Secreted protein profile from HepG2 cells incubated by S(−) and R(+) enantiomers of chiral drug...

8
RESEARCH ARTICLE Secreted protein profile from HepG2 cells incubated by S(À) and R(1) enantiomers of chiral drug warfarin – An analysis in cell-based system and clinical samples Jing Bai 1 , Chi Bun Ching 1 , Balram Chowbay 2 and Wei Ning Chen 1 1 School of Chemical and Biomedical Engineering, College of Engineering, Nanyang Technological University, Singapore, Singapore 2 National Cancer Center Singapore, Singapore Received: April 9, 2010 Revised: July 14, 2010 Accepted: August 2, 2010 Purpose: Warfarin is a commonly prescribed oral anticoagulant with narrow therapeutic index. It interferes with the vitamin K cycle to achieve anti-coagulating effects. Warfarin has two enantiomers, S(À) and R(1) and undergoes stereoselective metabolism, with the S(À) enantiomer being more effective. Our target is to discover the biological differences of the two enantiomers for better warfarin therapy. Experimental design: We reported the extracellular protein profile in HepG2 cells incubated with S(À) and R(1) warfarin, using iTRAQ-coupled 2-D LC-MS/MS. In addition, clinical sera from 30 patients taken warfarin were also analyzed by the same method as a long-term batch. Results: In cell line batch in samples incubated with S(À) and R(1) warfarin alone, inter-a-trypsin inhibitor heavy chain H4, apolipoprotein A-I and a-2-HS-glycoprotein showed variations in cells incubated with S(À) warfarin and R(1) warfarin. For other proteins like a-2-macroglobulin and Fibrinogen g chain, the expressions each were found to be the same in cells incubated with either S(À) or R(1) warfarin. Clinical results showed the same trends for protein ratio changes. Conclusion and clinical relevance: Our results indicated that those proteins may interfere with blood coagulation process, as well as contribute to the warfarin’s side-effect response. Taken together, our findings provided molecular evidence on a comprehensive protein profile on warfarin–cell interaction which may shed new lights on future improvement of warfarin therapy. Keywords: a-2-HS-glycoprotein / Inter-a-trypsin inhibitor / iTRAQ-coupled LC-MS/MS proteomics / Secreted protein profile / Warfarin enantiomers 1 Introduction Warfarin is a commonly prescribed oral anticoagulant in patients with prosthetic valve disease, venous thromboem- bolism and stroke [1]. Vitamin K cycle, an essential process for the maturation of several liver-secreted blood coagulating factors, including prothrombin and factor VII, IX, X, is inhibited by warfarin. In vitamin K cycle, vitamin K is first reduced by vitamin K reductase to vitamin K hydroquinone, which acts as a mediator to activate the enzyme g-glutamyl carboxylase. g-Glutamyl carboxylase-modified several coagulating factors post-translationally, via a vitamin K-dependent way. The Glutamic acid residues on those coagulating factors are modified into g-carboxyglutamic acid (Gla), resulting in the maturation of those coagulating factors. Gla residue will mediate those coagulating factors to bind to Ca 21 in the blood stream. The binding is needed for activation of platelets as well as other downstream coagulation factors and pathways [1, 2]. Once completed the g-carboxylation reaction, vitamin K hydroquinone is oxidized to vitamin K epoxide. Another Abbreviations: DIC, disseminated intravascular coagulation; Gla, g-carboxyglutamic acid; MGP, matrix Gla protein; SCX, strong cation exchange Correspondence: Dr. Wei Ning Chen, Nanyang Technological University, School of Chemical and Biomedical Engineering, 62 Nanyang Drive, Singapore 637459, Singapore E-mail: [email protected] Fax: 165-67947553 & 2010 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.clinical.proteomics-journal.com 808 Proteomics Clin. Appl. 2010, 4, 808–815 DOI 10.1002/prca.201000027

Transcript of Secreted protein profile from HepG2 cells incubated by S(−) and R(+) enantiomers of chiral drug...

RESEARCH ARTICLE

Secreted protein profile from HepG2 cells incubated by

S(�) and R(1) enantiomers of chiral drug warfarin – An

analysis in cell-based system and clinical samples

Jing Bai1, Chi Bun Ching1, Balram Chowbay2 and Wei Ning Chen1

1 School of Chemical and Biomedical Engineering, College of Engineering, Nanyang Technological University,Singapore, Singapore

2 National Cancer Center Singapore, Singapore

Received: April 9, 2010

Revised: July 14, 2010

Accepted: August 2, 2010

Purpose: Warfarin is a commonly prescribed oral anticoagulant with narrow therapeutic

index. It interferes with the vitamin K cycle to achieve anti-coagulating effects. Warfarin has

two enantiomers, S(�) and R(1) and undergoes stereoselective metabolism, with the S(�)

enantiomer being more effective. Our target is to discover the biological differences of the two

enantiomers for better warfarin therapy.

Experimental design: We reported the extracellular protein profile in HepG2 cells incubated

with S(�) and R(1) warfarin, using iTRAQ-coupled 2-D LC-MS/MS. In addition, clinical sera

from 30 patients taken warfarin were also analyzed by the same method as a long-term batch.

Results: In cell line batch in samples incubated with S(�) and R(1) warfarin alone,

inter-a-trypsin inhibitor heavy chain H4, apolipoprotein A-I and a-2-HS-glycoprotein showed

variations in cells incubated with S(�) warfarin and R(1) warfarin. For other proteins like

a-2-macroglobulin and Fibrinogen g chain, the expressions each were found to be the same in

cells incubated with either S(�) or R(1) warfarin. Clinical results showed the same trends for

protein ratio changes.

Conclusion and clinical relevance: Our results indicated that those proteins may interfere

with blood coagulation process, as well as contribute to the warfarin’s side-effect response.

