• No results found

Age determination of body fluids by mass spectrometry

N/A
N/A
Protected

Academic year: 2021

Share "Age determination of body fluids by mass spectrometry"

Copied!
87
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

Age determination of body fluids by

mass spectrometry

Katarzyna Szykuła

Master Thesis

2015 – 2016

(2)

2

MSc Chemistry

Analytical Sciences

Master Thesis

Age determination of body fluids by mass spectrometry

by

Katarzyna Szykuła

June 2016

Supervisor:

Second examiner:

Prof. dr. Garry Corthals Dr. Michelle Camenzuli

Analytical Chemistry Group

(3)

3

Acknowledgements

I would like to express my gratitude to professor Garry Corthals for his supervision and guidance

during this Master’s project. I have enjoyed my time carrying out this research and I would like to

thank everyone from the Analytical Chemistry group for their help and kindness. Additionally, I would

like to thank Dr. Michelle Camenzuli for reviewing this thesis.

Moreover, I would like to thank professor Maurice Aalders and his group for their help and the

opportunity to do experiments at Academic Medical Center.

(4)

4

Abstract

In forensic science, biological traces have a significant importance for forensic investigators. They can

provide crucial information about the donor or about a crime itself, for example what happened on a

crime scene or when the criminal incident was committed. In order to estimate the latter one, aging

of body fluids found on a crime scene is studied. This research investigated aging process of four

body fluids: blood, saliva, semen and fingermarks. As all of them are protein-containing fluids, this

type of compounds was examined by using mass spectrometry. Samples of aforementioned body

fluids were analyzed at different age, and subsequently, the numbers of proteins identified as well as

their quantification were compared. For blood, saliva and semen, the number of unique proteins was

changing with the age of a sample. Additionally, some proteins showed variations in quantity when

sample was getting older. These findings can indicate that protein levels are changing over time in

biological traces, and these biomolecules can be potential biomarkers for age estimation of specific

body fluid found on a crime scene.

(5)

5

Table of Contents

Acknowledgements ... 3

Abstract ... 4

1. Introduction ... 7

1.1 Forensic biological traces ... 7

1.1.1 Blood ... 7

1.1.2 Saliva ... 8

1.1.3 Semen ... 8

1.1.4 Fingermarks ... 8

1.2 Mass spectrometry ... 9

1.2.1 Proteomics ... 9

1.2.2 Quantitation in proteomics ... 11

1.2.3 Proteomics in forensic science ... 13

2. Materials and methods ... 15

2.1 Materials ... 15

2.2 Methods ... 15

2.2.1 Blood ... 15

2.2.2 Saliva ... 17

2.2.3 Semen ... 17

2.2.4 Fingermarks ... 17

3. Results and Discussion ... 19

3.1 Blood... 19

3.1.1 Method optimization ... 19

3.1.2 Examining aging of blood ... 20

3.2 Saliva ... 25

3.3 Semen ... 30

3.4 Fingermarks ... 34

4. Conclusions and future research ... 35

5. References ... 36

Appendix 1. ... 39

Appendix 2. ... 44

Appendix 3. ... 51

Appendix 4. ... 57

Appendix 5. ... 61

(6)

6

Appendix 6. ... 65

Appendix 7. ... 70

Appendix 8. ... 76

(7)

7

1. Introduction

orensic science is an enormously developing field nowadays. As an interdisciplinary study, it

utilizes the knowledge and technology from several fields, such as chemistry, physics, medicine

and mathematics to solve crimes. On a crime scene, investigators can find many forensically

important traces. Traces like: pieces of glass, hairs, fibers or paints are defined as physical traces. On

the other hand, body fluids, which can be commonly found on a crime scene as well, belong to the

group of biological traces. All traces found on a crime scene can play a crucial role during

investigations. They can provide information about the suspect/victim or what happened during the

criminal incident. However, this thesis will focus only on body fluids, therefore, only biological

evidence will be further described in details.

Examining biological evidence has a high importance for investigators as they can provide

intelligence about the donor (by DNA profiling) or about the crime of investigation. The latter one is

often studied by analyzing the bloodstain pattern, which can give the information about events that

occurred on the crime scene [1].

Moreover,

by studying the aging of body fluids, it can be estimated

when a crime took place, which can narrow down the time intervals for detectives.

To date, several

attempts have been performed to study body fluids aging by using mainly spectroscopic [2, 3] or

chromatographic methods [4, 5]. However, these studies did not provide with sufficient information

about the aging process. Therefore, the aim of this research was to use mass spectrometry

technique, suitable for trace analysis, to study the aging of body fluids.

1.1 Forensic biological traces

1.1.1 Blood

This body fluid is composed of blood cells (red cells, white cells and platelets) suspended in blood

plasma. The red color of blood is due to an iron-containing protein – hemoglobin, which its main

function is to transport the oxygen from lungs to tissues[6]. As blood contains proteins, lipids,

electrolytes, metabolites, hormones and more [6], this fluid is the most extensively studied, not only

in medical fields. In forensics, studying blood traces has various purposes. Besides obtaining DNA

profile, or studying blood stain pattern [1], also aging of this body fluid has been examined.

The idea to study blood aging is not new. Many attempts were made to investigate aging process by

studying different blood components with various methods, for example RNA with reverse

transcription polymerase chain reaction (RT-PCR) [7], erythrocytes with electron paramagnetic

resonance (EPR) [3] or globulins with immunoelectrophoresis [8]. More recent, three hemoglobin

derivatives: oxy-hemoglobin (oxy-Hb), methemoglobin (met-Hb) and hemichrome (HC) were studied

[2, 9, 10]. As the fractions of these derivatives are changing in time (fraction of oxy-Hb decreases and

met-Hb and HC increase), Bremmer et al.[2] applied reflectance spectroscopy to record spectra of

blood stains and calculated these fractions. They presented the potential of this method, as they

were able to discriminate age of blood stains with larger time intervals (e.g. 20 days). Edelman et al.

applied hyperspectral imaging to record the spectra of the described hemoglobin derivatives and

(8)

8

observed a decreased error compared to reflectance spectroscopy [10]. However, Bremmer et al. [9]

showed that conversion of oxy-Hb to met-Hb and HC is dependent on temperature and humidity.

Moreover, both methods were successful only when spectra were recorded against a white

background.

1.1.2 Saliva

This very essential body fluid has many functions, but the most important are: food solubilization,

lubrication, antibacterial, buffering and food digestion. Saliva is produced by salivary glands and it

contains water, electrolytes, proteins and other organic molecules [11]. Its chemical composition has

been studied widely, mostly for cancer [12], autoimmune diseases [13], hereditary diseases [14], or

viral diseases [15]. Saliva can also be used for drug analysis [16].

On a crime scene, this oral body fluid can be found for instance on food or cigarettes. In forensic

science, it is mainly studied for DNA profiling and aging studies have never been reported before.

Nevertheless, some study has shown that when saliva is in ex situ conditions [17], proteins can

degrade over time. This information is of importance and can indicate that saliva proteins have

potential to be used as a biomarker for age determination of saliva.

1.1.3 Semen

The main composition of semen is seminal fluid which may contain spermatozoa [18]. This fluid

contains high concentration of sugars (fructose and glucose) and proteins [19]. As study showed,

in a

single individual seminal fluid, over 900 proteins can be identified [20] which can suggest that human

seminal fluid proteome can be used for biomarker studies, for example in cancer [21] or infertility

research[22].

In forensic science, this biological evidence can be found on a crime scene in a case of

sexual assaults. Semen has a high evidential importance as from this fluid donor DNA profile can be

obtained.

