page 117: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

Bibliography

107 | P a g e

of Self‐Assembled Monolayers on Titanium and Gold. Langmuir 2008, 24(13):6774‐6784. 106. Blättler TM, Pasche S, Textor M, Griesser HJ: High Salt Stability and Protein Resistance of Poly(l‐

lysine)‐g‐poly(ethylene glycol) Copolymers Covalently Immobilized via Aldehyde Plasma Polymer Interlayers on Inorganic and Polymeric Substrates. Langmuir 2006, 22(13):5760‐5769.

107. Jennissen HP, Laub M: Development of an universal affinity fusion tag (Poly‐DOPA) for immobilizing recombinant proteins on biomaterials. Materialwissenschaft und Werkstofftechnik 2007, 38(12):1035‐1039.

108. Malisova B, Tosatti S, Textor M, Gademann K, Zürcher S: Poly(ethylene glycol) Adlayers Immobilized to Metal Oxide Substrates Through Catechol Derivatives: Influence of Assembly Conditions on Formation and Stability. Langmuir 2010, 26(6):4018‐4026.

109. Unsworth LD, Sheardown H, Brash JL: Protein‐Resistant Poly(ethylene oxide)‐Grafted Surfaces: Chain Density‐Dependent Multiple Mechanisms of Action. Langmuir 2008, 24(5):1924‐1929.

110. Sofia SJ, Premnath V, Merrill EW: Poly(ethylene oxide) Grafted to Silicon Surfaces: Grafting Density and Protein Adsorption. Macromolecules 1998, 31(15):5059‐5070.

111. Gennes PGd: Conformations of Polymers Attached to an Interface. Macromolecules 1980, 13:1069‐1075.

112. Luo N, Hutchison JB, Anseth KS, Bowman CN: Surface‐Initiated Photopolymerization of Poly(ethylene glycol) Methyl Ether Methacrylate on a Diethyldithiocarbamate‐Mediated Polymer Substrate. Macromolecules 2002, 35(7):2487‐2493.

113. Taylor W, Jones RAL: Producing High‐Density High‐Molecular‐Weight Polymer Brushes by a Grafting to Method from a Concentrated Homopolymer Solution. Langmuir 2010, 26(17):13954‐13958.

114. Minko S, Patil S, Datsyuk V, Simon F, Eichhorn K‐J, Motornov M, Usov D, Tokarev I, Stamm M: Synthesis of Adaptive Polymer Brushes via a Grafting‐To Approach from Melt. Langmuir 2002, 18(1):289‐296.

115. Uchida K, Hoshino Y, Tamura A, Yoshimoto K, Kojima S, Yamashita K, Yamanaka I, Otsuka H, Kataoka K, Nagasaki Y: Creation of a mixed poly(ethylene glycol) tethered‐chain surface for preventing the nonspecific adsorption of proteins and peptides. Biointerphases 2007, 2(4):126‐130.

116. Hey MJ, Ilett SM, Davidson G: Effect of temperature on poly(ethylene oxide) chains in aqueous solution. A viscometric, 1H NMR and Raman spectroscopic study. Journal of the Chemical Society, Faraday Transactions 1995, 91(21):3897‐3900.

117. Görgényi M, Dewulf J, Van Langenhove H, Héberger K: Aqueous salting‐out effect of inorganic cations and anions on non‐electrolytes. Chemosphere 2006, 65(5):802‐810.

118. Merrett K, Cornelius RM, McClung WG, Unsworth LD, Sheardown H: Surface analysis methods for characterizing polymeric biomaterials. Journal of Biomaterials Science, Polymer Edition 2002, 13(6):593‐621.

119. Ma Z, Mao Z, Gao C: Surface modification and property analysis of biomedical polymers used for tissue engineering. Colloids and Surfaces B: Biointerfaces 2007, 60(2):137‐157.

120. Ratner BD, Chilkoti A, Castner DG: Contemporary methods for characterizing complex biomaterial surfaces. Clinical Materials 1992, 11(1‐4):25‐36.

121. Kannan RY, Salacinski HJ, Vara DS, Odlyha M, Seifalian AM: Review paper: Principles and Applications of Surface Analytical Techniques at the Vascular Interface. Journal of Biomaterials Applications 2006, 21(1):5‐32.

122. Green RJ, Davies J, Davies MC, Roberts CJ, Tendler SJB: Surface plasmon resonance for real time in situ analysis of protein adsorption to polymer surfaces. Biomaterials 1997, 18(5):405‐413.

123. Elwing H: Protein absorption and ellipsometry in biomaterial research. Biomaterials 1998, 19(4‐5):397‐406.

124. Martin Halter MG, Marcus Textor, Janos Vörös, and H. Michelle Grandin: Enhanced optical waveguide

Page 118: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

Bibliography

108 | P a g e

light mode spectroscopy via detection of fluorophore absorbance Review of Scientific Instruments 2006, 77(10):6.

125. King RJ, Talim SP: A Comparison of Thin Film Measurement by Guided Waves, Ellipsometry and Reflectometry. Optica Acta: International Journal of Optics 1981, 28(8):1107‐1123.

126. Johnston KS, Karlsen SR, Jung CC, Yee SS: New analytical technique for characterization of thin films using surface plasmon resonance. Materials Chemistry and Physics 1995, 42(4):242‐246.

127. Piegari A, Masetti E: Thin film thickness measurement: A comparison of various techniques. Thin Solid Films 1985, 124(3‐4):249‐257.

128. Castner DG, Hinds K, Grainger DW: X‐ray Photoelectron Spectroscopy Sulfur 2p Study of Organic Thiol and Disulfide Binding Interactions with Gold Surfaces. Langmuir 1996, 12(21):5083‐5086.

129. Sauerbrey G: Use of quartz oscillators for weighing thin layers and Microweighing. Journal of Physics A Hadrons and Nuclei 1959, 155(2):206‐222.

130. Voinova MV, Jonson M, Kasemo B: 'Missing mass' effect in biosensor's QCM applications. Biosensors and Bioelectronics 2002, 17(10):835‐841.

131. Marx KA: Quartz Crystal Microbalance: A Useful Tool for Studying Thin Polymer Films and Complex Biomolecular Systems at the Solution−Surface Interface. Biomacromolecules 2003, 4(5):1099‐1120.

132. Menz B, Knerr R, Göpferich A, Steinem C: Impedance and QCM analysis of the protein resistance of self‐assembled PEGylated alkanethiol layers on gold. Biomaterials 2005, 26(20):4237‐4243.

133. Saito N, Matsuda T: Protein adsorption on self‐assembled monolayers with water‐soluble non‐ionic oligomers using quartz‐crystal microbalance. Materials Science and Engineering: C 1998, 6(4):261‐266.

134. Lee BS, Chi YS, Lee K‐B, Kim Y‐G, Choi IS: Functionalization of Poly(oligo(ethylene glycol) methacrylate) Films on Gold and Si/SiO2 for Immobilization of Proteins and Cells: SPR and QCM Studies. Biomacromolecules 2007, 8(12):3922‐3929.

135. Blümmel J, Perschmann N, Aydin D, Drinjakovic J, Surrey T, Lopez‐Garcia M, Kessler H, Spatz JP: Protein repellent properties of covalently attached PEG coatings on nanostructured SiO2‐based interfaces. Biomaterials 2007, 28(32):4739‐4747.

136. C.Dixon M: Quartz Crystal Microbalance with Dissipation Monitoring: Enabling Real‐Time Characterization of Biological Materials and Their Interactions. Journal of Bimolecular Technologies 2008, 19(3):151‐158.

137. Troughton EB, Bain CD, Whitesides GM, Nuzzo RG, Allara DL, Porter MD: Monolayer films prepared by the spontaneous self‐assembly of symmetrical and unsymmetrical dialkyl sulfides from solution onto gold substrates: structure, properties, and reactivity of constituent functional groups. Langmuir 1988, 4(2):365‐385.

138. Van Oss CJ, Chaudhury MK, Good RJ: Interfacial Lifshitz‐van der Waals and polar interactions in macroscopic systems. Chemical Reviews 1988, 88(6):927‐941.

139. Kakiuchi T, Sato K, Iida M, Hobara D, Imabayashi S‐i, Niki K: Phase Separation of Alkanethiol Self‐Assembled Monolayers during the Replacement of Adsorbed Thiolates on Au(111) with Thiols in Solution. Langmuir 2000, 16(18):7238‐7244.

140. Baralia GG, Duwez A‐S, Nysten B, Jonas AM: Kinetics of Exchange of Alkanethiol Monolayers Self‐Assembled on Polycrystalline Gold. Langmuir 2005, 21(15):6825‐6829.

