Proteomic characterization of epithelial-like extracellular vesicles in

Dec 26, 2018 - Proteomic characterization of epithelial-like extracellular vesicles in advanced endometrial cancer ... cell line and characterized the...
1 downloads 0 Views 1MB Size
Subscriber access provided by The University of British Columbia Library

Article

Proteomic characterization of epithelial-like extracellular vesicles in advanced endometrial cancer Javier Mariscal, Patricia Fernández-Puente, Valentina Calamia, Alicia Abalo, Maria Santacana, Xavier Matias-Guiu, Rafael Lopez-Lopez, Antonio Gil-Moreno, Lorena Alonso-Alconada, and Miguel Abal J. Proteome Res., Just Accepted Manuscript • DOI: 10.1021/acs.jproteome.8b00750 • Publication Date (Web): 26 Dec 2018 Downloaded from http://pubs.acs.org on January 1, 2019

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 34 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

1

2

Proteomic characterization of epithelial-like

3

extracellular vesicles in advanced endometrial

4

cancer

5 6

Javier Mariscal1, Patricia Fernandez-Puente2, Valentina Calamia2, Alicia Abalo1,

7

Maria Santacana3, Xavier Matias-Guiu3, Rafael Lopez-Lopez1, Antonio Gil-Moreno4,

8

Lorena Alonso-Alconada1, and Miguel Abal1

9 10

1

11

(IDIS), University Hospital of Santiago (SERGAS), Santiago de Compostela, Spain

12

2

13

Investigación Biomédica de A Coruña (INIBIC), University Hospital A Coruña

14

(SERGAS), Universidade da Coruña, A Coruña, Spain

15

3

16

CIBERONC, Lleida, Spain

17

4

18

Universitat Autonoma de Barcelona, CIBERONC, Barcelona, Spain

Translational Medical Oncology, CIBERONC, Health Research Institute of Santiago

Proteomics Group-PBR2-ProteoRed/ISCIII, Rheumatology Division, Instituto de

Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLLEIDA,

Biomedical Research Group in Gynecology, Vall Hebron Research Institute (VHIR),

19 20 21

1 ACS Paragon Plus Environment

Journal of Proteome Research 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 2 of 34

22

ABSTRACT: Endometrial cancer (EC) is the most frequent gynecological cancer.

23

Tumor dissemination affecting around 20% of EC patients is characterized at the primary

24

carcinoma by epithelial-to-mesenchymal transition (EMT) associated with myometrial

25

infiltration. At distant sites, the interaction of circulating tumor cells (CTCs) with the

26

microenvironment results crucial for metastatic colonization, with a participation of the

27

extracellular vesicles (EVs). We comprehensively approached these primary and

28

secondary sites to study the impact of tumor EVs on the metastatic efficiency of CTCs in

29

EC. Tumor EVs in circulation reproduce the epithelial phenotype predominant in the

30

primary carcinoma, while CTCs are characterized by an EMT phenotype. We modelled

31

this EMT-related clinical scenario in the Hec1A endometrial cell line and characterized

32

the epithelial-like EVs in circulation by SILAC proteome analysis. Identification of

33

proteins involved in cell-cell and cell-matrix interaction and binding, together with in

34

vitro evidences of an improved adhesion of CTC to a functionalized endothelium suggest

35

a contribution of the epithelial-like EVs in the homing of CTCs at metastatic sites.

36

Accordingly, adhesion protein LGALS3BP was found significantly enriched in

37

circulating EVs from a cohort of EC patients with high risk of recurrence by targeted

38

proteomics (MRM), highlighting their potential as liquid biopsy in EC.

39 40 41

KEYWORDS: Endometrial metastasis; EVs proteomics; SILAC; Circulating Tumor

42

Cells.