For identification purposes, forensic investigators utilize the proteins’ natural fluorescence

from semen. However, there is yet no literature published to use this biomolecules in age

determination of seminal fluid.

1.1.4 Fingermarks

This type of traces is often found a crime scene. The chemical composition of fingermarks is diverse.

They contain elements (e.g. sodium, potassium, chloride), amino acids, proteins, fatty acids and

other lipids [23]. Forensic scientists study fingermarks to obtain as much information as possible

about the donors. For instance, from analysis of fingermarks information about the sex [24] or drug

abused [25] can be obtained. Similar to the other body fluids, the knowledge of the age of

fingermarks can be of significant importance for investigators. Wolstenholme et al. [26] attempted to

determine the age of fingermarks by investigating the degradation of olenaic acid. The other

research group, van Dam et al. [27], showed that the fluorescence signal is changing with the age of

the sample. However, the estimation error of aged samples was relatively high - only 55% of aged

male fingermarks estimated within the error of 1.9 days.

(9)

9

1.2 Mass spectrometry

Mass spectrometry (MS) is a widely used method in analytical chemistry. Its very intense

development can be proved by five Nobel Prizes, starting from 1906 with J.J.Thomson for his work on

the electrical conductivity of gases. Subsequently, other prizes were for isotope discovery by mass

spectrograph, for cyclotron and for the development of the ion trap technique. The last Nobel Prize

in mass spectrometry, until now, was awarded to J. Bennet and K. Tanaka in 2002. The first one for

developing soft desorption ionization methods, and the second one for developing a new method for

mass spectrometric analyses of biological macromolecules; both extremely important in protein

analysis. Mass spectrometry, as a versatile technique, has found its application in various disciplines,

such as toxicology, forensic science, clinical or environmental chemistry. Its main advantage is high

sensitivity [28], which can be critical, especially, in forensic science for analyzing biological traces,

which often are found on the crime scene in scarce or mixed.

1.2.1 Proteomics

Proteomics is the field of science which studies the biological processes in organisms by protein

analysis. A proteome is the entire protein complement expressed by a genome. However, in contrast

to genes, the nature of proteins is not static but dynamic. Proteins can undergo modifications

(phosphorylation, oxidation), their abundance can change or they can form clusters. Those variations

are dependent on the physiological state of the organism. Therefore, the proteome reflects the state

of a cell or a tissue, and it is crucial to not only identify all the proteins but also to measure their

changes as well as pathways of change[29].

Protein analysis is a challenging task, as proteome is not only very complex because of all interactions

and modifications described before, but also, compared to genes, proteins cannot undergo

amplifications, which is the limiting factor when a sample amount is scarce. Hence, only appropriate

tools (i.e. high sensitivity) can be applied in proteomics, which makes mass spectrometry a suitable

method.

In proteomics, there are two ionization methods in widespread use: matrix-assisted laser

desorption/ionization (MALDI) and electrospray ionization (ESI). Both of them can generate ions from

large nonvolatile biomolecules. MALDI and ESI are knows as soft ionization methods, which means

that there is a minimal or no fragmentation of the analytes. The first one is mostly used for peptide

mass fingerprinting or tissue imaging. In MALDI, sample is mixed with a matrix, which molecules have

a strong absorption at the applied laser wavelength. The absorbed energy is transferred to the

analyte, which is desorbed and ionized together with matrix molecules. This solid-phase technique

mostly generates singly charged ions. Electrospray ionization is a liquid technique, which is easy to

couple to such separation methods as liquid chromatography or electrophoresis. Here, a high voltage

is applied, which generates highly charged droplets of the analyte. In contrast to MALDI, ESI

generates multiply charged ions [28].

The typical workflow in proteomics for LC-MS/MS analysis is presented on the Fig. 1. Briefly, proteins

in a sample undergo digestion into peptides, mostly by using enzyme digestion, trypsin for example.

Those peptides prior to MS/MS analysis are separated with reverse-phase liquid chromatography. In

(10)

10

MS/MS analysis, the precursor ions as well as product ions are scanned, which provides crucial

information about the structure of a sample. After the analysis, by using appropriate software,

protein and peptide identification can be obtained.

Figure 1. Typical workflow in proteomics for LC-MS/MS analysis. Adapted from reference [30].

The instrument used in this project was TripleTOF mass spectrometer. The scheme below presents

the features in this instrument. Briefly, QJet and Q0 are used for ion focusing, Q1 selects the

precursor ion and Q2 is the collision cell, where the ions collide with neutral gas molecules

(nitrogen). That formed product ions leave the collision cell and are re-accelerated and focused by

ions optics into a parallel beam which constantly enters TOF analyzer. There, a pulsed electric field is

pushing ions orthogonally to their previous direction, and then they acquire their final energy [31].

The accelerated ions travel through the tube, where at the opposite site is an electrostatic ion mirror

which reverses the ions direction to the detector. Ions gain the same kinetic energy, however, their

velocities are different, due to the mass differences. And consequently, ions reach the detector at

different time.

MS1

(11)

11

Figure 2. The scheme of triple TOF MS features. (a) The major platform features in details. (b) an image of this platform[32].

1.2.2 Quantitation in proteomics

Nowadays, only protein identification is no longer sufficient in proteomics studies. In order to gain

more knowledge about living organisms, tissues or cells, it is important to quantify the proteins. Their

abundances can change over a time or under specific conditions, such as disease for example. In

proteomics, there are two quantitative approaches, one which is based on labeling the sample and

the second one is label-free. Labeling techniques can be divided into three categories such as:

metabolic labeling, chemical labeling and addition of standard peptides. In those approaches, labeled

peptides have the same chemical properties as unlabeled ones, with the only difference in their

masses. In metabolic labeling, such as stable isotope labeling by amino acids in cell culture (SILAC) for

example, samples of “light” and “heavy” amino acids are mixed at early stage. In chemical labeling,

the label can be introduced at protein or peptide levels. One of commonly used chemical labeling is

the isotope-coded affinity tag (ICAT). This label is bound to sulfhydryl groups of cysteine residues and

is useful to study oxidation or reduction level of proteins. However, as cysteine belongs to low

abundant amino acids in proteins, it reduces the number of peptides that can be examined. The

other type of chemical labeling is isobaric mass tags. There are two tags commercially available:

tandem mass tags (TMT) and isobaric tags for relative and absolute quantitation (iTRAQ)[33]. The

third technique of labeling approach in quantitative proteomics is by using synthetic isotope-labeled

peptides as a standard for absolute quantification (AQUA). These reference peptides allow to precise

measure the level of proteins and posttranslationally modified proteins. For AQUA, selected reaction

monitoring (SRM) analysis is performed [34].

In label-free approach, two quantitation techniques can be distinguished. First one is based on under

the curve (AUC) or signal intensity measurements, both based on precursor ion spectra. With these

methods chromatographic alignment is required. The second technique, spectral counting, is based

on counting the number of peptides assigned to a protein in MS/MS analysis. In spectral counting

methods, more abundant peptides are selected for fragmentation, and they produce a higher

abundance of MS/MS spectra. This process is proportional to protein amount in data-dependent

(12)

12

acquisition. In order to assess protein abundance in the analyzed sample, protein abundance index

(PAI) is used. This index is defined as the number of identified peptides divided by the number of

observable generated peptides for every protein within a preferred mass range [35]. In order to

obtain absolute quantitation, PAI can be converted into exponentially modified emPAI, which is

equally to exponential form of PAI minus one, and this is proportional to protein content in analyzed

samples [36]. As in spectral counting, longer proteins generate more peptides than smaller ones, this

also can be used for quantification. In this case, normalized spectral abundance factor (NSAF) is used

which takes into account the length of the protein. In order to calculate NSAF, the number of spectral

counts for protein is divided by its length and this is defined as spectral abundance factor (SAF),

which has to be normalized by division by the sum of all SAFs for all proteins in a mixture [37].

