• No results found

Gene expression profiling of bronchial brushes is associated with the level of emphysema measured by computed tomography-based parametric response mapping

N/A
N/A
Protected

Academic year: 2021

Share "Gene expression profiling of bronchial brushes is associated with the level of emphysema measured by computed tomography-based parametric response mapping"

Copied!
22
0
0

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

Hele tekst

(1)

University of Groningen

Gene expression profiling of bronchial brushes is associated with the level of emphysema

measured by computed tomography-based parametric response mapping

Rathnayake, Senani N H; Hoesein, Firdaus A A Mohamed; Galban, Craig J; Ten Hacken,

Nick H T; Oliver, Brian G G; van den Berge, Maarten; Faiz, Alen

Published in:

American Journal of Physiology - Lung Cellular and Molecular Physiology DOI:

10.1152/ajplung.00051.2020

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below.

Document Version

Final author's version (accepted by publisher, after peer review)

Publication date: 2020

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Rathnayake, S. N. H., Hoesein, F. A. A. M., Galban, C. J., Ten Hacken, N. H. T., Oliver, B. G. G., van den Berge, M., & Faiz, A. (2020). Gene expression profiling of bronchial brushes is associated with the level of emphysema measured by computed tomography-based parametric response mapping. American Journal of Physiology - Lung Cellular and Molecular Physiology, 318(6), L1222-L1228.

https://doi.org/10.1152/ajplung.00051.2020

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

Gene expression profiling of bronchial brushes is associated with the level of 1

emphysema measured by computed tomography-based parametric response mapping 2

Senani N. H. Rathnayake1,6*, Firdaus A. A. Mohamed Hoesein2, Craig J. Galban3,

3

Nick H. T. ten Hacken4,5, Brian G. G. Oliver6,7, Maarten Van den Berge4,5#, and Alen

4

Faiz1,4,5,6#

5

1. University of Technology Sydney, Respiratory Bioinformatics and Molecular Biology

6

(RBMB), School of Life Sciences, Sydney, Australia

7

2. Utrecht University, Division of Heart and Lungs, Department of Respiratory

8

Medicine, University Medical center, Utrecht, The Netherlands

9

3. The University of Michigan, Department of Radiology, Ann Arbor, Michigan, USA

10

4. The University of Groningen, Groningen Research Institute for Asthma and

11

COPD(GRIAC), University Medical center Groningen, Groningen, The Netherlands.

12

5. The University of Groningen, Department of Pulmonary Diseases, University Medical

13

center Groningen, Groningen, The Netherlands

14

6. The University of Sydney, Respiratory Cellular and Molecular Biology, Woolcock

15

Institute of Medical Research, Sydney, Australia

16

7. The University of Technology Sydney, School of Life Sciences, Sydney, Australia

17

*= Corresponding author 18

#= These authors contributed equally to this manuscript 19

Correspondence address: 20

Senani N.H. Rathnayake

21

Respiratory Bioinformatics and Molecular Biology (RBMB), School of Life Sciences,

22

University of Technology Sydney, Thomas St, Ultimo NSW 2007

23 Email: senani.rathnayakemudiyanselage@uts.edu.au 24 25 26 27 28

(3)

Abstract 29

Parametric response mapping (PRM) is a computed tomography (CT) based method to

30

phenotype COPD patients. It is capable of differentiating emphysema related air trapping

31

with non-emphysematous air trapping (small airway disease), which helps to identify the

32

extent and localization of the disease. Most studies evaluating the gene expression in smokers

33

and COPD patients related this to spirometric measurements, but none have investigated the

34

relationship with CT-based measurements of lung structure. The current study aimed to

35

examine gene expression profiles of brushed bronchial epithelial cells in association with the

36

PRM-defined CT based measurements of emphysema (PRMEmph) and small airway disease

37

(PRMfSAD). Using the TIP study cohort (COPD = 12 and asymptomatic smokers = 32), we 38

identified a gene expression signature of bronchial brushings, which was associated with

39

PRMEmph in the lungs. One hundred thirty-three genes were identified to be associated with

40

PRMEmph. Among the most significantly associated genes, CXCL11 is a potent chemokine

41

involved with CD8+ T cell activation during inflammation in COPD, indicating that it may

42

play an essential role in the development of emphysema. The PRMEmph signature was then 43

replicated in two independent datasets. Pathway analysis showed that the PRMEmph signature

44

is associated with proinflammatory and notch signaling pathways. Together these findings

45

indicate that airway epithelium may play a role in the development of emphysema and/or

46

may act as a biomarker for the presence of emphysema. In contrast, its role in relation to

47

functional small airways disease is less clear.

