NOVEL BIOMEDICAL APPLICATIONS

OF MALDI AND ELECTROSPRAY

MASS SPECTROMETRY

by

Kim Yoke Ching Fung

A dissertation submitted in fulfillment of the requirements for the degree of Doctor of Philosophy

University of New South Wales Sydney, Australia 2001 I hereby declare that this submission is my own work and to the best of my knowledge it contains no material previously published or written by another person, nor material which to a substantial extent has been accepted for the award of any other degree or diploma at UNSW or any other educational institution, except where due acknowledgement is made in the thesis. Any contribution made to the research by others, with whom I have worked at UNSW or elsewhere, is explicitly acknowledged in the thesis.

I also declare that the intellectual content of this thesis is the product of my own work, except to the extent that assistance from others in the project's design and conception or in style, presentation and linguistic expression is acknowledged.

December, 2001 ACKNOWLEDGEMENTS

I would like to express my appreciation to the following people:

• Prof. Mark Duncan for the support, advice and guidance with every aspect of my degree and for the many opportunities presented to me over the past four years.

• Dr. George Smythe for being my co-advisor and for the support you have given me throughout my degree.

• Dr. Carol Morris and Dr. Robert Sack for providing the tear samples and for your advice and insight regarding the data generated.

• Dr. David Friedman for your help with the prostate fluid experiments.

• Martin Bucknall for your help with the studies on quantitative MALDI.

• Dr. Joseph Zirrolli for your day-to-day support, and for sharing your vast knowledge of mass spectrometry with me.

• Staff and fellow students (past and present) from the labs I have interacted with during the course of my work for the great times experienced both in the lab and socially.

Finally, I would like to thank my family and close friends for their continual love and support as well as for all the good times we have shared. ABSTRACT

Over the last decade, developments in mass spectrometry have made the analysis of high molecular weight involatile biomolecules, including peptide and proteins, a reality.

Concurrent expansions in protein and genomic databases, as well as progress in bioinformatics, have enabled proteins to be unambiguously identified using sensitive mass spectrometric techniques such as matrix assisted laser desorption ionisation

(MALDI) and electrospray ionisation (ESI).

This thesis investigates the application of mass spectrometry, in combination with techniques such as gel electrophoresis and liquid chromatography, for the analysis of peptides and proteins in biological fluids. Peptides and proteins in human tear fluid were characterised using a combination ofMALDI-TOFMS and ESI-LC/MS/MS. Direct analysis of tear fluid by MALDI-TOFMS detected over 30 peptides and proteins and

MALDI-PSD identified homologous peptides derived from the C-terminus oflacrimal -rich protein. ESI-LC/MS/MS of tryptic peptides identified many more proteins, including known constituents of human tear and proteins previously not reported as components of the tear film.

Seminal fluid proteins were separated by gel electrophoresis and identified by peptide mass fingerprinting. Although over 300 protein spots were detected, only 32 unique proteins were identified including semenogelin I, semenogelin II, prostate specific

antigen and prostate secreted seminal plasma protein. The remaining proteins were

identified as protein isoforms or modified proteins. Additional analysis by MALDI- TOFMS and ESI-LC/MS/MS identified many proteolytic products of semenogelin I and

semenogelin II.

The utility of mass spectrometry for the quantification of proteins and other biomolecules was also explored. The relative abundance of more than 20 proteins in open and closed eye tear fluid was measured using ESI-LC/MS following esterification of proteolytic peptides with isotopomers of butanol.

Also, the potential ofMALDI-TOFMS for the absolute quantification of various biomolecules is explored. Rat growth hormone, human insulin, LW-hemorphin- 7, the

catecholamines epinephrine and norepinephrine, and homovanillic acid were quantified

in biological tissue or fluid using stable isotopomers or structural homologues as

internal standards. The role of mass spectrometry in global protein identification and

quantification is also discussed. TABLE OF CONTENTS

LIST OF FIGURES ••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••• V

LIST OF TABLES •••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••• VIII

CHAPTER 1: INTRODUCTION 1

1.1. Introduction .•.••.•.••.•.••.•.••..•.•.•••.•...... •...... •...•.••.•.••.•..••.••.•.••.••..•.••.•.....••.••..•.••.••.. 2 1.2. Matrix assisted laser desorption ionisation ...... 3 1.3. Electrospray mass spectrometry ...... 10 1.4. Ion dissociation and tandem mass spectrometry ...... 16 1.5. MALDI and ESI in the biological sciences ...... 19 1.5.1. Quantification of biomolecules by mass spectrometry ...... 19 1.5.2. Analysis ofbiomolecules in complex mixtures ...... 21 1.6. The emergence of Proteomics ...... 25 1.7. Aims ••••••••••••••••••••••••••••••••••••.•.••.•.••..•..•.•.••.••.••••.••.•.••.•.••.•.••.•.••..••••.••.•.•••••••••••••••••••.•• 27

CHAPTER 2: METHODS 31

2.1. Instrumentation················································~······················································ 32 2.2. MALDI matrices •.••••••••••••••••••••••••••••••••••••••••••••••••••••••••.•.•••••••••••.••••.•.••.•.••.•.•••••••••••• 33 2.3. Proteolytic en.zymes •.••••••.•••••.•.••.•.••.•.••.••.•.••.•.•.••...•••.••••••••••••••••.••••••••..••••.••••.•.••••••• 33 2.4. Protein identification criteria .••.•.••.•.••.••.•.•••••••••••••••.••.•.••.•..•.•.••.•..••••..•••.••••••••••••••• 34 2.4.1. MALDI-TOF mass spectrometry ...... 34 2.4.2. Electrospray mass spectrometry ...... 34 2.S. Database search criteria •.••..••.•.•••••••••••••....•.•.•••••••••••••••.••.•.••.•.••.•.••.•.••.•.••.•.••.•.•..•.• 35 2.6. Identification of protein constituents in human tear ...... 36 2.6.1. Sample collection ...... 36 2.6.2. Analysis of whole tear sample...... 36

i 2.6.2.1. MALDI analysis ...... 36 2.6.2.2. Electrospray mass spectrometry...... 37 2.6.3. Isolation and detection oflacrimal proline-rich protein ...... 37 2.6.4. Quantification of tear fluid proteins ...... 38 2.6.4.1. Apomyoglobin ...... 38 2.6.4.2. Tear fluid proteins ...... 39 2. 7. Analysis of the peptide and protein components in human seminal plasma ..... 41 2. 7 .1. Sample collection ...... 41 2.7.2. Two dimensional gel electrophoresis ...... 41 2.7.3. One dimensional gel electrophoresis ...... 42 2.7.4. Mass spectrometry ofunfractionated seminal fluid ...... 43 2. 7 .5. Protein identification ...... 43 2.7.5.1. MALDI-TOF mass spectrometry ...... 43 2.7.5.2. ESI mass spectrometry ...... 44 2.8. Quantification of biomolecules by MALDI-TOFMS ...... 45 2.8.1. Analysis of growth hormone in rat pituitary tissue ...... 45 2.8.2. Quantification of insulin in human pancreatic tissue ...... 45 2.8.3. Analysis ofhemorphins in human adrenal and pheochromocytoma tissues ...... 46 2.8.4. Analysis of catecholamines in human adrenal and pheochromocytoma tissue ...... 47 2.8.5. Analysis ofhomovanillic acid in human urine samples ...... 48 2.8.6. Data acquisition and processing ...... 49

CHAPTER 3: MASS SPECTROMETRY AND PROTEIN IDENTIFICATION:

APPLICATION TO HUMAN TEAR FLUID 50

3.1. Introduction ...... 51 3.2. Methods ...... 53 3.3. Results ...... 54 3.3 .1. The peptide components of human open eye tear ...... 54 3.3.2. The detection and identification oflacrimal proline-rich protein ...... 76 3.3.3. Analysis ofunfractionated human tear proteins by ESI-LC/MS/MS ...... 80 3.4. Discussion ...... 108

11 CHAPTER 4: COMPREHENSIVE PEPTIDE AND PROTEIN IDENTIFICATION:

APPLICATION TO THE ANALYSIS OF HUMAN SEMINAL FLUID 111

4.1. Introduction ...... 112 4.2. Methods ...... •...... 115 4.3. Results ...... 115 4.3.1. Detection and identification of proteins by two dimensional gel electrophoresis 115 4.3.2. Detection and identification of proteins by one dimensional gel electrophoresis 130 4.3.3. Direct analysis of seminal fluid by MALDI-TOFMS and ESI-MS ...... 145 4.4. Discussion ...... 153

CHAPTER 5: QUANTIFICATION OF BIOMOLECULES BY MALDI-TOF MASS

SPECTROMETRY 157

S.1. Introduction ...... 158 5.2. Methods ...... 160 5.3. Results ...... 161 5.3.1. Quantification of proteins in biological tissue ...... 161 5.3.2. Quantification of peptides in biological samples ...... 166 5.3.2.1. Insulin in human pancreatic tissue ...... 166 5.3.2.2. LVV-hemorphin-7 in human adrenal gland ...... 173 5.3.3. Quantification oflow mass analytes in biological samples ...... 175 5.3.3.1. Catecholamines in human adrenal gland ...... 175 5.3.3.2. Homovanillic acid in human urine ...... 178 5.4. Discussion ...... 182

iii CHAPTER 6: ALTERNATIVE APPROACHES TO PROTEIN QUANTIFICATION 187

6.1. Introduction ...... 188 6.2. Methods ...... 192 6.3. Results ...... 193 6.3.1. Detn.onstration of the approach ...... 193 6.3.2. Identification of esterified peptides in open and closed eye tear...... 200 6.3.3. Relative quantification of proteins in open and closed eye tear fluid ...... 203 6.4. Discussion ...... 211

CHAPTER 7: DISCUSSION 214

7.1. Discussion ...... 215 7.2. Protein identification strategies ...... 216 7 .2.1. Two dimensional gel electrophoresis and peptide mass fingerprinting ...... 218 7.2.2. Direct analysis of proteins by ESI-LC/MS/MS ...... 220 7.2.3. Detection and identification of endogenous peptides in biological samples ...... 222 7.3. Characterising protein post-translational modifications ...... 224 7 .3 .1. Detection and isolation of modified proteins ...... 225 7.3.2. Identifying post-translational modifications ...... 227 7 .4. Protein quantification ...... 229 7.4.1. Relative quantification of proteins ...... 230 7.4.2. Quantification of intact proteins ...... 232 7.5. Alternative technologies for proteomic studies ...... 234 7.6. Conclusions ...... 237 7.7. Summary ...... 238

CHAPTER 8: REFERENCES 245

References ...... 246

iv LIST OF FIGURES

CHAPTERl Figure 1.1. A schematic diagram of the MALDI ionisation process...... 5 Figure 1.2. A schematic diagram of a MALDI-TOF mass spectrometer with delayed extraction...... 9 Figure 1.3. A schematic diagram of the electrospray ionisation process...... 12 Figure 1.4. A comparison ofESI and MALDI spectra of horse apomyoglobin...... 13

CHAPTER3 Figure 3.1. A MALDI-TOF mass spectrum of human tear...... 55 Figure 3.2. Post-source decay spectra for the abundant tear peptides...... 57 Figure 3.3. The sequence for the intact lacrimal proline-rich protein...... 75 Figure 3.4. An example of a MALDI-TOF mass spectrum for one of the SEC fractions of human tear fluid ...... 77 Figure 3.5. Amino acid coverage of the lacrimal proline-rich protein following its isolation by size exclusion chromatography...... 79 Figure 3.6. The base peak chromatogram of the tryptic peptides detected in human tear plotted over four distinct mass ranges ...... 81

CHAPTER4 Figure 4.1. An annotated silver-stained two dimensional gel of seminal fluid proteins separated over the pi range 4 - 7 ...... 116 Figure 4.2. An annotated silver-stained two dimensional gels of seminal fluid proteins separated over the pi range 6 - 11 ...... ·...... 117 Figure 4.3. Base peak chromatogram for 4 different regions of the 1D SDS-PAGE gel of seminal fluid proteins ...... 131 Figure 4.4. The amino acid sequences of the histone 2A family of proteins and the tryptic peptides that were detected by ESI-LC/MS/MS ...... 141

V Figure 4.5. The amino acid sequences of the histone 2B family of proteins and the tryptic peptides that were detected by ESI-LC/MS/MS ...... 143 Figure 4.6. A MALDI mass spectrum ofunfractionated seminal fluid ...... 146 Figure 4.7. The amino acid sequence of semenogelin I showing those peptides identified by ESI-LC/MS/MS ...... 149 Figure 4.8. The amino acid sequence of semenogelin II showing those peptides identified by ESI-LC/MS/MS ...... 150

CHAPTERS Figure 5.1. A representative MALDI mass spectrum for growth hormone in a rat pituitary extract...... 163 Figure 5.2. A representative standard curve obtained for methionyl rat growth hormone without an internal standard ...... 164 Figure 5.3. A representative standard curve obtained for methionyl rat growth hormone using ~-lactoglobulin as the internal standard ...... 165 Figure 5.4. Representative MALDI-TOF mass spectra for the analysis of a mixture of human insulin and porcine insulin...... 167 Figure 5.5. The amino acid sequence for human and porcine insulin...... 169 Figure 5.6. Representative standard curves obtained for human insulin using porcine insulin as the internal standard ...... 170 Figure 5. 7. A comparison of LVV-hemorphin- 7 concentrations in human adrenal gland determined by MALDI-TOFMS and ESI-MS ...... 174 Figure 5.8. A representative MALDI-TOF mass spectrum for norepinephrine, epinephrine and their respective internal standards in human adrenal extracts...... 17 6 Figure 5.9. A representative MALDI mass spectrum for homovanillic acid detected in human urine ...... 179 Figure 5.10. Homovanillic acid concentrations in human urine determined by MALDI­ TOFMS vs homovanillic acid values determined by a commercial clinical laboratory ...... 180

VI CHAPTER6

Figure 6.1. A schematic showing the addition of a butyl ester to the C-terminus of a tryptic peptide...... 195 Figure 6.2. A representative MALDI mass spectrum of horse apomyoglobin following digestion with trypsin...... 196

Figure 6.3. A MALDI-TOF mass spectrum showing do-butylated and d10-butylated tryptic peptide pairs derived from the proteins present in open and closed eye tear respectively...... 201 Figure 6.4. An example of a fragment ion spectrum for an esteri:fied tryptic peptide detected in open eye tear...... 202 Figure 6.5. An example of a mass spectrum of esterified open and closed eye tear showing at least three different co-eluting tryptic peptide pairs ...... 204 Figure 6.6. The extracted total ion chromatogram for one of the co-eluting peptide pairs at a retention time window of 60 - 62 minutes ...... 206

vu LIST OF TABLES

CHAPTER2 Protein database search criteria ...... 35

CBAPTER3 Table 3.1. Fragment ions detected in the post-source decay spectra of the peptide [M+H]+ 1,629.69 Da...... 59 Table 3.2. Fragment ions detected in the post-source decay spectra of the peptide [M+H]+ 1,443.74 Da...... 62 Table 3.3. Fragment ions detected in the post-source decay spectra of the peptide [M+H]+ 1,330.52 Da...... 65 Table 3.4. Fragment ions detected in the post-source decay spectra of the peptide [M+H]+ 1,105.51 Da...... 68 Table 3.5. Fragment ions detected in the post-source decay spectra of the peptide [M+H]+ 848.37 Da...... 71 Table 3.6. The amino acid sequence of five of the most abundant peptides detected in human tear...... 74 Table 3.7. Proteins identified by ESI-LC/MS/MS that have been previously reported as components of the human tear film ...... 84 Table 3.8. Proteins that have been detected in human ocular tissue but not in the tear film ...... 93 Table 3.9. Proteins identified by ESI-LC/MS/MS that are reportedly elevated in the tear fluid of patients with various pathological conditions...... 94 Table 3.10. Proteins identified by ESI-LC/MS/MS that have been reported in the tear film of animal species other than human...... 95 Table 3.11. Proteins identified by ESI-LC/MS/MS that have not previously been reported as components of the human tear film ...... 96

Vlll CHAPTER4 Table 4.1. Proteins in seminal fluid that were identified by 2DE (pi range 4 - 7) ...... 118 Table 4.2. Co-migrating proteins in seminal fluid that were identified by 2DE (pi range 4 - 7) ...... 124 Table 4.3. Proteins in seminal fluid that were identified by 2DE (pi range 6- 11) ...... 126 Table 4.4. Co-migrating proteins in seminal fluid that were identified by 2DE (pi range 6 - 11 ) ...... 129 Table 4.S. Proteins found in seminal fluid following separation by 1D gel electrophoresis, in-gel digestion and ESI-LC/MS/MS analysis ...... 132 Table 4.6. Peptides that were detected by MALDI-TOFMS in unfractionated seminal fluid and subsequently identified by ESI-LC/MS/MS ...... 148 Table 4.7. Peptides in unfractionated seminal fluid that were identified by ESI­ LC/MS/MS and database searching...... 151

CHAPTERS Table S.1. Insulin concentrations measured in human pancreatic tissue obtained from two cadavers ...... 172 Table S.2. Concentrations of norepinephrine and epinephrine in human adrenal tissue and pheochromocytoma tissue...... 177

CHAPTER6 Table 6.1. The relative abundance oftryptic peptides derived from apomyoglobin ...... 199 Table 6.2. The relative abundances oftryptic peptides derived from tear lipocalin ...... 208 Table 6.3. Proteins that were found to be more abundant in open eye tear in comparison to closed eye tear...... 209 Table 6.4. Proteins that were found to be more abundant in closed eye tear in comparison to open eye tear...... 210

ix CHAPTER 1

INTRODUCTION

I 1.1. Introduction

Recent technical advances have established the importance of mass spectrometry in the biological sciences. In particular, the development of sensitive and versatile ionisation techniques in the late 1980s, primarily matrix assisted laser desorption ionisation

(MALDI) [Karas and Hillenkamp, 1988] and electrospray ionisation (ESI) [Whitehouse

et al., 1985], and their coupling to various mass analysers have lead to affordable, powerful and versatile analytical instruments. Prior to the development of techniques

such as plasma desorption (PDMS) and fast atom bombardment (F AB) mass

spectrometry, only molecules that were oflow mass (i.e.,< 1,000 Da), volatile, or able to be made volatile by chemical derivatisation procedures, could be analysed easily

[Macfarlane and Torgerson, 1976; Barber et al., 1981]. The introduction ofMALDI and

ESI have allowed the mass limitations of these techniques to be overcome and many

additional classes of molecules can now be analysed, including high mass biomolecules

(up to 1,000,000 Da) [Beavis and Chait, 1990; Schriemer and Li, 1996]. This has had

tremendous impact in the biological sciences because molecules such as peptides,

proteins and oligonucleotides can now be routinely examined by mass spectrometry.

Most importantly, both of these techniques, described as soft ionisation processes, result

in minimal fragmentation, and have enabled accurate intact mass determinations of

biomolecules to be performed for the first time [Karas and Hillenkamp, 1988; Fenn et

al., 1989].

The versatility of time-of-flight (TOP) and ESI mass spectrometry (ESI-MS) has

increased considerably due to factors such as the implementation of delayed extraction

(time-lag focusing) [Wiley and McLaren, 1955; Brown and Lennon, 1995; Whittal and

Li, 1995; Bahr et al., 1997; Takach et al., 1997] and incorporation of an electrostatic

2 mirror (reflectron) [Mamyrin et al., 1973; Takach et al., 1997]. These have lead to dramatic improvements in resolution and mass accuracy for MALDI sources coupled to

TOF mass analysers (MALDI-TOFMS). In addition, electrospray ionisation has enabled the introduction of samples in a liquid phase and hence has facilitated the coupling of on-line separation techniques to mass spectrometry [Banks, 1997; Niessen, 1998;

Choudhary et al., 2000; Dalluge, 2000]. Consequently, mass spectrometry has emerged as a popular and versatile analytical tool in the biomedical sciences. Utilised together with other techniques, mass spectrometry now enables molecules involved in disease processes to be characterised and potential biomarkers for diagnostic, prognostic or therapeutic purposes to be identified.

1.2. Matrix assisted laser desorption ionisation

Desorption and ionisation of molecules from solid sample supports was first described

in the 1970s by Macfarlane et al. (1976) and Posthumus et al. (1978). These processes,

known as plasma desorption and laser desorption, employed the use of radioactive

decay products of californium-252 or a laser emitter, respectively, to promote ionisation

of sample molecules. Together with the introduction of F AB in 1981 [Barber et al.,

1981 ], analysis of involatile molecules, including proteins and oligonucleotides, became

possible for the first time [Sundqvist et al., 1984a; Sundqvist et al., 1984b;

Tsarbopoulos, 1989; Tsarbopoulos et al., 1991].

Advances in and refinements of these ionisation techniques resulted in the advent of

MALDI. MALDI, first described by Karas and Hillenkamp [Karas and Hillenkamp,

1988] and independently by Tanaka and coworkers (1988), involves the co-

3 crystallisation of the analyte with a matrix followed by laser ablation of the

sample/matrix mixture. By employing a vast excess of a matrix molecule, the extensive

fragmentation observed with plasma desorption and laser desorption ionisation is overcome. Molecules employed as the matrix are typically small organic acids, such as

cinnamic acid derivatives, capable of absorbing energy at the wavelength (usually in the

UV spectrum) of the laser being used [Beavis and Chait, 1989a; Beavis and Chait,

1989c; Juhasz et al., 1993]. The matrix serves to absorb the initial energy of the laser

and then to transfer this energy to the analyte molecule, resulting in ablation of both the

sample and matrix molecules and their ejection into the gaseous phase [Talrose et al.,

1999; Karas et al., 2000b]. During this process ions are formed, however, the exact mechanism involved with ion formation is still unclear [Talrose et al., 1999; Karas et al., 2000b]. Figure 1.1 shows a mechanistic diagram describing the MALO I ionisation process. Although the matrix serves to promote ionisation and detection of the intact

analyte molecule, it has also been observed that ion signals derived from the matrix can

suppress the signal for the analyte of interest, or lead to the formation of analyte-matrix

adducts [Beavis and Chait, 1989b].

4 Figure 1.1. A schematic diagram of the MALDI ionisation process.

Ti rne-of- f Ii grit t·1ass Ana l_yser

• ••••••••••••••

Desorbed 'pi ume' of _,,.,--- m':itrix 3nd analyte ions • /_,/..-,/ = An31~.ite • • = M':itrix • -• = Cation (e.g. Na+ or H+) • • • ...... Sample • • - •. - • • pl11te - . - • . -- .. \I • ;, • • • • • . • -·• .­ . • \ ..- . • • • • • · . • . . ·- .. · -·- .- ·- . . -.• ..- - . . -...... - ·. - . ·•. . -~

© 2000 PAUL GATES

Source: http://www-methods.ch.cam.ac.uk/meth/ms/theory/maldi.html

5 Characteristic of desorption/ionisation techniques, MALDI also requires samples to be introduced into the mass spectrometer in the solid phase and this hinders interfacing

MALDI to on-line separation techniques such as high performance liquid chromatography (HPLC). Because MALDI predominantly forms the singly protonated molecule and induces minimal fragmentation [Karas and Hillenkamp, 1988], the resulting mass spectrum is easy to interpret: the predominant ion in a MALDI mass spectrum corresponds to the singly protonated analyte (i.e., [M+Ht in positive ion mode). The simplicity of the MALDI mass spectrum, the sensitivity of the technique and the speed of analysis make this an ideal tool for quality control analysis of synthetic compounds, such as recombinant proteins, where both the intact mass of the analyte and the relative purity of the sample can be rapidly determined [Kanazawa et al., 1999;

Villanueva et al., 2001]. Many other classes of molecules are amenable to analysis by

MALDI-TOF mass spectrometry (MALDI-TOFMS), including synthetic polymers [Wu and Odom, 1998; Marie et al., 2000], a wide range oflow molecular weight natural products [Duncan et al., 1993; Gimon et al., 1994; LeRiche et al., 2001], lipids

[Fujiwak:i et al., 1999; Schiller et al., 1999; Ayorinde et al., 2000; Petkovic et al., 2001],

oligosaccharides [Stahl et al., 1994; Harvey, 1996; Papac et al., 1996; Dai et al., 1997;

Harvey et al., 1998; Mo et al., 1998; Charlwood et al., 1999a; Charlwood et al., 1999b;

Colangelo and Orlando, 1999; Finke et al., 1999; Geyer et al., 1999; Pfenninger et al.,

1999; Sato et al., 2000; Sumi et al., 2000; Jacobs and Dahlman, 2001], peptides and

proteins [Nguyen et al., 1995; Zaluzec et al., 1995; Roepstorff, 2000], as well as

oligonucleotides [Nordhoff et al., 1992; Pieles et al., 1993; Tang et al., 1993a;

Hathaway, 1994; Nordhoff et al., 1994; Wang and Biemann, 1994; Wu et al., 1994;

Christian et al., 1995; Fitzgerald and Smith, 1995; Bentzley et al., 1996; Dai et al.,

1996; Fu et al., 1996; Juhasz et al., 1996; Roskey et al., 1996; Smimov et al., 1996;

6 Ball and Packman, 1997; Faulstich et al., 1997; Haff and Smirnov, 1997; Hahner et al.,

1997; Miketova and Schram, 1997; Zhu et al., 1997; Bleczinski and Richert, 1998; Fei et al., 1998; Owens et al., 1998; Van Ausdall and Marshall, 1998; Cotter et al., 1999;

Langley et al., 1999; F ei and Smith, 2000; Ragas et al., 2000; Bray et al., 2001].

Although MALDI is not easily coupled to on-line separation techniques, it offers many other advantages including tolerance toward contaminants such as high concentrations of salts, buffers and detergents common to many biochemical preparations, and it is able to detect numerous components in complex mixtures in a single analysis and over an extended mass range [Beavis and Chait, 1990; Wang and Chait, 1994]. Also, the uniqueness of this process has inspired numerous novel applications ofMALDI­

TOFMS. For example, researchers have described direct sampling from membrane supports [Strupat et al., 1994; Vestling and Fenselau, 1994; Blackledge and Alexander,

1995; Liu et al., 1995; Gharahdaghi et al., 1996; Schreiner et al., 1996; McComb et al.,

1997; Guittard et al., 1999; Shen et al., 2001], the direct analysis of single cells and microorganisms [Holland et al., 1999; Lynn et al., 1999; Li et al., 2000a] and the direct

analysis of constituents from intact biological tissue [Caprioli et al., 1997; Chaurand et

al., 1999b; Jespersen et al., 1999; Li et al., 1999; Stoeckli et al., 1999; Stoeckli et al.,

2001]. This has enabled characterisation of abundant endogenous peptides as well as

regional molecular mapping of protein components in biological tissues and organisms.

In addition, MALDI-TOFMS has been utilised for the examination of non-covalent

complexes, such as protein-protein interactions and DNA-protein complexes [Tang et

al., 1995; Jensen et al., 1996; Farmer and Caprioli, 1998; Lin et al., 1998; Thiede and

von Janta-Lipinski, 1998; Kiselar and Downard, 2000].

7 Since its inception, MALDI has been successfully interfaced to several different types of mass analysers, including the quadrupole ion trap [Lee and Lubman, 1995; Qin et al.,

1996; Qin and Chait, 1997; Doroshenko and Cotter, 1998] and fourier transform

[Mciver et al., 1994], but the most popular combination is with the time-of-flight (TOF) mass analyser. Additionally, MALDI has been coupled to hybrid mass analysers, such as the quadrupole time-of-flight [Harvey et al., 2000; Krutchinsky et al., 2000; Loboda et al., 2000; Shevchenko et al., 2000; Baldwin et al., 2001; Griffin et al., 2001;

Kirpekar and Krogh, 2001]. However, the MALDI-TOFMS combination is affordable, practical, offers high mass accuracy, high resolution and high sensitivity over an unlimited mass range. The practical utility has also been enhanced significantly due to the advent of delayed extraction [Wiley and McLaren, 1955; Brown and Lennon, 1995;

Whittal and Li, 1995; Bahr et al., 1997]; and with the implementation of the reflectron which refocuses ions based on their energy (and effectively increases the flight length

and time), superior mass resolution (FWHM > 10000) and therefore improved mass

accuracy, are routinely achieved [Mamyrin et al., 1973; Kinsel et al., 1997; Takach et

al., 1997]. Mass errors of less than 10 ppm are routine for the characterisation of low

mass pharmacologically active compounds as well as for peptides and proteins [Fukai et

al., 2000; Lake et al., 2000; Andalo et al., 2001]. Figure 1.2 shows a schematic of a

MALDI-TOF mass spectrometer.

8 Figure 1.2. A schematic diagram of a MALDI-TOF mass spectrometer with delayed extraction.

Variable-voltage Laser grid attenuator Reflector Reffeotor (electrostallc detector mirror) Sample plate ~

Main ,ource chamber

.,_...... ~ Video carMra Linear Timed Ion detector Aperture {grounded) $elector Collision ·celt (optiOnal)

- - .... Ion path in ren.ct.or mode · • • ·...,. Lasorpeth

Source: Perseptive Biosystems. This is the configuration of the Perseptive DE-STR

system used throughout this project. This system differs only in detail from the DE-PRO

that was also employed for a portion of the work described in this thesis.

9 1.3. Electrospray mass spectrometry

In contrast to MALDI, electrospray ionisation involves the introduction of a sample in the liquid phase. Figure 1.3 shows a schematic of the electrospray process. Prior to the development of electrospray ionisation, analytes in solution could only be introduced into the mass spectrometer and analysed using techniques such as continuous flow F AB or thermospray [Blakley et al., 1980; Caprioli et al., 1986; Caprioli, 1990]. Although the principles of electrospray ionisation were originally described by Dole et al. (1968), its application to mass spectrometry was first demonstrated by Whitehouse and coworkers (1985) when they successfully detected biomolecules such as gramicidin S, cyclosporin and substance P. They also proposed the potential ofESI for interfacing liquid chromatography to a mass spectrometer [Whitehouse et al., 1985; Fenn et al.,

1989].

The formation of ions by this process has been widely studied and involves the

application of a voltage to an electrospray capillary, thereby imparting a charge to the

liquid stream carrying the analyte(s) of interest. The application of an electric field leads

to the formation of a Taylor cone at the capillary tip and dispersion of charged droplets.

Desolvation results in smaller droplets thereby increasing the charge density on the

surface of each. As the radius of the droplet continues to decrease, the charge repulsion

(i.e., Coulombic repulsion) of each individual droplet exceeds the cohesive surface

tension (i.e., the charge density approaches the Rayleigh limit) forcing it to disintegrate

into numerous droplets of smaller radii, a process known as Coulomb fission [Gaskell,

1997; Cole, 2000; Karas et al., 2000a; Kebarle, 2000]. This process ultimately leads to

each analyte molecule retaining multiple charges (protons). The mass spectrum

generated is characterised by multiple mass to charge (m/z) ratios for the population of

10 analyte molecules [Fenn et al., 1989; Smith et al., 1990; Cole, 2000]. Figure 1.4 shows an example of an ESI mass spectrum for myoglobin compared with a MALDI mass spectrum of the same molecule.

11 Figure 1.3. A schematic diagram of the electrospray ionisation process.

Taylor cone

Ca1hodo

Electrospray capillary maintained at high voltage I .., ~"--. . T . .,~ • ,+• .. + - • "" ....Y - •• + / ••• .. + " •.,.,./ • .. t- t .. .. 4 // ..' ... / -~. Anode

I/\ .. 0...,_

Powot Supply

Coulomb fission occurs as the Desolvation of charged droplets charge density exceeds the surface tension of each droplet

Source: Griffiths et al. , 2001

12 Figure 1.4. A comparison ofESI and MALDI spectra of horse apomyoglobin.

A. An ESI mass spectrum showing the multiple charge states detected for horse

apomyoglobin (16,951 Da). The insert shows the deconvoluted mass spectrum.

16951.0 ..... C a, u C Ill '0 C :I .0cc a, > i ai +16 a: 0 1 .5 15500 16000 16500 17000 17500 18000 18500 19000 100 Mass (Da) +15 +17 1131.1 +14 .2 1211.9 +1 94 .9 +13 1 .o ;;e !!.. +1 8 .3 +12 a, u ffi.s 141 .5 C Ill '0 C :I .0 cc +21 .2 +11 t 1541.9 i ai +22 a: 77 .5

+9 1 .2

0 600 700 800 900 1000 1100 1200 1300 1400 1500 1600 1700 1800 1900 inh.

13 Figure 1.4. A comparison ofESI and MALDI spectra of horse apomyoglobin.

B. A MALDI mass spectrum obtained for horse apomyoglobin showing the singly

([M+Ht 16,952 Da) and doubly ([M+2H]2+ 8,476 Da) charged species.

16952.56 100

8476.78 -0~

9200 12400 15600 18800 22000 Mass (mlz)

14 When compared to MALDI mass spectra, those generated by ESI are generally more

difficult to interpret, especially when analysing large molecules, e.g., proteins, oligonucleotides, and other polymers. Due to the more complex nature of ESI mass

spectra, multicomponent samples are usually examined only after separation. However, because this ionisation mode is compatible with on-line separation techniques, such as

HPLC and capillary electrophoresis, it offers a distinct advantage over MALDI [Banks,

1997; Niessen, 1998; Choudhary et al., 2000; Dalluge, 2000]. For example, facile

identification of endogenous peptides and proteins in biological fluids such as urine and blood filtrate has been reported [Evershed et al., 1993; Valaskovic et al., 1996; Heine et al., 1997; Raida et al., 1999]. Similar to MALDI, this mode of ionisation is also

amenable to the analysis of many different classes of molecules (e.g., peptides, proteins,

oligonucleotides, lipids, drug molecules and alkaloids), and the accurate intact mass of molecules can also be determined [Gelpi, 1995]. Because ESI is a soft ionisation

technique, it has also been employed for the characterisation of non-covalent complexes

[Jaquinod et al., 1993; Bayer et al., 1994; Bakhtiar and Steams, 1995; Pocsfalvi et al.,

1997; Pramanik et al., 1998]. By combining ESI with tandem mass spectrometry, or in­

source collision induced dissociation (CID), structural information for various organic

molecules, such as natural products, drug metabolites and peptides can also be acquired

with relative ease [Smith et al., 1990; Griffiths et al., 2001].

The most noticeable recent advancement in ESI is the development oflow flow rates ( <

1 µL/min) for sample introduction, commonly termed nano-electrospray (nano-ESI,

nanospray) [Gale and Smith, 1993; Andren et al., 1994; Emmett and Caprioli, 1994;

Davis et al., 1995; Wilm and Mann, 1996; Griffiths, 2000; Karas et al., 2000a]. This

substantial reduction in flow rate contributes to less sample consumption and has been

15 shown to offer greater sensitivity. For instance, only a few microlitres of a sample

solution is required to obtain molecular weight and structural information [Wilm and

Mann, 1996; Karas et al., 2000a]. Routine analysis of trace levels of peptides, carbohydrates and lipids have been reported and as a result, nanospray has been readily adopted in the biological sciences [Stenfors et al., 1997; Gatlin et al., 1998; Lhoest et al., 1998; Griffiths, 2000; Karas et al., 2000a; Rafatro et al., 2000; Zamfir et al., 2000].

Because nanospray allows the investigator to work with low volumes and consumes less

sample, it is ideally suited to applications where the sample is limited and multiple MS

experiments are performed. For instance, one area of interest is the mapping of sites of proteins involved in signal transduction pathways [Neubauer

and Mann, 1999; Ogueta et al., 2000].

1.4. Ion dissociation and tandem mass spectrometry

Both MALDI and ESI are soft ionisation techniques that result in minimal

fragmentation because of the low internal energy of the analyte ion [Karas and

Hillenkamp, 1988; Fenn et al., 1989]. In order to obtain structural information, these

ionisation techniques have been coupled with processes capable of increasing the

internal energy of ions and thereby inducing fragmentation [Griffiths et al., 2001]. For

ESI the most commonly employed analysers are currently the triple quadrupole and the

quadrupole ion trap [Griffiths et al., 2001]; however, commercially available hybrids,

such as the quadrupole time-of-flight [Shevchenko et al., 1997; Doroshenko and Cotter,

1998; Harvey et al., 2000; Krutchinsky et al., 2000; Loboda et al., 2000; Shevchenko et

al., 2000; Griffin et al., 2001; Shevchenko et al., 2001] are increasingly popular. TOF­

TOF instruments incorporating a collision cell between the two flight tubes are now

16 commercially available and have generated considerable interest [Medzihradszky et al.,

2000].

Since its inception, tandem mass spectrometry (MS/MS) has been applied in the pharmaceutical industry to characterise bioactive natural products and their metabolites,

and in quantitative drug studies; more recently, however, it has had tremendous impact

in the area of peptide sequencing and protein identification. Peptide sequencing by tandem mass spectrometry was first demonstrated in the mid-1980s [Tomer et al., 1984;

Cody et al., 1985; Hunt et al., 1985; Hunt et al., 1986; Hunt et al., 1987; Biemann,

1988] and this has lead to the refinement of many strategies, based on the generation of peptides by enzymatic or chemical cleavage, to unambiguously identify proteins. In

addition, the ability of tandem mass spectrometry to overcome some of the difficulties

associated with Edman sequencing - N-tenninally blocked or post-translationally

modified peptides - has contributed to its widespread use in protein analysis [Biemann

and Scoble, 1987; Biemann, 1988]. The recent explosion in the number of protein

sequences that are widely available, coupled with the development of powerful

computer search algorithms, has enabled rapid identification of proteins in biological

samples based on the sequence of relatively few(< 10) amino acids [Larsen and

Roepstorff, 2000].

The maturation of the quadrupole ion trap mass spectrometer over the last decade has

facilitated interest and development in MS/MS analysis [March, 1997]. In contrast to

the triple quadrupole mass spectrometer, the quadrupole ion trap is able to isolate and

store ions of interest (precursor ion) by manipulation of the radio frequency (RF)

potential and direct current (DC) voltage applied to the ring electrodes and end cap

17 respectively [Cooks et al., 1991; Jonscher and Yates, 1997]. The selected trapped precursor ions are subsequently decomposed prior to their ejection and detection.

Because newly generated fragment ions can also be selectively isolated and stored, multiple fragmentation cycles are possible (MSn) [Cooks et al., 1991]. In addition, the high efficiency with which quadrupole ion trap mass spectrometers can trap and fragment ions has contributed to their popularity in the area of protein and peptide characterisation [Davis and Lee, 1998; McGinley et al., 2000].

The mechanism involved in generating fragment ion information by MALDI-TOFMS differs to that described for the triple quadrupole or quadrupole ion trap mass

spectrometers. This process, termed post-source decay (PSD), involves the detection of

fragment ions formed by the metastable decomposition of precursor ions following their

acceleration into the first field-free region of the TOP mass spectrometer [Spengler et al., 1992; Kaufmann et al., 1993; Chaurand et al., 1999a]. Excess intramolecular energy

gained by the precursor ion during the ionisation process is released by decomposition

of the molecule. Because the fragment ions are formed in the field-free region and retain

the original velocity of the precursor ion, they contain a distribution of kinetic energies

dependant on their mass (KE = ½ mv2) and can only be detected following refocusing in

an electrostatic mirror (i.e., by reflectron TOP mass spectrometry) [Spengler et al.,

1992; Kaufmann et al., 1993; Cornish and Cotter, 1994; Cordero et al., 1995; Chaurand

et al., 1999a]. Post-source decay analysis has been used extensively for both peptide and

oligonucleotide sequencing [Bentzley et al., 1996; Mo et al., 1997; Gross et al., 1998;

Owens et al., 1998; Fournier et al., 2000; Gevaert et al., 2001; Gross et al., 2001; Talbo

et al., 2001], as well as for the characterisation of other biomolecules such as

oligosaccharides and small drug molecules [Lemoine et al., 1996; Garozzo et al., 1997;

18 Okamoto et al., 1997; Mo et al., 1998; Rouse et al., 1998; Yamagaki et al., 1998;

Yamagaki and Nakanishi, 1998; Mizuno et al., 1999; Viseux et al., 1999; Broberg et al.,

2000; Garozzo et al., 2000; Spina et al., 2000; Yamagaki and Nakanishi, 2000].

1.5. MALDI and ESI in the biological sciences

1.5.1. Quantification of biomolecules by mass spectrometry

Prior to the availability of ESI, quantitative analysis by mass spectrometry was mostly limited to low mass molecules amenable to separation by gas chromatography (GC)

[Ragunathan et al., 1999]. However, the introduction ofFAB and the subsequent development of continuous flow F AB enabled direct quantification of compounds such

as peptides and steroids and their metabolites [Barber et al., 1981; Gaskell et al., 1983;

Caprioli et al., 1986; Lisek et al., 1989; Gaskell, 1990; Kusmierz et al., 1990; Chen et

al., 1991; Dass et al., 1991; Ikarashi et al., 1991; Ikarashi and Maruyama, 1991;

Desiderio, 1992; Davoli et al., 1993]. Although GC-MS is still widely applied in

quantitative analysis in many areas (e.g., clinical testing, forensic or toxicological

sciences, and environmental analyses) the introduction ofESI coupled to on-line liquid

chromatography has overcome many of the obstacles associated with both GC-MS and

F AB analysis, such as mass limitations and the thermal lability and/or limited volatility

of some molecules [Niwa, 1986; Weykamp et al., 1989; Dehennin, 1990; Goldberger

and Cone, 1994; Gleispach et al., 1995; Lehrer, 1998; Risby and Sehnert, 1999;

Shindelman et al., 1999; Nexo et al., 2000; Richardson, 2000; Speed et al., 2000;

Kikuchi et al., 2001; Takayasu et al., 2001]. As a result, quantitative analyses by ESI

coupled to liquid chromatography have become routine in many settings, notably the

pharmaceutical industry, where it is predominantly applied to pharmacokinetic studies

19 [Gelpi, 1995; Tiller et al., 1997; Ayrton et al., 1998; Maurer, 1998; Lee and Kerns,

1999; Muck, 1999; Rashed et al., 1999; Vreken et al., 1999; Wong et al., 1999; Jemal et al., 2000; Jemal and Mulvana, 2000; Kato et al., 2000a; Kato et al., 2000b; Kissmeyer et al., 2000; Lau et al., 2000; Miketova et al., 2000; Riffel et al., 2000; Valentin-Blasini et al., 2000; Van Eeckhout et al., 2000; Xu et al., 2000; Holstege et al., 2001].

MALDI is an alternative ionisation technique to ESI that is also compatible with non­ volatile and/or high molecular weight analytes. Although it is not widely used for quantitative analysis, it possesses advantages that make it a potentially useful quantitative tool. For example, it allows analysis of molecules over an extensive mass range and it is capable of analysing complex sample mixtures without the need for

extensive sample preparation. In addition, almost any class of molecule can be analysed

and MALDI mass spectra are relatively easy to interpret, i.e., the predominant species is

the singly charged molecule. Also, analyses are fast: a single sample can be examined

within minutes and chemical modification of analytes prior to analysis is unnecessary.

These properties make quantitative MALDI-TOFMS attractive in many different areas,

for example, for rapid clinical testing and for quantification of large intact biomolecules

such as polypeptides and oligonucleotides [Duncan et al., 1993; Gusev et al., 1993;

Tang et al., 1993b; Muddiman et al., 1994; Nelson et al., 1994; Bruenner et al., 1996;

Gusev et al., 1996; Muddiman et al., 1996; Ling et al., 1998; Desiderio et al., 2000;

Gobom et al., 2000].

There has been a limited number of studies exploring the applicability of MALO1-

TOFMS as a quantitative tool [Duncan et al., 1993; Gusev et al., 1993; Harvey, 1993;

Tang et al., 1993b; Muddiman et al., 1994; Nelson et al., 1994; Jespersen et al., 1995;

20 Abell and Spoms, 1996; Bruenner et al., 1996; Gusev et al., 1996; Muddiman et al.,

1996; Tang et al., 1996; Camafeita et al., 1997; Wu et al., 1997; Kazmaier et al., 1998;

Ling et al., 1998; Sugiyama et al., 1999; Wang et al., 1999; Desiderio et al., 2000;

Gobom et al., 2000; Wang and Spoms, 2000; Horak et al., 2001]. These studies highlight many potential problems that have restricted its application including the

limited dynamic range, the incompatibility of the ionisation mode with on-line

chromatography, and the variability in the ion current measured between successive

laser shots and from sample-to-sample. Together, these factors limit its utility and

compound the errors involved with quantitative determinations. In addition, non­ uniform co-crystallisation of matrix and sample can also lead to poor reproducibility of results. Numerous approaches, such as electrospray deposition of sample and matrix to

improve matrix/analyte homogeneity [Preston et al., 1993; Vorm et al., 1994; Gusev et

al., 1995; Nicola et al., 1995; Hensel et al., 1997] and extensive studies concerning

matrix and internal standard selection [Gusev et al., 1993; Gusev et al., 1996;

Wilkinson et al., 1997; Horak et al., 2001], have been undertaken to increase the

applicability ofMALDI-TOFMS for quantitative analysis.

1.5.2. Analysis of biomolecules in complex mixtures

The ready availability of MALDI and ESI instruments has permitted a change in focus

from the analysis of low molecular weight organic biomolecules to the examination of

higher mass, involatile biomolecules, such as proteins. These molecules are responsible

for cell structure, cell growth and division, as well as intra- and inter- cellular regulation

and communication [Alberts, 1998]. Prior to the implementation ofMALDI and ESI,

the mass analysis of intact peptides and proteins was restricted to an upper mass limit of

21 approximately 20,000 Da at best [Hakansson et al., 1983; Fales et al., 1985; Craig et al.,

1987; Andrews et al., 1988; Jonsson et al., 1988; Roepstorff et al., 1988; Nielsen and

Roepstorff, 1989; Nielsen et al., 1989; Roepstorff, 1989; Tsarbopoulos et al., 1991].

The concurrent development of MALDI and ESI ionisation modes, and on-going refinements in mass analysers such as the time-of-flight and quadrupole ion trap, have

enabled vast expansions in the mass range capabilities of mass spectrometry, as well as improvements in mass resolution, accuracy and sensitivity.

The development of MALDI and ESI has revolutionised the way proteins are examined.

Previously, proteins were characterised using a combination of physical and biochemical methods, such as isolation and purification by HPLC, immunoaffinity

chromatography and gel electrophoresis [Hage, 1999; Weller, 2000]. Techniques such

as X-ray crystallography, nuclear magnetic resonance, amino acid analysis and Edman

sequencing were typically employed to determine properties such as the amino acid

sequence, mass, and three dimensional structure. Although these techniques are still

widely used today, they require large amounts of highly purified material and are not

suitable for the analysis of trace components in complex biological samples.

Today, separation and visualisation of proteins in complex biological samples by two

dimensional gel electrophoresis (2DE) has become a pivotal process preceding analysis

by mass spectrometry [Hanash et al., 1991]. This technique, first developed by

O'Farrell (1975), involves separation of proteins in a sample by their isoelectric point

(first dimension), followed by their molecular weight (second dimension). In addition to

aiding in protein separation, this procedure can be used to detect post-translational

modifications [Neubauer and Mann, 1999; Sickmann et al., 2001], or to provide an

22 estimate of the relative amounts of each protein in matched sample sets [Lahm and

Langen, 2000; Grus et al., 2001]. The protein(s) of interest are then excised, chemically or enzymatically cleaved at specific sites, and then the accurate mass capabilities of

MALDI-TOFMS are utilised to generate unique peptide mass maps or fingerprints

[James et al., 1993; Mann et al., 1993]. These data are then used to search databases containing the sequences of known proteins, permitting unambiguous identification of proteins [Henzel et al., 1993; James et al., 1993; Cottrell, 1994]. Aside from masses alone, structural information (sequence) can also be obtained by either ESI-LC/MS/MS or MALDI-PSD, and this provides further information that identifies a protein

[Shevchenko et al., 1996b; Wilm et al., 1996; Chaurand et al., 1999a; Keough et al.,

2000; Medzihradszky et al., 2001].

One of the first large scale studies involving protein identification by 2DE and mass

spectrometry was described using yeast as the model system [Shevchenko et al., 1996a].

This study demonstrated that given a completely sequenced genome, up to 90% of

visualised proteins can be identified with high confidence by peptide mass

fingerprinting and database searching alone. Partial amino acid sequences obtained by

nanospray tandem mass spectrometry were also employed to search databases

containing expressed sequence tags (ESTs) and this enabled additional, and some novel

proteins, to be identified.

Alternative strategies for global protein analysis based on mass spectrometry are

emerging. The most notable include the analysis of complex protein mixtures by ESI

mass spectrometry following enzymatic digestion. Peptides are first separated by

reverse phase liquid chromatography, or by multiple chromatographic separations in

23 series, then analysed by tandem mass spectrometry [McCormack et al., 1997; Yates et al., 1997; Link et al., 1999; Yates et al., 2000; Davis et al., 2001; Washburn et al.,

2001]. This approach aims to identify every protein in a sample in a single chromatographic analysis and bypasses the need for prior electrophoretic separation.

Although this technique is capable of rapidly identifying proteins, post-translational modifications are usually not detected or identified, and the in vivo form of each protein is assumed but not determined.

The protein identification strategies outlined above assume the primary structure of proteins and peptides based on limited data and do not offer any insight into their biological role in a chosen system. With the knowledge that each gene is capable of

expressing multiple forms of the same protein, mass spectrometric techniques are

continuously being developed and applied to provide additional information such as the

nature, site and extent of post-translational modifications (e.g., phosphorylation,

, oxidation, truncation), and both covalent and non-covalent protein­

protein interactions (e.g., protein complexes) [Miyazaki et al., 1996; Vinh et al., 1997;

Kamo et al., 1998; Terazaki et al., 1998; Zand et al., 1998; Boeshans et al., 1999;

Borchers and Tomer, 1999; Bordini and Hamdan, 1999; Jaquinod et al., 1999; Vinh et

al., 1999; Wilkins et al., 1999; Jonsson et al., 2000; Pritzker et al., 2000; Song et al.,

2000; Woods and Huestis, 2001]. To define biological functions of an individual

protein, established biochemical techniques such as enzyme activity assays and

bioassays are still required.

24 1.6. The emergence of Proteomics

The completion of the draft form of the Human Genome Project has encouraged interest in the expressed gene products as scientists scramble to determine the biological

function of each gene as well as its potential importance in health and disease.

Proteomics is a global term used to describe the process of identifying the total protein content of an organism or system (e.g., cell culture system, biological tissue or fluid), as well as determining the function or interaction of these proteins [Wasinger et al., 1995].

Traditionally, the biological activity of proteins in a chosen disease is deciphered one by one, but the emergence of proteomics has shifted the focus toward the examination of overall protein expression in a given system, i.e., the concurrent analysis of multiple proteins in the same sample. Differential or comparative proteomics has also developed

so that differences in protein expression between two (or more) different states of a biological system can be monitored and identified.

Proteomics has expanded beyond the health sciences to include areas such as plant

biology, virology, microbiology and parasitology [Thiellement et al., 1999; Barrett et

al., 2000; Cash, 2000; Gutierrez, 2000; Jan van Wijk, 2000; Panter et al., 2000; Peltier

et al., 2000; Gallardo et al., 2001; Peck et al., 2001]. This progression in proteomic

studies has been fueled by rapid expansions in the genomic databases of various

organisms, the ready availability of these data, and on-going developments in

bioinformatics. Completion of the genomic sequences of various organisms such as C.

elegans, S. cerevisiae, D. melanongastor and the availability of the draft sequence of the

human genome [Goffeau et al., 1996; Consortium, 1998; Adams et al., 2000; Venter et

al., 2001], has enabled cross-species homology searches that have aided in the

identification of unknown proteins in organisms with incomplete genomes, as well as

25 tentative assignment of protein structure and function [Cordwell et al., 1997]. Progress in bioinformatics has led to the implementation of computer search algorithms that

enable mass spectrometric data to be searched directly against both the protein and

genomic databases [Pappin et al., 1993; Yates et al., 1993; Eng et al., 1994; Mann and

Wilm, 1994; Yates et al., 1995a; Yates et al., 1995b; Taylor and Johnson, 1997; Clauser

et al., 1999; Perkins et al., 1999; Zhang and Chait, 2000]. It is therefore now feasible to rapidly identify almost any protein in a sample, and to gain some insight into biological

function. For example, the molecular events within a cell (e.g., signal transduction pathways, cell signaling) can be elucidated, potential biomarkers of disease can be

discovered, and protein-protein interactions (e.g., protein complexes) can be studied

[Rasmussen et al., 1997; Celis et al., 1999; Godovac-Zimmermann et al., 1999;

Ostergaard et al., 1999; Page et al., 1999; Soskic et al., 1999; Cans et al., 2000; Celis et

al., 2000; Fung et al., 2000; Lewis et al., 2000; Li et al., 2000b; Verma et al., 2000;

Guillonneau et al., 2001; Rohlff, 2001].

Proteomics is primarily descriptive in nature at this time (i.e., it is directed toward the

identification of proteins), and challenges still remain to develop strategies capable of

identifying and quantifying proteins at low copy numbers. An even greater challenge is

precisely determining differences in protein expression between two different states of

the same biological system. Such strategies are becoming increasingly important as the

applications of proteomics in the biomedical sciences escalate.

To measure the relative quantities of proteins, techniques have emerged that involve

protein labeling with differential mass tags and subsequent detection of the derivatised

peptides by mass spectrometry. For example, changes in protein expression that occurs

26 between two different states of the same biological system can be monitored using isotope coded affinity tags (ICAT} [Gygi et al., 1999]. Proteins in each sample are quantified following proteolytic digestion and derivatisation with differential mass tags.

Peptides containing the mass tag are then isolated and analysed by ESI-LC/MS/MS. A

similar technique has been described where C-terminal residues and acidic side chains of amino acid residues such as and are converted to methyl

esters prior to analysis [Goodlett et al., 2001]. Other approaches involve quantification of gel isolated proteins, for instance, by N- or C- terminal labeling oftryptic peptides

[Munchbach et al., 2000; Wang et al., 2001; Yao et al., 2001; Wang et al., 2002]. While these techniques are able to determine modest changes in the relative expression of

individual proteins, there is currently no practical method to accurately determine the

absolute quantities of the hundreds of proteins in a sample.

1.7. Aims

This thesis focuses on the development of mass spectrometric techniques to identify and

quantify peptides and proteins in complex biological systems and also evaluates the

utility of current proteomic approaches such as 2DE and direct analysis by ESI­

LC/MS/MS. Although these existing strategies aim to detect and identify proteins in

biological tissue or fluid, there are still difficulties associated with the identification of

low abundant proteins and the quantification of biologically relevant proteins. The

utility ofMALDI-TOFMS and ESI-MS for the detection and analysis of peptides in

biological fluids was explored, and mass spectrometric methods for the absolute and

relative quantification of proteins and other biomolecules were developed.

27 Because it is recognised that 2DE and peptide mass fingerprinting allow the visualisation and identification of multiple proteins in a given biological system, it is the preferred approach for protein analysis. This technique, however, is not applicable to the detection and identification of several protein classes, such as membrane proteins, basic proteins, as well as high mass proteins. Therefore, in order to provide a complete description of the protein complement of a biological system, 2DE must be integrated with other protein identification strategies. Consequently, during this work several

strategies were applied to the separation of proteins in biological fluids such as human tear and seminal plasma, and a combination of mass spectrometric approaches was

employed to examine the peptide and protein constituents.

To overcome the difficulties associated with 2DE, other techniques have been proposed

such as the direct analysis of proteins by ESI-LC/MS/MS following proteolytic

digestion [Yates et al., 1997; Link et al., 1999]. Even though this technique can be used

to identify multiple proteins in a single analysis, it does not provide quantitative

information and only establishes the presence of a protein or parts of a protein in a

sample. Additionally, information on possible protein modification is not easily

determined. Therefore, in this work this approach was combined with gel

electrophoresis (1D and 2D) and MALDI-TOFMS to characterise peptides and proteins

in biological fluids.

The ability to quantify changes in protein expression, whether in absolute or relative

terms, is an important aspect of proteomics and there are some emerging techniques that

attempt to address this issue. For example, metabolic labeling of proteins for in vitro

studies [Oda et al., 1999; Conrads et al., 2001; Smith et al., 2001] and the adoption of

28 differential mass tags such as ICAT [Gygi et al., 1999], have shown some promise for

accurately estimating the relative changes in protein expression in a biological system.

While these techniques take advantage of the sensitivity and specificity offered by mass

spectrometry, they are limited in their applicability to particular sample types, or are targeted toward selected proteins. Therefore, protein quantification methods that can be universally applied, either to quantify specific proteins or to quantify proteins on a

global scale, and that enable accurate and precise measurements are still needed. An

approach for the global quantification of proteins involving esterification of proteolytic peptides with isotopomers ofbutanol and analysis by ESI-LC/MS was developed. This technique was employed to measure the relative abundances of proteins in open and

closed eye tear fluid.