2 ACS Paragon Plus Environment

Page 3 of 34 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

43

1. INTRODUCTION

44

Endometrial Cancer (EC) constitutes the fourth leading cancer in women with more than

45

60,000 new cases and 10,000 deaths estimated in the United States in 2018.1 EC originates

46

in the inner layer of the uterus when epithelial cells lining the myometrium start to

47

proliferate abnormally and, although most EC are diagnosed early, up to 20% of the

48

lesions progress to a high-grade carcinoma.1 Compared to 90% in patients diagnosed with

49

confined disease, 5-year survival in advanced disease drops up to 15% as a result of

50

primary myometrial infiltration followed by dissemination of aggressive tumor cells.2

51

Myometrial invasion results from an epithelial-to-mesenchymal (EMT) process

52

characterized by a switch from E-Cadherin-positive epithelial tumor cells, strictly

53

organized in glands, transitioning into mesenchymal-like tumor cells able to migrate and

54

invade the surrounding myometrium and reach the vasculature.3 The presence of these

55

tumor cells in circulation, or CTCs in peripheral blood, has been recently reported by our

56

group and others in high risk-EC patients with the FDA-approved CellSearch®

57

technology.4–6 Notably, the molecular characterization of these disseminated tumor cells

58

has identified an EMT signature characterized by a plasticity phenotype.6 EMT at the

59

invasive front of EC is a well described event driving cancer cell aggressiveness, therapy

60

resistance and metastasis.7

61

Alternatively, tumor-secreted extracellular vesicles (EVs) have been identified as main

62

mediators of cell-to-cell communication between tumor and stromal cells both in local

63

and distant microenvironments participating in the formation of the pre-metastatic niche

64

prior to CTC colonization.8 Tumor exosomes, a subpopulation of 40-150 nm EVs released

65

from the tumor cells following the fusion of multivesicular late endosomes / lysosomes

66

with the plasma membrane, have been shown to be representative of the tumor of origin9

67

and important mediators of intercellular signaling including EMT and the tropism of

3 ACS Paragon Plus Environment

Journal of Proteome Research 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 4 of 34

68

disseminated disease to pre-conditioned tissues.10 In this scenario, EC-derived EVs may

69

not only be carriers of molecules of utility for early diagnosis and monitoring of patients,

70

but also actively participate in the recruitment of tumor supporting cells and CTCs in a

71

pro-metastatic microenvironment. Recent discoveries have reported increased secretion

72

of EVs associated with high-grade disease10–12 and the biological relevance of tumor EVs

73

in disease progression and metastatic potential.13 However, identification of the cell of

74

origin and biological profiling of circulating tumor EVs remains challenging.

75

To study this, we comprehensively approached EMT in EC and the contribution of the

76

epithelial and mesenchymal components to the tumor EVs shed in circulation. By

77

combining EMT modelling and quantitative mass spectrometry analysis by stable isotope

78

labeling with amino acids in cell culture (SILAC), we were able to characterize the

79

predominant epithelial-like tumor EVs. We further explored by multiple reaction

80

monitoring (MRM) the potential of these EVs as biomarkers of disease in plasma from a

81

cohort of EC patients with high risk of recurrence.

82

4 ACS Paragon Plus Environment

Page 5 of 34 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

83

2. MATERIALS & METHODS

84

2. 1. Histology and Immunohistochemistry analysis & cell culture of EC cell lines.

85

EC tissue sections from patients (n= 5) were selected where the non-invasive, the

86

superficial invasive, the invasive front and areas with deep invasion could be

87

distinguished. Consecutive sections were obtained from tissue blocks, sectioned at a

88

thickness of 3 μm, dried for 1h at 65ºC before pre-treatment procedure of

89

deparaffinization, rehydration and epitope retrieval in the Pre-Treatment Module, PT-

90

LINK (Agilent Technologies-DAKO, Santa Clara, CA, USA) at 95°C for 20 min in 50x

91

Tris/EDTA buffer, pH 9. Before staining the sections, endogenous peroxidase was

92

blocked. The antibodies used were against Cytokeratin (Ready to Use, clone AE1/AE3,),

93

E-cadherin (Ready to Use, clone NHC-38, Agilent Technologies-DAKO), SNAIL (1:100,

94

Polyclonal, ABCAM) and HMGA2 (1:300, Polyclonal, Thermo Fisher scientific). After

95

incubation, the reaction was visualized with the EnVisionTM FLEX Detection Kit

96

(Agilent Technologies-DAKO). Sections were counterstained with hematoxylin.