48 49 50 51 52 53 54 55 56 57

(4)

Introduction

58

Chronic Obstructive Pulmonary Disease (COPD) is considered as one of the major

non-59

communicable diseases in the world (20). The persistent airflow limitation is associated with

60

inflammatory responses, which are initially to noxious particles (22). These factors together

61

result in an accelerated decline in lung function (16). COPD is a heterogeneous disease in

62

which fibrosis and loss of small airways and emphysema are two major pathological

63

characteristics of the disease (17).

64

Current theories behind the development of the emphysematous phenotype of COPD include

65

protease antiprotease imbalance, chronic airway inflammation, and dysregulation of oxidative

66

stress (9, 35). These mechanisms are thought to cause the characteristic symptoms of

67

emphysema, including abnormal inflammatory responses together with alveolar destruction,

68

which leads to a reduction of the alveolar-capillary exchange area (29).

69

Parametric response mapping (PRM) is a novel computed tomography (CT) based method to

70

phenotype lung diseases (23). Application of PRM to paired inhaled/exhaled CT scans is

71

capable of differentiating emphysema from non-emphysematous air trapping due to

72

functional small airway disease (14, 23, 24).

73

Gene expression signatures have been studied in different diseases to identify the underlying

74

mechanisms and biological pathways associated with the disease of interest (25, 28). These

75

gene expression profiles of bronchial brushes provide a global picture of the airways, and

76

they can help understand the mechanisms involved in the development of emphysema (18,

77

29).

78

Several studies have assessed gene expression in smokers and COPD patients and related this

79

to spirometric measurements (18, 29-31). However, none have investigated the relationship

80

with CT-based measurements of lung structure. In the present study, gene expression profiles

81

of bronchial epithelial cells were investigated in association with the severity of PRM-defined

82

emphysema (PRMEmph) and functional small airway disease (PRMfSAD).

83 84 85 86

(5)

Methods

87

Study population 88

The study population was a subset of subjects included in the Top Institute Pharma (TIP)

89

study (3) who underwent bronchoscopy. The TIP study was approved by the ethics

90

committee of UMCG and registered under the National Clinical Trial (NCT) identifier:

91

NCT00850863. All these selected subjects were >35 years of age and current or ex-smokers

92

consist of 12 COPD subjects and 32 asymptomatic smokers who had provided written

93

informed consent. The spirometric measurements were collected according to the

94

international guidelines described in (21) and (37). The clinical characteristics of the current

95

study population are described in table 1.

96

Bronchial brushes sample collection and processing 97

Bronchoscopically derived bronchial brushings were collected from the first, and second

98

subsegmental branches of the left lower lobe and total RNA was extracted with the

99

miRNeasy Mini Kit (Qiagen, Valencia, CA). From each sample, 100–200 ng total RNA was

100

processed and examined with Affymetrix Gene Chip Human Gene 1.0 ST, as previously

101

described (GSE97010) (3).

102

CT images acquisition for PRM 103

The inspiratory and expiratory low dose chest CT scans were taken using multi-detector CT

104

scanners at full inspiration, and normal expiration. Then the CT image processing was done

105

using PRM. Detail protocols used for CT scan acquisition and PRM processing were

106

previously described in (15). PRM scores are presented as the percent volume of the total

107

lung. PRM processing for inhaled and exhaled CT images of a single patient is illustrated in

108

figure 1A.

109

Bioinformatic Analysis 110

Microarray data analyses were done using the Bioconductor-limma package in R software

111

version 3.5.1. Gene expression of the bronchial brushings were correlated to different CT

112

scan variables (PRMEmph and PRMfSAD scores) and Forced Expiratory Volume in one second

113

(FEV1) %predicted using the R package Limma (V3.38.3). Linear models were applied after

114

corrected for gender and packyears. The False Discovery Rate (FDR) less than 0.05

115

considered as statistical significance.