At this time, there are no established mass spectrometric methods for the absolute

quantification of proteins in biological samples. Currently, protein quantification

methods generally utilise specific antibody-based methods to determine in vivo levels.

As a consequence, only a limited number of targeted proteins can be quantified and

their identity must be known and an appropriate antibody available. Although

immunochemical techniques offer a sensitive approach to protein quantification, they

are not able to distinguish proteins that bind non-specifically and between multiple

forms of a protein that may arise in vivo. Mass spectrometry, however, offers the

sensitivity and specificity required for the absolute quantification of many different

biomolecules, including peptides and proteins. MALDI-TOFMS is able to detect

biomolecules over an extended mass range, requires minimal sample preparation prior

to analysis, and is able to analyse many classes of biomolecules. Therefore, the

application of MALDI-TOFMS to the absolute quantification of a wide range of

29 analytes in biological matrices was explored: homovanillic acid in human urine; epinephrine and norepinephrine in adrenal gland extract; insulin in human pancreatic tissue; LW-hemorphin- 7 in adrenal gland extract and growth hormone in rat pituitary gland. The outcomes and implications of these studies are presented and discussed in detail.

30 CHAPTER2

METHODS

31 2.1. Instrumentation

All MALDI analyses were performed on a Voyager DE-STR or DE-PRO time of flight mass spectrometer (Perseptive Biosystems, Framingham, MA). All spectra were acquired in positive ion mode using standard instrument parameters unless otherwise indicated. Each spectrum was composed of an average of 100 individual laser shots. For peptide analyses, spectra were collected in reflectron mode using an acceleration voltage of20 kV. When the DE-STR was employed, the extraction grid voltage was set to 63 % and a delay time of 225 nsec was used. Analysis on the DE-PRO was performed with the extraction grid set at 76 % and the delay time set to 100 nsec.

Post-source decay (PSD) analyses were performed at an acceleration voltage of20 kV

and a grid voltage of 70% was applied. A delay time of 100 nsec was employed.

Fragment ion spectra were collected at mirror ratio decrements of 0.85 and were

stitched together, using the software provided, to form the composite PSD spectrum.

Proteins were analysed in linear mode with the acceleration voltage set to 25 kV and the

grid voltage set to 92 %. A delay time of750 nsec was used. A standard 100 position

MALDI sample plate was used for all analyses.

All standards used for mass calibration were obtained from Perseptive Biosystems

(Framingham, MA) with the exception of substance P and bovine insulin oxidised B

chain (Sigma Chemical Co., St Louis, MO).

32 All electrospray mass spectrometry was performed on a quadrupole ion trap mass spectrometer (Finnigan LCQ DECA, San Jose, CA) operated in a data dependant mode with dynamic exclusion enabled. Ion trap parameters were optimised by tuning to the doubly charged ion of angiotensin II (m/z 523.7; 1 pmol/µL; Sigma Chemical Co., St

Louis, MO). The heated capillary was maintained at a temperature of 200°C. Tandem mass spectra were obtained using a collision energy of28 %. For flow rates greater than

10 µL/min, a voltage of 3.5 kV was applied to the electrospray needle. For analyses at

0.5 µL/min, the electrospray needle was maintained at 1. 75 - 2.0 kV.

2.2. MALDI matrices

All MALDI matrices were used as supplied and were obtained from Sigma Chemical

Co (St Louis, MO), with the exception of caffeic acid (ICN Biomedicals Inc., Aurora,

OH). MALDI matrices included: a.-cyano-4-hydroxycinnamic acid (5 mg/mL in 80 %

CH3CN: 0.1 % CF3COOH), caffeic acid, ferulic acid and sinapinic acid prepared at 10

mg/mL (80 % CH3CN: 0.1 % CF3COOH). 2,5-Dihydroxybenzoic acid was also

employed and was prepared at 10 mg/mL in 100 % CH3CN.

2.3. Proteolytic enzymes

Modified porcine trypsin was obtained from Promega (Madison, WI) and

endoproteinase Asp-N was obtained from Calbiochem (La Jolla, CA). Both proteolytic

enzymes were resuspended in NH4HCO3 (0.01 µg/µL, 25 mM, pH 8), and used as

supplied.

33 2.4. Protein identification criteria

2.4.1. MALDI-TOF mass spectrometry

Peptide mass fingerprints were searched against the non-redundant protein databases using MS-FIT (Protein Prospector, http://ucsf.prospector.edu/). Positive identifications were assigned when the significance score between the first and second match was greater than one order of magnitude. Fragment ion spectra generated by MALDI-PSD were searched against the non-redundant protein databases using MS-TAG (Protein

Prospector). (See below for database search criteria.)

2.4.2. Electrospray mass spectrometry

Fragment ion spectra generated by ESI-LC/MS/MS were searched against the human

subset of the non-redundant protein databases using SEQUEST (ThermoFinnigan, San

Jose, CA). Peptide matches were considered to be significant when XCorr scores

(matching efficiency between the experimentally generated spectrum and the theoretical

spectrum) exceeded 1.5, when the DCn scores (similarity between the best match and

subsequent matches) exceeded 0.1, and when the Sp value was greater than 100.

Positive protein identification also required that multiple peptides (> 2) were detected

for each protein.

34 2.5. Database search criteria

Protein databases were searched using the following criteria unless otherwise noted:

MS-FIT MS-TAG SEQUEST Enzyme Trypsin No enzyme Trypsin Max. number missed 1 NIA 1 cleavage sites Homo Homo Species Homo sapiens sapiens sapiens Protein molecular Unrestricted Unrestricted Unrestricted weight range Protein pi range Unrestricted NIA Unrestricted modification carbamidomethyalation None None (static modification) Possible ( differential) None None None peptide modifications Peptide mass type Monoisotopic Monoisotopic Monoisotopic Fragment ion mass type NIA Monoisotopic Monoisotopic Peptide mass tolerance ± lO0ppm ±0.25 Da ± 1 Da Fragment ion tolerance NIA ±0.5 Da No error Min. number peptides 4 NIA 2 required for a match Min. number fragment NIA NIA 20 ion count Max.number unmatched fragment NIA 30% NIA ions b- andy- Fragment ion type NIA All series

35 2.6. Identification of protein constituents in human tear

2.6.1. Sample collection

Open eye reflex tear and closed eye tear were collected from healthy individuals using glass microcapillary tubes. Samples were then stored at -80°C until required.

2.6.2. Analysis of whole tear sample

2.6.2.1. MALDI analysis

Aliquots (0.1 - 0.2 µL) of untreated (unfractionated) tear samples were analysed by

MALDI-TOFMS using either a-cyano-4-hydroxycinnamic acid or sinapinic acid as matrix. Samples were analysed in linear and reflector positive ion modes. Spectra were

externally mass calibrated to substance P ([M+Ht 1,347.74 Da), bovine insulin

([M+Ht 5,734.74 Da) and horse apomyoglobin ([M+Ht 16,952.56 Da).

Post-source decay (PSD) was performed on the most abundant components with a mass

ofless than 2000 Da in the MALDI spectrum. Potential precursor peptide masses were

calibrated in reflector mode using the protonated monoisotopic mass of substance P

([M+Ht 1,347.74 Da) and bovine insulin oxidised B chain ([M+Ht 3,494.65 Da). The

timed ion selector was set to allow detection of the fragment ions derived from the

chosen precursor peptides. Multiple PSD spectra were obtained for each chosen peptide.

The fragment ions derived from each peptide was then searched against the non­

redundant protein databases using MS-TAG (Protein Prospector) to determine the

peptide sequence and the parent protein.

36 2.6.2.2. Electrospray mass spectrometry

An aliquot (5 µL) of open eye tear was treated with trypsin (37°C, overnight), and an aliquot containing the resulting tryptic peptides (1 µL) was analysed by ESI­

LC/MS/MS on an ion trap mass spectrometer. Chromatography was performed on a

Cl8 reverse phase column (150 x 1 mm ID) (Vydac, Hesperia, CA) using a binary solvent system. Peptides were eluted over an increasing linear gradient of acidified

CH3CN (0.01 % CF3COOH) (Solvent B) over two hours. Solvent A consisted of aqueous 0.01 % CF3COOH. A flow rate of50 µL/min was employed. The charge state of each eluting peptide was determined, and then each ion was fragmented to generate

MS/MS spectra. To identify the proteins in tear fluid, each MS/MS spectrum was

searched against the human subset of the non-redundant protein database (SEQUEST,

ThermoFinnigan, San Jose, CA).

2.6.3. Isolation and detection of lacrimal proline-rich protein

(This section of the experimental work was undertaken in collaboration with Dr. Robert Sack.)

Open eye tear was fractionated by size exclusion chromatography and selected fractions

were analysed by MALDI-TOFMS to determine the mass of each protein. Sinapinic

acid was used as the matrix. Spectra were externally mass calibrated to bovine insulin

([M+Ht 5,734.74 Da), thioredoxin ([M+Ht 11,674.48 Da) and horse apomyoglobin

([M+Ht 16,952.56 Da). Fractions were then incubated concurrently with trypsin (37°C,

2 hrs) and Asp-N (37°C, 2 hrs) and the resulting peptides analysed by MALDI-TOFMS

with a.-cyano-4-hydroxycinnamic acid used as the matrix. Spectra were internally mass

calibrated to known tryptic fragments of either lysozyme or tear lipocalin.

37 The digested HPLC fractions were also analysed by electrospray LC/MS/MS to determine the identity of each peptide detected by MALDI-TOFMS. Peptides were separated by on-line reverse chromatography using capillary columns (100 mm x 150

µm ID) packed with R2 POROS (Perseptive Biosystems, Framingham, MA). Peptides were eluted over 60 minutes using a linear gradient consisting of acidified CH3CN (0.05

% CF3COOH) (0 - 100 %). The charge state of each eluting peptide was determined before being fragmented to produce MS/MS spectra. Each MS/MS spectrum was then

searched against the human subset of the non-redundant protein database using

SEQUEST. SEQUEST search parameters were adjusted to enable both trypsin and Asp­

N enzyme cleavage.

2.6.4. Quantification of tear fluid proteins

2.6.4.1. Apomyoglobin

Horse skeletal apomyoglobin (20 µg; 98 % purity; ICN Biomedicals Inc., Aurora, OH)

was resuspended in NRiHCO3 (20 µL, 25 mM, pH 8) and digested using trypsin (1 µg,

37°C, overnight). Tryptic peptides were divided into equal aliquots and then dried under

vacuum. Peptides were then esterified with either acidified butanol or d10-butanol (lM,

20 µL, 70°C, 3 hrs) under anhydrous conditions. Anhydrous butanol (Sigma Chemical

Co, St Louis, MO) or d10-butanol (Cambridge Isotope Laboratory, Andover, MA) was

acidified by the addition of anhydrous acetyl chloride (140 µL; Alltech Associates Inc.,

Deerfield, IL) to the respective alcohol (lM, 1 mL).

Following esterification, peptides were dried under vacuum and reconstituted into

aqueous 0.1 % CF3COOH (20 µL) prior to analysis by MALDI-TOFMS and ESI-MS.

38 To ensure complete esterification of the tryptic peptides, an aliquot (0.5 µL) of each

sample was first analysed by MALDI-TOFMS with a.-cyano-4-hydroxycinnamic acid

as the matrix. An aliquot (5 µL) of each was also analysed by ESI-LC/MS/MS. Each

MS/MS spectrum was searched against the non-redundant protein databases to determine the identity of each tryptic peptide (as the butyl ester). Peptides were separated by reverse phase chromatography using a capillary column (100 mm x 150

µm ID) packed with R2 POROS under the conditions previously described.

Following this, the two samples were combined and analysed by ESI-LC/MS under the

same chromatographic conditions. The relative amount of each tryptic peptide was

determined by comparing the peak area of each pair of co-eluting peptides from the respective extracted ion chromatogram.

2.6.4.2. Tear fluid proteins

Proteins in matched open and closed eye tear (n = 2) were precipitated (acetone; 10

volumes) and the protein pellet collected after centrifugation (10,000 g, 10 min). The

pellet was reconstituted in NRJHC03 (20 µL, 25 mM, pH 8) and an aliquot

( corresponding to 10 µg protein) of each sample was incubated with trypsin (0.1 µg,

37°C, overnight). Tryptic peptides were dried under reduced pressure prior to

esterification with either acidified butanol (open eye tear) or acidified d10-butanol

( closed eye tear). Each sample was then dried under vacuum and reconstituted in

aqueous 0.1 % CF3COOH (20 µL). An aliquot of the esterified peptides was then

analysed by ESI-LC/MS/MS on a capillary column (100 mm x 150 µm ID) packed with

R2 POROS under the conditions previously described. Esterified peptides from each

sample were identified by SEQUEST. SEQUEST parameters were adjusted to allow for

39 butanol esterification of the C-tenninal residues, aspartic acid and glutamic acid residues.

Following identification of the esterified tryptic peptides, equal amounts of the two samples were combined and analysed by ESI-LC/MS to determine the relative abundances of the chosen proteins in the sample. Co-eluting esterified peptide pairs were identified and their relative abundances were determined by calculating the peak area ratio for each peptide and its isotopomer in the extracted total ion chromatogram.

Multiple peptides were used to estimate the amount of each protein in the sample. To determine the precision with which these measurements could be made, the ratios obtained for multiple peptides derived from the same parent protein were averaged, and the standard deviation detennined.

40 2. 7. Analysis of the peptide and protein components in human

seminal plasma

2.7.1. Sample collection

Azospennic semen samples were collected from vasectomised human males (n = 7) and

stored at -80°C. Proteins were precipitated with CH3OH : CHCh : H2O ( 4: I :3) and centrifuged before analysis by two dimensional gel electrophoresis (2OE).

Seminal fluid was also collected from normal young males (n = 5) and pooled prior to incubation (37°C, 30 min). Proteins in this sample were separated by 10

electrophoresis, and analysed by ESI-LC/MS/MS and MALDI-TOFMS.

2. 7 .2. Two dimensional gel electrophoresis

(Experimental work detailed in this section was performed in collaboration with Dr. David

Friedman.)

The proteins (100 - 200 µg total) in pooled seminal fluid ( containing protease

inhibitors) were precipitated and the pellet isolated following centrifugation (10,000 g,

10 min). The protein pellet was then resuspended in 2OE rehydration buffer (200 µL,

8M urea, 2 % CHAPS, 18 mM OTT} and passively rehydrated into 18 cm immobilised

pH gradient strips (IPG 4-7 or 6-11; Amersham Pharmacia Biotech, Piscataway, NJ) for

at least 16 hours. First dimension isoelectric focusing was performed for 24 hours

(Multiphor II system; Amersham Pharmacia, Piscataway, IL). Cysteine side-chains were

then reduced by incubating the focused strips in equilibration solution (6 M Urea, 50

mM Tris, pH 8.8, 30% glycerol, 2% SOS, bromophenol blue) containing 1%

dithiothreitol {OTT; 20 min; Sigma Chemical Co., St Louis, MO) followed by

41 alkylation by incubation in equilibration solution containing 2.5% iodoacetamide (20 min; Sigma Chemical Co., St Louis, MO). Second dimension separation was performed

overnight (Protean II XL electrophoresis system, Bio-Rad Laboratories, Hercules, CA) using 8-16 % polyacylamide gradient gels. Proteins were stained with silver as

described by Shevchenko et al. (1996b).

Prior to in-gel digestion with trypsin, protein spots were excised and the silver removed

as described by Gharahdaghi et al. (1999). The gel pieces were then dehydrated with

50% CH3CN in NH.iHCO3 (50 mM) and 100 % CH3CN, followed by vacuum

centrifugation for 5 minutes. Gel pieces were then re-hydrated with trypsin (0.1 µg) in

NH4HCO3 (25 mM) and incubated overnight (30°C). Tryptic peptides were extracted

with CH3CN: 0.1% aqueous CF3COOH (60:40, 15 µL), and dried under vacuum. The

peptides were reconstituted in aqueous 0.1 % CF3COOH (8 µL) and de-salted with

ZipTip C18 pipette tips (Millipore Corporation, Bedford, MA). Peptide mass

fingerprints were then generated by MALDI-TOFMS.

2. 7 .3. One dimensional gel electrophoresis

Seminal fluid (corresponding to ea. 200 µg protein) was diluted into loading buffer (15

µL; 50 mM Tris pH 6.8, 5 % ~-mercaptoethanol, 10 % glycerol, 10 % SDS, 1%

bromophenol blue) before separation by 1D gel electrophoresis. Molecular weight

markers (Kaleidoscope prestained standards; Bio-Rad Laboratories, Hercules, CA) were

loaded into the adjacent lane. Proteins were separated for 50 min at a constant voltage

(180 V) and then visualised by incubation in Coomassie Brilliant Blue (CH3OH :

CH3COOH, 15 min; Fisher Scientific, Houston, TX). Discrete protein bands, as well as

42 regions ofless intense stain (i.e., 26 different bands in total), were excised and destained

(50 % CH3CN in NH.iHC03, 25 mM, pH 8) prior to in-gel digestion (trypsin, 0.3 µg, overnight, 37°C). An aliquot of each digest (5 µL) was then analysed by ESI­

LC/MS/MS.

2. 7 .4. Mass spectrometry of unfractionated seminal fluid

Diluted seminal fluid collected from normal young males (1: 10, 25 mM NH.iHC03, pH

8) was analysed by MALDI-TOFMS with either a-cyano-4-hydroxycinnamic acid or

sinapinic acid (10 mg/mL, CH3CN: 0.1 % aqueous CF3COOH, 80:20) as the matrix.

Spectra were externally mass calibrated to the protonated monoisotopic masses of

angiotensin I ([M+Ht 1,296.68 Da), ACTH clip 18-39 ([M+Ht 2,465.19 Da) and

bovine insulin ([M+Ht 5,734.74 Da). To identify the peptides present in seminal fluid

an aliquot (5 µL) was also analysed by ESI-LC/MS/MS under the conditions described

below.

2. 7 .5. Protein identification

2.7.5.1. MALDI-TOF mass spectrometry

Proteins in seminal fluid (obtained from azospermic men) separated by 2DE were

analysed by MALDI-TOFMS following in-gel tryptic digestion. Peptide mass maps

were collected in reflectron mode with a-cyano-4-hydroxycinnamic acid as matrix and

spectra were internally calibrated to the protonated monoisotopic masses of several

trypsin autolysis peptides ([M+Ht 842.51, 1,045.55 and 2,211.42 Da). Proteins were

43 identified by searching protonated peptide masses (i.e., the [M+Ht values) against the non-redundant protein databases using Protein Prospector (MS-FIT).

2. 7.5.2. ESI mass spectrometry

Peptides derived from tryptic digestion of bands visualised by one dimensional gel electrophoresis, and whole unfractionated seminal fluid, were analysed in a data dependant mode on a quadrupole ion trap mass spectrometer fitted with a nanospray source. Peptides were separated by reverse phase chromatography using a capillary column (100 mm x 150 µm ID) packed with POROS R2. Peptides were eluted with an increasing linear gradient of acidified CH3CN (0.05 % CF3COOH) over one hour at a flow rate of - 0.5 µL/min. The second solvent consisted of aqueous 0.05 % CF3COOH.

The charge state of each eluting peptide was determined prior to fragmentation to produce a product ion spectrum. Proteins were identified by searching each fragment

ion spectrum against the human subset of the non-redundant protein database using the

SEQUEST software. For the identification of peptides in unfractionated seminal fluid,

SEQUEST parameters were adjusted to allow searches with 'No enzyme' specified.

44 2.8. Quantification of biomolecules by MALDI-TOFMS

2.8.1. Analysis of growth hormone in rat pituitary tissue

Male Sprague-Dawley rats (n = 2, > 300 g body weight) were decapitated and their pituitary glands (ea. 15 -20 mg) were surgically removed and stored at -20°C until required. Whole glands were homogenised (probe sonication) and the protein

components extracted into aqueous 0.1 % CF3COOH (1.5 mL). The internal standard,

~-lactoglobulin (15 nmol; Sigma Chemical Co., St. Louis, MO), was then added.

Recombinant methionyl rat growth hormone(> 97 % monomer, 21,943 Da, Gropep Pty.

Ltd., Adelaide, Australia) was used in place of rat growth hormone to generate the

standard curve. Standard solutions containing the recombinant methionyl rat growth

hormone (0, 10, 20, 40, 100, 150 and 200 pmol) and ~-lactoglobulin (100 pmol) were

prepared in aqueous 0.1 % CF3COOH (20 µL). Standard solutions and samples were

diluted 1:5 in caffeic acid solution before analysis, and 1 µL of the final solution was

applied to the MALDI target. Spectra were acquired in linear mode across the mass

range 5,000 - 40,000 Da.

2.8.2. Quantification of insulin in human pancreatic tissue

(This section of the experimental work was undertaken in collaboration with Martin Bucknall.)

Control human pancreatic tissue was obtained from cadavers (n = 2) and frozen at

-20°C until analysis. Sections of each tissue (3 x ea. 50 mg) were homogenised and the

proteins extracted into aqueous 0.1 % CF3COOH (1 mL). Porcine insulin (2.5 nmol;

Sigma Chemical Co., St Louis, MO) was added as the internal standard. The extracts

were passed through C18 solid phase extraction cartridges (Sep-Pak; Waters

45 Corporation, Milford, MA) pre-conditioned with CH3CN: H2O: CF3COOH (80:20:0.1;

5 mL) and equilibrated with aqueous 0.1 % CF3COOH (10 mL). Insulin was eluted in

· CH3CN: H2O: CF3COOH (80:20:0.1; 1 mL). Samples were evaporated under reduced pressure to yield aliquots of ea. 50 µL. These were diluted in sinapinic acid matrix

solution (1 :20).

Standard solutions containing human insulin (0, 40,120,400, 1,200, 2,800, 4,000 pmol)

and porcine insulin (200 pmol) were prepared in aqueous 0.1 % CF3COOH (20 µL)

containing bovine serum albumin (5 nmol/mL; Sigma Chemical Co., St Louis, MO).

Aliquots (1 µL) of each sample and standard solution were applied to the MALDI target

for analysis. Spectra were acquired in both linear and reflector modes.

2.8.3. Analysis of hemorphins in human adrenal and pheochromocytoma tissues

(This section of the experimental work was undertaken in collaboration with Martin Bucknall.)

Control adrenal glands (n = 10) and surgically removed, histologically verified

pheochromocytoma (n = 3) were collected and the peptides extracted as described in

Cerpa-Poljak et al. (1997). The extracts were stored at-20°C until required. Prior to

analysis, the extracts were mixed with the internal standard rat LVV-hemorphin- 7 (300

pmol/mg tissue; Chiron Technologies Pty., Ltd., Clayton, Australia), and diluted 1 :30

with a.-cyano-4-hydroxycinnamic acid solution. All MALDI-TOFMS was performed in

linear mode.

A single tissue extract, previously found to have the lowest endogenous

LVV-hemorphin- 7 concentration, was employed as the matrix for the preparation of a

46 standard addition calibration curve. Aliquots of this sample were spiked with human

LVV-hemorphin- 7 (0, 10, 30, 60, 100, 300 and 600 pmol/mg tissue; Chiron

Technologies Pty., Ltd., Clayton, Australia) and a fixed amount of the internal standard

(300 pmol/mg tissue). Control samples (n = 3) were also included in the analysis. These were prepared from each of the pheochromocytoma extracts by adding internal standard and spiking with human LVV-hemorphin- 7 (100 pmol/mg tissue). The hemorphin content of each extract had previously been determined by electrospray mass spectrometry [Cerpa-Poljak et al., 1997].

2.8.4. Analysis of catecholamines in human adrenal and pheochromocytoma tissue

(This section of the experimental work was undertaken in collaboration with Martin Bucknall.)

Frozen tissue samples (-70°C; ea. 100 mg each) from control adrenal glands (n = 3) and histologically verified pheochromocytoma (n = 3) were extracted into aqueous 0.1 %

CF3COOH (1 mL) as previously described [Cerpa-Poljak et al., 1997].

Aliquots (10 µL) from each tissue extract were mixed with the internal standards [2H3]­

epinephrine (2 nmol) and [2H3]-norepinephrine (3 nmol) (Cambridge Isotope

Laboratories, Andover, MA) and diluted 1 :25 in ferulic acid matrix solution. Samples

were mixed, cooled (4°C, 10 minutes) and centrifuged (14,000 g, 10 min) to precipitate

protein. Aliquots (0.25 µL) of the supernatant were applied directly on the MALDI

target for analysis.

An extract of surgically resected adrenal cortex was employed as the matrix for the

preparation of a standard addition calibration curve. Aliquots of this sample, each

47 corresponding to 5 mg of tissue, were spiked with 0, 0.5, 1.5, 3, 5, 15, 30, 45, 60, 90 nmol of both epinephrine (Sigma Chemical Co., St Louis, MO) and norepinephrine

(Sigma Chemical Co., St Louis, MO), together with fixed amounts of the internal

standards ([2H3]-epinephrine (20 nmol) and [2H3]-norepinephrine (30 nmol)). These

spiked extracts were diluted 1 :25 in ferulic acid solution prior to analysis. All mass

spectra were acquired in reflector mode at a reduced acceleration voltage (6 kV).

2.8.S. Analysis of homovanillic acid in human urine samples

Urine samples were collected from 19 healthy volunteers and adjusted to pH 2.5 (HCl,

lM). The internal standard, [2Hs]-homovanillic acid ([2H5]-HVA; 100 nmol; Cambridge

Isotope Laboratory, Andover, MA), was added to aliquots of each sample (50 µL) and

the mixture extracted into ethyl acetate (4 volumes, 3 times). Extracts were combined,

dried under reduced pressure, and the residue reconstituted into dilute HCI (0.1 M, 50

µL ). Samples were diluted 1: 10 in 2,5-dihydroxybenzoic acid matrix solution and

applied to the MALDI target. Each urine sample was also independently analysed for

homovanillic acid (HVA} content at a commercial clinical chemistry laboratory.

Standard solutions containing HVA (0, 25, 50, 100, 500 and 1,000 pmol; Sigma

Chemical Co., St. Louis, MO) and [2H5]-HVA (100 pmol) were prepared in water and

diluted 1 :5 in the matrix solution (2,5-dihydroxybenzoic acid) prior to analysis (final

volume 20 µL). An aliquot (1 µL) of each sample and standard solution was applied to

the MALDI target. Spectra were acquired in reflectron mode at a reduced acceleration

voltage (6 kV) and over the mass range 180-190 Da.

48 2.8.6. Data acquisition and processing

Spectra were acquired in autosampler mode with the laser operated at a fixed fluence.

Laser firing patterns started at the center of each sample spot and spiraled progressively outwards to the spot perimeter. The laser was programmed to fire once at each location in the firing pattern. For each sample spot, spectra obtained from 256 laser shots, and meeting the specified acceptance criteria, were averaged to form a single spectrum.

Acceptance criteria consisted of a signal-to-noise ratio (S / N) greater than 3 and absolute ion intensities between 5,000 and 62,000 counts over a restricted mass range inclusive of both the analyte and internal standard. Spectra from ten sample spots were

collected for each standard solution and biological sample.

From each individual spectrum the heights of the relevant peaks (i.e., analyte and

internal standard) were determined and the analyte to internal standard peak height ratio

was calculated. Each standard curve was constructed by plotting the calculated peak

height (or peak area) ratio (y-axis) against the molar ratio of analyte to internal standard

(x-axis). The mean value and standard deviation for each standard were determined and

plotted to generate the standard curve.

For homovanillic acid, peak area ratios of analyte to internal standard were also

determined to construct a standard curve. The equation for each standard curve was

calculated using an unweighted linear regression.

49 CHAPTER3

MASS SPECTROMETRY AND

PROTEIN IDENTIFICATION:

APPLICATION TO HUMAN TEAR

FLUID

50 3.1. Introduction

The human tear film is believed to be composed of three layers: a superficial layer composed of lipids secreted by the Meibomian gland, a middle aqueous component consisting primarily of peptides and proteins, and an inner mucosa! layer [Coyle et al.,

1989; Wollensak: et al., 1990]. Together these components are believed to play an essential role in the protection of the external ocular surface from potential pathogens, as well as to provide nourishment to the corneal surface [Coyle et al., 1989; Wollensak: et al., 1990]. The blinking action serves to spread the tear film over the entire surface of the eye, as well as to remove contaminants and waste products by drainage into the nasolacrimal duct.

There are many physiological factors that can alter the balance of the protein components in the tear film, or affect the rate of tear production from the lacrimal gland or its accessory glands. These include specific ocular pathologies such as glaucoma, dry eye syndrome, Sjorgren's syndrome and conjunctivitis, as well as systemic diseases such as diabetes and rheumatoid arthritis [Jensen et al., 1986; Coyle and Sibony, 1987;

Coyle et al., 1987; Khalil et al., 1988; Mavra et al., 1990; Domingo et al., 1998;

Goebbels, 2000; Thakur and Willcox, 2000]. In addition to these, it is also believed that procedures routinely undertaken to improve visual acuity, such as laser eye surgery or the use of contact lenses, can alter the constituents of the tear film [Tervo et al., 1995;

Tervo et al., 1997; Tuominen et al., 1999; Thakur and Willcox, 2000]. Currently, little information is available concerning the nature and extent of changes in protein expression in various disease states.

51 Although it has been proposed that changes in protein expression in tear fluid can be used for diagnostic purposes [Molloy et al., 1997], the protein composition of the normal tear film is poorly defined. Large scale studies involving the identification of tear proteins by two dimensional gel electrophoresis have been conducted employing either amino acid analysis or Edman sequencing [Mii et al., 1992; Molloy et al., 1997].

Although many proteins were detected by this approach, only a small subset of these

could be identified due to post-translational processing such as glycosylation and N­ terminal processing.

Other studies have focused on the identification of protein components by techniques

such as radioimmunoassay, gel electrophoresis, high performance liquid

chromatography, immunochemical techniques, and enzyme activity assays [Coyle and

Sibony, 1986; Boukes et al., 1987; Fullard, 1988; Coyle et al., 1989; Baier et al., 1990;

Wollensak: et al., 1990; Kuizenga et al., 1991b; Mii et al., 1992; Molloy et al., 1997;

Behndig et al., 1998; Jumblatt et al., 1999]. As a result of these investigations,

lactotransferrin, tear lipocalin (von Ebner's gland protein), secretory IgA and lysozyme

have been identified as the major protein components of the normal tear film [Janssen

and van Bijsterveld, 1983b; Kijlstra et al., 1983; Fullard and Kissner, 1991]. In addition

to these, various enzymes and serum proteins have also been detected and identified

[Crouch et al., 1991].

As discussed earlier in this thesis, advances in mass spectrometry have contributed to

substantial progress in peptide and protein identification in biological tissues and fluids.

Both MALDI-TOFMS and ESI-MS provide the sensitivity and specificity required to

detect and identify multiple components in trace quantities of complex biological

52 samples. Post-translational modifications can also be characterised, and many proteins

can be identified in a single analysis.

In this study, the peptide and protein constituents of the normal human tear film were

investigated by a combination ofMALDI-TOFMS and ESI-LC/MS. Because MALDI­

TOFMS enables the intact mass of molecules to be accurately determined, it was

employed to survey samples and establish a profile of the peptides and proteins present

in untreated human tear. In addition, MALDI-PSD was used to identify several of the major peptide constituents of tear. Signal suppression effects are significant when

MALDI-TOFMS is employed to examine complex samples, and this limits the amount

of information available. Therefore, ESI-LC/MS/MS was also employed to identify

protein constituents following proteolytic digestion.

3.2. Methods

The methods employed for this study are detailed in Chapter 2 (pages 36- 38).

53 3.3. Results

3.3.1. The peptide components of human open eye tear

Human tear fluid (n = 7) was directly analysed by MALDI-TOFMS using, separately, both a.-cyano-4-hydroxycinnamic acid and sinapinic acid as the matrix. Over 30

components were detected by this technique (Figure 3.1). Lysozyme and tear lipocalin, two of the most abundant proteins in the tear film, were detected and tentatively identified based on their theoretical molecular weight. In addition to these, numerous other components were detected below 5,000 Da.

To establish the identity of the major low mass constituents(< 2,000 Da), sequence information for five of the more abundant peptide species was obtained by post-source

decay (PSD). The PSD spectrum for each of the five peptides is shown in Figure 3.2.

Indicated on each PSD spectrum are the b- and y- series ions. In addition to these,

internal sequence ions and immonium ions were also detected. The mass and amino acid

sequence for the fragment ions detected for each peptide are listed in Tables 3.1 - 3.5.

54 Figure 3.1. A MALDI-TOF mass spectrum of human tear.

An aliquot of human tear (0.2 µL) was analysed by MALDI-TOFMS using a-cyano-4- hydroxycinnamic acid as the MALDI matrix. Multiple components were detected in the mass range of 500 - 20,000 Da including proteins such as lysozyme ([M+Ht 14,678

Da) and tear lipocalin ([M+Ht 17,438 Da), as well as many other peptide constituents.

(As shown, the mass range 2,000- 5,000 Dais magnified by a factor 10.) The peptides

selected for post-source decay analysis are indicated.

55 56

5000

lipocalin

4400

(17438.1)

Tear

I

l

4049.2

3800

3749.3

10

X

(14687.8)

Lysozyme

3336.9

14906.4

3200 /

II

2940.1

2876.5

272315 I

I I I

2600

2423.7

12411.7

I

I

(m/z)

~7·

I

....

:

.

!2J4.5 :I

I

2000

Mass

I

1786.9

tear.

1629.9

human

of

1400 1700

8713.4

1330.7

1326.8

for

\

spectrum

\----1

7.5

1100

mass \

\ 1

7340.7

1

chosen

analysis

PSD

800

Peptides

MALDI-TOF

A

3.1.

0500

100

100

II) C

> ea

G)

s

~

0 ~

'ii - .5 - a: Figure - Figure 3.2. Post-source decay spectra for the abundant tear peptides.

A. A post-source decay spectrum for the peptide [M+Ht 1,629.69 Da.

B. A post-source decay spectrum for the peptide [M+Ht 1,443.74 Da.

C. A post-source decay spectrum for the peptide [M+Ht 1,330.52 Da.

D. A post-source decay spectrum for the peptide [M+Ht 1,105.51 Da.

E. A post-source decay spectrum for the peptide [M+Ht 848.45 Da.

Indicated on each spectrum are the fragment ions corresponding to either b- and y­

series ions as well as immonium ions specific to particular amino acids. The mass and

amino acid sequence for the fragment ions detected for each peptide are listed in Tables

3.1 - 3.5.

57 58

II) ~

600

.0

1700

~

~'

I

II) 1

""" ,-

.0

V--

I

l I\.

' u

[M+Ht +

co 1629.69

:E .... ;

w

! ·V

'

'

'

~

I

I

I:'

1

NN ,,....

>>

"""

,-

490

I

,,

1480

N

,- '

+

,,,

co ,- .0

I I l,

-:I'

,/'

\

.0

N or-

.0

'

.

'

.

'~'.//~'II~.

. ~·.:. I

.... ,-

>,

I

C')

>,

;

...

' .i

'

, 380

1260

~~~~,~~

I

I

""" ,- .i

·

.,

CJ)

I

·

·

I

CJ) 1

.

>>

""" I C') ,-

Da). WI CU

""" ,-

I

111'

I

{m/z)

, •

CJ)

~~

.·~~

,- """

.0

1,629.69

,; ....

·

peptides.

Mass

"'

'

~I

.0

CJ) 1 I

"'

270

""" ,-

1040 tear I

I I ' "'

""" ,-

.0 Y~

'

([M+Ht

i

abundant

'1'L! I ...

>,

'

peptide

the

.1

tear ~~wi

for

a

...

,

for

160

820

spectra

..

decay

spectrum

/

I

·- ::J:

CD

.0 uL~

>

..J

I CD

decay """ ,-

.0

::::,u,u,0) G)

..J

CD

CU 0 ..

a.

Post-source

0) .. I CD

<(

CU

""" ,-

.,

so

3.2.

0

•J:""'""'"'·~

100

100

post-source

A

C

"#. i - - I 'tu -~ ~

Figure

A. Table 3.1. Fragment ions detected in the post-source decay spectra of the peptide

[M+Ht 1,629.69 Da.

Experimental [M+Ht values are the average (± SD) of at least three separate determinations.

A. Fragment ions corresponding to b- and y- series ions.

THEORETICAL EXPT. THEORETICAL EXPT. BIONS YIONS [M+Ht [M+Ht [M+Ht [M+Ht B12 + H2O 1,443.7 1,444.4 ± 0.6 Y12 1,514.8 1,515.2 ± 0.6 B11+H2O 1,330.7 1,329.4 ± 0.9 Y12-NH3 1,497.8 1,498.6 ± 0.6 B11 1,312.7 1,312.2 ± 0.6 Y11 1,358.7 1,359.2 ± 0.4 B9 1,087.5 1,087.7 ± 0.3 Y9 1,190.6 1,190.6 ± 0.5 B9-NH3 1,070.5 1,071.2 ± 0.6 Y9-NH3 1,173.6 1,172.8 ± 0.5 A9-NH3 1,042.5 1,043.3 ± 0.6 Ys-NH3 1,017.5 1,018.4 ± 0.4 B6 733.4 734.1 ± 0.5 Ys 672.3 672.0 ± 0.9 B6-NH3 716.4 717.1±0.5 *Y1 205.1 205.4 A6 705.4 705.6 ± 0.2 ~-NH3 688.4 688.5 ± 0.4 Bs 596.3 597.2 ± 0.7 Bs-NH3 579.3 580.4 ± 0.9 B4 440.2 439.6 ± 0.1 B4-NH3 423.2 422.9 ± 0.8 *~ 412.2 412.4 B3-NH3 352.2 352.6 ± 0.2 B2 272.1 272.6 ± 0.4 B2-NH3 255.1 255.4 ± 0.1

• Due to the low abundance of this peptide in some samples, these fragment ions were detected in only one of the PSD spectra.

59 Table 3.1. Fragment ions detected in the post-source decay spectra of the peptide

[M+Ht 1,629.69 Da.

Experimental [M+Ht values are the average (± SD) of at least three separate determinations.

B. Internal sequence ions.

INTERNAL THEORETICAL EXPERIMENTAL SEQUENCE IONS rM+Ht [M+Ht PA 169.1 169.3 ± 0.1 PQ-28 198.1 198.5 ± 0.2 AR-NH3 211.1 211.6 ± 0.3 PQorQP 226.1 226.5 ± 0.1 HP 235.1 235.4 ± 0.4 EQorQE 258.1 258.4 ± 0.1 RPAorPAR 325.2 325.6± 0.2 RPA-NH3 308.2 308.7 ± 0.3 PAR-NH3 EQPorPQE 355.1 355.4 ± 0.2 QEQ 386.2 386.4 ± 0.3 PARH-NH3 445.2 445.3 ± 0.2 QEQPorPQEQ 483.2 483.6 ± 0.3 *HPQE 492.2 492.5 PQEQP 580.3 580.4 ± 0.9 HPQEQ 620.3 620.6 ± 0.4 RHPQE-NH3 631.3 630.8 ± 0.8 HPQEQP 717.3 717.0 ± 0.3

* Due to the low abundance of this peptide in some samples, this fragment ion was detected in only one of the PSD spectra.

60

61 61

750 750

1480 1480

.E .E

I I

\ \

! !

vut vut

[M+Ht [M+Ht

1443.74 1443.74

a, a,

I I

(U (U

......

,... ,...

LJ1 LJ1

I I

+ +

:c :c

:i :i

......

,-- co co

,... ,...

+ +

I I

......

......

610 610

1334 1334

1u 1u

I I

......

......

>> >>

......

,... ,...

v--

I I

......

......

.0 .0

,... ,...

~ ~

¼~~~~ ¼~~~~

470 470

1188 1188

0 0

~ ~

: :

I I

......

,... ,...

.d .d

(m/z) (m/z)

0, 0,

.0 .0

Da). Da).

Mass Mass

!I !I

. .

0, 0,

I I

! !

! !

0, 0,

>.0 >.0

0) 0)

I I I

:I :I

1,443.74 1,443.74

(U (U

......

,...,...,... ,...,...,...

peptides peptides

330 330

1042 1042

tear tear

a) a)

I I

> >

([M+Ht ([M+Ht

N N

I I

5 5

abundant abundant

......

.0 .0

,... ,...

peptide peptide

X X

the the

tear tear

for for

a a

for for

896 896

190 190

spectra spectra

decay decay

spectrum spectrum

~ ~

e> e>

c( c(

......

I I

> >

......

,... ,...

en en

-

:c :c

decay decay

Post-source Post-source

~ ~

0 0

a. a.

1y~~~~AAh~~~~wv~~ 1y~~~~AAh~~~~wv~~

3.2. 3.2.

750 750

0 0

post-source post-source

100 100

100 100

A A

~ ~

C C

0 0

a: a:

i i

! !

'i 'i

~ ~

0 0

::,!! ::,!!

-

-

Figure Figure B. B. Table 3.2. Fragment ions detected in the post-source decay spectra of the peptide

[M+Ht 1,443.74 Da.

Experimental [M+Ht values are the average(± SD) of three separate determinations.

A. Fragment ions corresponding to b- and y- series ions.

THEORETICAL EXPT. THEORETICAL EXPT. BIONS YIONS [M+Ht [M+Ht [M+Ht [M+Ht B11 +H2O 1,330.6 1,329.8 ± 0.1 Yu 1,328.7 1,329.8 ± 0.1 * B11 -NH3 1,295.6 1,294.9 Y11 -NH3 1,311.7 1,311.6 ± 0.1 *B9 1,087.5 1,088.3 Y10 1,172.6 1,172.5±0.1 *B9-NH3 1,070.5 1,070.7 Y9-NH3 1,058.5 1,059.1 ± 0.5 A9-NH3 1,042.5 1,042.7 ± 0.2 Ys 1,004.5 1,004.8 ± 0.4 Bs 958.5 958.6± 0.4 Y1-NH3 831.4 831.2 ± 0.1 B6 733.4 733.3 ± 0.4 *Y6 711.4 712.4 ~-NH3 688.4 688.3 ± 0.4 *Y2 229.2 229.4 *Bs-NH3 579.3 579.8 *B4 440.2 440.1 *B4-NH3 423.2 423.1 *B2 272.1 272.1 B2-NH3 255.1 255.3 ± 0.1

• Due to the low abundance of this peptide in some samples, these fragment ions were detected in only one of the PSD spectra.

62 Table 3.2. Fragment ions detected in the post-source decay spectra of the peptide

[M+Ht 1,443.74 Da.

Experimental [M+Ht values are the average(± SO) of three separate determinations.

B. Internal sequence ions.

INTERNAL THEORETICAL EXPERIMENTAL

SEQUENCE IONS [M+Ht [M+Ht PA 169.1 169.2 ± 0.2 PQ 226.1 226.4 ± 0.1 *PQ-28 198.1 198.3 HP 235.1 235.3 ± 0.1 PQEorEQP 355.2 355.3 ± 0.1 RPA-NH3 308.2 308.5 ± 0.2 PAR-NH3 PQEQ 483.2 483.5 ± 0.1 PARHPQ-NH3 670.3 671.7 ± 0.9 RHPQEQP 873.4 872.2 ± 0.1

• Due to the low abundance of this peptide in some samples, this fragment ion was detected in only one of the PSD spectra.

63 I

700

64

1370

I

cl

.....

.....

I~

~/\

~

I

[M+Ht

+

1330.78

co .....

..... :c

+

1~

i

U')

11

.c

~~,,.~~

\

I U')

>-

11

.....

,...

0

~ll~

+ \

co ,...

.c

570

1236

~·~r~

\~

0

.c

I

~

I \

0

......

.....

,...

.c

~~

I

0 ....

co

.....

.....

~~

. I\

1

.

l1

440

1102

en

!1

.c

c:n

I

Wl,M,)~~,;J

......

o,

>-.0

I

en

(m/z)

>-

......

.

~~y~~

I

Mass

Da) ~\

\

1,330.52

310

968

peptides.

I

tear

\~lm;~~~

I

([M+Ht

~

...... i .....

w

~ ,..

>-

abundant

!

peptide

the

tear

for

a

180

834

for

spectra

en

:i..

decay

spectrum

. \~~~~

,,,

·- :c

co

~~~~

decay

.c

0 ...

n.

I co

......

.c

Post-source

50

0

3.2.

100

1001

post-source

A

C

C Cl) > ea

~

.! ,:::e.

~ ·;; - :;::; 'i a:

Figure

C. Table 3.3. Fragment ions detected in the post-source decay spectra of the peptide

[M+Ht 1,330.52 Da.

Experimental [M+Ht values are the average (± SD) of at least two separate

determinations.

A. Fragment ions corresponding to b- and y- series ions.

THEORETICAL EXPT. THEORETICAL EXPT. BIONS YIONS [M+Ht [M+Ht [M+Ht [~+Ht B1o+H2O 1,233.6 1,233.7 ± 0.1 Y10 1,215.6 1,215.8 ± 0.1

B10 1,215.6 1,215.7 ± 0.1 *Y10-NH3 1,198.6 1,198.7 B10-NH3 1,198.6 1,198.7 ± 0.1 Y9 1,059.5 1,060.1 ± 0.4 A10-NH3 1,170.6 1,170.6±0.1 *Y9-NH3 1,042.5 1,042.6

B9 1,087.5 1,088.0 ± 0.6 *Y1 891.4 891.4 *B9-NH3 1,070.5 1,070.8 Ys-NH3 581.2 580.6 ± 0.4 A9 1,059.5 1,060.1 ± 0.4 Y4-NH3 484.2 484.1 ± 0.1 *A9-NH3 1,042.5 1042.6 Y2-NH3 227.1 226.9 ± 0.5 *B6 733.4 733.9 *B6-NH3 716.4 717.2 *A6-NH3 688.4 688.3 *~-NH3 598.3 597.5 Bs-NH3 581.3 580.6 ± 0.4 *B2 272.1 272.7 B2-NH3 255.1 255.5 ± 0.1 A2-NH3 227.1 226.9± 0.5

* Due to the low abundance of this peptide in some samples, these fragment ions were detected in only one of the PSD spectra.

65 Table 3.3. Fragment ions detected in the post-source decay spectra of the peptide

[M+Ht 1,330.52 Da.

Experimental [M+Ht values are the average (± SD) of at least two separate determinations.

B. Internal sequence ions.

INTERNAL THEORETICAL EXPERIMENTAL

SEQUENCE IONS [M+Ht [M+Ht PA 169.1 169.3 ± 0.2 PQ-28 198.1 198.4 ± 0.3 *PQ-NH3 209.1 209.2 HP 235.1 235.4 ± 0.3 RPA-NH3 308.2 308.6 ± 0.2 PAR-NH3 PQE 355.2 355.7 ± 0.2 *HPQE 492.2 492.8 *ARHPQ-NH3 562.3 563.2

• Due to the low abundance of this peptide in some samples, these fragment ions were detected in only one of the PSD spectra.

66 570

1150

U'I

67

co

[MtHt I

1105.51 +

co

'I""

:c: ...... - :5

+

466

1034

co

I

>-

Cl)

+

'I""

co

.0

I co

>-

r--

'I""

Cl)

.0

;.

362

918

(m/z)

,.._

Da).

>-

"'

.0

-~·

Mass

258

802

I

1,105.51

,.._

'I""

.E

peptides.

tear

([M+Ht

,.._._,

abundant -~~~~ij~,~~~

peptide

the

tear

for

a

154

686

for

0, ...

spectra

<(

"'

:c: ·-

decay

spectrum

0,

<(

U'I

.0

0

......

decay

0..

i

Post-source

50

0570

0

100

100

3.2.

>- C

C > a,

G)

post-source

s

~ -

~ 'i

:;:::: - G)

a:

A

Figure

D. Table 3.4. Fragment ions detected in the post-source decay spectra of the peptide

[M+Ht 1,105.51 Da.

Experimental [M+Ht values are the average(± SD) of three separate determinations.

A. Fragment ions corresponding to b- and y- series ions.

THEORETICAL EXPT. THEORETICAL EXPT. BIONS YIONS [M+Ht [M+Ht [M+Ht [M+Ht Bs +H2O 976.5 976.5 ± 0.5 Ys 990.5 990.8 ± 0.5 Bs 958.5 959.9± 0.6 *Ys-NH3 973.5 973.6 * B6 733.4 734.8 Y1 834.4 834.6 ± 0.5 *A6 705.4 705.4 Ys 666.3 667.1 ± 0.1 A6-NH3 688.4 688.7 ± 0.4 Y4 510.2 510.6 ± 0.3 Bs 596.3 596.4 ± 0.6 Y4-NH3 493.2 492.8 ± 0.5 *B4 440.2 439.5 Y3 373.2 373.6 ± 0.3 B2 272.1 272.3 ± 0.1 B2-NH3 255.1 255.2 ± 0.2

* Due to the low abundance of this peptide in some samples, these fragment ions were detected in only one of the PSD spectra.

68 Table 3.4. Fragment ions detected in the post-source decay spectra of the peptide

[M+Ht 1,105.51 Da.

Experimental [M+Ht values are the average(± SD) of three separate determinations.

B. Internal sequence ions.

INTERNAL THEORETICAL EXPERIMENTAL

SEQUENCE IONS [M+Ht [M+Ht PA 169.1 169.3 ± 0.1 *PQ-28 198.1 198.1 PQ 226.1 226.2 ± 0.1 HP 235.1 235.1 ± 0.3 RPAorPAR 325.2 325.5 ± 0.2 RPA-NH3 308.2 308.4 ± 0.2 PAR-NH3 ARHPorPARH 462.3 462.3 ± 0.4

• Due to the low abundance of this peptide in some samples, this fragment ion was detected in only one of the PSD spectra.

69 70

1

400

870

~

1

848.37

[M+Ht

I

+

T""

a, == ..... :x: .....

+

330

n&

~~

CO

a, T""

.c

I

ID

>-

,_8 I

I ,J

I

o

T"" .....

1,.8c5+

>-

T"" r-,.

COCOi..

I

as

(\I

.c

I

CO

as

r-,.

T""

I 260 (\I

682

T"" .....

.c

(\I

>-

(m/z)

Da).

Mass

848.37

peptides.

II)

.c

190

588

tear

II')

>-

([M+Ht

abundant

peptide

the

tear

for

a

for

~b~~~~

C, ... t'

~

spectra

120

'

494

(I)

:x: ·-

,-.d•

decay

,.

spectrum

...

<:I'

>-

decay

I

<:I'

>

T"" .....

0 ...

0..

Post-source

3.2.

__;~fw~~,w~lw~~Jw~1

~

post-source

400

0

100 A

100

Figure

E.

C

i CU

~

'#- - ·;;; - I :;:: I Table 3.5. Fragment ions detected in the post-source decay spectra of the peptide

[M+Ht848.37 Da.

Experimental [M+Ht values are the average (± SD) of at least two determinations.

A. Fragment ions corresponding to b- and y- series ions.

THEORETICAL EXPT. THEORETICAL EXPT. BIONS YIONS [M+Ht [M+Ht [M+Ht [M+Ht B6+H2O 751.4 751.4 ± 0.1 y6 733.4 733.5 ± 0.1 B6 733.4 733.5 ± 0.1 Y6-NH3 716.4 716.7 ± 0.1 B6-NH3 716.4 716.7 ± 0.1 Ys 577.3 577.6 ± 0.1 ¾, 705.4 705.7 ± 0.1 Y4 480.3 480.3 ± 0.1 A6-NH3 688.4 688.2 ± 0.2 Y4-NH3 463.2 463.1 ± 0.5 Bs + H20 614.3 614.5 ± 0.4 Y3 409.2 409.1 ± 0.1 Bs 596.3 596.4 ± 0.1 Y2 253.1 253.2 ± 0.1 As 568.3 568.3 ± 0.1 84 440.2 439.3 ± 0.1 B2 272.1 272.4 ± 0.1 B2-NH3 255.1 255.2 ± 0.1

71 Table 3.5. Fragment ions detected in the post-source decay spectra of the peptide

[M+Ht 848.37 Da.

Experimental [M+Ht values are the average (± SO) of at least two determinations.

B. Internal sequence ions.

INTERNAL THEORETICAL EXPERIMENTAL

SEQUENCE IONS [M+Ht [M+Ht PA 169.1 169.2 ± 0.1 AR-NH3 211.1 211.8 ± 0.8 RPAorPAR 325.2 325.2 ± 0.2 RPA-NH3 308.2 308.2 ± 0.2 PAR-NH3 PARH-NH3 445.2 445.1 ± 0.4 RPARH 618.4 618.5 ± 0.2

72 Post-source decay primarily results in fragmentation about the amide bond of the peptide backbone resulting in the formation of b- and y- series ions [Roepstorff and

Pohlman, 1984; Biemann, 1988; Biemann, 1992]. In addition to this, fragment ions

corresponding to amino acid side chain losses, as well as low mass immonium ions

specific to individual amino acids, are also often observed [Biemann, 1992]. The PSD

spectra for all peptides analysed in this study revealed an almost complete b- and y­

series of ions, as well as several internal fragment ions and immonium ions.

Comparison of each PSD spectrum revealed fragment ions that were common to each peptide, indicating that these shared at least some sequence homology. De novo

sequencing, in addition to database searching using MS-TAG (Protein Prospector),

indicated that these peptides share a common N-terminus and that the largest peptide

([M+Ht 1,629.83 Da) is sequentially cleaved from the C-terminus to give rise to the

smaller peptides. The amino acid sequences of each of these peptides are shown in

Table 3.6.

To determine the origin of these peptides, the fragment ion data were searched against

the NCBI non-redundant database using MS-TAG (Protein Prospector). These peptides

matched the sequence of the C-terminus of a proline-rich protein reported to be

synthesized by the lacrimal gland (Accession number 2135904) [Dickinson and Thiesse,

1995]. Previously, only the mRNA for this protein had been detected in the human

lacrimal gland, and this is the first report of the presence of the protein itself in human

tear fluid. The amino acid sequence for the lacrimal proline-rich protein is shown in

Figure 3.3.

73 Table 3.6. The amino acid sequence of five of the most abundant peptides detected in human tear.

These peptides were sequenced by post-source decay. Database searching revealed that

these peptides originated from the C-terminus of a lacrimal proline-rich protein.

Experimental [M+Ht values are the average (± SD) of seven determinations.

THEORETICAL EXPERIMENTAL PEPTIDE SEQUENCE [M+Ht (Da) [M+Ht (Da) DRP ARHPQEQPLW 1,629.83 1,629.69 ± 0.40

DRPARHPQEQPL 1,443.75 1,443.74 ± 0.03

DRPARHPQEQP 1,330.66 1,330.52 ± 0.16

DRPARHPQE 1,105.55 1,105.51 ± 0.07

DRPARHP 848.45 848.37 ± 0.13

74 Figure 3.3. The amino acid sequence for the intact lacrimal praline-rich protein.

This protein consists of 134 amino acid residues and has a theoretical molecular weight of 15,096 Da. Indicated is the C-terminal peptide (13 amino acids) that forms the largest of the five peptides detected in open eye tear.

MLLVLLSVVL LALSSAQSTD NDVNYEDFTF TIPDVEDSSQ

RPDQGPQRPP PEGLLPRPPG DSGNQDDGPQ QRPPKPGGHH

RHPPPPPFQN QQRPPQRGHR QLSLPRFPSV SLQEASSFFR

RDRPARHPQE QPLW

Figure legend.

C-te1minal peptide detected in the MALDI-TOF mass spectrum of open eye tear.

75 3.3.2. The detection and identification of lacrimal proline-rich protein

To investigate the possible presence of the intact lacrimal proline-rich protein, tear fluid was separated by size exclusion chromatography (SEC) and the fractions of interest

(i.e., those corresponding to the approximate theoretical mass of the protein of interest) were further characterised by mass spectrometry. MALDI-TOFMS analysis of each fraction indicated the presence of multiple components. The molecular weights of the proteins detected were consistent with the reported theoretical masses oflysozyme

(14,693 Da) and tear lipocalin (17,444 Da). In addition to these, several other protein species consistent in mass to truncated forms of tear lipocalin were also detected in the fractions analysed [Fullard and Kissner, 1991]. Although a signal for the theoretical mass of the intact secreted form of the lacrimal proline-rich protein (13,458 Da) was not detected in any of the fractions analysed, a protein of mass 11,731 ± 3 Da (n = 10) was

evident in some of these fractions. Figure 3.4 shows an example of a MALDI-TOF mass

spectrum for one of the SEC fractions.

76 77

25000

lipocalin

lipocalin

tear

of

tear

fluid. protein

21300

form

tear

Intact

proline-rich

Truncated

ofhuman

/

~.,11-r.;tw;t~~,rlvM\~~of.v'N./M¼,

\

lacrimal .....