Taken together, our findings provided molecular evidence on a comprehensive protein profile

on warfarin–cell interaction which may shed new lights on future improvement of warfarin

therapy.

Keywords:

a-2-HS-glycoprotein / Inter-a-trypsin inhibitor / iTRAQ-coupled LC-MS/MS

proteomics / Secreted protein profile / Warfarin enantiomers

1 Introduction

Warfarin is a commonly prescribed oral anticoagulant in

patients with prosthetic valve disease, venous thromboem-

bolism and stroke [1]. Vitamin K cycle, an essential process

for the maturation of several liver-secreted blood coagulating

factors, including prothrombin and factor VII, IX, X, is

inhibited by warfarin.

In vitamin K cycle, vitamin K is first reduced by vitamin K

reductase to vitamin K hydroquinone, which acts as a

mediator to activate the enzyme g-glutamyl carboxylase.

g-Glutamyl carboxylase-modified several coagulating factors

post-translationally, via a vitamin K-dependent way. The

Glutamic acid residues on those coagulating factors are

modified into g-carboxyglutamic acid (Gla), resulting in the

maturation of those coagulating factors. Gla residue will

mediate those coagulating factors to bind to Ca21 in the blood

stream. The binding is needed for activation of platelets as

well as other downstream coagulation factors and pathways

[1, 2]. Once completed the g-carboxylation reaction, vitamin K

hydroquinone is oxidized to vitamin K epoxide. Another

Abbreviations: DIC, disseminated intravascular coagulation; Gla,

g-carboxyglutamic acid; MGP, matrix Gla protein; SCX, strong

cation exchange

Correspondence: Dr. Wei Ning Chen, Nanyang Technological

University, School of Chemical and Biomedical Engineering, 62

Nanyang Drive, Singapore 637459, Singapore

E-mail: [email protected]

Fax: 165-67947553

& 2010 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.clinical.proteomics-journal.com

808 Proteomics Clin. Appl. 2010, 4, 808–815DOI 10.1002/prca.201000027

enzyme, vitamin K 2,3 epoxide reductase, reduces vitamin K

epoxide to its reduced form, and the cycle repeats.

Warfarin inhibits vitamin K cycle and formation of Gla

domain through interfering with vitamin K 2,3 epoxide

reductase activity by binding to the enzyme’s disulfide

motif, thus prevents the formation of free sulfhydryls lead-

ing to the inactivation of the enzyme [3, 4]. Besides those

coagulating factors, there are a number of clearly defined

vitamin K-dependent g-carboxylation proteins that do not

response to blood coagulation, osteocalcin [5] and matrix Gla

protein (MGP) [6]. They all require Gla domains for

maturation and the maturation are inhibited by warfarin.

Warfarin is a racemic drug consists of S(�) and R(1)

enantiomers, with S(�) form being more effective in vitro(three to five times) [7, 8]. Clinically, racemic warfarin is

normally used to treat patients without enantiomeric separa-

tion, as warfarin enantiomers have activities that qualitatively

similar but quantitatively different [9]. In vivo situation indi-

cated that 60–70% anticoagulating effects in racemic warfarin

were contributed from S(�) enantiomer [7]. There are repor-

ted that steady-state doses required to maintain the same

anticoagulating effect, for S(�), racemic and R(1) warfarin

were around 6.5, 9.5 and 18 (mg/70 kg/day), respectively [10].

Racemic warfarin was in between of S(�) and R(1) warfarin

while more similar with the S(�) enantiomer. The biological

half life for racemic warfarin is also in between of S(�) and

R(1) warfarin [11]. In racemic warfarin, the effect of one

enantiomer is not altered by the presence of the other.

Proteomic analysis involves large-scale identification and

characterization of proteins. Recent developments on MS

and chromatography provide to be useful tools in protein

profile analysis. A combination system of a 2-D nano flow

LC (a strong cation exchange (SCX) column with a reversed

phase column) coupled with tandem MS (MS/MS), proves

to be more sensitive in proteomic analysis [12–14]. iTRAQ

have several advantages over other protein-labeling techni-

ques [15]. This tag allows four samples to be analyzed in one

experiment. It has proved to be a powerful tool on study

large-scale protein profiles in pharmaco-proteomics [16–18].

In this study, we used iTRAQ 4-plex strategy (with reporter

group m/z 114–117) to semi-quantify the differences in secreted

protein levels between untreated human liver cells and those

incubated by S(�) and R(1) warfarin, respectively. To better

reflect the in vivo condition, similar analysis was also carried out

in cells incubated with warfarin in the presence of vitamin K.

Protein expression was verified by Western blot analysis. In

addition, we treated 30 patients with racemic warfarin and the

proteins from their serum samples were analyzed with the

same method. This batch of experiment was designed as a

clinical long-term verification of protein secreted on warfarin

treatment. We mainly focus on understanding the biological

differences between S(�) and R(1) warfarin by analyzing the

secreted proteins from cell line incubated with individual

enantiomers of warfarin, and to identify the drug targeted

markers. In complementary, long-term clinical data gave more

evidences and support on the tendency of protein changes.

2 Materials and methods

2.1 Cell culture

HepG2 cells were purchased from American Type Culture

Collection and cultured in MEM (supplemented with 10%

FBS, penicillin (100 U/mL), and streptomycin (100 U/mL)).

All culture media and media supplements were purchased

from Life Technologies. Cells were maintained at 371C in an

atmosphere of 5% CO2. For the first set of experiment, cells

were incubated only by either S(�) or R(1) warfarin

(commercially available in Sigma Aldrich, Product No.:

UC213 and UC 214, purities Z97% (HPLC)). Once cells

were reaching 80% confluence (10 cm tissue culture dish),

the cells were incubated with either S (�) or R (1) warfarin

at a concentration of 20mM, respectively, for 24 h in the

absence of FBS. Before drug incubation, the cells were

washes by PBS for two times to avoid the contamination of

FBS. For the second set, cells were supplied with physiolo-

gical concentration of vitamin K (Sigma Aldrich) (0.5 mM)

for 12 h, then again 20 mM of either S(�) or R(1) enantio-

mer of warfarin was used to incubate the cells separately for

another 24 h, in the absence of FBS. Culture media was

obtained and centrifuged at 3000� g for 10 min to remove

dead cells and debris.