(6)

117

Gene Set Enrichment Analysis (GSEA) 118

GSEA gives the quantification of the association of gene sets with the differential expression

119

changes. In this study, GSEA was done using GSEA V.2.0.14 to compare the PRMEmph 120

signature to the difference in bronchial brush gene expression between COPD and non-COPD

121

individuals, using two previously published publicly available independent datasets. These

122

datasets are accessible through following GEO Series accession numbers in the National

123

Center for Biotechnology Information Gene Expression Omnibus (NCBI GEO). (cohort one

124

and cohort two as described below). Cohort one consists of current and ex-smokers with and

125

without COPD (COPD=87, non-COPD =151) (GSE37147) (31). The cohort 2 composed of

126

COPD and non-COPD subjects (COPD= 8, non-COPD =14) (GSE56342) (36). 127

Gene Set Variation Analysis (GSVA) 128

GSVA analysis allows us to explore the effect of genes associated with PRMEmph signature on

129

each patient. The GSVA analysis was done using GSVA (1.34.0) package in R software

130

version 3.5.1, by looking at the genes that were positively and negatively associated with

131

PRMEmph signature separately.

132

Pathway analysis 133

Pathway analysis was done to identify the pathways related to significant genes associated

134

with PRMEmph score. This analysis was done using the g: Profiler web base tool (26).

135 136 137

(7)

Results

138

Association of bronchial brush gene expression with PRM scores and FEV1 %predicted 139

Initially, we investigated the gene expression profiles of bronchial epithelial cells in relation

140

to PRMEmph, PRMfSAD, and the FEV1 % predicted. A total number of 133 genes were 141

associated with PRMEmph scores, with 82 genes (61.65%) positively associated and 51

142

(38.35%) genes negatively associated (FDR < 0.05). In contrast, no genes were significantly

143

associated with PRMfSAD, and FEV1% predicted. The top 20 genes associated with PRMEmph

144

were tabulated in table 2. A volcano plot present in figure 1B represents the differentially

145

expressed genes in bronchial brushings related to emphysema (PRMEmph score), and the 146

heatmap in figure 1C represents the significant genes associated with PRMEmph score,

147

respectively.

148

Association of identified PRMEmph signature with other clinical parameters and 149

independent datasets 150

We next compared the overlap between the identified signatures using GSEA and GSVA

151

analysis. The GSEA results for genes significantly associated with FEV1 % predicted, and

152

PRMfSAD are illustrated in Figure 1D & E. These results show a high overlap between genes 153

associated with PRMEmph score, FEV1% predicted and PRMfSAD. This is reflected with the

154

high correlation between FEV1% predicted with PRMEmph scores (r = -0.508, p-value =

155

0.000507, n= 44), and PRMEmph with PRMfSAD scores (r = 0.852, p-value = 2.2e-16, n= 44).

156

We then compared the gene expression signature of PRMEmph with an independent dataset of

157

COPD status signature (GSE37147). Those genes positively associated with PRMEmph scores

158

were enriched among genes expressed in bronchial brushings of the COPD cohort (Figure

159

1F). The PRMEmph signature was then compared with another independent dataset, consisting

160

of gene expression profiles of COPD status in small airway epithelium (GSE56342). The

161

resulted GSEA plot in Figure 1G shows a similar pattern as in our dataset, further confirming

162

the identified gene expression signature of PRMEmph replicated in different independent

163

cohorts. The GSVA results further confirm that there is a continuous relationship in the

164

change of gene expression patterns associated with PRMEmph scores (figure 2A and B). 165

Pathways associated with PRMEmph signature 166

Pathway analysis shows that the PRMEmph score is associated with cytokine-mediated 167

signalling pathways, interferon pathways and NOTCH signalling pathways. Both

(8)

mediated signalling and interferon signalling pathways got increased. In contrast,

169

extracellular metric, collagen and NOTCH signalling related pathways got decreased

170

associated with PRMEmph signature (FDR<0.05) (table 3).

171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186 187 188 189 190 191 192 193

(9)

Discussion

194

The current study examines gene expression profiles of bronchial brushings in association

195

with PRM-defined CT measurements of emphysema and small airway disease. The CXCL11

196

gene which produced by the airway epithelium (13), and it is known for its role as a

197

prominent chemokine in CD8+ T cell activation during inflammation in COPD was found as

198

one of the most significantly associated genes with PRMEmph scores, indicating that CXCL11 199

may play an essential role in the development of emphysema. The identified PRMEmph

200

signature was then replicated in two independent datasets, providing evidence that the airway

201

epithelium may play a role in the development of emphysema and/or may act as a biomarker

202

for the presence of emphysema.