,,

~

of /~

17600

/

fractions

\!\;

!

form

I

1M~.O

_!\\

SEC

the

(m/z)

of

Truncated

Mass

one

for

13900

/

¼f#~~~~r1lv~~~~}/t

11733.1

spectrum

/I

I~

J

I

mass

10200

~~~~

~

~~

I

MALDI-TOF

ofa

'flMRIMIJ

6500

O+------~

100

U)

C

i

example

I

~ ~

a: "i ;

- -

An

3.4.

Figure To confirm the identity of the proteins in the SEC fractions that were detected by

MALDI-TOFMS, an aliquot from each was treated with both trypsin and Asp-N.

Analysis detected peptide masses that are predicted to be derived from lysozyme and tear lipocalin, as well as peptides predicted to originate from lacrimal proline-rich protein.

Following this, the amino acid sequence of the proteolytic peptides from these fractions was obtained by ESI-LC/MS/MS. Each MS/MS spectrum was searched against the non­ redundant protein databases, and this confirmed the presence oflysozyme and tear lipocalin, as well as the presence of the lacrimal proline-rich protein. Using this

approach, greater than 50% coverage of the lacrimal proline-rich protein was obtained

(Figure 3.5).

The molecular weight of the intact secreted form of this lacrimal proline rich protein is

reported to be 13,458 Da and this corresponds to residues 17 - 134, with residue 17

being . Although the secreted form of the lacrimal proline-rich protein

was not detected by MALDI-TOFMS in any of the SEC fractions analysed, a protein of

mass 11,731.3 ± 3 .1 Da (n = 10) was detected in all of the fractions that showed

evidence for the presence of the pro line-rich protein derived peptides. This same species

(i.e., 11,731 ± 3 Da) is also evident in the MALDI-TOF mass spectrum of intact human

tear. (See Figure 3.1, p 56.) This mass is consistent with the molecular weight of

residues 18 - 121 (11,733 Da) of the intact lacrimal proline-rich protein, indicating that

the N-terminal pyroglutamic acid residue is also removed. Additionally, residues 122-

134 (i.e., [M+Ht 1,629.83 Da), corresponding to the C-terminal peptide detected in

tear, is found in all the tear samples examined (Figure 3 .5).

78 Figure 3.5. Amino acid coverage of the lacrimal proline-rich protein following its isolation by size exclusion chromatography.

Coverage was determined by proteolytic cleavage using trypsin and Asp-N. The resulting peptides were detected by MALDI-TOFMS and ESI-LC/MS/MS was used to identify the parent protein. Also indicated is the portion of the lacrimal proline-rich protein that is detected in the MALDI-TOF mass spectrum of open eye tear fluid (i.e., residues 18 - 121).

18 MLLVLLSVVL LALSSAQETD NDVNYEDFTF TIPDVEDSSQ

RPDQGPQRPP PEGLLPRPPG DSGNQDDGPQ QRPPKPGGHH

121 ~!~P!_!'9_!'1_~~~~~~~ g~~1:_P_!U'PSV SLQEASSwt

DRPARHPQE QPLW

Figure legend.

C-terminal peptide detected in unfractionated tear.

Peptides identified following Asp-N/trypsin digestion of SEC fractions.

79 3.3.3. Analysis of unfractionated human tear proteins by ESI-LC/MS/MS

To confirm the identity of the proteins detected by MALDI-TOFMS, and to identify other proteins present in tear film, ESI-LC/MS was performed. This was necessary because signal suppression - a common problem associated with MALDI-TOFMS of complex mixtures - limits what can be determined directly. Just as importantly,

MALDI-TOFMS only provides molecular weight information, so at best offers tentative identification of proteins. Therefore, additional studies were undertaken. An aliquot of tear sample was digested with trypsin and the resulting tryptic peptides were then directly analysed by ESI-LC/MS/MS using reverse phase chromatography.

In excess of 2,000 tryptic peptides were detected, matched by SEQUEST, and identified

as constituent peptides of known proteins. The base peak chromatogram for the tryptic

digest of human tear is shown in Figure 3.6. Positive protein identification was based on

the detection and sequencing of multiple peptides derived from the same protein. Over

100 known tear constituents were detected and identified (Table 3.7, p 84), including

serum-based proteins, cytokines, growth factors, immunoglobulins, proteins with

antibacteriolytic or antimicrobial function, a large number of enzymes, and tear-specific

proteins such as lactotransferrin and tear lipocalin.

80 Figure 3.6. The base peak chromatogram of the tryptic peptides (m/z 500-2,000) detected in human tear plotted over four distinct mass ranges.

To identify proteins in human tear, an aliquot (10 µL) was digested with trypsin and analysed by ESI-LC/MS/MS. Shown is the total ion chromatogram obtained when the tryptic peptides were separated by reverse phase liquid chromatography and detected by mass spectrometry. This chromatogram has been divided into four mass ranges. The x­ axis indicates retention time (minutes) and the y-axis indicates the relative abundance of each chromatographic peak.

81 Fig ure 3.6. The base peak c hromatogram of the tryptic peptides (m/ z SOO - 2,000) detected in human tear p lotted over four distinct mas s ranges.

90.59

92.35

97.16

r06.184 106.84 84.87 106.21 110.00

104.06

90.59

137.20 m/z 1625 - 2000 92.35

100 84.87 m/z 1250- 1625

::,g 25 -0 30.34 48.31 49.87 -Cl) I tl) \I 1,tj 1,.t__ _ 0 C: ea "C C: :::, .0 et Cl) > .:: ea 130.20 cii m/z500 - 875 139.15 a: 79.38 I 1II f 1.37 32.24 52.81 77.79 11 ij 111 64.95 \ I I I ' 35.90 51 ~ 1 I /\ I' 111 11 i.'I I' )' 1 \ I I I I' I I I I I I I I 1 I I I I I I j I I I I I I I I I I j I I I I I I I I I I I I I I I I I I I I I I I I I 50 30 40 60 70 80 90 100 110 120 130 140 150 Time (min) 82 This study has identified over 300 proteins not previously reported as a component of human tear. However, subsets of these proteins have been detected in various human ocular tissues (e.g., lacrimal gland, conjunctiva, cornea) or have been identified in the tear film of individual with pathological conditions (e.g., dry eye syndrome, rheumatoid

arthritis). These proteins are listed in Tables 3.8 (p 93) and 3.9 (p 94) respectively.

Proteins detected in the tear film or ocular tissue of other animal species are listed in

Table 3.10 (p 95). In addition to these, proteins that have not previously been reported

as components of human tear were detected and identified: these are listed in Table 3.11

(p 96). These include various plasma proteins, immunoglobulin receptor proteins,

cytokines, growth factors, proline-rich proteins, mucins, enzymes and other biologically

active peptides and proteins.

83 Table 3.7. Proteins identified by ESI-LC/MS/MS that have been previously reported as components of the human tear film.

# PEPTIDES PROTEIN REFERENCE DETECTED van Haeringen and a.-N-acetylglucosaminidase precursor Glasius, 1976; van 2 Accession number: 4505327 Haeringen and Glasius, 1980 a.-1-antichymotrypsin precursor Prause, 1983; Sathe et 4 Accession number: 112874 al., 1998. a.-1-antitrypsin precursor ( a.-1- Berta, 1982; Prause, antiproteinase) 2 1983; Gupta et al., Accession number: 1703025 1988; Sathe et al., 1998 van Haeringen and Glasius, 1974; Van a-amylase precursor (salivary) 5 Haeringen and Glasius, Accession number: 1351933 1974; Van Haeringen et al., 1975 a.-2-macroglobulin precursor Prause, 1983; Sathe et 15 Accession number: 112911 al., 1998 van Haeringen and a.-mannosidase II 4 Glasius, 1980; Kitaoka Accession number: 3122380 et al., 1985 van Haeringen and a.-mannosidase IIX 5 Glasius, 1980; Kitaoka Accession number: 3123244 et al., 1985 van Haeringen and P-glucuronidase precursor 7 Glasius, 1980; Kitaoka Accession number: 114963 et al., 1985 P-2-microglobulin precursor Markusse et al., 1992; 3 Accession number: 114773 Markusse et al., 1993

84 # PEYrIDES PROTEIN REFERENCE DETECTED Amyloid protein precursor 3 Van Setten et al., 1996 Accession number: 112927 Angiotensin converting enzyme precursor 7 Vita et al., 1981 Accession number: 113045 Antileukoproteinase precursor 3 Sathe et al., 1998 Accession number: 113636 Arylsulfatase D precursor van Haeringen and 2 Accession number: 1703425 Glasius, 1980 Calcitonin gene related peptide I precursor or Calcitonin gene related peptide II precursor Mertaniemi et al., 1995; 2 Accession number: 115487 Tervo et al., 1995 Accession number: 115482 Ceruloplasmin precursor 11 Seal, 1985 Accession number: 116117 Complement C 1Q subcomponent precursor, Mondino and Zaidman, chain A 2 1983; Willcox et al., Accession number: 399139 1997 Complement Cl Q subcomponent precursor, Mondino and Zaidman, chainB 4 1983; Willcox et al., Accession number: 399140 1997 Complement CIR component precursor Mondino and Zaidman, 9 Accession number: 115205 1983 Imanishi et al., 1982; Complement C 1 Scomponent precursor Mondino and Zaidman, ( C 1esterase) 5 1983; Sack et al., 1992; Accession number: 115205 Willcox et al., 1997 Mondino and Zaidman, Complement C3 precursor 19 1983; Willcox et al., Accession number: 116594 1997 Mondino and Zaidman, Complement C4 precursor 15 1983; Willcox et al., Accession number: 116602 1997

85 # PEYfIDES PROTEIN REFERENCE DETECTED Complement C5 precursor 13 Willcox et al., 1997 Accession number: 116602 Complement C9 precursor 7 Willcox et al., 1997 Accession number: 116607 Complement decay accelerating factor precursor 3 Lass et al., 1990 Accession number: 1345942 Complement factor B precursor 7 Willcox et al., 1997 Accession number: 584908 Cystatin C precursor Barka et al., 1991; Reitz 3 Accession number: 118183 et al., 1998 Barka et al., 1991; Cystatin SN precursor 4 Isemura et al., 1991; Accession number: 118188 Molloy et al., 1997 Cytosolic phospholipase A2 Nevalainen, 1993; Qu 2 Accession number: 1352707 and Lehrer, 1998 Elafin 3 Sathe et al., 1998 Accession number: 119262 Eosinophil cationic protein precursor 2 Leonardi et al., 1995 Accession number: 119124 Eosinophil granule major basic protein precursor 2 Udell et al., 1981 Accession number: 11929 Eosinophil lysophospholipase 2 Udell et al., 1981 Accession number: 547870 Eosinophil peroxidase precursor Fullard, 1988; Fullard 10 Accession number: 1352738 and Tucker, 1991 Epidermal growth factor precursor Fullard, 1988; Fullard 7 Accession number: 119226 and Tucker, 1991 Fibroblast growth factor (basic 22kDa form) 2 van Setten, 1996 Accession number: 482272 Hayashi and Sueishi, Fibronectin precursor 16 1988; Sakata et al., Accession number: 2506872 1997; Sack et al., 1999

86 # PEPTIDES PROTEIN REFERENCE DETECTED Van Haeringen and Glutamate dehydrogenase I precursor 3 Glasius, 1974; van Accession number: 118541 Haeringen and Glasius, 1976 Granulocyte macrophage colony stimulating factor precursor 3 Thakur et al., 1998 Accession number: 117561

Heparin binding growth factor- I precursor 4 van Setten, 1996 Accession number: 122737

Heparin binding growth factor-2 precursor 3 van Setten, 1996 Accession number: 122742

Hepatocyte growth factor precursor 10 Li et al., 1996 Accession number: 123116

Van Haeringen and Hexokinase, type II Glasius, 1974; van 5 Accession number: 1708361 Haeringen and Glasius, 1976 Little et al., 1969; McClellan et al., 1973; Coyle and Sibony, 1986; Fullard, 1988; Coyle et Immunoglobulin alpha chain 1 7 al., 1989; Tchah, 1989; Friedman, 1990; Mii et al., 1992; Reitz et al., 1998 Brauninger and lmmunoglobulin epsilon chain C region Centifanto, 1971; 3 Accession number: 119512 McClellan et al., 1973; Friedman, 1990

87 # PEYfIDES PROTEIN REFERENCE DETECTED Little et al., 1969; McClellan et al., 1973; Seal, 1985; Coyle and Immunoglobulin gamma chain 1 2 Sibony, 1986; Fullard, 1988; Coyle et al., 1989; Friedman, 1990; Reitz et al., 1998 Immunoglobulin J chain 5 Friedman, 1990 Accession number: 400044

McClellan et al., 1973; Coyle and Sibony, 1986; Immunoglobulin mu precursor 1 17 Fullard, 1988; Friedman, 1990; Fullard and Tucker, 1991 Interleukin 2 precursor 3 Uchio et al., 2000 Accession number: 124325

Interleukin 6 precursor 3 Thakur et al., 1998 Accession number: 124347

Van Haeringen and Isocitrate dehydrogenase, mitochondrial Glasius, 1974; van precursor 4 Haeringen and Glasius, Accession number: 6166246 1976

Latent transforming growth factor ~ binding protein 19 Yoshino et al., 1996 Accession number: 1196439 Leukocyte elastase inhibitor precursor 3 Sathe et al., 1998 Accession number: 266344

Lipophilin A precursor Molloy et al., 1997; 2 Accession number: 6831580 Lehrer et al., 1998

88 # PEPTIDES PROTEIN REFERENCE DETECTED Berta, 1982; Janssen and van Bijsterveld, 1983b; Kijlstra et al., 1983; Seal, 1985; Jensen et al., 1986; Fullard, 1988; Coyle et al., 1989; Kijlstra et al., 1989; Lactotransferrin precursor 35 Baier et al., 1990; Accession number: 6175096 Fullard and Tucker, 1991; Kuizenga et al., 1991b; Kuizenga et al., 1991a; Mii et al., 1992; Sack et al., 1992; Molloy et al., 1997; Reitz et al., 1998 van Haeringen and Glasius, 1974; Van Haeringen and Glasius, L-Lactate dehydogenase M chain precursor 2 1974; Kahan and Accession number: 126047 Ottovay, 1975; van Haeringen and Glasius, 1976; Jacq et al., 1982 Lysosomal cx-glucosidase precursor van Haeringen and 4 Accession number: 3122374 Glasius, 1980 van Haeringen and Lysosomal cx-mannosidase precursor 3 Glasius, 1980; Kitaoka Accession number: 3122374 et al., 1985 Mammoglobin B precursor (lipophilin C) Molloy et al., 1997; 2 Accession number: 6831582 Lehrer et al., 1998 Muc5AC (gastric mucin) [Jumblatt et al., 1999; 6 Accession number: 15026993 McKenzie et al., 2000]

89 # PEPTIDES PROTEIN REFERENCE DETECTED Van Haeringen and Glasius, 1974; Stuchell etal., 1981; Berta, 1982; Lysozyme precursor Seal, 1985; Fullard, 19 Accession number: 126615 1988; Coyle et al., 1989; Baier et al., 1990; Molloy et al., 1997; Reitz et al., 1998 van Haeringen and Glasius, 1974; Van Malate dehydrogenase, cytoplasmic 3 Haeringen and Glasius, Accession number: 1708967 1974; van Haeringen and Glasius, 1976 Mucin 5 precursor (tracheobronchial) 11 Jumblatt et al., 1999 Accession number: 1346603 Mucin SB Jumblatt et al., 1999; 2 Accession number: 631466 McKenzie et al., 2000 Phospholipase A2 precursor, membrane Nevalainen, 1993; Qu associated 5 and Lehrer, 1998 Accession number: 129483 Plasminogen activator, precursor urokinase type 5 Schenkels et al., 1991 Accession number: 137112 Plasminogen precursor Thorig et al., 1983; 11 Accession number: 130316 Tozser and Berta, 1991 Polymeric immunoglobulin receptor Coyle and Sibony, 1986; precursor, secretory component 11 Kuizenga et al., 1991a Accession number: 1730570 Berta, 1982; Coyle et Prolactin-inducible protein precursor 5 al., 1989; Fullard and Accession number: 134170 Tucker, 1991 Prolactin precursor Frey et al., 1981; Frey et 3 Accession number: 130930 al., 1986

90 # PEPTIDES PROTEIN REFERENCE DETECTED van Haeringen and Glasius, 1974; Van Pyruvate kinase, isozymes R/L 2 Haeringen and Glasius, Accession number: 8247933 1974; van Haeringen and Glasius, 1976 van Haeringen and Pyruvate kinase, M 1 isozyme or Glasius, 1974; Van Pyruvate kinase, M2 isozyme 2 Haeringen and Glasius, Accession number: 266427 1974; van Haeringen Accession number: 125604 and Glasius, 1976 Stuchell et al., 1981; Berta, 1982; Prause, 1983; Fullard, 1988; Serotransferrin precursor Coyle et al., 1989; Baier 8 Accession number: 136191 et al., 1990; Fullard and Tucker, 1991; Kuizenga et al., 1991b; Mii et al., 1992 Serum albumin precursor Prause, 1983; Gupta et 12 Accession number: 113576 al., 1988 Superoxide dismutase precursor, extracellular 2 Behndig et al., 1998 Accession number: 134635 Transcobalamin I precursor 2 Carmel, 1972 Accession number: 135533 Transforming growth factor ~-1 precursor Gupta et al., 1996; 3 Accession number: 135674 Yoshino et al., 1996 Transforming growth factor ~-2 precursor Gupta et al., 1996; 8 Accession number: 135679 Yoshino et al., 1996 Vitronectin precursor 6 Sack et al., 1994 Accession number: 139653 Zn-cx-2-glycoprotein precursor 11 Molloy et al., 1997 Accession number: 141596

91 # PEPTIDES PROTEIN REFERENCE DETECTED Bonavida et al., 1969; Berta, 1982; Janssen and van Bijsterveld, 1983b; Janssen and van Bijsterveld, 1983a; von Ebner's gland protein precursor Fullard, 1988; Baier et (tear lipocalin) 14 al., 1990; Fullard and Accession number: 401346 Kissner, 1991; Fullard and Tucker, 1991; Kuizenga et al., 1991b; Baguet et al., 1992; Molloy et al., 1997; Reitz et al., 1998

1 Due to the high degree of homology between isoforms of these proteins the peptides detected could not be assigned to a unique protein without ambiguity.

92 Table 3.8. Proteins that have been detected in human ocular tissue but not in the tear

film.

No. PEPTIDES PROTEIN REFERENCE DETECTED P-defensin 1 precursor 2 Haynes et al., 1999 Accession number: 1705448 P-defensin 2 precursor 4 Haynes et al., 1999 Accession number: 3023383 Apolipoprotein D precursor 8 Holzfeind et al., 1995 Accession number: 114034 Basic proline rich protein Dickinson and Thiesse, 2 Accession number: 1836022 1996 CD44 antigen precursor 5 Yoshida et al., 1996 Accession number: 2507241 Clusterin Nishida et al., 1996; 2 Accession number: 116533 Dota et al., 1999 Immunoglobulin delta chain C region precursor 2 Friedman, 1990 Accession number: 118945 Interleukin 1-a. precursor 2 Pflugfelder et al., 1999 Accession number: 124297 Mucin 1 precursor ( epithelial) 2 Inatomi et al., 1995 Accession number: 547937 Mucin 2 precursor 12 McKenzie et al., 2000 Accession number: 2506877 pHL ElFl (lacrimal proline-rich protein) Dickinson and Thiesse, 2 Accession number: 2135904 1995 Platelet derived growth factor precursor, A chain 3 Nguyen et al., 1997 Accession number: 129719 Transforming growth factor-P-3 precursor Nishida et al., 1995; 6 Accession number: 135684 Nguyen et al., 1997

93 Table 3.9. Proteins identified by ESI-LC/MS/MS that are reportedly elevated in the tear fluid of patients with various pathological conditions.

No. PEPTIDES PROTEIN REFERENCE DETECTED 52kDR052 (Sjogren syndrome type A antigen) 3 Pflugfelder et al., 1986 Accession number: 123250 FASL receptor precursor 3 Tuominen et al., 1999 Accession number: 119833 Granzyme A precursor Tsubota et al., 1994a; 2 Accession number: 121581 Tsubota et al., 1994b Perforin 1 precursor Tsubota et al., 1994a; 6 Accessionnumber: 129819 Tsubota et al., 1994b Sjogren syndrome Type B antigen (Lupus LA protein) 4 Pflugfelder et al., 1986 Accession number: 125985

94 Table 3.10. Proteins identified by ESI-LC/MS/MS that have been reported in the tear film of animal species other than human.

No. PEPTIDES PROTEIN REFERENCE DETECTED Cysteine rich secretory protein 3 precursor 3 Haendler et al., 1999 Accession number: 1706135 Endothelin-1 precursor 2 Takashima et al., 1996 Accession number: 119610 Pancreatic lipase related protein 1 precursor 4 Remington et al., 1999 Accession number: 1708837 Pancreatic lipase related protein 2 precursor 3 Remington et al., 1999 Accession number: 1708840 Plasma retinol-binding protein precursor 5 Lee et al., 1992 Accession number: 132404 Retinoic acid binding protein I precursor, cellular 2 Lee et al., 1992 Accession number: 266904 Retinoic acid binding protein II precursor, cellular 3 Lee et al., 1992 Accession number: 132401 Retinol binding-protein II precursor, cellular 3 Lee et al., 1992 Accession number: 1710092

95 Table 3.11. Proteins identified by ESI-LC/MS/MS that have not previously been reported as components of the human tear film.

No. PEPTIDES PROTEIN DETECTED cx-1-antitrypsin related protein precursor 2 Accession number: 112891 cx-1-acid glycoprotein 1 precursor 3 Accession number: 112877 cx-1-acid gl ycoprotein 2 precursor 4 Accession number: 231458 cx-1 b-glycoprotein precursor 6 Accession number: 112892 cx-1-microglobulin / inter-ex-trypsin inhibitor precursor, light chain 4 Accession number: 122801 cx-2-antiplasmin precursor 4 Accession number: 112907 cx-defensin 4 precursor 2 Accession number: 399352 cx-defensin 5 precursor 2 Accession number: 399353 cx-defensin 6 precursor 2 Accession number: 399354 cx-fetoprotein precursor 4 Accession number: 120042 ~-2-glycoprotein (hemopexin) 3 Accession number: 170182 ~-2-glycoprotein I precursor (apolipoprotein H) 3 Accession number: 543826 ~-mannosidase precursor 2 Accession number: 3024091 ~-secretase precursor 3 Accession number: 6685248

96 No. PEPTIDES PROTEIN DETECTED y-interferon inducible protein IP-30 precursor 3 Accession number: 280788 Afamin precursor ( a-albumin) 3 Accession number: 1168366 Aldehyde dehydrogenase 6 2 Accession number: 1352246 Aldehyde dehydrogenase, cytosolic ( class 1) 2 Accession number: 118495 Aldehyde dehydrogenase, mitochondrial precursor 2 Accession number: 118504 Aldehyde reductase ( al dose reductase) 2 Accession number: 113596 Amyloid-like protein 1 precursor 4 Accession number: 2829415 Amyloid-like protein 2 precursor 3 Accession number: 1703344 Androgen induced growth factor (FGF-8) 7 Accession number: 1206791 Angiogenin precursor 2 Accession number: 113873 Angiotensinogen precursor 5 Accession number: 113880 Angiotensinogen - mutant 2 Accession number: 4261988 Antithrombin III precursor 4 Accession number: 113936 Apolipoprotein A-I precursor 3 Accession number: 113992 Apolipoprotein B precursor 16 Accession number: 114014 Apolipoprotein E precursor 2 Accession number: 114039

97 NO. PEPTIDES PROTEIN DETECTED Azurocidin precursor ( cationic antimicrobial peptide, CAP3 7) 5 Accession number: 416746 Bacterial permeability increasing protein precursor 11 Accession number: 2822120 C4B binding protein ex chain precursor 2 Accession number: 416733 Carboxypepetidase Al precursor 2 Accession number: 399196 Carboxypepetidase A2 precursor 2 Accession number: 1352189 Carboxypepetidase B precursor (pancreas specific) 5 Accession number: 3915628 Carboxypeptidase D 3 Accession number: 4503007 Carboxypeptidase H precursor 2 Accession number: 115892 Carboxypeptidase M precursor 3 Accession number: 14916957 Carboxypeptidase N precursor 2 Accession number: 1154432 Caspase 9 precursor 2 Accession number: 1264432 Catalase 6 Accession number: 115702 Cathepsin E precursor 2 Accession number: 115723 Cathepsin G precursor 4 Accession number: 115725 Cathepsin K precursor 5 Accession number: 1168793

Cathepsin L2 precursor 2 Accession number: 12644075

98 NO. PEPTIDES PROTEIN DETECTED Cathepsin S precursor 2 Accession number: 115748 Cerebrin-50 2 Accession number: 2134902 Choriomammotropin like protein 2 Accession number: 106319 Chromogranin A precursor 4 Accession number: 6832905 Chymotrypsinogen B precursor 2 Accession number: 117617 Coagulation factor V precursor 16 Accession number: 462045 Coagulation factor VII precursor 4 Accession number: 119766 Coagulation factor VIII precursor 10 Accession number: 119767 Coagulation factor XI precursor 3 Accession number: 119762 Coagulation factor XII precursor 3 Accession number: 119763 Coagulation factor XIIIa precursor 5 Accession number: 119720 Complement C2 precursor 5 Accession number: 3915642 Complement C6 precursor 13 Accession number: 116609 Complement C7 precursor 10 Accession number: 87197 Complement C8 a chain precursor 5 Accession number: 729167

Complement C8 ~ chain precursor 5 Accession number: 116612

99 NO. PEPTIDES PROTEIN DETECTED Complement Factor H precursor 13 Accession number: 116131 Complement Factor I precursor 6 Accession number: 116133 Complement activating component of RA-reactive factor precursor 2 Accession number: 1352202 Cysteine proteinase 3 Accession number: 2134965 Cysteine rich protein 1 precursor 2 Accession number: 48161 Defensin HNP2 homologue (fragment) 1 Accession number: 2134986 Endothelin-converting enzyme 1 precursor 2 Accession number: 1706563 FALL-39 precursor (antimicrobial protein, CAP-18) 5 Accession number: 1706745 Fibroblast growth factor-4 precursor 4 Accession number: 122750 Fibroblast growth factor-6 precursor 2 Accession number: 1169676 Fibroblast growth factor-11 precursor 3 Accession number: 2494457 Fibroblast growth factor-12 precursor 3 Accession number: 2494459 Fibroblast growth factor-13 precursor 4 Accession number: 2494461 Fibroblast growth factor-14 precursor 3 Accession number: 2494463 Fibroblast growth factor-17 precursor 3 Accession number: 601514 7 Fibrinogen a/a.-E chain precursor 9 Accession number: 1706799

100 No. PEPTIDES PROTEIN DETECTED Fibrinogen ~ chain precursor 7 Accession number: 399492 Fibrinogen gamma-A chain precursor or Fibrinogen gamma-B chain precursor 5 Accession number: 120142 Accession number: 120146 Follistatin precursor 2 Accession number: 120547 Follistatin-related protein precursor 3 Accession number: 2498390 Fructose bisphosphate aldolase 2 Accession number: 113611 Glial cell line-derived neurotrophic factor precursor 2 Accession number: 729567 Glutamate decarboxylase, 67kDa isoform 3 Accession number: 1352213 Growth/differentiation factor 5 precursor 2 Accession number: 1346125 Hepatocyte growth factor activator precursor 2 Accession number: 547643 Hepatocyte growth factor-like protein precursor (macrophage stimulatory protein) 3 Accession number: 123114 Hepatoma-derived growth factor 3 Accession number: 1708159 Hepcidinprecursor 3 Accession number: 10720397

High affinity immunoglobulin E FC receptor ~-subunit precursor 2 Accession number: 232084 Immunoglobulin a. FC receptor precursor 5 Accession number: 119800 Immunoglobulin y FC receptor precursor 2 Accession number: 119871

101 NO. PEPTIDES PROTEIN DETECTED Immunoglobulin mu heavy chain disease protein precursor 3 Accession number: 127506 Immunoglobulin mu switch region binding protein 2 precursor 7 Accession number: 1082534 Inhibin 13 B chain precursor 2 Accession number: 1708437 Insulin degrading enzyme 3 Accession number: 124157 Insulin like growth factor II precursor 2 Accession number: 124255 Insulin like growth factor binding protein V precursor 3 Accession number: 124070 Insulin like growth factor binding protein complex acid labile chain precursor 4 Accession number: 543800 Interferon a-2 precursor 2 Accession number: 124449 Interferon a-8 (interferon a-b2) precursor 2 Accession number: 417188 Interleukin 1-13 convertase precursor 3 Accession number: 266321 Interleukin 7 precursor 4 Accession number: 124354 Interleukin 12 a chain precursor 3 Accession number: 266319 Interleukin 12 13 chain precursor 4 Accession number: 266320 Interleukin 13 precursor 2 Accession number: 462408 Interleukin 17 precursor 2 Accession number: 2498481 lnterphotoreceptor retinoid-binding protein precursor (interstitial) 3 Accession number: 124894

102 NO. PEPTIDES PROTEIN DETECTED Kallikrein (glandular kallikrein) 3 Accession number: 125170 Kallikrein (plasma prekallikrein, kininogen) 6 Accession number: 125184 Kallistatin precursor (kallikrein inhibitor) 3 Accession number: 1708609 Keratinocyte growth factor precursor (FGF-7) 4 Accession number: 122756 Kininogen precursor 5 Accession number: 125507 Lens epithelium-derived growth factor 3 Accession number: 11360305 Leucine-rich a-2-glycoprotein 2 Accession number: 16418467 Low affinity immunoglobulin yFC receptor 11-C precursor 3 Accession number: 399476 Lysosomal acid lipase/cholesterol ester hydrolase precursor 3 Accession number: 585405 Lysosomal protective protein precursor (cathepsin A, carboxypeptidase C) 3 Accession number: 131081 Macrophage colony stimulating factor- I precursor 6 Accession number: 117558 Macrophage inflammatory protein 2-a precursor 3 Accession number: 127085 Maspin precusor 4 Accession number: 547892 Mast cell carboxypeptidase A precursor 8 Accession number: 115887 Melanotransferrin precursor 9 Accession number: 136204 Membrane cofactor protein precursor 2 Accession number: 1708964

103 NO. PEPTIDES PROTEIN DETECTED Meprin A a-subunit precursor 2 Accession number: 2499910 Meprin A ~-subunit precursor 3 Accession number: 2499912 Monocyte chemotactic protein 3 precursor ( eotaxin precursor) 2 Accession number: 1706661 Myeloperoxidase precursor 4 Accession number: 129825 Neuromedin B precursor 3 Accession number: 1346684 Neutrophil cytosol factor 2 3 Accession number: 1346609 Pepsinogen A precursor 2 Accession number: 129792

Phosphatidylinositol transfer protein ~ precursor 2 Accession number: 1346772 Pigment epithelium-derived factor precursor 5 Accession number: 1352735 Plasminogen activator inhibitor 1 precursor 2 Accession number: 129576 Plasminogen activator inhibitor 2 precursor 5 Accession number: 1352712 Plasma protease C 1 inhibitor precursor 4 Accession number: 124096 Plasma protease inhibitor precursor 3 Accession number: 400068 Plastin precursor, L- 3 Accession number: 2493466 Plastin precursor, T- 3 Accession number: 2506254 Platelet-activating factor acetylhydolase 1B subunit 2 Accession number: 3024345

104 NO. PEPTIDES PROTEIN DETECTED Platelet derived epithelial cell growth factor precursor 2 Accession number: 129719 Proenk:ephalin A precursor 6 Accession number: 129770 Proline rich protein (PRCC) 3 Accession number: 2498802 Prostacyclin synthase precursor (prostaglandin 12 synthase) 2 Accession number: 2493373 Prostaglandin G/H synthase 1 precursor 3 Accession number: 129899 Prostaglandin D2 synthase precursor 3 Accession number: 730305 Prothrombin precursor ( coagulation factor II) 4 Accession number: 135807 Pyruvate carboxylase precursor 2 Accession number: 1709947 Renin precursor (angiotensinogenase) 2 Accession number: 132326 Salivary peroxidase or lactoperoxidase 9 Accession number: 2117625 Accession number: 4264341 Secretogranin I precursor (chromogranin B) 4 Accession number: 134461 Secretagranin II precursor ( chromogranin C) 7 Accession number: 134464 Semaphorin 3C precursor 2 Accession number: 8134685 Semaphorin 3F precursor 2 Accession number: 8134696 Serine hydroxymethyltransferase, cytosolic 3 Accession number: 462184 Serine protease hepsin 2 Accession number: 123057

105 No. PEPTIDES PROTEIN DETECTED Somatoliberin precursor (OH-releasing factor) 2 Accession number: 134521 Somatotropin precursor 10 Accession number: 134703 Stem cell factor precursor (mast cell growth factor) 3 Accession number: 4580470 Succinate dehydrogenase flavoprotein subunit, mitochondrial precursor 4 Accession number: 1169337 Tissue factor precursor precursor 2 Accession number: 135666 Tissue factor pathway inhibitor 1 precursor 3 Accession number: 125932 Transforming growth factor-~ 4 precursor 4 Accession number: 13124811 Transforming growth factor~ induced protein IG-H3 precursor 9 Accession number: 2498193 Tumour necrosis factor-C precursor (lymphotoxin ~) 3 Accession number: 549088 Tumour necrosis factor, a induced protein 1, B12 protein 3 Accession number: 2833248 Tumour necrosis factor inducible protein TSG-6 precursor 2 Accession number: 1351315 Tumour necrosis factor, a induced protein 3 3 Accession number: 112894 Vasoactive intestinal peptide precursor 3 Accession number: 138574 Vitamin K dependant protein C precursor 5 Accession number: 131067 Vitamin K dependant protein S precursor 3 Accession number: 131086 Vascular endothelial growth factor-B precursor 3 Accession number: 1718152

106 No. PEPTIDES PROTEIN DETECTED Vascular endothelial growth factor-C precursor 4 Accession number: 1718154 Vascular endothelial growth factor precursor 3 Accession number: 137821 von Willebrand factor precursor 13 Accession number: 401413

107 3.4. Discussion

Mass spectrometry, in particular MALDI-TOFMS and ESI-LC/MS/MS, provides a sensitive and rapid approach to the detection and identification of protein components in complex biological samples. By utilising these techniques, several hundred peptides and proteins were detected and identified in a small volume of human tear: less than 10 µL was required for the entire study. Peptides in human tear were detected by MALDI­

TOFMS, sequenced by MALDI-PSD, then identified by database searching. Proteins were analysed by ESI-LC/MS/MS following enzymatic digestion with trypsin, and identified by searching fragment ion spectra against the non-redundant protein databases using SEQUEST.

By combining the approaches ofMALDI-TOFMS and ESI-LC/MS/MS a

comprehensive analysis of the peptide and protein components in biological tissues and

fluids can be performed. MALDI-TOFMS is ideal for the detection of peptides and

proteins in complex samples, but due to the signal suppression effects inherent to the

MALDI ionisation process, not all components can be detected. Nevertheless, the mass

of the most abundant endogenous peptides and proteins can be determined with

accuracy. Over 30 peptides and proteins were detected in human tear using MALDI­

TOFMS, and the most abundant peptides were shown to be derived from the C-terminus

of a lacrimal proline-rich protein previously not detected as a component of human tear

[Dickinson and Thiesse, 1995]. In addition to these peptides, a truncated form of this

protein corresponding to residues 18 - 121 of the full length protein was also detected.

The biological significance of the cleavage of this protein to yield a homologous family

of peptides is unknown.

108 By employing ESI-LC/MS/MS a more comprehensive characterisation of the protein constituents can be obtained. Although the direct analysis of peptides and proteins by

ESI-LC/MS/MS aims to identify all the proteins in a given sample, it does not routinely detect or identify post-translational modifications such as phosphorylation, glycosylation or truncation to form bioactive products. For instance, the presence of glycoproteins and truncated proteins, such as truncated forms of tear lipocalin, in the tear film has been reported [Coyle et al., 1989; Fullard and Kissner, 1991; Kuizenga et al., 1991a; Jumblatt et al., 1999; McKenzie et al., 2000]. Although more than 400 proteins were identified in this study, it should be noted that ESI-LC/MS/MS can not identify all protein isoforms nor establish all protein modifications.

There have been many investigations concerning the protein composition of the normal

tear film and in total, approximately 100 proteins have been identified and reported.

Previously, the protein components of human tear were characterised by techniques

such as gel electrophoresis, HPLC and/or immunochemical techniques [Boukes et al.,

1987; Fullard, 1988; Coyle et al., 1989; Baier et al., 1990; Wollensak et al., 1990;

Kuizenga et al., 1991b; Mii et al., 1992; Behndig et al., 1998; Jumblatt et al., 1999].

These techniques require large amounts of sample and extensive purification of the

proteins of interest. In this study over 400 proteins were detected and identified in a

single ESI-LC/MS/MS analysis, including most of those that have been previously

reported as components of the tear film. The biological function of many of the proteins

identified by this approach, and their possible role in the pathogenesis of ocular disease

is yet to be established. This study also identified proteins in normal tear fluid that have

previously only been detected in pathological states, indicating that these proteins are

109 also present at low concentrations in normal tear. Further investigations involving analysis of tear film derived from individuals diagnosed with these conditions would be advantageous in establishing the utility of these proteins as potential biomarkers of disease.

110 CHAPTER4

COMPREHENSIVE PEPTIDE AND

PROTEIN IDENTIFICATION:

APPLICATION TO THE ANALYSIS

OF HUMAN SEMINAL FLUID

111 4.1. Introduction

Proteomics is the process of defining the proteome [Wasinger et al., 1995] of a biological system and is generally determined following the separation of a complex protein mixture by two dimensional gel electrophoresis (2DE), proteolytic digestion of

the individual proteins, and subsequent identification by mass spectrometry and

database searching [James et al., 1993; Mann et al., 1993]. Analysis of proteins by 2DE

involves the separation and focusing of proteins in a biological sample based on their

isoelectric point (pi; first dimension) and following this, migration through a

polyacrylamide gel and separation according to molecular weight (second dimension).

Although 2DE was first introduced in 1975 [O'Farrell, 1975], its popularity as a protein

separation tool was enhanced considerably by two developments: improvements in

isoelectric focusing (IEF) technology (i.e., the introduction of carrier ampholytes and

immobilised pH gradient strips) enabling improved resolution of proteins in the first

dimension [Bjellqvist et al., 1982; Qorg et al., 1988; Steinberg et al., 2000]; and

improved ionisation methods for mass spectrometry, in particular, matrix assisted laser

desorption ionisation (MALDI) and electrospray ionisation (ESI). These two techniques

enabled rapid and unambiguous identification of subpicomole amounts of the separated

protein. As a result, 2DE has become the most widely accepted technique for the·

separation and simultaneous visualisation of multiple proteins in complex samples

[Klose and Kobalz, 1995; Lopez, 1999].

A variety of different chemical staining techniques have been developed that are able to

detect low amounts of protein following their separation by 2DE. The most widely

employed stains include Coomassie blue and silver staining, but more recently a variety

of fluorescent stains have been developed that reportedly offer greater sensitivity and

112 the ability to detect low nanograms of proteins [Shevchenko et al., 1996; Lopez et al.,

2000; Lauber et al., 2001]. Because many hundreds of proteins can be resolved by 2DE and then detected using these stains, relative changes in protein expression can be estimated on a global scale (i.e., alterations in the levels of many different proteins can be monitored simultaneously). For example, changes in protein expression between

cells maintained under different conditions, or between normal and disease tissue can be

quantified [Kuster et al., 1997; Neubauer and Mann, 1999; Lahm and Langen, 2000;

Sickmann et al., 2001].

Prior to the use of mass spectrometry, techniques such as Edman degradation and amino

acid analysis were employed to identify proteins separated on gels. The success of these techniques, however, was limited because relatively large quantities of pure proteins

were required and even then, post-translational modifications presented a serious

obstacle. Developments in mass spectrometry, primarily MALDI-TOFMS and ESI-MS,

have overcome these limitations because they allow peptides and proteins to be ionised

with high efficiency. Currently, the most widely accepted technique for protein

identification following 2DE involves the use ofMALDI-TOFMS to generate a peptide

mass fingerprint or mass map of the proteolytically digested protein of interest [James et

al., 1993; Mann et al., 1993]. These data are then searched against a database containing

the sequences of known proteins digested with the same enzyme in silico. This strategy

is versatile and powerful and has also been employed successfully to identify proteins

that have been post-translationally modified, e.g., N-terminal , glycosylation,

or phosphorylation [Mills et al., 2001; Sickmann et al., 2001].

113 Although 2DE remains the preferred approach for the global representation of a proteome, it has shortcomings. These include the inability to adequately resolve low molecular weight and basic proteins, the limited solubility of hydrophobic and high mass proteins, co-migration of proteins, and inadequate sensitivity for low abundant proteins [Patton, 1999]. To overcome these problems alternative strategies have been

adopted such as the separation of tryptic peptides by multidimensional liquid

chromatography and identification by tandem mass spectrometry [McCormack et al.,

1997; Yates et al., 1997; Link et al., 1999; Yates et al., 2000; Davis et al., 2001;

Washburn et al., 2001]. Although these approaches are able to identify multiple proteins rapidly, even in complex mixtures, they are not able to easily detect post-translational modifications or to differentiate between multiple forms of the same protein present in

vivo.

To fully characterise the peptide and protein constituents of biological tissues or fluids

requires a comprehensive strategy involving several separation, detection and

identification techniques. In this study a combined approach involving gel

electrophoresis, MALDI-TOFMS and ESI-LC/MS/MS was used to characterise the

peptide and protein constituents of human seminal fluid. Proteins were separated by

either 1D or 2D gel electrophoresis prior to identification by ESI-LC/MS/MS and

MALDI-TOFMS respectively. The peptide components were also investigated by direct

analysis ofunfractionated seminal fluid by MALDI-TOFMS. This approach enables the

accurate mass of peptides to be determined, and ESI-LC/MS/MS was employed

thereafter to obtain amino acid sequence information for each peptide.

114 4.2. Methods

Toe methods employed for this study can be found in Chapter 2 (pages 41 -44).

4.3. Results

4.3.1. Detection and identification of proteins by two dimensional gel

electrophoresis

Separation of proteins in normal human seminal fluid by 2DE followed by visualisation

with silver staining indicated the presence of over 300 discrete protein spots. Figures 4.1

and 4.2 show examples of two annotated silver stained gels of seminal fluid proteins

focused over two different pH ranges (pi range 4 - 7 and 6 - 11 respectively).

Of the 300 discrete protein spots that were excised and digested, 169 were positively

identified by peptide mass fingerprinting and database searching. Of these 169 proteins,

only 32 unique proteins were identified. The other proteins were identified as post­

translational variants of these. Additionally, some proteins were found to co-migrate so

that two or more proteins could be identified under a single spot. Some of the proteins

identified include serum proteins (e.g., serum albumin, Zn-cx-2-glycoprotein,

fibronectin), enzymes and proteins believed to be specific to seminal fluid (e.g., prostate

specific antigen, prostate secreted seminal plasma, semenogelin I, semenogelin II}.

Listed in Tables 4.1 - 4.4 are the proteins that were identified in each gel, including

proteins that were found to co-migrate.

115 Figure 4.1. An annotated silver-stained two dimensional gel of semjnal fluid proteins

separated over the pl range 4 - 7. Indicated are the protein spots that were excised,

digested with trypsin, and identified by peptide mass fingerprinting.

Increasing pi

I ? 0 () 5 i 4 e 6 10 12 0 9 0 0 bi 3 0 13 4 15 ~ . 0 0 0 18 19 16 17 0 0 200 bi 8 2:\ 24 25 26 29 3S 31 32 ::::J tJ 68 0 u 0 035 Cl) 3p 3~ 36 36 31b 3% 4(\) 0 0 42 o41 E 't) CJ 96 8 ~8 49 0 0 43 Cl 53 C: 500 ~1°s2 . '54 ·c:;; l'0 55 65 ~ 56 S7 u o o 58 C: sf 6z 0 0 61 82 ~ 0 66 6J 68 64

72 0 78 77 0 0 79 6 88 ~ 0

94 0 104

116 Figure 4.2. An annotated silver-stained two dimensional gels of seminal fl uid proteins

separated over the pI range 6 - 11. Indicated are the protein spots that were excised,

digested with trypsin, and identified by peptide mass fingerprinting.

Increasing pi

') 0 o-

3 0 d

... 5 6. ~ 0 7 10 en , ~ 0 "ai ~- ...== .!2 8 17 :::, = I S (J 0 .S! 01 9 0 ') E en C: 1'i ')6 "iii c::, C'O 28 (I)... 27 (J C: 3

32 :n 35 0 0 36 39 040 38 0 37 c::::> JI. 0 44 4 .'i 42 -g 0

0 47

48 0

11 7 Table 4.1. Proteins in seminal fluid that were identified by 2DE (pi range 4 - 7).

Listed below are the identified proteins with their corresponding experimentally

determined molecular weight and isoelectric point derived from the gel.

EXPT. EXPT. SPOT PROTEIN MOLECULAR ISOELECTRIC # WEIGHT (kDa) POINT cx-1-antitrypsin Accession number: 1703025 19 45 5.7 Theoretical molecular weight: 46.7 kDa Theoretical isoelectric point: 5.3 cx-enolase Accession number: 119339 32 45 7.0 Theoretical molecular weight: 4 7.1 kDa Theoretical isoelectric point: 7 .0

Antioxidant protein 2 74 20 6.4 Accession number: 1718024 Theoretical molecular weight: 24.9 kDa Theoretical isoelectric point: 6.0 75 20 6.5

Carboxypeptidase B Accession number: 115892 94 15 4.0 Theoretical molecular weight: 53.1 kDa Theoretical isoelectric point: 5.0 Cathe~sinB Accession number: 115711 41 40 6.3 Theoretical molecular weight: 37.8 kDa Theoretical isoelectric point: 5.9 34 45 6.4 CathepsinD 37 43 5.2 Accession number: 115717 Theoretical molecular weight: 44.5 kDa 38 43 5.4 Theoretical isoelectric point: 6.1 39 43 5.6

118 EXPT. EXPT. SPOT MOLECULAR ISOELECTRIC PROTEIN # WEIGHT (kDa) POINT 53 40 6.0 Clusterin Accession number: 116533 55 37 5.2 Theoretical molecular weight: 52.5 kDa Theoretical isoelectric point: 5.9 65 35 6.0

Cystatin S 107 10 4.5 Accession number: 399336 Theoretical molecular weight: 16.2 kDa 108 10 4.7 Theoretical isoelectric point: 4.9 8 65 5.7

Fibronectin 13 65 6.2 Accession number: 2506872 Theoretical molecular weight: 262.6 kDa 14 65 6.3 Theoretical isoelectric point: 5.4 15 65 6.4

Glycodelin Accession number: 130701 78 20 5.5 Theoretical molecular weight: 20.6 kDa Theoretical isoelectric point: 5.3 Heat shock 27 kDa protein number: 123571 Accession 72 20 6.2 Theoretical molecular weight: 22.3 kDa Theoretical isoelectric point: 7 .8 Heat shock 70 kDa, protein 1B 4885431 Accession number: 7 65 5.5 Theoretical molecular weight: 70.0 kDa Theoretical isoelectric point: 5.5 1 70 6.5

4 70 6.7 Lactotransferrin Accession number: 6175096 5 70 7.0 Theoretical molecular weight: 78.3 kDa Theoretical isoelectric point: 8.5 33 43 6.3

35 43 6.5

119 EXPT. EXPT. SPOT MOLECULAR ISOELECTRIC PROTEIN # WEIGHT (kDa) POINT Isocitrate dehydrogenase (NADP+ - dependant) Accession number: 1352423 36 43 6.7 Theoretical molecular weight: 46.6 kDa Theoretical isoelectric point: 6.3 Proapolipoprotein A-I Accession number: 113992 77 20 5.3 Theoretical molecular weight: 30.7 kDa Theoretical isoelectric point: 5.5 96 13 5.1 104 13 4.5 105 13 4.6 Prolactin-inducible protein 106 13 4.6 Accession number: 134170 109 10 4.9 Theoretical molecular weight: 16.5 kDa 110 13 5.0 Theoretical isoelectric point: 8.2 111 13 5.1 113 13 5.4 115 13 5.5

Prostate secreted seminal plasma protein Accession number: 131436 116 10 5.5 Theoretical molecular weight: 12.8 kDa Theoretical isoelectric point: 5.3 54 35 6.4 67 25 6.4 68 25 6.7 71 23 6.8 Prostate specific antigen 74 20 6.4 Accession number: 130989 79 17 6.0 Theoretical molecular weight: 28.7 kDa 80 17 6.2 Theoretical isoelectric point: 7 .6 81 17 6.4 87 17 5.8 91 15 6.7 99 13 6.2

120 EXPT. EXPT. SPOT PROTEIN MOLECULAR ISOELECTRIC # WEIGHT (kDa) POINT Prostate Accession number: 3915892 83 17 6.4 Theoretical molecular weight: 77 .1 kDa Theoretical isoelectric point: 6.3 18 45 5.6 20 45 5.0 21 45 5.1 22 45 5.2 23 45 5.3 24 45 5.4 25 45 5.5 26 45 5.6 27 45 5.7 Prostatic acid phosphatase Accession number: 130730 28 45 5.8 Theoretical molecular weight: 44.5 kDa 29 45 6.0 Theoretical isoelectric point: 5.8 56 35 5.1 57 33 5.2 58 30 5.3 59 35 5.0 60 33 5.1 61 30 5.2 62 30 5.5 101 13 6.7

102 13 6.7

Protein phosphatase, receptor type, cr Accession number: 14765223 Theoretical molecular weight: 217 .1 kDa 76 20 6.3 Theoretical isoelectric point: 6.0

121 EXPT. EXPT. SPOT PROTEIN MOLECULAR ISOELECTRIC # WEIGHT (kDa) POINT 84 17 6.7 85 17 6.7 88 17 5.8 89 15 6.6 90 15 6.6 92 15 6.8 Semenogelin I 93 15 6.9 Accession number: 134426 Theoretical molecular weight: 52.1 kDa 100 13 6.3 Theoretical isoelectric point: 9 .3 114 13 5.4 117 10 5.6 118 10 5.6 119 10 5.6 120 13 5.8 121 10 5.9 95 13 5.0 Semenogelin II 97 13 5.5 Accession number: 401079 98 13 5.8 Theoretical molecular weight: 65.5 kDa 112 13 5.2 Theoretical isoelectric point: 9.0 114 13 5.4

Serotransferrin 2 70 6.6 Accession number: 136191 Theoretical molecular weight: 77 .0 kDa 6.7 Theoretical isoelectric point: 6.8 3 70 44 40 5.0 Zn-a-2-glycoprotein 45 40 5.1 Accession number: 141596 46 40 5.2 Theoretical molecular weight: 33.9 kDa 50 40 5.2 Theoretical isoelectric point: 5.6 55 37 5.2

122 EXPT. EXPT. SPOT PROTEIN MOLECULAR ISOELECTRIC # WEIGHT (kDaJ POINT 9 65 5.8 10 65 5.8 11 65 5.9 12 65 6.0 16 60 6.1 17 60 6.3 30 45 6.3 31 45 6.4 40 43 5.8 42 40 5.6 43 40 5.7 47 40 5.3 Serum albumin Accession number: 113576 48 40 5.4 Theoretical molecular weight: 69.3 kDa 49 40 5.5 Theoretical isoelectric point: 5.9 51 37 5.3 52 37 5.4 53 40 6.0 62 30 5.5 63 25 5.6 64 25 5.6 66 25 6.0 69 23 6.4 70 23 6.6 73 20 6.3 86 15 5.4 103 10 7.0

123 Table 4.2. Proteins in seminal fluid that were identified by 2DE (pi range 4 - 7). These proteins were found to comigrate.

EXPT. EXPT. SPOT PROTEIN MOLECULAR ISO ELECTRIC # WEIGHT (kDa) POINT Serum albumin Accession number: 113576 Theoretical molecular weight: 69 .3 kDa Theoretical isoelectric point: 5.9 53 40 6.0 Clusterin Accession number: 116533 Theoretical molecular weight: 52.5 kDa Theoretical isoelectric point: 5.9 Clusterin Accession number: 116533 Theoretical molecular weight: 52.5 kDa Theoretical isoelectric point: 5.9 55 37 5.2 Zn-Cl-2-glycoprotein Accession number: 141596 Theoretical molecular weight: 33.9 kDa Theoretical isoelectric point: 5.6 Prostatic acid phosphatase Accession number: 130730 Theoretical molecular weight: 44.5 kDa Theoretical isoelectric point: 5.8 62 30 5.5 Serum albumin Accession number: 113576 Theoretical molecular weight: 69.3 kDa Theoretical isoelectric point: 5.9

124 EXP'f. EXP'f. SPOT MOLECULAR ISO ELECTRIC PROTEIN # WEIGHT (kDa) POINT Antioxidant protein 2 Accession number: 1718024 Theoretical molecular weight: 24.9 kDa Theoretical isoelectric point: 6.0 74 20 6.4 Prostate specific antigen Accession number: 130989 Theoretical molecular weight: 28.7 kDa Theoretical isoelectric point: 7 .6 Semenogelin I Accession number: 134426 Theoretical molecular weight: 52.1 kDa Theoretical isoelectric point: 9 .3 114 13 5.4 Semenogelin II Accession number: 401079 Theoretical molecular weight: 65.5 kDa Theoretical isoelectric point: 9.0

125 Table 4.3. Proteins in seminal fluid that were identified by 2DE (pi range 6 - 11 ).

Listed below are the identified proteins with their corresponding experimentally determined molecular weight and isoelectric point derived from the gel.

EXYr. EXYr. SPOT PROTEIN MOLECULAR ISOELECTRIC # WEIGHT (kDa) POINT a-1-antichymotrypsin Accession number: 112874 42 18 6.0 Theoretical molecular weight: 47.6 kDa Theoretical isoelectric point: 5.3 a-1-antitrypsin 10 45 9.0 Accession number: 1703025 Theoretical molecular weight: 46.7 kDa 48 Theoretical isoelectric point: 5.3 10 7.5

Clusterin Accession number: 116533 12 36 6.4 Theoretical molecular weight: 52.5 kDa Theoretical isoelectric point: 5.9 Fibronectin 18 35 9.3 Accession number: 2506872 Theoretical molecular weight: 262.6 kDa 19 35 9.5 Theoretical isoelectric point: 5.4

Glyceraldehyde 3-phosphate dehydrogenase Accession number: 14761208 17 36 8.8 Theoretical molecular weight: 36.0 kDa Theoretical isoelectric point: 8.5

1 78 7.4 2 70 7.4 Lactotransferrin 3 60 7.0 Accession number: 6175096 4 55 7.7 Theoretical molecular weight: 78.3 kDa Theoretical isoelectric point: 8.5 8 41 6.4 15 36 7.7 16 36 8.5

126 EXPT. EXPT. SPOT MOLECULAR ISOELECTRIC PROTEIN # WEIGHT (kDa) POINT

Isocitrate dehydrogenase (NADP+ -dependant) 5 45 6.5 Accession number: 1352423 Theoretical molecular weight: 46.6 kDa Theoretical isoelectric point: 6.3 6 45 7.0

Phosphoglycerate kinase Accession number: 129904 7 45 8.5 Theoretical molecular weight: 44.8 kDa Theoretical isoelectric point: 8.8

11 36 6.3

Prostatic acid phosphatase 13 36 6.4 Accession number: 130730 Theoretical molecular weight: 44.5 kDa 14 36 6.5 Theoretical isoelectric point: 5.8 41 20 6.0

20 30 7.0 21 29 6.4 22 29 6.5 23 29 7.5 Prostate specific antigen Accession number: 130989 24 27 6.5 Theoretical molecular weight: 28.7 kDa 29 25 6.4 Theoretical isoelectric point: 7.6 30 25 6.4 31 25 6.3 32 23 6.5 39 20 6.7 37 20 10.5 Semenogelin I 38 20 11.0 Accession number: 134426 Theoretical molecular weight: 52.1 kDa 45 18 8.3 Theoretical isoelectric point: 9.3 47 15 8.5

127 EXPT. EXPT. SPOT PROTEIN MOLECULAR ISOELECTRIC # WEIGHT (kDa) POINT 33 23 7.8 34 23 8.5 35 23 10.5 36 20 10.5 Semenogelin II 7.5 Accession number: 401079 40 20 Theoretical molecular weight: 65.5 kDa 43 17 6.4 Theoretical isoelectric point: 9.0 44 17 7.8 46 15 10.5 47 15 8.5 48 10 7.5

Serum albumin 25 27 7.0 Accession number: 113576 26 27 7.3 Theoretical molecular weight: 69 .3 kDa Theoretical isoelectric'point: 5.9 28 27 7.3

Sorbitol dehydrogenase Accession number: 4033691 9 38 8.5 Theoretical molecular weight: 38.3 kDa Theoretical isoelectric point: 8.2 Triosephosphate isomerase Accession number: 136060 27 27 7.0 Theoretical molecular weight: 26. 7 kDa Theoretical isoelectric point: 6.5

128 Table 4.4. Proteins in seminal fluid that were identified by 2DE (pi range 6 - 11 ).

These proteins were found to comigrate.

EXPT. EXPT. SPOT PROTEIN MOLECULAR ISO ELECTRIC # WEIGHT (kDa) POINT Phosphoglycerate kinase Accession number: 129904 Theoretical molecular weight: 44.8 kDa Theoretical isoelectric point: 8.8 7 45 8.5 Plasma serine protease inhibitor Accession number: 400068 Theoretical molecular weight: 45.7 kDa Theoretical isoelectric point: 9.3

Semenogelin I Accession number: 134426 Theoretical molecular weight: 52.1 kDa Theoretical isoelectric point: 9 .3 47 15 8.5 Semenogelin II Accession number: 401079 Theoretical molecular weight: 65.5 kDa Theoretical isoelectric point: 9.0 a.-1-antitrypsin Accession number: 1703025 Theoretical molecular weight: 46.7 kDa Theoretical isoelectric point: 5.3 48 10 7.5 Semenogelin II Accession number: 401079 Theoretical molecular weight: 65.5 kDa Theoretical isoelectric point: 9.0

129 4.3.2. Detection and identification of proteins by one dimensional gel electrophoresis

To identify proteins not detected by the 2DE approach, such as high molecular weight

and hydrophobic proteins, seminal fluid proteins were also separated by one

dimensional gel electrophoresis according to their molecular weight. Specific regions of

the gel were excised and digested in-gel with trypsin. The resulting tryptic peptides

were analysed by ESI-LC/MS/MS and identified by database searching. Figure 4.3

shows the base peak chromatogram for peptides derived from proteins in four different molecular weight regions of the ID gel (i.e., regions corresponding to the molecular

weight ranges of 20, 40, 50 and 60 kDa). Peptides derived from multiple proteins (10-

15) were identified in each gel region.

In comparison to 2DE, many more proteins (95) were identified by this approach.

Proteins found included plasma proteins, enzymes, and proteins believed to be specific

to either the seminal vesicle or prostate gland. The majority of the proteins previously

detected and identified by 2DE and peptide mass fingerprinting were also identified.

Some of the proteins identified, such as serum albumin, fibronectin, semenogelin I and

semenogelin II were identified in more than one molecular weight region of the gel

indicating that these proteins are present in at least two distinct forms. These proteins

were also identified in multiple forms by 2DE. Table 4.5 (p 132) lists those proteins

identified by this technique and includes their theoretical and experimentally determined

molecular weight.

130 131

kDa

20

-

kDa

40

-

kDa

07

_

93

50

1,.,,..,,_\.-,

..

.._

.

kDa

_,_

-

24

......

97

~

94

-

60

83

proteins.

·

"'

,,,.

-

._

-_..,

.

.

.I\.

39

95

.

....

002

l fluid

81

92

.

09 . ,,,

0

,1

100

I

a

1

73 .

·t

I

I

·I\

,11

11 .

1

min I

·

_

ij

I

I

42

se

5

_

8

74

06

90

.

8

of

81

~

.

'

83 l

I '

I

62.31

71

ge

-!l~Qey

1

70 67

43

"

80 55

95

89 ~··"

. ,

,

15

,

73

•.

52

.-

'

)

..

62

.

51

,,,.,.,"

85

,1,1

...

5640

73

'L1,}j1li.1~1,1-~.,."'<·I·

k ,.

II 1

1

,

42

,,

.

·

70

,

SDS-PAGE

I

08

J

j

u

I

I

s922

56

..

42.25

!

1

o1

iii

63 1

,,11,~j\ ·

j\/~,l.\ 1D

\

84

1

I

\

~a

44

57

55r1

.

ll

41

3830

!

I ~

t

42

438t

62 .

the

i,

50

60

I

,i

48

I \

33

of

95 :,,,.1.l\

84

'"

,

I

:

,

91

4122

59

1,

:.

11

l

5

, 21

28

'

r

.I

I

,.

ll

36

"

44

77

L

(min)

50

M

~

[

25

42

~

,.

regions

:

__

34 34

98

95

.J.

53

11\i)ll,,,

Time

.

32

nt

23

_

80

......

_..,,~

22

lli~-•,.,. :

·

38 35 38

40

1

ffere

98

04

.

·

59

14

1

37

32

di

½

34

j

29

IIL;,;;.:

1

l

34

82

15~3 1

4

im

8

98 26.89

30

for

65

7

31

30

35

93

25.97

6

25

.,_l!.,,1,,,

11

02

,'.

31

·-,.

"1

97

_...

,

5.80

15

,.,

20

,L

......

..

~

.

1487

chromatogram

~077

I

95

ll

11(\ii~l_i;)

1

.,,

13

10

ak

10

e

p

07

ase

j

.,

0

B

0 9

10

90 30 80 70

60 40 50

20

100

a, ~ >

C ni

3

~ "C ~

cc

4.3.

Figure Table 4.5. Proteins found in seminal fluid following separation by ID gel electrophoresis, in­ gel digestion and ESI-LC/MS/MS analysis. Also listed are the theoretical and experimentally determined molecular weights for each protein.

THEORETICAL EXPT. PROTEIN MOLECULAR MOLECULAR WEIGHT (kDa) WEIGHT (kDa) Acidic epididymal glycoprotein-like 1 28.4 85 Accession number: 4501983 cx.-enolase 47.1 50 Accession number: 119339 cx.-1-acid glycoprotein 1 23.4 50 Accession number: 9257232 46.6 43

I -antitrypsin ex.- 46.6 60 Accession number: 1703025 46.6 50

Arachidonate 12-lipoxygenase, 12R type 80.3 < 10 Accession number: 12230234

~-2-microglobulin 13.7 60 Accession number: 114773 Bactericidal permeability increasing protein 19.3 20 Accession number: 2822120

Calcineurin binding protein I 246.3 < 10 Accession number: 6912458 Carbonic anhydrase 39.4 50 Accession number: 4502515 Carboxypeptidase D 152.4 < 10 Accession number: 4503007 Cathepsin C 51.8 20 Accession number: 4503141 Cathepsin F 53.3 20 Accession number: 6042196

132 THEORETICAL EXPT. PROTEIN MOLECULAR MOLECULAR WEIGHT (kDa) WEIGHT (kDa)

Cathepsin G 28.8 40 Accession number: 4503149

Ceruloplasmin 122.2 60 Accession number: 4557485

52.5 85 Clusterin Accession number: 4502905 52.5 50

52.5 40 192.4 85 Complement C4A Accessionnumber: 14577919 192.4 20

Complement component C5 188.2 60 Accession number: 4502507 Complement component C9 63.1 20 Accession number: 4502511 Complement factor B 85.4 220 Accession number: 4502397 Corticotropin releasing factor-binding protein 36.1 10 Accession number: 544099 CYR61 protein 42.0 20 Accession number: 13638596 Epididymal secretory protein E3 alpha 14.9 85 Accession number: 7227896 262.6 220 Fibronectin Accession number: 2506872 262.6 85

Fibroblast growth factor receptor 2 92.0 60 Accession number: 4503709 Galectin 3 binding protein 65.3 60 Accession number: 5031863

yEnolase 47.3 50 Accession number: 5803011

133 THEORETICAL EXPT. PROTEIN MOLECULAR MOLECULAR WEIGHT (kDa) WEIGHT (kDa) Gastricin (pepsinogen C) 42.4 40 Accession number: 4505757 Gastrin-releasing peptide 16.1 20 Accession number: 4504159 Glucose phosphate isomerase 63.2 50 Accession number: 4504087 Glutathione S-transferase A2 25.7 20 Accession number: 121733 Glutathione S-transferase mu 1 25.7 20 Accession number: 11428198 Glutathione S-transferase p 1-1 25.7 < 10 Accession number: 2981694 Glyceraldehyde 3-phosphate dehydrogenase 36.0 40 Accession number: 7669492 Glycerol kinase, testes specific 60.5 85 Accession number: 12230075 Glycodelin 20.6 50 Accession number: 13639329 Growth/differentiation factor 15 34.1 20 Accessionnumber: 13124261 Heat shock 20 kDa like protein 22.4 20 Accession number: 6016272 Heat shock 60 kDa protein, mitochondrial 61.1 85 Accession number: 129379 Heat shock 70 kDa protein-like-I 70.4 85 Accession number: 5031769 Heat shock protein (HSPl 10 family) 94.5 85 Accession number: 7656894 Heat shock protein 105 kDa 96.8 50 Accession number: 2495344

HistoneHlA 22.1 20 Acccession number: 121916

134 THEORETICAL EXP'f. PROTEIN MOLECULAR MOLECULAR WEIGHT (kDa) WEIGHT (kDa)

Histone HIB 21.8 60 Accession number: 121919

HistoneHID 21.3 50 Accession number: 417101

Histone H2A family 14.0 40 Accession number: 121968 Accession number: 121959 Accession number: 12585257 14.0 50 Accession number: 12585251 Accession number: 12643341 Accession number: 121986 14.0 60 Accession number: 121971 Accession number: 121970 Histone H2B family 14.0 25 Accession number: 122055 Accession number: 7387743 14.0 40 Accession number: 7387741 Accession number: 7387742 Accession number: 462236 14.0 50 Accession number: 7387739 Accession number: 7387740 14.0 60 Accession number: 122026 Accession number: 7387736 Accession number: 7404367 14.0 85 Accession number: 9973351 hSMP-1 (sperm membrane protein) 55.1 85 Accession number: 1836035 lg alpha-I chain C region 37.6 60 Accession number: 113584 Inhibin alpha chain 39.7 25 Accession number: 4504697 Inhibin beta A chain 47.4 60 Accession number: 4504699 Inhibin beta B chain 45.1 40 Accession number: 1708437 Insulin-like growth factor binding protein 2 35.1 60 Accession number: 124058 Interleukin 4 17.5 20 Accession number: 4504669

135 THEORETICAL EXPT. PROTEIN MOLECULAR MOLECULAR WEIGHT (kDa) WEIGHT (kDa) 1-plastin 70.3 10 Accession number: 4505897 Isocitrate dehydrogenase 1 (NADP+) soluble 46.7 25 Accession number: 51744 71 Kallikrein 14 27.4 < 10 Accession number: 11545747 Kallistatin 48.6 25 Accession number: 5453888 Keratinocyte growth factor (FGF-7) 22.4 < 10 Accession number: 4503705 Kinesin heavy chain 109.7 220 Accession number: 4758648

76.3 25 Lactotransferrin

Accession number: 6175096 76.3 85

Major sperm fibrous sheath protein 93.5 50 Accession number: 3777541

Malate dehydrogenase, mitochondrial 35.5 85 Accession number: 5174541 Myeloperoxidase 83.8 85 Accession number: 4557759 Neutrophil cytosol factor 4 39.0 20 Accession number: 4505345 Nitric oxide synthase, inducible 131.1 25 Accession number: 1352513 Nucleobindin 2 50.2 50 Accession number: 4826870 Peroxiredoxin 2 21.9 85 Accession number: 5902726 Phosphatidylserine-specific phospholipase Al-a. 49.7 50 Accession number: 7706661

136 THEORETICAL EXPT. PROTEIN MOLECULAR MOLECULAR WEIGHT (kDa) WEIGHT (kDa) Phosphoglycerate kinase, testes specific 44.8 < 10 Accession number: 129904 Plasma serine protease inhibitor 45.6 50 Accession number: 400068 Plasminogen activator, tissue type 62.9 220 Accession number: Proactivator polypeptide (prosaposin) 58.1 60 Accession number: 11386147 Prolactin-inducible protein 16.6 10 Accession number: 4505821 16.6 25 68.7 85 Prostagladin G/H synthase 1 68.7 60 Accession number: 11386141 68.7 20 Prostate secreted seminal plasma protein 12.8 20 Accession number: 45570623 Prostate specific antigen 28.6 60 Accession number: 4758398 Prostatic acid phosphatase 44.6 70 Accession number: 6382064 44.6 50 Protein- gamma-glutamyltransferase 77.2 60 Accession number: 4759228 Putative peroxisomal antioxidant enzyme 16.9 85 Accession number: 3915613 52.1 220 52.1 60 52.1 50

Semenogelin I 52.1 40 Accession number: 4506883 52.1 < 10 52.1 10 52.1 25 52.1 20

137 THEORETICAL EXPT. PROTEIN MOLECULAR MOLECULAR WEIGHT (kDa) WEIGHT (kDa) 65.4 220 65.4 60 65.4 50

Semenogelin II 65.4 40 Accession number: 4506885 65.4 10 65.4 20 65.4 < 10 65.4 25 Serine hydroxymethyltransferase, mitochondrial 56.0 220 Accession number: 6226865 76.9 10 Serotransferrin Accession number: 4557871 76.9 85

69.4 10 69.4 < 10 Serum albumin 69.4 220 Accession number: 4557519 69.4 60 69.4 85 13.1 85 Sperm histone P2 (protamine) Accession number: 123700 13.1 40

Sperm membrane protein BS-63 198.7 220 Accession number: 5809678 Tektin 2 49.7 60 Accession number: 7657643 Testis-specific bromodomain protein 107.8 220 Accession number: 4502453 50.7 20 Thymopoietin, isoforms beta/gamma Accession number: 1174690 50.7 < 10

138 THEORETICAL EXP'f. PROTEIN MOLECULAR MOLECULAR WEIGHT (kDa) WEIGHT (kDa) 46.3 40 Thyroxine-binding globulin ( clade A) Accession number: 11422666 46.3 60

Transforming growth factor-~ induced protein IG-H3 74.7 50 Accession number: 4507467 Triosephosphate isomerase 26.7 20 Accession number: 4507645 8.6 220 Accession number: 4507761 Uromodulin 69.7 50 Accession number: 137116 Vascular endothelial growth factor 46.8 85 Accession number: 4558049 Vitamin K-dependant protein S 75.1 60 Accession number: 131086

Zn-a.-2-glycoprotein 33.9 40 Accession number: 141596

139 This technique also identified the presence of histones in seminal fluid, in particular the histone 2A and 2B family of proteins. Multiple peptides from each of these proteins were detected, corresponding to 44% and 26% sequence coverage of the proteins in the histone 2A and 2B families respectively. Because proteins in each of these families share greater than 95% sequence homology, analysis by ESI-LC/MS/MS was not able to distinguish between the different histone proteins present, i.e., a unique tryptic peptide fragment for each of these proteins was not detected by ESI-LC/MS/MS. The amino acid sequence for the proteins in the histone 2A and histone 2B families, and the tryptic peptides from each that were detected and identified, are shown in Figures 4.4 and 4.5 respectively.·

140 Figure 4.4. The amino acid sequences of the histone 2A family of proteins and the tryptic peptides that were detected (underlined) by ESI-LC/MS/MS.

Each protein in the histone 2A family consists of 130 amino acids, with the exception of

H2A.E and H2A.X (128 and 143 amino acids respectively). Highlighted are the unique sequences within each protein. The tryptic peptides detected are underlined.

141 Figure 4.4. The amino acid sequences of proteins in the histone 2A family. The tryptic peptides detected by ESI-LC/MS/MS are indicated (underlined).

HISTONE PROTEIN SEQUENCE H2A.A MSGRGKQGGKARAKAKTRSSRAGLQFPVGRVHRLLRKGNYSERVGAGAPV H2A.C / D/I/N/P MSGRGKQGGKARAKAKTRSSRAGLQFPVGRVHRLLRKGNYAERVGAGAPV H2A .E MSGRGKQGGKARAKAKTRSSRAGLQFPVGRVHRLLRKGNYAERVGAGAPV H2A.G MSGRGKQGGKARAKAKTRSSRAGLQFPVGRVHRLLRKGNYSERVGAGAPV H2A.L MSGRGKQGGKARAKAKSRSSRAGLQFPVGRVHRLLRKGNYAERVGAGAPV H2A . M MSGRGKQGGKARAKAKTRSSRAGLQFPVGRVHRLLRKAHYSERVGAGAPV H2A.0 MSGRGKQGGKARAKAKSRSSRAGLQFPVGRVHRLLRKGNYAERVGAGAPV H2A.X MSGRGKTGGKARAKAKSRSSRAGLQFPVGRVHRLLRKGHYAERVGAGAPV

H2A.A YLAAVLEYLTAEILELAGNAARDNKKTRIIPRHLQLAIRNDEELNKLLGR H2A.C / D/I/N/P YLAAVLEYLTAEILELAGNAARDNKKTRIIPRHLQLAIRNDEELNKLLGK H2A.E YLAAVLEYLTAEI LELAGNAARDNKKTRIIPRHLQLAIRNDEELNKLLGK H2A . G YLAAVLEYLTAEILELAGNAARDNKKTRIIPRHLQLAIRNDEELNKLLGK H2A.L YLAAVLEYLTAEILELAGNAARDNKKTRII PRHLQLAIRNDEELNKLLGK H2A.M YLAAVLEYLTAEILELAGNAARDNKKTRIIPRHLQLAI RNDEELNKLLGR H2A.0 YMAAVLEYLTAEILELAGNAARDNKKTRIIPRHLQLAIRNDEELNKLLGK H2A.X YLAAVLEYLTAEILELAGNAARDNKKTRIIPRHLQLAIRNDEELNKLLGG

H2A.A VTIAQGGVLPNIQAVLLPKKTESHHKAKGK H2A.C / D/I/N/P VTIAQGGVLPNIQAVLLPKKTESHHKAKGK H2A . E VTIAQGGVLPNIQAVLLPKKTESHHKTK H2A.G VTIAQGGVLPNIQAVLLPKKTESHHKAKGK H2A.L VTIAQGGVLPNIQAVLLPKKTESHHKAKGK H2A.M VTIAQGGVLPNIQAVLLPKKTESHHKAKGK H2A. 0 VTIAQGGVLPNIQAVLLPKKTESHHKAKGK H2A. X VTIAQGGVLPNIQAVLLPKKTSATVGPKAPSGGKKATQASQEY

142 Figure 4.5. The amino acid sequences of the histone 2B family of proteins and the tryptic peptides that were detected (underlined) by ESI-LC/MS/MS.

Each protein in the histone 2B family consists of 126 amino acids and shares greater than 95% sequence homology. Highlighted are the unique sequences within each protein. The tryptic peptides that were detected are underlined.

143 Figure 4.5. The amino acid sequences of the histone 2B family of proteins and the tryptic peptides that were detected (underlined) by ESI-LC/MS/MS.

HISTONE PROTEIN SEQUENCE H2B.A/G/K MPEPAKSAPAPKKGSKKAVTKAQKKDGKKRKRSRKESYSVYVYKVLKQVH H2B.C MPELAKSAPAPKKGSKKAVTKAQKKDGKKRKRSRKESYSVYVYKVLKQVH B2B.D MPEPSKSAPAPKKGSKKAVTKAQKKDGKJCRKRSRKESYSVYVYKVLKQVH B2B.B MPEPVKSAPVPKKGSKKAINKAQKKDGKKRKRSRKESYSVYVYKVLKQVH B2B.P MPEPSKSAPAPKKGSKKAITKAQKKDGKKRKRSRKESYSZYVYKVLKQVH B2B.J MPDPAKSAPAPKKGSKKAVTKAQKKDGKKRKRSRKESYSVYVYKVLKQVH H2B.L MPBPSKSAPAPKKGSKKAVTKAQKKDGKKRKRTRKESYSVYVYKVLKQVH B2B.N MPDPAKSAPAPKKGSKKAVTKAQKKDGKERKRSRKESYSZYVYKVLKQVH B2B.Q MPEPAKSAPAPKKGSKKAVTKAQKKDGKKRKRSRKESYSZYVYKVLKQVH B2B.R MPEPAKSAPAPKKGSKKAVTKAQKKDGKKRKRSRKESYSZYVYKVLKQVH B2B.S MPEPAKSAPAPKKGSKKAVTKAQKKDGRKRKRSRKESYSVYVYKVLKQVH

B2B.A/G/K PDTGISSKAMGIMNSFVNDIFERIAGEASRLAHYNKRSTITSREIQTAVR H2B.C PDTGISSKAMGIMNSFVNDIFERIASEASRLAHYNKRSTITSREIQTAVR B2B.D PDTGISSKAMGIMNSFVNDIFERIAGEASRLAHYNKRSTITSREIQTAVR H2B.B PDTGISSKAMGIMNSFVNDIFERIAGEASRLAHYNKRSTITSREIQTAVR H2B.P PDTGISSKAMGIMNSFVNDIFERIAGEASRLAHYNKRSTITSREIQTAVR B2B.J PDTGISSKAMGIMNSFVNDIFERIAGEASRLAHYNKRSTITSREIQTAVR H2B.L PDTGISSKAMGIMNSFVNDIFERIAGEASRLAHYNKRSTITSREIQTAVR H2B.N PDTGISSKAMGIMNSFVNDIFERIAGEASRLAHYNKRSTITSREIQTAVR H2B.Q PDTGISSKAMGIMNSFVNDIFERIAGEASRLAHYNKRSTITSREIQTAVR H2B.R PDTGISSKAMGIMNSFVNDIFERIAGEASRLAHYNKRSTITSREIQTAVR H2B.S PDTGISSKAMGIMNSFVNDIFERIAGEASRLPHYNKRSTITSREIQTAVR

B2B.A/G/K LLLPGELAKHAVSEGTKAVTKYTSSK H2B.C LLLPGELAKHAVSEGTKAVTKYTSSK B2B.D LLLPGELAKHAVSEGTKAVTKYTSSK B2B.B LLLPGELAKHAVSEGTKAVTKYTSSK H2B.P LLLPGELAKHAVSEGTKAVTKYTSSK B2B.J LLLPGELAKHAVSEGTKAVTKYTSSK B2B.L LLLPGELAKHAVSEGTKAVTKYTSSK B2B.N LLLPGELAKHAVSEGTKAVTKYTSSK H2B.Q LLLPGELAKHAVSEGTKAVTKYTSSK B2B.R LLLPGELAKHAVSEGTKAVTKYTSAK H2B.S LLLPGELAKHAVSEGTKAVTKYTSAK

144 4.3.3. Direct analysis of seminal fluid by MALDI-TOFMS and ESI-MS

Direct analysis ofunfractionated seminal fluid by MALDI-TOFMS revealed the presence of numerous components in the mass range 500 - 10,000 Da. Due to the signal suppression effects of the MALDI ionisation process, components greater than 10,000

Da were not detected. Because MALDI-TOFMS only provides the accurate molecular weight of molecules, capillary ESI-LC/MS/MS was employed to identify these peptides.

Shown in Figure 4.6 is the MALDI mass spectrum (linear mode) of the peptide

components of seminal fluid. The majority of the peptides detected by MALDI-TOFMS

and subsequently identified by ESI-LC/MS/MS are proteolytic peptides derived from

either semenogelin I or semenogelin II. (These peptides are listed in Table 4.6, p 148.)

Seminal basic protein ([M+Ht 5,757.65 Da), an endogenous bioactive peptide derived

from semenogelin I, was also detected [Robert and Gagnon, 1999].

Analysis of the peptides in seminal fluid by ESI-LC/MS/MS and database searching

identified 50 peptides. Of these, 35 peptides were derived from either semenogelin I or

semenogelin II. Figures 4. 7 (p 149) and 4.8 (p 150) show the amino acid sequence of

semenogelin I and II respectively, and the peptides that were identified and that are

derived from each protein.

Also detected were peptides derived from seminal fluid proteins that had previously

been identified following separation by either 1D- or 2D- gel electrophoresis. The

peptides that were identified by ESI-LC/MS/MS and database searching are listed in

Table 4.7 (pl51).

145 Figure 4.6. A MALDI mass spectrum ofunfractionated seminal fluid.

An aliquot (0.2 µL) of diluted human seminal fluid (1:10) was analysed by MALDI­

TOFMS (with a.-cyano-4-hydroxycinnamic acid as the MALDI matrix). When analysed in the linear mode, numerous peptides below 10,000 Da were detected. Indicated are those peptides that were identified by ESI-LC/MS/MS as proteolytic peptides derived from either semenogelin I or semenogelin II. (See Table 4.6, p 148.)

146

147 147

3000 3000

7300 7300

S1E,S2D S1E,S2D

7000.60 7000.60

2769.39 2769.39

2560 2560

6440 6440

6271.36 6271.36

i i

5809.64 5809.64

! !

Da Da

protein protein

2120 2120

\ \

5560 5560

5754.65 5754.65

basic basic

928.88 928.88

[M+Ht [M+Ht

Seminal Seminal

Mass(m/z) Mass(m/z)

fluid. fluid.

692.74 692.74

, ,

1680 1680

4m 4m

1

s

seminal seminal

S1B S1B