2.2 Patient selection

Thirty local patients were recruited for this pilot study.

The study population consisted of patients receiving long-term

warfarin anticoagulant therapy at the SGH Anticoagulant

Clinic. Patients with severe congestive heart failure (NYHA

class 3 or greater), liver cirrhosis and thyroid disease were

excluded from the study. Eligible patients were required to

have a stable maintenance dose of warfarin for at least 1

month (defined as no fluctuation in warfarin requirement for

1 month) and INR value variations r15% over 1 month, and

had no bleeding episodes. Patients on medication known to

interact with warfarin (NSAIDs, antiepileptics, sulphona-

mides, rifampicin and amiodarone) were excluded. The

selection of patients was approved by the Institutional Review

Board (i.e. Helsinki Declaration) to assure protection of

subjects. Serum from a healthy volunteer was collected as the

control sample for clinical group. All patients’ serum samples

were compared with the control sample.

2.3 Protein concentration, digestion and labeling

with iTRAQ reagents

In the cell line batch, culture media was transferred into

5 kD ultra-filter concentrators (Sartorius, Germany) and

centrifuged for 30 min at 41C in swing bucket rotor. The

protein concentration for the concentrated samples was

measured by 2-D Quant Kit (GE Healthcare). BSA was used

Proteomics Clin. Appl. 2010, 4, 808–815 809

& 2010 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.clinical.proteomics-journal.com

to draw a standard curve as a control. For each sample,

100 mg proteins were separated and four volumes of acetone

(Fisher Scientific) precipitation were done at �201C for 2 h.

After that, acetone was removed by centrifugation and

proteins were suspended in dissolution buffer (0.5 M tri-

ethylammonium bicarbonate). Then proteins were dena-

tured by denaturant (2% SDS), followed by cleavage of

disulfide bonds (50 mM tris-(2-carboxyethyl)phosphine). All

these reagents came from iTRAQ 4-plex Reagent Kit

(Applied Biosystems). Those steps were to make sure that all

the proteins were maintained in their denatured forms.

Then each sample was digested by 20mL of 0.25 mg/mL

sequence grade modified trypsin (Promega) solution at 371C

overnight, labeled with the iTRAQ reagents and incubated

as follows: For the first set (w/o vitamin K), HepG2 without

treatment iTRAQ 5 114; HepG2 incubated with the S (�)

enantiomer 5 iTRAQ 116; HepG2 incubated with the R(1)

enantiomer 5 iTRAQ 117. Then the three labeled samples

were mixed in one plastic tube. For the second set (w/vita-

min K), HepG2 without treatment iTRAQ 5 114; HepG2

incubated with vitamin K only 5 iTRAQ 115; HepG2 incu-

bated with vitamin K then S(�) enantiomer 5 iTRAQ 116;

HepG2 incubated with vitamin K then R(1) enantiomer 5

iTRAQ 117. Then the four labeled samples were mixed and

were ready for LC-MS/MS analysis.

For the analysis of clinical serum samples, protein

concentrating by concentrator was not necessary. Briefly,

blood samples were centrifuged, supernatant stored at

�801C until use. And serum protein was quantified using

2-D Quant Kit. For each patient sample, 100 mg proteins

were separated, precipitated and digested by the same

method above. All samples were analyzed in three inde-

pendent analyses.

2.4 2-D nano-LC/MS/MS procedure

Three individual iTRAQ experiments for cell-line batch and

clinical batch by the following protocol were performed. The

separation and detection of peptides was performed by

Agilent 1200 nanoflow LC interfaced with a QSTAR XL

mass spectrometer (Applied Biosystems/MDS Sciex)

system. In the LC separation, first, 3mL of the combined

peptide mixture was injected onto the PolySulfoethyl A SCX

column (0.32� 50 mm, 5 mm). Buffer D which is a series of

increasing concentration of KCl salt solution from 10 mM to

1 M (10, 20, 30, 40, 50, 60, 80, 100, 300, 500, 1000 mM) used

to elute the retained peptides in a stepwise manner from the

SCX column by sequential injection [16, 17]. Each KCl

elution of peptides took up 100 min. Totally, eleven runs

made up a whole program. In the first run (Position 1),

some of the peptides bound to the SCX column during the

flow, whereas those did not bind were trapped in the

ZORBAX 300SB-C18 enrichment column (trap column)

(0.3� 5 mm, 5 mm) and washed by buffer A (5% ACN, 0.1%

formic acid) at 0.5 mL/min to remove the excess reagent.

There were two enrichment columns in total, which was

alternatively switched into the solvent path of the nano

pump with positions 1 and 2, between the eleven runs by a

10-port valve. In the second run with an increased of KCL

solution (Position 2), the 10-port switching valve switched to

another position. The previous enrichment column which

trapped the unbound peptides in the first run was switched

into the solvent path of the nanopump. Peptides were eluted

using the buffer B (0.1% formic acid) and the buffer C (95%

ACN, 0.1% formic acid) with a nanoflow gradient starting

with 5% of the buffer C and increasing up to 80% of the

same buffer C over 100 min at a flow rate of 500 nL/min.