141. Magonov SN, Reneker DH: Characterization of polymer surfaces with atomic force microscopy. Annu Rev Mater Sci 1997, 27:175‐222.

142. Hamilton‐Brown P, Gengenbach T, Griesser HJ, Meagher L: End Terminal, Poly(ethylene oxide) Graft Layers: Surface Forces and Protein Adsorption. Langmuir 2009, 25(16):9149‐9156.

143. Li S, Cao P, Colorado R, Yan X, Wenzl I, Shmakova OE, Graupe M, Lee TR, Perry SS: Local Packing Environment Strongly Influences the Frictional Properties of Mixed CH3‐ and CF3‐Terminated

Page 119: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

Bibliography

109 | P a g e

Alkanethiol SAMs on Au(111). Langmuir 2005, 21(3):933‐936. 144. Cappella B, Dietler G: Force‐distance curves by atomic force microscopy. Surface Science Reports 1999,

34(1‐3):1‐104. 145. Derjaguin BV, Muller VM, Toporov YP: Effect of contact deformations on the adhesion of particles.

Journal of Colloid and Interface Science 1975, 53(2):314‐326. 146. Pittenger B EN, Su C: Quantitative mechanical property mapping at the nanoscale with PeakForce

QNM Application Note Veeco Instruments Inc. 147. Sweers K, van der Werf K, Bennink M, Subramaniam V: Nanomechanical properties of alpha‐synuclein

amyloid fibrils: a comparative study by nanoindentation, harmonic force microscopy, and Peakforce QNM. Nanoscale Research Letters 2011, 6(1):270.

148. J. A. Woollam Co. I: Ellipsometry tutrotial. In: Application notes. 149. Web‐page: Ellipsometry In., vol. http://academic.brooklyn.cuny.edu/physics/holden/ellipsometry.htm. 150. Unsworth LD, Tun Z, Sheardown H, Brash JL: Chemisorption of thiolated poly(ethylene oxide) to gold:

surface chain densities measured by ellipsometry and neutron reflectometry. Journal of Colloid and Interface Science 2005, 281(1):112‐121.

151. Sauer K, Camper AK, Ehrlich GD, Costerton JW, Davies DG: Pseudomonas aeruginosa Displays Multiple Phenotypes during Development as a Biofilm. In., vol. 184; 2002: 1140‐1154.

152. Unsworth LD, Sheardown H, Brash JL: Polyethylene oxide surfaces of variable chain density by chemisorption of PEO‐thiol on gold: Adsorption of proteins from plasma studied by radiolabelling and immunoblotting. Biomaterials 2005, 26(30):5927‐5933.

153. Uchida K, Otsuka H, Kaneko M, Kataoka K, Nagasaki Y: A Reactive Poly(ethylene glycol) Layer To Achieve Specific Surface Plasmon Resonance Sensing with a High S/N Ratio: The Substantial Role of a Short Underbrushed PEG Layer in Minimizing Nonspecific Adsorption. Analytical Chemistry 2005, 77(4):1075‐1080.

154. Chi Ying Lee LJG, David W. Grainger, and David G. Castner: Mixed DNA/oligo(ethylene glycol) functionalized gold surfaces improve DNA hybridization in complex media Biointerphases 2006, 1(2):11.

155. Herne TM, Tarlov MJ: Characterization of DNA Probes Immobilized on Gold Surfaces. Journal of the American Chemical Society 1997, 119(38):8916‐8920.

156. Brunner S, Caseri WR, Suter UW, Hähner G, Brovelli D, Spencer ND, Vinckier A, Rau IU, Galda P, Rehahn M: Preparation and Characterization of Ultrathin Layers of Substituted Oligo‐ and Poly(p‐phenylene)s and Mixed Layers with Octadecanethiol on Gold and Copper. Langmuir 1999, 15(19):6333‐6342.

157. Tosatti S, Michel R, Textor M, Spencer ND: Self‐Assembled Monolayers of Dodecyl and Hydroxy‐dodecyl Phosphates on Both Smooth and Rough Titanium and Titanium Oxide Surfaces. Langmuir 2002, 18(9):3537‐3548.

158. Bozzini S, Petrini P, Tanzi MC, Zürcher S, Tosatti S: Poly(ethylene glycol) and Hydroxy Functionalized Alkane Phosphate Mixed Self‐Assembled Monolayers to Control Nonspecific Adsorption of Proteins on Titanium Oxide Surfaces. Langmuir 2009, 26(9):6529‐6534.

159. Amstad E, Zurcher S, Mashaghi A, Wong JY, Textor M, Reimhult E: Surface Functionalization of Single Superparamagnetic Iron Oxide Nanoparticles for Targeted Magnetic Resonance Imaging. Small 2009, 5(11):1334‐1342.

160. Stranick SJ, Parikh AN, Tao YT, Allara DL, Weiss PS: Phase Separation of Mixed‐Composition Self‐Assembled Monolayers into Nanometer Scale Molecular Domains. The Journal of Physical Chemistry 1994, 98(31):7636‐7646.

161. Otsuka H, Nagasaki Y, Kataoka K: Characterization of Aldehyde‐PEG Tethered Surfaces: Influence of PEG Chain Length on the Specific Biorecognition. Langmuir 2004, 20(26):11285‐11287.

Page 120: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

Bibliography

110 | P a g e

162. Lee C‐Y, Gong P, Harbers GM, Grainger DW, Castner DG, Gamble LJ: Surface Coverage and Structure of Mixed DNA/Alkylthiol Monolayers on Gold: Characterization by XPS, NEXAFS, and Fluorescence Intensity Measurements. Analytical Chemistry 2006, 78(10):3316‐3325.

163. Cole MA, Thissen H, Losic D, Voelcker NH: A new approach to the immobilisation of poly(ethylene oxide) for the reduction of non‐specific protein adsorption on conductive substrates. Surface Science 2007, 601(7):1716‐1725.

164. Ayyoob Arpanaeia SP, Peter Kingshotta, Nete Bernbomd, Jan J. Enghilda, Yoke Yin Ng, Lone Gram, Flemming Besenbachera,and Rikke L. Meyera: Muscle proteins from fish minimise bacterial adhesion on a variety of material surfaces. Submitted.

165. Tokumitsu S, Liebich A, Herrwerth S, Eck W, Himmelhaus M, Grunze M: Grafting of Alkanethiol‐Terminated Poly(ethylene glycol) on Gold. Langmuir 2002, 18(23):8862‐8870.

166. Dalsin JL, Lin L, Tosatti S, Vörös J, Textor M, Messersmith PB: Protein Resistance of Titanium Oxide Surfaces Modified by Biologically Inspired mPEG−DOPA. Langmuir 2004, 21(2):640‐646.

167. Pasche S, De Paul SM, Vörös J, Spencer ND, Textor M: Poly(l‐lysine)‐graft‐poly(ethylene glycol) Assembled Monolayers on Niobium Oxide Surfaces: A Quantitative Study of the Influence of Polymer Interfacial Architecture on Resistance to Protein Adsorption by ToF‐SIMS and in Situ OWLS. Langmuir 2003, 19(22):9216‐9225.

168. Ostuni E, Chapman RG, Liang MN, Meluleni G, Pier G, Ingber DE, Whitesides GM: Self‐Assembled Monolayers That Resist the Adsorption of Proteins and the Adhesion of Bacterial and Mammalian Cells. Langmuir 2001, 17(20):6336‐6343.

169. Hangler M, Burmølle M, Schneider I, Allermann K, Jensen B: The serine protease Esperase HPF inhibits the formation of multispecies biofilm. Biofouling 2009, 25(7):667‐674.

170. Pettitt ME, Henry SL, Callow ME, Callow JA, Clare AS: Activity of Commercial Enzymes on Settlement and Adhesion of Cypris Larvae of the Barnacle Balanus amphitrite, Spores of the Green Alga Ulva linza, and the Diatom Navicula perminuta. Biofouling 2004, 20(6):299‐311.