203

The top five genes differentially expressed in bronchial brushes related to PRMEmph scores

204

include SLCO1B3, SPRR1A, FKBP5, CXCL11, and CLEC4E. CXCL11 is a T-cell

205

chemoattractant and one of the most effective ligands of CXCR3 on CD8+ T cell and CD4+ T 206

cells (5). CD8+ T cell activation has previously been associated with the development of 207

emphysema by inducing alveolar cell apoptosis (2) via producing perforins and granzyme B

208

(6, 12). In addition, the CXCL11 gene was previously identified as a highly expressed gene in

209

the sputum of COPD patients (8). FKBP5 is a negative regulator of the glucocorticoid

210

receptor and therefore regulates corticosteroid anti-inflammatory functions (11, 27). This

211

gene has previously been found as corticosteroid sensitive gene, and its upregulation with

212

PRMEmph may be due to a higher dose of corticosteroid use in patients with a high level of

213

emphysema; thus, it could be more of a treatment effect rather than disease effect (27). The

214

SLCO1B3 gene, which encodes a transmembrane receptor that mediates the

sodium-215

independent uptake of endogenous and xenobiotic compounds, mainly in the liver (32), while

216

the CLEC4E gene encodes a protein which belongs to C-type lectin domain family 4 (7), but

217

for these two genes roles related to COPD, is yet to be explained.

218

The GSEA results, which show the association of PRMEmph gene expression signature with 219

FEV1% predicted and PRMfSAD on the gene set level, show a similar overlapping pattern with 220

the PRMEmph signature, indicating possible similar mechanisms associated with these

221

measurements of the lung (18, 29, 31).

222

The PRMEmph associated signature was shown to be associated with COPD in two 223

independent datasets from the upper and lower airways. This result follows the theory of

(10)

“united airway field of injury,” providing evidence that this signature may common

225

throughout the compartments of the lung (4, 31).

226

The pathway analysis revealed top pathways associated with PRMEmph score include

227

cytokine-mediated signalling pathways and NOTCH signalling pathways which are well

228

known for their role in COPD (2). Cytokine-mediated signalling pathways are responsible for

229

the increased inflammation in COPD. In contrast, NOTCH signalling pathway plays a

230

significant role in lung epithelial morphogenesis, and it is found to be downregulated in

231

COPD patients and cause the lung epithelial metaplasia which leads to mucosal hyperplasia

232

(1, 2, 10, 19, 33, 34).

233

The limitation of this study is the small number of patients tested in the discovery cohort,

234

however despite these low number of patients the identified signature was able to be observed

235

in two independent datasets of bronchial brushes from COPD, indicating the robustness of the

236

PRMEmph signature. The lack of significance in PRMfSAD may be due to its variability within 237

the GOLD status of COPD and possible multifactorial causes for the development of small

238

airways disease. In addition, the bronchial brushes were collected from the 1st and second

239

subsegmental branches of the left lower lobe of the lung which may not accurately reflect the

240

transcriptomic changes occurring in the peripheral small airways, which are inaccessible to

241

bronchoscopy. Furthermore, our replication study was conducted on COPD status and not

242

PRM, as this data is currently not available for airway gene expression datasets.

243

In conclusion, we have identified a gene expression signature of bronchial brushings, which

244

is associated with PRMEmph signature in the lungs. In contrast, we did not find gene 245

expression levels to be significantly associated with PRMfSAD. These findings indicate that 246

airway epithelium may play a role in the development of emphysema and/or may act as a

247

biomarker for the presence of emphysema, but not or to a lesser extent for functional small

248

airways disease.

(11)

References

250

1. Barnes PJ. The cytokine network in chronic obstructive pulmonary disease. Am J 251

Respir Cell Mol Biol 41: 631-638, 2009.

252

2. Barnes PJ. Inflammatory mechanisms in patients with chronic obstructive pulmonary 253

disease. J Allergy Clin Immunol 138: 16-27, 2016.

254

3. Billatos E, Faiz A, Gesthalter Y, LeClerc A, Alekseyev YO, Xiao X, Liu G, Ten 255

Hacken NHT, Heijink IH, Timens W, Brandsma CA, Postma DS, van den Berge M, 256

Spira A, and Lenburg ME. Impact of acute exposure to cigarette smoke on airway gene 257

expression. Physiol Genomics 50: 705-713, 2018.