This gradient wash was to make sure most of the peptides

could be eluted out and further separation was achieve in

analytical Zorbax 300SB C-18 reversed-phase column

(analytical column, 75mm� 50 mm, 3.5 mm), which sepa-

rated peptides according to their hydrophobic–hydrophilic

properties. In the MS detection, the ionization method was

electrospray ionization. Survey scans were acquired from

m/z 300 to 1500 with up to two precursors whose intensity

415 were selected for MS/MS analysis. In addition, the

same peptide could not be selected within 60 s. The MS/MS

was scanned from m/z 100 to 2000 using dynamic exclu-

sion, and the rolling collision energy was used to promote

fragmentation. Finally, detection was performed on the

AnalystQS (Applied Biosystems) software for every 100-min

run coming from LC.

2.5 Protein identification and data analysis

ProteinPilotTM 2.0(Applied Biosystems, Software Revision

50861) was used to analyze iTRAQ experiments. Each MS/

MS spectrum was identified using MS/MS data interpreta-

tion algorithms in the software and then searched against

the Swiss-Prot protein database for the species of Homosapiens (UniProt_sprot_20070123). The searches were

conducted under the following parameters: fixed modifica-

tion of methylmethanethiosulfate-labeled cysteine, fixed

iTRAQ modification of free amine in the amino terminus

and lysine, variable iTRAQ modifications of tyrosine and

allowing serine and threonine residues undergoing side

reaction with the iTRAQ reagent. Other parameters such as

tryptic cleavage specificity, allowance for number of missed

cleavage, parent ion and fragment ion mass accuracy are

built-in functions of ProteinPilot software. As it is described

in the protocol from our previous study [19], relative protein

quantification of iTRAQ was calculated by the ratio of the

areas under the peaks at 114, 115, 116 and 117 Da. A total of

114, 115, 116 and 117 were the masses of iTRAQ tags

reagents, respectively. The relative amount of a peptide in

each sample was calculated by dividing the peak areas

observed at m/z 116 and 117 by 114 (HepG2 cells incubated

with warfarin only); and m/z 114, 116, 117 by 115 (HepG2

cells incubated with warfarin in the presence of vitamin K).

The following criteria were set up: unused protein score was

810 J. Bai et al. Proteomics Clin. Appl. 2010, 4, 808–815

& 2010 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.clinical.proteomics-journal.com

more than 1.3 (95% confidence) per experiment, p-value had

to be less than 0.05; at least two unique peptides

whose confidence was more than 95% needed to be

detected; and protein quantification data with relative

expression of 41.1 or o0.9 was chosen. As for certain

proteins, 10% changes may result in significant changes in

protein functions. The error factor, which is a statistic value

describing a peptide’s 95% confidence interval, must be less

than 2 for the detected proteins. The proteins detected were

examined by Protein ID of the ProteinPilot software.

Those peptides without any modification of free amine in

the amino terminus or without iTRAQ modification were

not selected for calculation of the protein ratios. The

generated ratios of all detected proteins were normalized by

bias correction and other built-in function tools in

the software, to correct the small differences in protein

loading.

2.6 Western blot analysis for culture media proteins

Total protein secreted from HepG2 culture media extracted

from the experiments for the LC-MS/MS analysis was used

in the Western blot analysis. Apolipoprotien A-I detected in

LC-MS/MS were analyzed by Western blot. The separation

was performed by 12% SDS-PAGE. This protein expression

was examined on two individual sets: set 1, R(1) enantio-

mer, and S(�) enantiomer-incubated cells without the

presence of vitamin K; set 2, R(1) enantiomer, and the S(�)

enantiomer-incubated cells in the presence of vitamin K.

Then protein was transferred onto nitrocellulose

membrane, and the detection of the respective protein was

carried out by using specific antibody (Santa Cruz sc-69755).

The results were visible using Supersignal West solutions

(1856136, Pierce). The developed X-ray film was then scan-

ned using Densitometer (Bio-Rad) and analyzed by Qual-

ityOne software (Bio-Rad).

3 Results and discussion

To better understand the warfarin-induced anti-coagulating

effects, the secretome of warfarin incubated hepatocytes was

analyzed as most liver-originated coagulation related

proteins are secreted. To this end, secreted proteins were

isolated from cells incubated with individual enantiomers of

warfarin and subject to 2-D LC-MS/MS analysis. A total of

76 unique proteins, in which 49 were secreted proteins,

were obtained together for both sets with and without the

presence of vitamin K. A total of five proteins showed

changes in their level. The low number of detected proteins

in the culture medium was consistent with the nature of our

analysis, namely the culture medium which contains far less

proteins compared with the intracellular cell environment,

which was around 600 unique proteins in our previous

intracellular study [19]. Tab

le1.

Pro

tein

sid

en

tifi

ed

ince

ll-l

ine

base

dexp

eri

men

ts

Warf

ari

nW

arf

ari

n1

vit

am

inK

Acc

ess

ion

nu

m.

an

dn

am

e

%C

ov.

Avg

.S:C

(SD

)p

(S:C

)A

vg

.R:C

(S.D

.)p

(R:C

)N

um

.o

fd

ete

ctio

nN

um

.o

f95%

pep

tid

es

%C

ov.

Avg

.S:V

(SD

)p

(S:V

)A

vg

.R:V

(S.D

.)p

(R:V

)N

um

.o

fd

ete

ctio

nN

um

.o

f95%

pep

tid

es

P01023a-

2-m

acr

og

lob

uli

n48%

1.1

67

0.1

50.0

81.1

97

0.0

20.0

52

629.5

%1.17

0.0

80.0

41.0

97

0.0

50.0

32

6P

02765a-

2-H

S-g

lyco

pro

tein

52%

0.8

67

0.0

10.0

20.9

27

0.0

30.0

13

443.1

%0.7

97

0.0

40.0

10.9

37

0.0

50.0

72

4P

02647

Ap

oli

po

pro

tein

A-I

63.7

%0.87

0.0

80.0

60.7

37

0.0

20.0

22

340.5

%0.8

27

0.0

10.0

20.7

67

0.0

30.0

12

3P

02679

Fib

rin

og

eng

chain

14.8

%0.8

87

0.0

70.0

50.8

67

0.0

50.0

62

2–

––

12

Q14624

Inte

r-a-

tryp

sin

inh

ibit

or

heavy

chain

H4

43.6

%1.1

17

0.0

40.0

41.0

47

0.0

40.0

42

2–

––

12

Left

,w

arf

ari

n,

Pro

tein

sid

en

tifi

ed

at

least

twic

ean

dh

ave

exp

ress

ion

rati

ovari

ed

at

least

10%

(po

0.0

5),

inth

ese

to

fexp

eri

men

tw

ith

ou

tV

itam

inK

pre

-in

cub

ati

on

were

incl

ud

ed

.C

5co

ntr

olg

rou

p,S

5S

(�)