171. Biology IUoBaM: Nomenclature and Classification of Enzymes In. San Diego: Academic press; 1992. 172. Crapisi A, Lante A, Pasini G, Spettoli P: Enhanced microbial cell lysis by the use of lysozyme

immobilized on different carriers. Process Biochemistry 1994, 28(1):17‐21. 173. Charabarty ABaAM: Role of alginate lyase in cell detachment of Pseudomonas aeruginosa. Journal of

Applied environmental Microbiology 1994, 60(7):2355‐2359. 174. Fuqua C, Greenberg EP: Listening in on bacteria: acyl‐homoserine lactone signalling. Nat Rev Mol Cell

Biol 2002, 3(9):685‐695. 175. Kaplan JB: Methods for the treatment and prevention of bacterial biofilms. Expert Opinion on

Therapeutic Patents 2005, 15(8):955‐965. 176. Cherry JR, Fidantsef AL: Directed evolution of industrial enzymes: an update. Current Opinion in

Biotechnology 2003, 14(4):438‐443. 177. Rao MB, Tanksale AM, Ghatge MS, Deshpande VV: Molecular and Biotechnological Aspects of

Microbial Proteases. Microbiol Mol Biol Rev 1998, 62(3):597‐635. 178. Gupta, Beg, Lorenz: Bacterial alkaline proteases: molecular approaches and industrial applications.

Applied Microbiology and Biotechnology 2002, 59(1):15‐32. 179. Araújo R, Cavaco‐Paulo A, Casal M: Strategies towards the Functionalization of Subtilisin E from

Bacillus subtilis for Wool Finishing Applications. Engineering in Life Sciences 2008, 8(3):238‐249. 180. Kumar CG, Takagi H: Microbial alkaline proteases: From a bioindustrial viewpoint. Biotechnology

Advances 1999, 17(7):561‐594. 181. Magnus Andersson OA, Bernt Eric Uhlin, and Erik Fällman Optical tweezers for single molecule force

spectroscopy on bacterial adhesion organelles. Proceedings of SPIE 2006, 6326(1):632620‐632620.

Page 121: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

Bibliography

111 | P a g e

182. Alzohairy AAKaMA: Recent Advances and Applications of Immobilized Enzyme Technologies: A Review. Research Journal of Biological Sciences 2010, 5(8):565‐575.

183. Brode PF, Erwin CR, Rauch DS, Lucas DS, Rubingh DN: Enzyme behavior at surfaces. Site‐specific variants of subtilisin BPN' with enhanced surface stability. Journal of Biological Chemistry 1994, 269(38):23538‐23543.

184. Kleijn Mike NW: The asdsorption of proteins from aqueous solution on solid surfaces. Heterogeneous Chemistry 1995.

185. Nakanishi K, Sakiyama T, Imamura K: On the adsorption of proteins on solid surfaces, a common but very complicated phenomenon. Journal of Bioscience and Bioengineering 2001, 91(3):233‐244.

186. Roach P, Farrar D, Perry CC: Interpretation of Protein Adsorption:Surface‐Induced Conformational Changes. Journal of the American Chemical Society 2005, 127(22):8168‐8173.

187. Wang J, Bhattacharyya D, Bachas LG: Improving the activity of immobilized subtilisin by site‐directed attachment through a genetically engineered affinity tag. Fresenius' Journal of Analytical Chemistry 2001, 369(3):280‐285.

188. Tomoko Tada KM, Eri Yoshida, Naoki Tanaka, and Shigeru Kunugi: SH‐Group Introduction to the N‐terminal of Subtilisin and Preparation of Immobilized and Dimeric Enzymes. Bull Chem Soc Jpn 2002, 75(10):2247–2251.

189. Tasso M, Pettitt ME, Cordeiro AL, Callow ME, Callow JA, Werner C: Antifouling potential of Subtilisin A immobilized onto maleic anhydride copolymer thin films. In., vol. 25: Taylor & Francis; 2009: 505 ‐ 516.

190. Liu J, Wang J, Bachas LG, Bhattacharyya D: Activity Studies of Immobilized Subtilisin on Functionalized Pure Cellulose‐Based Membranes. Biotechnology Progress 2001, 17(5):866‐871.

191. Rusmini F, Zhong Z, Feijen J: Protein Immobilization Strategies for Protein Biochips. Biomacromolecules 2007, 8(6):1775‐1789.

192. ExPASy ProtParam tool [http://web.expasy.org/protparam/] 193. Protein database NCfBI. In.: U.S. National Library of Medicine. 194. Cheng F, Gamble LJ, Grainger DW, Castner DG: X‐ray Photoelectron Spectroscopy, Time‐of‐Flight

Secondary Ion Mass Spectrometry, and Principal Component Analysis of the Hydrolysis, Regeneration, and Reactivity of N‐Hydroxysuccinimide‐Containing Organic Thin Films. Analytical Chemistry 2007, 79(22):8781‐8788.

195. Vermette P, Meagher L: Immobilization and Characterization of Poly(acrylic acid) Graft Layers. Langmuir 2002, 18(26):10137‐10145.

196. Staros JV, Wright RW, Swingle DM: Enhancement by N‐hydroxysulfosuccinimide of water‐soluble carbodiimide‐mediated coupling reactions. Analytical Biochemistry 1986, 156(1):220‐222.

197. Grabarek Z, Gergely J: Zero‐length crosslinking procedure with the use of active esters. Analytical Biochemistry 1990, 185(1):131‐135.

198. CHROMOGENIC SUBSTRATES UNIVERSITY 199. Gong P, Grainger DW: Comparison of DNA immobilization efficiency on new and regenerated

commercial amine‐reactive polymer microarray surfaces. Surface Science 2004, 570(1‐2):67‐77. 200. IUBMB: International enzyme units and isoenzyme nomenclature. J clin Path 1963, 16:391‐393. 201. Haurowitz.F (ed.): The Chemistry and Function of Proteins. New York: Academic Press; 1963. 202. Iwase T, Uehara Y, Shinji H, Tajima A, Seo H, Takada K, Agata T, Mizunoe Y: Staphylococcus epidermidis

Esp inhibits Staphylococcus aureus biofilm formation and nasal colonization. Nature 2010, 465(7296):346‐349.

203. Dutta P, Ray N, Roy S, Dasgupta AK, Bouloussa O, Sarkar A: Covalent immobilization of active lysozyme on Si/glass surface using alkoxy Fischer carbene complex on SAM. Organic & Biomolecular Chemistry

Page 122: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

Bibliography

112 | P a g e

2011, 9(14):5123‐5128. 204. Wu Y, Daeschel MA: Lytic Antimicrobial Activity of Hen Egg White Lysozyme Immobilized to

Polystyrene Beads. Journal of Food Science 2007, 72(9):M369‐M374. 205. Eckert CA, Knutson BL, Debenedetti PG: Supercritical fluids as solvents for chemical and materials

processing. Nature 1996, 383(6598):313‐318. 206. Beckman EJ: Supercritical and near‐critical CO2 in green chemical synthesis and processing. The

Journal of Supercritical Fluids 2004, 28(2‐3):121‐191. 207. Bahrami M, Ranjbarian S: Production of micro‐ and nano‐composite particles by supercritical carbon

dioxide. The Journal of Supercritical Fluids 2007, 40(2):263‐283. 208. Kazarian SG: Polymer processing with supercritical fluids. Polymer science, Ser C 2000, 42(1):78‐101. 209. Brudi K, Dahmen N, Schmieder H: Partition coefficients of organic substances in two‐phase mixtures

of water and carbon dioxide at pressures of 8 to 30 MPa and temperatures of 313 to 333 K. The Journal of Supercritical Fluids 1996, 9(3):146‐151.

210. Lou X, Janssen H‐G, Cramers CA: Temperature and pressure effects on solubility in supercritical carbon dioxide and retention in supercritical fluid chromatography. Journal of Chromatography A 1997, 785(1‐2):57‐64.

211. Kirby CF, McHugh MA: Phase Behavior of Polymers in Supercritical Fluid Solvents. Chemical Reviews 1999, 99(2):565‐602.

212. Drohmann C, Beckman EJ: Phase behavior of polymers containing ether groups in carbon dioxide. The Journal of Supercritical Fluids 2002, 22(2):103‐110.

213. Liu T, Hu G‐H, Tong G‐s, Zhao L, Cao G‐p, Yuan W‐k: Supercritical Carbon Dioxide Assisted Solid‐State Grafting Process of Maleic Anhydride onto Polypropylene. Industrial & Engineering Chemistry Research 2005, 44(12):4292‐4299.

214. Kunita MH, Rinaldi AW, Girotto EM, Radovanovic E, Muniz EC, Rubira AF: Grafting of glycidyl methacrylate onto polypropylene using supercritical carbon dioxide. European Polymer Journal 2005, 41(9):2176‐2182.