~~~~~~""'"~,M~..,._,Jt,JWJ,lw,,i).~~,.,!Jl,;.l,~~.Ji.J...... ,-~~ ~~~~~~""'"~,M~..,._,Jt,JWJ,lw,,i).~~,.,!Jl,;.l,~~.Ji.J...... ,-~~

1

1460.74 1460.74

S1H S1H

37 37

.

4140

I I

1

S1A S1A

129910 129910

I I

ofunfractionated ofunfractionated

3890.94 3890.94

. .

5.51 5.51

,1226,65 ,1226,65

3810.46 3810.46

~o ~o

118•. 118•.

1240 1240

tl7!4 tl7!4

~••llllt.l""'-'

spectrum spectrum

S1G S1G

1010.55 1010.55

mass mass

r r

32!0.78 32!0.78

j

310< 310<

,~.w~u~u-~ ,~.w~u~u-~

S1F,S2E S1F,S2E

MALDI MALDI

A A

~ ~

oJ""""'""''·J.~-

0

100 100

100 100

4.6. 4.6.

~ ~

~ ~

~ ~

a: a:

"ii "ii i i

e... e...

! !

- Figure Figure Table 4.6. Peptides that were detected by MALDI-TOFMS in unfractionated seminal fluid (see Figure 4.6, p 147) and subsequently identified by ESI-LC/MS/MS.

EXPT.MASS PEPTIDE PROTEIN [M+Ht FRAGMENT SIA Semenogelin I 1,404.38 361-373

SIB Semenogelin I 1,640.40 222-235

SIC Semenogelin I 2,446.54 80-107

SID Semenogelin I 2,481.87 195 -215

SlE Semenogelin I 2,893.95 428-453

SlF Semenogelin I 3,083.53 81-107

SlG Semenogelin I 3,211.02 195 -221

SlH Semenogelin I 4,309.32 390-427

Sll Semenogelin I 4,623.85 68-107

S2A Semenogelin II 1,591.71 86-99

S2B Semenogelin II 2,307.80 68 - 85

S2C Semenogelin II 2,509.96 195 -215

S2D Semenogelin II 2,893.95 481-507

S2E Semenogelin II 3,083.53 248-273

S2F Semenogelin II 4,469.78 236-273

148 Figure 4.7. The amino acid sequence of semenogelin I showing those peptides

identified by ESI-LC/MS/MS. Highlighted in red is the portion of the protein that

corresponds to seminal basic protein (residues 108 - 159).

MKPNIIFVLSLLLILEKQAAVMGQKGGSKGRLPSEFSQFPHGQKGQHYSGQKGKQQTESK

GSFSIQYTYHVDANDHDQSRKSQQYDLNALHKTTKSORHLGGS OOLLHNKQEGRDHDKSK 68 ______107 81 07 86~------,07

GHFHRVVIHHKGGKAHRGTQNPSQDQGNSPSGKGISSQY160.,__SNTEERLWVHGLSKEQTSVSG______160------160 ------161 f&.KGRKQGGSQSSYVLQTEELVAN-KQQRETKNSHQNKGHYQNVVEVREEHSSKVOTSLCP 182 192 215 222 235 --183 193 215 185 - 182 AHQDKLQHGSKDIFSTODELLVYNKNQHQTKNLNQDQQHGRKANKISYQSSSTEERRLHY 248 ______273 253 273

GENGVQKDVSQSSIYSQTEEKAQGKSQKQITIPSQEQEHSQKANKISYQSSSTEERRLHY

GENGVOKDVSORSIYSOTEKLVAGKSOI OAPNPKOEPWHGENAKGESGOSTNREQDLLSH 361 ---- 373 376 409 362 368 376 410 376 381 390 EQKGRHQHGSHGGLDIVIIEOEDDSDRHLAOHLNNDRNPLFT 428 453 428 462 427 ----427 427

149 Figure 4.8. The amino acid sequence of sernenogelin II showing those peptides

identified by ESI-LC/MS/MS.

MKSIILFVLSLLLILEKQAAVMGQKGGSKGQLPSGSSQFPHGQKGQHYFGQKDQQHTKSK

GSFSIQHTYHVDINDHDWTRKSOOYDLNALHKATKSKQHLGGSQQLLNYKQEGRDHDKSK . ~ • w 70 86______99

GHFHMIVIHHKGGQAHHGTQNPSQDQGNSPSGKGLSSQCSNTEKRLWVHGLSKEQASASG

AQKGRTQGGSQSSYVLOTEELVVNKOORETKNSHONKGHYQNVVDVREEHSSKLQTSLHP 192 215 22 229 23 194 215

AHODRLOHGPKDIFTTODELLVYNKNOHOTKNLSQDQEHGRKAHKISYPSSRTEERQLHH 273 253 273

GEKSVQKDVSKGSISIQTEEKIHGKSQNQVTIHSQDQEHGHKENKISYQSSSTEERHLNC

GEKGIQKGVSKGSISIQTEEQIHGKSQNQVRIPSQAQEYGHKENKISYQSSSTEERRLNS

GEKDVQKGVSKGS ISIQTEEKIHGKSQNQVTIPSQDQEHGHKENKMSYQSSSTEERRLNY

GGKSTOKDVSOSSISFOIEKLVEGKS~IQTPNPNQDQWSGQNAKGKSGQSADSKQDLLSH m Ot 481 509 488,------5·09

EQKGRYK~ESSESHNIVITEHEVAODDHLTOOYNEDRNPIST ~ ~ ~9 582

150 Table 4.7. Peptides in unfractionated seminal fluid that were identified by ESI-LC/MS/MS and database searching. These peptides were not evident by direct MALDI-TOFMS analysis of the unfractionated seminal fluid.

PROTEIN PEYfIDE FRAGMENT(S) Antibacterial protein F ALL-39 72-96 Accession number: 1706745

cl+-independent phospholipase A2 short form 383-399 Accession number: 7670058 Coagulation factor V 1397-1418 Accession number: 462045 Complement component C4A 863-880 Accession number: 116602 Complement component C 1R component 109-151 Accession number: 115204 Fibronectin 2280-2304 Accession number: 2506872 Glycoprotein hormones alpha chain 77-102 Accession number: 121312 Heat shock protein 70 testes variant 612-639 Accession number: 3461866 Histone Hl' 24-47 Accession number: 121897 Histone HlB 98-151 Accession number: 121919 Platelet-activating factor acetylhydrolase 187-209 Accession number: 2497687 Prolactin-inducible protein 34-47 Accession number: 134170 Prostasin 78-98 Accession number: 2833277 Sperm membrane protein BS-63 510-519 Accession number: 5809678

151 PROTEIN PEPTIDE FRAGMENT(S) 68-107, 81-107, 86-107, 160-182, 160-183, 160-185, 161-182, 192-215,

Semenogelin I 193-215, 222-235, 248-273, 253-273, Accession number: 4506883 361-373, 362-368, 376-409, 376-410, 376-427, 381-427, 390-427, 428-453, 428-462 68-85, 68-99, 70-99, 86-99, 192-215,

Semenogelin II 194-215, 225-229, 236-273, 253-273, Accession number: 4506885 481-507, 481-509, 488-509, 548-573, 549-582 Testis-specific protein TEX28 177-192 Accession number: 6136075

152 4.4. Discussion

A comprehensive analysis of the peptide and protein constituents of human seminal fluid was performed by combining gel electrophoresis (1D and 2D) with MALDI­

TOFMS, ESI-LC/MS/MS and database searching. Proteins in seminal fluid were separated by 2DE and identified by peptide mass fingerprinting. Additionally, proteins were also separated by ID gel electrophoresis and identified by ESI-LC/MS/MS.

Peptides in seminal fluid were also detected following direct analysis by MALDI­

TOFMS and identified by ESI-LC/MS/MS. Over 100 unique peptides and proteins were

identified in seminal fluid by this combined approach.

Separation of seminal fluid proteins by 2DE allowed visualisation of over 300 protein

spots and enabled the approximate molecular weight and isoelectric point of each protein to be determined. This technique also enabled proteins that had been post­

translationally modified, for example, phosphorylated, glycosylated or truncated

proteins, to be separated and subsequently identified. Some of the modifications are not

evident by other protein separation and identification techniques, such as the direct

multidimensional liquid chromatography tandem mass spectrometry approach.

In total, only 32 unique proteins were identified by peptide mass fingerprinting and

database searching. These included prolactin-inducible protein, prostate secreted

seminal plasma protein, serum albumin, fibronectin, and lactotransferrin. Multiple

isoforms of proteins such as prostatic acid phosphatase and prostate specific antigen are

known to be present in human seminal plasma [Taga et al., 1983; Zhang et al., 1995;

Wang et al., 1999], and some of these isoforms, as well as their degradation products,

were resolved and detected by 2DE in this study. In addition, multiple truncated forms

153 of semenogelin I and II were detected, consistent with proteolytic activity in seminal fluid. Semenogelin I and II and fibronectin are believed to be the main constituents of seminal coagulum and are rapidly degraded upon ejaculation by serine proteases such as prostate specific antigen [Malm et al., 1996; Peter et al., 1998; Robert and Gagnon,

1999]. This process of semen liquefaction plays an important role in sperm motility

(Robert and Gagnon, 1999].

Separation by 1D gel electrophoresis enabled 95 unique proteins to be identified, including some that are not amenable to separation by 2DE, such as high molecular weight proteins and basic proteins. Additionally, many of the proteins were identified in multiple molecular weight regions of the gel, indicating that they are present in several modified forms in seminal fluid. For example, proteins detected in a region lower than their theoretical mass have likely undergone ; other proteins found to migrate

at a higher mass than predicted may be present as glycosylated forms or as covalent

complexes with other proteins. For instance, prostate specific antigen has been reported

to form complexes in seminal fluid with protein C inhibitor, as well as with

antiproteases in serum such as a.-1-antichymotrypsin and a.-1-macroglobulin [Lilja et

al., 1991; Christensson and Lilja, 1994; Kise et al., 1996; Leinonen et al., 1996; Seregni

et al., 1996; Charrier et al., 1999; Zhang et al., 2000]. The nature of the complex

between prostate specific antigen and these proteins has not been completely defined.

Although gel electrophoresis can be successfully applied to separate and detect proteins,

it is not able to adequately resolve low molecular weight polypeptides(< 10,000 Da) or

endogenous peptide components. In this study, the direct analysis ofunfractionated

seminal fluid by MALDI-TOFMS revealed the presence of over 50 peptide components.

154 The identity of some of these constituents, and of other peptides not evident by

MALDI-TOFMS, was subsequently established by ESI-LC/MS/MS. The majority of the peptides detected were identified as fragments of either semenogelin I or semenogelin II, providing further evidence that these proteins undergo extensive proteolysis. These peptides are primarily generated by cleavage C-terminal to glutamine residues, tyrosine and leucine residues, indicating the presence of enzymes with

chymotryptic activity, e.g., prostate specific antigen [Malm et al., 1996; Peter et al.,

1998].

Analysis by MALDI-TOFMS also detected the presence of seminal basic protein, a bioactive peptide derived from semenogelin I that is believed to inhibit sperm motility

[Robert and Gagnon, 1999]. Although seminal fluid reportedly contains other bioactive

peptides such as oxytocin [Goverde et al., 1998], angiotensin II [O'Mahony et al.,

2000], calcitonin [Slavakis and Papadimas, 2000], somatostatin-64 [Odum and Johnsen,

1994] and thyrotropin-releasing hormone [Khan and Smyth, 1993; Cockle et al., 1994;

Gkonos et al., 1994], as well as antimicrobial peptides such as human cationic

antimicrobial peptide-18 {hCAP-18) [Malm et al., 2000], these were not detected by the

techniques used here. The inability to detect these peptides by either MALDI-TOFMS

or ESI-LC/MS/MS may be due to the high abundance of semenogelin peptides, or be

due to degradation of the peptides by the proteases present in seminal fluid.

Although numerous peptides in seminal fluid were identified, some of the abundant

constituents detected by MALDI-TOFMS remain unknown. The inability to identify

some of these peptides may be due to post-translational modifications such as C- or N­

terminal modifications (e.g., amidation, acetylation), amino acid side chain

155 modifications, and/or the presence of intramolecular bridges (e.g., oxytocin).

Because the available protein sequences are direct translations from DNA sequences, these potential modifications are not considered in the available protein databases.

156 CHAPTERS

QUANTIFICATION OF

BIOMOLECULES BY MALDI-TOF

MASS SPECTROMETRY

157 5.1. Introduction

Since its inception and commercial availability, the versatility of MALDI-TOFMS has been demonstrated convincingly by its extensive use for qualitative analysis of many different biomolecules. For example, MALDI-TOFMS has been employed for the characterisation of synthetic polymers [Wu and Odom, 1998; Marie et al., 2000], peptide and protein analysis [Nguyen et al., 1995; Zaluzec et al., 1995; Roepstorff,

2000], DNA and oligonucleotide sequencing [Bentzley et al., 1996; Faulstich et al.,

1997; Miketova and Schram, 1997], and the characterisation of recombinant proteins

[Kanazawa et al., 1999; Villanueva et al., 2001]. Recently, applications ofMALDI­

TOFMS have extended to include the direct analysis of biological tissues and single cell organisms with the aim of characterising endogenous peptide and protein constituents

[Caprioli et al., 1997; Chaurand et al., 1999; Jespersen et al., 1999; Li et al., 1999; Lynn et al., 1999; Stoeckli et al., 1999; Li et al., 2000; Stoeckli et al., 2001].

The properties that make MALDI-TOFMS a popular qualitative tool - its ability to

analyse molecules across an extensive mass range, high sensitivity, minimal sample

preparation and rapid analysis times - also make it a potentially useful quantitative tool.

MALDI-TOFMS also enables non-volatile and thermally labile molecules to be

analysed with relative ease. These factors make it worthwhile to explore the potential of

MALDI-TOFMS for quantitative analysis in clinical settings, for toxicological

screenings, as well as for environmental analysis. In addition, the application of

MALDI-TOFMS to the quantification of peptides and proteins is particularly relevant.

The ability to quantify intact proteins in biological tissue and fluids presents a particular

challenge in the expanding area of proteomics. Investigators urgently require methods to

accurately measure the absolute quantity of proteins.

158 While there have been reports of quantitative MALDI-TOFMS applications, there are many problems inherent to the MALDI ionisation process that have restricted its use

[Duncan et al., 1993; Gusev et al., 1993; Harvey, 1993; Tang et al., 1993; Muddiman et al., 1994; Nelson et al., 1994; Jespersen et al., 1995; Muddiman et al., 1995; Nicola et al., 1995; Abell and Spoms, 1996; Bruenner et al., 1996; Gusev et al., 1996; Muddiman et al., 1996; Tang et al., 1996; Wu et al., 1997; Kazmaier et al., 1998; Ling et al., 1998;

Desiderio et al., 2000; Gobom et al., 2000; Wang and Spoms, 2000; Horak et al., 2001].

These limitations primarily stem from factors such as the sample/matrix heterogeneity that is believed to contribute to the large variability seen in the observed signal intensity for analytes, the limited dynamic range due to detector saturation, and difficulties associated with coupling MALDI-TOFMS to on-line separation techniques such as liquid chromatography. Combined, these factors are thought to compromise the

accuracy, precision and utility with which quantitative determinations can be made.

Because of these difficulties, examples of quantitative applications ofMALDI-TOFMS have been limited. Most of the studies to date have focused on the quantification oflow

mass analytes, in particular, alkaloids or active ingredients in agricultural or food

products [Wang and Spoms, 1999; Wang et al., 1999; Jiang and V asanthan, 2000;

Wang et al., 2000; Yang and Chien, 2000; Wittmann and Heinzle, 2001]; other studies

have demonstrated the potential ofMALDI-TOFMS for the quantification of

biologically relevant analytes such as neuropeptides, proteins, antibiotics, or various

metabolites in biological tissue or fluid [Duncan et al., 1993; Muddiman et al., 1994;

Nelson et al., 1994; Muddiman et al., 1995; Muddiman et al., 1996; Wu et al., 1997;

Gobom et al., 2000; Mirgorodskaya et al., 2000].

159 This study explores the utility ofMALDI-TOFMS for the quantification of five different biomolecules in biological tissue or fluid: homovanillic acid in human urine; catecholamines (epinephrine and norepinephrine) in human adrenal tissue; insulin in human pancreatic tissue; LVV-hemorphin- 7 in human adrenal gland; and growth hormone in the rat pituitary gland. These samples cover a wide mass range and represent practical examples of quantification in complex real world biological matrices.

5.2. Methods

The methods employed for this study can be found in Chapter 2 (pages 45 - 49).

160 5.3. Results

5.3.1. Quantification of proteins in biological tissue

Growth hormone in rat pituitary tissue (n = 2) was successfully quantified using ~­ lactoglobulin as the internal standard. Figure 5.1 shows a MALDI mass spectrum of rat pituitary extract spiked with ~-lactoglobulin (15 nmol) using caffeic acid as the matrix.

Also evident were the doubly charged species for these proteins, as well as other minor protein components from the pituitary gland.

Figure 5.2 shows the standard curve obtained for rat methionyl growth hormone when the internal standard was not employed. This standard curve was constructed by plotting the average signal intensity for 10 determinations (y-axis) against the amount of methionyl growth hormone applied to the MALDI target (x-axis).

Figure 5.3 shows the standard curve obtained for rat methionyl growth hormone with

~-lactoglobulin incorporated as the internal standard. In this case, peak height ratios of

analyte to internal standard were used to construct the standard curve. Greater precision

was achieved when an internal standard was employed. The standard curve is linear (R2

= 0.997, slope= 1.17) over the chosen concentration range (1-21 µmol/g tissue, 0-

200 pmol met-GH), but has a non-zero intercept.

Growth hormone was measured in two different rat pituitary glands and values were

determined to be 2.40 ± 0.40 and 2.49 ± 0.25 µmol/g tissue. The values obtained in this

study are in agreement with published values for growth hormone in rat pituitary tissue

161 (2.3 - 3.2 µmol/g tissue) [Dickerman et al., 1971]. Coefficients of variation for these measurements were approximately 20% (n = 10).

162 Figure 5.1. A representative MALDI mass spectrum for growth hormone ([M+Ht

21,808 Da) in a rat pituitary extract. ~-Lactoglobulin ([M+Ht 18,364 Da) was added as

an internal standard and caffeic acid was used as the MALDI matrix.

~-Lactoglobulin ([M+Ht 18364.0 Da) 100

Rat growth hormone G> fl ([M+Hr 21808.9 Da) > ; 'I "i '1 a: 11 ! I I~ 16155.9 I \ 1\ I \ 1! I ' i~ I~

17600 19200 20800 22400 24000 Mass (m/z)

163 Figure S.2. A representative standard curve obtained for methionyl rat growth hormone

(met-rGH, 0 - 200 pmol) without an internal standard. The plot is the average peak height ratios for met-rGH (y-axis) plotted against amount (x-axis) applied to the

MALDI target. Error bars represent standard deviations.

25000 ,------~

y = 65. 77x + 755.07 20000 R2 = 0.687

~ 15000 ,n i .5 ~

D.= 10000

5000

0 50 100 150 200 250 Concentration rat met-GH (pmol)

164 Figure 5.3. A representative standard curve obtained for methionyl rat growth hormone

(met-rGH, 0 - 200 pmol) using ~-lactoglobulin (100 pmol) as the internal standard. The plot is the average peak height ratios for met-rGH to ~-lactoglobulin (n = 10) plotted

against molar concentration ratio. Error bars represent standard deviation.

3.5 ~------~

3.0 y = 1.17x - 0.14 R2 = 0.997

.5 2.5 0 :i ·-!.2.Cl ~ g> 2.0 ·-I!.!!u .! c:o.. .5~ -::C 1.5 1B \? a. .:. EID 1.0

0.5

0.0 0.5 1.0 1.5 2.0 2.5 Concentration ratio met-rGH / ~-lactoglobulin

165 5.3.2. Quantification of peptides in biological samples

5.3.2.1. Insulin in human pancreatic tissue

Insulin was quantified in human pancreatic tissue (n = 2) by incorporating porcine insulin as the internal standard. Shown in Figure 5.4 are the MALDI-TOF mass spectra of a pancreatic tissue extract obtained in both linear and reflector modes. Samples were analysed using sinapinic acid as the MALDI matrix. Complete resolution of human insulin and the internal standard (porcine insulin) was achieved in both modes of analysis. The amino acid sequences for porcine and human insulin are shown in Figure

5.5 (p 169). These polypeptides differ by one amino acid and the corresponding mass difference for this amino acid substitution is 30 Da. (Human insulin has a residue at position 109 whereas porcine insulin has an alanine residue at this.position.)

Figure 5.6 (p 170) shows the standard curve obtained for insulin in both linear (R2 =

0.999, slope= 1.08) and reflector modes (R2 = 0.999, slope= 0.98) using peak height ratios of analyte to internal standard. For samples analysed in reflector mode, the peak heights of each isotopic form in each respective insulin isotope cluster were summed to

calculate the peak height ratios. The values obtained for insulin in each of the pancreatic

tissue samples are shown in Table 5.1 (p 172) and are the average often separate

determinations.

166 Figure 5.4. Representative MALDI-TOF mass spectra for the analysis of a mixture of human insulin ([M+Ht 5,809 Da) and porcine insulin ([M+Ht 5,778 Da).

A. Human and porcine insulin detected in linear mode with sinapinic acid as the

MALDI matrix.

Porcine insulin ([M+Ht Da) 6000 sns.s

5000

,4000

Cl) E :::, Human insulin 0 (.) 3000 ([M+Ht 5809.0 Da)

2000

1000

0

5720 5740 5760 5780 5800 5820 58,40 5860 Mass (m/Z)

167 Figure S.4. Representative MALDI-TOF mass spectra for the analysis of a mixture of human insulin ([M+Ht 5,809 Da) and porcine insulin ([M+Ht 5,778 Da).

B. Detection of human and porcine insulin in reflector mode using sinapinic acid as the

MALDI matrix.

2500

Porcine insulin

2000

1500

~:::, Human insulin ..... 0 (.) •n~ r-- 1000 CD r-- . . a::i en to C !ii CD~ fB Lli Cil m 500 §

0 5775 5780 5785 5790 5795 5800 5805 5810 5815 Mass (mfz)

168 Figure 5.5. The amino acid sequence for human and porcine insulin.

Insulin is composed of two subunits joined by 2 intramolecular disulfide bonds. Insulin

A chain consists of 21 amino acids and insulin B chain consists of 30 amino acids.

Shown are the amino acid sequences for porcine and human insulin A and B chains. The sequence for these polypeptides differ by one amino acid (highlighted).

Amino acid sequence

HUMAN INSULIN A CHAIN GIVEQCCTSICSLYQLENYCN

PORCINE INSULIN A CHAIN GIVEQCCTSICSLYQLENYCN

HUMAN INSULIN B CHAIN FVNQHLCGSHLVEALYLVCGERGFFYTPKT

PORCINE INSULIN B CHAIN FVNQHL CGSHLVEAL Y LVCGERGFFY TPKA

169 Figure 5.6. Representative standard curves obtained for human insulin (0 - 600 pmol) using porcine insulin (200 pmol) as the internal standard. These are plots of average peak intensity ratios for human insulin to porcine insulin (n = 10). Error bars represent

standard deviation.

A. Standard curve for human insulin obtained in linear mode.

as-.------

3 y = 1.08x: + 0.04 .5 R2:0.999 ]25 0 .5 .::e l!.5 ~I:! 2 -o c_• 0. ,! C c=1.5 -::::, .:.=1n I .5 a. i 1 E ::::, ::c 0.5

0-----1------t------t-----+----+----+----' 0 0.5 1.5 2 25 3 Concemalion ratio HLmat Insulin/ porcine insulin

170 Figure 5.6. Representative standard curves obtained for human insulin (0 - 600 pmol) using porcine insulin (200 pmol) as the internal standard. These are plots of average peak intensity ratios for human insulin to porcine insulin (n = 10). Error bars represent standard deviation.

B. Standard curve for human insulin obtained in reflector mode.

3.5 ~------~

3 y = 0.98x + 0.04 C R2 =0.999 ·-::::,o= 1ij IO .. c2.5 ~-; e-10 C a, 2 .. e0 .5 Q. ~­as C !_ =§ 1.5 "0 ~ Cl) ·- E c E as 1 ::::, E ,,, ::::, ::c 0.5

o~----+-----+-----+------1------+------1--...J 0 0.5 1.5 2 2.5 3 Concentration ratio Human insulin/ porcine Insulin

171 Table 5.1. Insulin concentrations measured in human pancreatic tissue obtained from two cadavers. Each pancreatic tissue was divided into three sections and analysed in both linear and reflector modes. The concentration of insulin is the average of ten determinations from each tissue sample.

LINEARMODE REFLECTOR MODE

HUMANINSULIN HUMAN INSULIN CV(%) CV(%) (nmol/g tissue) (nmol/g tissue) A 20.9 1.6 19.7 6.1

Pancreas 1 B 18.7 1.4 18.2 7.1

C 16.8 2.9 16.l 6.8

A 12.6 3.3 12.1 6.4

Pancreas 2 B 15.2 2.3 13.6 11.2

C 12.9 3.0 10.7 6.6

172 5.3.2.2. L VV-hemorphin-7 in human adrenal gland

LW-hemorphin- 7 was quantified in whole human adrenal tissue (n = 10), adrenal cortex (n = 10), adrenal medulla (n = 9), and in pheochromocytoma tissue (n = 3). All analyses were performed in linear mode using cx.-cyano-4-hydroxycinnamic acid as the

MALDI matrix.

To determine the concentration of LW-hemorphin- 7 in pancreatic tissue, a standard addition curve was constructed using a pancreatic tissue extract as the matrix. Linear standard curves (R2 = 0.999) were routinely obtained over the concentration range 7 to

500 nmol/g tissue (0-600 nmol/g tissue of human LW-hemorphin-7 added). The structural homologue, rat LW-hemorphin-7 (300 nmol/g tissue), was used as the internal standard.

Figure 5.7 shows the results obtained by MALDI-TOFMS analysis when compared to electrospray analysis of the same extracts [Cerpa-Poljak et al., 1997]. To determine the concentration of LW-hemorphin- 7 by MALDI-TOFMS, each tissue sample was analysed 10 times and the average concentration calculated. The values obtained by

MALDI-TOFMS for LW-hemorphin-7 were found to correlate well with the electrospray data generated from the same samples [Cerpa-Poljak et al., 1997].

173 Figure 5. 7. A comparison of L W-hemorphin-7 concentrations in human adrenal gland

determined by MALDI-TOFMS and ESI-MS. The values obtained by MALDI-TOFMS

are the average of 10 separate determinations. Co-efficients of variation (CV) for each measurement were less than 10%.

800-.------~

700 R2 = 0.796 C 0 • i 600 'E Cl) U) g 'ii' "i 500 0:::, > u • 'ii .!! C ":" ;, as 400 C :::. (I) :c I:& Q. C I ...... ci5 300 ~ w .! 200

~ 100

0 50 100 150 200 250 300 350 400 LVV-hemorphin-7 concentration (nrnoVg tissue) MALDI-TOFMS analysis

174 5.3.3. Quantification of low mass analytes in biological samples

5.3.3.1. Catecholamines in human adrenal gland

The catecholamines, epinephrine and norepinephrine, were quantified in normal human adrenal gland (n = 3) and pheochromocytoma tissues (n = 3) using ferulic acid as the

MALDI matrix. Standard addition curves (concentration range 0- 18 µmol/g tissue) for both epinephrine and norepinephrine were constructed using an extract of adrenal cortex as the matrix. Linear standard curves were obtained for both epinephrine (R2 = 0.999) and norepinephrine (R2 = 0.998) when the respective stable isotopomers were used as the internal standard. For epinephrine, [2H3]-epinephrine (4 µmol/g tissue) was used as the internal standard; [2H3]-norepinephrine (6 µmol/g tissue) was used as the internal standard for norepinephrine. Figure 5.8 shows an example of epinephrine, norepinephrine and their internal standards in an adrenal tissue extract. All spectra were acquired in reflector mode. Table 5.2 shows the results for the quantification of epinephrine and norepinephrine in normal adrenal tissue and pheochromocytoma tissue.

175 Figure S.8. A representative MALDI-TOF mass spectrum for norepinephrine,

epinephrine and their respective internal standards in human adrenal extracts.

Samples were analysed in reflector mode and ferulic acid was used as the MALDI

matrix.

Epinephrine [2H3]-Epinephrine ([M+H( 184.0972 Da) ([M+H( 187.1161 Da) 100 ~ I

-0~

Norepinephrine ([M+H( 170.0823 Da) [2H3]-N~repinephrine

([M-,H] 173.1005 Da) /' I

172 176 181 185 190 Mass (m/z)

176 Table 5.2. Concentrations ofnorepinephrine and epinephrine in hwnan adrenal tissue (n

= 3) and pheochromocytoma tissue (n = 3). Values are the average of 10 separate determinations.

NOREPINEPHRINE EPINEPHRINE

MEANVALUE MEAN VALUE CV(%) CV(%) (nmol/g tissue) (nmol/g tissue) 233 16 279 6 Adrenal gland A

Adrenal gland B 510 13 582 10

Adrenal gland C 499 17 690 19

Pheochromocytoma 1 3,989 7 184 16

Pheochromocytoma 2 6,708 8 2,854 8

Pheochromocytoma 3 59,940 5 5,735 4

177 5.3.3.2. Homovanillic acid in human urine

Homovanillic acid was quantified in urine obtained from healthy volunteers (n = 19).

An example of a MALDI-TOF mass spectrum showing the molecular ion of homovanillic acid and its internal standard ([2H5]-homovanillic acid, 100 pmol) in a urine sample is shown in Figure 5.9. All spectra were acquired in reflector mode using

2,5-dihydroxybenzoic acid as the MALDI matrix. Because the molecular ion for both molecules was the most abundant species in the mass spectrum, the peak height and peak area for these ions were used for all calculations. Linear standard curves

(concentration range 0- 1,000 pmol) were obtained when either peak height (R2 =

0.999, slope= 0.97) or peak area (R2 = 0.999, slope= 1.05) ratios_were calculated.

Standards and samples were analysed 10 times to obtain an average value. The co­

efficient of variation was less than 10% for each sample analysed.

The same 19 urine samples were also independently analysed for homovanillic acid

content at a clinical laboratory by high performance liquid chromatography (HPLC).

These values correlated well with those obtained by MALDI-TOFMS when either peak height or peak area ratios were used. Figure 5 .10 shows the results for both peak height

and peak area ratio values.

178 Figure S.9. A representative MALDI mass spectrum for homovanillic acid detected in

human urine. The internal standard [2H5]-homovanillic acid (I 00 pmol) was added prior

to analysis. Samples were analysed in reflector mode using 2,5-dihydroxybenzoic acid

as the MALDI matrix.

[2H5]-Homovanilllc acid (M+. 187.0896 Da) Homovanillic acid 100 (M+. 182.0577 Da)

~ ui C f 5 Cl) ;> 'i a:

179 Figure S.10. Homovanillic acid concentrations in human urine (n = 19) determined by

MALDI-TOFMS vs homovanillic acid values determined by a commercial clinical

laboratory (HPLC method). The values obtained by MALDI-TOFMS analysis are the

average of 10 measurements for each sample.

A. Homovanillic acid concentrations obtained using peak height ratios.

12000

R2 = 0.915 C 0 10000 =! a, C 'i -Cl) u f 8000 C C o ::::r as u E u, :5! ::& 6000 u tE u. as C 0 .!:! - '7 ~ 9 4000 as CC • g ::& • E 0 2000 ::c

0+-----'.___-~-~--~--~------0 1000 2000 3000 4000 5000 6000 7000 8000 Homovanillic acid concentration (nmoVml}

180 Figure 5.10. Homovanillic acid concentrations in human urine (n = 19) determined by

MALDI-TOFMS vs homovanillic acid values determined by a commercial clinical laboratory (HPLC method). The values obtained by MALDI-TOFMS analysis are the average of 10 measurements for each sample.