Before applying a specific quantitation method, it is essential to consider which parameters are

crucial in the performed analysis.

For example, it has to be determined what proteome coverage is

desired in the analyzed sample. This and other parameters for quantitative methods are overviewed

in the table below.

Table 1. Overview of quantitation methods applied in proteomics [33].

Method Proteome coverage Quantitative accuracy Quantitation type

Metabolic labeling

SILAC Medium Precise Relative

Chemical labeling

ICAT Poor Precise Relative

iTRAQ, TMT Medium Medium Relative

Standard peptide Poor Precise Relative/Absolute

Label-free

Ion intensities (PCP) Good Medium Relative

Spectrum count Good Poor Relative

Derived indices (APEX, emPAI)

Good Poor Relative/Absolute

Figure 3 shows that from all proteins in a sample, only a certain amount can be identified.

Furthermore, from this amount, even a smaller fraction can be successfully quantified.

It emphasizes

how challenging protein analysis is and how important it is to perform appropriate sample

preparation with suitable enrichment and purification steps. Additionally, it also indicates how much

of the proteome is still unknown.

(13)

13

Figure 3. Graphical representation of the amount of proteins that can be identified relative to all proteins in a sample [33].

1.2.3 Proteomics in forensic science

In forensic science it is very important to identify as many traces as possible which can be found on a

crime scene. The information of the trace’s origin can facilitate to reconstruct what has happened on

a crime scene. Especially, very crucial is the analysis of biological matrices as they can contain DNA,

which can have a huge value for investigators. However, as mostly those matrices are the mixtures of

several body fluids, such analysis is a complicated process. To identify such mixtures, an advanced

and sensitive method, like mass spectrometry, is needed. With this method, such body fluids as

blood, saliva, semen, vaginal fluids, urine and nasal secretions can be identified by a protein-specific

biomarker, which is unique for every body fluid [38, 39]. Moreover, analyzing biological traces with

MS is not destructive for DNA, which is an another advantage [40]. Hence, identification of the

source of a biological trace can be performed prior to DNA analysis.

The other approach of applying proteomics in forensic field is investigating organ-specific proteome.

This studies are especially useful in a case of ballistic reconstruction of the event [41] as the

information about which organ was damaged with the bullet can help to reconstruct the bullet’s

projectile.

Table 2. Organ-specific proteins [41].

Organ Protein

Heart

myosin binding protein C,cardiac-type glycogen phosphorylase, brain form troponin I, cardiac muscle

myosin light chain 3

pyruvate dehydrogenase E1 component subunit beta, mitochondrial

calpastatin

NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 4

cytochrome c oxidase subunit 4 isoform 1, mitochondrial

Kidney

calbindin

Na(+)/H(+) exchange regulatory cofactor NHE-RF3 low-density lipoprotein receptor-related protein 2 villin 1

retinyl ester hydrolase type 1

membrane metalloendopeptidase variant 2 phosphotriesteraserelated protein plastin-1

(14)

14

Liver

hydroxymethylglutaryl- CoA synthase, mitochondrial carbamoyl-phosphate synthase [ammonia], mitochondrial phenylalanine-4- hydroxylase 3-oxo-5-beta-steroid 4-dehydrogenase catechol Omethyltransferase acetyl-CoA acetyltransferase cytochrome P450 2E1 dimethylaniline monooxygenase Lung plastin-2 cathelicidin-4 tubulin beta chain

cysteine and glycine-rich protein 1 prostaglandin F synthase 2 myosin light polypeptide 6 calpain-2 catalytic subunit alpha-actinin-1

Muscle

bridging integrator 1

myosin-binding protein C, slow-type fructose-1,6- bisphosphatase isozyme 2 myosin-binding protein C, fast-type troponin C, skeletal muscle (fast type)

myosin regulatory light chain 2, skeletal muscle isoform nebulin

PDZ and LIM domain protein 3

As it can be seen, protein studies by mass spectrometry are common used in forensic science. It can

provide with very substantial information about the donor of a sample, or about the circumstances

that the crime happened. However, one of the very important information obtained from body fluids

has not been yet investigated using mass spectrometry – age determination of body fluids by protein

analysis. This thesis presents a novel approach of studying the aging process with LC-MS/MS method.

(15)

15

2. Materials and methods

2.1 Materials

All samples were taken from healthy donors and kept in the dark at room temperature.

Chemicals:

Acetonitrile, methanol, water, formic acid (Biosolve)

Ammonium bicarbonate, DTT, iodoacetamide, Tris(2-carboxyethyl)phosphine hydrochloride, sodium

deoxycholate, Brilliant Blue G (Sigma Aldrich)

Sequencing Grade Modified Trypsin (Promega)

4-20% Tris-glycine precast gel, Tricine-Glycine SDS Running Buffer (Novex)

4-15% Tris-HCl precast gel (Biorad)

Apparatus:

Samples were collected on: Human ID Bloodstain Card; glass microfibers filter (Whatman) or into 2.0

ml Eppendorf vials.

Solid-phase extraction was performed with C18 solid-phase extraction cartridges, 4mm/1mL

(Empore).

LC system consist of nanoLC 425; separation column C18, 3µm, 120Å, 0.075x150mm, and trap

column C18, 3µm, 120Å, 350µmx0.5mm (Eksigent). Flow rate: 300nL/minute, injection volume 1-5

µL.

MS/MS analysis was performed with Triple TOF 5600+ (ABSciex) in positive mode. TOF m/z range:

400-1250, product ion m/z range 100-2000.

2.2 Methods

2.2.1 Blood

Dried blood spots were prepared by spotting 15µl of blood on Whatman filter paper. Samples

preparation for LC-MS/MS analysis was performed according to the protocol described by Chambers,

A.G., et al. [42]. Briefly, dried blood spots were excised from the filter paper and transferred to 1.5 ml

Eppendorf tubes. Fresh sample (15µl) was pipetted directly to the vial. In order to dissolve dried

blood spots and dilute fresh blood, 800µl of 25mM ammonium bicarbonate was added to the vials

and vortexed. Subsequently, 100 µl of 10% sodium deoxycholate and 10 µl of 0.5 M

Tris(2-carboxyethyl)phosphine hydrochloride were added and samples were incubated for an hour at 60°C.

Then, samples were alkylated for one hour in the dark (covered with aluminum foil) in room

temperature after addition of 52.63µl 200mM iodoacetamide. In order to consume remain

iodoacetamide, 55.4µl of 200mM dithiotreitol (DTT) was added and samples were incubated for 30

minutes at 37°C. Next, 10 µl of trypsin solution was added and protein digestion was performed

(16)

16

overnight at 37°C. Protein concentration in samples, before digestion, was estimated with Bradford

assay (Biorad). After digestion, sodium deoxycholate was precipitated by adding 40 µl of 2% formic

acid and the samples were centrifuged at 4000 g for 20 minutes. The supernatant was collected into

the new vials. Prior to LC-MS/MS analysis, samples were desalted by using solid-phase extraction.

Cartridges were first pre-washed twice with 50% ACN/water and 0.1% TFA. Afterwards, samples

were loaded and washed with 0.1% TFA. Analytes were eluted with 0.1% formic acid in 60% ACN.

The collected samples were, subsequently, evaporated to dryness and peptides were reconstituted

with water to obtain a final protein concentration of 0.2µg/µl.

In order to obtain the highest number of protein identification, three different LC methods were first

tested with fresh blood samples. For all methods, the same chemicals were used; mobile phase A:

0.1% formic acid, mobile phase B: 100% acetonitrile.