215. Jia X, McCarthy TJ: Buried Interface Modification Using Supercritical Carbon Dioxide. Langmuir 2002, 18(3):683‐687.

216. Gu W, Tripp CP: Reaction of Silanes in Supercritical CO2 with TiO2 and Al2O3. Langmuir 2006, 22(13):5748‐5752.

217. Tripp CP, Combes JR: Chemical Modification of Metal Oxide Surfaces in Supercritical CO2: The Interaction of Supercritical CO2 with the Adsorbed Water Layer and the Surface Hydroxyl Groups of a Silica Surface. Langmuir 1998, 14(26):7348‐7352.

218. Mishima K, Matsuyama K, Nagatani M: Solubilities of poly(ethylene glycol)s in the mixtures of supercritical carbon dioxide and cosolvent. Fluid Phase Equilibria 1999, 161(2):315‐324.

219. Daneshvar M, Kim S, Gulari E: High‐pressure phase equilibria of polyethylene glycol‐carbon dioxide systems. The Journal of Physical Chemistry 1990, 94(5):2124‐2128.

220. Rijnkels M, Kooiman PM, Krimpenfort PJ, de Boer HA, Pieper FR: Expression analysis of the individual bovine beta‐, alpha s2‐ and kappa‐casein genes in transgenic mice. Biochem J 1995, 311(3):929‐937.

221. Mir MA: Lysozyme : a brief review. Postgraduate Medical Journal 1977, 53:257.259. 222. Krebs HA: CHEMICAL COMPOSITION OF BLOOD PLASMA AND SERUM. Annu Rev Biochem 1950,

19:409‐430. 223. Brandrup J, Immergut EH, Grulke EA, Abe A, Bloch DR: Polymer Handbook (4th Edition). In.: John Wiley

& Sons. 224. Cumpson PJ: Estimation of inelastic mean free paths for polymers and other organic materials: use of

quantitative structure–property relationships. Surface and Interface Analysis 2001, 31(1):23‐34.

Page 123: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

Bibliography

113 | P a g e

225. Laibinis PE, Bain CD, Whitesides GM: Attenuation of photoelectrons in monolayers of n‐alkanethiols adsorbed on copper, silver, and gold. The Journal of Physical Chemistry 1991, 95(18):7017‐7021.

226. Piehler J, Brecht A, Valiokas R, Liedberg B, Gauglitz G: A high‐density poly(ethylene glycol) polymer brush for immobilization on glass‐type surfaces. Biosensors and Bioelectronics 2000, 15(9‐10):473‐481.

227. Ostaci R‐V, Damiron D, Al Akhrass S, Grohens Y, Drockenmuller E: Poly(ethylene glycol) brushes grafted to silicon substrates by click chemistry: influence of PEG chain length, concentration in the grafting solution and reaction time. Polymer Chemistry 2011, 2(2):348‐354.

228. Mehne J, Markovic G, Pröll F, Schweizer N, Zorn S, Schreiber F, Gauglitz G: Characterisation of morphology of self‐assembled PEG monolayers: a comparison of mixed and pure coatings optimised for biosensor applications. Analytical and Bioanalytical Chemistry 2008, 391(5):1783‐1791.

Page 124: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

114

Appendix

Mixed poly (ethylene glycol) (PEG) and oligo (ethylene glycol)

(OEG) layers on gold as non-fouling surfaces created by

backfilling

Status of article:

Submitted to ‘Biointerphases’

Arcot R. Lokanathana, Shuai Zhanga, Viduthalai R. Reginaa, Martin A. Colea, Ryosuke Ogakia‡,

Mingdong Donga, Flemming Besenbachera, Rikke L. Meyera, Peter Kingshottb‡

a- The Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, Ny Munkegade

120, Aarhus 8000, Denmark.

b- Industrial Research Institute Swinburne (IRIS), Swinburne University of Technology,

Burwood Road, Hawthorn 3122, Australia.

‡ Corresponding authors:

I. Ryosuke Ogaki

Phone number: +45 8942 3556,

Email: [email protected]

II. Peter Kingshott

Formerly at iNANO, Aarhus University, Denmark

Currently at IRIS, Swinburne University of Technology, Australia

Phone number: +61 9214 5033

Email: [email protected]

Page 125: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

115

ABSTRACT

Backfilling a self-assembled monolayer (SAM) of long PEG with short PEG is a

well known strategy to improve its non-fouling potential. Here we show, using x-ray

photoelectron spectroscopy (XPS), contact angle and atomic force microscopy (AFM)

that backfilling PEG thiol monolayer with OEG terminated alkane thiol molecules results

in underbrush formation. We also confirm the absence of phase separated arrangement

which is commonly observed with backfilling experiments involving SAM of short chain

alkane thiol with long chain alkane thiol. Furthermore it was found that OEG addition

caused less PEG desorption when compared to alkane thiol. The non-fouling potential of

surfaces was tested through serum adsorption and bacterial adhesion studies. We

demonstrate that the mixed monolayer with PEG and OEG is better than PEG at resisting

protein adsorption and bacterial adhesion, and conclude that backfilling PEG with OEG

resulting in the underbrush formation enhances the non-fouling potential of PEG.

KEYWORDS

Non-fouling, PEG, Oligo (ethylene glycol), mixed monolayers, XPS, QCM-D

I. INTRODUCTION

Non-fouling coatings are immensely important to a variety of biomedical

applications such as implants, drug delivery, tissue engineering and biosensing 1. They

Page 126: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

116

are also important for water purification, food processing and marine industries 2. One

goal of a non-fouling coating is to reduce biofilm formation on surfaces while

maintaining functionality. Biofilm formation is found to be facilitated by a conditioning

layer (adsorbed layer of biomolecules from the surrounding environment), which is

followed by reversible physiochemical attachment of microbes 3. Among the various

strategies used in making non-fouling coatings, preventing initial bioadhesion is a

popular approach that generally involves the use of a hydrophilic polymeric coating. The

concept is to present an interfacial steric barrier between the substrate and the

environment in order to prevent conditioning layer dependent biofouling. Polyethylene

glycol (PEG) is widely used to functionalize surfaces in order to render them non-fouling.

The osmotic and elastic properties of PEG along with its neutral charge, non toxic nature

make it an effective and safe choice to minimize fouling in a number of biologically

relevant applications 4. The ability for a given PEG functionalized surface to resist

fouling is thought to be mainly dependent on the graft density and molecular weight

(MW) of PEG used, both of which are key properties that decide the conformation of

PEG chains on surfaces 5 6 7 . For example in the case of high or low graft density, PEG

chains are arranged in either the ‘brush’ or ‘mushroom’ conformation respectively 8. A

pancake conformation is observed for very low graft densities. Self-assembled

monolayers (SAMs) of PEG chains are known to resist non specific adsorption best when

present in the brush conformation 7. Studies have shown that covalently immobilized

PEG coatings provide superior antifouling properties, probably since they cannot easily

be displaced by biomolecules or conditioning layer molecules 9. PEG functionalization

techniques can be broadly classified into ‘grafting to’ and ‘grafting from’ approaches.

Page 127: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

117

The ‘grafting from’ technique generally gives higher graft density since the limiting

factor is diffusion of monomer onto the reactive ends of growing chains whereas in the

case of ‘grafting to’ the limitation is diffusion of entire polymer chains to the reactive

substrate 10. Several strategies such as cloud point grafting 11, grafting in homopolymer

solutions 12, grafting from polymeric melts 13 and underbrush formation by backfilling

with shorter molecules 14 have been used to increase the graft density for ’grafting to’

techniques. The backfilling approach, unlike the other three strategies that depend on

minimization of excluded volume interactions, is a simple method wherein the interchain

spaces present in layers of high MW PEG chains are filled with shorter PEG chains that

can diffuse to the surface. In a study of 5 kDa MW PEG by Uchida et al., it was shown

that backfilling with shorter PEG (MW 2 kDa) improved the ability of the surface to

resist non specific protein adsorption 14 15.

However the mechanism of formation of mixed layers is not yet fully understood.

The possibility of the formation of domains exists (Figure 1a) where the mechanism of

addition is similar to that of the replacement phenomena observed for short alkane thiol

SAM when they are exposed to longer alkane thiol molecules 16. Here, replacement of

short alkane thiol by longer alkane thiols begins at grain boundaries and is followed by

gradual replacement of entire domains, resulting in phase separated arrangement 16 17.

The replacement of shorter alkane thiols with longer alkane thiols in SAMs is

thermodynamically favored due to the higher number of intermolecular Van der Waals

interactions formed with longer alkane thiol molecule. Uchida et al. studied the

backfilling of high MW PEG layers with low MW PEG and assumed that the addition of

low MW species results in formation of well-mixed, homogeneous mixed layers 14 15.