B. Homovanillic acid concentrations obtained using peak area ratios.

7000 R2 = 0.942 6000 C 0 • .:;

l! .!5000,n ..C >- pj 'ii C C 0 :::r as 4000 u E u, "Cl ::::, ::E 0 LL. uas ! e3000 ~ cI ·cas ..I > ;2000 0 E 0 ::c 1000

0 0 1000 2000 3000 401)0 5000 6000 7000 8000 Homovanilllc acid concentration (nmol/ml)

181 5.4. Discussion

MALDI-TOFMS has been applied successfully to the quantitative analysis of several biologically relevant molecules ranging in molecular weight from 170 - 22 kDa. Linear calibration curves (R2 > 0.99) were routinely obtained for each analyte over the chosen concentration range. Each analyte was detected and quantified in a biological matrix with minimal sample preparation. The accuracy of the approach was demonstrated by comparison of these results with literature values, or by independent analysis. In each case, the values obtained by MALDI-TOFMS were in good agreement with those reported or obtained by other techniques [Dickerman et al., 1971; Cerpa-Poljak et al.,

1997].

This study has also shown the potential ofMALDI-TOFMS for the quantification of low mass analytes, especially in a clinical diagnostic setting. Factors such as the speed

of analysis(< 1 min per sample) and minimal sample preparation make MALDI­

TOFMS appropriate for these types of analyses. Additionally, the accurate mass

capability of MALDI-TOFMS offers an additional level of specificity when quantifying

low mass analytes because mass assignments within 5 ppm can be achieved routinely.

In this mode, elemental composition can confirm assignment (using internal calibration

in reflector mode). In this study, levels ofhomovanillic acid measured by MALDI­

TOFMS were found to correlate well with those obtained by HPLC analysis in a

commercial laboratory setting. Both peak height ratios and peak area ratios were used

for the quantification of homovanillic acid and similar results were obtained when either

was employed. This indicates that for the quantification of low mass analytes, either one

can be used to obtain accurate and precise measurements. Additionally, elevated levels

of epinephrine and/or norepinephrine were measured in pheochromocytoma tissue when

182 compared to normal adrenal glands, with the exception of epinephrine in one of the pheochromocytoma samples analysed. This may occur because cortisol, produced by the adrenal cortex, is a necessary factor for the conversion of norepinephrine to epinephrine via phenylethanolamine N-methyltransferase. A large medullary tumour grows at the expense of the adrenal cortex reducing the availability of this co-factor, thereby reducing the synthesis of epinephrine by the adrenal gland.

The utility ofMALDI-TOFMS for the absolute quantification of peptides and proteins in biological tissue has also been demonstrated. Growth hormone in rat pituitary tissue was quantified employing ~-lactoglobulin as the internal standard and caffeic acid as the

MALDI matrix. Although broadening of the signal obtained for both growth hormone and ~-lactoglobulin was observed (see Figure 5.1, p 163), possibly due to protein heterogeneity and the formation of protein/matrix adducts [Beavis and Chait, 1989], the amount of growth hormone in rat pituitary gland could still be accurately determined.

Human insulin and LVV-hemorphin- 7 were also quantified in pancreatic tissue and adrenal gland extract respectively. In both cases, structural homologues were employed

as internal standards. Insulin in pancreatic tissue was quantified using both linear and reflector modes and similar results were obtained when either mode was employed.

Although an improved level of precision was achieved in linear mode, either mode of

analysis can still be used for peptide quantification. Peptide quantification by MALDI­

TOFMS was also demonstrated with L VV-hemorphin-7. The results obtained by

MALDI-TOFMS were found to compare well with those obtained by ESI-MS analysis

of the same extracts [Cerpa-Poljak et al., 1997]. However, the values measured in this

183 present study were found to be slightly lower than those previously obtained, possibly due to degradation of the adrenal extract during storage (2 years).

In this study, internal standards were employed to reduce variations that arise from heterogeneity in the sample/matrix co-crystallisation process. For the low mass analytes, isotopomers were employed as the internal standard, and structural analogues were used for the analysis of LVV-hemophin- 7 and insulin. In the case of growth hormone, homologous proteins, such as human growth hormone, were initially investigated as potential internal standards, but the two species could not be adequately resolved by

MALDI-TOFMS. Therefore, a structurally unrelated protein of similar mass (i.e.,

~-lactoglobulin) was employed as the internal standard. Here the precision was significantly lower than those observed when structural analogues and isotopomers were employed. These differences are likely due to the different physical and chemical properties between the two proteins, notably the differences in ionisation efficiencies of the two proteins, as well as differences in solubility of the proteins in the chosen matrix solvent (80 % CH3CN) [Cohen and Chait, 1996]. Although acceptable mass accuracy was obtained in this instance, the ideal internal standard for quantitative analyses is an isotopomer, thereafter a close structural analogue. Based on these results, and as might be expected, compounds with little chemical or physical similarity are less than ideal

when employed as internal standards; however, they do allow for greater precision than

when peak intensity alone is plotted against concentration (see Figures 5.2 and 5.3, p

164- 165).

The incorporation of an internal standard also serves to increase the precision with

which a value can be measured. Precision may be improved further by employing

184 alternative methods of sample/matrix deposition, such as electrospray deposition of sample and matrix [Hensel et al., 1997], the addition of additives such as nitrocellulose

[Preston et al., 1993; Gusev et al., 1995], or the application of sample and matrix using the fast-evaporation technique [Vorm et al., 1994; Nicola et al., 1995]. These techniques result in the formation of smaller crystals uniformally distributed, and therefore this minimises observable variations in signal intensity between successive laser shots, as well the variations observed from sample to sample.

Although proteins can be accurately quantified using this technique, there are limitations: a restricted dynamic range and limited sensitivity. Due to the signal suppression effects of the MALDI process, the limit of detection of a protein is dependant on the complexity of the sample matrix. A highly purified protein sample containing only the protein of interest and the internal standard yields lower limits of detection and improve sensitivity. This issue is important when attempting to measure

circulating biomarkers that are often present in low abundance with respect to other

serum proteins (e.g., serum albumin and hemoglobin). It has been shown that by

combining MALDI-TOFMS with immunoaffinity or other sample purification steps, the

dynamic range and sensitivity can be improved [Nelson et al., 1995; Tubbs et al., 2001].

The potential quantitative applications ofMALDI-TOFMS are unlimited. On-going

technical advances will likely provide solutions to some of the current obstacles, and

these will allow MALDI-TOFMS to be used routinely in quantitative settings. MALDI­

TOFMS can offer high sensitivity and specificity, and this study has shown that it can

also be used for diagnostic purposes where the presence of a particular analyte has

either markedly increased or decreased in a pathological state. Also, proteins such as

185 circulating growth hormone and insulin are currently measured using immunochemical techniques, and these techniques alone may not provide sufficient specificity for a

confident diagnosis [Strasburger and Dattani, 1997; Strasburger, 1998; Chevenne et al.,

1999]. By utilising MALDI-TOFMS, these and other biomarkers can be accurately quantified.

186 CHAPTER6

ALTERNATIVE APPROACHES TO

PROTEIN QUANTIFICATION

187 6.1. Introduction

Different approaches to protein characterisation have been described, but among the most commonly adopted today is detection by mass spectrometry, followed by identification through database searching. Strategies include separation of proteins by gel electrophoresis, or chromatographic separation of proteolytic peptides generated from complex protein mixtures. By employing two dimensional gel electrophoresis

(2DE), protein post-translational modifications, such as phosphorylation, glycosylation

and truncation can be visualised [Larsen and Roepstorff, 2000; Sarioglu et al., 2000]

and information concerning changes in protein expression between samples of similar biological origin can be obtained [Gros et al., 2001]. Although 2DE is the most widely

accepted technique for global protein separation, the ability to obtain an accurate measurement of absolute amounts of protein, or to determine relative levels with

acceptable precision, remain a considerable challenge.

Various approaches have been employed to determine the relative concentrations of

proteins in biological samples. The majority of these involve isolation and purification

of the protein(s) of interest prior to quantification. Standard biochemical approaches for

protein quantification include immunochemical techniques such as ELISA,

radioimmunnoassays, or derivatisation with specific reagents to promote detection by

fluorescence following protein isolation [Krull et al., 1997; Weller, 2000]. Even though

these techniques can offer high sensitivity, they must be targeted toward particular

proteins and therefore lack versatility. In contrast, recent advances in proteomic

technologies now enable proteins to be quantified on a global scale. Because proteomic

approaches allow many hundreds of proteins to be separated and identified in a few

steps, the ability to quantify these with precision and accuracy is an important objective.

188 2DE and densitometry have been employed to estimate the relative abundances of proteins in tissue obtained from normal and disease states, or between wild type and treated cells [Newsholme et al., 2000; Poirier et al., 2001; Yoo et al., 2001]. Because many proteins can be simultaneously detected, this approach also enables the presence of protein isoforms and modified proteins to be monitored. Many detailed studies investigating methods to improve the precision of quantitative measurements made by

2DE have been reported. These have been focused on the development of computer algorithms to improve protein spot detection and matching [Pleissner et al., 1999;

Kriegel et al., 2000], as well as investigations into compatible protein stains that offer high sensitivity, linearity and a wide dynamic range [Yan et al., 1999; Berggren et al.,

2000; Lopez et al., 2000; Yan et al., 2000]. Although this approach is widely applicable

for both in vivo and in vitro studies, many limitations exist. For instance, it is widely recognised that a two dimensional gel does not provide a complete representation of the

entire proteome of a chosen organism [Gygi et al., 2000]. Further, hydrophobic proteins, basic proteins, high mass and low mass proteins are not readily amenable to

2DE separations, and at times, co-migration of proteins may lead to inaccurate estimates

of protein expression levels.

Alternatively, methods to quantify proteolytic peptides by mass spectrometry following

differential isotope labeling have also been developed. For example, the relative

quantities of gel separated proteins obtained from matched biological samples have

been determined by isotopic labeling of either the N- or C- terminal residue of

proteolytically derived peptides [Munchbach et al., 2000; Yao et al., 2001]. MALDI­

TOFMS and peptide mass fingerprinting is then used to identify the protein and to

189 determine the relative amounts of each. In addition to enabling proteins to be identified and quantified, this approach also facilitates interpretation of tandem mass spectral data and protein identification by de novo sequencing [Munchbach et al., 2000].

Additionally, proteins have been quantified by combining proteins obtained from cells cultured in normal medium with those obtained by metabolic labeling of proteins in cell cultures maintained in an isotopically enriched medium [Oda et al., 1999; Conrads et al., 2001; Smith et al., 2001]. Here the proteins are electrophoretically separated, proteolytically digested, and the relative abundance of each protein (i.e., labeled and unlabeled) is quantified by ESI-LC/MS. Although demonstrated to offer precise and accurate quantification of proteins, and has the potential for quantification of modified proteins, [Oda et al., 1999], the utility of this approach is limited to monitoring protein

expression in in vitro studies.

Other techniques that provide a global approach to protein quantification, such as

isotope-coded affinity tagging (ICAT), have also been developed. These allow the

relative amounts of many proteins to be determined simultaneously when two samples

containing an identical set of proteins are compared [Gygi et al., 1999]. Proteins in each

sample are proteolytically digested and the resulting peptides are reacted with mass tags

that are chemically identical, but differ in mass. Peptides containing the mass tags are

then purified, combined and analysed. In contrast to techniques involving metabolic

labeling of proteins, this approach is more universally applicable; however, it involves

the incorporation of a mass tag that is highly specific for one amino acid residue (i.e.,

cysteine) and therefore only those proteins containing cysteine residues can be

quantified. More recently, a similar technique has been described which is more general

190 in nature [Goodlett et al., 2001]. Instead of a single specific amino acid being derivatised, here all C-tenninal residues, and the acidic side chains of aspartic acid and glutamic acid residues, are converted to methyl esters prior to analysis and quantification. These procedures do not require individual proteins to be separated prior to analysis, and in contrast to approaches involving quantification of electrophoretically separated proteins, rely solely on the separation of peptides by liquid chromatography.

Even though many of these methods have been successfully applied to the determination of relative amounts of proteins in model systems, they have not been readily adopted. This is due to the level of precision that can be achieved, limitations relating to the proteins that can be quantified, less than ideal yields following the derivatisation or mass tagging procedure, and the complexity of the overall process. For example, incorporation of chemical derivatives may confound analysis due to unexpected side reactions or incomplete derivatisation [Krull et al., 1997].

Quantification strategies that overcome these difficulties, offer accurate and precise measurements of changes in protein expression, and that are widely applicable to many different biological systems are therefore required.

This study describes a procedure to determine the relative abundances of proteins: here applied to human tear fluid obtained under two different biological conditions. Because tear is believed to perform different biological functions under different conditions, human tear fluid derived from the open eye and closed eye state reportedly contain the same protein components but at different concentrations [Sack et al., 2000]. During the open eye state, the tear film primarily serves to protect the external ocular surface from potential pathogens, provide nourishment for the corneal surface, and to remove waste

191 products resulting from cellular respiration [Coyle et al., 1989; Wollensak et al., 1990]; during eye closure, tear flow is static, and both the nourishment of the corneal and conjunctiva! surfaces and removal of potentially toxic waste products are less efficient.

Therefore, the primary function of tear fluid proteins during prolonged eye closure is to maintain the viability of the ocular surface [Sack et al., 2000].

The analytical process described here involves the esterification of proteolytically derived peptides with butanol, either native or deuterated butanol (d10-butanol), and subsequent analysis by ESI-LC/MS to measure the relative amounts of each protein.

Carboxylic acid groups of glutamic acid and aspartic acid residues are specifically targeted, as well as the C-terminal residue of each peptide. In contrast to ICAT, this approach is not selective for a specific amino acid residue and as a result, potentially all proteins in a sample can be quantified. In addition, this approach offers multiple determinations of relative levels for each protein because multiple derivatised peptides are formed and can be measured. Proteins in open and closed eye tear fluid were chosen as the model system to demonstrate the potential of this approach for determining the relative quantities of proteins in a complex biological sample.

6.2. Methods

Details of the methods employed for this study can be found in Chapter 2 (pages 38 -40).

192 6.3. Results

6.3.1. Demonstration of the approach

To demonstrate the feasibility of the approach, horse apomyoglobin was chosen as a model protein. An aliquot of protein was digested using trypsin and the resulting peptides divided into two portions (ratio of 1: 1) prior to derivatisation with either acidified butanol (do) or acidified deuterated butanol (d10-butanol). This resulted in the esterification of the C-terminus of all peptides, as well as all acidic amino acid residues, i.e., glutamic acid and aspartic acid (Figure 6.1). The addition of each butyl ester resulted in an increase in the peptide mass of 56 Da for do-butanol, and 65 Da for d10- butanol. Following derivatisation, samples were combined and analysed by MALDI­

TOFMS and ESI-LC/MS.

One of the difficulties associated with this study was to ensure complete derivatisation of the peptide mixture. Therefore, analysis by MALDI-TOFMS was performed to assess the conversion of the free acids of the tryptic peptides to the butyl ester under the

conditions employed (i.e., 3hrs, 70°C). Figure 6.2A (p 196) shows a representative

MALDI mass spectrum of an apomyoglobin tryptic digest prior to esterification. Tryptic

peptides derived from the predicted sequence of apomyoglobin are indicated. The

remaining peptides are the result of non-specific enzymatic cleavage of apomyoglobin,

or arise from impurities in the protein sample. (Analysis of the tryptic peptides by ESI­

LC/MS/MS identified contaminant proteins such as tropomyosin, hemoglobin and

phosphatidylethanolamine binding protein.) Figure 6.2B (p 197) shows a MALDI mass

spectrum of the same digest following derivatisation with acidified do-butanol.

Complete derivatisation of all tryptic peptides was achieved under the conditions

193 employed. Peptides derived from apomyoglobin that were detected at multiples of 56

Da higher in mass due to butyl esterification are highlighted.

194

195 195

and and

acid acid

glutamic glutamic

3 3

D D

as as

C C

D2

such such

C

D2

oH oH

C) C)

ESI-LC/MS ESI-LC/MS

°

C

,o ,o

'

70

D2

BY BY

residues residues

, ,

C C

-

OC

hrs

X X

~ ~

3 3

-

Acidic Acidic

-butanol/HCI -butanol/HCI

M, M,

X X

10

d

(1 (1

-

IDENTIFY IDENTIFY

X X

peptide. peptide.

-

l l

X X

-x-~c, -x-~c,

-

X

-

tryptic tryptic

a a

X X

H2N H2N

-

of of

s s

u

H2N H2N

ANALYSE ANALYSE

C-termin

e e

and and

h

t

ESI-LC/MS ESI-LC/MS

to to

BY BY

70°C) 70°C)

oH oH

ester ester

l l

,o ,o

'

COMBINE COMBINE

OCH2CH2CH2CH3 OCH2CH2CH2CH3

hrs, hrs,

C C

buty

3 3

~ ~

-

a a

X X

C' C'

-butanol/HCI -butanol/HCI

M, M,

of of

0

-

d

(1 (1

X-

X X

-

-

l l X

X X

addition addition

-

-

residue residue

X X

X X

the the

-

acid acid

X X

N - N

2

-

H

mino mino

N N

ESI-LC/MS ESI-LC/MS

showing showing

2

a

esterified. esterified.

H

ny ny

BY BY

a

also also

are are

notes notes

Schematic Schematic

e

d

acid acid

DENTIFY DENTIFY

6.1. 6.1.

'X' 'X'

I

rtic rtic

a

p

as Figure Figure 196

in

3000

analysed

were

indicated.

are

2602.32

Samples

97-108

2500

trypsin.

apomyoglobin

with

from

digestion

derived

1983.01

79-96

2000

1885.98

103-118

following

peptides

-

I.I

(m/z)

•.

1854.92

tryptic

Mass

80-96

1606.85

17-31

apomyoglobin

matrix.

1500

119-133

predicted

the

horse

as

The

of

1271.67 1502.67

I

32-42

acid

l~\111>

'

1158.59

I

33-42

"

spectrum

esterification.

to mass

1000

941.49

I

146-153

prior

MALDI

748.43

'!

134-139

digest

I

I

a-cyano-4-hydroxycinnamic

I

tryptic

F

500

0

using

1 representative

i

i

'#.

~

a: - -

"i :;

A

mode

6.2.

Apomyoglobin

Figure

reflector

A. in

197

in

increased

that

analysed

2500

were

apomyoglobin

Samples

2166.23

32-47

from

trypsin.

2100

derived

with

1889.08

32-45

1832.05

peptides

17-31

digestion

Those

1700

following

1434.91

64-n

(m/z)

do-butanol.

Mass

apomyoglobin

acidified

matrix.

1300

the

highlighted.

with

horse

as

are

1174.82

43-50

of

acid

spectrum

derivatisation

esterification

mass

900

860.55

134-139

-1139

butyl

789.51

following

to

135

MALDI

due

digest

Da

676.41

56

a-cyano-4-hydroxycinnamic

136-139

tryptic

of

0500

using

1

representative

C ~

fl)

~

~

0

·;;;

- a: - i

:;

A

mode

multiples

6.2.

by

Apomyoglobin

Figure

reflector

B.

mass Following derivatisation, the esterified peptides were combined (ratio of 1: 1). To determine the relative quantity of each peptide, an aliquot of the combined apomyoglobin tryptic digest was analysed by ESI-LC/MS. The deuterated peptides were found to have a slightly earlier retention time than the corresponding unlabeled peptide.

Co-eluting peptide pairs showing an appropriate mass difference were detected (SIN >

3), with the observed mass difference being dependant on the number of incorporated butyl esters and the charge state of each peptide. The relative amount of each co-eluting peptide pair was determined by integrating the area under the extracted ion chromatograms for the do- and d10- butylated peptides.

Analysis by ESI-LC/MS detected many co-eluting tryptic peptide pairs that were derived from apomyoglobin following digestion and derivatisation with do-butanol or d10-butanol. Some of these peptides were a result of non-specific enzymatic cleavage of apomyoglobin, or were peptides derived from contaminant proteins in the sample. Some of the esterified tryptic peptides derived from apomyoglobin, as well as their measured peak area ratios, are listed in Table 6.1. The average observed ratio (do-/ d10- butyl peptide esters) was determined to be 1.03 ± 0.19 (CV 18 %). This indicates that this technique can be used to precisely and accurately determine the relative amount of proteins in a complex sample.

198 Table 6.1. The relative abundance oftryptic peptides derived from apomyoglobin.

Following derivatisation with either do- or d10- butanol, equal aliquots were combined and analysed by ESI-LC/MS. The observed ratio for do-/ d10- butylated peptides was determined to be 1.03 ± 0.19 (CV 18 %).

TRYPTIC CHARGE OBSERVED RATIO

PEPTIDE STATE (do-butanol / d10-butanol) 17-31 +2 0.97

32-42 +2 0.89

43-45 +l 1.29

48-50 +l 1.13

51-56 +l 1.11

57-63 +l 0.74

97-102 +l 1.32

99-102 +l 0.79

134-139 +l 1.14

140-145 +l 0.97

148-153 +l 0.91

199 6.3.2. Identification of esterified peptides in open and closed eye tear

Aliquots (10 µg of total protein) from open and closed eye tear fluid obtained from healthy individuals were digested using trypsin and the resulting peptides derivatised with either acidified do- or d10- butanol respectively. To ensure complete derivatisation of the peptides an aliquot of each was analysed by MALDI-TOFMS prior to quantification by ESI-LC/MS. Figure 6.3 shows a MALDI mass spectrum of a combined aliquot of open and closed eye tear following tryptic digestion and butanol esterification. Some of the peptide pairs, identified by a mass difference of 9 Da ( or multiple of), are highlighted.

Esterified peptides derived from both open and closed eye tear were analysed separately by ESI-LC/MS/MS and identified by database searching prior to quantification. This

ensured that the same proteins could be detected and identified in both types of tear

fluid. Figure 6.4 (p 202) shows an example of a fragment ion spectrum of an esterified peptide derived from open eye tear. Database searching using SEQUEST identified this

as the tryptic peptide 107 - 120 of lacrimal proline-rich protein containing two butyl

esters. Some of the other proteins detected include growth factors, immunoglobulins,

lactotransferrin, lysozyme, tear lipocalin, and many serum proteins. Due to the complex

nature of human tear, and the limited solubility of some peptides in butanol, only a

subset of the proteins previously identified by ESI-LC/MS/MS were detected and

identified (see Chapter 3). In this case, only the most abundant proteins (ea. 50 proteins)

in tear fluid were identified in open or closed eye tear fluid following butanol

esterification (cf. with the 400 identified previously).

200

201 201

in in

,.;l'/f. ,.;l'/f.

'

2200 2200

,14 ,14

.

present present

l'/

15 15

.

......

14 14

.

2052.24 2052.24

987 987

l l

,It:" ,It:"

1

I I

proteins proteins

i i

2034

the the

' '

i

j j

from from

~/ ~/

1 0 0 1

1880 1880

.

derived derived

08 08

"

1976

13.96 13.96

pairs pairs

1732

1 1

peptide peptide

tryptic tryptic

531.99 531.99

1560 1560

1513.86 1513.86

) )

m/z

(

d10-butylated d10-butylated

nd nd

Mass Mass

a

~~---i~~/~~1'~1 ~~---i~~/~~1'~1

1315.99 1315.99

~ ~

~

\ \

1297.88 1297.88

91 91

75 75

.

. .

79 79

1240 1240

.

~ ~

51

do-butylated do-butylated

1160

1111 1111

1 1

. .

I I

. .

i i

\ \

1093.63 1093.63

l~j l~j

showing showing

. .

936.71 936.71

spectrum spectrum

927.65 927.65

920 920

mass mass

.~-~ .~-~

58 58

56 56

.

.

respectively

815.66 815.66

744.64 744.64

790

797

180870 180870

tear tear

/ /

eye eye

MALDI-TOF MALDI-TOF

5\ 5\

.

A A

closed closed

726

6.3. 6.3.

,------,------,------,------,-----~------,------~-----,-----~-----~ ,------,------,------,------,-----~------,------~-----,-----~-----~

600 600

and and

0

n n

100 100

e

>, >,

ea ea > >

Q) Q)

C: C:

Q) Q) Ill Ill C: C:

-

Q) Q)

a: a:

~ ~

0 0

:;: :;:

~ ~

-

p

o Figure Figure 202

protein.

glutamic

Da).

1

' '

esterified

proline-rich

1,713.0

Y12

contains an

oflacrimal

([M+Ht

also 1

120)

Y11

tear

-

and

.

eye

_

(107

Y10

.

open

esterified

I

in

_

yg

peptide

11,

I

detected

tryptic

a

Ya

I

C-terminally

as 11

I

is

peptide

1ll,1

I

I

mlz

1

I

peptide

Y1

tryptic

identified

I

,I

11

The

I

esterified

peptide was peptide

an

800

1.Ul

I

Ys

for

This

ill.1.111

R*

Ys

1i111,I

spectrum

FPSVSLQEASSFFR.

detected.

I,

is

ion

was

A S S F F

E*

ions

peptide

Q

fragment

the

a

ofy-

of

of

series

sequence

F PS V S L example

An

acid

complete

6.4.

residue.

amino

almost

100

C ea g C

::,

G> i

-;ft.

c(

.a - 'g

i - a:

Figure

An

acid

The 6.3.3. Relative quantification of proteins in open and closed eye tear fluid

Following esterification and identification, combined aliquots of the do- and d10- butylated peptides were analysed by capillary ESI-LC/MS. Co-eluting peptides exhibiting an expected mass shift were identified and their relative amounts determined by comparing their respective peak areas obtained from each extracted ion chromatogram. The observed mass difference between each corresponding peptide pair is dependant on the number ofbutanol esters incorporated, as well as the charge state of the peptides.

Over 100 co-eluting peptide pairs were detected (SIN> 3) representing more than 20 different proteins in the combined tear fluid. This enabled multiple peptide pairs derived

from the same protein in tear fluid to be used for quantification. Figure 6.5 shows an

example of an averaged mass spectrum showing at least three different co-eluting peptide pairs (retention time of 60 - 62 minutes). The corresponding extracted ion

chromatogram for one of these peptide pairs is shown in Figure 6.6 (p 206). By

comparing the area under each peak, this peptide (identified as tryptic peptide 120- 125

from lysozyme) was estimated to be five times more abundant in open eye tear.

203 Figure 6.5. An example of a mass spectrum (m/z 600 - 1,300) of esterified open and

closed eye tear showing at least three different co-eluting tryptic peptide pairs.

These peptides eluted over a retention time window of 60 - 62 minutes and were

derived from lysozyme (m/z 797.5 and 815.6, residues 120-125; mlz 790.5 and 808.6, residues 19-23) and lactotransferrin (m/z 1,206.5 and 1,215.6, residues 110- 119).

The extracted ion chromatograms for one of these peptide pairs are shown in Figure 6.6

(p 206).

204 Figure 6.5. An example of a mass spectrum (m/z 600 - 1,300) of esterified tear samples (open and closed eye) showing at least three different co-eluting tryptic peptide pairs.

797.5

~ -0 Cl) 7~0.5 0 C: ea 'O C: :::J .0 < Cl) .:> 808.6 ea 'ii a: '815.6

1206.5 1215.6

600 700 800 900 1000 1100 1200 m/z

205 206

butyl

tes.

u

two

s

min

in

ta

62

*

n

-

R*

co

60

130

G I R

of

Q

peptide

P

P Q G I

dow

is

n

120

h

D*

D*

T

wi

.

e

m

ti

n

tio

DPQGIR

n

e

t

ce

e

n

r

100 110

e

a

t

qu

se

s a

e

ir

90

a

th

p

e

as

h

ptid

80

nd

e

p

e a

g

m

(min)

lutin

70

ysozy

m/z815.6

co-e

m/z797.5

Time

m l

e

h

t

60

fro

of

25

e

1

n

o

-

50

20

for

1

m

e

d

40

atogra

pepti

m

ro

h

30

c

tryptic

n

e

io

h

t

l

tear

as

20

tota

tear

eye

eye

10

identified

Closed

extracted

Open

B.

A.

was

The

0

6.6.

10

10

peptide

::::l

C1I

C: ::: 0

C:

C1I

Ill

Ill

~ -

"O <(

.D

~ Qi a:

. -

Figure

This

esters(*). Multiple peptides{> 3) derived from the same protein in each sample were used to determine the variation in the measured changes in the relative abundance for each protein. Table 6.2 shows an example of the peak area ratios obtained from multiple tryptic peptides of tear lipocalin. Listed are the tryptic peptides derived from this protein and the relative peak area ratio for each of these.

Some of the proteins found to be equally abundant in open and closed eye tear fluid include lactotransferrin, lacrimal proline-rich protein and various complement proteins

(e.g., C2, C4, C7). Table 6.3 (p 209) lists those proteins that were determined to be more abundant in open eye tear. Listed in Table 6.4 (p 210) are those proteins that were found to be more abundant in closed eye tear. These proteins were primarily serum­ based proteins.

207 Table 6.2. The relative abundances of tryptic peptides derived from tear lipocalin.

This protein was found to be 1.7 (± 0.4, CV 22 %) times more abundant in open eye tear.

TRYYfIC -,r OBSERVED m/z ¥ OBSERVED mlz CHARGE * OBSERVED RATIO PEPTIDE Open eye tear Closed eye tear STATE (open eye: closed eye) 95 - 101 823.4 841.5 +1 1.5: 1

84- 88 671.5 689.5 +1 1.9 : 1

89-94 814.5 832.6 +1 1.7: 1

102-108 927.6 936.6 +1 2.4: 1

156 - 166 649.4 658.3 +2 1.4: 1

1 Observed mlz for do-butyl ester.

¥ Observed mlz for d10-butyl ester.

• Average observed ratio of do- I d10- butyl esters.

208 Table 6.3. Proteins that were found to be more abundant in open eye tear in comparison to closed eye tear.

* OBSERVED RATIO PROTEIN (open eye: closed eye)

Lysozyme 5: 1

Tear lipocalin 1.7: 1

Transforming growth factor ~-2 1.5:1

Transcobalamin 1 2: 1

Fibroblast growth factor 1 1.5 : 1

Hepatocyte growth factor 1.5:1

Catalase 5:1

Prolactin inducible protein 1.5:1

• Average observed ratio of do-/ d1 0- butyl esters.

209 Table 6.4. Proteins that were found to be more abundant in closed eye tear in comparison to open eye tear.

* OBSERVED RATIO PROTEIN (open eye: closed eye) Plasminogen 1 : 10

Serotransferrin 1 : 3

von Willebrand factor 1 : 10

Serum albumin 1 : 3

Apolipoprotein A 1: 2

Immunoglobulin A 1 : 3

Lymphotoxin a 1: 2

Interferon fi 1 : 3

• Average observed ratio of do-/ dw- butyl esters.

210 6.4. Discussion

Proteins in open and closed eye tear were identified and the relative abundances of some of these were determined following proteolytic digestion and esterification with either butanol or deuterium labeled butanol. Esterification results in an increase in mass of multiples of either 56 Da ( do-butanol) or 65 Da ( d 10-butanol) due to modification of the

C-terminus of each peptide, as well as aspartic acid and glutamic acid residues. The observed mass difference between each peptide pair is dependant on the charge state of the peptides, as well as the number of butyl groups incorporated in each peptide. The use ofbutanol and its deuterated isotopomer ensures a measurable mass difference between higher order charge states of the same peptide and controls for variations in physiochemical properties and ionisation efficiencies for each co-eluting peptide pair.

Current methods of protein quantification include the use of appropriate antibodies, autoradiography or densitometry and require complete separation of the proteins of interest, usually by chromatographic approaches or gel electrophoresis [Krull et al.,

1997; Weller, 2000]. The technique described here enables multiple proteins to be quantified in a single analysis and does not require prior separation of proteins.

Additionally, multiple peptides derived from the same protein can be used to measure

independently the relative abundance of each protein, and therefore improve the

accuracy and precision of the quantitative determinations.

In this study the accuracy and precision of the approach was determined using horse

apomyoglobin as the model protein. Following digestion and derivatisation with either

do- or d10- butanol, equal aliquots of each were combined. Analysis of the combined

sample by ESI-LC/MS revealed that the observed ratio (1.03) for this analysis was not

211 significantly different from the expected ratio (1.00). Furthermore, the co-efficient of variation for these determinations was found to be 18%. This indicates that this technique can be used to accurately measure changes in protein expression and enables modest changes (i.e., - 2 fold increase or decrease) in protein abundances to be assessed. Although the deuterium labeled peptides co-eluted at a slightly earlier retention time than the corresponding unlabeled peptide [Masters et al., 1988; Gygi et al., 1999; Goodlett et al., 2001], this did not affect the accuracy of the quantitative determination.

This technique was employed to measure the relative abundance of over 20 different proteins in tear fluid obtained from the open and closed eye states. At least three different peptides derived from the same parent protein were employed for each measurement. Several serum-based proteins were found to be elevated in closed eye

tear, and in comparison, various growth factors and lacrimal gland derived proteins

were found to be more abundant in open eye tear. These findings are supported by other

studies, where serum proteins such as albumin, vitronectin and fibronectin, have been

found to increase in abundance in closed eye tear [Sack et al., 1994; Fukuda et al.,

1996; Sack et al., 1997]. Other proteins such as lactotransferrin and lacrimal proline­

rich protein were equally abundant in both open and closed eye tear [Sack et al., 1992].

In contrast to other techniques, such as ICAT, that have been developed for the global

quantification of peptides and proteins, the approach described here is not selective for a

particular amino acid residue (i.e., cysteine) [Gygi et al., 1999]. Instead, this technique

involves a simple chemical reaction that is directed toward acidic functional groups

(i.e., C-terminus of all peptides). As a result, all proteins present in a sample can be

212 quantified and multiple peptides derived from each can then be used to determine the relative changes in protein expression. However, because the ICAT reagent incorporates a biotin affinity tag, labeled peptides can be selectively isolated using avidin affinity chromatography. By reducing the complexity of the sample, quantification of proteins present at low abundance can be facilitated. For instance, in this study only a limited number of proteins in tear fluid could be detected and their relative quantities determined. Also of consideration is the dynamic range of this quantification technique.

This is dependant on the complexity of the sample as well as the instrument employed.

Although an ion trap mass spectrometer offers limited dynamic range and precision in comparison with other mass spectrometers, the dynamic range can be improved by reducing sample complexity.

Protein quantification techniques that are based on relative levels of proteolytic peptides, such as quantification using butyl esterification, are not able to distinguish between different in vivo forms of the intact protein, and therefore can be misleading.

However, the technique described here can be applied to quantify any pair of separated proteins, for example, in comparative or functional proteomic studies where protein

isoforms and modified proteins can be separated by 2DE. This technique is highly

versatile and can be employed to measure the relative amounts of proteins in a variety

of biological systems.

213 CHAPTER 7

DISCUSSION

214 7 .1. Discussion

Over the last decade, mass spectrometry has emerged as a powerful tool for protein and peptide analysis. Both MALDI-TOFMS and ESI-MS have matured to the point where they offer the sensitivity and specificity required to detect peptides and proteins in complex biological samples and these complementary approaches are now considered the primary tools for peptide and protein characterisation. The ability to obtain data such as peptide mass fingerprints and/or partial amino acid sequence on trace quantities of a protein has proved invaluable in the area of protein analysis. Further, extensive protein databases and database search algorithms that utilise the mass spectrometric data can now unambiguously identify proteins on a global scale. This allows the discovery of potentially important protein candidates that may be relevant to a particular biological process or disease.

These developments, coupled with the ready availability of commercial mass spectrometers, have enabled protein identification to become routine in many laboratory settings. However, the dynamic nature of the protein complement of a cell still poses significant challenges. The techniques to detect, characterise and quantify biologically relevant proteins that are differentially expressed or post-translationally modified are not readily available, and biochemical methods remain essential to determine the

significance of a particular protein in a biological or disease process, or its potential

impact on therapeutic strategies.

Proteomics has emerged as an important technology for protein characterisation and

involves the use of integrated protein separation and identification strategies. The comer

stones are gel electrophoresis and mass spectrometry, but there are continual

215 refinements and concurrent development of alternative approaches. For instance, comparative proteomics, which involves the analysis of protein expression between two

(or more) different states of a biological system, shows considerable promise for the identification of biomarkers of disease, and to determine potentially important proteins involved in specific cellular functions. This approach requires complete resolution of protein isoforms, unambiguous identification of each protein and its modification, as well as an accurate measurement of the changes in the expression of each protein.

Before studies such as these become routine, the limitations of the current strategies must be addressed, or alternative technologies considered.

7 .2. Protein identification strategie~

To completely describe a proteome, practical integrated strategies to separate, detect

and identify peptide and protein constituents are required. Unambiguous identification

of multiple proteins represents the initial step in elucidating their potential biological

role, and there have been many different strategies described to undertake this process

[Shevchenko et al., 1996; Yates et al., 1997; Link et al., 1999; Patton, 1999]. Although

these strategies employ a variety of separation procedures, such as electrophoretic or

chromatographic techniques, mass spectrometry is now common to the protein

identification process. Prior to mass spectrometry, proteins were identified using Edman

degradation or by amino acid analysis but these techniques neccesitated high sample

purity and homogeneity to be successful. Mass spectrometry, in particular MALDI and

ESI mass spectrometry, provides the sensitivity and certainty necessary to

unequivocally identify a protein in biological samples without these restrictions.

Proteins can be identified following enzymatic or chemical degradation to generate 216 unique peptides that can then be mass measured by MALDI-TOFMS (i.e., a peptide mass fingerprint or peptide mass map) [Henzel et al., 1993; J atnes et al., 1993; Mann et al., 1993; Pappin et al., 1993; Yates et al., 1993; Cottrell, 1994], or the atnino acid sequence of one or more of the proteolytic peptides can be obtained using tandem mass spectrometry [Mann and Wilm, 1994; Yates et al., 1995]. Protein identifications are then achieved by searching these data directly against the publicly available protein or genomic databases.

Although many protein detection and identification strategies have been described, most of these are not suitable for the analysis of endogenous levels of free peptides. These peptides are often not retained by gel electrophoresis, or ifESI-LC/MS/MS is

employed, these endogenous peptides are not able to be distinguished from those that were generated by the initial proteolytic step. Peptides, such as neuropeptides and

growth factors are usually secreted into the general circulation and interact with receptors, usually in an endocrine or paracrine manner, to regulate many physiological

processes [Verhaert et al., 2001]. As a result, strategies that enable facile analysis of

peptides are important in monitoring the function of biological systems. By directly

analysing biological tissues or fluids by MALDI-TOFMS, the accurate mass of

endogenous peptides can be determined, and mnino acid sequence information can be

obtained by MALDI-PSD, by another form of tandem mass spectrometry or by de novo

sequencing.

217 7.2.1. Two dimensional gel electrophoresis and peptide mass f"mgerprinting

The current accepted approach for protein identification involves separation by gel electrophoresis, in particular two dimensional gel electrophoresis (2DE), followed by peptide mass fingerprinting and database searching. This strategy, first described in

1993 [Henzel et al., 1993; James et al., 1993; Mann et al., 1993; Pappin et al., 1993;

Yates et al., 1993 ], involves generating a set of peptide masses by proteolytic digestion and MALDI-TOFMS, then matching these experimentally derived peptide masses with those calculated from in silico digestion of all the proteins in the database. For a positive identification, it has been suggested that a minimum of 4 peptides accurate to within 10 ppm of their theoretically calculated mass is required [Clauser et al., 1999]. This criterion is easily achieved with current MALDI-TOFMS technology. In addition to this, sequence tags derived from tandem mass spectrometry can also be used to confirm an identity [Mann and Wilm, 1994; Wilkins et al., 1998]. Importantly, as it stands, no single strategy is capable of delivering all the data that is necessary to determine the relevant information in a proteomics study.

As a part of this study, two dimensional gel electrophoresis combined with peptide mass fingerprinting was employed as a first step to detect and unambiguously identify proteins in human seminal fluid. Separation by 2DE allowed detection of more than 300 discrete protein spots, however, subsequent analysis by MALDI-TOFMS identified only

32 unique proteins. The remainder of the proteins detected were found to be post­ translationally modified forms of these 32 proteins, e.g., multiple forms of prostate

specific antigen and prostatic acid phosphatase were detected. Additionally, numerous

truncated forms of serum albumin, semenogelin I and semenogelin II were identified.

These :findings indicate that there is extensive proteolytic activity in seminal fluid, and

218 this is supported by the numerous enzymes that were also identified. The detection of multiple forms of any single protein would not be possible using any of the current alternative protein identification strategies, such as ESI-LC/MS/MS analysis of proteolytically derived peptides.

Although one dimensional gel electrophoresis in combination with capillary ESI­

LC/MS/MS identified over 100 different proteins in seminal fluid, some modified proteins could not be detected or identified in this way. By contrast, high mass proteins

and basic proteins that are not amenable to 2DE separations, such as fibronectin and histone proteins, were identified by this technique. This approach also identified many more proteins than 2DE and these data highlight some of the problems associated with the 2DE process. For example, some protein classes are known to stain poorly with

Coomassie or silver staining techniques and this reduces their ability to be detected

[Rabilloud, 2000]. Additionally, some proteins are present at low abundance and are not readily detectable by 2DE; identification of other protein spots is confounded by the presence of comigrating proteins. For instance, at least 7 excised spots were found to

contain more than one protein when seminal fluid was separated by 2DE (see Tables 4.2

and 4.4, p 124 and 129 respectively). Consistent with this finding, the presence of at

least 6 unique proteins has been reported within one discrete protein spot [Gygi et al.,

2000].

Therefore, improvements in current 2DE technology are required to address issues such

as difficulties in detecting low abundant proteins, the co-migration of proteins, and the

inability to adequately resolve high mass proteins, hydrophobic proteins and basic

proteins. Solutions are being continuously described in the literature, including sample

219 pre-fractionation strategies and the application of narrow range immobilised pH gradient strips that improve protein separation and enable detection of low abundant proteins [Zuo and Speicher, 2000; Zuo et al., 2001]. Also, sensitive fluorescent protein stains, such as Sypro Red and Sypro Orange, capable of detecting low femtomoles of a protein, have also been described [Steinberg et al., 1996a; Steinberg et al., 1996b;

Harvey et al., 1998; Berggren et al., 2000; Guttman et al., 2000; Lopez et al., 2000;

Patton, 2000; Steinberg et al., 2000a; Steinberg et al., 2000b; Valdes et al., 2000; Yan et al., 2000; Lauber et al., 2001; Tonge et al., 2001]. Although the exact nature of the interaction between proteins and these stains has not been defined, these fluorescent stains are believed to bind noncovalently with proteins. In contrast to conventional silver and Coomassie blue staining, the staining properties of fluorescent dyes are not dependant on any specific chemical interaction with amino acid residues [Rabilloud,

2000; Yan et al., 2000]. As a result, protein detection using fluorescent stains is less dependant on amino acid sequence. These stains also have the added advantage of being able to detect proteins over a wide linear dynamic range, thereby facilitating quantitative protein analysis [Lopez et al., 2000; Yan et al., 2000].

7.2.2. Direct analysis of proteins by ESI-LC/MS/MS

To overcome the problems associated with 2DE some investigators have adopted an

approach involving direct analysis of proteolytically derived peptides following

chromatographic separation [McCormack et al., 1997; Yates et al., 1997; Link et al.,

1999; Yates et al., 2000; Davis et al., 2001; Washburn et al., 2001]. In contrast to 2DE

and peptide mass fingerprinting, separation of proteins prior to analysis is not required

and amino acid sequence data for each peptide can be obtained by ESI tandem mass

220 spectrometry. These data are then compared with the available protein and genomic databases. This technique involves minimal sample preparation and because of the increased information content of sequence data, the probability of detecting and identifying proteins present at low abundance is enhanced. Also, by combining different chromatographic separations in series, such as ion exchange chromatography followed by reverse phase chromatography, a greater number of peptides can be detected than with a single stage of chromatography. This two dimensional chromatography mass spectrometric approach for protein identification has been termed MuDPIT [Yates et al.,

1997; Link et al., 1999]. This technique enables proteins that are not amenable to electrophoretic separation, such as hydrophobic proteins or high mass proteins, to be identified. However, because this technique only requires the sequence ofup to 6 amino acids for a positive protein identification [Wilkins et al., 1998], it does not routinely deliver high sequence coverage of the protein, nor are post-translational modifications usually identified.

In the work detailed in this thesis, LC/MS/MS of proteolytically generated peptides was employed to identify proteins in the human tear film. An aliquot of tear fluid was digested with trypsin and the proteins identified by capillary ESI-LC/MS/MS and database searching. This approach enabled more than 400 different proteins to be unambiguously identified, including known constituents of human tear such as tear

lipocalin, lactotransferrin, lysozyme and secretory lgA. Proteins that have not been previously detected in tear fluid were also detected and identified. The tear fluid is also

reported to contain a large number of modified proteins, such as glycoproteins,

truncated proteins and N-terminally processed proteins as well as isoforms of proteins

such as lactotransferrin and Zn-a-2-glycoprotein [Coyle et al., 1989; Fullard and

221 Kissner, 1991; Kuizenga et al., 1991; Molloy et al., 1997; Jumblatt et al., 1999;

McKenzie et al., 2000]. Although numerous proteins were identified, modified proteins

and protein isoforms could not be distinguished.

This study has also demonstrated that LC/MS/MS is a rapid approach for the

identification of proteins in complex biological samples where tandem mass spectra

only are obtained on one or two peptide(s) derived from the precursor protein.

Additionally, this approach may be superior to 2DE in some respects, especially because peptide mass fingerprinting does not always yield a positive protein

identification due to the presence of contaminants or peptides derived from more than one protein.

7.2.3. Detection and identification of endogenous peptides in biological samples

The techniques described above are able to identify proteins with high confidence;

however, they are not compatible for the analysis of endogenous peptide components.

Peptide components in biological fluids or tissues can only be detected by direct

analysis of the sample employing either MALDI-TOFMS or ESI-MS. This allows the

accurate masses of the endogenous peptide components to be determined, and post­

source decay (PSD) or tandem mass spectrometry can also be employed to obtain

information pertaining to the amino acid sequence of each individual peptide [Jespersen

et al., 1999; V erhaert et al., 2001].

In this thesis, the direct analysis of human tear fluid by MALDI-TOFMS enabled the

intact mass of numerous abundant peptide and protein components to be accurately

222 determined. In addition, PSD analysis allowed the sequence of five of the more abundant peptides to be elucidated. Further investigations revealed that these peptides were derived from the C-terminus of a lacrimal proline-rich protein that had not previously been detected in human tear fluid [Dickinson and Thiesse, 1995].

Subsequent studies employing size exclusion chromatography and MALDI-TOFMS analysis confirmed the presence and identity of a truncated form of this protein in tear fluid. This demonstrates the potential ofMALDI-TOFMS in identifying the circulating form of peptides and proteins in biological fluids, especially because peptides and low mass proteins are often not detected by standard gel electrophoretic techniques.

A combination ofMALDI-TOFMS and capillary ESI-LC/MS/MS was also employed to detect and identify the endogenous peptide constituents in human seminal fluid.

MALDI-TOFMS enabled the accurate intact mass of peptides to be determined and

ESI-LC/MS/MS enabled the identity of these peptides to be ascertained. Multiple peptide fragments of semenogelin I and semenogelin II were detected, including

seminal basic protein, a bioactive peptide derived from semenogelin I [Robert and

Gagnon, 1999]. Human seminal fluid is known to contain many proteolytic enzymes,

and the presence of these peptides is indicative of the high level of enzymatic activity.

In this case, the ability to detect peptide components may prove valuable in deciphering

the biological role of many of the protein constituents. For instance, semenogelin I and

II are known to be the major components of the seminal coagulum which is rapidly

degraded by proteases upon ejaculation [Malm et al., 1996; Peter et al., 1998; Robert

and Gagnon, 1999]. The presence of these peptide fragments, and the observation that

the terminal amino acid residues of these peptides were consistent with cleavage by

223 prostate specific antigen supports their putative role in sperm motility [Robert and

Gagnon, 1999].

7.3. Characterising protein post-translational modifications

Although the human genome has been reported to encode for approximately 30,000 unique proteins, it is widely believed that many more proteins exist in vivo [Kellner,

2000]. This discrepancy is due to protein modifications, and the synthesis of protein variants and isoforms as a result of gene splicing and frameshift mutations.

Modifications such as these can not be predicted from DNA sequences and can only be determined using proteomic strategies. There are many documented protein modifications that may occur in the cell. These include oxidation, amino acid

substitution, acetylation, acylation, amidation, phosphorylation and glycosylation.

Proteins often require specific modifications to prolong their stability or halflife, to determine their subcellular localisation or to effect specific biological function (e.g.,

cleavage of a prepro-protein precursor to its bioactive form). In addition, some of these modifications, such as the phosphorylation of proteins involved in signal transduction

pathways, are often reversible. Hence, the ability to isolate and detect the modified

protein(s) of interest, as well as to identify the type and site of modification, will aid our

understanding of many biological processes.

224 7.3.1. Detection and isolation of modified proteins

The separation of seminal fluid proteins by 2DE identified multiple forms of many proteins. For example, multiple forms of serum albumin, prostate specific antigen, lactotransferrin and prostatic acid phosphatase were detected and these modified proteins were found to migrate at a different isoelectric point and/or molecular weight when compared to their native form, i.e., when compared to their theoretical isoelectric point and mass (Chapter 4, Figures 4.1 - 4.2 and Tables 4.1 - 4.4, p 116 - 129).

Although 2DE remains the most effective biochemical method for the separation of complex protein mixtures and enables visualisation of modified proteins, it only provides the approximate molecular weight for each protein [Klose and Kobalz, 1995;

Lopez, 1999]. Characterisation of post-translational modifications can be aided by obtaining the accurate molecular weight of the intact protein prior to proteolysis.

Mass spectrometry is a sensitive technique for accurately determining the molecular weights of proteins. For instance, MALDI and ESI enable large non volatile molecules to be ionised and mass analysers, such as the time-of-flight mass analyser, offer superior resolution and mass accuracy than gel electrophoresis. The interfacing ofMALDI or

ESI with the appropriate mass analyser allows modified proteins to be identified by the

distinct mass shift from its predicted mass. For example, phosphorylation would result

in a mass increase of 80 Da, and acetylation results in a mass increase of 42 Da.

However, small mass differences, such as those that occur as result of amidation

(increase in 1 Da) or amino acid substitutions (e.g., a substitution of glutamine for

glutamic acid results in a mass increase of 1 Da) are not easily measurable by mass

spectrometric analysis of intact proteins. However, these modifications often result in pi

changes allowing detection and identification by 2DE [Larsen and Roepstorff, 2000].

225 Despite the popularity and widespread use of 2DE, there are many difficulties associated with improving the compatibility of electrophoretically separated proteins with direct analysis by mass spectrometry. The efficiency with which intact proteins can be extracted from gel pieces, whether by electroelution or by chemical means, is low

[Eckerskom et al., 1997; Mirza et al., 2000], and direct MALDI analysis of gel pieces often does not yield the sensitivity or mass resolution required to confidently assign a mass and hence identify a particular type of modification [Ogorzalek Loo et al., 1997].

To overcome these problems, alternative methods for protein separation and isolation that are more compatible with mass spectrometric analyses are therefore required.

Alternative methods for protein purification and isolation include various chromatographic techniques (e.g., reverse phase, affinity chromatography, ion exchange

chromatography). There have been several reports of coupling on-line chromatographic

separation, in particular reverse phase chromatography, to mass spectrometry for the

analysis of whole proteins. For instance, Opiteck et al. (1997) have developed a technique that utilises on-line chromatographic separation interfaced to a quadrupole

mass spectrometer for the direct analysis of complex protein mixtures. Alternatively,

there have been reports of successful direct deposition of chromatographic eluent on to

a MALDI target for subsequent analysis, thereby eliminating the need for fraction

collection [Zhang and Caprioli, 1996; Miliotis et al., 2000]. Although this has only been

demonstrated for low mass molecules such as peptides, it can potentially be adapted for

the separation of complex protein mixtures. In addition to chromatographic analysis,

other investigators have attempted to directly analyse intact proteins by MALDI-

226 TOFMS following separation by solution phase isoelectric focusing although this approach still requires manual :fraction collection [Loo et al., 1999; Walker et al., 2001].

7.3.2. Identifying post-translational modifications

Although the identity of modified proteins can be achieved using standard proteomic techniques, these do not adequately address the difficulties associated with the

characterisation of extensively modified proteins. For example, peptide mass

fingerprinting (or mapping) involves the matching of a set of proteolytically derived peptide masses to those derived from known proteins entered into a specific database.

The available protein databases contain the primary amino acid sequence of genetically

encoded proteins and do not include information concerning potential modifications.

Therefore, if a protein is extensively modified, the resulting set of proteolytic peptide(s)

containing the modification(s) will not match the theoretically calculated peptide mass(es ), thereby hindering identification of the protein [Larsen and Roepstorff, 2000].

High sequence coverage of a protein is also required before a modification can be

confidently assigned, and although this is best achieved by peptide mass fingerprinting,

it is often hindered by the reduced ionisation efficiencies of some proteolytic peptides

and signal suppression effects that occur during the MALDI ionisation process. As a

result, modifications can often only be localised to a small segment of the protein.

However, modifications can still be identified by comparing the experimentally

determined peptide masses with those predicted from the database, and the modification

predicted from the shift in mass from its theoretical value.

227 Alternatively, the nature and site of modification can be established with peptide sequence information derived from either MALDI-PSD or ESI tandem mass spectrometry. This strategy has been most successfully employed to identify sites of protein phosphorylation and glycosylation [Larsen and Roepstorff, 2000; Yanagida et al., 2000; Oda et al., 2001]. Nevertheless, the specific site of modification can be difficult to determine, especially because tandem mass spectrometry rarely yields the full sequence of a peptide. Both computer search algorithms and de novo sequencing methods are usually required for accurate interpretation of the mass spectral data and confirmation of the specific site of modification. In some instances, the site of modification can be localised to a particular amino acid residue (e.g., glycosylation at threonine, serine or residues, amidation at acidic residues, or N-terminal

acetylation) simplifying interpretation of the product ion spectrum.

Both MALDI-TOFMS and ESI-MS are valuable for the detection and identification of modified proteins. For example, in this thesis, MALDI-TOFMS was used to detect and

identify a truncated form oflacrimal proline-rich protein in human tear fluid following

its isolation by size exclusion chromatography. The intact mass of the protein was

determined by MALDI-TOFMS and then ESI-LC/MS/MS was employed to confirm the

identity of the protein following digestion with multiple enzymes (trypsin and Asp-N).

This strategy established that the mass of the secreted form of this protein (11,733 Da)

was consistent with residues 18- 121 of the predicted protein (from the DNA sequence)

and that the 13 C-terminal residues were cleaved in vivo to form a series of homologous

peptides. Proteolysis of the isolated protein with multiple enzymes confirmed the

identity of the protein and enabled more than 50% sequence coverage of the protein

(See Figure 3.5, p 79). The truncated form of the lacrimal proline-rich protein could not

228 be detected when tear fluid was proteolytically digested and directly analysed by ESI­

LC/MS/MS. Only two peptides corresponding to residues 82-97 and 127-134 of the full length protein were detected by this approach.

The trend toward lower flow liquid chromatography (flow rates of30 nL/min have been reported [Wilm and Mann, 1996]) is rapidly establishing ESI-MS as the tool of choice, especially when this ionisation technique is coupled with hybrid mass analysers such as the quadrupole time-of-flight: low flow liquid chromatography allows both separation of individual peptides and high sensitivity. Further, high mass accuracy can be obtained with a quadrupole time-of-flight mass analyser [Morris et al., 1997; Borchers et al.,

2000; Kristensen et al., 2000; Ma et al., 2001; Macek et al., 2001; Steen et al., 2001].

The ability to obtain the sensitivity and mass accuracy that these instruments can provide is essential in the area of functional or comparative proteomics where modified proteins may be important determinants of cellular function.

7.4. Protein quantification

Another important requirement of a protein analysis strategy is the ability to accurately

quantify the protein(s) of interest; or at least to be able to measure changes in protein

expression in response to various stimuli. Quantification by mass spectrometry is

capable of offering a level of sensitivity and specificity not available by other

commonly employed protein quantification methods such as chromatographic,

immunoaffinity or immunochemical techniques, but this area is not well developed at

this time. Because there is a poor correlation between measured mRNA levels and

protein expression in the cell [Anderson and Seilhamer, 1997; Gygi et al., 1999b],

229 protein quantification is particularly important. In addition, quantification is essential in areas where proteomics is applied to the search for biomarkers or indices of therapeutic efficacy, or when alterations in the level of expression of modified proteins may result in perturbations in cellular function.