97

Appropriate negative controls including no primary antibody were also tested.

98

The human EC cell line Hec1A (ATCC® HTB-122™) and the derivative cell line

99

overexpressing ETV5 transcription factor (Hec1A-ETV5) were cultured in McCoy’s 5A

100

media (Gibco, Grand Island, NY, USA) supplemented with 10% (v/v) FBS and 1% (v/v)

101

PenStrep (100 U/mL penicillin - 100 µg/mL streptomycin; Gibco). Cells were maintained

102

in humidified atmosphere at 37°C and 5% CO2. ETV5 transfected cells (EX-F080000-

103

Lv105, NM_004454; GeneCopoieia, Rockville, MD, USA) were selected in 500 µg/mL

104

G-418 (Gibco). This cell line has been previously generated and thoroughly

105

characterized.14,15

106

2.2.

107

in supplemented media until 70% confluence, washed 3 times in PBS and cultured in

EV production and collection. Hec1A and Hec1A-ETV5 cell lines were grown

5 ACS Paragon Plus Environment

Journal of Proteome Research 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 6 of 34

108

FBS-free McCoy’s 5A to produce the EVs. After 48h, small EVs were harvested (cell

109

viability at collection >95%) by differential centrifugation as referred by Théry et al.16

110

Briefly, conditioned medium was initially cleared of floating (300x g, 10 min) and dead

111

cells (2,500x g, 15 min, 4°C) at low speed centrifugation and immediately cleared of cell

112

debris and large EVs (10,000x g, 30 min, 4°C, k-Factor: 2567.7) in a SW32Ti swinging

113

rotor (Beckman Coulter, Brea, CA, USA). Small vesicles were then collected at high-

114

speed ultracentrifugation (100,000x g, 90 min, 4°C, k-Factor: 256.8), washed in 0.22 µm

115

Ø-filtered PBS (100,000x g, 90 min, 4°C, k-Factor: 256.8) and resuspended in the

116

appropriate buffer. Alternatively, circulating small EVs were purified from human plasma

117

by drawing after tourniquet with a 21-gauge needle and 15 mL of peripheral blood

118

collected and immediately cleared of blood cells (1,250x g, 15 min, no brake); 12 mL of

119

buffer solution (Ca2+ and Mg2+ depleted PBS, 0.1% BSA, 2 mM EDTA) were then added

120

and plasma further cleared of cell debris (2,500x g, 15 min, 4°C), and frozen at -80°C

121

until EVs purification. When needed, plasma samples were thawed at 4°C, cleared of

122

large EVs (30 min, 4°C, k-Factor: 2567.7) and small EVs harvested at 100,000x g (90

123

min, 4°C, k-Factor: 256.8). Small EVs used for MRM development were resuspended in

124

6M urea/2M thiourea buffer. For retrospective analyses, plasma was processed as referred

125

and pelleted EVs directly resuspended in AllPrep® DNA/ RNA/ Protein Mini lysis buffer

126

(Qiagen, Hilden, Germany).

127

2.3.

128

EV batches were routinely assessed by Dynamic Light Scattering (DLS) in a ZetaSizer

129

Nano ZS bioanalyzer (Malvern, Worcestershire, UK). 1:10 diluted EVs were measured

130

(25°C, 5 runs of 10 seconds per sample, backscattering 173°, attenuator 8) in disposable

131

cuvettes. PSD and count rate (kilo counts per second, kcps) are represented as the mean

132

value of 3 replicates. Alternatively, EV size and shape were visualized by transmission

Characterization of EVs. Number and Particle Size Distribution (PSD) among

6 ACS Paragon Plus Environment

Page 7 of 34 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

133

electron microscopy (TEM, JEOL JEM-2010, 120 KV; Tokyo, Japan). To do so, fresh

134

pelleted vesicles were directly deposited onto formvar-coated grids, washed, air dried and

135

stained with phosphotungstic acid (5 min, pH 6.8) prior imaging.