Table 3. Gradient profile for method 1 [43].

Time [min] Mobile phase A% Mobile phase B%

0 97.3 2.7 1.5 93.7 6.3 16 86.5 13.5 18 86.2 13.8 33 77.5 22.5 38 59.5 40.5 39 19 81 42.9 19 81 43 97.3 2.7

Table 4. Gradient profile for method 2.

Time [min] Mobile phase A% Mobile phase B%

0 100 0

10 95 5

40 40 60

45 20 80

60 100 0

Table 5. Gradient profile for method 3 [42].

Time [min] Mobile phase A% Mobile phase B%

0 95 5

90 55 45

92 10 90

(17)

17

2.2.2 Saliva

Prior to LC-MS/MS analysis, proteins in saliva samples were first separated with SDS-PAGE using

precast 4-15% Tris-HCl gel. 10 µl of saliva was mixed with 10 µl of reducing sample buffer and

incubated for 5 minutes in 95°C. Then samples were applied to the gel lanes (20 µl per lane). Samples

were run at a voltage of 100V for 75 minutes. Afterwards, proteins were stained with Coomassie blue

and the gel was destained with distilled water.

Selected bands were excised from the gel and in-gel digestion was performed. Firstly, Coomassie

blue staining was removed from gel bands by incubation with 200µl of 0.04M NH

4

HCO

3

/50% ACN.

Next, proteins in gel bands were reduced with 100µl of 20mM DTT for 30 minutes in 37°C.

Afterwards, 100µl of 55mM iodoacetamide was added to every sample for alkylation in room

temperature for 20 minutes. Afterwards, proteins were digested with 10µl of 0.02µg/µl of trypsin

solution for 20 minutes on ice and subsequently 20µl of 40mM ammonium bicarbonate/10% ACN

was added and digestion was performed overnight in 37°C. Next day, digested proteins were

extracted from the gel bands with first 60µl ACN for 15 minutes in 37°C and then with 150µl of

50%ACN/5%HCOOH also for 15 minutes at the same temperature. Finally, peptides were dried and

dissolved in 10 µl of water prior to LC-MS/MS analysis and 94 minutes method was used, the same as

for blood samples.

2.2.3 Semen

Semen samples were analyzed similarly to saliva samples described in section 2.2.2. Firstly, proteins

were separated with SDS-PAGE (however, in this case, samples were first diluted 21 times with

phosphate-buffered saline) and later in-gel digestion was performed according to the same protocol.

LC-MS/MS analysis of semen peptides was performed with 20 minutes method (see Table 6).

Table 6. Gradient profile for semen samples.

Time [min] Mobile phase A% Mobile phase B%

0 95 5 15 70 30 16 5 95 18 5 95 20 95 5

2.2.4 Fingermarks

For fingermark samples collection, donors (males and females) were asked to put their finger onto a

glass surface, without previous washing their hands, in order to obtain “realistic” conditions.

Deposited fingermarks were later swabbed with cotton swabs. Swabs were subsequently transferred

into Eppendorf vials, 900 µl of 25mM ammonium bicarbonate was added and samples were

incubated at 37°C for 30 minutes. Afterwards, swabs were removed and samples were centrifuged

for 15 minutes at 3000g. The supernatants were collected and in-solution digestion was performed

(18)

18

according to the same protocol as for blood samples. For LC-MS/MS analysis, 66 minutes method

was used (see Table 7).

Table 7. Gradient profile for fingermark samples.

Time [min] Mobile phase A% Mobile phase B%

0 90 10

60 0 100

65 0 100

(19)

19

3. Results and Discussion

Due to problems with the instrumentation, which had occurred during this research project, not all

samples were analyzed.

This chapter presents the results obtained from analysis of aging body fluids by mass spectrometry.

3.1 Blood

3.1.1 Method optimization

Prior to blood samples analysis, LC-MS/MS conditions were initially optimized with fresh blood

samples. Three different gradients were tested and the number of identified proteins and peptides

was compared. The graphs below present the obtained results.

Figure 4. Number of proteins identified in fresh blood with three different methods.

Figure 5. Number of peptides identified in fresh blood with three different methods. 0 50 100 150 200

Proteins identified

43 minutes method 60 minutes method 94 minutes method

0 200 400 600 800 1000 1200

Peptides identified

(20)

20

From the foregoing graphs can be derived that the highest number of identification was obtained by

using 94 minutes method. With this gradient, 179 proteins and 953 peptides were identified. The 60

minutes method provided a significant lower number of proteins and peptide, 117 and 748

respectively. Finally, application of the shortest gradient resulted in 167 identified proteins and 748

identified peptides.

3.1.2 Examining aging of blood

Blood samples were analyzed at three different ages: fresh blood sample (liquid), 1 day old (stain)

and 7 days old blood (stain) with LC-MS/MS. Subsequently, the number of proteins and peptides

from every sample were compared (see Table 8). All measurements were performed in triplicates

and data was generated using human thorough protein database with Protein Pilot

TM

software using

false discovery rate (FDR) 1%.

Table 8. Number of proteins and peptides identified in aged blood samples, in triplicates.

Sample Proteins #1 Proteins #2 Proteins #3 Peptides #1 Peptides #2 Peptides #3 Fresh blood 115 131 139 1280 1712 1646 1 day old 119 130 128 1552 1522 1512 7 days old 129 110 111 1458 1369 1058

Coefficient of variation (CV) in every sample for proteins and peptides, respectively, is as follow: fresh

sample 9.5%, 15%; one day old sample 4.7%, 1.4%; and seven days old sample 9.1%, 16.2%.

In the next step, data was generated using other software. First, Search Gui 2.7.1, and subsequently,

Peptide Shaker 1.8.1. Two combined search engines were used: Open Mass Spectrometry Search

Algorithm (OMSSA) and MS-GF+. The used database for identification was Target-Decoy version

Homo sapiens protein database + common contaminants. Quality control filtering was performed

with 1% FDR. Data from three technical replicates were pooled together.

The number of identified proteins from all three samples was plotted and presented in the Figure 4.

The complete lists of identified proteins in blood samples (using Search Gui and Peptide Shaker) can

be found in Appendices 1-3.

(21)

21

Figure 6. Comparison of proteins in all three blood samples.

78 proteins were detected in every blood sample. Nonetheless, the number of unique proteins in

every sample is different. These number decreased from 14 proteins in fresh blood to 12 protein in

one day old sample and finally to 4 proteins in 7 days old blood.

Protein quantification was performed by using normalized spectrum abundance factor (NSAF).

10 proteins were found with alterations in NSAF values. 9 proteins have decreasing NSAF value (Fig.

7-15) and for one it is increasing (Fig. 16).

Figure 7. NSAF values for alpha-1 acid glycoprotein-2. 0 0,02 0,04 0,06 0,08 0,1

fresh 1 day old 7 days old

NS

A

F

(22)

22

Figure 8. NSAF values for alpha-2-macroglobulin.

Figure 9. NSAF values for ankyrin-1.

Figure 10. NSAF values for apolipoprotein B-100. 0 0,005 0,01 0,015 0,02 0,025 0,03 0,035

fresh 1 day old 7 days old

NS A F

Alpha-2-macroglobulin

0 0,001 0,002 0,003 0,004 0,005 0,006 0,007

fresh 1 day old 7 days old

NS A F

Ankyrin-1

0 0,0005 0,001 0,0015 0,002 0,0025 0,003 0,0035

fresh 1 day old 7 days old

NS

A

F

(23)

23

Figure 11. NSAF values for band 3 anion transport protein.