Page 128: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

118

The aspect of replacement and the extent of replacement are expected to be low since it is

thermodynamically unfavorable for a longer PEG molecule to be replaced by a shorter

PEG molecule.

Here we have used oligo (ethylene glycol) (OEG) terminated alkane thiols to backfill

PEG thiol SAMs on Au surfaces. Since alkane thiols can from SAMs driven by

intermolecular Van der Waals interactions and are kinetically more reactive than high

MW PEG thiols our aim is to thoroughly explore whether phase separated domains are

formed (Figure 1a) and also the extent of PEG desorption. The schematic presentation of

thiolated PEG, OEG and alkane molecules are presented in Figure 1c. Although SAMs

of OEG have been shown to be very efficient in resisting protein adsorption 18 for short

durations, it has been identified that applications involving immobilization of bioactive

molecules such as enzymes would benefit from longer PEG chains due to higher mobility

for biorecognition 19.Thus we expect our combination of PEG-COOHSAM backfilled with

OEG3 will be an ideal support for immobilizing bioactive molecules.

We will use the following notations for SAM of PEG and for the backfilled surface:

PEG-COOHSAM, OEG3-SAM, and RSAM correspond to SAMs formed with PEG-

COOH, OEG3, and R molecules

PEG-COOHSAM+OEG3: PEG-COOHSAM backfilled with OEG3

PEG-COOHSAM+R: PEG-COOHSAM backfilled with R

II. MATERIALS AND METHODS

A. Chemicals

Page 129: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

119

α-carboxyl-ω-thiol poly(ethylene glycol) (PEG-COOH, 99% purity) was

purchased from Laysan Bio Inc. (Alabama, USA), and Hydroxy-terminated tri (ethylene

glycol) undecane thiol (OEG3, 99% purity) was purchased from Assemblon (Washington,

USA). Fetal bovine serum (FBS), octane thiol (R) (99% purity), SYBR Green II,

phosphate buffered saline (PBS) buffer tablets, 25% ammonium hydroxide (NH4OH),

30% hydrogen peroxide (H2O2) and absolute ethanol were purchased from Sigma Aldrich

(Aarhus, Denmark). Ultrapure MilliQ (MQ) water with resistivity of 18.2 MΩ was used

for making buffers and aqueous ethanolic solutions.

B. Substrate preparation and functionalization

Gold substrates were prepared by sputtering 50 nm gold layer onto a 3 nm

titanium adhesion layer on silicon wafers. Sputtering was done using a RF sputtering

system with Ti and Au targets of 10 cm diameter (2.54 W cm−2) inside a standard

chamber maintained at an Ar pressure of 2 × 10−3 mbar. The substrate-target separation

distance was 7 cm, while the deposition rates for Ti and Au were 0.4 nm s−1 and 1 nm s−1

respectively. The gold surfaces were cleaned by UV/ozone treatment for 30 minutes,

followed by treatment with basic piranha solution (H2O: NH4OH: H2O2 in ratio 4:1:1) at

70˚-80˚C for 5 minutes and then rinsed with MQ water. (CAUTION: Piranha solution

reacts violently upon contact with organic solutions). Cleaned gold slides were immersed

for 3 h at room temperature in 0.2 mM PEG-COOH solution (75% ethanol), followed by

rinsing with MQ water to remove any physically adsorbed molecules. Backfilling

involved immersion of PEG-COOHSAM for 3 h at room temperature in 1mM absolute

Page 130: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

120

ethanolic solution of OEG3 or R, followed by rinsing with ethanol and drying with a jet

of nitrogen. The SAMs of OEG3 and R for control experiments were made by immersing

cleaned gold slides in 1mM ethanolic solution of OEG3 or R for a period of 3 h followed

by rinse with ethanol and drying with nitrogen gas.

C. X-ray photoelectron spectroscopy

XPS spectra were recorded using a Kratos Axis UltraDLD instrument (Kratos Ltd,

Telford, UK) equipped with a monochromated aluminum anode (Al kα 1486 eV)

operating at 150 W power (15 kV and 10 mA) with pass energies of 80 eV and 160 eV

for high resolution and survey spectra respectively. A hybrid lens mode was employed

during analysis (electrostatic and magnetic). The XPS spectra were measured at three

areas on each sample and the take-off angle for all measurements was 0°. The measured

binding energy positions were charge corrected with reference to 285.0 eV,

corresponding to the C-C/C-H species. Quantification and curve fitting was conducted

using CasaXPS software. A linear background with a Gaussian to Lorentzian ratio of 30

was used to fit all spectra. The S 2p high resolution spectra were deconvoluted into two

doublets corresponding to bound and unbound thiol each with full width half maximum

of 1.2 eV). The lower binding energy doublet with its 2p3/2 at 161.9 eV was assigned to

bound thiol while the higher binding energy doublet with its 2p3/2 at 163.5 eV

corresponded to unbound thiol as described by Castner et al. 20. The representative high

resolution spectra are included in supplementary information (Figure S1b). Overlayer

thickness and graft density were calculated using the following equations 5 6 21:

⎟⎠⎞

⎜⎝⎛ −

=θλ sin

exp dII

O

Eqn-1

Page 131: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

121

79.0)(104.14207.0)(117.3

KeVHnon

ringsVO

EN

N

⎥⎥⎥

⎢⎢⎢

⎡+

+=

χλ Eqn-2

21

⎟⎟⎠

⎞⎜⎜⎝

⎛=

dNML

Aρ Eqn-3a 2

1L

S = Eqn-3b

Where Io –signal intensity from bare infinitely thick substrate, I – substrate signal after

functionalization, d overlayer thickness, λ – attenuation length of Au 4f photoelectron in

PEG polymer, θ take off angle of analyzer, oχv – atomic connectivity index of polymer

repeat unit, Nrings – number of aromatic rings, Nnon-H – number of atoms in repeat unit

excluding hydrogen atoms, EKeV – kinetic energy of photoelectron, L – interchain

distance, M – molecular weight of PEG chain, ρ – density assumed to be 1 gm/cm3, NA –

Avogadro number, S – graft density defined as number of chains per unit area.

D. Contact angle measurements

Static contact angles were measured on all surfaces using water, formamide and α-

bromonapthalene. All images of liquid drops on surfaces were recorded using a KRUSS

DSA100 (KRUSS GmbH, Hamburg, Germany), followed by drop shape analysis using

ImageJ software. Reported contact angles are the average of at least 6 measurements at

different surface positions (data are included in supplementary information Table S1a).

The values of γLd, γL

+, γL− for the three liquids used are given in supplementary

information (Table S1b). The interfacial Lifshitz van der Waals (γd) and polar

components (γ+, γ−) of surface tension of a surface can be determined by measuring

Page 132: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

122

contact angles with three different liquids followed by solving the corresponding

equations whose general form is given by 22,

( ) ( ) ( ) ( )[ ]+−−+ ++=+ LSLSdL

dSL γγγγγγλθ

21cos1 Eqn 4

θ – Measured contact angle. γL, γLd, γL

+ and γL− represent total surface tension, Lifshitz

van der Waals component, electron acceptor component, electron donor component of

liquid respectively. γS, γSd, γS

+, γS− and represent total surface tension, Lifshitz van der

Waals component, electron acceptor component, electron donor component of sample

surface respectively.

E. Atomic force microscopy

All of the AFM images were recorded using a commercial Nanoscope VIII

MultiMode SPM system (Bruker AXS, Santa Barbara, CA) in MQ water. Topography

images were recorded using Quantitative Nano-Mechanical Mapping (QNM) under

Peakforce Tapping mode 23. Ultrasharp silicon nitride cantilevers (triangular, Mpp-

12120-10, Bruker AXS) were used with a typical resonance frequency of 150 KHz in air,

a spring constant of 5 N/m and a normal tip radius of 8 nm. All AFM images were

recorded with 512 × 512 pixels resolution per image, and they were flattened and

analyzed with Scanning Probe Image Processor software (SPIPTM, Image Metrology

ApS, version 5.1.3, Lyngby, Denmark).

F. Quartz crystal microbalance with dissipation

QCM-D studies were performed using a Q-Sense E4 system (Gothenburg, Sweden).