136

Immunoblot analysis of EVs was performed by direct lysis (25 mM Tris-HCl, pH 7.6,

137

150 mM NaCl, 1% NP-40, 1% DOC, 0.1% SDS) of EVs in presence of protease inhibitors

138

(4 mM NaF, 2mM NaSOV4, 1x Sigma Protease Inhibitor Cocktail and 1 mM

139

phenylmethanesulfonyl fluoride (PMSF)). Protein extracts were quantified by BCA assay

140

and 10 µg of protein resolved in SDS-PAGE and probed with the indicated antibodies.

141

Antibodies used were CD9 (sc-13118; Santa Cruz, Dallas, TX, USA), CD81 (sc-7637;

142

Santa Cruz), PDCD6IP (NB-100-65678; Novus Biologicals, Littleton, CO, USA) and

143

HSP90B1 (NBP1-04346; Novus Biologicals).

144

2.4. Stable isotope labeling with amino acids in cell culture (SILAC). Hec1A and

145

Hec1A-ETV5 cells were cultured in SILAC-DMEM 4.5 g/L glucose deficient in arginine

146

(Arg) and lysine (Lys), supplemented with 2 mM L-glutamine (Ref. 280001200), 10%

147

dialyzed FBS (dFBS; Ref. 281000800) and 1% PenStrep. Labeling media was

148

supplemented with 28 mg/L L-arginine-HCl-13C6 (Arg+6; Ref. 201203902) and 73 mg/L

149

L-lysine-HCl-4,4,5,5-D4 (Lys+4; Ref. 211103913) (middle); or L-arginine-HCl-13C615N4

150

(Arg+10; Ref. 201603902) and L-lysine

151

(Silantes, Munich, Germany). Cells were subcultured until homogeneous labeling (>12

152

cell doubling accumulated); normal cellular growth under labeling conditions, and

153

complete substitution of the naturally light isotopes (labeling efficiency near 100%), were

154

assessed (data not shown). For LC-MALDI-MS analysis of the samples, EV protein

155

lysates from both cell lines were mixed in 1:1 (w/w). One replicate of forward (Hec1A

156

middle / Hec1A-ETV5 heavy) and reverse labeling (Hec1A heavy / Hec1A-ETV5

157

middle), combining technical and biological replicates due to the limited recovery of

13C 15N 6 2

(Lys+8; Ref. 211603902) (heavy)

7 ACS Paragon Plus Environment

Journal of Proteome Research 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 8 of 34

158

sample, were approached. Protein was resolved in 10% SDS-PAGE, stained with 0.1%

159

Coomassie G-250 (Thermo Scientific, Waltham, MA, USA) and protein lanes cut into 1

160

mm3 pieces. Tryptic digestion and extraction were performed as described by Shevchenko

161

et al.17 Briefly, two gel sections were de-stained (50% acetonitrile (ACN) / 50% 25 mM

162

NH4HCO3), reduced with 10 mM DTT (45 min, 56°C), alkylated with 50 mM

163

iodoacetamide (IAA, 15 min in dark) and in-gel digested with 6 ng/µL sequencing grade

164

modified trypsin (16h, 37°C; Promega, Madison, WI, USA). Peptides were then extracted

165

(50% ACN, 0.2% TFA), desalted, concentrated in a nu-tipC18 column (Glygen,

166

Columbia, MA, USA) and eluted using 70% ACN, 0.1% TFA. Finally, samples were

167

dried and resuspended in 7 µL 2% ACN, 0.1% TFA.