warf

ari

nin

cub

ate

dg

rou

p,R

5R

(1)

warf

ari

nin

cub

ate

dg

rou

p.In

this

tab

le,th

eco

ntr

olg

rou

pis

the

un

treate

dsa

mp

le.R

igh

t,W

arf

ari

n1

vit

am

inK

,P

rote

ins

iden

tifi

ed

at

least

twic

ean

dh

ave

exp

ress

ion

rati

ovari

ed

at

least

10%

(po

0.0

5),

inth

ese

to

fexp

eri

men

tw

ith

Vit

am

inK

pre

-in

cub

ati

on

.V

5vit

am

inK

-tre

ate

dg

rou

p,

S5

vit

am

inK

incu

bate

dth

en

S(�

)w

arf

ari

nin

cub

ate

dg

rou

p,

R5

vit

am

inK

incu

bate

dth

en

R(1

)w

arf

ari

nin

cub

ate

dg

rou

p.

Inth

ista

ble

,th

eco

ntr

ol

gro

up

isth

evit

am

inK

-tre

ate

dsa

mp

le.

%co

v5

perc

en

tag

eo

fco

vera

ge.

Avg

.S:C

5A

vera

ge

S:C

;A

vg

.R:C

5A

vera

ge

R:C

.A

vg

.S:V

5A

vera

ge

S:V

;A

vg

.R:V

5A

vera

ge

R:V

.N

um

.o

fd

ete

ctio

n5

Nu

mb

er

of

dete

ctio

no

fa

part

icu

lar

pro

tein

inth

eth

ree

ind

ep

en

den

tLC

-MS

/MS

run

s.N

um

.o

f95%

pep

tid

es

5N

um

ber

of

95%

pep

tid

es

dete

cted

ina

part

icu

lar

pro

tein

.

Proteomics Clin. Appl. 2010, 4, 808–815 811

& 2010 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.clinical.proteomics-journal.com

Results showed that most proteins displayed similar

changes in cells incubated with either S(�) or R(1)

warfarin. Some of the proteins showed changes to one but

not both enantiomers of warfarin. Results from the selection

are summarized in Table 1 for cells incubated with the

individual enantiomers of warfarin, and cells incubated with

the individual enantiomers of warfarin in the presence of

vitamin K. Most proteins displayed changes in their level

varying from 15 to 25%. A peptide fragment information

from a-2-HS glycoprotein (in the set with the presence of

vitamin K) is shown in Fig. 1.

The secretome analysis in our cell-based system provided

us with information on proteins that were secreted at the

early stage of warfarin incubation. Conversely, similar

analysis from patients receiving warfarin therapy would

provide valuable validation on the significance of the early

secretome in long-term warfarin therapy. To this end, we

extracted serum proteins from patients who have been

treated with warfarin for more than 1 month. The warfarin

used in the therapy was of racemic nature, as it no warfarin

in individual enantiomers was available for clinical use. It

was nevertheless useful for verification of our cell-based

secretome analysis.

A total of 163 unique proteins were identified in the

clinical samples. Interestingly, the five proteins which were

detected in the cell line batch were also detected in the

clinical batch (Fig. 2). A Box and Whisker Plot was drawn to

explain the clinical data distribution. The detailed data of

serum protein expression from 30 patients can be found in

Supporting Information. The fact that the tendency of either

up- or down-regulated for a particular protein was the same

in both settings indicated that changes in these five proteins

were related to warfarin treatment instead of random

stimulations. However, the exact ratio of protein expressions

in cell line and clinical batch was different probably as a

result of the intrinsic differences between cell lines and

patients or the racemic warfarin in clinical samples. Quan-

tification assessment was performed by ProteinPilot soft-

ware. Protein profiles generated by this software were

undergone a filtering, by statistical criteria described in

Section 2. The list of proteins in both cell-line and clinical

samples with the list of 95% peptides, charged state and

protein functions was summarized in the Supporting

Information.

3.1 Up-regulation of Inter-a-trypsin inhibitor heavy

chain H4 and a-2-macroglobulin

There were two proteins inter-a-trypsin inhibitor heavy

chain H4 and a-2-macroglobulin up-regulated regardless of

the presence of vitamin K. In cell line batch, inter-a-trypsin

inhibitor heavy chain H4 was up-regulated slightly in cells

Figure 1. A representative MS/

MS spectrum of a peptide,

CDSSPDSAEDVR. It was iden-

tified in the cell-line batch, with

the presence of vitamin K. The

m/z 114 represents untreated

sample, m/z 115 represents Vk

incubated sample, m/z 116

represents S(�) warfarin incu-

bated sample after Vk expo-

sure, m/z 117 represents R(1)

warfarin incubated sample

after Vk exposure.

Figure 2. Pharmacoproteomics analysis of patients receiving

warfarin. Five proteins identified in the cell line batch were

verified by clinical data. Two of them were up-regulated,

whereas the rest were down-regulated.