258

4. Boudewijn IM, Faiz A, Steiling K, Van Der Wiel E, Telenga ED, Hoonhorst 259

SJM, Ten Hacken NHT, Brandsma C-A, Kerstjens HAM, Timens W, Heijink IH, 260

Jonker MR, De Bruin HG, Sebastiaan Vroegop J, Pasma HR, Boersma WG, Wielders 261

P, Van Den Elshout F, Mansour K, Spira A, Lenburg ME, Guryev V, Postma DS, and 262

Van Den Berge M. Nasal gene expression differentiates COPD from controls and overlaps 263

bronchial gene expression. Respiratory Research 18: 2017.

264

5. Chen Hong WD, Wang Xiangdong Role of Airway Epithelium-Origin Chemokines 265

and their Receptors in COPD. Journal of Epithelial Biology & Pharmacology 3: 26-33, 2010.

266

6. Chrysofakis G, Tzanakis N, Kyriakoy D, Tsoumakidou M, Tsiligianni I, 267

Klimathianaki M, and Siafakas NM. Perforin Expression and Cytotoxic Activity of 268

Sputum CD8+ Lymphocytes in Patients With COPD. 125: 71-76, 2004.

269

7. Clement M, Basatemur G, Masters L, Baker L, Bruneval P, Iwawaki T, 270

Kneilling M, Yamasaki S, Goodall J, and Mallat Z. Necrotic Cell Sensor Clec4e Promotes 271

a Proatherogenic Macrophage Phenotype Through Activation of the Unfolded Protein

272

Response. Circulation 134: 1039-1051, 2016.

273

8. Costa C, Rufino R, Traves SL, Lapa ESJR, Barnes PJ, and Donnelly LE. CXCR3 274

and CCR5 chemokines in induced sputum from patients with COPD. Chest 133: 26-33, 2008.

275

9. Demedts IK, Demoor T, Bracke KR, Joos GF, and Brusselle GG. Role of 276

apoptosis in the pathogenesis of COPD and pulmonary emphysema. Respir Res 7: 53, 2006.

277

10. EMPHZong D, Ouyang R, Li J, Chen Y, and Chen P. Notch signaling in lung 278

diseases: focus on Notch1 and Notch3. Ther Adv Respir Dis 10: 468-484, 2016.

279

11. Faiz A, Postma DS, Koppelman GH, Hiemstra PS, Sterk PJ, Timens W, Steiling 280

K, Spira A, Heijink IH, and van den Berge M. FKBP5 a candidate for corticosteroid 281

insensitivity in COPD. European Respiratory Journal 48: OA1779, 2016.

(12)

12. Fenwick PS, Macedo P, Kilty IC, Barnes PJ, and Donnelly LE. Effect of JAK 283

Inhibitors on Release of CXCL9, CXCL10 and CXCL11 from Human Airway Epithelial

284

Cells. PLoS One 10: e0128757, 2015.

285

13. Fenwick PS, Macedo P, Kilty IC, Barnes PJ, and Donnelly LEJPO. Effect of JAK 286

inhibitors on release of CXCL9, CXCL10 and CXCL11 from human airway epithelial cells.

287

10: e0128757, 2015.

288

14. Galban CJ, Han MK, Boes JL, Chughtai KA, Meyer CR, Johnson TD, Galban S, 289

Rehemtulla A, Kazerooni EA, Martinez FJ, and Ross BD. Computed tomography-based 290

biomarker provides unique signature for diagnosis of COPD phenotypes and disease

291

progression. Nat Med 18: 1711-1715, 2012.

292

15. Hoff BA, Pompe E, Galban S, Postma DS, Lammers JJ, Ten Hacken NHT, 293

Koenderman L, Johnson TD, Verleden SE, de Jong PA, Mohamed Hoesein FAA, van 294

den Berge M, Ross BD, and Galban CJ. CT-Based Local Distribution Metric Improves 295

Characterization of COPD. Sci Rep 7: 2999, 2017.

296

16. Huertas A, and Palange P. COPD: a multifactorial systemic disease. 5: 217-224, 297

2011.