168

2.5. NanoLC-MALDI-TOF/TOF. Peptide fractions were resolved using RP-LC in a

169

Tempo nanoLC system (Eksigent, Dublin, CA, USA). Peptide mixture was injected on a

170

C18 precolumn (Michrom, 0.5x2 mm) coupled to a reversed-phase column (Integrafit

171

C18, Proteopep II, 75 µm id, 10.2 cm, 5 µm, 300 Å pore size; New Objective, Woburn,

172

MA, USA), eluted in a linear gradient of 5-50% ACN (0.35 µL/min, 45 min) in α-cyano

173

matrix (4 mg/mL, 1.2 µL/min), and deposited onto a MALDI plate (Sun Collect;

174

Sunchrom, Friedrichsdorf, Germany). Chromatograms corresponding to each gel section

175

were composed of 180 spots. MS runs for each chromatogram were acquired and

176

analyzed in a MALDI-TOF/TOF instrument (4800 Sciex; Framingham, MA, USA) as

177

described by Mateos et al.18 A total of 1,500 laser shots were accumulated for each TOF-

178

MS spectrum at fixed laser setting (3,600 kV). Selection of precursors was approached

179

by Job-Wide interpretation method and peptide fragmentation approached with a laser

180

intensity of 4,400 kV and 2,000 shots per spectrum.

181

Paragon™ algorithm (ProteinPilot 4.5; Sciex) was used for protein identification by

182

searching in the UniProtKB/Swiss-Prot database 2015_05 (http://www.expasy.ch/sprot;

8 ACS Paragon Plus Environment

Page 9 of 34 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

183

547,599 sequences, 195,014,757 residues, Homo sapiens). Alternatively, hits were

184

contrasted with UniProt_all database for the estimation of contaminants. Search

185

parameters included SILAC, IAA alkylation, in-gel trypsin digestion and urea

186

denaturalization as specific factors. Proteins identified with ≥2 distinct peptides with 95%

187

confidence and a ProtScore ≥1.3 were considered for relative quantification. Exosome

188

datasets were manually inspected for proteins identified with one single peptide and

189

normalized against 5%-trimmed mean. Finally, abnormal extreme quantifications are

190

discarded, and differences were considered significant when mean log2 absolute protein

191

ratio ≥ 1.18 and p value ≤0.05 according to a two-tailed t-test.

192

2.6. Patient recruitment. Subjects were recruited from June 2013 to May 2016 in Vall

193

d’Hebron University Hospital (Barcelona, Spain), University Hospital of Santiago de

194

Compostela (Santiago de Compostela, Spain), Arnau de Vilanova Hospital (Lleida,

195

Spain) and MD-Anderson Cancer Center Madrid (Madrid, Spain) (supplemental Table

196

S1). Samples were collected in EDTA Vacutainer tubes (Becton Dickinson, Franklin

197

Lakes, NJ, USA) and processed in less than 48 hours. All participants provided informed

198

consent according to the Institutional Ethical Committee (Galicia, Spain). Circulating

199

exosomes from age-matched healthy women were included as controls.

200

2.7. Multiple Reaction Monitoring of exosome biomarkers. Circulating EVs were

201

isolated from human plasma samples as previously described. Protein digestion and

202

peptide clean-up was approached as described by Fernandez-Puente et al.19 Briefly,

203

protein was reduced in 10 mM DTT, alkylated in 50 mM IAA and in-solution digested

204

with trypsin (0.5 µg, 16h, 37°C). Samples were then desalted in C18 stage tips and

205

reconstituted in 25 µL of mobile phase buffer A (0.1% formic acid / 2% ACN) for MRM

206

development analysis and in a mixture of Stable Isotope-labeled Standard (SIS) peptides

207

(7 µL) for MRM validation. SIS or heavy peptides incorporated a fully atom labeled 13C

9 ACS Paragon Plus Environment

Journal of Proteome Research 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 10 of 34

208

and 15N isotope at the C-terminal lysine, (13C6, 15N2-Lys) (K) or arginine (13C6, 15N4-Arg)

209

(R) position of each (tryptic) peptide, resulting in a mass shift of +8 or +10 Da

210

respectively. SIS peptides (crude purity) were acquired from JPT (JPT Peptide

211

Technologies GmnH, Berlin, Germany).