The PEG-COOH functionalization followed by backfilling with OEG3 or R was

Page 133: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

123

performed on QSX 301 crystals (Q-Sense) coated with 100 nm gold which had

approximate resonance frequency of 5 MHz. Serum adsorption experiments were

performed in triplicate. Functionalized gold coated crystal surfaces were equilibrated at

37 °C in PBS (100 mM pH 7.4) prior to exposure to serum solution. All serum

adsorptions studies were carried out using 10% fetal bovine serum (FBS) at 37 °C. Once

a stable baseline was attained, the surfaces were exposed to 750 μL of protein solution

delivered at a flow rate of 100 μL/minute followed by rinse with PBS buffer. The shift in

resonance frequency corresponding to the 7th overtone was used to monitor the

adsorption. After measurement QCM crystals exposed to serum were rinsed with MQ

water to remove any salts before drying under a jet of nitrogen for XPS analysis.

G. Bacterial adhesion assay

A Staphylococcus aureus (ATCC 12598) starter culture was inoculated from agar

plates and grown in 3 ml of 1% tryptic soy broth (TSB) medium in 50 ml conical bottom

tube by incubating overnight in a shaker at 37 °C. An inoculum prepared by adding 1 ml

of starter culture to 100 ml 1% TSB medium was incubated in a shaker at 37 °C until

cultures were in the late-exponential growth phase (OD600 = 1.0). Cells were then

harvested by centrifugation (5 minutes at 3000 x g), washed twice and resuspended in

PBS (pH 7.4) and diluted to obtain an OD600 of 0.5. Surfaces were incubated with the

bacterial suspensions in 24 well plates for 5 h at 37°C and gently washed three times with

sterile PBS to remove non-adherent cells. The adherent cells on the surfaces were stained

with ~10 µl 20x SYBR Green II RNA stain (2 µl ml-1 of 10.000x SYBR Green II stock),

covered with glass cover slips and sealed with nail polish to avoid evaporation. Slides

were stored in the dark at 4 °C until analyzed. Adherent cells were counted by

Page 134: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

124

fluorescence microscopy using a Zeiss Axiovert 200M epifluorescence microscope (Carl

Zeiss GmbH, Jena, Germany) equipped with Zeiss filter set 10 and 63x oil immersion

objective. Cells were counted in 109 µm2 grids on triplicates of sample on a total of

fifteen random positions on each of the surface type.

III. RESULTS AND DISCUSSION

A. Extent of addition and coverage in backfilled monolayer

XPS was used to monitor the chemical changes occurring on Au substrates

functionalized with PEG-COOH upon exposure to OEG3 or R molecules. The respective

relative elemental and chemical species calculated from survey and high resolution XPS

scans are summarized in Table 1. The substrate photoelectrons are attenuated by the

overlayer and hence the substrate signal has an inverse relationship with overlayer

coverage as shown in Eq- 1. The Au 4f signal was used to obtain quantitative data on the

addition of molecules to the PEG-COOHSAM. The Au 4f signal of PEG-COOHSAM+OEG3

is 3.6 % lower than that of PEG-COOHSAM, indicating that OEG3 molecules have added

to the PEG-COOHSAM. From the chemical structure of OEG3 and R molecules it can be

observed that they have lower O/C and higher alkane (285.0 eV)/PEG (286.7 eV) ratios

when compared to PEG-COOH and hence any addition during backfilling results in a

corresponding change in the detected O/C and alkane/PEG ratios. The alkane/PEG ratio

was calculated from the peak areas of components corresponding to binding energies

285.0 eV (alkane) and 286.7 eV (PEG) obtained from high resolution C 1s spectra shown

in supplementary information Figure S1a. Addition of OEG3 is further supported by a

decrease in O/C ratio from 0.49 to 0.42 and increase in alkane/PEG ratio from 0.1 to 0.3

as shown in Table 1. The PEG-COOHSAM+R system exhibits a decrease in O/C ratio

Page 135: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

125

from 0.49 to 0.40 and increase in alkane/PEG ratio from 0.1 to 0.4 similar to PEG-

COOHSAM+OEG3 except for the Au signal which increased by 2.6 %. The increase in

substrate signal indicates a decrease in overlayer thickness that could be caused by

desorption of some PEG-COOH molecules during backfilling by R molecules.

Based on O/C ratios obtained from XPS of PEG-COOHSAM, OEG3-SAM, and RSAM,

a change in O/C ratio as a function of relative coverage of PEG-COOH and OEG3 or R

was plotted assuming a linear relationship (Figure 2). By correlating the experimentally

obtained values of O/C ratios for PEG-COOHSAM+OEG3 and PEG-COOHSAM+R, we

estimate the coverage of OEG3 or R on the backfilled PEG-COOHSAM surface to be 29%

and 18% respectively. It must be noted that this estimation assumes that the

intermolecular spaces in PEG-COOHSAM ideally are free for any contamination free,

which in real laboratory conditions is not the case and thus we overestimate the number

of added molecules. Here, however, we mainly focus on the relative differences rather

than the absolute values. Despite the greater number of OEG3 molecules being added to

the PEG-COOHSAM, the decrease in carboxyl peak area (Table 1) in C1s spectra of PEG-

COOHSAM+OEG3 and PEG-COOHSAM+R are comparable. This suggests that desorption

of PEG-COOH is lower when OEG3 is added in comparison to when R is added. It is

known that relative atomic percentage of S from SAMs of PEG thiol depends not only on

the stoichiometric ratio but also on the extent of dehydration and collapse of brushes

during grafting of chains as described by Larry D. Unsworth et al. 8. The amount of S in

PEG-COOHSAM was 2.1 %, which is higher than the corresponding stoichiometric ratio

of <1%, very similar to what was reported by Unsworth et al. 8 with hydroxyl terminated

2 kDa PEG thiol molecules under cloud point conditions. Though R and OEG3 molecules

Page 136: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

126

have S/C ratios of 0.11 and 0.045 respectively, which is higher than the ratio for PEG-

COOHSAM (0.008), the backfilling of PEG-COOHSAM with either R or OEG3 did not

significantly change the relative atomic percentage of S (Table 1). Hence the S content

was not helpful in providing additional information about the backfilling process. The

percentages of bound thiol on functionalized surfaces were determined as described

above in the methods section 2.3. The amount of bound sulfur was found to increase by a

small extent (increase of 3.5%) upon backfilling PEG-COOHSAM with R, and for OEG3 it

increased by 1.6%. This small increase in bound sulfur could be due to Au-thiol bond

formation of backfilled molecules, also indicating that a significant proportion of added

molecules have chemisorbed to the gold surface and not physically adsorbed. The

deconvoluted high resolution S2p spectra of various samples are included in the

supplementary information (Figure S1b).

B. Arrangement of backfilled molecules

As discussed earlier, backfilling process could result in mixed monolayer with two

possible arrangements namely underbrush and island. We used contact angle based

surface energy measurements to find out the arrangement of backfilled molecules.

Contact angle measurements have very high surface sensitivity (<1nm) 24 and allow the

determination of the surface energy of materials 22. The components of surface tension

can be calculated by measurement of contact angles of three different liquids using Eq- 4

22. The contact angles measured for various surfaces using three liquids are given in the

supplementary information (Table S1b). The surface tension components; Lifshitz van

der Waals component (γSd), electron acceptor component (γS

), electron donor component

(γS−) for various surfaces are shown in Table 2. The hydrophobicity of a surface is

Page 137: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

127

determined by its γSd value, and the lower the value, the more hydrophobic a surface is.

Since alkane thiol molecules are hydrophobic a SAM of alkane thiol would have lower

γSd value than PEG or OEG based SAMs and this is apparent for the values determined

for RSAM and OEG3-SAM (Table 2). If alkane thiol molecules form islands instead of being

homogeneously mixed during the backfilling of the PEG-COOHSAM, then the γSd value of

PEG-COOHSAM+R would significantly decrease. The γSd value 44.0 mJ/m2 for PEG-

COOHSAM+R is virtually the same as that of PEG-COOHSAM that has a γSd value 44.0

mJ/m2 indicating that hydrophobic islands are not present. The γS− value of a surface

gives a quantitative idea about the presence of electron donor groups, with the greater the

value of γS− the higher the number of surface electron donor groups. PEG and OEG have

oxygen atoms and thus have high γS− values when compared to the alkane thiol (Table 2).

Backfilling PEG-COOHSAM with R results in a 7 mJ/m2 decrease in γS− indicating that a

significant number of PEG-COOH molecules have desorbed from the surface. The values

for γSd and γS

− for PEG-COOHSAM after addition of OEG3 do not change significantly

since both molecules have very similar values and could not easily be followed by

contact angle measurements.