298

17. Izquierdo-Alonso JL, Rodriguez-Gonzalezmoro JM, de Lucas-Ramos P, Unzueta 299

I, Ribera X, Anton E, and Martin A. Prevalence and characteristics of three clinical 300

phenotypes of chronic obstructive pulmonary disease (COPD). Respir Med 107: 724-731,

301

2013.

302

18. Jeong I, Lim JH, Oh DK, Kim WJ, and Oh YM. Gene expression profile of human 303

lung in a relatively early stage of COPD with emphysema. Int J Chron Obstruct Pulmon Dis

304

13: 2643-2655, 2018.

305

19. Kiyokawa H, and Morimoto M. Notch signaling in the mammalian respiratory 306

system, specifically the trachea and lungs, in development, homeostasis, regeneration, and

307

disease. Dev Growth Differ 2019.

308

20. Mannino DM, and Buist AS. Global burden of COPD: risk factors, prevalence, and 309

future trends. The Lancet 370: 765-773, 2007.

310

21. Miller MR, Hankinson J, Brusasco V, Burgos F, Casaburi R, Coates A, Crapo R, 311

Enright Pv, Van Der Grinten C, and Gustafsson P. Standardisation of spirometry. 312

European respiratory journal 26: 319-338, 2005.

313

22. Pauwels RA, and Rabe KF. Burden and clinical features of chronic obstructive 314

pulmonary disease (COPD). The Lancet 364: 613-620, 2004.

(13)

23. Pompe E, Galban CJ, Ross BD, Koenderman L, Ten Hacken NH, Postma DS, 316

van den Berge M, de Jong PA, Lammers JJ, and Mohamed Hoesein FA. Parametric 317

response mapping on chest computed tomography associates with clinical and functional

318

parameters in chronic obstructive pulmonary disease. Respir Med 123: 48-55, 2017.

319

24. Pompe E, van Rikxoort EM, Schmidt M, Rühaak J, Estrella LG, Vliegenthart R, 320

Oudkerk M, de Koning HJ, van Ginneken B, and de Jong PA. Parametric response 321

mapping adds value to current computed tomography biomarkers in diagnosing chronic

322

obstructive pulmonary disease. American Journal of Respiratory and Critical Care Medicine

323

191: 1084-1086, 2015.

324

25. Rathnayake SNH, Van den Berge M, and Faiz A. Genetic profiling for disease 325

stratification in chronic obstructive pulmonary disease and asthma. Curr Opin Pulm Med 25:

326

317-322, 2019.

327

26. Raudvere U, Kolberg L, Kuzmin I, Arak T, Adler P, Peterson H, and Vilo J. 328

g:Profiler: a web server for functional enrichment analysis and conversions of gene lists

329

(2019 update). Nucleic Acids Res 47: W191-W198, 2019.

330

27. Russo P, Tomino C, Santoro A, Prinzi G, Proietti S, Kisialiou A, Cardaci V, Fini 331

M, Magnani M, Collacchi F, Provinciali M, Giacconi R, Bonassi S, and Malavolta M. 332

FKBP5 rs4713916: A Potential Genetic Predictor of Interindividual Different Response to

333

Inhaled Corticosteroids in Patients with Chronic Obstructive Pulmonary Disease in a

Real-334

Life Setting. International Journal of Molecular Sciences 20: 2024, 2019.

335

28. Schadt EE, Lamb J, Yang X, Zhu J, Edwards S, Guhathakurta D, Sieberts SK, 336

Monks S, Reitman M, Zhang C, Lum PY, Leonardson A, Thieringer R, Metzger JM, 337

Yang L, Castle J, Zhu H, Kash SF, Drake TA, Sachs A, and Lusis AJ. An integrative 338

genomics approach to infer causal associations between gene expression and disease. Nat

339

Genet 37: 710-717, 2005.

340

29. Spira A, Beane J, Pinto-Plata V, Kadar A, Liu G, Shah V, Celli B, and Brody JS. 341

Gene Expression Profiling of Human Lung Tissue from Smokers with Severe Emphysema.

342

31: 601-610, 2004.

343

30. Steiling K, Van Den Berge M, Hijazi K, Florido R, Campbell J, Liu G, Xiao J, 344

Zhang X, Duclos G, Drizik E, Si H, Perdomo C, Dumont C, Coxson HO, Alekseyev YO, 345

Sin D, Pare P, Hogg JC, McWilliams A, Hiemstra PS, Sterk PJ, Timens W, Chang JT, 346

Sebastiani P, O’Connor GT, Bild AH, Postma DS, Lam S, Spira A, and Lenburg ME. A 347

Dynamic Bronchial Airway Gene Expression Signature of Chronic Obstructive Pulmonary

(14)

Disease and Lung Function Impairment. American Journal of Respiratory and Critical Care

349

Medicine 187: 933-942, 2013.