212

Protein digests with SIS peptides were analyzed by LC-MS/MS in a nanoLC system

213

(TEMPO) coupled to a 5500-QTRAP instrument (Sciex). A label-free EM ER analysis

214

(Enhanced Mass / Enhanced Resolution) was performed in EV samples to detect target

215

peptides. Initial transitions for each peptide included, MS/MS spectra from the EM ER

216

analysis, peptides previously identified by MALDI-TOF/TOF in shotgun experiments

217

and sequences frequently observed in the open-source Global Proteome Machine

218

Database.20 This analysis involved performing an information-dependent LC/MS/MS

219

experiment on the EV extract and searching the acquired data against protein databases

220

by Protein Pilot software 4.5 (Sciex). Several candidate peptide precursors and fragment

221

ion masses were selected per each protein and analyzed with Skyline 1.3 software

222

(MacCoss, Seattle, WA, USA).21 Results were pooled and an optimized set of MRM

223

transitions covering a total of 4 and 3 peptides for ANXA2 and LGALS3BP respectively

224

were acquired (supplemental Table S3). Mass spectrometer was interfaced with a nano-

225

spray source equipped with an uncoated fused silica emitter tip (20 μm inner diameter,

226

10 μm tip; New Objective, Woburn, MA) and operated in positive ion mode. MS source

227

parameters were set as follows: IS 2,600 V, IHT 150°C, GS2= 0, CUR= 20, GS1= 25 psi

228

and high CAD. MS compound parameters were set to 10 for the EP and to 15 for CXP.

229

Skyline was used to predict and optimize collision energies for each peptide, both analyte

230

and SIS. Q1 and Q3 were set to unit/unit resolution (0.7 Da) and pause between mass

231

ranges set to 3 ms. To confirm the identity of the peptides, a MRM Information Dependent

232

Acquisition (IDA) experiment was performed for each peptide. Mass spectrometer was

10 ACS Paragon Plus Environment

Page 11 of 34 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Proteome Research

233

instructed to switch from MRM to EPI scanning mode when an individual MRM signal

234

exceeded 1,000 counts. Each precursor was fragmented a maximum of twice before being

235

excluded for 10s and the masses were scanned from 250 to 1,000 Da. The rolling collision

236

energy (CE) option was employed to automatically ramp up the CE value in the collision

237

cell as the m/z value increased. One replicate was analyzed for each sample. SIS were

238

used for relative quantification using one-point calibration to determine basically fold

239

changes between the different cancer samples compared to control donors. The

240

concentration of the SIS stock solution was 10 fmol/µL for all the peptides except for the

241

ASHEEVEGLVEK (LGALS3BP) that was 1 pmol/µL due to the low signal of the peptide

242

compared to the other two LGALS3BP targeted peptides. A blank solvent injection was

243

run between each sample injection to avoid carry over in the nanoLC-MRM system. The

244

linearity, LOD and LOQ were not performed due to the low concentration of the exosome

245

samples matrix. The MRM method was developed for research use, which aligns this

246

work with a Tier 2 Targeted Measurement as described.22

247

Data analysis for the target proteins was performed using Skyline for method refinement,

248

optimization and peak integration. Raw files were imported to Skyline and the

249

chromatograms were manually inspected to ensure correct peak detection and accurate

250

integration. Co-elution of all the transitions of the target or light peptide with the SIS

251

peptide was considered accurate MRM signal. Peak area ratios between light and heavy

252

peptide was used to calculate the relative peak area ratio of each peptide in the conditions

253

of the study. Reports of peak area ratios between the light and heavy peptides of each

254

peptide in the serum samples were exported by Skyline to MS Excel. The % CV were

255

calculated for the RT of the targeted proteins. The peptide RT observed for LGALS3BP

256

and ANXA2 was very reproducible with a %CV