Figure 12. NSAF values for Ig gamma-4 chain region.

Figure 13. NSAF values for haptoglobin. 0 0,01 0,02 0,03 0,04 0,05 0,06 0,07

fresh 1 day old 7 days old

NS

A

F

Band 3 anion transport protein

0 0,01 0,02 0,03 0,04 0,05 0,06

fresh 1 day old 7 days old

NS

A

F

Ig gamma-4 chain C region

0 0,002 0,004 0,006 0,008 0,01

fresh 1 day old 7 days old

NS

A

F

(24)

24

Figure 14. NSAF values for hemopexin.

Figure 15. NSAF values for putative uncharacterized protein.

Figure 16. NSAF values for putative bisphosphoglycerate mutase.

From all aforementioned proteins, two characterize with significant changes in NSAF values:

apolipoprotein B-100 and haptoglobin. The first one is responsible for lipids transporting and

proteins and phospholipids binding (See Table 9). The latter one is accountable for capturing and

0 0,01 0,02 0,03 0,04 0,05 0,06

fresh 1 day old 7 days old

NS A F

Hemopexin

0 0,02 0,04 0,06 0,08 0,1

fresh 1 day old 7 days old

NS

A

F

Putative uncharacterized protein

0,019 0,02 0,021 0,022 0,023 0,024 0,025 0,026

fresh 1 day old 7 days old

NS

A

F

(25)

25

combining free plasma hemoglobin and it allows for hepatic cyclin of heme iron which prevents the

damage of kidneys.

For two proteins, alpha-1 acid glycoprotein-2 and putative uncharacterized proteins, the NSAF value

is decreasing notably during first 24 hours, however, posteriorly, this decline is less noticeably.

Alpha-1 acid glycoprotein-2 is transporting protein, however, the function of putative

uncharacterized proteins in humans is still unknown. In other mammals, mouse for example, it has a

regulation function of proteins activity.

Only bisphosphoglycerate mutase protein demonstrates an increasing trend in NSAF values over

examined time period. It can be explained by the function of this protein, to regulate hemoglobin

oxygen affinity, which is stronger with the increasing age of the sample.

Table 9. Functions of proteins with changing NSAF values. Information took from http://www.uniprot.org/.

Protein Function

Alpha-1 acid glycoprotein-2 Transporting protein; binding hydrophobic ligands and synthetic drugs

Alpha-2-macroglobulin Inhibiting proteinases by unique 'trapping' mechanism

Ankyrin-1 Attaching integral membrane proteins to cytoskeletal

elements

Apolipoprotein B-100 Lipid transporter activity, binding proteins and phospholipids,

Band 3 anion transport protein Transporting protein, mediator in electroneutral anion exchange across the cell membrane; structural protein

Bisphosphoglycerate mutase regulating hemoglobin oxygen affinity, exhibiting mutase and phosphatase activities

Ig gamma-4 chain C region Antigen binding

Haptoglobin Capturing and combining with free plasma

hemoglobin (preventing kidney damage)

Hemopexin Binding heme and transporting it to the liver for

breakdown and iron recovery Putative uncharacterized protein Unknown function in homo sapiens

3.2 Saliva

Saliva samples at ages of one day old, one week old and three weeks old were analyzed.

Firstly, proteins were separated with SDS-PAGE (Fig. 17). The arrows indicate from which part of the

gel bands were excised. Per sample, three bands were excised and each band was analyzed

separately. Afterwards, results from three bands were pooled together.

(26)

26

Figure 17. SDS-PAGE of saliva samples.

Data were generated according to the second step with blood analysis, by using First Search Gui 2.7.1

and Peptide Shaker 1.8.1 software. The number of all identified proteins in all three samples is

presented in the Venn diagram below. The all lists of proteins identified in saliva samples can be

found in Appendices 4-6.

Figure 18. Comparison of proteins in all three saliva samples.

39 proteins were present in every saliva sample, nonetheless, the number of unique proteins

detected is increasing with the age of saliva samples, from 9 unique proteins in one day old sample to

22 in three weeks old.

(27)

27

Similarly to blood samples, saliva samples also underwent quantification with NSAF. For eight

proteins, an alteration of NSAF was observed (See Fig. 19-26).

Figure 19. NSAF values for keratin, type I cytoskeletal 13.

Figure 20. NSAF values for keratin, type I cytoskeletal 17.

Figure 21. NSAF values for keratin, type II cytoskeletal 2 epidermal. 0 0,02 0,04 0,06 0,08 0,1 0,12 0,14 0,16

one day one week 3 weeks

NS

A

F

Keratin, type I cytoskeletal 13

0 0,005 0,01 0,015 0,02 0,025

one day one week 3 weeks

NS

A

F

Keratin, type I cytoskeletal 17

0 0,005 0,01 0,015 0,02 0,025

one day one week 3 weeks

NS

A

F

Keratin, type II cytoskeletal 2

epidermal

(28)

28

Figure 22. NSAF values for keratin, type II cytoskeletal 3.

Figure 23. NSAF values for keratin, type II cytoskeletal 6A.

Figure 24. NSAF values for lactotransferrin. 0 0,01 0,02 0,03 0,04 0,05

one day one week 3 weeks

NS

A

F

Keratin, type II cytoskeletal 3

0 0,005 0,01 0,015 0,02 0,025 0,03 0,035 0,04

one day one week 3 weeks

NS

A

F

Keratin, type II cytoskeletal 6A

0 0,002 0,004 0,006 0,008 0,01 0,012

one day one week 3 weeks

NSA

F

(29)

29

Figure 25. NSAF values for leukocyte elastase inhibitor.

Figure 26. NSAF values for tubulin alpha-4A chain.

Interestingly, in this case, there are more proteins in which NSAF values were increasing with the age

of the sample. Moreover, most of proteins with variation in NSAF are keratin type proteins. The most

significant changes occur for keratin, type I cytoskeletal 13 with upward trend and for keratin, type II

3 with the downward trend. Furthermore, those two proteins have similar function: structural

molecule activity. The other two proteins with considerable alterations are leukocyte elastase

inhibitor and tubulin alpha-4A chain. The first one is responsible for the activity of neutrophil

proteases elastase, and the latter one for GTPase activity and GTP binding.

Table 10. Functions of proteins with changing NSAF values. Information took from http://www.uniprot.org/.

Protein Function

Keratin, type I cytoskeletal 13 Structural molecule activity; cellular response to retinoic acid; tongue morphogenesis

Keratin, type I cytoskeletal 17 Structural molecule activity

Keratin, type II cytoskeletal 2 epidermal Associated with keratinocyte activation; proliferation and keratinization

Keratin, type II cytoskeletal 3 Structural molecule activity; intermediate filament cytoskeleton organization 0 0,01 0,02 0,03 0,04 0,05

one day one week 3 weeks

NS

A

F

Leukocyte elastase inhibitor

0 0,005 0,01 0,015 0,02 0,025 0,03 0,035

one day one week 3 weeks

NS

A

F

(30)

30

Keratin, type II cytoskeletal 6A Involved in the activation of follicular keratinocytes after wounding

Lactotransferrin Iron ion binding

Leukocyte elastase inhibitor Regulating the activity of the neutrophil proteases elastase

Tubulin alpha-4A chain GTPase activity, GTP binding

3.3 Semen

Semen samples (fresh, one week old and two weeks old) were analyzed according to the same

protocol as used for saliva samples. Proteins were first separated with SDS-PAGE, then 7 bands per

lane were excised and analyzed separately. At the end, results were pooled together for every

sample.

(31)

31

The Venn diagram below depicts the identified proteins in all semen samples. The lists of all

identified proteins in semen samples can be found in Appendices 7-9.