7 .4.1. Relative quantification of proteins

There are examples describing the measurement of the relative expression levels of individual proteins by densitometry following 2DE separation [Newsholme et al., 2000;

Poirier et al., 2001; Yoo et al., 2001]. Although it maybe possible to estimate the relative abundance of a protein in this manner, the reproducibility, dynamic range, accuracy and precision with which this determination can be made is less than ideal

[Tonge et al., 2001]. These studies are highly dependant on gel to gel reproducibility

and the accuracy of gel comparison and matching software algorithms available. These methods also assume efficient staining of all proteins present in the sample, i.e., that all proteins have the same chemical response to the chosen stain [Pleissner et al., 1999;

Kriegel et al., 2000; Voss and Haberl, 2000]. A recent study has highlighted the

limitations of gel to gel reproducibility with respect to protein spot pattern and spot

density and the consequences for protein quantification [Voss and Haberl, 2000].

Mass spectrometric techniques that measure the relative abundance of proteins by

differential mass tagging of proteolytically derived peptides have been described

[Schnolzer et al., 1996; Gygi et al., 1999a; Ji et al., 2000; Munchbach et al., 2000;

Goodlett et al., 2001; Wang et al., 2001; Yang et al., 2001; Wang et al., 2002]. These

strategies can be employed to determine the differences in the relative expression of

230 proteins on a global scale, or they can be applied to specific proteins following their separation by 2DE. These methods, however, are only capable of measuring modest changes in protein expression and require efficient labeling of all peptides. The quantification of modified proteins presents additional analytical challenges because these proteins are often present at low abundances and modifications may be transient in nature.

One study described in this thesis involved the development of a technique that allows the relative abundance of multiple proteins in a sample to be determined. Proteins in open and closed eye tear fluid were reacted with either do-butanol or d10-butanol resulting in esterification of the C-terminal residues of each peptide as well as aspartic acid and glutamic acid residues. The relative amounts of more than 20 proteins in open and closed eye tear fluid were measured using this approach, and multiple peptides from each protein were employed in the analysis. In contrast to the ICAT approach, this technique allows all peptides in the mixture to be quantified because it does not target a

single amino acid residue, e.g., cysteine. Because all peptides can be used for quantification, this approach has the potential to improve the accuracy with which

changes in protein expression can be measured. Although this approach is able to

accurately measure changes in protein expression, subtle changes are not able to be precisely determined (i.e., less than 2 fold differences). Approaches such as this,

however, presents an alternative for the quantification of biologically important proteins

that have been previously isolated using standard chromatographic or electrophoretic

approaches.

231 7 .4.2. Quantification of intact proteins

Most of the approaches available for determining the relative level of proteins in a sample involve derivatisation of proteolytic peptides and subsequent detection by ESI or

MALDI mass spectrometry [Schnolzer et al., 1996; Gygi et al., 1999a; Oda et al., 1999;

Ji et al., 2000; Munchbach et al., 2000; Conrads et al., 2001; Goodlett et al., 2001;

Smith et al., 2001; Wang et al., 2001; Yao et al., 2001; Wang et al., 2002]. There is currently no established inass spectrometric technique available to measure the relative or absolute levels of intact proteins.

MALDI-TOFMS has been well established as a sensitive approach for qualitative analysis of peptides and proteins. In addition to rapid analysis times and high mass accuracy, analysis of peptides and proteins by MALDI-TOFMS primarily yields the singly protonated molecule simplifying interpretation of the data. In this thesis the absolute quantification of proteins and peptides has been examined by MALDI-TOFMS following their extraction from an appropriate biological matrix (i.e., rat growth hormone in pituitary gland, insulin in human pancreatic tissue and L VV-hemorphin-7 in adrenal gland extract). Linear calibration curves were obtained for all analyses and the values obtained by MALDI-TOFMS for each analyte were consistent with those reported by others [Dickerman et al., 1971; Cerpa-Poljak et al., 1997].

Because MALDI-TOFMS is able to measure the intact mass of proteins, it can also be

employed to determine the degree of modification of a chosen protein provided that the

two species, i.e., the modified and unmodified form, can be adequately resolved.

However, problems may arise due to the heterogeneity of the modification; for example,

a protein may contain multiple potential sites for phosphorylation and care must be

232 taken to ensure that only one modified species is being detected and quantified. To overcome these types of difficulties, adequate separation of the protein of interest is required.

Selection of the appropriate internal standard is also an important factor for the accurate

quantification of proteins by MALDI-TOFMS. The optimum standard is a homologous protein, e.g., obtained from another species, or from the same protein family.

Alternatively, the protein of interest can be purified following its expression in cell

culture maintained in isotopically enriched media. Internal standards such as these will minimise problems associated with different ionisation efficiencies and other physical

and chemical properties important in the isolation and purification steps.

Although it has been demonstrated that proteins can be accurately quantified by this

approach, this application ofMALDI-TOFMS requires additional development. As it

stands, it is limited by the dynamic range and the poor sensitivity for high mass

molecules. Issues relating to dynamic range and protein solubility can be overcome by

selecting the appropriate matrix molecule and solvents [Cohen and Chait, 1996],

especially because the number of potential organic molecules that can be employed as

suitable MALDI matrices is continuously expanding [Fitzgerald et al., 1993; Bai et al.,

1996; Krause et al., 1996; Zhu et al., 1996; Nonami et al., 1998; Ayorinde et al., 1999;

Vandell and Limbach, 1999].

MALDI-TOFMS offers many advantages for the quantitative analysis of proteins, such

as minimal sample preparation and fast analysis times, but it is not amenable to on-line

separation techniques. Therefore, proteins must still be purified and isolated off-line to

233 yield the most accurate and precise results. This relies on traditional protein purification techniques such as various chromatographic separations and/or immunoaffinity separations. For example, glycoproteins can be purified using lectin chromatography or affinity-based methods and phosphorylated peptides and proteins can be isolated by employing immobilised metal affinity chromatography [Kobata, 1994; Hage, 1999; Li and Dass, 1999; Posewitz and Tempst, 1999].

The use of microscale sample preparation methods such as on-target immunoaffinity capture of both analyte and internal standard has been described [Nelson et al., 1995;

Tubbs et al., 2001]. This approach involves modification of a MALO I target, e.g., by the incorporation of antibodies that are targeted toward the protein of interest. This requires low sample volume and potentially offers fast and specific analysis because extensive purification and concentration of the analyte is unnecessary. In addition, the mass accuracy offered by MALDI-TOFMS can be employed to increase selectivity. The ability ofMALDI-TOFMS to discriminate proteins based on mass becomes important when quantifying potential biomarkers, especially since antibodies are capable of recognising multiple epitopes and are not able to distinguish between multiple forms of the same protein that are present in vivo. By employing MALDI-TOFMS, only the protein of interest is quantified.

7 .5. Alternative technologies for proteomic studies

Current interest in proteomics is primarily concerned with the identification of proteins,

differential expression of proteins in tissues or fluids obtained from normal and disease

states, and in cell culture systems maintained under different conditions. Although mass

234 spectrometry has been established as the tool of choice for the detection and

identification of peptides and proteins, it is well recognised that current separation

techniques, such as 2DE or HPLC, are biased toward abundant proteins. As a result,

approaches to enrich for particular classes of proteins are being investigated and are

proposed to facilitate the detection of biologically important proteins that may provide

an insight into cellular function or disease mechanism. For example, tissue samples are

composed of many different cell types. Isolation and enrichment of specific cell populations from tissue samples by laser capture microdissection is valuable in

comparative proteomic studies where abnormal cells can be selectively separated from

surrounding non-affected cells. This enables potential biomarkers to be established and may also provide an insight into the possible role of proteins in the disease mechanism

[Banks et al., 1999; Simone et al., 2000; Bichsel et al., 2001; Paweletz et al., 2001].

Proteins from the isolated cell population can then be extracted and analysed by

common proteomic techniques.

Similarly, direct analysis of intact biological tissue by MALDI-TOFMS allows regional

mapping of the protein components of a tissue or cell. Proteins can be isolated to a

specific cellular compartment or to a particular tissue region and this technique also

enables the in vivo form of peptides and proteins to be determined [Caprioli et al., 1997;

Chaurand et al., 1999; Jespersen et al., 1999; Li et al., 1999; Stoeckli et al., 1999;

Stoeckli et al., 2001]. This approach also shows immense potential for detecting and

establishing possible biomarkers for the diagnosis of disease, for example, following

biopsy.

235 The development and integration of other technologies, such as microfluidics and micro fabrication technology, will overcome some of the limitations of current sample preparation techniques and may lead to improvements in the capabilities of mass spectrometry for protein analysis. At present, there is much interest in coupling microfluidic devices or protein chip arrays to mass spectrometry, primarily to MALDI and/or ESI [Figeys and Aebersold, 1997; Figeys et al., 1998a; Figeys et al., 1998b;

Figeys and Aebersold, 1999; Ekstrom et al., 2000; Pinto et al., 2000; Zhang et al., 2000;

Ekstrom et al., 2001; Figeys and Pinto, 2001]. These techniques are being developed to address issues in protein analysis relating to the automation of some aspects of the protein identification process such as sample preparation (e.g., sample purification and enrichment), enzymatic digestion of proteins, and peptide and/or protein separation prior to mass spectrometric analysis. Although the implementation of these devices is proposed to increase sample throughput and improve sensitivity [Figeys et al., 1998a;

Ekstrom et al., 2001], current concerns are focused on the practical aspects of sample preparation that will be needed to successfully utilise these devices, i.e., the amount of sample required and the final volume necessary for analysis.

236 7 .6. Conclusions

The ability to confidently identify proteins represents the initial stage in elucidating the biological function(s) of a protein, and hence its potential importance, in a chosen biological system. The primary structure of a protein is determined by the genetic code of an organism; however, additional information pertaining to secondary or tertiary structure, post-translational modifications, or interactions with other biological molecules must be investigated to understand the underlying mechanisms of a biological process or disease.

The major obstacles that exist in the field of proteomics pertain to protein quantification and establishing the nature and extent of protein modification. Currently, MALDI­

TOFMS and ESI-MS are central components in the development of strategies to overcome these difficulties. The integration of mass spectrometric techniques into established strategies will ensure progress in the ability to relate gene function to protein expression, to direct therapeutic strategies, and to provide an insight into the mechanisms associated with normal biological function or dysfunction. Even though there are many difficulties associated with the analysis of a dynamic biological system where protein expression is constantly changing in response to multiple stimuli, the

application of mass spectrometry will be pivotal in deciphering the interactions that

exist between these and other biomolecules.

237 7.7. Summary

Over the last decade, mass spectrometry has developed to enable the analysis of a wide range of biomolecules over an extensive mass range. In large part this is due to developments in instrumentation and ionization techniques such as electrospray ionization (ESI) and matrix-assisted laser desorption ionization (MALDI) that have enabled the analysis of high mass molecules such as peptides and proteins. Because of these advances, and coupled with the completion of the human genome project, interest in protein and peptide analysis has escalated as investigators attempt to identify potential new protein drug targets and to gain additional insight into disease mechanisms. As a result, there has been considerable interest in the development of methods to detect, identify and quantify peptides and proteins in biological samples.

This thesis focuses on the development of mass spectrometric-based proteomic methods and their potential application in the biological sciences. A variety of strategies were examined and compared, including gel electrophoresis, ESI-LCMSMS, MALDI­

TOFMS and peptide mass fingerprinting.

Proteins in human prostatic fluid were characterized using two dimensional gel electrophoresis (2DE) and peptide mass fingerprinting. Proteins were first separated by their isoelectric point (1 st dimension, pi), and are then by their molecular weight using

SOS-PAGE (2nd dimension). Two dimensional separation was performed over two

different pi ranges (pi 4-7 and 6-11 ), enabling better resolution of proteins, and

allowing more proteins to be detected and identified. Over 100 proteins were visualized

using this technique. Of these, 73 were positively identified by peptide mass

fingerprinting, and these represented 20 unique proteins. For example, multiple forms of

prostate specific antigen, prostatic acid phosphatase, clusterin and prolactin-induced

238 protein were all identified. These results highlight the primary strength of 2DE where protein isoforms and modified proteins can be readily detected. Due to some of the limitations of 2DE, such as co-migrating proteins and the inability to resolve basic proteins or high mass proteins, proteins in prostatic fluid were also separated by 1D gel electrophoresis, digested with trypsin and analysed by ESI-LCMSMS. This approach enabled more than 100 proteins to be identified, including proteins that had also been identified by 2DE. Multiple proteins were identified in each band that was excised.

Although high molecular weight proteins and basic proteins could be detected and identified, in contrast to 2DE, modified forms of proteins could not be detected by this approach. In addition to gel electrophoresis, an unfractionated sample was also analysed directly by MALDI-TOFMS and ESI-LCMSMS to identify the peptide components in seminal fluid. These peptides and small proteins, which included seminal basic peptide and peptide fragments of semenogelins I and II, were not detected or identified by either of the electrophoretic approaches.

In another study, the protein constituents of human tear were characterised by ESI­

LCMSMS and database searching following complete proteolysis of the sample with

trypsin. Analysis of proteolytic peptides by ESI-LCMSMS overcomes some of the

shortcomings inherent to 2DE, such as protein solubility and losses due to sample

handling, and as a result may allow less abundant proteins to be detected and identified.

This approach enabled more than 100 proteins to be identified in less than 10 µL of tear

fluid, including many novel proteins that have not been identified in human tear before.

Some of the proteins identified include lysozyme, prolactin-induced protein, slgA, tear

lipocalin, lactoferrin, complement proteins, serum albumin and various growth factors

such as EGF and bFGF. Multiple peptides derived from the same protein were required

239 to identify each protein. Although this technique is able to identify many proteins in a single analysis, it is not able to detect or identify proteins that have been modified. This technique also results in low sequence coverage when compared to the 2DE approach.

In addition, human tear was analysed directly by MALDI-TOFMS. As a consequence, the 5 most abundant peptides in tear, which had not previously been described, were sequenced by post-source decay (PSD). These peptides were identified as a homologous family corresponding to the C-terminal portion of a novel lacrimal proline-rich protein.

Further fractionation and purification of tear enabled detection of the remaining N­ terminal portion of the lacrimal proline-rich protein. The intact mass of the in vivo form of this protein was accurately determined to be 11,731 Da (in comparison to the predicted 13,478 Da), consistent with cleavage of the 13 C-terminal residues to form the peptides found in tear. Although this protein displays some homology with a salivary proline-rich protein, the biological function and significance of these proteins is not known. This particular study demonstrated that although ESI-LCMSMS is able to identify many proteins in a small volume of sample, it is riot able to accurately identify the circulating in vivo form(s) of proteins.

The study of human tear was extended further to explore a method to determine the

relative quantities of proteins in biological samples on a global scale. This method

involved the esterification of acidic functional groups with either do- or d10-butanol

under anhydrous conditions following complete proteolysis of the sample. In contrast to

other protein quantification techniques that utilize differential mass tags ( e.g., ICAT),

the method described here is not selective for any specific amino acid residue and

therefore allows all peptides to be used for quantification. This method, however, also

240 assumes both complete proteolysis and derivatisation of proteins in the sample. The accuracy and precision of the technique was determined by combining equal amounts of a standard protein, horse apomyoglobin, following proteolysis and esterification (1.03 ±

0.19, CV 20%).

By adopting this method, protein expression in normal open eye tear fluid was compared to proteins in tear collected following extended eye closure. Although the protein constituents of open and closed eye tear are the same, it is believed that during prolonged periods of eye closure ( e.g., during sleep) the protein constituents of tear fluid change to reflect an induced state of inflammation resulting from the static nature of the tear fluid during this time. Tear fluid functions to lubricate and supply nutrients to the corneal surface, and to remove cellular debris and waste products. During periods of eye closure, the blink action no longer serves to encourage the continuous flow of tear fluid over the anterior eye surface.

In this study, proteins in open eye tear fluid were esterifed with do-butanol and the proteins for closed eye tear esterified with isotopically labeled d10-butanol. This resulted in a mass difference of 9 Da between corresponding peptides derived from each sample.

Following esterification, the samples were combined and analysed by ESI-LCMSMS to

determine the relative abundance of each protein in the sample. The peak area ratios of

multiple peptide pairs derived the same protein were used to determine the relative

abundance of each protein in each sample. Proteins that were found to be equally

abundant in open and closed eye tear include various complement proteins, lactoferrin

and lacrimal proline-rich protein. Proteins found to increase in abundance in open eye

tear fluid include tear lipocalin and lysozyme. In contrast, proteins such as plasminogen,

241 von Willebrand factor and slgA were found to be up to 10 times more abundant in closed eye tear. Co-efficients of variation for all determinations were less than 20%.

This thesis also explores the potential of MALDI-TOFMS for the absolute

quantification of biomolecules, including urinary metabolites, peptides and proteins.

MALDI-TOFMS is used extensively for qualitative analysis, such as DNA analysis and peptide and protein identification; however, its use for quantitative analysis is limited,

especially in a biological setting. Although this thesis demonstrated that MALDI­

TOFMS can be used successfully for the absolute quantification of a variety of molecules in a biological setting, some limitations still exist, especially for the

quantification oflarge biomolecules such as proteins. For instance, low sensitivity for high molecular weight proteins, a limited dynamic range and low precision need to be

overcome before it becomes a routine procedure. Despite these limitations, MALDI­

TOFMS can be used to accurately quantify biomolecules over an extensive mass range.

Homovanillic acid was quantified in human urine, and epinephrine and norepinephrine

quantified in adrenal tissue by employing stable isotope labeled internal standards. The

peptides LVV-hemorphin- 7 and insulin were quantified in human adrenal gland extract

and human pancreatic tissue respectively by employing structural homologues as

internal standards (i.e., rat LVV-hemorphin- 7 and porcine insulin respectively). In

addition, growth hormone was quantified in rat pituitary gland by incorporating P­

lactoglobulin as the internal standard. This study revealed that MALDI-TOFMS is a

good quantitative tool provided that the appropriate internal standard is used. For

instance, correlation coefficients (r2) greater than 0.99 were routinely achieved with

each chosen analyte and coefficients of variation (CV) were less than 10%, with the

242 exception of growth hormone where the CV was approximately 20%. The accuracy of the overall technique was determined by independent analysis of the same samples by a commercial laboratory ( e.g., in the case with homovanillic acid), by comparison with a different technique (LVV-hemorphin- 7) or by comparison with literature values

(insulin, epinephrine, norepinephrine and growth hormone).

This thesis assesses the most common methods for protein identification and highlights the strengths and weaknesses of each. Overall, it was determined that more than one

strategy is required to comprehensively identify all proteins in a complex biological

sample. 2DE is best utilized to visualize proteins in a sample, including those that have been post-translationally modified, because this often results in a change in pi and/or molecular weight of the protein, and hence changes in gel mobility. However,

shortcomings include its inability to separate basic proteins, acidic proteins, high molecular weight proteins, small proteins and peptides and hydrophobic proteins.

Alternative strategies such as direct analysis by ESI-LCMSMS following proteolytic

digestion are therefore also required. Neither of these techniques, however, is able to

detect peptides and/or small proteins(< 10000 Da) in a sample. For the characterization

of small proteins and peptides, direct analysis by MALDI-TOFMS is suitable. Peptides

can be further sequence by PSD or ESI-LCMSMS and identified by de novo sequencing

and/or database searching. In addition, protein and peptide quantification using mass

spectrometry was explored using two different approaches: relative quantification of

proteins was explored by esterification of tryptic peptides using differential mass tags,

and absolute quantification of peptides and proteins was explored using MALDI­

TOFMS. The choice of appropriate quantification methodology is dependant on factors

such as sample type and complexity. The methods described here are only in the

243 preliminary stages of development, but they appear robust, accurate and simple. An integrated strategy that allows complete characterization of all peptides and proteins, as well as their quantification, is important when attempting to establish new biomarkers of disease, potential drug targets, or disease mechanisms.

244 CHAPTERS

REFERENCES

245 References:

Abell, D. C., Sporns, P. (1996). Rapid quantitation ofpotato glycoalkaloids by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. Journal of Agricultural & Food Chemistry 44(8):2292-2296.

Adams, M. D., Celniker, S. E., Holt, R. A., Evans, C. A., Gocayne, J. D., Amanatides, P. G., Scherer, S. E., Li, P. W., Hoskins, R. A., Galle, R. F., George, R. A., Lewis, S. E., Richards, S., Ashburn.er, M., Henderson, S. N., Sutton, G. G., Wortman, J. R., Yandell, M. D., Zhang, Q., Chen, L. X., Brandon, R. C., Rogers, Y. H., Blazej, R. G., Champe, M., Pfeiffer, B. D., Wan, K. H., Doyle, C., Baxter, E. G., Helt, G., Nelson, C. R., Gabor, G. L., Abril, J. F., Agbayani, A., An, H.J., Andrews-Pfannkoch, C., Baldwin, D., Ballew, R. M., Basu, A., Baxendale, J., Bayraktaroglu, L., Beasley, E. M., Beeson, K. Y., Benos, P. V., Berman, B. P., Bhandari, D., Bolshakov, S., Borkova, D., Botchan, M. R., Bouck, J., Brokstein, P., Brottier, P., Burtis, K. C., Busam, D. A., Butler, H., Cadieu, E., Center, A., Chandra, I., Cherry, J.M., Cawley, S., Dahlke, C., Davenport, L.B., Davies, P., de Pablos, B., Delcher, A., Deng, Z., Mays, A. D., Dew, I., Dietz, S. M., Dodson, K., Doup, L. E., Downes, M., Dugan-Rocha, S., Dunkov, B. C., Dunn, P., Durbin, K. J., Evangelista, C. C., Ferraz, C., Ferriera, S., Fleischmann, W., Fosler, C., Gabrielian, A. E., Garg, N. S., Gelbart, W. M., Glasser, K., Glodek, A., Gong, F., Gorrell, J. H., Gu, Z., Guan, P., Harris, M., Harris, N. L., Harvey, D., Heiman, T. J., Hernandez, J. R., Houck, J., Hostin, D., Houston, K. A., Howland, T. J., Wei, M. H., lbegwam, C., Jalali, M., Kalush, F., Karpen, G. H., Ke, Z., Kennison, J. A., Ketchum, K. A., Kimmel, B. E., Kodira, C. D., Kraft, C., Kravitz, S., Kulp, D., Lai, Z., Lasko, P., Lei, Y., Levitsky, A. A., Li, J., Li, Z., Liang, Y., Lin, X., Liu, X., Mattei, B., McIntosh, T. C., McLeod, M. P., McPherson, D., Merkulov, G., Milshina, N. V., Mobarry, C., Morris, J., Moshrefi, A., Mount, S. M., Moy, M., Murphy, B., Murphy, L., Muzny, D. M., Nelson, D. L., Nelson, D. R., Nelson, K. A., Nixon, K., Nusskern, D. R., Pacleb, J. M., Palazzolo, M., Pittman, G. S., Pan, S., Pollard, J., Puri, V., Reese, M. G., Reinert, K., Remington, K., Saunders, R. D., Scheeler, F., Shen, H., Shue, B. C., Siden-Kiamos, I., Simpson, M., Skupski, M. P., Smith, T., Spier, E., Spradling, A. C., Stapleton, M., Strong, R., Sun, E., Svirskas, R., Tector, C., Turner, R., Venter, E., Wang, A. H., Wang, X., Wang, Z. Y., Wassarman, D. A., Weinstock, G. M., Weissenbach, J., Williams, S. M., WoodageT, Worley, K. C., Wu, D., Yang, S., Yao, Q. A., Ye, J., Yeh, R. F., Zaveri, J. S., Zhan, M., Zhang, G., Zhao, Q., Zheng, L., Zheng, X. H., Zhong, F. N., Zhong, W., Zhou, X., Zhu, S., Zhu, X., Smith, H. 0., Gibbs, R. A., Myers, E.W., Rubin, G. M., Venter, J.C. (2000). The genome sequence ofDrosophila melanogaster. Science 287(5461):2185-2195.

246 Alberts, B. (1998). The cell as a collection ofprotein machines: preparing the next generation ofmolecular biologists. Cell 92(3):291-294.

Andalo, C., Bocchini, P., Pozzi, R., Galletti, G. C. (2001). Accurate mass measurement of synthetic analogues ofprazosine by matrix-assisted laser desorption/ionisation time-of­ .flight mass spectrometry. Rapid Commun Mass Spectrom 15(9):665-669.

Anderson, L., Seilhamer, J. (1997). A comparison ofselected mRNA and protein abundances in human liver. Electrophoresis 18(3-4):533-537.

Andren, P. E., Emmett, M. R., Caprioli, R. M. (1994). Micro-e/ectrospray: zeptomole/attomole per microlitre sensitivity for peptides. J Am Soc Mass Spectrom S:867-869.

Andrews, P. C., Alai, M., Cotter, R. J. (1988). The use ofplasma desorption time-of-flight mass spectrometry to screen for products ofprohormone processing in crude tissue extracts. Anal Biochem 174(1):23-31.

Ayorinde, F. 0., Garvin, K., Saeed, K. (2000). Determination ofthe fatty acid composition of saponi.fied vegetable oils using matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. Rapid Commun Mass Spectrom 14(7):608-615.

Ayorinde, F. 0., Hambright, P., Porter, T. N., Keith, Q. L., Jr. (1999). Use ofmeso­ tetrakis(pentafluorophenyl)porphyrin as a matrix for low molecular weight alkylphenol ethoxylates in laser desorption/ ionization time-of-flight mass spectrometry. Rapid Commun Mass Spectrom 13(24):2474-2479.

Ayrton, J., Dear, G. J., Leavens, W. J., Mallett, D. N., Plumb, R. S. (1998). Use ofgeneric fast gradient liquid chromatography-tandem mass spectroscopy in quantitative bioanalysis. J Chromatogr B Biomed Sci Appl 709(2):243-254.

Baguet, J., Claudon-Eyl, V., Gachon, A. M. (1992). Tear protein G originates from denatured tear specific prealbumin as revealed by two-dimensional electrophoresis. Curr Eye Res 11(11):1057-1065.

Bahr, U., Stahl-Zeng, J., Gleitsmann, E., Karas, M. (1997). Delayed extraction time-of-flight MALDI mass spectrometry ofproteins above 25,000 Da. J Mass Spectrom 32(10): 1111- 1116.

247 Bai, J., Liang, X., Liu, Y. H., Zhu, Y., Lubman, D. M. (1996). Characterization oftwo new matrices for matrix-assisted laser desorption/ionization mass spectrometry. Rapid Commun Mass Spectrom 10(7):839-844.

Baier, G., Wollensak, G., Mur, E., Redl, B., Stoffler, G., Gottinger, W. (1990). Analysis of human tear proteins by different high-performance liquid chromatographic techniques. J Chromatogr 525(2):319-328.

Bakhtiar, R., Steams, R. A. (1995). Studies on non-covalent associations ofimmunosuppressive drugs with serum albumin using pneumatically assisted electrospray ionization mass spectrometry. Rapid Commun Mass Spectrom 9(3):240-244.

Baldwin, M. A., Medzihradszky, K. F., Lock, C. M., Fisher, B., Settineri, T. A., Burlingame, A. L. (2001). Matrix-assisted laser desorption/ionization coupled with quadrupole/orthogonal acceleration time-of-flight mass spectrometry for protein discovery, identification, and structural analysis. Anal Chem 73(8):1707-1720.

Ball, R. W., Packman, L. C. (1997). Matrix-assisted laser desorption ionization time-of-flight mass spectrometry as a rapid quality control method in oligonucleotide synthesis. Anal Biochem 246(2): 185-194.

Ban1cs, J. F. (1997). Recent advances in capillary electrophoresis/electrospraylmass spectrometry. Electrophoresis 18(12-13):2255-2266.

Ban1cs, R. E., Dunn, M. J., Forbes, M. A., Stanley, A., Pappin, D., Naven, T., Gough, M., Harnden, P., Selby, P. J. (1999). The potential use oflaser capture microdissection to selectively obtain distinct populations ofcells for proteomic analysis--preliminary findings. Electrophoresis 20(4-5):689-700.

Barber, M., Bordolli, R. S., Sedgwick, R. D., Tyler, A. N. (1981 ). Fast atom bombardment of solids (F.A.B.): A new ionization source for mass spectrometry. J Chem Soc Chem Commun 7:325-327.

Barka, T., Asbell, P.A., van der Noen, H., Prasad, A. (1991). Cystatins in human tear fluid. Curr Eye Res 10(1):25-34.

Barrett, J., Jefferies, J. R., Brophy, P. M. (2000). Parasite proteomics. Parasitol Today 16(9):400-403.

248 Bayer, E., Bauer, T., Schmeer, K., Bleicher, K., Maier, M., Gaus, H.J. (1994). Analysis of double-stranded oligonucleotides by electrospray mass spectrometry. Anal Chem 66(22):3858-3863.

Beavis, R. C., Chait, B. T. (1989a). Cinnamic acid derivatives as matrices for ultraviolet laser desorption mass spectrometry ofproteins. Rapid Commun Mass Spectrom 3(12):432-435.

Beavis, R. C., Chait, B. T. (1989b). Factors affecting the ultraviolet laser desorption of proteins. Rapid Commun Mass Spectrom 3(7):233-237.

Beavis, R. C., Chait, B. T. (1989c). Matrix-assisted laser-desorption mass spectrometry using 355 nm radiation. Rapid Commun Mass Spectrom 3(12):436-439.

Beavis, R. C., Chait, B. T. (1990). High-accuracy molecular mass determination ofproteins using matrix-assisted laser desorption mass spectrometry. Anal Chem 62(17): 1836-1840.

Behndig, A., Svensson, B., Marklund, S. L., Karlsson, K. (1998). Superoxide dismutase isoenzymes in the human eye. Invest Ophthalmol Vis Sci 39(3):471-475.

Bentzley, C. M., Johnston, M. V., Larsen, B. S., Gutteridge, S. (1996). Oligonucleotide sequence and composition determined by matrix-assisted laser desorption/ionization. Anal Chem 68(13):2141-2146.

Berggren, K., Chernokalskaya, E., Steinberg, T. H., Kemper, C., Lopez, M. F., Diwu, Z., Haugland, R. P., Patton, W. F. (2000). Background-free, high sensitivity staining of proteins in one- and two-dimensional sodium dodecyl sulfate-polyacrylamide gels using a luminescent ruthenium complex. Electrophoresis 21(12):2509-2521.

Berta, A. (1982). Apolyacrylamide-gel electrophoretic study ofhuman tear proteins. Graefes Arch Clin Exp Ophthalmol 219(2):95-99.

Bichsel, V. E., Liotta, L. A., Petricoin, E. F., 3rd (2001). Cancer proteomics:from biomarker discovery to signal pathway profiling. Cancer J 7(1 ):69-78.

Biemann, K. (1988). Contributions ofmass spectrometry to peptide and protein structure. Biomed Environ Mass Spectrom 16(1-12):99-111.

Biemann, K. (1992). Mass spectrometry ofpeptides and proteins. Annu Rev Biochem 61:977- 1010.

249 Biemann, K., Scoble, H. A. (1987). Characterization by tandem mass spectrometry ofstructural modifications in proteins. Science 237(4818):992-998.

Bjellqvist, B., Ek, K., Righetti, P. G., Gianazza, E., Gorg, A., Westermeier, R., Postel, W. (1982). lsoelectricfocusing in immobilized pH gradients: principle, methodology and some applications. J Biochem Biophys Methods 6(4):317-339.

Blackledge, J. A., Alexander, A. J. (1995). Polyethylene membrane as a sample support for direct matrix-assisted laser desorption/ionization mass spectrometric analysis ofhigh mass proteins. Anal Chem 67(5):843-848.

Blakley, C.R., Carmody, J.C., Vestal, M. L. (1980). Combined liquid chromatograph/mass spectrometer for involatile biological samples. Clin Chem 26(10):1467-1473.

Bleczinski, C. F., Richert, C. (1998). Monitoring the hybridization of the components of oligonucleotide mixtures to immobilized DNA via matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. Rapid Commun Mass Spectrom 12(22):1737-1743.

Boeshans, K. M., Resing, K. A., Hunt, J.B., Ahn;N. G., Shabb, J.B. (1999). Structural characterization ofthe membrane-associated regulatory subunit oftype I cAMP­ dependent protein kinase by mass spectrometry: identification ofSer81 as the in vivo phosphorylation site ofRlalpha. Protein Sci 8(7):1515-1522.

Bonavida, B., Sapse, A. T., Sercarz, E. E. (1969). Specific tear prealbumin: a unique lachrymal protein absent from serum and other secretions. Nature 221(178):375-376.

Borchers, C., Peter, J. F., Hall, M. C., Kunkel, T. A., Tomer, K. B. (2000). Identification ofin­ gel digested proteins by complementary peptide mass fingerprinting and tandem mass spectrometry data obtained on an electrospray ionization quadrupole time-of-flight mass spectrometer. Anal Chem 72(6):1163-1168.

Borchers, C., Tomer, K. B. (1999). Characterization ofthe noncovalent complex ofhuman immunodeficiency virus glycoprotein 120 with its cellular receptor CD4 by matrix­ assisted laser desorption/ionization mass spectrometry. Biochemistry 38(36): 11734- 11740.

Bordini, E., Hamdan, M. (1999). Investigation ofsome covalent and noncovalent complexes by matrix-assisted laser desorption/ionization time-of-flight and electrospray mass spectrometry. Rapid Commun Mass Spectrom 13(12):1143-1151.

250 Boukes, R. J., Boonstra, A., Breebaart, A. C., Reits, D., Glasius, E., Luyendyk, L., Kijlstra, A. (1987). Analysis ofhuman tear protein pro.files using high performance liquid chromatography (HPLC). Doc Ophthalmol 67(1-2):105-113.

Brauninger, G. E., Centifanto, Y. M. (1971). lmmunog/obulin E in human tears. Am J Ophthalmol 72(3):558-561.

Bray, M. S., Boerwinkle, E., Doris, P.A. (2001). High-throughput multiplex SNP genotyping with MALDI-TOF mass spectrometry: Practice, problems and promise. Hum Mutat 17(4):296-304.

Broberg, S., Broberg, A., Duus, J. 0. (2000). Matrix-assisted laser desorption/ionization time­ of-flight mass spectrometry ofoligosaccharides derivatized by reductive amination and N,N-dimethylation. Rapid Commun Mass Spectrom 14(19): 1801-1805.

Brown, R. S., Lennon, J. J. (1995). Mass resolution improvement by incorporation ofpulsed ion extraction in a matrix-assisted laser desorption/ionization linear time-of-flight mass spectrometer. Anal Chem 67(13):1998-2003.

Bruenner, B. A., Yip, T. T., Hutchens, T. W. (1996). Quantitative analysis ofoligonucleotides by matrix-assisted laser desorption/ionization mass spectrometry. Rapid Commun Mass Spectrom 10(14):1797-1801.

Camafeita, E., Alfonso, P., Mothes, T., Mendez, E. (1997). Matrix-assisted laser desorption/ionization time-of-flight mass spectrometric micro-analysis: the first non­ immunological alternative attempt to quantify gluten gliadins in food samples. J Mass Spectrom 32(9):940-947.

Cans, C., Mangano, R., Barila, D., Neubauer, G., Superti-Furga, G. (2000). Nuclear tyrosine phosphorylation: the beginning ofa map. Biochem Pharmacol 60(8):1203-1215.

Caprioli, R. M. (1990). Bombardment mass spectrometry. Anal Chem 62(8):477A-485A.

Caprioli, R. M., Fan, T., Cottrell, J. S. (1986). Continuous-flow sample probe for fast atom bombardment mass spectrometry. Anal Chem 58:2949-2954.

Caprioli, R. M., Farmer, T. B., Gile, J. (1997). Molecular imaging ofbiological samples: localization ofpeptides and proteins using MALDI-TOF MS. Anal Chem 69(23):4751- 4760.

251 Carmel, R. (1972). The vitamin B 12 -binding proteins ofsaliva and tears and their relationship to other vitamin B 12 binders. BiochimBiophys Acta 263(3):747-752.

Cash, P. (2000). Proteomics in medical microbiology. Electrophoresis 21(6):1187-1201.

Celis, J.E., Celis, P., Ostergaard, M., Basse, B., Lauridsen, J.B., Ratz, G., Rasmussen, H. H., Omtoft, T. F., Hein, B., Wolf, H., Celis, A. (1999). Proteomics and immunohistochemistry de.fine some ofthe steps involved in the squamous differentiation of the bladder transitional epithelium: a novel strategy for identifying metaplastic lesions. Cancer Res 59(12):3003-3009.

Celis, J.E., Wolf, H., Ostergaard, M. (2000). Bladder squamous cell carcinoma biomarkers derived from proteomics. Electrophoresis 21(11 ):2115-2121.

Cerpa-Poljak, A., Lahnstein, J., Mason, K. E., Smythe, G. A., Duncan, M. W. (1997). Mass spectrometric identification and quantification ofhemorphins extracted from human adrenal and pheochromocytoma tissue. J Neurochem 68(4):1712-1719.

Charlwood, J., Birrell, H., Organ, A., Camilleri, P. (1999a). A chromatographic and mass spectrometric strategy for the analysis ofoligosaccharides: determination ofthe glycan structures in porcine thyroglobulin. Rapid Commun Mass Spectrom 13(8):716-723.

Charlwood, J., Tolson, D., Dwek, M., Camilleri, P. (1999b). A detailed analysis ofneutral and acidic carbohydrates in human milk. Anal Biochem 273(2):261-277.

Charrier, J.P., Tournel, C., Michel, S., Dalbon, P., Jolivet, M. (1999). Two-dimensional electrophoresis ofprostate-specific antigen in sera ofmen with prostate cancer or benign prostate hyperplasia. Electrophoresis 20(4-5): 1075-1081.

Chaurand, P., Luetzenkirchen, F., Spengler, B. (1999a). Peptide and protein identification by matrix-assisted laser desorption ionization (MALDI) and MALDI-post-source decay time­ of-flight mass spectrometry. J Am Soc Mass Spectrom 10(2):91-103.

Chaurand, P ., Stoeckli, M., Caprioli, R. M. (1999b). Direct pro.filing ofproteins in biological tissue sections by MALDI mass spectrometry. Anal Chem 71(23):5263-5270.

Chen, S., Pieraccini, G., Moneti, G. (1991). Quantitative analysis ofthe molecular species of monosialogangliosides by continuous-flow fast-atom bombardment mass spectrometry. Rapid Commun Mass Spectrom 5(12):618-621.

252 Chevenne, D., Trivin, F., Porquet, D. (1999). Insulin assays and reference values. Diabetes Metab 25(6):459-476.

Choudhary, G., Apffel, A., Yin, H., Hancock, W. (2000). Use ofon-line mass spectrometric detection in capillary electrochromatography. J Chromatogr A 887(1-2):85-101.

Christensson, A., Lilja, H. (1994). Complex formation between protein C inhibitor and prostate­ specific antigen in vitro and in human semen. Eur J Biochem 220(1):45-53.

Christian, N. P., Colby, S. M., Giver, L., Houston, C. T., Arnold, R. J., Ellington, A. D., Reilly, J.P. (1995). High resolution matrix-assisted laser desorption/ionization time-of-flight analysis ofsingle-stranded DNA of27 to 68 nucleotides in length. Rapid Commun Mass Spectrom 9(11):1061-1066.

Clauser, K. R., Baker, P., Burlingame, A. L. (1999). Role ofaccurate mass measurement (+I­ i Oppm) in protein identification strategi,es employing MS or MS/MS and database searching. Anal Chem 71(14):2871-2882.

Cockle, S. M., Prater, G. V., Thetford, C.R., Hamilton, C., Malone, P.R., Mundy, A. R. (1994). Peptides related to thyrotrophin-releasing hormone (TRH) in human prostate and semen. Biochim Biophys Acta 1227(1-2):60-66.

Cody, R. B., Jr., Amster, I. J., McLafferty, F. W. (1985). Peptide mixture sequencing by tandem Fourier-transform mass spectrometry. Proc Natl Acad Sci US A 82(19):6367-6370.

Cohen, S. L., Chait, B. T. (1996). Influence ofmatrix solution conditions on the MALDI-MS analysis ofpeptides and proteins. Anal Chem 68(1):31-37.

Colangelo, J., Orlando, R. (1999). On-target exoglycosidase digestions/MALDI-MS for determining the primary structures ofcarbohydrate chains. Anal Chem 71(7):1479-1482.

Cole, R. B. (2000). Some tenets pertaining to electrospray ionization mass spectrometry. J Mass Spectrom 35(7):763-772.

Conrads, T. P., Alving, K., Veenstra, T. D., Belov, M. E., Anderson, G. A., Anderson, D. J., Lipton, M. S., Pasa-Tolic, L., Udseth, H. R., Chrisler, W. B., Thrall, B. D., Smith, R. D. (2001 ). Quantitative analysis ofbacterial and mammalian proteomes using a combination ofcysteine affinity tags and l 5N-metabolic labeling. Anal Chem 73(9):2132-2139.

253 The C.elegans Sequencing Consortium. (1998). Genome sequence ofthe nematode C.elegans: Aplatformfor investigating biology. Science 282(5396):2012-2018.

Cooks, R. G., Glish, G. L., McLuckey, S. A., Kaiser, R. E. (1991). Ion trap mass spectrometry. Chem Eng News 69(12):26-41.

Cordero, M. M., Cornish, T. J., Cotter, R. J., Lys, I. A. (1995). Sequencing peptides without scanning the reflectron: post-source decay with a curved-field reflectron time-of-flight mass spectrometer. Rapid Commun Mass Spectrom. 9(14):1356-61.

Cornish, T. J., Cotter, R. J. (1994). A curved field reflectron time-of-flight mass spectrometer for the simultaneous focusing ofmetastable product ions. Rapid Commun Mass Spectrom. 8(9):781-5.

Cordwell, S. J., Wasinger, V. C., Cerpa-Poljak, A., Duncan, M. W., Humphery-Smith, I. (1997). Conserved motifs as the basis for recognition ofhomologous proteins across species boundaries using peptide-mass fingerprinting. J Mass Spectrom 32(4):370-378.

Cotter, R. J., Fancher, C., Cornish, T. J. (1999). Miniaturized time-of-flight mass spectrometer for peptide and o/igonucleotide analysis. J Mass Spectrom 34(12):1368-1372.

Cottrell, J. S. (1994). Protein identification by peptide mass fingerprinting. Pept Res 7(3):115- 124.

Coyle, P. K., Sibony, P.A. (1986). Tear immunoglobulins measured by ELISA. Invest Ophthalmol Vis Sci 27(4):622-625.

Coyle, P. K., Sibony, P.A. (1987). Viral specificity ofmultiple sclerosis tear immunoglobulins. J Neuroimmunol 14(2):197-203.

Coyle, P. K., Sibony, P.A., Johnson, C. (1987). Increased monomeric immunoglobulin A levels in tears from multiple sclerosis patients. Ann Neurol 21(2):211-214.

Coyle, P. K., Sibony, P.A., Johnson, C. (1989). Electrophoresis combined with immunologic identification ofhuman tear proteins. Invest Ophthalmol Vis Sci 30(8):1872-1878.

Craig, A. G., Engstrom, A., Bennich, H., Kamensky, I. (1987). Plasma desorption mass spectrometry coupled with conventional peptide sequencing techniques. Biomed Environ Mass Spectrom 14(11):669-673.

254 Crouch, R. K., Goletz, P., Snyder, A., Coles, W. H. (1991). Antioxidant enzymes in human tears. J Ocul Pharmacol 7(3):253-258.

Dai, Y., Whittal, R. M., Bridges, C. A., Isogai, Y., Hindsgaul, 0., Li, L. (1997). Matrix-assisted laser desorption ionization mass spectrometry for the analysis ofmonosulfated oligosaccharides. Carbohydr Res 304(1):1-9.

Dai, Y., Whittal, R. M., Li, L., Weinberger, S. R. (1996). Accurate mass measurement of oligonucleotides using a time-lag focusing matrix-assisted laser desorption/ionization time-of-flight mass spectrometer. Rapid Commun Mass Spectrom 10(14): 1792-1796.

Dalluge, J. J. (2000). Mass spectrometry for direct determination ofproteins in cells: applications in biotechnology and microbiology. Fresenius J Anal Chem 366(6-7):701- 71 l.

Dass, C., Kusmierz, J. J., Desiderio, D. M. (1991). Mass spectrometric quantification of endogenous beta-endorphin. Biol Mass Spectrom 20(3):130-138.

Davis, M. T., Beierle, J., Bures, E. T., McGinley, M. D., Mort, J., Robinson, J. H., Spahr, C. S., Yu, W., Luethy, R., Patterson, S. D. (2001): Automated LC-LC-MS-MS platform using binary ion-exchange and gradient reversed-phase chromatography for improved proteomic analyses. J Chromatogr B Biomed Sci Appl 752(2):281-291.

Davis, M. T., Lee, T. D. (1998). Rapid protein identification using a microscale electrospray LCIMS system on an ion trap mass spectrometer. J Am Soc Mass Spectrom 9(3):194-201.

Davis, M. T., Stahl, D. C., Hefta, S. A., Lee, T. D. (1995). A microscale electrospray interface for on-line, capillary liquid chromatography/tandem mass spectrometry ofcomplex peptide mixtures. Anal Chem 67(24):4549-4556.

Davoli, E., Fanelli, R., Bagnati, R. (1993). Purification and analysis ofdrug residues in urine samples by on-line immunoaffinity chromatography/high-performance liquid chromatography/continuous-flow fast atom bombardment mass spectrometry. Anal Chem 65(19):2679-2685.

Dehennin, L. (1990). Estrogens, androgens, and progestins in follicular fluid from preovulatory follicles: identification and quantification by gas chromatography/mass spectrometry associated with stable isotope dilution. Steroids 55(4): 181-184.

255 Desiderio, D. M. (1992). Quantitative analytical mass spectrometry ofendogenous neuropeptides in human pituitaries. Life Sci 51(3): 169-76.

Desiderio, D. M., Wirth, U., Lovelace, J. L., Fridland, G., Umstot, E. S., Nguyen, T. M., Schiller, P. W., Szeto, H. S., Clapp, J. F. (2000). Matrix-assisted laser desorption/ionization mass spectrometric quantification ofthe mu opioid receptor agonist DAMGO in ovine plasma. J Mass Spectrom 35(6):725-733.

Dickerman, E., Dickerman, S., Meites, J. (1971): Influence of age, sex and estrous cycle on pituitary and plasma growth hormone levels in rats. In Pecile, A., Muller, E. E. (eds): "Growth and Growth Hormone - International Symposium on Growth Hormone." Amsterdam: Excerpta Medica, pp 252-260.

Dickinson, D. P., Thiesse, M. (1995). A major human lacrimal gland mRNA encodes a new proline-rich protein family member. Invest Ophthalmol Vis Sci 36(10):2020-2031.

Dickinson, D. P., Thiesse, M. (1996). cDNA cloning ofan abundant human lacrimal gland mRNA encoding a novel tear protein. Curr Eye Res 15(4):377-386.

Dole, M., Hines, R. L., Mack, R. C., Mobley, R. C., Ferguson, L. D., Alice, M. B. (1968). Molecular beams ofmacroions. J Chem Phys 49:2240-2249.

Domingo, I., Coll, J., Ribas-Montobio, J., Marrugat, J., Rubies-Prat, J. (1998). Lacrimal immunoglobulins in rheumatoid arthritis patients with or without Sjogren's syndrome. Ophthalmologica 212(1):30-33.

Doroshenko, V. M., Cotter, R. J. (1998). A quadrupole ion trap/time-of-flight mass spectrometer with a parabolic reflectron. J Mass Spectrom 33(4):305-318.

Dota, A., Nishida, K., Quantock, A. J., Kinoshita, S. (1999). Clusterin in human corneal endothelium and aqueous humor. Exp Eye Res 69(6):705-708.

Duncan, M. W., Matanovic, G., Cerpa-Poljak, A. (1993). Quantitative analysis of/ow molecular weight compounds ofbiological interest by matrix-assisted laser desorption ionization. Rapid Commun Mass Spectrom 7(12):1090-1094.

Eckerskom, C., Strupat, K., Schleuder, D., Hochstrasser, D., Sanchez, J.C., Lottspeich, F., Hillenkamp, F. (1997). Analysis ofproteins by direct-scanning infrared-MALDI mass spectrometry after 2D-PAGE separation and electroblotting. Anal Chem 69(15):2888- 2892.

256 Ekstrom, S., Ericsson, D., Onnerfjord, P., Bengtsson, M., Nilsson, J., Marko-Varga, G., Lauren, T. (2001). Signal amplification using "spot-on-a-chip" technology for the identification of proteins via MALDJ-TOF MS. Anal Chem 73(2):214-219.

Ekstrom, S., Onnerfjord, P., Nilsson, J., Bengtsson, M., Laurell, T., Marko-Varga, G. (2000). Integrated microanalytical technology enabling rapid and automated protein identification. Anal Chem 72(2):286-293.

Emmett, M. R., Caprioli, R. M. (1994). Micro-electrospray mass spectrometry: ultra-high sensitivity analysis ofpeptides and proteins. J Am Soc Mass Spectrom 5:605-613.

Eng, J. K., McCormack, A. L., Yates, J. R. I. (1994). An approach to correlate tandem mass spectral data ofpeptides with amino acid sequences in a protein database. J. Am. Soc. Mass Spectrom. 5:976-989.

Evershed, R. P., Robertson, D. H., Beynon, R. J., Green, B. N. (1993). Application of electrospray ionization mass spectrometry with maximum-entropy analysis to allelic :fingerprinting' ofmajor urinary proteins. Rapid Commun Mass Spectrom 7(10):882-886.

Fales, H. M., McNeal, C. J., Macfarlane, R. D., Shimohigashi, Y. (1985). Californium-252 plasma-desorption mass spectrometry ofpolymethylenediamine-linked enkephalin peptides. Anal Chem 57(8):1616-1621.

Farmer, T. B., Caprioli, R. M. (1998). Determination ofprotein-protein interactions by matrix­ assisted laser desorption/ionization mass spectrometry. J Mass Spectrom 33(8):697-704.

Faulstich, K., Womer, K., Brill, H., Engels, J. W. (1997). A sequencing method for RNA oligonucleotides based on mass spectrometry. Anal Chem 69(21):4349-4353.

Fei, Z., Ono, T., Smith, L. M. (1998). MALDJ-TOF mass spectrometric typing ofsingle nucleotide polymorphisms with mass-tagged ddNTPs. Nucleic Acids Res 26(11):2827- 2828.

Fei, Z., Smith, L. M. (2000). Analysis ofsingle nucleotide polymorphisms by primer extension and matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. Rapid Commun Mass Spectrom 14(11):950-959.

Fenn, J.B., Mann, M., Meng, C. K., Wong, S. F., Whitehouse, C. M. (1989). Electrospray ionization/or mass spectrometry oflarge biomolecules. Science 246(4926):64-71.

257 Figeys, D., Aebersold, R. (1997). High sensitivity identification ofproteins by electrospray ionization tandem mass spectrometry: initial comparison between an ion trap mass spectrometer and a triple quadrupole mass spectrometer. Electrophoresis 18(3-4):360- 368.

Figeys, D., Aebersold, R. (1999). Microfabricated modules for sample handling, sample concentration and flow mixing: application to protein analysis by tandem mass spectrometry. J Biomech Eng 121(1):7-12.

Figeys, D., Gygi, S. P., McKinnon, G., Aebersold, R. (1998a). An integrated microfluidics­ tandem mass spectrometry system for automated protein analysis. Anal Chem 70(18):3728-3734.

Figeys, D., Lock, C., Taylor, L., Aebersold, R. (1998b). Microfabricated device coupled with an electrospray ionization quadrupole time-of-flight mass spectrometer: protein identifications based on enhanced-resolution mass spectrometry and tandem mass spectrometry data. Rapid Commun Mass Spectrom 12(20): 1435-1444.

Figeys, D., Pinto, D. (2001 ). Proteomics on a chip: promising developments. Electrophoresis 22(2):208-216.

Finke, B., Stahl, B., Pfenninger, A., Karas, M., Daniel, H., Sawatzki, G. (1999). Analysis of high-molecular-weight oligosaccharides from human milk by liquid chromatography and MALDI-MS. Anal Chem 71(17):3755-3762.

Fitzgerald, M. C., Parr, G. R., Smith, L. M. (1993). Basic matrices for the matrix-assisted laser desorption/ionization mass spectrometry ofproteins and oligonucleotides. Anal Chem 65(22):3204-321 l.

Fitzgerald, M. C., Smith, L. M. (1995). Mass spectrometry ofnucleic acids: the promise of matrix-assisted laser desorption-ionization (MALDI) mass spectrometry. Annu Rev Biophys Biomol Struct 24:117-140.

Fournier, I., Chaurand, P., Holbach, G., Lutzenkirchen, F., Spengler, B., Tabet, J.C. (2000). Sequencing ofa branched peptide using matrix-assisted laser desorption/ionization time­ of-flight mass spectrometry. J Mass Spectrom 35(12):1425-1433.

Frey, W. H., Nelson, J. D., Frick, M. L., Elde, R. P. (1986): Prolactin immunoreactivity in human tears and lacrimal gland: Possible implications for tear production. In Holly, F. J.

258 (ed): "The Preocular Tear Film in Health, Disease, and Contact Lens Wear." Lubbock, Texas: The Dry Eye Institute, pp 798-807.

Frey, W. H. d., DeSota-Johnson, D., Hoffman, C., McCall, J. T. (1981). Effect ofstimulus on the chemical composition ofhuman tears. Am J Ophthalmol 92(4):559-567.

Friedman, M. G. (1990). Antibodies in human tears during and after infection. Surv Ophthalmol 35(2): 151-157.

Fu, D. J., Broude, N. E., Koster, H., Smith, C. L., Cantor, C.R. (1996). Efficient preparation of short DNA sequence ladders potentially suitable for MALDI-TOF DNA sequencing. Genet Anal 12(3-4):137-142.

Fujiwaki, T., Yamaguchi, S., Sukegawa, K., Taketomi, T. (1999). Application ofdelayed extraction matrix-assisted laser desorption ionization time-of-flight mass spectrometry for analysis ofsphingolipids in tissues from sphingolipidosis patients. J Chromatogr B Biomed Sci Appl 731(1):45-52.

Fukai, T ., Kuroda, J., Nomura, T. (2000). Accurate mass measurement oflow molecular weight compounds by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. J Am Soc Mass Spectrom 11(5):458-463.

Fukuda, M., Fullard, R. J., Willcox, M. D., Baleriola-Lucas, C., Bestawros, F., Sweeney, D., Holden, B. A. (1996). Fibronectin in the tear film. Invest Ophthalmol Vis Sci 37(2):459- 467.

Fullard, R. J. (1988). Identification ofproteins in small tear volumes with and without size exclusion HPLC fractionation. Curr Eye Res 7(2): 163-179.

Fullard, R. J., Kissner, D. M. (1991). Purification ofthe isoforms oftear specific prealbumin. Curr Eye Res 10(7):613-628.

Fullard, R. J., Tucker, D. L. (1991). Changes in human tear protein levels with progressively increasing stimulus. Invest Ophthalmol Vis Sci 32(8):2290-2301.

Fung, E.T., Wright, G. L., Jr., Dalmasso, E. A. (2000). Proteomic strategies for biomarker identification: progress and challenges. Curr Opin Mol Ther 2(6):643-650.

Gale, D. C., Smith, R. D. (1993). Small volume and low flow-rate electrospray ionization mass spectrometry ofaqueous samples. Rapid Commun Mass Spectrom 7: 1017-1021.

259 Gallardo, K., Job, C., Groot, S. P., Puype, M., Demol, H., Vandekerckhove, J., Job, D. (2001). Proteomic analysis ofarabidopsis seed germination and priming. Plant Physiol 126(2):835-848.

Garozzo, D., Nasello, V., Spina, E., Sturiale, L. (1997). Discrimination ofisomeric oligosaccharides and sequencing ofunknowns by post source decay matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. Rapid Commun Mass Spectrom 11(14): 1561-1566.

Garozzo, D., Spina, E., Cozzolino, R., Cescutti, P., Fett, W. F. (2000). Studies on the primary structure ofshort polysaccharides using SEC MALDI mass spectroscopy. Carbohydr Res 323(1-4): 139-146.

Gaskell, S. J. (1990). Quantification ofsteroid conjugates usingfast atom bombardment mass spectrometry. Steroids 55(10):458-462.

Gaskell, S. J. (1997). Electrospray: Principles and Practice. J Mass Spectrom 32:677-688.

Gaskell, S. J., Brownsey, B. G., Brooks, P. W., Green, B. N. (1983). Fast atom bombardment mass spectrometry ofsteroid sulphates: qualitative and quantitative analyses. Biomed Mass Spectrom 10(3):215-219.