812 J. Bai et al. Proteomics Clin. Appl. 2010, 4, 808–815

& 2010 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.clinical.proteomics-journal.com

incubated with S(�) warfarin (11%) compared to R(1)

warfarin (4%). In clinical batch, the protein was also up-

regulated in all patients. A more detailed data can be found

in on-line Supporting Information. Inter-a-trypsin inhibitor

is one of the plasma serine-protease inhibitor [20], with no

reported link to the effect of warfarin. Recent evidence

suggests that this protein may well play a role in the process

of anticoagulation. First, the coagulation factors are gener-

ally serine proteases. As an inhibitor, inter-a-trypsin inhi-

bitor may interact with coagulating factors thus inhibit their

normal functions. Second, the substrate of plasma kalli-

krein, a serine protease that generates plasmin from plas-

minogen to enhance blood coagulation, has a significant

sequence identity to heavy chains of the inter-a-trypsin

inhibitor superfamily [21]. Third, it has been known that

this protein acts as a probable component pertaining to

disseminated intravascular coagulation (DIC), which is a

disease leading to the formation of small blood clots inside

the blood vessels throughout the body. The DIC patients

have lower level of this protein expression compared with

normal group [22]. Warfarin has been used to treat patients

with DIC to prevent further formation of blood clotting [23].

In our study, the elevated protein expression level may

associate with warfarin treatment. We thus proposed that

the expression level of inter-a-trypsin inhibitor may be

related to warfarin therapy.

The other up-regulated protein detected in our analysis

was a-2-macroglobulin. The up-regulation was observed in

cells incubated with either S(�) or R(1) warfarin compared

with control cells (absence of warfarin), but no significant

changes between cells incubated with S(�) warfarin and

those incubated with R(1) warfarin. Clinical data gave

support on the tendency of the protein changes, although a

decreased level was observed in 6 of the 30 clinical samples

(shown in the Box and Whisker plot and in Supporting

Information). The inconsistency may be a result of intrinsic

differences among patients selected for this analysis. This

protein is synthesized in the liver and secreted into plasma.

It plays an important role in blood coagulation. Although

this protein is not a vitamin K-dependent protein, it acts as

an inhibitor of coagulation by inhibiting thrombin forma-

tion [24]. Prothrombin, the precursor of thrombin, is a

vitamin K-dependent coagulating factor, whose expression is

inhibited by warfarin. Thus the elevation of this protein may

reflect the thrombin regulation by warfarin. It has also been

reported that elevation of this protein level may in part

provide protection from thrombosis [25].

3.2 Down-regulation of a-2-HS-glycoprotein,

apolipoprotein A-I and fibrinogen c chain

There were three down-regulated proteins in both cell line

and clinical samples. For a-2-HS-glycoprotein, it was down-

regulated in cells incubated with S(�) warfarin and to a

greater extent compared with those incubated with R(1)

warfarin. Our results therefore suggested a differential

effect of warfarin enantiomers on a-2-HS-glycoprotein.

Clinically, this protein also showed down-regulated for 15%

in average. However, the up-regulation was found in 2 of the

30 clinical samples. The inconsistency may be due to the

intrinsic differences between patients.

This protein is secreted by hepatocytes. It forms a

complex with MGP and acts as a carrier for this insoluble

protein in serum [26, 27]. MGP has been shown to be an

inhibitor of vascular calcification [28]. It is a protein that

requires vitamin K dependent-g-carboxylation for its activa-

tion. This activation is inhibited by warfarin. It has been

reported in animal model that warfarin induces vascular

calcification by inhibiting MGP expression [29–31]. In our

study, the down-regulation of a-2-HS-glycoprotein upon

warfarin incubation may associate with reduced MGP

expression. As detection of this insoluble MGP in proteo-

mics is difficult, identification of its carrier instead would be

an alternative way to monitor the side-effect of warfarin. Our

study also provided evidences on S(�) and R(1) warfarin

may contribute differently in warfarin’s side effect on

vascular calcification.

Another down-regulated protein, apolipoprotein A-I, was

down-regulated in cells incubated with S(�) warfarin to a

lesser extent compared with those incubated with R(1)

warfarin. To examine whether the differences in the changes

Figure 3. Western blot analysis of protein expression for the

protein apolipoprotein A-I. (A) Protein expression in the

experiment without vitamin K pre-incubation. (B) Protein

expression in the experiment with vitamin K pre-incubation.

Ctrl 5 sample without any treatment, S-warf 5 the sample incu-

bated by S(�) warfarin, R-warf 5 the sample incubated by R(1)

warfarin; Vk 5 the sample only incubated by vitamin K, Vk1S-

warf 5 the sample incubated by vitamin K then S(�) warfarin, Vk

1R-warf 5 the sample incubated by vitamin K then R(1)

warfarin.

Proteomics Clin. Appl. 2010, 4, 808–815 813

& 2010 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.clinical.proteomics-journal.com

were significant, Western blot analysis was carried out for this

protein (Fig. 3). Results were obtained from cells incubated

with individual enantiomers of warfarin and those incubated

with individual enantiomers of warfarin in the presence of

vitamin K. Consistently with the LC-MS/MS analysis, apoli-

poprotein A-I showed a reduced level in cells incubated with

warfarin, compared with the control cells. Our Western blot

analysis therefore confirmed that the level of this protein was

down-regulated more in cells incubated with R(1) warfarin

compared to those incubated with S(�) warfarin.

Taken together, the change identified in LC-MS/MS

analysis was supported by our Western blot analysis.

Quantitative information calculated is shown in Fig. 3. Our

results therefore suggested a differential effect of warfarin

enantiomers on apolipoprotein A-I. The inconsistency

observed in the clinical samples was similar to a-2-HS-

glycoprotein, in that the up-regulation was found in 10 of

the 30 clinical samples. Again, this could be a result of

intrinsic differences among individual patients.

Apolipoprotein A-I, is a well-known major component of

high density lipoprotein in the plasma and involved in the

transportation of plasma lipids [32]. It has been reported that

elevated plasma lipids appear to be procoagulant and may be

associated with either clinically significant thrombosis or

elevated haemostatic markers of hypercoagulability [33].