Figure 28. Comparison of proteins in all three semen samples.

In the case of this body fluid, 41 common proteins for all aging samples were found, however, the

number of the unique proteins is variable. In fresh samples 36 unique proteins were identified. In 7

days old samples this number increased to 37. Nevertheless, this number decreased to only 11

unique proteins in two weeks old samples.

Proteins in all semen samples were quantified with NSAF and 6 proteins showed variations in NSAF

values (Fig. 29-34).

Figure 29. NSAF values for clusterin. 0 0,005 0,01 0,015 0,02 0,025

fresh one week two weeks

NS

A

F

(32)

32

Figure 30. NSAF values for dermcidin.

Figure 31. NSAF values for keratin, type I cytoskeletal 10.

Figure 32. NSAF values for keratin, type II cytoskeletal 6A. 0

0,05 0,1 0,15 0,2

fresh one week two weeks

NS A F

Dermcidin

0 0,01 0,02 0,03 0,04 0,05 0,06

fresh one week two weeks

NS

A

F

Keratin, type I cytoskeletal 10

0 0,002 0,004 0,006 0,008 0,01 0,012

fresh one week two weeks

NS

A

F

(33)

33

Figure 33. NSAF values for semenogelin-2.

Figure 34. NSAF values for serum albumin.

Keratin proteins are also present in semen samples with the downward or upward trend for type I

and type II, respectively. However, there are two proteins which NSAF is changing significantly. First

one, dermcidin, with upward trend, has antimicrobial properties. The second one, with reducing

NSAF, is semenogelin-2, which is involved in gel matrix formation. This result can indicate why the

older semen is more liquid then the fresh one.

Table 11. Functions of proteins with changing NSAF values. Information took from http://www.uniprot.org/.

Protein Function

Clusterin

Maintains partially unfolded proteins in a state appropriate for subsequent refolding by other chaperones

Dermcidin Displays antimicrobial activity; exhibits proteolytic activity

Keratin, type I cytoskeletal 10 Cellular response to calcium ion

Keratin, type II cytoskeletal 6A Involved in the activation of follicular keratinocytes after wounding

Semenogelin-2 Participates in the formation of a gel matrix 0 0,005 0,01 0,015 0,02 0,025

fresh one week two weeks

NS A F

Semenogelin-2

0 0,005 0,01 0,015 0,02 0,025 0,03 0,035

fresh one week two weeks

NS

A

F

(34)

34

Serum albumin Transporting

3.4 Fingermarks

7 female (2 donors) and 6 male (one donor) fingermarks samples were analyzed at different age:

fresh (only female), one day old, 10 days old and 17 days old. Results were generated with Protein

Pilot software. Table 12 presents proteins identified in each sample.

Table 12. Proteins identified in every fingermark sample.

Sample Proteins

Fresh female fingermark keratin, type I cytoskeletal (9, 10, 14, 16); keratin, type II cytoskeletal (1, 1b, 2 epidermal, 5, 6A, 78); dermcidin;

keratinocyte proline-rich protein; filaggrin-2;

skin-specific protein 32; loricrin;

junction plakoglobin

One day old female fingermark keratin, type II cytoskeletal 1

One day old male fingermark keratin, type I cytoskeletal (9, 10, 13, 14, 15, 16, 17); keratin, type II cytoskeletal (1, 2 epidermal); charged multivesicular body protein 2a; serum albumin;

phosphofurin acidic cluster sorting protein 1 10 days old female fingermark Keratin, type I cytoskeletal 9;

keratin, type II cytoskeletal (1, 2 epidermal, 5, 6A, 73); apoptosis-enhancing nuclease;

10 days old male fingermark Keratin, type I cytoskeletal 9;

keratin, type II cytoskeletal (1, 2 epidermal); sodium/potassium-transporting ATPase subunit alpha-4;

dermcidin

17 days old female fingermark Keratin, type I cytoskeletal (9, 10, 14, 16, 17); keratin, type II cytoskeletal (1, 2 epidermal, 5); dermcidin

17 days old male fingermark Keratin, type I cytoskeletal 10; prolactin-inducible protein

All samples contain keratin proteins, which is a protein found in skin cells. Interestingly, some

samples contain proteins which were not found in other fingermarks. This can suggest that those

proteins originate from contamination, as the donors deposited their fingermarks directly on the

glass slides, without previous finger preparation.

The results from fingermark samples could not be quantified as the signal to noise ratio in the

spectra was too low.

(35)

35

4. Conclusions and future research

For the first time mass spectrometry method was applied to study the aging of blood, saliva, semen

and fingerprints. In this research, protein changes were investigated, which occurs when the body

fluid is aging. It was shown that for some proteins, their concentration is increasing with time, for

example biphosphoglycerate mutase in blood, or keratins, type II in saliva. There were also several

proteins with decreasing concentration, such as apolipoprotein B-100 or semengelin-2. In the present

study, it was observed that for saliva, more proteins increased in concentration in comparison to

blood and semen. With regard to fingermarks samples, in this case, different extraction method is

required to obtain enough protein concentration that can be measured. This work presented the

potential of mass spectrometry technique for age determination of four body fluids commonly found

on a crime scene, although more research is needed in this area.

For future research, it is recommended to broaden the range of analyzed samples. In the case of

investigating proper biomarkers, it is important to expand studies to several donors from both sexes

with a wider range of age.

Additionally, in forensic science it is significant to investigate fluids on

different substrates. Also, environmental conditions are crucial, for example how proteins undergo

degradation at different temperatures and humidity levels. Furthermore, label-free quantification

methods used in this study, can also suggest that more accurate methods should be applied.

(36)

36

5. References

1.

Peschel, O., et al., Blood stain pattern analysis. Forensic science, medicine, and pathology,

2011. 7(3): p. 257-270.

2.

Bremmer, R.H., et al., Age estimation of blood stains by hemoglobin derivative determination

using reflectance spectroscopy. Forensic science international, 2011. 206(1): p. 166-171.

3.

Sakurai, H., et al., Dating of human blood by electron spin resonance spectroscopy.

Naturwissenschaften, 1989. 76(1): p. 24-25.

4.

Inoue, H., et al., Identification of fetal hemoglobin and simultaneous estimation of bloodstain

age by high-performance liquid chromatography. International journal of legal medicine,

1991. 104(3): p. 127-131.

5.

Andrasko, J., The estimation of age of bloodstains by HPLC analysis. Journal of Forensic

Science, 1997. 42(4): p. 601-607.

6.

Kumar, V., et al., Clinical pathology and their potential application in disease diagnosis.

International Journal of Agricultural Sciences, 2013. 3(9): p. 005-015.

7.

Bauer, M., S. Polzin, and D. Patzelt, Quantification of RNA degradation by semi-quantitative

duplex and competitive RT-PCR: a possible indicator of the age of bloodstains? Forensic

science international, 2003. 138(1): p. 94-103.

8.

Rajamannar, K., Determination of the age of bloodstains using immunoelectrophoresis.

Journal of Forensic Science, 1977. 22(1): p. 159-164.

9.

Bremmer, R.H., et al., Biphasic oxidation of oxy-hemoglobin in bloodstains. PloS one, 2011.

6(7): p. e21845.

10.

Edelman, G., T.G.v. Leeuwen, and M.C. Aalders, Hyperspectral Imaging for the Age

Estimation of Blood Stains at the Crime Scene. Indian Internet Journal of Forensic Medicine &

Toxicology, 2014. 12(1): p. 22-22.

11.

Kaufman, E. and I.B. Lamster, The diagnostic applications of saliva—a review. Critical Reviews

in Oral Biology & Medicine, 2002. 13(2): p. 197-212.