Gatlin, C. L., Kleemann, G. R., Hays, L. G., Link, A. J., Yates, J. R., 3rd (1998). Protein identification at the low femtomo/e level from silver-stained gels using a new.fritless electrospray inte,face for liquid chromatography-microspray and nanospray mass spectrometry. Anal Biochem 263(1):93-101.

Gelpi, E. (1995). Biomedical and biochemical applications ofliquid chromatography-mass spectrometry. J Chromatogr A 703(1-2):59-80.

Gevaert, K., Demol, H., Martens, L., Hoorelbeke, B., Puype, M., Goethals, M., Van Damme, J., De Boeck, S., Vandekerckhove, J. (2001). Protein identification based on matrix assisted laser desorption/ionization-post source decay-mass spectrometry. Electrophoresis 22(9): 1645-1651.

Geyer, H., Schmitt, S., Wuhrer, M., Geyer, R. (1999). Structural analysis ofglycoconjugates by on-target enzymatic digestion and MALDI-TOF-MS. Anal Chem 71(2):476-482.

Gharahdaghi, F., Kirchner, M., Fernandez, J., Mische, S. M. (1996). Peptide-mass pro.files of polyvinylidene difluoride-bound proteins by matrix-assisted laser desorption/ionization

260 time-of-flight mass spectrometry in the presence ofnonionic detergents. Anal Biochem 233(1):94-99.

Gharahdaghi, F., Weinberg, C.R., Meagher, D. A., Imai, B. S., Mische, S. M. (1999). Mass spectrometric identification ofproteins from silver-stained polyacrylamide gel: a method for the removal ofsilver ions to enhance sensitivity. Electrophoresis 20(3):601-605.

Gimon, M. E., Kinsel, G. R., Edmondson, R. D., Russell, D. H., Prout, T. R., Ewald, H. A. (1994). Matrix-assisted laser desorption/ionization time-of-flight mass spectrometry of pac/itaxel and related taxanes. J Nat Prod 57(10):1404-1410.

Gkonos, P. J., Kwok, C. K., Block, N. L., Roos, B. A. (1994). Identification ofthe human seminal TRH-like peptide pGlu-Phe-Pro-NH2 in normal human prostate. Peptides 15(7):1281-1283.

Gleispach, H., Huber, E., Fauler, G., Kerbl, R., Urban, C., Leis, H.J. (1995). Neuroblastoma screening: labeling ofHVA and VMAfor stable isotope dilution gas chromatography­ mass spectrometry. Nutrition 11(5 Suppl):604-606.

Gobom, J., Kraeuter, K. 0., Persson, R., Steen, H:, Roepstorff, P., Elcman, R. (2000). Detection and quantification ofneurotensin in human brain tissue by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. Anal Chem 72(14):3320-3326.

Godovac-Zimmermann, J., Soskic, V., Poznanovic, S., Brianza, F. (1999). Functional proteomics ofsign.al transduction by membrane receptors. Electrophoresis 20(4-5):952- 961.

Goebbels, M. (2000). Tear secretion and tear film function in insulin dependent diabetics. Br J Ophthalmol 84(1):19-21.

Goffeau, A., Barrell, B. G., Bussey, H., Davis, R. W., Dujon, B., Feldmann, H., Galibert, F., Hoheisel, J. D., Jacq, C., Johnston, M., Louis, E. J., Mewes, H. W., Murakami, Y., Philippsen, P., Tettelin, H., Oliver, S. G. (1996). Life with 6000 genes. Science 274(5287):546, 563-547.

Goldberger, B. A., Cone, E. J. (1994). Confirmatory tests for drugs in the workplace by gas chromatography-mass spectrometry. J Chromatogr A 674(1-2):73-86.

Goodlett, D.R., Keller, A., Watts, J. D., Newitt, R., Yi, E. C., Purvine, S., Eng, J. K., Haller, P., Aebersold, R., Kolker, E. (2001). Differential stable isotope labeling ofpeptides for

261 quantitation and de novo sequence derivation. Rapid Commun Mass Spectrom 15(14):1214-1221.

Gorg, A., Postel, W., Gunther, S. (1988). The current state oftwo-dimensional electrophoresis with immobilized pH gradients. Electrophoresis 9(9):531-546.

Goverde, H.J., Bisseling, J. G., Wetzels, A. M., Braat, D. D., Pesman, G. J., Sweep, F. C., Meuleman, E. J. (1998). A neuropeptide in human semen: oxytocin. Arch Androl 41(1):17-22.

Griffin, T. J., Gygi, S. P., Rist, B., Aebersold, R., Loboda, A., Jilkine, A., Ens, W., Standing, K. G. (2001). Quantitative proteomic analysis using a MALDI quadrupole time-of-flight mass spectrometer. Anal Chem 73(5):978-986.

Griffiths, W. J. (2000). Nanospray mass spectrometry in protein and peptide chemistry. Exs 88:69-79.

Griffiths, W. J., Jonsson, A. P., Liu, S., Rai, D. K., Wang, Y. (2001). Electrospray and tandem mass spectrometry in biochemistry. Biochem J 3SS(Pt 3):545-561.

Gross, J., Hillenkamp, F., Wan, K. X., Gross, M. L. (2001). Metastable decay ofnegatively charged oligodeoxynucleotides analyzed with ultraviolet matrix-assisted laser desorption/ionization post-source decay and deuterium exchange. J Am Soc Mass Spectrom 12(2): 180-192.

Gross, J., Leisner, A., Hillenkamp, F., Hahner, S., Karas, M., Schafer, J., Lutzenkirchen, F., Nordhoff, E. (1998). Investigations ofthe metastable decay ofDNA under ultraviolet matrix-assisted laser desorption/ionization conditions with post-source-decay analysis and hydrogen/deuterium exchange. J Am Soc Mass Spectrom 9(9):866-878.

Grus, F. H., Sabuncuo, P., Augustin, A. J. (2001). Analysis oftear protein patterns ofdry-eye patients usingjluorescent staining dyes and two-dimensional quantification algorithms. Electrophoresis 22(9): 1845-1850.

Guillonneau, F., Guieysse, A. L., Le Caer, J.P., Rossier, J., Praseuth, D. (2001). Selection and identification ofproteins bound to DNA triple-helical structures by combination of2D­ electrophoresis and MALDI-TOF mass spectrometry. Nucleic Acids Res 29(11):2427- 2436.

262 Guittard, J., Hronowski, X. L., Costello, C. E. (1999). Direct matrix-assisted laser desorption/ionization mass spectrometric analysis ofglycosphingolipids on thin layer chromatographic plates and transfer membranes. Rapid Commun Mass Spectrom 13(18): 1838-1849.

Gupta, A., Monroy, D., Ji, Z., Yoshino, K., Huang, A., Pflugfelder, S. C. (1996). Transforming growth factor beta-I and beta-2 in human tear fluid. Curr Eye Res 15(6):605-614.

Gupta, A. K., Sarin, G. S., Mathur, M. D., Ghosh, B. (1988). Alpha 1-antitrypsin and serum albumin in tear fluids in acute adenovirus conjunctivitis. Br J Ophthalmol 72(5):390-393.

Gusev, A. I., Wilkinson, W. R., Proctor, A., Hercules, D. M. (1993). Quantitative analysis of peptides by matrix-assisted laser desorption ionization time-of-flight mass spectrometry. Applied Spectroscopy 47(8):1091-1092.

Gusev, A. I., Wilkinson, W. R., Proctor, A., Hercules, D. M. (1995). Improvement ofsignal reproducibility and matrixlcomatrix effects in MALDI analysis. Anal Chem 67(6):1034- 1041.

Gusev, A. I., Wilkinson, W.R., Proctor, A., Hercules, D. M. (1996). Direct quantitative analysis ofpeptides using matrix assisted laser desorption ionization. Fresenius Journal of Analytical Chemistry 354(4):455-463.

Gutierrez, J. A. (2000). Genomics: from novel genes to new therapeutics in parasitology. Int J Parasitol 30(3):247-252.

Guttman, A., Ronai, Z., Csapo, Z., Gerstner, A., Sasvari-Szekely, M. (2000). Rapid analysis of covalently and non-covalently fluorophore-labeled proteins using ultra-thin-layer sodium dodecylsulfate gel electrophoresis. J Chromatogr A 894(1-2):329-335.

Gygi, S. P., Corthals, G. L., Zhang, Y., Rochon, Y., Aebersold, R. (2000). Evaluation oftwo­ dimensional gel electrophoresis-based proteome analysis technology. Proc Natl Acad Sci U S A 97(17):9390-9395.

Gygi, S. P., Rist, B., Gerber, S. A., Turecek, F., Gelb, M. H., Aebersold, R. (1999a). Quantitative analysis ofcomplex protein mixtures using isotope-coded affinity tags. Nat Biotechnol 17(10):994-999.

Gygi, S. P., Rochon, Y., Franza, B. R., Aebersold, R. (1999b). Correlation between protein and mRNA abundance in yeast. Mol Cell Biol 19(3):1720-1730.

263 Haendler, B., Toda, I., Sullivan, D. A., Schleuning, W. D. (1999). Expression oftranscripts for cysteine-rich secretory proteins (CRISPs) in the murine lacrimal gland. J Cell Physiol 178(3):371-378.

Haff, L. A., Smimov, I. P. (1997). Single-nucleotide polymorphism identification assays using a thermostable DNA polymerase and delayed extraction MALDI-TOF mass spectrometry. Genome Res 7(4):378-388.

Hage, D. S. (1999). Affinity chromatography: a review ofclinical applications. Clin Chem 45(5):593-615.

Hahner, S., Ludemann, H. C., Kirpekar, F., Nordhoff, E., Roepstorff, P., Galla, H.J., Hillenkamp, F. (1997). Matrix-assisted laser desorption/ionization mass spectrometry (MALDI) ofendonuclease digests ofRNA. Nucleic Acids Res 25(10):1957-1964.

Hakansson, P., Kamensky, I., Kjellberg, J., Sundqvist, B., Fohlman, J., Peterson, P.A. (1983). The observation ofquasi-molecular ions from a protein neurotoxin (MW 7821) using 1271-plasma desorption mass spectroscopy. Biochem Biophys Res Commun 110(2):519- 524.

Hanash, S. M., Strahler, J. R., Neel, J. V., Hailat, N., Melhem, R., Keim, D., Zhu, X. X., Wagner, D., Gage, D. A., Watson, J. T. (1991). Highly resolving two-dimensional gels for protein sequencing. Proc Natl Acad Sci US A 88(13):5709-5713.

Harvey, D. J. (1993). Quantitative aspects ofthe matrix-assisted laser desorption mass spectrometry ofcomplex oligosaccharides. Rapid Commun Mass Spectrom 7(7):614-619.

Harvey, D. J. (1996). Matrix-assisted laser desorption/ionisation mass spectrometry of oligosaccharides and glycoconjugates. J Chromatogr A 720(1-2):429-446.

Harvey, D. J., Bateman, R.H., Bordoli, R. S., Tyldesley, R. (2000). Ionisation and fragmentation ofcomplex glycans with a quadrupole time-of-flight mass spectrometer fitted with a matrix-assisted laser desorption/ionisation ion source. Rapid Commun Mass Spectrom 14(22):2135-2142.

Harvey, D. J., Kuster, B., Naven, T. J. (1998a). Perspectives in the glycosciences-matrix­ assisted laser desorption/ionization (MALDI) mass spectrometry ofcarbohydrates. Glycoconj J 15(4):333-338.

264 Harvey, M. D., Bandilla, D., Banlcs, P.R. (1998b). Subnanomolar detection limit for sodium dodecyl sulfate-capillary gel electrophoresis using a fluorogenic, noncovalent dye. Electrophoresis 19(12):2169-2174.

Hathaway, G. M. (1994). Characterization ofmodified and normal deoxyoligonuc/eotides by MALDI, time-of-flight mass spectrometry. Biotechniques 17(1):150-155.

Hayashi, K., Sueishi, K. (1988). Fibrinolytic activity and species ofplasminogen activator in human tears. Exp Eye Res 46(2):131-137.

Haynes, R. J., Tighe, P. J., Dua, H. S. (1999). Antimicrobial defensin peptides ofthe human ocular surface. Br J Ophthalmol 83(6):737-741.

Heine, G., Raid.a, M., Forssmann, W. G. (1997). Mapping ofpeptides and protein.fragments in human urine using liquid chromatography-mass spectrometry. J Chromatogr A 776(1):117-124.

Hensel, R.R., King, R. C., Owens, K. G. (1997). Electrospray sample preparation for improved quantitation in matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. Rapid Commun Mass Spectr6m 11(16):1785-1793.

Henzel, W. J., Billeci, T. M., Stults, J. T., Wong, S. C., Grimley, C., Watanabe, C. (1993). Identifying proteins from two-dimensional gels by molecular mass searching ofpeptide fragments in protein sequence databases. Proc Natl Acad Sci US A 90(11):5011-5015.

Holland, R. D., Duffy, C.R., Rafii, F., Sutherland, J.B., Heinze, T. M., Holder, C. L., Voorhees, K. J., Lay, J. 0., Jr. (1999). Identification ofbacterial proteins observed in MALDI TOF mass spectra from whole cells. Anal Chem 71(15):3226-3230.

Holstege, D. M., Puschner, B., Le, T. (2001). Determination ofgrayanotoxins in biological samples by LC-MSIMS. J Agric Food Chem 49(3):1648-1651.

Holzfeind, P., Merschak, P., Dieplinger, H., Redl, B. (1995). The human lacrimal gland synthesizes apolipoprotein D mRNA in addition to tear prealbumin mRNA, both species encoding members ofthe lipocalin superfamily. Exp Eye Res 61(4):495-500.

Horak, J., Werther, W., Schmid, E. R. (2001). Optimisation ofthe quantitative determination of chlormequat by matrix-assisted laser desorption/ionisation mass spectrometry. Rapid Commun Mass Spectrom 15(4):241-248.

265 Hunt, D. F., Shabanowitz, J., Yates, J. R., 3rd, Melver, R. T., Jr., Hunter, R. L., Syka, J.E., Amy, J. (1985). Tandem quadrupole-Fourier transform mass spectrometry of oligopeptides. Anal Chem 57(13):2728-2733.

Hunt, D. F., Shabanowitz, J., Yates, J. R., 3rd, Zhu, N. Z., Russell, D. H., Castro, M. E. (1987). Tandem quadrupole Fourier-transform mass spectrometry ofoligopeptides and small proteins. Proc Natl Acad Sci U SA 84(3):620-623.

Hunt, D. F., Yates, J. R., 3rd, Shabanowitz, J., Winston, S., Hauer, C.R. (1986). Protein sequencing by tandem mass spectrometry. Proc Natl Acad Sci US A 83(17):6233-6237. lkarashi, Y., Itoh, K., Maruyama, Y. (1991). Application ofFRJTfast atom bombardment liquid chromatography/mass spectrometry for the determination ofacetylcholine levels in rat brain regions. Biol Mass Spectrom 20(1):21-25. lkarashi, Y., Maruyama, Y. (1991). Applicability ofhigh-performance liquid chromatography­ continuous-flow fast atom bombardment mass spectrometry for simultaneous quantitation ofmultiple neurochemicals. J Chromatogr 587(2):306-313. lmanishi, J., Takahashi, F., Inatomi, A., Tagami, H., Yoshikawa,. T., Kondo, M. (1982). Complement levels in human tears. Jpn J Ophthalmol 26(2):229-233.

Inatomi, T., Spurr-Michaud, S., Tisdale, A. S., Gipson, I. K. (1995). Human corneal and conjunctiva/ epithelia express MUCJ mucin. Invest Ophthalmol Vis Sci 36(9):1818-1827.

Isemura, S., Saitoh, E., Sanada, K., Minakata, K. (1991). Identification offull-sized forms of salivary (S-type) cystatins (cystatin SN, cystatin SA, cystatin S, and two phosphorylated forms ofcystatin S) in human whole saliva and determination ofphosphorylation sites of cystatin S. J Biochem (Tokyo) 110(4):648-654.

Jacobs, A., Dahlman, 0. (2001). Enhancement ofthe quality ofMALDI mass spectra ofhighly acidic oligosaccharides by using a nafion-coated probe. Anal Chem 73(3):405-410.

Jacq, C., Liotet, S., Warnet, V. N., Arrata, M. (1982). Lactic deshydrogenase isoenzymes in the ocular tissues and liquids. Ophthalmologica 184(3):174-178.

James, P., Quadroni, M., Carafoli, E., Gonnet, G. (1993). Protein identification by mass profile fingerprinting. Biochem Biophys Res Commun 195(1):58-64.

266 Jan van Wijk, K. (2000). Proteomics ofthe chloroplast: experimentation and prediction. Trends Plant Sci S(l0):420-425.

Janssen, P. T., van Bijsterveld, 0. P. (1983a). Origin and biosynthesis ofhuman tear fluid proteins. Invest Ophthalmol Vis Sci 24(5):623-630.

Janssen, P. T., van Bijsterveld, 0. P. (1983b). The relations between tear fluid concentrations of lysozyme, tear-specific prealbumin and lactoferrin. Exp Eye Res 36(6):773- 779.

Jaquinod, M., Holtet, T. L., Etzerodt, M., Clemmensen, I., Thogersen, H. C., Roepstorff, P. (1999). Mass spectrometric characterisation ofpost-translational modification and genetic variation in human tetranectin. Biol Chem 380(11):1307-1314.

Jaquinod, M., Potier, N., Klarskov, K., Reymann, J.M., Sorokine, 0., Kieffer, S., Barth, P., Andriantomanga, V., Biellmann, J. F., Van Dorsselaer, A. (1993). Sequence ofpig lens a/dose reductase and electrospray mass spectrometry ofnon-covalent and covalent complexes. Eur J Biochem 218(3):893-903.

Jemal, M., Huang, M., Mao, Y., Whigan, D., Schuster, A. (2000). Liquid chromatographylelectrospray tandem mass' spectrometry method for the quantitation of fosinoprilat in human serum using automated 96-well solid-phase extraction for sample preparation. Rapid Commun Mass Spectrom 14(12):1023-1028.

Jemal, M., Mulvana, D. E. (2000). Liquid chromatographic-electrospray tandem mass spectrometric method for the simultaneous quantitation ofthe prodrugfosinopril and the active drugfosinoprilat in human serum. J Chromatogr B Biomed Sci Appl 739(2):255- 271.

Jensen, 0. L., Gluud, B. S., Birgens, H. S. (1986). The concentration oflactoferrin in tears of normals and ofdiabetics. Acta Ophthalmol (Copenh) 64(1):83-87.

Jensen, 0. N., Kulkarni, S., Aldrich, J. V., Barofsky, D. F. (1996). Characterization ofpeptide­ oligonucleotide heteroconjugates by mass spectrometry. Nucleic Acids Res 24(19):3866- 3872.

Jespersen, S., Chaurand, P., van Strien, F. J., Spengler, B., van der Greef, J. (1999). Direct sequencing ofneuropeptides in biological tissue by MALDI-PSD mass spectrometry. Anal Chem 71(3):660-666.

267 Jespersen, S., Niessen, W. M.A., Tjaden, U. R., van der Greef, J. (1995). Quantitative bioanalysis using matrix-assisted laser desorption/ionization mass spectrometry. J Mass Spectrom 30:357-364.

Ji, J., Chakraborty, A., Geng, M., Zhang, X., Amini, A., Bina, M., Regnier, F. (2000). Strategy for qualitative and quantitative analysis in proteomics based on signature peptides. J Chromatogr B Biomed Sci Appl 745(1):197-210.

Jiang, G., Vasanthan, T. (2000). MALDI-MS and HPLC quantification ofoligosaccharides of lichenase-hydrolyzed water-soluble beta-glucanfrom ten barley varieties. J Agric Food Chem 48(8):3305-3310.

Jonscher, K. R., Yates, J. R., 3rd (1997). The quadrupole ion trap mass spectrometer--a small solution to a big challenge. Anal Biochem 244(1):1-15.

Jonsson, A. P., Griffiths, W. J., Bratt, P., Johansson, I., Stromberg, N., Jomvall, H., Bergman, T. (2000). A novel Ser O-glucuronidation in acidic proline-rich proteins identified by tandem mass spectrometry. FEBS Lett 475(2):131-134.

Jonsson, G., Hedin, A., Hakansson, P., Sundqvist; B. U. (1988). Competition between protein/protein interactions and protein/substrate interactions studied by plasma desorption mass spectrometry. Rapid Commun Mass Spectrom 2(8):154-156.

Juhasz, P., Costello, C. E., Biemann, K. (1993). Matrix-assisted laser desorption ionization mass spectrometry with 2-(4-hydroxyphenylazo)-benzoic acid matrix. J Am Soc Mass Spectrom 4:399-409.

Juhasz, P., Roskey, M. T., Smimov, I. P., Haff, L. A., Vestal, M. L., Martin, S. A. (1996). Applications ofdelayed extraction matrix-assisted laser desorption ionization time-of­ flight mass spectrometry to oligonucleotide analysis. Anal Chem 68(6):941-946.

Jumblatt, M. M., McKenzie, R. W., Jumblatt, J.E. (1999). MUC5AC mucin is a component of the human precorneal tear film. Invest Ophthalmol Vis Sci 40(1):43-49.

Kahan, I. L., Ottovay, E. (1975). Lactate dehydrogenase oftears and corneal epithelium. Exp Eye Res 20(2):129-133.

Kamo, M., Kawakami, T., Tsugita, A. (1998). Novel detection methods for modification sites using chemical cleavages and mass spectrometry. J Protein Chem 17(6):511.

268 Kanazawa, K., Ashida, K., Itoh, M., Nagai, H., Sasaki, H., Fukuda, M. (1999). Establishment of a method for mapping ofN-linked oligosaccharides and its use to analyze industrially produced recombinant erythropoietin. Biol Phann Bull 22(4):339-346.

Karas, M., Bahr, U., Dulcks, T. (2000a). Nano-electrospray ionization mass spectrometry: addressing analytical problems beyond routine. Fresenius J Anal Chem 366(6- 7):669- 676.

Karas, M., Gluckmann, M., Schafer, J. (2000b). Ionization in matrix-assisted laser desorption/ionization: singly charged molecular ions are the lucky survivors. J Mass Spectrom 35(1):1-12.

Karas, M., Hillenkamp, F. (1988). Laser desorption ionization ofproteins with molecular masses exceeding 10,000 daltons. Anal Chem 60(20):2299-2301.

Kato, K., Jingu, S., Ogawa, N., Higuchi, S. (2000a). Determination ofpibutidine metabolites in human plasma by LC-MS/MS. J Pharm Biomed Anal 24(2):237-249.

Kato, K., Jingu, S., Ogawa, N., Higuchi, S. (2000b). Development and validation ofa liquid chromatographic-tandem mass spectrometric method for the determination ofpibutidine in human urine. J Chromatogr B Biomed Sci Appl 740(2):187-193.

Kaufmann, R., Spengler, B., Lutzenkirchen, F. (1993). Mass spectrometric sequencing oflinear peptides by product-ion analysis in a rejlectron time-of-flight mass spectrometer using matrix-assisted laser desorption ionization. Rapid Commun Mass Spectrom 7(10):902- 910.

Kazmaier, T., Roth, S., Zapp, J., Harding, M., Kuhn, R. (1998). Quantitative analysis ofmalto­ oligosaccharides by MALDI-TOF mass spectrometry, capillary electrophoresis and anion exchange chromatography. Fres J Anal Chem 361(5):473-478.

Kebarle, P. (2000). A briefoverview ofthe present status ofthe mechanisms involved in electrospray mass spectrometry. J Mass Spectrom 35(7):804-817.

Kellner, R. (2000). Proteomics. Concepts and perspectives. Fresenius J Anal Chem 366(6- 7):517-524.

Keough, T., Lacey, M. P., Fieno, A. M., Grant, R. A., Sun, Y., Bauer, M. D., Begley, K. B. (2000). Tandem mass spectrometry methods for definitive protein identification in proteomics research. Electrophoresis 21(11):2252-2265.

269 Khalil, H. A., de Keizer, R. J., Kijlstra, A. (1988). Analysis oftear proteins in Graves' ophthalmopathy by high performance liquid chromatography. Am J Ophthalmol 106(2): 186-190.

Khan, z., Smyth, D. G. (1993). Isolation and identification ofN-terminally extended forms of5- oxoprolylglutamylprolinamide (Glp-Glu-Pro-NH2), a thyrotropin-releasing-hormone (TRH)-like peptide present in human semen. Eur J Biochem 212(1 ):35-40.

Kijlstra, A., Jeurissen, S. H., Koning, K. M. (1983). Lactoferrin levels in normal human tears. Br J Ophthalmol 67(3):199-202.

Kijlstra, A., Kuizenga, A., van der Velde, M., van Haeringen, N. J. (1989). Gel electrophoresis ofhuman tears reveals various forms oftear lactoferrin. Curr Eye Res 8(6):581-588.

Kikuchi, M., Sato, M., Ito, T., Honda, M. (2001). Application ofa new analytical method using gas chromatography and gas chromatography-mass spectrometry for the azide ion to human blood and urine samples ofan actual case. J Chromatogr B Biomed Sci Appl 752(1):149-157.

Kinsel, G. R., Edmondson, R. D., Russell, D. H. (1997). Profile andflight time analysis of bovine insulin clusters as a probe ofmatrix-assisted laser desorption/ionization ion formation dynamics. J Mass Spectrom 32(7):714-722.

K.irpekar, F., Krogh, T. N. (2001). RNAfragmentation studied in a matrix-assisted laser desorption/ionisation tandem quadrupole/orthogonal time-of-flight mass spectrometer. Rapid Commun Mass Spectrom 15(1):8-14.

Kise, H., Nishioka, J., Kawamura, J., Suzuki, K. (1996). Characterization ofsemenogelin II and its molecular interaction with prostate-specific antigen and protein C inhibitor. Eur J Biochem 238(1):88-96.

Kiselar, J. G., Downard, K. M. (2000). Preservation and detection ofspecific antibody-peptide complexes by matrix-assisted laser desorption ionization mass spectrometry. J Am Soc Mass Spectrom 11(8):746-750.

Kissmeyer, A. M., Sonne, K., Binderup, E. (2000). Determination ofthe vitamin D analog EB 1089 (seocalcitol) in human and pig serum using liquid chromatography-tandem mass spectrometry. J Chromatogr B Biomed Sci Appl 740(1):117-128.

270 K.itaoka, M., Nakazawa, M., Hayasaka, S. (1985). Lysosomal enzymes in human tear fluid collected by filter paper strips. Exp Eye Res 41(3):259-265.

Klose, J., Kobalz, U. (1995). Two-dimensional electrophoresis ofproteins: an updated protocol and implications for a.functional analysis ofthe genome. Electrophoresis 16(6):1034- 1059.

Kobata, A. (1994). Affinity chromatography with use ofimmobilized lectin columns. Biochem Soc Trans 22(2):360-364.

Krause, J., Stoeckli, M., Schlunegger, U. P. (1996). Studies on the selection ofnew matrices for ultraviolet matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. Rapid Commun Mass Spectrom 10(15):1927-1933.

Kriegel, K., Seefeldt, I., Hoffmann, F., Schultz, C., Wenk, C., Regitz-Zagrosek, V., Oswald, H., Fleck, E. (2000). An alternative approach to deal with geometric uncertainties in computer analysis oftwo-dimensional electrophoresis gels. Electrophoresis 21(13):2637- 2640.

Kristensen, D. B., Imamura, K., Miyamoto, Y., Yoshizato, K. (2000). Mass spectrometric approaches for the characterization ofproteins on a hybrid quadrupole time-of-flight (Q­ TOF) mass spectrometer. Electrophoresis 21(2):430-439.

Krull, I. S., Strong, R., Sosic, Z., Cho, B. Y., Beale, S. C., Wang, C. C., Cohen, S. (1997). Labeling reactions applicable to chromatography and electrophoresis ofminute amounts ofproteins. J Chromatogr B Biomed Sci Appl 699(1-2): 173-208.

Krutchinsky, A. N., Zhang, W., Chait, B. T. (2000). Rapidly switchable matrix-assisted laser desorption/ionization and electrospray quadrupole-time-of-flight mass spectrometry for protein identification. J Am Soc Mass Spectrom 11(6):493-504.

Kuizenga, A., van Haeringen, N. J., Kijlstra, A. (1991a). Identification oflectin binding proteins in human tears. Invest Ophthalmol Vis Sci 32(13):3277-3284.

Kuizenga, A., van Haeringen, N. J., Kijlstra, A. (1991b). SDS-Minigel electrophoresis ofhuman tears. Effect ofsample treatment on protein patterns. Invest Ophthalmol Vis Sci 32(2):381-386.

271 Kusmierz, J. J., Sumrada, R., Desiderio, D. M. (1990). Fast atom bombardment mass spectrometric quantitative analysis ofmethionine-enkephalin in human pituitary tissues. Anal Chem 62(21):2395-2400.

Kuster, B., Wheeler, S. F., Hunter, A. P., Dwek, R. A., Harvey, D. J. (1997). SequencingofN­ linked oligosaccharides directly from protein gels: in-gel deg/ycosy/ationfollowed by matrix-assisted laser desorption/ionization mass spectrometry and normal-phase high­ performance liquid chromatography. Anal Biochem 250(1):82-101.

Lahm, H. W., Langen, H. (2000). Mass spectrometry: a tool for the identification ofproteins separated by gels. Electrophoresis 21(11):2105-2114.

Lake, D. A., Johnson, M. V., McEwen, C. N., Larsen, B. S. (2000). Sample preparation for high throughput accurate mass analysis by matrix-assisted laser desorption/ionization time-of­ .flight mass spectrometry. Rapid Commun Mass Spectrom 14(11):1008-1013.

Langley, G. J., Herniman, J.M., Davies, N. L., Brown, T. (1999). Simplified sample preparation for the analysis ofoligonuc/eotides by matrix-assisted laser desorption/ionisation time-of-flight mass spectrometry. Rapid Commun Mass Spectrom 13(17):1717-1723.

Larsen, M. R., Roepstorff, P. (2000). Mass spectrometric identification ofproteins and characterization oftheir post-translational modifications in proteome analysis. Fresenius J Anal Chem366(6-7):677-690.

Lass, J. H., Walter, E. I., Burris, T. E., Grossniklaus, H. E., Roat, M. I., Skelnik, D. L., Needham, L., Singer, M., Medof, M. E. (1990). Expression oftwo molecular forms ofthe complement decay-accelerating/actor in the eye and /acrimal gland. Invest Ophthalmol Vis Sci 31(6):1136-1148.

Lau, B. P., Scott, P. M., Lewis, D. A., Kanhere, S. R. (2000). Quantitative determination of ochratoxin A by liquid chromatographylelectrospray tandem mass spectrometry. J Mass Spectrom 35(1):23-32.

Lauber, W. M., Carroll, J. A., Dufield, D.R., Kiesel, J. R., Radabaugh, M. R., Malone, J.P. (2001). Mass spectrometry compatibility oftwo-dimensional gel protein stains. Electrophoresis 22(5):906-918.

272 Lee, H., Lubman, D. M. (1995). Sequence-specific fragmentation generated by matrix-assisted laser desorption/ionization in a quadrupole ion trap/reflectron time-of-flight device. Anal Chem 67(8):1400-1408.

Lee, M. S., Kerns, E. H. (1999). LCIMS applications in drug development. Mass Spectrom Rev 18(3-4): 187-279.

Lee, S. Y., Ubels, J. L., Soprano, D. R. (1992). The lacrimal gland synthesizes retinol-binding protein. Exp Eye Res 55(1 ): 163-171.

Lehrer, M. (1998). The role ofgas chromatography/mass spectrometry. Instrumental techniques in forensic urine drug testing. Clin Lab Med 18(4):631-649.

Lehrer, R. I., Xu, G., Abduragimov, A., Dinh, N. N., Qu, X. D., Martin, D., Glasgow, B. J. (1998). Lipophilin, a novel heterodimeric protein ofhuman tears. FEBS Lett 432(3):163- 167.

Leinonen, J., Zhang, W. M., Stenman, U. H. (1996). Complexformation betweenPSA isoenzymes and protease inhibitors. J Urol 155(3):1099-1103.

Lemoine, J., Chirat, F., Dornon, B. (1996). Structural analysis ofderivatized oligosaccharides using post-source decay matrix-assisted laser desorption/ionization mass spectrometry. J Mass Spectrom 31(8):908-912.

Leonardi, A., Borghesan, F., Faggian, D., Secchi, A., Plebani, M. (1995). Eosinophil cationic protein in tears ofnormal subjects and patients affected by vernal keratoconjunctivitis. Allergy 50(7):610-613.

LeRiche, T., Osterodt, J., Volmer, D. A. (2001). An experimental comparison ofelectrospray ion-trap and matrix-assisted laser desorption/ionization post-source decay mass spectra for the characterization ofsmall drug molecules. Rapid Commun Mass Spectrom 15(8):608-614.

Lewis, T. S., Hunt, J.B., Aveline, L. D., Jonscher, K. R., Louie, D. F., Yeh, J.M., Nahreini, T. S., Resing, K. A., Ahn, N. G. (2000). Identification ofnovel MAP kinase pathway signaling targets by functional proteomics and mass spectrometry. Mol Cell 6(6):1343- 1354.

Lhoest, G. J., de Jong, A. P., Meiring, H. D., Maton, N., Latinne, D., Verbeeck, R. K., Otte, J. B., Zurini, M. ( 1998). Isolation, identification and immunosuppressive activity ofa new

273 IMM-125 metabolite.from human liver microsomes. Identification ofits cyclophilin A­ IMM-125 metabolite complex by nanospray tandem mass spectrometry. J Mass Spectrom 33(10):936-942.

Li, L., Garden, R. W., Romanova, E. V., Sweedler, J. V. (1999). In situ sequencing ofpeptides from biological tissues and single cells using MALDI-PSDICID analysis. Anal Chem 71(24):5451-5458.

Li, L., Garden, R. W., Sweedler, J. V. (2000a). Single-cell MALDI: a new tool for direct peptide pro.filing. Trends Biotechnol 18(4):151-160.

Li, Q., Weng, J., Mohan, R.R., Bennett, G. L., Schwall, R., Wang, Z. F., Tabor, K., Kim, J., Hargrave, S., Cuevas, K. H., Wilson, S. E. (1996). Hepatocyte growth.factor and hepatocyte growth.factor receptor in the /acrimal gland, tears, and cornea. Invest Ophthalmol Vis Sci 37(5):727-739.

Li, S., Dass, C. (1999). Iron(III)-immobilized metal ion affinity chromatography and mass spectrometry for the purification and characterization ofsynthetic phosphopeptides. Anal Biochem 270(1 ):9-14.

Li, Y., Li, M., Xing, G., Hu, Z., Wang, Q., Dong, C., Wei, H., Fan, G., Chen, J., Yang, X., Zhao, S., Chen, H., Guan, K., Wu, C., Zhang, C., He, F. (2000b). Stimulation ofthe mitogen-activated protein ldnase cascade and tyrosine phosphorylation of the epidermal growth.factor receptor by hepatopoietin. J Biol Chem 275(48):37443-37447.

Lilja, H., Christensson, A., Dahlen, U., Matikainen, M. T., Nilsson, 0., Pettersson, K., Lovgren, T. (1991). Prostate-specific antigen in serum occurs predominantly in complex with alpha 1-antichymotrypsin. Clin Chem 37(9):1618-1625.

Lin, S., Cotter, R. J., Woods, A. S. (1998). Detection ofnon-covalent interaction ofsingle and double stranded DNA with peptides by MALDI-TOF. Proteins Suppl 2: 12-21.

Ling, Y. C., Lin, L., Chen, Y. T. (1998). Quantitative analysis ofantibiotics by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. Rapid Commun Mass Spectrom 12(6):317-327.

Link, A. J., Eng, J., Schieltz, D. M., Carmack, E., Mize, G. J., Morris, D. R., Garvik, B. M., Yates, J. R., 3rd (1999). Direct analysis ofprotein complexes using mass spectrometry. Nat Biotechnol 17(7):676-682.

274 Lisek, C. A., Bailey, J.E., Benson, L. M., Yaksh, T. L., Jardine, I. (1989). Quantitation of endogenous substance P by on-line microcolumn liquid chromatography/continuous-flow fast-atom bombardment mass spectrometry. Rapid Commun Mass Spectrom 3(2):43-46.

Little, J.M., Centifanto, Y. M., Kaufman, H. E. (1969). Immunoglobulins in human tears. Am J Ophthalmol 68(5):898-905.

Liu, Y. H., Bai, J., Liang, X., Lubman, D. M., Venta, P. J. (1995). Use ofa nitrocellulose film substrate in matrix-assisted laser desorption/ionization mass spectrometry for DNA mapping and screening. Anal Chem 67(19):3482-3490.

Loboda, A. V., Krutchinsky, A. N., Bromirski, M., Ens, W., Standing, K. G. (2000). A tandem quadrupole/time-of-flight mass spectrometer with a matrix-assisted laser desorption/ionization source: design and performance. Rapid Commun Mass Spectrom 14(12):1047-1057.

Loo, J. A., Brown, J., Critchley, G., Mitchell, C., Andrews, P. C., Ogorzalek Loo, R.R. (1999). High sensitivity mass spectrometric methods for obtaining intact molecular weights from gel-separated proteins. Electrophoresis 20(4-5):743-748.

Lopez, M. F. (1999). Proteome analysis. I Gene products are where the biologi,cal action is. J Chromatogr B Biomed Sci Appl 722(1-2):191-202.

Lopez, M. F., Berggren, K., Chemokalskaya, E., Lazarev, A., Robinson, M., Patton, W. F. (2000). A comparison ofsilver stain and SYPRO Ruby Protein Gel Stain with respect to protein detection in two-dimensional gels and identification by peptide mass profiling. Electrophoresis 21(17):3673-3683.

Lynn, E. C., Chung, M. C., Tsai, W. C., Han, C. C. (1999). Identification ofEnterobacteriaceae bacteria by direct matrix-assisted laser desorptiomlionization mass spectrometric analysis ofwhole cells. Rapid Commun Mass Spectrom 13(20):2022-2027.

Ma, Y., Lu, Y., Zeng, H., Ron, D., Mo, W., Neubert, T. A. (2001). Characterization of phosphopeptides from protein digests using matrix-assisted laser desorption/ionization time-of-flight mass spectrometry and nanoelectrospray quadrupole time-of-flight mass spectrometry. Rapid Commun Mass Spectrom 15(18):1693-1700.

Macek, B., Hofsteenge, J., Peter-Katalinic, J. (2001 ). Direct determination ofglycosylation sites in O-facosylated glycopeptides using nano-electrospray quadrupole time-of-flight mass spectrometry. Rapid Commun Mass Spectrom 15(10):771-777.

275 Macfarlane, R. D., Torgerson, D. F. (1976). Californium-252 plasma desorption mass spectroscopy. Science 191(4230):920-925.

Malm, J., Hellman, J., Magnusson, H., Laurell, C. B., Lilja, H. (1996). Isolation and characterization ofthe major gel proteins in human semen, semenogelin I and semenogelin II. Eur J Biochem 238(1):48-53.

Malm, J., Sorensen, 0., Persson, T., Frohm-Nilsson, M., Johansson, B., Bjartell, A, Lilja, H., Stahle-Backdahl, M., Borregaard, N., Egesten, A (2000). The human cationic antimicrobial protein (hCAP-18) is expressed in the epithelium ofhuman epididymis, is present in seminal plasma at high concentrations, and is attached to spermatozoa. Infect Immun 68(7):4297-4302.

Mamyrin, B. A, Karataev, V. I., Shmikk, D. V., Zagulin, V. A (1973). The mass reflectron, a new nonmagnetic time-of-flight mass spectrometer with high resolution. Sov Phys JETP 37:45.

Mann, M., Hojrup, P., Roepstorff, P. (1993). Use ofmass spectrometric molecular weight information to identify proteins in sequence databases. Biol Mass Spectrom 22(6):338- 345.

Mann, M., Wilm, M. (1994). Error-tolerant identification ofpeptides in sequence databases by peptide sequence tags. Anal Chem 66(24):4390-4399.

March, R. E. (1997). An introduction to quadrupole ion trap mass spectrometry. J Mass Spectrom 32:351-369.

Marie, A, Fournier, F., Tabet, J. C. (2000). Characterization ofsynthetic polymers by MALDI­ TOFIMS: investigation into new methods ofsample target preparation and consequence on mass spectrumfinger print. Anal Chem 72(20):5106-5114.

Markusse, H. M., Huysen, J.C., Nieuwenhuys, E. J., Swaak, A J. (1992). Beta 2 microglobulin in tear fluid from patients with primary Sjogren's syndrome. Ann Rheum Dis 51(4):503- 505.

Markusse, H. M., van Haeringen, N. J., Swaak, A J., Hogeweg, M., de Jong, P. T. (1993). Tear fluid analysis in primary Sjogren's syndrome. Clin Exp Rheumatol 11(2):175-178.

276 Masters, C. F., Markey, S. P., Mefford, I. N., Duncan, M. W. (1988). Separation ofdeuteriated isotopomers ofdopamine by ion-pair reversed-phase high-performance liquid chromatography. Anal Chem 60(19):2131-2134.

Maurer, H. H. (1998). Liquid chromatography-mass spectrometry in forensic and clinical toxicology. J Chromatogr B Biomed Sci Appl 713(1):3-25.

Mavra, M., Thompson, E. J., Nikolic, J., Krunic, A., Ranin, J., Levie, Z., Keir, G., Luxton, R., Youl, B. D. (1990). The occurrence ofoligoclonal JgG in tears from patients with MS and systemic immune disorders. Neurology 40(8):1259-1262.

McClellan, B. H., Whitney, C.R., Newman, L. P., Allansmith, M. R. (1973). Jmmunoglobulins in tears. Am J Ophthalmol 76(1):89-101.

McComb, M. E., Oleschuk, R. D., Manley, D. M., Donald, L., Chow, A., O'Neil, J. D., Ens, W., Standing, K. G., Perreault, H. (1997). Use ofa non-porous polyurethane membrane as a sample support for matrix-assisted laser desorption/ionization time-of-flight mass spectrometry ofpeptides and proteins. Rapid Commun Mass Spectrom 11(15):1716-1722.

McCormack, A. L., Schieltz, D. M., Goode, B., Yang, S., Barnes, G., Drubin, D., Yates, J. R., 3rd (1997). Direct analysis and identification ofproteins in mixtures by LC/MSIMS and database searching at the low-femtomole level. Anal Chem 69(4):767-776.

McGinley, M. D., Davis, M. T., Robinson, J. H., Spahr, C. S., Bures, E. J., Beierle, J., Mort, J., Patterson, S. D. (2000). A simplified device for protein identification by microcapillary gradient liquid chromatography-tandem mass spectrometry. Electrophoresis 21(9):1678- 1684.

Mciver, R. T ., Jr., Li, Y., Hunter, R. L. (1994). Matrix-assisted laser desorption/ionization with an external ion source Fourier-transform mass spectrometer. Rapid Commun Mass Spectrom 8(3):237-241.

McKenzie, R. W., Jumblatt, J. E., Jumblatt, M. M. (2000). Quantification ofMUC2 and MUC5AC transcripts in human conjunctiva. Invest Ophthalmol Vis Sci 41(3):703-708.

Medzihradszky, K. F., Campbell, J.M., Baldwin, M.A., Falick, A. M., Juhasz, P., Vestal, M. L., Burlingame, A. L. (2000). The characteristics ofpeptide collision-induced dissociation using a high-performance MALDI-TOFITOF tandem mass spectrometer. Anal Chem 72(3):552-558.

277 Medzihradszky, K. F., Leffler, H., Baldwin, M.A., Burlingame, A. L. (2001). Protein identification by in-gel digestion, high-performance liquid chromatography, and mass spectrometry: peptide analysis by complementary ionization techniques. J Am Soc Mass Spectrom 12(2):215-221.

Mertaniemi, P., Ylatupa, S., Partanen, P., Tervo, T. (1995). Increased release of immunoreactive ca/citonin gene-related peptide (CGRP) in tears after excimer laser keratectomy. Exp Eye Res 60(6):659-665.

Mii, S., Nakamura, K., Takeo, K., Kurimoto, S. (1992). Analysis ofhuman tear proteins by two­ dimensional electrophoresis. Electrophoresis 13(6):379-382.

Miketova, P., Schram, K. H. (1997). Mass spectrometry ofnucleotides and o/igonucleotides. Mol Biotechnol 8(3):249-253.

Miketova, P., Schram, K. H., Whitney, J., Li, M., Huang, R., Kerns, E., Valcic, S., Timmermann, B. N., Rourick, R., Klohr, S. (2000). Tandem mass spectrometry studies of green tea catechins. Identification ofthree minor components in the polypheno/ic extract ofgreen tea. J Mass Spectrom 35(7):860-869.

Miliotis, T., Kjellstrom, S., Onnerfjord, P., Nilsson, J., Laurell, T., Edholm, L. E., Marko-Varga, G. (2000). Protein identification platform utilizing micro dispensing technology interfaced to matrix-assisted laser desorption ionization time-of-flight mass spectrometry. J Chromatogr A 886(1-2):99-110.

Mills, P. B., Mills, K., Johnson, A. W., Clayton, P. T., Winchester, B. G. (2001). Analysis by matrix assisted laser desorption/ionisation-time offlight mass spectrometry ofthe post­ translational modifications ofalpha 1-antitrypsin isoforms separated by two-dimensional polyacry/amide gel electrophoresis. Proteomics 1(6):778-786.

Mirgorodskaya, 0. A., Kozmin, Y. P., Titov, M. I., Komer, R., Sonksen, C. P., Roepstorff, P. (2000). Quantitation ofpeptides and proteins by matrix-assisted laser desorption/ionization mass spectrometry using (18)0-labe/ed internal standards. Rapid Commun Mass Spectrom 14(14):1226-1232.

Mirza, U. A., Liu, Y. H., Tang, J. T., Porter, F., Bondoc, L., Chen, G., Pramanik, B. N., Nagabhushan, T. L. (2000). Extraction and characterization ofadenovirus proteins from sodium dodecylsulfate polyacrylamide gel electrophoresis by matrix-assisted laser desorption/ionization mass spectrometry. J Am Soc Mass Spectrom 11(4):356-361.

278 Miyazaki, A., Nakanishi, T., Kishikawa, M., Shimizu, A., Ohba, Y., Tanaka, A., Sasaki, H. (1996). Post-translational modification from to aspartic acid-residue on a variant hemoglobin, Hb Bristol, a proofby ESI-MS-MS. J Mass Spectrom 31(11):1311- 1313.

Mizuno, Y., Sasagawa, T., Dohmae, N., Takio, K. (1999). An automated interpretation of MALDIITOF postsource decay spectra ofoligosaccharides. 1. Automated peak assignment. Anal Chem 71(20):4764-4771.

Mo, W., Takao, T., Sakamoto, H., Shimonishi, Y. (1998). Structural analysis of oligosaccharides derivatized with 4-aminobenzoic acid 2-(diethylamino)ethyl ester by matrix-assisted laser desorption/ionization mass spectrometry. Anal Chem 70(21):4520- 4526.

Mo, W., Takao, T., Shimonishi, Y. (1997). Accurate peptide sequencing by post-source decay matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. Rapid Commun Mass Spectrom 11(17): 1829-1834.

Molloy, M. P., Bolis, S., Herbert, B. R., Ou, K., Tyler, M. I., vanDyk, D. D., Willcox, M. D., Gooley, A. A., Williams, K. L., Morris, C. A., Walsh, B. J. (1997). Establishment ofthe human reflex tear two-dimensional polyacrylamide gel electrophoresis reference map: new proteins ofpotential diagnostic value. Electrophoresis 18(15):2811-2815.

Mondino, B. J., Zaidman, G. W. (1983). Hemolytic complement in tears. Ophthalmic Res 15(4):208-211.

Morris, H. R., Paxton, T., Panico, M., McDowell, R., Dell, A. (1997). A novel geometry mass spectrometer, the Q-TOF, for low-femtomole/attomole-range biopolymer sequencing. J Protein Chem 16(5):469-479.

Muck, W. (1999). Quantitative analysis ofpharmacokinetic study samples by liquid chromatography coupled to tandem mass spectrometry (LC-MSIMS). Phannazie 54(9):639-644.

Muddiman, D. C., Gusev, A. I., Martin, L. B., Hercules, D. M. (1996). Direct quantification of cocaine in urine by time-of-flight mass spectrometry. Fresenius Journal of Analytical Chemistry 354(1):103-110.

279 Muddiman, D. C., Gusev, A. I., Proctor, A., Hercules, D. M., Venkataramanan, R., Diven, W. (1994). Quantitative measurement ofcyclosporin A in blood by time-of-flight mass spectrometry. Anal Chem 66(14):2362-2368.

Muddiman, D. C., Gusev, A. I., Stoppeklangner, K., Proctor, A., Hercules, D. M., Tata, P., Venkataramanan, R., Diven, W. (1995). Simultaneous quantification ofcyclosporin A and its major metabolites by time-of-flight second~ry-ion mass spectrometry and matrix­ assisted laser desorption/ionization mass spectrometry utilizing data analysis techniques - comparison with high-performance liquid chromatography. Journal of Mass Spectrometry 30(10):1469-1479.

Munchbach, M., Quadroni, M., Miotto, G., James, P. (2000). Quantitation and facilitated de novo sequencing ofproteins by isotopic N-terminal /abeling ofpeptides with a .fragmentation-directing moiety. Anal Chem 72(17):4047-4057.

Nelson, R. W., Krone, J. R., Bieber, A. L., Williams, P. (1995). Mass spectrometric immunoassay. Analytical Chemistry 67(7):1153-1158.

Nelson, R. W., McLean, M.A., Hutchens, T. W. (1994). Quantitative determination ofproteins by matrix-assisted laser desorption ionization time-of-flight mass spectrometry. Analytical Chemistry 66(9):1408-1415.

Neubauer, G., Mann, M. (1999). Mapping ofphosphorylation sites ofgel-isolated proteins by nanoelectrospray tandem mass spectrometry: potentials and limitations. Anal Chem 71(1 ):235-242.

Nevalainen, T. J. (1993). Serum phospholipases A2 in inflammatory diseases. Clin Chem 39(12):2453-2459.

Newsholme, S. J., Maleeff, B. F., Steiner, S., Anderson, N. L., Schwartz, L. W. (2000). Two­ dimensional electrophoresis ofliver proteins: characterization ofa drug-induced hepatomegaly in rats. Electrophoresis 21(11):2122-2128.

Nexo, E., Engbaek, F., Deland, P. M., Westby, C., O'Gorman, P., Johnston, C., Kase, B. F., Guttormsen, A. B., Alfheim, I., McPartlin, J., Smith, D., Moller, J., Rasmussen, K., Clarke, R., Scott, J.M., Refsum, H. (2000). Evaluation ofnovel assays in clinical chemistry: quantification ofplasma total homocysteine. Clin Chem 46(8 Pt 1): 1150-1156.

Nguyen, D. H., Beuerman, R. W., Thompson, H. W., DiLoreto, D. A. (1997). Growth/actor and neurotrophicfactor mRNA in human /acrimal gland. Cornea 16(2):192-199.

280 Nguyen, D. N., Becker, G. W., Riggin, R. M. (1995). Protein mass spectrometry: applications to analytical biotechnology. J Chromatogr A 705(1):21-45.

Nicola, A. J., Gusev, A. I., Proctor, A., Jackson, E. K., Hercules, D. M. (1995). Application of the fast-evaporation sample preparation method for improving quantification of angiotensin II by matrix-assisted laser desorption/ionization. Rapid Commun Mass Spectrom 9(12):1164-1171.

Nielsen, P. F., Roepstorff, P. (1989). Suppression effects in peptide mapping by plasma desorption mass spectrometry. Biomed Environ Mass Spectrom 18(2):131-137.

Nielsen, P. F., Roepstorff, P., Clausen, I. G., Jensen, E. B., Jonassen, I., Svendsen, A., Balschmidt, P., Hansen, F. B. (1989). Plasma desorption mass spectrometry, an analytical tool in protein engineering: characterization ofmodified insulins. Protein Eng 2(6):449-457.

Niessen, W. M. (1998). Advances in instrumentation in liquid chromatography-mass spectrometry and related liquid-introduction techniques. J Chromatogr A 794(1-2):407- 435.

Nishida, K., Kawasaki, S., Adachi, W., Kinoshita, S. (1996). Apolipoprotein J expression in human ocular surface epithelium. Invest Ophthalmol Vis Sci 37(11 ):2285-2292.

Nishida, K., Sotozono, C., Adachi, W., Yamamoto, S., Yokoi, N., Kinoshita, S. (1995). Transforming growth factor-beta 1, -beta 2 and-beta 3 mRNA expression in human cornea. Curr Eye Res 14(3):235-241.

Niwa, T. (1986). Metabolic pro.filing with gas chromatography-mass spectrometry and its application to clinical medicine. J Chromatogr 379:313-345.

Nonami, H., Tanaka, K., Fukuyama, Y., Erra-Balsells, R. (1998). beta-Carboline alkaloids as matrices for UV-matrix-assisted laser desorption/ionization time-of-flight mass spectrometry in positive and negative ion modes. Analysis ofproteins ofhigh molecular mass, and ofcyclic and acyclic oligosaccharides. Rapid Commun Mass Spectrom 12(6):285-296.

Nordhoff, E., lngendoh, A., Cramer, R., Overberg, A., Stahl, B., Karas, M., Hillenkamp, F., Crain, P. F. (1992). Matrix-assisted laser desorption/ionization mass spectrometry of nucleic acids with wavelengths in the ultraviolet and infrared. Rapid Commun Mass Spectrom 6(12):771-776.

281 Nordhoff, E., Kirpekar, F., Karas, M., Cramer, R., Hahner, S., Hillenkamp, F., Kristiansen, K., Roepstroff, P., Lezius, A. (1994). Comparison oflR- and UV-matrix-assisted laser desorption/ionization mass spectrometry ofoligodeoxynucleotides. Nucleic Acids Res 22(13):2460-2465.

Oda, Y., Huang, K., Cross, F. R., Cowbum, D., Chait, B. T. (1999). Accurate quantitation of protein expression and site-specific phosphorylation. Proc Natl Acad Sci U S A 96( 12):6591-6596.

Oda, Y., Nagasu, T., Chait, B. T. (2001 ). Enrichment analysis ofphosphorylated proteins as a tool for probing the phosphoproteome. Nat Biotechnol 19(4):379-382.

Odum, L., Johnsen, A. H. (1994). Human seminal plasma contains somatostatin-64. Biochem J 303(Pt 1):263-268.

O'Farrell, P.H. (1975). High resolution two-dimensional electrophoresis ofproteins. J Biol Chem 250(10):4007-4021.

Ogorzalek Loo, R.R., Mitchell, C., Stevenson, T.1., Martin, S. A., Hines, W. M., Juhasz, P., Patterson, D. H., Peltier, J.M., Loo, J. A., Andrews, P. C. (1997). Sensitivity and mass accuracy for proteins analyzed directly from polyacrylamide gels: implications for proteome mapping. Electrophoresis 18(3-4):382-390.

Ogueta, S., Rogado, R., Marina, A., Moreno, F., Redondo, J.M., Vazquez, J. (2000). Identification ofphosphorylation sites in proteins by nanospray quadrupole ion trap mass spectrometry. J Mass Spectrom 35(4):556-565.

Okamoto, M., Takahashi, K., Doi, T., Takimoto, Y. (1997). High-sensitivity detection and postsource decay of2-aminopyridine-derivatized oligosaccharides with matrix-assisted laser desorption/ionization mass spectrometry. Anal Chem 69(15):2919-2926.

O'Mahony, 0. A., Djahanbahkch, 0., Mahmood, T., Puddefoot, J. R., Vinson, G. P. (2000). Angiotensin II in human seminal fluid. Hum Reprod 15(6): 1345-1349.

Opiteck, G. J., Lewis, K. C., Jorgenson, J. W., Anderegg, R. J. (1997). Comprehensive on-line LCILC/MS ofproteins. Anal Chem 69(8):1518-1524.

Ostergaard, M., Wolf, H., Omtoft, T. F., Celis, J.E. (1999). Psoriasin (SJ00A7): a putative urinary marker for the follow-up ofpatients with bladder squamous cell carcinomas. Electrophoresis 20(2):349-354.

282 Owens, D. R., Bothner, B., Phung, Q., Harris, K., Siuzdak, G. (1998). Aspects of oligonuc/eotide and peptide sequencing with MALDI and electrospray mass spectrometry. Bioorg Med Chem 6(9):1547-1554.

Page, M. J., Amess, B., Townsend, R. R., Parekh, R., Herath, A., Brusten, L., Zvelebil, M. J., Stein, R. C., Waterfield, M. D., Davies, S. C., O'Hare, M. J. (1999). Proteomic definition ofnormal human luminal and myoepithelial breast cells purified from reduction mammop/asties. Proc Natl Acad Sci US A 96(22):12589-12594.