Thus, the down-regulation of apolipoprotein A-I may indicate

that the enhancement of anticoagulation effects by warfarin.

Our results also showed that Fibrinogen g chain was

down-regulated without any differences in cells incubated

with either S(�) or R(1) warfarin, regardless of the presence

of vitamin K. In clinical study, this protein was also down-

regulated with a slightly larger ratio (40%). Fibrinogen

which is synthesized in the liver is a major component in

blood coagulation involving in formation of blood clots. The

down-regulation of this protein indicated an enhanced level

of anticoagulation by warfarin.

4 Concluding remarks

We reported the extracellular protein profile in HepG2 cells

incubated with S(�) and R(1) warfarin, using iTRAQ-

coupled 2-D LC-MS/MS and followed by validation with

Western blot analysis. In addition, the observed warfarin-

associated protein changes were analyzed in serum samples

of patients receiving warfarin therapy. Our results indicated

that inter-a-trypsin inhibitor heavy chain H4, apolipoprotein

A-I and a-2-HS-glycoprotein showed changes in cells

incubated with either S(�) or R(1) warfarin. For other

proteins like a-2-macroglobulin and Fibrinogen g chain, the

changes were observed in cells incubated with either S(�)

or R(1) warfarin compared with control cells (absence of

warfarin), but no significant changes between cells incubated

with S(�) warfarin and those incubated with R(1) warfarin.

Our results indicated that those proteins may interfere with

blood coagulation process, and possibly contribute to the

warfarin’s side effect on vascular calcification. Taken toge-

ther, our findings provided molecular evidence on a secreted

protein profile on warfarin–cell interaction which may shed

new lights on future improvement of warfarin therapy.

This work was supported by NMRC/EDG/0033/2008 (WNChen). J. Bai is a recipient of graduate research scholarship fromNanyang Technological University, Singapore.

The authors have declared no conflict of interest.

5 References

[1] Hirsh, J., Dalen, J. E., Anderson, D. R., Poller, L. et al., Oral

anticoagulants: mechanism of action, clinical effectiveness,

and optimal therapeutic range. Chest 2001, 119, 8S–21S.

[2] Wu, S. M., Cheung, W. F., Frazier, D., Stafford, D. W.,

Cloning and expression of the Cdna for human gamma-

glutamyl carboxylase. Science 1991, 254, 1634–1636.

[3] Tie, J. K., Stafford, D. W., Vitamin K, Elsevier Academic

Press Inc, San Diego 2008, pp. 103–130.

[4] Wajih, N., Hutson, S. M., Wallin, R., Disulfide-dependent

protein folding is linked to operation of the vitamin K cycle

in the endoplasmic reticulum - A protein disulfide isomer-

ase-VKORC1 redox enzyme complex appears to be

responsible for vitamin K-1 2,3-epoxide reduction. J. Biol.

Chem. 2007, 282, 2626–2635.

& 2010 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.clinical.proteomics-journal.com

Clinical Relevance

This manuscript describes the biological effects

differences of S(�) and R(1) warfarin. Clinically, this

drug is widely used to prevent prosthetic valve

disease, venous thromboembolism and stroke.

However, warfarin has a very wide therapeutic range.

There are many factors which have been proved to

affect warfarin clinical dosage. Enantiomer drugs

interaction with the body has been found to be

responsible for dosage variation to some extent. Thus

we analyzed both cell-line-based secreted proteins

and proteins from clinical patients’ sera, to identify

candidate drug-stimulated biomarkers, as well as, to

recognize protein markers which might contribute to

warfarin’s side effect. The design of both short-term

(cell-line) and long-term (clinical) experiments has

further confirmed the protein variation. These find-

ings may contribute to the improvement of warfarin

therapy in terms of optimal dosage.

814 J. Bai et al. Proteomics Clin. Appl. 2010, 4, 808–815

[5] Price, P. A., Otsuka, A. S., Poser, J. W., Kristaponis, J.,

Raman, N., Characterization of a gamma-carboxyglutamic

acid-containing protein from bone. Proc. Natl. Acad. Sci.

USA 1976, 73, 1447–1451.

[6] Price, P. A., Williamson, M. K., Primary structure of bovine

matrix Gla protein, a new vitamin-K-dependent bone

protein. J. Biol. Chem. 1985, 260, 4971–4975.

[7] Takahashi, H., Echizen, H., Pharmacogenetics of warfarin

elimination and its clinical implications. Clin. Pharmacoki-

net. 2001, 40, 587–603.

[8] Fasco, M. J., Principe, L. M., R-warfarin and S-warfarin

inhibition of vitamin-K and vitamin-Kz2,3-epoxide reductase

activities in the Rat. J. Biol. Chem. 1982, 257, 4894–4901.

[9] Rentsch, K. M., The importance of stereoselective determi-

nation of drugs in the clinical laboratory. J. Biochem.

Biophys. Methods 2002, 54, 1–9.

[10] Wingard, L. B., Oreilly, R. A., Levy, G., Comparative phar-

macokinetics of coumarin anticoagulants. 34. Pharmacoki-

netics of warfarin enantiomers – search for intrasubject

correlations. Clin. Pharmacol. Ther. 1978, 23, 212–217.

[11] Oreilly, R. A., Studies on optical enantiomorphs of warfarin

in man. Clin. Pharmacol. Ther. 1974, 16, 348–354.

[12] Lafaye, A., Junot, C., Ramounet-Le Gall, B., Fritsch, P. et al.,

Profiling of sulfoconjugates in urine by using precursor ion

and neutral loss scans in tandem mass spectrometry.

Application to the investigation of heavy metal toxicity in

rats. J. Mass Spectrom. 2004, 39, 655–664.