12.

Zhang, L., et al., Development of transcriptomic biomarker signature in human saliva to

detect lung cancer. Cellular and molecular life sciences, 2012. 69(19): p. 3341-3350.

13.

Hernández-Molina, G., et al., Chemokine saliva levels in patients with primary Sjögren’s

syndrome, associated Sjögren’s syndrome, pre-clinical Sjögren’s syndrome and systemic

autoimmune diseases. Rheumatology, 2011: p. ker019.

14.

Sánchez-Pablo, M.-A., V. González-García, and A. del Castillo-Rueda, Study of total stimulated

saliva flow and hyperpigmentation in the oral mucosa of patients diagnosed with hereditary

hemochromatosis. Series of 25 cases. Med Oral Patol Oral Cir Bucal, 2012. 1(17): p. e45-9.

15.

Slots, J. and H. Slots, Bacterial and viral pathogens in saliva: disease relationship and

infectious risk. Periodontology 2000, 2011. 55(1): p. 48-69.

16.

Wong, R.C., M. Tran, and J.K. Tung, Oral fluid drug tests: effects of adulterants and foodstuffs.

Forensic science international, 2005. 150(2): p. 175-180.

17.

Esser, D., et al., Sample stability and protein composition of saliva: implications for its use as

a diagnostic fluid. Biomarker insights, 2008. 3.

18.

Al-Dosary, A.M., et al., Y-STR Profiling of Semen Stain Evidences of Azoospermic Individuals.

International Journal of Forensic Science & Pathology, 2015: p. 210-214.

19.

Owen, D.H. and D.F. Katz, A review of the physical and chemical properties of human semen

and the formulation of a semen simulant. Journal of andrology, 2005. 26(4): p. 459-469.

20.

Pilch, B. and M. Mann, Large-scale and high-confidence proteomic analysis of human seminal

plasma. Genome biology, 2006. 7(5): p. R40.

21.

Auger, J., N. Sermondade, and F. Eustache, Semen quality of 4480 young cancer and systemic

disease patients: baseline data and clinical considerations. Basic and clinical andrology, 2016.

(37)

37

22.

Qian, L., et al., IL-18 levels in the semen of male infertility: semen analysis. International

journal of biological macromolecules, 2014. 64: p. 190-192.

23.

Bailey, M.J., et al., Chemical characterization of latent fingerprints by matrix-assisted laser

desorption ionization, time-of-flight secondary ion mass spectrometry, mega electron volt

secondary mass spectrometry, gas chromatography/mass spectrometry, X-ray photoelectron

spectroscopy, and attenuated total reflection Fourier transform infrared spectroscopic

imaging: an intercomparison. Analytical chemistry, 2012. 84(20): p. 8514-8523.

24.

Huynh, C., et al., Forensic identification of gender from fingerprints. Analytical chemistry,

2015. 87(22): p. 11531-11536.

25.

Hazarika, P., et al., Multiplexed detection of metabolites of narcotic drugs from a single latent

fingermark. Analytical chemistry, 2010. 82(22): p. 9150-9154.

26.

Wolstenholme, R., et al., Study of latent fingermarks by matrix‐assisted laser

desorption/ionisation mass spectrometry imaging of endogenous lipids. Rapid

communications in mass spectrometry, 2009. 23(19): p. 3031-3039.

27.

van Dam, A., et al., Oxidation monitoring by fluorescence spectroscopy reveals the age of

fingermarks. Angewandte Chemie International Edition, 2014. 53(24): p. 6272-6275.

28.

de Hoffmannn, E. and V. Stroobant, Mass Spectrometry: Principles & Applications. 3 ed. 2007:

Wiley&Sons.

29.

Aebersold, R. and D.R. Goodlett, Mass spectrometry in proteomics. Chemical reviews, 2001.

101(2): p. 269-296.

30.

Meissner, F. and M. Mann, Quantitative shotgun proteomics: considerations for a

high-quality workflow in immunology. Nature immunology, 2014. 15(2): p. 112-117.

31.

Chernushevich, I.V., A.V. Loboda, and B.A. Thomson, An introduction to quadrupole–

time‐of‐flight mass spectrometry. Journal of Mass Spectrometry, 2001. 36(8): p. 849-865.

32.

Andrews, G.L., et al., Performance characteristics of a new hybrid quadrupole time-of-flight

tandem mass spectrometer (TripleTOF 5600). Analytical chemistry, 2011. 83(13): p.

5442-5446.

33.

Schulze, W.X. and B. Usadel, Quantitation in mass-spectrometry-based proteomics. Annual

review of plant biology, 2010. 61: p. 491-516.

34.

Gerber, S.A., et al., Absolute quantification of proteins and phosphoproteins from cell lysates

by tandem MS. Proceedings of the National Academy of Sciences, 2003. 100(12): p.

6940-6945.

35.

Rappsilber, J., et al., Large-scale proteomic analysis of the human spliceosome. Genome

research, 2002. 12(8): p. 1231-1245.

36.

Ishihama, Y., et al., Exponentially modified protein abundance index (emPAI) for estimation of

absolute protein amount in proteomics by the number of sequenced peptides per protein.

Molecular & Cellular Proteomics, 2005. 4(9): p. 1265-1272.

37.

Paoletti, A.C., et al., Quantitative proteomic analysis of distinct mammalian Mediator

complexes using normalized spectral abundance factors. Proceedings of the National

Academy of Sciences, 2006. 103(50): p. 18928-18933.

38.

Legg, K.M., et al., Discovery of highly specific protein markers for the identification of

biological stains. Electrophoresis, 2014. 35(21-22): p. 3069-3078.

39.

Igoh, A., Y. Doi, and K. Sakurada, Identification and evaluation of potential forensic marker

proteins in vaginal fluid by liquid chromatography/mass spectrometry. Analytical and

bioanalytical chemistry, 2015. 407(23): p. 7135-7144.

40.

Van Steendam, K., et al., Mass spectrometry-based proteomics as a tool to identify biological

matrices in forensic science. International journal of legal medicine, 2013. 127(2): p. 287-298.

41.

Dammeier, S., et al., Mass-Spectrometry-Based Proteomics Reveals Organ-Specific Expression

Patterns To Be Used as Forensic Evidence. Journal of proteome research, 2015. 15(1): p.

(38)

38

42.

Chambers, A.G., et al., Comparison of proteins in whole blood and dried blood spot samples

by LC/MS/MS. Journal of The American Society for Mass Spectrometry, 2013. 24(9): p.

1338-1345.

43.

Chambers, A.G., et al., Multiple Reaction Monitoring Enables Precise Quantification of 97

(39)

39

The data presented in appendixes were generated by Search Gui 2.7.1 and Peptide Shaker 1.8.1.

Note that some proteins are identified more than one time per sample. emPAI - exponentially

modified protein abundance index; NSAF - normalized spectrum abundance factor; MW – molecular

weight.

Appendix 1.

All validated proteins in fresh blood sample.