350

31. Steiling K, Van Den Berge M, Hijazi K, Florido R, Campbell J, Liu G, Xiao J, 351

Zhang X, Duclos G, Drizik E, Si H, Perdomo C, Dumont C, Coxson HO, Alekseyev YO, 352

Sin D, Pare P, Hogg JC, McWilliams A, Hiemstra PS, Sterk PJ, Timens W, Chang JT, 353

Sebastiani P, O’Connor GT, Bild AH, Postma DS, Lam S, Spira A, and Lenburg ME. A 354

Dynamic Bronchial Airway Gene Expression Signature of Chronic Obstructive Pulmonary

355

Disease and Lung Function Impairment. 187: 933-942, 2013.

356

32. Tague LK, Byers DE, Hachem R, Kreisel D, Krupnick AS, Kulkarni HS, Chen

357

C, Huang HJ, and Gelman A. Impact of SLCO1B3 polymorphisms on clinical outcomes in 358

lung allograft recipients receiving mycophenolic acid. Pharmacogenomics J

10.1038/s41397-359

41019-40086-41390, 2019.

360

33. Tilley AE, Harvey BG, Heguy A, Hackett NR, Wang R, O'Connor TP, and 361

Crystal RG. Down-regulation of the notch pathway in human airway epithelium in 362

association with smoking and chronic obstructive pulmonary disease. Am J Respir Crit Care

363

Med 179: 457-466, 2009.

364

34. Tsao PN, Matsuoka C, Wei SC, Sato A, Sato S, Hasegawa K, Chen HK, Ling TY, 365

Mori M, Cardoso WV, and Morimoto M. Epithelial Notch signaling regulates lung 366

alveolar morphogenesis and airway epithelial integrity. Proc Natl Acad Sci U S A 113:

8242-367

8247, 2016.

368

35. Tuder RM, Yoshida T, Arap W, Pasqualini R, and Petrache I. State of the art. 369

Cellular and molecular mechanisms of alveolar destruction in emphysema: an evolutionary

370

perspective. Proc Am Thorac Soc 3: 503-510, 2006.

371

36. Vucic EA, Chari R, Thu KL, Wilson IM, Cotton AM, Kennett JY, Zhang M, 372

Lonergan KM, Steiling K, Brown CJ, McWilliams A, Ohtani K, Lenburg ME, Sin DD, 373

Spira A, Macaulay CE, Lam S, and Lam WL. DNA methylation is globally disrupted and 374

associated with expression changes in chronic obstructive pulmonary disease small airways.

375

Am J Respir Cell Mol Biol 50: 912-922, 2014.

376

37. Wanger J, Clausen J, Coates A, Pedersen O, Brusasco V, Burgos F, Casaburi R, 377

Crapo R, Enright P, and Van Der Grinten C. Standardisation of the measurement of lung 378

volumes. European respiratory journal 26: 511-522, 2005.

379 380 381

(15)

382 383

(16)

Figure Legends

384

Figure 1: Gene expression and GSEA results of bronchial brushings associated with 385

emphysema score. A) Parametric response mapping of one patient CT scans. Lung tissue 386

Inspiration and expiration CT scans, small airway disease in yellow (PRMfSAD), and

387

emphysematous lung tissue in red (PRMEmph). B) Volcano plot of differential gene expression 388

in bronchial brushings related to emphysema (PRMEmph) score. C) Heatmap shows genes

389

significantly altered associated with the PRMEmph score. The red and blue colours in the heat

390

map representing up and down-regulated gene-expression levels, respectively. Samples with

391

COPD are clustered under red, and non-COPD are under green. Samples grouped related to

392

PRMEmph score range from high to low represented in black to light grey colour gradient, 393

respectively. FEV1 % predicted value less than 50 represented in yellow and FEV1 %

394

predicted value range from 50 to 80 and 80 to133 were grouped under light blue and Purple,

395

respectively. Gene set enrichment analysis (GSEA) of genes significantly associated with

396

PRMEmph score related to D) FEV1% predicted E) PRMfSAD score associated genes in this

397

study, and related to COPD status in F) replicate data set 1(GSE37147) and G) replicate data

398

set 2 (GSE56342). In each GSEA plot, the colored bars represent the ranked t-values of the

399

association of bronchial gene expression. The red colour represents a positive association,

400

whereas blue represents a negative association with the signature. The black vertical lines

401

each represent a significantly differentially expressed gene.