C. AFM results

QNM-AFM has a very high lateral resolution and has been used to characterize the

surface morphology of the different SAM layers with the aim to check the presence of

phase separated domains. With the use of the peak forces less than 1 nN, QNM AFM is

able to map mechanical properties of surfaces quantitatively while simultaneously

capturing topographic images with the same resolutions as traditional tapping mode AFM

Page 138: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

128

25 23. All QNM-AFM measurements were done liquid condition. The topography and

modulus images of Au surface and various SAM are shown in Figure 3 (Figure 3), and

the corresponding surface roughness is summarized in Table 3. The topography of the

bare Au surface (Figure 3a) shows Au grains without any additional features on top of

grains. The recorded nanomechanical map of Au surface (Figure 3a’) demonstrates the

homogeneous Young’s modulus distribution as the whole image has nearly the same

range of stiffness showing the same color contrast. Comparing the topography images of

modified surfaces with the bare Au, one can clearly see the additional particle-like

features on all the modified surfaces (Figure 3b-f). In addition, the modulus images of the

modified surfaces show that many of the grain boundaries are slightly invisible due to the

SAMs. Furthermore, the surface roughness has been changed dramatically after

modification. Hence these evidences prove the successful formation of the SAMs.

Interestingly, the modulus of the modified surfaces also represents the surface modulus

variations, because of the slightly different color contrast (Figure 3b’-3f’). The small

variations in material properties observed in elastic modulus measurements are

particularly useful in the analysis of heterogeneous surface. However most importantly,

phase separations were not observed in all of images. This indicates that the backfilled

surfaces are homogeneous. Hence we conclude that the SH-R-OEG3 or SH-R molecules

form underbrush upon addition to SH-PEG-COOHSAM.

D. Non-fouling properties of PEG-COOH surfaces with and

without underbrush

Resistance towards non specific adsorption can be tested using simple model

systems such as single protein solutions. Examples have included collagen 26 and bovine

Page 139: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

129

serum albumin 27. More rigorous tests are conducted with serum27 or microbial cell

suspensions28. The non-fouling properties of the PEG-COOHSAM and mixed surfaces

were evaluated by serum adsorption and bacterial adhesion studies. Addition of smaller

OEG chains in theory increases the graft density protein resistance molecules within the

layer of diffuse PEG-COOHSAM molecules and hence is expected to improve its

resistance towards non specific adsorption.

QCM and XPS have been employed to quantitatively compare the serum adsorption

resistance properties of the SAMs. The frequency shift after serum exposure in QCM is

directly proportional to the amount of protein adsorbed 27 , and the relative atomic

percentage of nitrogen from XPS also enables a quantitative estimation of the amount of

adsorbed protein 26. In Figure 4a we show the data points corresponding to various

samples studied, where the X-axis shows the % N obtained from XPS and the Y-axis

shows the frequency shift from the QCM data. Upon serum exposure the QCM frequency

decreases by 7Hz for the PEG-COOHSAM+OEG3 surface, compared to the PEG-

COOHSAM and PEG-COOHSAM+R surfaces, which both exhibit a frequency shift of

about 15Hz. The lower frequency shift for PEG-COOHSAM+OEG3 proves that OEG3

addition improves the non-fouling abilities of PEG-COOHSAM surfaces. We also

speculate that protein adsorption would be lower if the PEG thiol molecules used did not

have a carboxyl end-group, increasing electrostatic interactions with oppositely charged

serum proteins and facilitating higher adsorption. Representative QCM kinetic plots are

included in the supplementary information (Figure S2). Figure 4a also shows the nitrogen

content on the X-axis, where the PEG-COOHSAM+OEG3 surface has 2% N compared to

PEG-COOHSAM and PEG-COOHSAM+R surfaces both showing approximately 5% N,

Page 140: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

130

which follows the same trend as seen with QCM. The OEG3-SAM had the highest ability to

resist serum adsorption which agrees with previous studies with OEG based SAMs 18.

The surfaces were tested for bacterial adhesion using a Staphylococcus aureus strain and

the results are shown in Figure 4b. Clearly, the PEG-COOHSAM+OEG3 (4.2 × 107

cells/cm2) surface is better at resisting bacterial adhesion when compared to PEG-

COOHSAM (7.2 × 107 cells/cm2) and PEG-COOHSAM+R (9.2 × 107 cells/cm2). Hence it

can be concluded that adding OEG3 molecules improve the non-fouling properties of the

PEG-COOHSAM. Though the OEG3-SAM has the least bacterial adhesion and serum

adsorption, we propose that PEG-COOHSAM+OEG3 system would be a better system for

applications involving immobilization of bioactive molecules such as enzymes and

further advantages of PEG-COOHSAM+OEG3 will be discussed in the next section.

E. Discussion

We have studied mixed monolayers of OEG3 on PEG-COOHSAM using XPS,

contact angle and AFM. The contact angle results support the notion that phase separated

surface arrangement of the two molecules does not occur proving that homogeneous

mixed layers are formed. There have been some reports on grafting 2 kDa PEG thiol

chains. For example Unsworth et al. 8 reported a maximum graft density of 0.58

chains/nm2 using methoxy capped 2 kDa PEG thiol chains under cloud point conditions,

and Tokumitsu et al. 29 reported a maximum graft density of 3.6 chains/nm2 using 2 kDa

PEG conjugated to undecane thiols. From our XPS data we have calculated a graft

density of PEG-COOHSAM to be 1.0 chain/nm2 using Eq 1-3. Thus, the PEG-COOH

chains in PEG-COOHSAM have a density that is not close-packed and there exists

Page 141: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

131

sufficient space for the smaller OEG molecules to fill the gaps, although in a good

solvent the PEG is likely to extend and form highly mobile brushes 8.

We used Carboxyl capped PEG molecules since we intend to use them for

immobilization of antibacterial enzymes in future studies. These molecules are expected

to contribute to the surface negative charge. We suspect that the non-fouling properties

would be better with hydroxyl or methoxy capped PEG molecules, which are neutrally

charged and thus minimize electrostatic interactions. Our mixed PEG-COOHSAM+OEG3

system is anticipated to be an ideal platform for immobilization of bioactive molecules,

since the OEG maximizes non-fouling potential while the longer PEG chains would

confer higher mobility and thus improved bioactivity capabilities to the immobilized

molecule 19. There have been reports on bacterial adhesion studies on OEG 30 and PEG 28

surfaces. Our adhesion studies with Staphylococcus aureus involved different incubation

time compared to that used by Nejadnik et al., also Ostuni et al. used a counting

technique involving removal of attached cells by sonication followed by counting the

colony forming units on culture plates that introduces possibility of 10 fold error in

estimation as mentioned by authors. Hence the absolute values of our bacterial adhesion

studies could not be compared with these earlier reported studies. We plan to immobilize

enzymes on backfilled PEG-COOHSAM+OEG3 surfaces and further understand the role of

backfilled OEG molecules.

IV. SUMMARY AND CONCLUSIONS

We have investigated backfilling of a PEG-COOHSAM with alkane and oligo

ethylene glycol terminated thiol molecules. From XPS and contact angle results we have

Page 142: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

132

shown that R molecules form mixed layers and the addition process is accompanied by

desorption of PEG-COOH molecules. OEG3 molecules, upon addition, also formed mixed

layers but less desorption of the PEG occurs, when compared to addition of R molecules.

The non-fouling properties of various surfaces were compared, and it was shown that the

PEG-COOHSAM+OEG3 surface found to be better than the PEG-COOHSAM surface with

respect to resisting serum adsorption and bacterial adhesion. Thus, the strategy involving

backfilling using OEG terminating alkane thiols is indeed capable of improving the non-

fouling properties of monolayers of PEG chains of significant graft densities.

ACKNOWLEDGMENTS

We gratefully acknowledge the Danish Strategic Research Council (grant no.

2106-07-0013), Alfa Laval, the Lundbeck Foundation and the Carlsberg Foundation for

financial support. The authors acknowledge Uffe B. Skov Sørensen, Associate Professor,

Institute for Medical Microbiology and Immunology, Aarhus University for providing S.

aureus strain and lab space to carry out the bacterial adhesion study.

1. D. G. Castner and B. D. Ratner, Surface Science 500 (1-3), 28-60 (2002).

2. I. Banerjee, R. C. Pangule and R. S. Kane, Advanced Materials 23 (6), 690-718 (2010).

3. K. Sauer, A. K. Camper, G. D. Ehrlich, J. W. Costerton and D. G. Davies, (2002), Vol. 184, pp. 1140-1154.

4. S. I. Jeon, J. H. Lee, J. D. Andrade and P. G. De Gennes, Journal of Colloid and Interface Science 142 (1), 149-158 (1991).