Panter, S., Thomson, R., de Bruxelles, G., Laver, D., Trevaskis, B., Udvardi, M. (2000). Identification with proteomics ofnovel proteins associated with the peribacteroid membrane ofsoybean root nodules. Mol Plant Microbe Interact 13(3):325-333.

Papac, D. I., Wong, A., Jones, A. J. (1996). Analysis ofacidic oligosaccharides and g/ycopeptides by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. Anal Chem 68(18):3215-3223.

Pappin, D. J.C., Hojrup, P., Bleasby, A. J. (1993). Rapid identification ofproteins by peptide­ massfingerprinting. Current Biology 3:327-332.

Patton, W. F. (1999). Proteome analysis. II. Protein subcellular redistribution: linking physiology to genomics via the proteome and separation technologies involved. J Chromatogr B Biomed Sci Appl 722(1-2):203-223.

Patton, W. F. (2000). A thousand points of light: the application offluorescence detection technologies to two-dimensional gel electrophoresis and proteomics. Electrophoresis 21(6):1123-1144.

Paweletz, C. P., Liotta, L. A., Petricoin, E. F., 3rd (2001). New technologies for biomarker analysis ofprostate cancer progression: Laser capture microdissection and tissue proteomics. Urology 57(4 Suppl 1):160-163.

Peck, S. C., Nuhse, T. S., Hess, D., Iglesias, A., Meins, F., Boller, T. (2001). Directed proteomics identifies a plant-specific protein rapidly phosphory/ated in response to bacterial and fungal elicitors. Plant Cell 13(6):1467-1475.

Peltier, J.B., Friso, G., Kalume, D. E., Roepstorff, P., Nilsson, F., Adamska, I., van Wijk, K. J. (2000). Proteomics ofthe chloroplast: systematic identification and targeting analysis of lumenal and peripheral thylakoid proteins. Plant Cell 12(3):319-341.

283 Perkins, D. N., Pappin, D. J., Creasy, D. M., Cottrell, J. S. (1999). Probability-based protein identification by searching sequence databases using mass spectrometry data. Electrophoresis 20(18):3551-3567.

Peter, A., Lilja, H., Lundwall, A., Malm, J. (1998). Semenogelin I and semenogelin II, the major gel-forming proteins in human semen, are substrates for transglutaminase. Eur J Biochem 252(2):216-221.

Petkovic, M., Schiller, J., Muller, M., Benard, S., Reichl, S., Arnold, K., Arnhold, J. (2001). Detection ofindividual phospholipids in lipid mixtures by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry: phosphatidylcholine prevents the detection offarther species. Anal Biochem 289(2):202-216.

Pfenninger, A., Karas, M., Finke, B., Stahl, B., Sawatzki, G. (1999). Matrix optimization for matrix-assisted laser desorption/ionization mass spectrometry ofoligosaccharides from human milk. J Mass Spectrom 34(2):98-104.

Pflugfelder, S. C., Jones, D., Ji, Z., Afonso, A., Monroy, D. (1999). Altered cytokine balance in the tear fluid and conjunctiva ofpatients with Sjogren's syndrome keratoconjunctivitis sicca. Curr Eye Res 19(3):201-211.

Pflugfelder, S. C., Wilhelmus, K. R., Osato, M. S., Matoba, A. Y., Font, R. L. (1986). The autoimmune nature ofaqueous tear deficiency. Ophthalmology 93(12):1513-1517.

Pieles, U., Zurcher, W., Schar, M., Moser, H. E. (1993). Matrix-assisted laser desorption ionization time-of-flight mass spectrometry: a powerful tool for the mass and sequence analysis ofnatural and modified oligonucleotides. Nucleic Acids Res 21(14):3191-3196.

Pinto, D. M., Ning, Y., Figeys, D. (2000). An enhanced microfluidic chip coupled to an electrospray Qstar mass spectrometer for protein identification. Electrophoresis 21(1):181-190.

Pleissner, K. P., Hoffmann, F., Kriegel, K., Wenk, C., Wegner, S., Sahlstrom, A., Oswald, H., Alt, H., Fleck, E. (1999). New algorithmic approaches to protein spot detection and pattern matching in two-dimensional electrophoresis gel databases. Electrophoresis 20(4- 5):755-765.

Pocsfalvi, G., Di Landa, G., Ferranti, P., Ritieni, A., Randazzo, G., Malorni, A. (1997). Observation ofnon-covalent interactions between beauvericin and oligonucleotides using

284 electrospray ionization mass spectrometry. Rapid Commun Mass Spectrom 11(3):265- 272.

Poirier, F., Pontet, M., Labas, V., le Caer, J. P., Sghiouar-lmam, N., Raphael, M., Caron, M., Joubert-Caron, R. (2001 ). Two-dimensional database ofa Burkitt lymphoma cell line (DG 75) proteins: protein pattern changes following treatment with 5'-azycytidine. Electrophoresis 22(9):1867-1877.

Posewitz, M. C., Tempst, P. (1999). Immobilized gallium(///) affinity chromatography of phosphopeptides. Anal Chem 71(14):2883-2892.

Posthumus, M.A., Kistermaker, P. G., Meuzelaar, H. C. L., ten Noever de Brauw, M. C. (1978). Laser desorption-mass spectrometry ofpolar nonvolatile bio-organic molecules. Anal Chem 50:985-981.

Pramanik, B. N., Bartner, P. L., Mirza, U. A., Liu, Y. H., Ganguly, A. K. (1998). Electrospray ionization mass spectrometry for the study ofnon-covalent complexes: an emerging technology. J Mass Spectrom 33(10):911-920.

Prause, J. U. (1983). Serum albumin, serum antiproteases and polymorphonuclear leucocyte neutral collagenolytic protease in the tear fluid ofnormal healthy persons. Acta Ophthalmol (Copenh) 61(2):261-271.

Preston, L. M., Murray, K. K., Russell, D. H. (1993). Reproducibility and quantitation of matrix-assisted laser desorption ionization mass spectrometry: effects ofnitrocellulose on peptide ion yields. Biol Mass Spectrom 22(9):544-550.

Pritzker, L. B., Joshi, S., Harauz, G., Moscarello, M. A. (2000). Deimination ofmyelin basic protein. 2. Effect ofmethylation ofMBP on its deimination by peptidylarginine deiminase. Biochemistry 39(18):5382-5388.

Qin, J., Chait, B. T. (1997). Identification and characterization ofposttranslational modifications ofproteins by MALDI ion trap mass spectrometry. Anal Chem 69(19):4002-4009.

Qin, J., Ruud, J., Chait, B. T. (1996). A practical ion trap mass spectrometer for the analysis of peptides by matrix-assisted laser desorption/ionization. Anal Chem 68(10):1784-1791.

285 Qu, X. D., Lehrer, R. I. (1998). Secretory phospholipase A2 is the principal bactericide for staphylococci and other gram-positive bacteria in human tears. Infect Immun 66(6):2791-2797.

Rabilloud, T. (2000). Detecting proteins separated by 2-D gel electrophoresis. Anal Chem 72(1 ):48A-55A.

Rafatro, H., Verbeeck, R. K., Gougnard, T. Y., De Jonghe, P. J., Rasoanaivo, P., Laurent, A., Lhoest, G. (2000). Isolation from rat urine and human liver microsomes and identification by electrospray and nanospray tandem mass spectrometry ofnew malagashanine metabolites. J Mass Spectrom 35(9):1112-1120.

Ragas, J. A., Simmons, T. A., Limbach, P.A. (2000). A comparative study on methods of optimal sample preparation for the analysis ofoligonucleotides by matrix-assisted laser desorption/ionization mass spectrometry. Analyst 125(4):575-581.

Ragunathan, N., Krock, K. A., Klawun, C., Sasaki, T. A., Wilkins, C. L. (1999). Gas chromatography with spectroscopic detectors. J Chromatogr A 856(1-2):349-397.

Raida, M., Schulz-Knappe, P., Heine, G., Forssmann, W. G. (1999). Liquid chromatography and electrospray mass spectrometric mapping ofpeptides from human plasma filtrate. J Am Soc Mass Spectrom 10(1):45-54.

Rashed, M. S., Rahbeeni, Z., Ozand, P. T. (1999). Application ofelectrospray tandem mass spectrometry to neonatal screening. Semin Perinatol 23(2):183-193.

Rasmussen, R. K., Ji, H., &ides, J. S., Moritz, R. L., Reid, G. E., Simpson, R. J., Dorow, D. S. (1997). Two-dimensional electrophoretic analysis ofhuman breast carcinoma proteins: mapping ofproteins that bind to the SH3 domain ofmixed lineage kinase MLK2. Electrophoresis 18(3-4):588-598.

Reitz, C., Breipohl, W., Augustin, A., Bours, J. (1998). Analysis oftear proteins by one- and two-dimensional thiri-layer iosoelectric focusing, sodium dodecyl sulfate electrophoresis and lectin blotting. Detection ofa new component: cystatin C. Graefes Arch Clin Exp Ophthalmol 236(12):894-899.

Remington, S. G., Lima, P.H., Nelson, J. D. (1999). Pancreatic lipase-related protein 1 mRNA in female mouse lacrimal gland. Invest Ophthalmol Vis Sci 40(6):1081-1090.

Richardson, S. D. (2000). Environmental mass spectrometry. Anal Chem 72(18):4477-4496.

286 Riffel, K. A., Song, H., Gu, X., Yan, K., Lo, M. W. (2000). Simultaneous determination ofa novel thrombin inhibitor and its two metabolites in human plasma by liquid chromatography/tandem mass spectrometry. J Pharm Biomed Anal 23(2-3):607-616.

Risby, T. H., Sehnert, S.S. (1999). Clinical application ofbreath biomarkers ofoxidative stress status. Free Radie Biol Med 27(11-12):1182-1192.

Robert, M., Gagnon, C. (1999). Semenogelin I: a coagulum forming, multifunctional seminal vesicle protein. Cell Mol Life Sci SS( 6-7):944-960.

Roepstorff, P. (1989). Plasma desorption mass spectrometry oflarge biomolecules. J Pharm Biomed Anal 7(2):247-253.

Roepstorff, P. (2000). MALDI-TOF mass spectrometry in protein chemistry. Exs 88:81-97.

Roepstorff, P., Fohlman, J. (1984). Proposal for a common nomenclature for sequence ions in mass spectra ofpeptides. Biomed Mass Spectrom 11(11 ):601.

Roepstorff, P., Nielsen, P. F., Klarskov, K., Hojrup, P. (1988). Applications ofplasma desorption mass spectrometry in peptide a~d protein chemistry. Biomed Environ Mass Spectrom 16(1-12):9-18.

Rohlff, C. (2001). Proteomics in neuropsychiatric disorders. Int J Neuropsychopharmacol 4(1):93-102.

Roskey, M. T., Juhasz, P., Smirnov, I. P., Takach, E. J., Martin, S. A., Haff, L. A. (1996). DNA sequencing by delayed extraction-matrix-assisted laser desorption/ionization time of flight mass spectrometry. Proc Natl Acad Sci US A 93(10):4724-4729.

Rouse, J.C., Strang, A. M., Yu, W., Vath, J.E. (1998). Isomeric differentiation ofasparagine­ linked oligosaccharides by matrix-assisted laser desorption-ionization postsource decay time-of-flight mass spectrometry. Anal Biochem 256(1):33-46.

Sack, R. A., Beaton, A., Sathe, S., Morris, C., Willcox, M., Bogart, B. (2000). Towards a closed eye model ofthe pre-ocular tear layer. Prog Retin Eye Res 19(6):649-668.

Sack, R. A., Beaton, A. R., Sathe, S. (1999). Diurnal variations in angiostatin in human tear fluid: a possible role in prevention ofcorneal neovascu/arization. Curr Eye Res 18(3):186-193.

287 Sack, R. A., Bogart, B. I., Beaton, A., Sathe, S., Lew, G. (1997). Diurnal variations in tear glycoproteins: evidence for an epithelial origin for the major non-reducible> or= 450 kDa sialoglycoprotein(s). Cu.rr Eye Res 16(6):577-588.

Sack, R. A., Tan, K. 0., Tan, A. (1992). Diurnal tear cycle: evidence for a nocturnal inflammatory constitutive tear fluid. Invest Ophthalmol Vis Sci 33(3):626-640.

Sack, R. A., Underwood, A., Tan, K. 0., Morris, C. (1994). Vitronectin in human tears-­ protection against closed eye induced inflammatory damage. Adv Exp Med Biol 350:345- 349.

Sakata, M., Sack, R. A., Sathe, S., Holden, B., Beaton, A. R. (1997). Polymorphonuclear leukocyte cells and elastase in tears. Cu.rr Eye Res 16(8):810-819.

Sarioglu, H., Lottspeich, F., Walk, T., Jung, G., Eckerskorn, C. (2000). as a widespread phenomenon in two-dimensional polyacrylamide gel electrophoresis of human blood plasma proteins. Electrophoresis 21(11):2209-2218.

Sathe, S., Sakata, M., Beaton, A. R., Sack, R. A. (1998). Identification, origins and the diurnal role ofthe principal serine protease inhibitors in human tear fluid. Cu.rr Eye Res 17(4):348-362.

Sato, Y., Suzuki, M., Nirasawa, T., Suzuki, A., Endo, T. (2000). Microsequencing ofglycans using 2-aminobenzamide and MALDI-TOF mass spectrometry: occu"ence ofunique linkage-dependentfragmentation. Anal Chem 72(6):1207-1216.

Schenkels, L. C., Rathman, W. M., Veerman, E. C., Nieuw Amerongen, A. V. (1991). Detection ofproteins related to a salivary glycoprotein (EP-GP). Concentrations in human secretions (saliva, sweat, tears, nasal mucus, cerumen, seminal plasma). Biol Chem Hoppe Seyler 372(5):325-329.

Schiller, J., Arnhold, J., Benard, S., Muller, M., Reichl, S., Arnold, K. (1999). Lipid analysis by matrix-assisted laser desorption and ionization mass spectrometry: A methodological approach. Anal Biochem 267(1):46-56.

Schnolzer, M., Jedrzejewski, P., Lehmann, W. D. (1996). Protease-catalyzed incorporation of 180 into peptide fragments and its application for protein sequencing by electrospray and matrix-assisted laser desorption/ionization mass spectrometry. Electrophoresis 17(5):945-953.

288 Schreiner, M., Strupat, K., Lottspeich, F., Eckerskorn, C. (1996). Ultraviolet matrix assisted laser desorption ionization-mass spectrometry ofelectroblotted proteins. Electrophoresis 17(5):954-961.

Schriemer, D. C., Li, L. (1996). Detection ofhigh molecular weight na"ow polydisperse polymers up to 1.5 million daltons by MALDI mass spectrometry. Anal Chem 68(17):2721-2725.

Seal, D. V. (1985). The effect ofageing and disease on tear constituents. Trans Ophthalmol Soc UK 104(Pt 4):355-362.

Seregni, E., Botti, C., Ballabio, G., Bombardieri, E. (1996). Biochemical characteristics and recent biological knowledge on prostate-specific antigen. Tumori 82(1):72-77.

Shen, Z., Thomas, J. J., Averbuj, C., Broo, K. M., Engelhard, M., Crowell, J.E., Finn, M. G., Siuzdak, G. (2001). Porous silicon as a versatile platform for laser desorption/ionization mass spectrometry. Anal Chem 73(3):612-619.

Shevchenko, A., Chemushevich, I., Ens, W., Standing, K. G., Thomson, B., Wilm, M., Mann, M. (1997). Rapid 'de novo 'peptide sequencing by a combination ofnanoelectrospray, isotopic labeling and a quadrupole/time-of-flight mass spectrometer. Rapid Commun Mass Spectrom 11(9):1015-1024.

Shevchenko, A., Jensen, 0. N., Podtelejnikov, A. V., Sagliocco, F., Wilm, M., Vorm, 0., Mortensen, P., Boucherie, H., Mann, M. (1996a). Linking genome and proteome by mass spectrometry: large-scale identification ofyeast proteins from two dimensional gels. Proc Natl Acad Sci US A 93(25):14440-14445.

Shevchenko, A., Loboda, A., Ens, W., Standing, K. G. (2000). MALDI quadrupole time-of­ flight mass spectrometry: a powerful tool for proteomic research. Anal Chem 72(9):2132- 2141.

Shevchenko, A., Sunyaev, S., Loboda, A., Bork, P., Ens, W., Standing, K. G. (2001). Charting the proteomes oforganisms with unsequenced genomes by MALDI-quadrupole time-of­ flight mass spectrometry and BLAST homology searching. Anal Chem 73(9):1917-1926.

Shevchenko, A., Wilm, M., Vorm, 0., Mann, M. (1996b). Mass spectrometric sequencing of proteins silver-stained polyacrylamide gels. Anal Chem 68(5):850-858.

289 Shindelman, J., Mahal, J., Hemphill, G., Pizzo, P., Coty, W. A. (1999). Development and evaluation ofan improved method for screening ofamphetamines. J Anal Toxicol 23(6):506-510.

Sickmann, A., Marcus, K., Schafer, H., Butt-Dorje, E., Lehr, S., Herkner, A., Suer, S., Bahr, I., Meyer, H. E. (2001 ). Identification ofpost-translationally modified proteins in proteome studies. Electrophoresis 22(9):1669-1676.

Simone, N. L., Paweletz, C. P., Charboneau, L., Petricoin, E. F., 3rd, Liotta, L. A. (2000). Laser capture microdissection: beyond.functional genomics to proteomics. Mol Diagn 5(4):301- 307.

Slavakis, A., Papadimas, J. (2000). Procalcitonin: does it play a role in male reproduction? Fertil Steril 74(6):1227-1228.

Smirnov, I. P., Roskey, M. T., Juhasz, P., Takach, E. J., Martin, S. A., Haff, L. A. (1996). Sequencing oligonucleotides by exonuclease digestion and delayed extraction matrix­ assisted laser desorption ionization time-of-flight mass spectrometry. Anal Biochem 238(1):19-25.

Smith, R. D., Loo, J. A., Edmonds, C. G., Barinaga, C. J., Udseth, H. R. (1990). New developments in biochemical mass spectrometry: electrospray ionization. Anal Chem 62(9):882-899.

Smith, R. D., Pasa-Tolic, L., Lipton, M. S., Jensen, P. K., Anderson, G. A., Shen, Y., Conrads, T. P., Udseth, H. R., Harkewicz, R., Belov, M. E., Masselon, C., Veenstra, T. D. (2001). Rapid quantitative measurements ofproteomes by Fourier transform ion cyclotron resonance mass spectrometry. Electrophoresis 22(9):1652-1668.

Song, E. J., Kim, Y. S., Chung, J. Y., Kim, E., Chae, S. K., Lee, K. J. (2000). Oxidative modification ofnucleoside diphosphate kinase and its identification by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. Biochemistry 39(33):10090- 10097.

Soskic, V., Gorlach, M., Poznanovic, S., Boehmer, F. D., Godovac-Zimmennann, J. (1999). Functional proteomics analysis ofsignal transduction pathways ofthe platelet-derived growth factor beta receptor. Biochemistry 38(6):1757-1764.

290 Speed, D. J., Dickson, S. J., Cairns, E. R., Kim, N. D. (2000). Analysis ofsix anticonvulsant drugs using so/id-phase extraction, deuterated internal standards, and gas chromatography-mass spectrometry. J Anal Toxicol 24(8):685-690.

Spengler, B., Kirsch, D., Kaufmann, R., Jaeger, E. (1992). Peptide sequencing by matrix­ assisted laser-desorption mass spectrometry. Rapid Commun Mass Spectrom 6(2):105- 108.

Spina, E., Cozzolino, R., Ryan, E., Garozzo, D. (2000). Sequencing ofoligosaccharides by collision-induced dissociation matrix-assisted laser desorption/ionization mass spectrometry. J Mass Spectrom 35(8): 1042-1048.

Stahl, B., Thurl, S., Zeng, J., Karas, M., Hillenkamp, F., Steup, M., Sawatzki, G. (1994). O/igosaccharides from human milk as revealed by matrix-assisted laser desorption/ionization mass spectrometry. Anal Biochem 223(2):218-226.

Steen, H., Kuster, B., Mann, M. (2001). Quadrupole time-of-flight versus triple-quadrupole mass spectrometry for the determination ofphosphopeptides by precursor ion scanning. J Mass Spectrom 36(7):782-790.

Steinberg, T. H., Chemokalskaya, E., Berggren, K., Lopez, M. F., Diwu, Z., Haugland, R. P., Patton, W. F. (2000a). Ultrasensitivefluorescenceprotein detection in isoelectric focusing gels using a ruthenium metal chelate stain. Electrophoresis 21(3):486-496.

Steinberg, T. H., Haugland, R. P., Singer, V. L. (1996a). Applications ofSYPRO orange and SYPRO red protein gel stains. Anal Biochem 239(2):238-245.

Steinberg, T. H., Jones, L. J., Haugland, R. P., Singer, V. L. (1996b). SYPRO orange and SYPRO red protein gel stains: one-step fluorescent staining ofdenaturing gels for detection ofnanogram levels ofprotein. Anal Biochem 239(2):223-237.

Steinberg, T. H., Lauber, W. M., Berggren, K., Kemper, C., Yue, S., Patton, W. F. (2000b). Fluorescence detection ofproteins in sodium dodecyl sulfate-polyacrylamide gels using environmentally benign,, nonfvcative, saline solution. Electrophoresis 21(3):497-508.

Stenfors, C., Hellman, U., Silberring, J. (1997). Characterization ofendogenous neuropeptide Y in rat hippocampus and its metabolism by nanospray mass spectrometry. J Biol Chem 272(9):5747-5751.

291 Stoeckli, M., Chaurand, P., Hallahan, D. E., Caprioli, R. M. (2001). Imaging mass spectrometry: a new technology for the analysis ofprotein expression in mammalian tissues. Nat Med 7(4):493-496.

Stoeckli, M., Farmer, T. B., Caprioli, R. M. (1999). Automated mass spectrometry imaging with a matrix-assisted laser desorption ionization time-of-flight instrument. J Am Soc Mass Spectrom 10(1):67-71.

Strasburger, C. J. (1998). Laboratory assessment ofGH. Growth Honn IGF Res 8 Suppl A:41- 46.

Strasburger, C. J., Dattani, M. T. (1997). New growth hormone assays: potential benefits. Acta Paediatr Suppl 423:5-11.

Strupat, K., Karas, M., Hillenkamp, F., Eckerskom, C., Lottspeich, F. (1994). Matrix-assisted laser desorption ionization mass spectrometry ofproteins electroblotted after polyacrylamide gel electrophoresis. Anal Chem 66(4):464-470.

Stuchell, R. N., Farris, R. L., Mandel, I. D. (1981). Basal and reflex human tear analysis. II. Chemical analysis: lactoferrin and lysozyme. Ophthalmology 88(8):858-861.

Sugiyama, E., Hara, A., Uemura, K. (1999). A quantitative analysis ofserum sulfatide by matrix-assisted laser desorption ionization time-of-flight mass spectrometry with delayed ion extraction. Anal Biochem 274(1):90-97.

Sumi, T., Sakaki, T., Ohba, H., Shibata, M. (2000). Application ofmatrix-assisted laser desorption/ionization time-of-flight mass spectrometry to insoluble glucose oligomers in decomposed cellulose. Rapid Commun Mass Spectrom 14(19):1823-1827.

Sundqvist, B., Kamensky, I., Hakansson, P., Kjellberg, J., Salehpour, M., Widdiyasekera, S., Fohlman, J., Peterson, P.A., Roepstorff, P. (1984a). Californium-252 plasma desorption time offlight mass spectroscopy ofproteins. Biomed Mass Spectrom 11(5):242-257.

Sundqvist, B., Roepstorff, P., Fohlman, J., Hedin, A., Hakansson, P., Kamensky, I., Lindberg, M., Salehpour, M., Sawe, G. (1984b). Molecular weight determinations ofproteins by californium plasma desorption mass spectrometry. Science 226(4675):696-698.

Taga, E. M., Moore, D. L., Van Etten, R. L. (1983). Studies on the structural basis ofthe heterogeneity ofhuman prostatic and seminal acid phosphatases. Prostate 4(2): 141-150.

292 Takach, E. J., Hines, W. M., Patterson, D. H., Juhasz, P., Falick, A. M., Vestal, M. L., Martin, S. A. (1997). Accurate mass measurements using MALDI-TOFwith delayed extraction. J Protein Chem 16(5):363-369.

Takashima, Y., Takagi, H., Takahashi, M., Reinach, P. S., Mircheff, A. K., Warren, D. W., Yoshimura, N. (1996). Endothelinprotein expression in tear glands ofthe rabbit. Curr Eye Res 15(7):768-773.

Takayasu, T., Ohshima, T., Kondo, T., Sato, Y. (2001). Intratracheal gas analysis for volatile substances by gas chromatography/mass spectrometry--app/ication to forensic autopsies. J Forensic Sci 46(1):98-104.

Talbo, G. H., Suckau, D., Malkoski, M., Reynolds, E. C. (2001). MALDI-PSD-MS analysis of the phosphorylation sites ofcaseinomacropeptide. Peptides 22(7): 1093-1098.

Talrose, V. L., Person, M. D., Whittal, R. M., Walls, F. C., Burlingame, A. L., Baldwin, M.A. (1999). Insight into absorption ofradiation/energy transfer in infrared matrix-assisted laser desorption/ionization: the roles ofmatrices, water and metal substrates. Rapid Commun Mass Spectrom 13(21):2191-2198.

Tanaka, K., Waki, H., Ido, Y., Akita, S., Yoshida, Y., Yoshida, T. (1988). Protein and polymer analyses up to mlz 100,000 by laser ionization time-of-flight mass spectrometry. Rapid Commun Mass Spectrom 2: 151-153.

Tang, K., Allman, S. L., Chen, C. H. (1993a). Matrix-assisted laser desorption ionization of oligonucleotides with various matrices. Rapid Commun Mass Spectrom 7(10):943-948.

Tang, K., Allman, S. L., Jones, R. B., Chen, C. H. (1993b). Quantitative analysis of biopolymers by matrix-assisted laser desorption. Analytical Chemistry 65(15):2164-2166.

Tang, X., Callahan, J. H., Zhou, P ., V ertes, A. (1995). Noncovalent protein--o/igonuc/eotide interactions monitored by matrix-assisted laser desorption/ionization mass spectrometry. Anal Chem 67(24):4542-4548.

Tang, X. D., Sadeghi, M. E., Olumee, Z., Vertes, A., Braatz, J. A., Mcilwain, L. K., Dreifuss, P. A. (1996). Detection and quantitation ofbeta-2-microglobulin glycosylated end products in human serum by matrix-assisted laser desorption/ionization mass spectrometry. Anal Chem 68(21):3740-3745.

293 Taylor, J. A., Johnson, R. S. (1997). Sequence database searches via de novo peptide sequencing by tandem mass spectrometry. Rapid Commun Mass Spectrom 11(9): 1067- 1075.

Tchah, H. (1989). Measurement ofIgA. level in normal human tears by enzyme-linked immunosorbent assay. Korean J Ophthalmol 3(2):70-74.

Terazaki, H., Ando, Y., Suhr, 0., Ohlsson, P. I., Obayashi, K., Yamashita, T., Yoshimatsu, S., Suga, M., Uchino, M., Ando, M. (1998). Post-translational modification oftransthyretin in plasma. Biochem Biophys Res Commun 249(1 ):26-30.

Tervo, T., Vesaluoma, M., Bennett, G. L., Schwall, R., Helena, M., Liang, Q., Wilson, S. E. (1997). Tear hepatocyte growth factor (HGF) availability increases markedly after excimer laser surface ablation. Exp Eye Res 64(4):501-504.

Tervo, T. M., Mertaniemi, P., Ylatupa, S., Tervo, K., Virtanen, T., Partanen, P. (1995). Release ofcalcitonin gene-related peptide in tears after excimer laser photorefractive keratectomy. J Refract Surg 11(2):126-128.

Thakur, A., Willcox, M. D. (2000). Contact lens 'wear alters the production ofcertain inflammatory mediators in tears. Exp Eye Res 70(3):255-259.

Thakur, A., Willcox, M. D., Stapleton, F. (1998). The proinflammatory cytokines and arachidonic acid metabolites in human overnight tears: homeostatic mechanisms. J Clin Immunol 18(1):61-70.

Thiede, B., von Janta-Lipinski, M. (1998). Noncovalent RNA-peptide complexes detected by matrix-assisted laser desorption/ionization mass spectrometry. Rapid Commun Mass Spectrom 12(23): 1889-1894.

Thiellement, H., Bahrman, N., Damerval, C., Plomion, C., Rossignol, M., Santoni, V., de Vienne, D., Zivy, M. (1999). Proteomicsfor genetic and physiological studies in plants. Electrophoresis 20(10):2013-2026.

Thorig, L., Wijngaards, G., Van Haeringen, N. J. (1983). Immunological characterization and possible origin ofplasminogen activator in human tear fluid. Ophthalmic Res 15(5):268- 276.

Tiller, P.R., Cunniff, J., Land, A. P., Schwartz, J., Jardine, I., Wakefield, M., Lopez, L., Newton, J. F., Burton, R. D., Folk, B. M., Buhrman, D. L., Price, P., Wu, D. (1997). Drug

294 quantitation on a benchtop liquid chromatography-tandem mass spectrometry system. J Chromatogr A 771(1-2):119-125.

Tomer, K. B., Crow, F. W., Gross, M. L., Kopple, K. D. (1984). Fast atom bombardment combined with tandem mass spectrometry for the determination ofcyclic peptides. Anal Chem 56(6):880-886.

Tonge, R., Shaw, J., Middleton, B., Rowlinson, R., Rayner, S., Young, J., Pognan, F., Hawkins, E., Currie, I., Davison, M. (2001). Validation and development offluorescence two­ dimensional differential gel electrophoresis proteomics technology. Proteomics 1(3):377- 96.

Tozser, J., Berta, A. (1991). Plasminogen activator inhibitors in human tears. Acta Ophthalmol (Copenh) 69(4):426-431.

Tsarbopoulos, A. (1989). Plasma desorption mass spectrometry ofnatural and recombinant peptides and proteins. Pept Res 2(4):258-266.

Tsarbopoulos, A., Pramanik, B. N., Reichert, P., Siegel, M. M., Nagabhushan, T. L., Trotta, P. P. (1991). 252Cf-plasma desorption and cesium-ion liquid secondary-ion mass spectrometric analysis ofrecombinant proteins. Rapid Commun Mass Spectrom 5(2):81- 85.

Tsubota, K., Saito, I., Miyasaka, N. (1994a). Expression ofgranzyme A and perforin in lacrimal gland ofSjogren's syndrome. Adv Exp Med Biol 350(1):637-640.

Tsubota, K., Saito, I., Miyasaka, N. (1994b). Granzyme A and perforin expressed in the lacrimal glands ofpatients with Sjogren's syndrome. Am J Ophthalmol 117(1):120-121.

Tubbs, K. A., Nedelkov, D., Nelson, R. W. (2001). Detection and quantification ofbeta-2- microglobulin using mass spectrometric immunoassay. Anal Biochem 289(1):26-35.

Tuominen, I., Vesaluoma, M., Teppo, A. M., Gronhagen-Riska, C., Tervo, T. (1999). Soluble Fas and Fas ligand in human tear fluid after photorefractive keratectomy. Br J Ophthalmol 83(12):1360-1363.

Uchio, E., Ono, S. Y., Ikezawa, Z., Ohno, S. (2000). Tear levels ofinterferon-gamma, interleukin (IL) -2, IL-4 and IL-5 in patients with vernal keratoconjunctivitis, atopic keratoconjunctivitis and allergic conjunctivitis. Clin Exp Allergy 30(1):103-109.

295 Udell, I. J., Gleich, G. J., Allansmith, M. R., Ackerman, S. J., Abelson, M. B. (1981). Eosinophil granule major basic protein and Charcot-Leyden crystal protein in human tears. Am J Ophthalmol 92(6):824-828.

Valaskovic, G. A., Kelleher, N. L., McLafferty, F. W. (1996). Attomole protein characterization by capillary electrophoresis-mass spectrometry. Science 273(5279):1199-1202.

Valdes, I., Pitarch, A., Gil, C., Bermudez, A., Llorente, M., Nombela, C., Mendez, E. (2000). Novel procedure for the identification ofproteins by mass fingerprinting combining two­ dimensional electrophoresis with fluorescent SYPRO red staining. J Mass Spectrom 35(6):672-682.

Valentin-Blasini, L., Blount, B. C., Rogers, H. S., Needham, L. L. (2000). HPLC-MS/MS method for the measurement ofseven phytoestrogens in human serum and urine. J Expo Anal Environ Epidemiol 10(6 Pt 2):799-807.

Van Ausdall, D. A., Marshall, W. S. (1998). Automated high-throughput mass spectrometric analysis ofsynthetic oligonucleotides. Anal Biochem 256(2):220-228.

Van Eeckhout, N., Perez, J.C., Claereboudt, J., Vandeputte, R., Van Peteghem, C. (2000). Determination oftetracyclines in bovine kidney by liquid chromatography/tandem mass spectrometry with on-line extraction and clean-up. Rapid Commun Mass Spectrom 14(4):280-285.

Van Haeringen, N. J., Ensink, F., Glasius, E. (1975). Amylase in human tear fluid: origin and characteristics, compared with salivary and urinary amylases. Exp Eye Res 21(4):395- 403.

Van Haeringen, N. J., Glasius, E. (1974). Enzymatic studies in lacrimal secretion. Exp Eye Res 19(2):135-139. van Haeringen, N. J., Glasius, E. (1974). Enzymes ofenergy-producing metabolism in human tear fluid. Exp Eye Res 18(4):407-409. van Haeringen, N. J., Glasius, E. (1976). The origin ofsome enzymes in tear fluid, determined by comparative investigation with two collection methods. Exp Eye Res 22(3):267-272. van Haeringen, N. J., Glasius, E. (1980). Lysosomal hydrolases in tears and the lacrimal gland: effect ofacetylsalicylic acid on the release from the lacrimal gland. Invest Ophthalmol Vis Sci 19(7):826-829.

296 van Setten, G. B. (1996). Basic.fibroblast growth factor in human tear fluid: detection of another growth/actor. Graefes Arch Clin Exp Ophthalmol 234(4):275-277.

Van Setten, G. B., Nilsson, L., Hahne, S., Johnston, J. A., Kvanta, A., Gandy, S. E., Naslund, J., Nordstedt, C. (1996). Beta-amyloid protein protein precursor expression in lacrimal glands and tear fluid. Invest Ophthalmol Vis Sci 37(13):2585-2593.

Vandell, V. E., Limbach, P.A. (1999). Polyamine co-matrices for matrix-assisted laser desorption/ionization mass spectrometry ofoligonucleotides. Rapid Commun Mass Spectrom 13(20):2014-2021.

Venter, J.C., Adams, M. D., Myers, E.W., Li, P. W., Mural, R. J., Sutton, G. G., Smith, H. 0., Yandell, M., Evans, C. A., Holt, R. A., Gocayne, J. D., Amanatides, P., Ballew, R. M., Huson, D. H., Wortman, J. R., Zhang, Q., Kodira, C. D., Zheng, X. H., Chen, L., Skupski, M., Subramanian, G., Thomas, P. D., Zhang, J., Gabor Miklos, G. L., Nelson, C., Broder, S., Clark, A. G., Nadeau, J., McKusick, V. A., Zinder, N., Levine, A. J., Roberts, R. J., Simon, M., Slayman, C., Hunkapiller, M., Bolanos, R., Delcher, A., Dew, I., Fasulo, D., Flanigan, M., Florea, L., Halpern, A., Hannenhalli, S., Kravitz, S., Levy, S., Mobarry, C., Reinert, K., Remington, K., Abu-Threideh~J., Beasley, E., Biddick, K., Bonazzi, V., Brandon, R., Cargill, M., Chandramouliswaran, I., Charlab, R., Chaturvedi, K., Deng, Z., Di Francesco, V., Dunn, P., Eilbeck, K., Evangelista, C., Gabrielian, A. E., Gan, W., Ge, W., Gong, F., Gu, Z., Guan, P., Heiman, T. J., Higgins, M. E., Ji, R.R., Ke, Z., Ketchum, K. A., Lai, Z., Lei, Y., Li, Z., Li, J., Liang, Y., Lin, X., Lu, F., Merkulov, G. V., Milshina, N., Moore, H. M., Naik, A. K., Narayan, V. A., Neelam, B., Nusskern, D., Rusch, D. B., Salzberg, S., Shao, W., Shue, B., Sun, J., Wang, Z., Wang, A., Wang, X., Wang, J., Wei, M., Wides, R., Xiao, C., Yan, C., Yao, A., Ye, J., Zhan, M., Zhang, W., Zhang, H., Zhao, Q., Zheng, L., Zhong, F., Zhong, W., Zhu, S., Zhao, S., Gilbert, D., Baumhueter, S., Spier, G., Carter, C., Cravchik, A., Woodage, T., Ali, F., An, H., Awe, A., Baldwin, D., Baden, H., Barnstead, M., Barrow, I., Beeson, K., Busam, D., Carver, A., Center, A., Cheng, M. L., Curry, L., Danaher, S., Davenport, L., Desilets, R., Dietz, S., Dodson, K., Doup, L., Ferriera, S., Garg, N., Gluecksmann, A., Hart, B., Haynes, J., Haynes, C., Heiner, C., Hladun, S., Hostin, D., Houck, J., Howland, T., Ibegwam, C., Johnson, J., Kalush, F., Kline, L., Koduru, S., Love, A., Mann, F., May, D., Mccawley, S., McIntosh, T., McMullen, I., Moy, M., Moy, L., Murphy, B., Nelson, K., Pfannkoch, C., Pratts, E., Puri, V., Qureshi, H., Reardon, M., Rodriguez, R., Rogers, Y. H., Romblad, D., Ruhfel, B., Scott, R., Sitter, C., Smallwood, M., Stewart, E., Strong, R., Suh, E., Thomas, R., Tint, N. N., Tse, S., Vech, C., Wang, G., Wetter, J., Williams, S., Williams, M., Windsor, S., Winn-Deen, E., Wolfe, K., Zaveri, J., Zaveri, K., Abril, J. F., Guigo, R., Campbell, M. J.,

297 Sjolander, K. V., Karlak, B., Kejariwal, A., Mi, H., Laz.areva, B., Hatton, T., Narechania, A., Diemer, K., Muruganujan, A., Guo, N., Sato, S., Bafna, V., lstrail, S., Lippert, R., Schwartz, R., Walenz, B., Yooseph, S., Allen, D., Basu, A., Baxendale, J., Blick, L., Caminha, M., Carnes-Stine, J., Caulk, P., Chiang, Y. H., Coyne, M., Dahlke, C., Mays, A., Dombroski, M., Donnelly, M., Ely, D., Esparham, S., Fosler, C., Gire, H., Glanowski, S., Glasser, K., Glodek, A., Gorokhov, M., Graham, K., Gropman, B., Harris, M., Heil, J., Henderson, S., Hoover, J., Jennings, D., Jordan, C., Jordan, J., Kasha, J., Kagan, L., Kraft, C., Levitsky, A., Lewis, M., Liu, X., Lopez, J., Ma, D., Majoros, W., McDaniel, J., Murphy, S., Newman, M., Nguyen, T., Nguyen, N., Nodell, M., Pan, S., Peck, J., Peterson, M., Rowe, W., Sanders, R., Scott, J., Simpson, M., Smith, T., Sprague, A., Stockwell, T., Turner, R., Venter, E., Wang, M., Wen, M., Wu, D., Wu, M., Xia, A., Zandieh, A., Zhu, X. (2001). The sequence ofthe human genome. Science 291(5507):1304-1351.

Verhaert, P., Uttenweiler-Joseph, S., de Vries, M., Loboda, A., Ens, W., Standing, K. G. (2001). Matrix-assisted laser desorption/ionization quadropole time-of-flight mass spectrometry: an elegant too/for peptidomics. Proteomics 1(1):118-131.

Verma, R., Chen, S., Feldman, R., Schieltz, D., Yates, J., Dohmen, J., Deshaies, R. J. (2000). Proteasomal proteomics: identification ofnucleotide-sensitive proteasome-interacting proteins by mass spectrometric analysis ofaffinity-purified proteasomes. Mol Biol Cell 11(10):3425-3439.

Vestling, M. M., Fenselau, C. (1994). Poly(vinylidene difluoride) membranes as the inte,face between laser desorption mass spectrometry, gel electrophoresis, and in situ proteolysis. Anal Chem. 66(4):471-477.

Villanueva, J., Canals, F., Querol, E., Aviles, F. X. (2001). Monitoring the expression and purification ofrecombinant proteins by MALDI-TOF mass spectrometry. Enzyme Microb Technol 29(1):99-103.

Vinh, J., Langridge, J. I., Bre, M. H., Levilliers, N., Redeker, V., Loyaux, D., Rossier, J. (1999). Stroctural characterization by tandem mass spectrometry ofthe posttranslational polyglycylation oftubulin. Biochemistry 38(10):3133-3139.

Vinh, J., Loyaux, D., Redeker, V., Rossier, J. (1997). Sequencing branched peptides with CIDIPSD MALDI-TOF in the low-picomole range: application to the structural study of the posttranslational polyglycylation oftubulin. Anal Chem 69(19):3979-3985.

298 Viseux, N., Costello, C. E., Dornon, B. (1999). Post-source decay mass spectrometry: optimized calibration procedure and structural characterization ofpermethylated oligosaccharides. J Mass Spectrom 34(4):364-376.

Vita, J.B., Anderson, J. A., Hulem, C. D., Leopold, I. H. (1981). Angiotensin-converting enzyme activity in ocular fluids. Invest Ophthalmol Vis Sci 20(2):255-257.

Vonn, 0., Roepstorff, P., Mann, M. (1994). Improved resolution and very high sensitivity in MALDI TOF ofmatrix surfaces made by fast evaporation. Analytical Chemistry 66(19):3281-3287.

Voss, T., Haberl, P. (2000). Observations on the reproducibility and matching efficiency oftwo­ dimensional electrophoresis gels: consequencesforcomprehensive data analysis. Electrophoresis 21(16):3345-3350.

Vreken, P., van Lint, A. E., Bootsma, A. H., Overmars, H., Wanders, R. J., van Gennip, A. H. (1999). Rapid diagnosis oforganic acidemias and fatty-acid oxidation defects by quantitative electrospray tandem-MS acyl-camitine analysis in plasma. Adv Exp Med Biol 466:327-337.

Walker, A. K., Rymar, G., Andrews, P. C. (2001). Mass spectrometric imaging ofimmobilized pH gradient gels and creation of "virtual" two-dimensional gels. Electrophoresis 22(5):933-945.

Wang, B. H., Biemann, K. (1994). Matrix-assisted laser desorption/ionization time-of-flight mass spectrometry ofchemically modified oligonucleotides. Anal Chem 66(11):1918- 1924.

Wang, J., Kalt, W., Spoms, P. (2000). Comparison between HPLC and MALDI-TOF MS analysis ofanthocyanins in highbush blueberries. J Agric Food Chem 48(8):3330-3335.

Wang, J., Spoms, P. (1999). Analysis ofanthocyanins in red wine and fruit juice using MALDI­ MS. J Agric Food Chem 47(5):2009-2015.

Wang, J., Spoms, P. (2000). MALDI-TOF MS quantification ofcoccidiostats in poultry feeds. J Agric Food Chem 48(7):2807-2811.

Wang, J., Spoms, P., Low, N. H. (1999a). Analysis offood oligosaccharides using MALDI-MS: quantification offructooligosaccharides. J Agric Food Chem 47(4):1549-1557.

299 Wang, R., Chait, B. T. (1994). High-accuracy mass measurement as a too/for studying proteins. Curr Opin Biotechnol 5(1):77-84.

Wang, S., Zhang, X., Regnier, F. E. (2002). Quantitative proteomics strategy involving the selection ofpeptides containing both cysteine and from tryptic digests ofcell lysates. J Chromatogr A. 949(1-2):153-62.

Wang, T. J., Linton, H.J., Sokoloff, R. L., Grauer, L. S., Rittenhouse, H. G., Wolfert, R. L. (1999b). Western blotting analysis ofantibodies to prostate-specific antigen: specificities for prostate-specific antigen and prostate-specific antigen fragments. Tumour Biol 20(Suppl 1):79-85.

Wang, Y. K., Ma, Z., Quinn, D. F., Fu, E.W. (2001). Inverse 180 labeling mass spectrometry for the rapid identification ofmarker/target proteins. Anal Chem 73(15):3742-50.

Washburn, M. P., Wolters, D., Yates, J. R., 3rd (2001). Large-scale analysis ofthe yeast proteome by multidimensional protein identification technology. Nat Biotechnol 19(3):242-247.

Wasinger, V. C., Cordwell, S. J., Cerpa-Poljak, A., Yan, J. X., Gooley, A. A., Wilkins, M. R., Duncan, M. W., Harris, R., Williams, K. L., Humphery-Smith, I. (1995). Progress with gene-product mapping ofthe Mollicutes: Mycoplasma genitalium. Electrophoresis 16(7):1090-1094.

Weller, M. G. (2000). Immunochromatographic techniques--a critical review. Fresenius J Anal Chem 366(6-7):635-645.

Weykamp, C. W., Penders, T. J., Schmidt, N. A., Borburgh, A. J., van de Calseyde, J. F., Wolthers, B. J. (1989). Steroid pro.file for urine: reference values. Clin Chem 35(12):2281-2284.

Whitehouse, C. M., Dreyer, R. N., Yamashita, M., Fenn, J.B. (1985). Electrospray interface for liquid chromatographs and mass spectrometers. Anal Chem 57(3):675-679.

Whittal, R. M., Li, L. (1995). High-resolution matrix-assisted laser desorption/ionization in a linear time-of-flight mass spectrometer. Anal Chem 67(13):1950-1954.

Wiley, W. C., McLaren, I. H. (1955). Time-of-flight mass spectrometer with improved resolution. Rev Sci Instrum 26:1150-1157.

300 Wilkins, M. R., Gasteiger, E., Gooley, A. A., Herbert, B. R., Molloy, M. P., Binz, P.A., Ou, K., Sanchez, J.C., Bairoch, A., Williams, K. L., Hochstrasser, D. F. (1999). High-throughput mass spectrometric discovery ofprotein post-translational modifications. J Mol Biol 289(3):645-657.

Wilkins, M. R., Gasteiger, E., Tonella, L., Ou, K., Tyler, M., Sanchez, J. C., Gooley, A. A., Walsh, B. J., Bairoch, A., Appel, R. D., Williams, K. L., Hochstrasser, D. F. (1998). Protein identification with N and C-terminal sequence tags in proteome projects. J Mol Biol 278(3):599-608.

Wilkinson, W.R., Gusev, A. I., Proctor, A., Houalla, M., Hercules, D. M. (1997). Selection of internal standards for quantitative analysis by matrix assisted laser desorption ionization (MALD/) time-of-flight mass spectrometry. Fresenius Journal of Analytical Chemistry 357(3):241-248.

Willcox, M. D., Morris, C. A., Thakur, A., Sack, R. A., Wickson, J., Boey, W. (1997). Complement and complement regulatory proteins in human tears. Invest Ophthalmol Vis Sci 38(1):1-8.

Wilm, M., Mann, M. (1996). Analytical properties ofthe nanoelectrospray ion source. Anal Chem 68(1):1-8.

Wilm, M., Shevchenko, A., Houthaeve, T., Breit, S., Schweigerer, L., Fotsis, T., Mann, M. (1996). Femtomole sequencing ofproteins from polyacrylamide gels by nano­ electrospray mass spectrometry. Nature 379(6564):466-469.

Wittmann, C., Heinzle, E. (2001). MALDI-TOF MS for quantification ofsubstrates and products in cultivations ofCorynebacterium glutamicum. Biotechnol Bioeng 72(6):642- 647.

Wollensak, G., Mur, E., Mayr, A., Baier, G., Gottinger, W., Stoffler, G. (1990). Effective methods for the investigation ofhuman tear film proteins and lipids. Graefes Arch Clin Exp Ophthalmol 228(1):78-82.

Wong, F. A., Edom, R. W., Duda, M., Tischio, J.P., Huang, M., Juzwin, S., Tegegne, G. (1999). Determination ofnorgestimate and its metabolites in human serum using high­ performance liquid chromatography with tandem mass spectrometric detection. J Chromatogr B Biomed Sci Appl 734(2):247-255.

301 Woods, A. S., Huestis, M.A. (2001). A study ofpeptide-peptide interaction by matrix-assisted laser desorption/ionization. J Am Soc Mass Spectrom 12(1):88-96.

Wu, J., Chatman, K., Harris, K., Siuzdak, G. (1997). An automated MALDI mass spectrometry approach for optimizing cyclosporin extraction and quantitation. Anal Chem 69(18):3767-3771.

Wu, K. J., Odom, R. W. (1998). Characterizing synthetic polymers by MALDI MS. Anal Chem 70(13):456A-461A.

Wu, K. J., Shaler, T. A., Becker, C. H. (1994). Time-of-flight mass spectrometry of underivatized single-stranded DNA oligomers by matrix-assisted laser desorption. Anal Chem 66(10):1637-1645.

Xu, X., Bartlett, M. G., Stewart, J. T. (2000). Determination ofondansetron and its hydroxy metabolites in human serum using solid-phase extraction and liquid chromatography/positive ion electrospray tandem mass spectrometry. J Mass Spectrom 35(11 ): 1329-1334.

Yamagaki, T., Mitsuishi, Y., Nakanishi, H. (1998). Structural analysis ofxyloglucan oligosaccharides by the post-source decay fragmentation method ofMALDI-TOF mass spectrometry: influence ofthe degree ofsubstitution by branched galactose, xylose, and fucose on the fragment ion intensities. Biosci Biotechnol Biochem 62(12):2470-2475.

Yamagaki, T., Nakanishi, H. (1998). Influence ofstereoisomeric glucose, galactose and mannose residues on fragmentation at their glycosidic linkages in post-source decay fragment analyses for oligosaccharides using matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. Rapid Commun Mass Spectrom 12(16):1069-1074.

Y amagaki, T ., Nakanishi, H. (2000). Post-source decay fragmentation analyses oflinkage isomers ofLewis-type oligosaccharides in curved-field reflectron matrix-assisted laser desorption/ionization time-of-flight mass spectrometry: combined in-source decay/post­ source decay experiments and relative ion abundance analysis. J Mass Spectrom 35(11):1300-1307.

Yan, J. X., Harry, R. A., Spibey, C., Dunn, M. J. (2000). Postelectrophoretic staining of proteins separated by two-dimensional gel electrophoresis using SYPRO dyes. Electrophoresis 21(17):3657-3665.

302 Yan, J. X., Sanchez, J.C., Tonella, L., Williams, K. L., Hochstrasser, D. F. (1999). Studies of quantitative analysis ofprotein expression in Saccharomyces cerevisiae. Electrophoresis 20(4-5):738- 742.

Yanagida, M., Miura, Y., Yagasaki, K., Taoka, M., Isobe, T., Takahashi, N. (2000). Matrix assisted laser desorption/ionization-time offlight-mass spectrometry analysis ofproteins detected by anti-phosphotyrosine antibody on two-dimensional-gels offibrolast cell lysates after tumor necrosis factor-alpha stimulation. Electrophoresis 21(9): 1890-1898.

Yang, Y., Chien, M. (2000). Characterization ofgrape procyanidins using high-performance liquid Chromatography/Mass spectrometry and matrix-assisted laser Desorption/Ionization time-of-flight mass spectrometry. J Agric Food Chem 48(9):3990- 3996.

Yao, X., Freas, A, Ramirez, J., Demirev, P. A, Fenselau, C. (2001). Proteolytic 180 labeling for comparative proteomics: model studies with two serotypes ofadenovirus. Anal Chem 73(13):2836-2842.

Yates, J. R., 3rd, Speicher, S., Griffin, P.R., Hunkapiller, T. (1993). Peptide mass maps: a highly informative approach to protein identification. Anal Biochem 214(2):397-408.

Yates, J. R., Eng, J. K., McCormack, AL. (1995a). Mining genomes: co"elating tandem mass spectra ofmodified and unmodified peptides to sequences in nucleotide databases. Anal Chem 67(18):3202-3210.

Yates, J. R., Eng, J. K., McCormack, AL., Schieltz, D. (1995b). Method to co"elate tandem mass spectra ofmodified peptides to amino acid sequences in the protein database. Anal Chem 67(8):1426-1436.

Yates, J. R., Link, A J., Schieltz, D. (2000). Direct analysis ofproteins in mixtures. Application to protein complexes. Methods Mol Biol 146(10):17-26.

Yates, J. R., McCormack, AL., Schieltz, D., Carmack, E., Link, A (1997). Direct analysis of protein mixtures by tandem mass spectrometry. J Protein Chem 16(5):495-497.

Yoo, B. C., Vlkolinsky, R., Engidawork, E., Cairns, N., Fountoulakis, M., Lubec, G. (2001). Differential expression ofmolecular chaperones in brain ofpatients with Down syndrome. Electrophoresis 22(6):1233-1241.

303 Yoshida, K., Nitatori, Y., Uchiyama, Y. (1996). Localization ofglycosaminoglycans and CD44 in the human lacrimal gland. Arch Histol Cytol 59(5):505-513.

Yoshino, K., Garg, R., Monroy, D., Ji, Z., Pflugfelder, S. C. (1996). Production and secretion of transforming growth factor beta (TGF-beta) by the human lacrimal gland. Curr Eye Res 15(6):615-624.

Zaluzec, E. J., Gage, D. A., Watson, J. T. (1995). Matrix-assisted laser desorption ionization mass spectrometry: applications in peptide and protein characterization. Protein Expr Purif 6(2): 109-123.

Zamfir, A., Konig, S., Althoff, J., Peter-Katalinc, J. (2000). Capillary electrophoresis and off­ line capillary electrophoresis-electrospray ionization quadrupole time-of-flight tandem mass spectrometry ofcarbohydrates. J Chromatogr A 895(1-2):291-299.

Zand, R., Li, M. X., Jin, X., Lubman, D. (1998). Determination ofthe sites ofposttranslational modifications in the charge isomers ofbovine myelin basic protein by capillary electrophoresis-mass spectroscopy. Biochemistry 37(8):2441-2449.

Zhang, B., Foret, F., Karger, B. L. (2000a). A microdevice with integrated liquid junction for facile peptide and protein analysis by capillary electrophoresislelectrospray mass spectrometry. Anal Chem 72(5):1015-1022.

Zhang, H., Caprioli, R. M. (1996). Capillary electrophoresis combined with matrix-assisted laser desorption/ionization mass spectrometry; continuous sample deposition on a matrix­ precoated membrane target. J Mass Spectrom 31(9):1039-1046.

Zhang, W., Chait, B. T. (2000). ProFound: an expert system for protein identification using mass spectrometric peptide mapping information. Anal Chem 72(11):2482-2489.

Zhang, W. M., Finne, P., Leinonen, J., Stenman, U. H. (2000b). Characterization and determination ofthe complex between prostate-specific antigen and alpha I-protease inhibitor in benign and malignant prostatic diseases. Scand J Clin Lab Invest Suppl(233):51-58.

Zhang, W. M., Leinonen, J., Kalkkinen, N., Dowell, B., Stenman, U. H. (1995). Purification and characterization ofdifferent molecular forms ofprostate-specific antigen in human seminal fluid. Clin Chem 41(11):1567-1573.

304 Zhu, Y., He, L., Srinivasan, J. R., Lubman, D. M. (1997). Improved resolution in the detection ofo/igonuc/eotides up to 60-mers in matrix-assisted laser desorptiowionization time-of­ flight mass spectrometry using pulsed-delayed extraction with a simple high voltage transistor switch. Rapid Commun Mass Spectrom 11(9):987-992.

Zhu, Y. F., Chung, C. N., Taranenko, N. I., Allman, S. L., Martin, S. A., Haff, L., Chen, C. H. (1996). The study o/2,3,4-trihydroxyacetophenone and 2,4,6-trihydroxyacetophenone as matrices for DNA detection in matrix-assisted laser desorptio11lionization time-of-flight mass spectrometry. Rapid Commun Mass Spectrom 10(3):383-388.

Zuo, X., Echan, L., Hembach, P., Tang, H. Y., Speicher, K. D., Santoli, D., Speicher, D. W. (2001 ). Towards global analysis ofmammalian proteomes using sample prefractionation prior to narrow pH range two-dimensional gels and using one-dimensional gels for insoluble and large proteins. Electrophoresis 22(9):1603-1615.

Zuo, X., Speicher, D. W. (2000). A method for global analysis ofcomplex proteomes using sample prefractionation by solution isoelectrofocusing prior to two-dimensional electrophoresis. Anal Biochem 284(2):266-278.

305