Accession

Protein Inference

Class Protein name

Sequence Coverage (%) #Validate d Peptides

emPAI NSAF MW (kDa)

P01024 Unrelated Proteins Complement C3 40.8899579 0739627 44 0.714125253 2913134 0.01152003971 2574301 187.0298753 1966258 F6KPG5 Single Protein Albumin (Fragment) 76.7521367 5213675 56 4.115178099 293143 0.06854929340 53836 66.48798147 102801 P01023 Single Protein Alpha-2-macroglobulin 47.3541383 98914515 44 1.249054605 8357808 0.02915375633 5619455 163.1878914 6924135 P11277 Related and Unrelated Proteins Spectrin beta chain, erythrocytic 17.5011698 64295743 26 0.249264143 7899512 0.00369787553 21113903 246.3157334 321971 A0A087W ZE4 Related Proteins Spectrin alpha chain, erythrocytic 1 15.7201646 09053498 23 0.177572498 21932586 0.00339278233 15763428 280.9387841 720546 A0A087W TM7 Related and Unrelated Proteins Apolipoprotei n B-100 6.88305709 0239411 18 0.091578487 87739664 0.00315395313 4692732 489.5273949 3234196 C8C504 Single Protein Beta-globin 95.9183673 4693877 23 33.14548873 833603 1.08852418504 30756 15.98734254 8192922 P02730 Related and Unrelated Proteins Band 3 anion transport protein 24.2590559 82436884 15 0.674482438 0115348 0.05793226863 3375625 101.7274121 8512044 P02042 Related Proteins Hemoglobin subunit delta 93.1972789 1156462 18 14.84893192 4611133 0.45956003636 3566 16.04528986 0997062 A0A0C4D GB6 Related and Unrelated Proteins Serum albumin 35.2649006 62251655 21 0.790655529 6923772 0.15593423608 704202 69.18136901 532891 A0A0K0K 1H8 Related and Unrelated Proteins Epididymis secretory sperm binding protein Li 71p 33.3810888 252149 19 0.739828052 9303642 0.04192341630 458897 77.02963611 452142 P02787 Related and Unrelated Proteins Serotransferri n 32.5214899 713467 19 0.739828052 9303642 0.02566325370 2962904 77.01362996 651181 P02787 Related and Unrelated Proteins Serotransferri n 32.9512893 9828081 19 0.739828052 9303642 0.02241122118 2637716 77.01362996 651181 P02671 Protein Single alpha chain Fibrinogen 28.521939953810627 17 0.5524271734347945 0.021934645114181275 94.91441497589778

P04040 Related Proteins Catalase 35.8633776 09108156 13 0.690714103 4735806 0.03718063723 218359 59.71875531 355211 P16157 Single Protein Ankyrin-1 13.1313131 31313131 15 0.207722655 13446264 0.00599593074 032388 206.1369244 395263 P02675 Unrelated Proteins Fibrinogen beta chain 50.3054989 8167006 16 0.887391822 1350972 0.04582730151 411469 55.89226078 115152

(40)

40

54 Proteins C4-B 75618375 45092833 24914566 9222188 I1VZV6 Single Protein Hemoglobin alpha 1 92.9577464 7887323 15 13.25102670 3029977 2.74934774039 3739 15.26995755 1186833 P02647 Related Proteins Apolipoprotei n A-I 55.4307116 10486895 17 1.801356761 1988681 0.15414244973 89475 30.75893250 0112536 Q3LR79 Single Protein Hemoglobin beta 93.3333333 3333333 15 30.62277660 1683793 1.06467847333 06542 11.47484278 9202496 A0A024R 6I7 Related Proteins Alpha-1-antitrypsin 40.1913875 5980861 13 1.198392648 8622894 0.13958117324 59462 46.67898813 8410034 Q9BWU5 Single Protein Mutant hemoglobin beta chain (Fragment) 85.7142857 1428571 13 14.19911082 9529339 1.16773237162 46961 11.49394786 3407726 D9YZU8 Single Protein Hemoglobin, gamma A 85.7142857 1428571 12 5.309573444 801933 0.24368578322 660492 16.11827504 126487 C9JC84 Related Proteins Fibrinogen gamma chain 33.8394793 92624725 10 0.687612475 7881477 0.05015322108 597598 52.30432646 809949 P00918 Related and Unrelated Proteins Carbonic anhydrase 2 48.0769230 7692308 9 1.154434690 0318838 0.08711270534 414638 29.22792218 6630536 Q4TZM4 Single Protein Hemoglobin beta chain (Fragment) 88.1188118 8118812 12 14.84893192 4611133 1.94561481731 5928 11.01563886 7792866 P02774 Unrelated Proteins Vitamin D-binding protein 36.9198312 2362869 9 0.429461338 35684486 0.01950723327 3056056 52.92902967 385641 P02790 Single Protein Hemopexin 36.5800865 8008658 11 0.778279410 0389228 0.05063651626 151628 51.64327297 890149 A0A075B 6N9 Related Proteins Ig mu chain C region (Fragment) 31.3465783 66445915 11 0.947483039 9087562 0.03887085137 085134 49.40858023 156175 P30041 Unrelated Proteins Peroxiredoxin -6 62.0535714 2857142 10 1.154434690 0318838 0.04061968537 4149665 25.01919029 056794 P06727 Single Protein Apolipoprotei n A-IV 15.1515151 51515152 5 0.242623671 9115049 0.01489071038 2513661 45.37146664 308862 P07195 Related Proteins L-lactate dehydrogenas e B chain 29.3413173 6526946 7 0.606506003 8537282 0.01930001682 6518603 36.61514296 895986 A0A087W U08 Related Proteins Haptoglobin 39.8576512 455516 9 0.995262314 9688795 0.03584200886 8324615 31.38786848 63898 P04114 Related Proteins Apolipoprotei n B-100 2.43261012 49178174 7 0.033029926 90742457 0.00166372245 1468622 515.2829109 17923 B7ZKJ8 Related and Unrelated Proteins ITIH4 protein 8.55614973 262032 5 0.139772519 80662686 0.00661953727 5064268 103.8162240 1008842 P11277 Related and Unrelated Proteins Spectrin beta chain, erythrocytic 3.04164716 89284044 5 0.043728062 944320634 0.00130995471 56802832 246.3157334 321971 P32119 Related and Unrelated Proteins Peroxiredoxin -2 49.4949494 94949495 10 1.993577294 7204895 0.16687101854 784764 21.87823782 783635 Q6MZU6 Related and Unrelated Proteins Putative uncharacteriz ed protein DKFZp686C15 213 27.5862068 96551726 9 0.701254279 8525889 0.07749920340 380885 51.06619379 122771 A0A075B 6N8 Related and Ig gamma-3 chain C region 34.4827586 2068966 9 0.750827031 7357244 0.02486747170 9576934 41.29932388 898452

Referenties

GERELATEERDE DOCUMENTEN

S-RQ4 To develop conceptual models as a representation of the findings deriving from the scoping review, that will facilitate transformation and innovation relevant to South

Zo realiseerden de bio- logische melkveebedrijven in Oostenrijk het hoogste inkomen terwijl ze veel kleiner zijn dan de biologische bedrijven in Denemarken en ook, zij het dat

vên variërend van 0,1 iot 0,4 meq/|, oílewe van vrjwe iiêt gêbuííerd tol matig gêbLrffêrd Wordt de soortensamenste Ing van beide clusteÍs veÍgeleken metde

Om te bepalen of indicatie uitbreiding van miglustat in aanmerking komt voor uitbreiding van bijlage 2 dient een oordeel te worden gegeven over de therapeutische waarde en

Wel bestaat er een digitaal kwaliteitssys- teem waarin kennis in de vorm van (handelings-) protocollen is opgeslagen. Een aanbeveling is om alle projecten die starten met

The portfolios were analyzed for a sample period of 14 years as well as sub- periods before and after the financial crisis of 2007-2008 using the Modern Portfolio Theory of

Om te kunnen voldoen aan de Europese norm voor fijnstofconcentraties in de buitenlucht dienen in Nederland maatregelen te worden doorgevoerd die de uitstoot van fijnstof (PM10,

Aangezien het vervroegen van het seizoen van 1 april naar 1 maart mogelijk wel effect kan hebben op verstoring van kwalificerende soorten, maar geen effect zal hebben op de