Page 143: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

133

5. S. J. Sofia, V. Premnath and E. W. Merrill, Macromolecules 31 (15), 5059-5070 (1998).

6. L. D. Unsworth, H. Sheardown and J. L. Brash, Biomaterials 26 (30), 5927-5933 (2005).

7. L. D. Unsworth, H. Sheardown and J. L. Brash, Langmuir 24 (5), 1924-1929 (2008).

8. L. D. Unsworth, Z. Tun, H. Sheardown and J. L. Brash, Journal of Colloid and Interface Science 281 (1), 112-121 (2005).

9. P. Kingshott, J. Wei, D. Bagge-Ravn, N. Gadegaard and L. Gram, Langmuir 19 (17), 6912-6921 (2003).

10. N. Luo, J. B. Hutchison, K. S. Anseth and C. N. Bowman, Macromolecules 35 (7), 2487-2493 (2002).

11. P. Kingshott, H. Thissen and H. J. Griesser, Biomaterials 23 (9), 2043-2056 (2002).

12. W. Taylor and R. A. L. Jones, Langmuir 26 (17), 13954-13958 (2010).

13. S. Minko, S. Patil, V. Datsyuk, F. Simon, K.-J. Eichhorn, M. Motornov, D. Usov, I. Tokarev and M. Stamm, Langmuir 18 (1), 289-296 (2002).

14. K. Uchida, Y. Hoshino, A. Tamura, K. Yoshimoto, S. Kojima, K. Yamashita, I. Yamanaka, H. Otsuka, K. Kataoka and Y. Nagasaki, Biointerphases 2 (4), 126-130 (2007).

15. K. Uchida, H. Otsuka, M. Kaneko, K. Kataoka and Y. Nagasaki, Analytical Chemistry 77 (4), 1075-1080 (2005).

16. T. Kakiuchi, K. Sato, M. Iida, D. Hobara, S.-i. Imabayashi and K. Niki, Langmuir 16 (18), 7238-7244 (2000).

17. G. G. Baralia, A.-S. Duwez, B. Nysten and A. M. Jonas, Langmuir 21 (15), 6825-6829 (2005).

18. P. Harder, M. Grunze, R. Dahint, G. M. Whitesides and P. E. Laibinis, Journal of Physical Chemistry B 102 (2), 426-436 (1998).

19. H. Otsuka, Y. Nagasaki and K. Kataoka, Langmuir 20 (26), 11285-11287 (2004).

20. D. G. Castner, K. Hinds and D. W. Grainger, Langmuir 12 (21), 5083-5086 (1996).

21. P. J. Cumpson, Surface and Interface Analysis 31 (1), 23-34 (2001).

22. C. J. Van Oss, M. K. Chaudhury and R. J. Good, Chemical Reviews 88 (6), 927-941 (1988).

23. K. Sweers, K. van der Werf, M. Bennink and V. Subramaniam, Nanoscale Research Letters 6 (1), 270 (2011).

24. E. B. Troughton, C. D. Bain, G. M. Whitesides, R. G. Nuzzo, D. L. Allara and M. D. Porter, Langmuir 4 (2), 365-385 (1988).

25. E. N. Pittenger B, Su C, Application Note Veeco Instruments Inc.

26. M. A. Cole, H. Thissen, D. Losic and N. H. Voelcker, Surface Science 601 (7), 1716-1725 (2007).

Page 144: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

134

27. B. Menz, R. Knerr, A. Göpferich and C. Steinem, Biomaterials 26 (20), 4237-4243 (2005).

28. M. R. Nejadnik, H. C. van der Mei, W. Norde and H. J. Busscher, Biomaterials 29 (30), 4117-4121 (2008).

29. S. Tokumitsu, A. Liebich, S. Herrwerth, W. Eck, M. Himmelhaus and M. Grunze, Langmuir 18 (23), 8862-8870 (2002).

30. E. Ostuni, R. G. Chapman, M. N. Liang, G. Meluleni, G. Pier, D. E. Ingber and G. M. Whitesides, Langmuir 17 (20), 6336-6343 (2001).

Table 1. Summary of XPS results. Au % - relative atomic percentage of Au. O/C - ratio

of relative atomic percentage oxygen and carbon. Alkane (285.0 eV)/PEG (286.7 eV) -

ratio of alkane and PEG components in C 1s high resolution spectra. S % - relative

atomic percentage of S. % bound S - percentage of bound sulfur from components in S

2p high resolution spectra.

Sample PEG-

COOHSAM

OEG3-SAM RSAM PEG-

COOHSAM +R

PEG-COOHSAM

+ OEG3

Au % 39.0±0.4 32.6±0.4 55.3±2.4 41.6±0.5 36.2±0.5

O/C 0.49±0.03 0.25±0.01 0.0±0.0 0.40±0.01 0.42±0.01

Alkane/PEG 0.1±0.0 0.8±0.0 21.0±0.0 0.4±0.0 0.3±0.0

S % 2.1±0.2 1.5±0.2 3.2±0.4 2.1±0.2 1.9±0.6

% bound S 78.3±0.7 80.7±1.0 88.1±1.0 81.8±1.0 79.9±0.7

Table 2. Surface tension components: the Lifshitz van der Waals component (γSd),

electron acceptor component (γS+), and electron donor component (γS

−) determined from

the surfaces of OEG3-SAM, RSAM, PEG-COOHSAM, PEG-COOHSAM+R and PEG-

COOHSAM+OEG3.

Page 145: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

135

Sample γSd mJ/m2 γS

+ mJ/m2 γS− mJ/m2

OEG3-SAM 43.45±0.06 0.85±0.05 45.66±1.49

RSAM 30.67 1.27 0.01±0.01 0.69±0.26

PEG-COOHSAM 44.24±0.04 0.92±0.05 43.97±1.56

PEG-COOHSAM+R 44.05±0.08 0.96±0.03 36.58±0.80

PEG-COOHSAM+OEG3 44.07±0.1 0.90±0.05 42.90±1.59

Table 3: Normalized Surface roughness (to Au) values for OEG3-SAM, RSAM, PEG-

COOHSAM, PEG-COOHSAM+R and PEG-COOHSAM+ OEG3. SqSAM/SqAu stands for ratio

of the means of the two surface roughness; SqAu= 0.48±0.02nm

Sample Roughness (Sq) (nm) SqSAM/SqAu

PEG-COOHSAM 0.69±0.04 1.44

OEG3-SAM 0.63±0.04 1.31

RSAM 0.21±0.02 0.44

PEG-COOHSAM+OEG3 0.81±0.05 1.69

PEG-COOHSAM+R 0.66±0.06 1.38

Figure Captions

Figure 1. (Colour online only) The schematic diagrams of back filled PEG SAM in island

type (a) and underbrush type arrangements (b).The chemical formula and assigned

abbreviations for various molecules used in self assembly and backfilling experiments

(c).

Figure 2. (Colour online only) Relative coverage of PEG-COOH and R or OEG3 on

backfilled surfaces as determined by correlating their XPS O/C ratios (on X axis) to a

plot obtained by extrapolating values of O/C ratio for 0% and 100% relative coverage (on

Y axis) assuming a linear relationship. The data points and + shown in the plot

Page 146: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

136

represent the experimentally obtained O/C ratios for PEG-COOHSAM+OEG3 and PEG-

COOHSAM+R respectively.

Figure 3. (Colour) Topography and Young’s modulus images recorded by QNM-AFM

measurements in MQ water. (a, a’) Au substrate, (b, b’) PEG-COOHSAM, (c, c’) OEG3-

SAM, (d, d’) RSAM, (e, e’) PEG-COOHSAM+OEG3, and (f, f’) PEG-COOHSAM+R. The scan

area of each image is 1µm×1µm.

Figure 4. (Colour) Non-fouling properties of the surfaces. (a) Shows XPS (X-axis – N %)

and QCM (Y-axis – frequency shift) based quantification after exposure to 10% FBS

adsorption, to PEG-COOHSAM, PEG-COOHSAM+R and PEG-COOHSAM+OEG3,

OEG3-SAM and RSAM surfaces. (b) Shows results of bacterial adhesion studies using

Staphylococcus aureus performed on PEG-COOHSAM, PEG-COOHSAM+R and PEG-

COOHSAM+OEG3, OEG3-SAM, RSAM. The X-axis shows the number of adherent cells on

the Log scale.

Figures

Table of contents image

Page 147: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

137

Figure 1.

Figure 2.

Page 148: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

138

Figure 3.

Page 149: Strategies for creating antifouling surfaces using self …pure.au.dk/.../files/40363364/Arcot_thesis_iNANO_School.pdf · 2011. 9. 28. · of bio-interfacial phenomena at the molecular

139

Figure 4.