[13] Jackson, P. J., Brownsill, R. D., Taylor, A. R., Walther, B.,

Use of electrospray-ionization and neutral loss liquid-chro-

matography tandem mass-spectrometry in drug-metabo-

lism studies. J. Mass Spectrom. 1995, 30, 446–451.

[14] Jemal, M., Ouyang, Z., Zhao, W. P., Zhu, M. S., Wu, W. W., A

strategy for metabolite identification using triple-quadru-

pole mass spectrometry with enhanced resolution and

accurate mass capability. Rapid Commun. Mass Spectrom.

2003, 17, 2732–2740.

[15] Zappacosta, F., Annan, R. S., N-terminal isotope tagging

strategy for quantitative proteomics: results-driven analysis of

protein abundance changes. Anal. Chem. 2004, 76, 6618–6627.

[16] Sui, J. J., Tan, T. L., Zhang, J. H., Ching, C. B., Chen, W. N.,

ITRAQ-coupled 2D LC-MS/MS analysis on protein profile in

vascular smooth muscle cells incubated with S- and

R-enantiomers of propranolol: possible role of metabolic

enzymes involved in cellular anabolism and antioxidant

activity. J. Proteome Res. 2007, 6, 1643–1651.

[17] Sui, J. J., Zhang, J. H., Tan, T. L., Ching, C. B., Chen, W. N.,

Comparative proteomics analysis of vascular smooth

muscle cells incubated with S- and R-enantiomers of

atenolol using iTRAQ-coupled two-dimensional LC-MS/MS.

Mol. Cell. Proteomics 2008, 7, 1007–1018.

[18] Zhang, J. H., Sui, J. J., Ching, C. B., Chen, W. N., Protein

profile in neuroblastoma cells incubated with S- and R-

enantiomers of ibuprofen by iTRAO-coupled 2-D LC-MS/MS

analysis: Possible action of induced proteins on Alzheimer’s

disease. Proteomics 2008, 8, 1595–1607.

[19] Bai, J., Sadrolodabaee, L., Ching, C. B., Chowbay, B., Chen,

W. N., A comparative proteomic analysis of HepG2 cells

incubated by S(-) and R(1) enantiomers of anti-coagulating

drug warfarin. Proteomics 2010, 10, 1463–1473.

[20] Flahaut, C., Mizon, C., Aumercier-Maes, P., Colson, P. et al.,

Disulphide bonds assignment in the inter-alpha-inhibitor

heavy chains - Structural and functional implications. Eur. J.

Biochem. 1998, 255, 107–115.

[21] Nishimura, H., Kakizaki, I., Muta, T., Sasaki, N. et al., Cdna

and deduced amino-acid-sequence of human Pk-120, a

plasma kallikrein-sensitive glycoprotein. FEBS Lett. 1995,

357, 207–211.

[22] Ishizaki, T., Hashizume, K., Matsubara, F., Tsukuda, K. et al.,

Inter-alpha-trypsin inhibitor – as a probable component

pertaining to disseminated intravascular coagulation (Dic).

Acta Haematologica Japonica 1980, 43, 587–592.

[23] Chen, K. T., Lin, H. J., Lee, W. J., Severe disseminated

intravascular coagulation caused by congestive heart fail-

ure and left ventricular thrombus. Eur. J. Emerg. Med. 2007,

14, 87–89.

[24] Deboer, J. P., Creasey, A. A., Chang, A., Abbink, J. J. et al.,

Alpha-2-macroglobulin functions as an inhibitor of fibrino-

lytic, clotting, and neutrophilic proteinases in sepsis – studies

using a baboon model. Infect. Immun. 1993, 61, 5035–5043.

[25] Cvirn, G., Gallistl, S., Muntean, W., Effects of alpha(2)-

macroglobulin and antithrombin on thrombin generation

and inhibition in cord and adult plasma. Thromb. Res. 2001,

101, 183–191.

[26] Price, P. A., Thomas, G. R., Pardini, A. W., Figueira, W. F.

et al., Discovery of a high molecular weight complex of

calcium, phosphate, fetuin, and matrix gamma-carbox-

yglutamic acid protein in the serum of etidronate-treated

rats. J. Biol. Chem. 2002, 277, 3926–3934.

[27] Wajih, N., Wallin, R., Identification of alpha 2-HS glycopro-

tein as a human serum carrier of matrix Gla protein (MGP)

and evidence for vitamin K4 as a modulator of gene

expression in proliferating vascular smooth muscle cells.

FASEB J. 2006, 20, A134–A134.

[28] Wajih, N., Borras, T., Xue, W., Hutson, S. M., Wallin, R.,

Processing and transport of matrix gamma-carbox-

yglutamic acid protein and bone morphogenetic protein-2

in cultured human vascular smooth muscle cells – evidence

for an uptake mechanism for serum fetuin. J. Biol. Chem.

2004, 279, 43052–43060.

[29] Price, P. A., Faus, S. A., Williamson, M. K., Warfarin-induced

artery calcification is accelerated by growth and vitamin D.

Arterioscler. Thromb. Vasc. Biol. 2000, 20, 317–327.

[30] Danziger, J., Vitamin K-dependent proteins, warfarin, and

vascular calcification. Clin. J. Am. Soc. Nephrol. 2008, 3,

1504–1510.

[31] Price, P. A., Faus, S. A., Williamson, M. K., Warfarin causes

rapid calcification of the elastic lamellae in rat arteries and

heart valves. Arterioscler. Thromb. Vasc. Biol. 1998, 18,

1400–1407.

[32] Duriez, P., Fruchart, J. C., High-density lipoprotein

subclasses and apolipoprotein A-I. Clin. Chim. Acta 1999,

286, 97–114.

[33] Miller, G., Lipoproteins and thrombosis: effects of lipid

lowering. Curr. Opin. Lipidol. 1995, 6, 38–42.

Proteomics Clin. Appl. 2010, 4, 808–815 815

& 2010 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.clinical.proteomics-journal.com