402

Abbreviations: logFC -Log2 fold change, n_Emph- normalized emphysema score.

403

FEV1_P_predicted- Forced Expiratory Volume in one-second Percentage predicted,

404

PRMEmph- Parametric Response Mapping derived scores of emphysema, PRM fSAD-

405

Parametric Response Mapping derived scores of small airway disease.

406 407

Figure 2: GSVA results of the top 10genes associated with PRM Emph scores. A) genes 408

negatively associated with PRM Emph scores B) genes positively associated with PRMEmph

409

score. The samples colored with red and black in the plot represent 32 asymptomatic “party”

410

smokers and 12 COPD patients, respectively.

411

Abbreviations: r=Spearman correlation value

(17)

Genes downregulated with PRMEmph score

(p<0.05)

Genes upregulated with PRMEmph score

(p<0.05)

F

G

Genes upregulated with PRMEmph score

(p<0.05)

Genes downregulated with PRMEmph score

(p<0.05)

D

Genes upregulated with PRMEmph score

(p<0.05) Gene Expression in bronchial brushings Genes decreased by FEV1% Genes increased by FEV1%

Genes downregulated with PRMEmph score

(p<0.05)

Genes upregulated with PRMEmph score

(p<0.05) Gene Expression in bronchial brushings Genes decreased in PRMfSAD Genes increased in PRMfSAD

Genes downregulated with PRMEmph score

(p<0.05)

E

A

(18)

0 10 20 30 40 -1.0 -0.5 0.0 0.5 1.0 Emphysema score Negati ve enri chment score

Asymptomatic “party” smokers COPD r = -0.3129 P. value = 0.0386 0 10 20 30 40 -1.0 -0.5 0.0 0.5 1.0 Emphysema score P o s it ive e n ri ch m e nt s c o re

Asymptomatic “party” smokers COPD

r = 0.3738 P. value = 0.0124

(19)

Table 1. Clinical characteristics of the current study population

Character Asymptomatic smokers COPD

n 32 12

Male subjects no. (%) 28(87.5) 12(100)

Current smoking, no. (%) 30(93.8) 10(83.3)

Age, mean (SD) 51.28(11) 65.42(7)

PRMEmph score, mean (SD) 1.23(1.25) 13.58(9.95)

FEV1% predicted, mean (SD) 107.94(12.29) 55.29(12.43)

PRMfSAD score, mean (SD) 10.62(10.97) 32.56(6.97)

Abbreviations: SD= standard deviation, PRMEmph- Parametric Response Mapping derived

scores of emphysema, FEV1%predicted= Forced Expiratory Volume in one-second percentage predicted, PRM fSAD- Parametric Response Mapping derived scores of small airway disease

(20)

Table 2. Statistical results of top significant genes found in bronchial brushings of party smokers and COPD patients associated with emphysema scores

Gene name Log FC t P.Value adj.P.Val

SLCO1B3 0.127806024 6.519990726 5.67E-08 5.89E-04

SPRR1A 0.063278543 6.413278613 8.15E-08 5.89E-04

FKBP5 0.06591885 6.3851007 8.96E-08 5.89E-04 CXCL11 0.066597346 6.16666158 1.88E-07 9.28E-04 CLEC4E 0.0693752 5.756387466 7.56E-07 0.002497 CLU -0.03213479 -5.656230211 1.06E-06 0.002497 SNTG2 0.031901105 5.655277552 1.06E-06 0.002497 CDH2 -0.043848573 -5.644123872 1.11E-06 0.002497 DQX1 0.031684446 5.615244924 1.22E-06 0.002497 C12orf36 0.052959543 5.603764761 1.27E-06 0.002497 MYO3A 0.037012707 5.409883752 2.43E-06 0.004359 ANKRD22 0.047548353 5.334362512 3.13E-06 0.004704 THSD4 -0.046401359 -5.333727566 3.14E-06 0.004704 DKK1 0.050502428 5.315200317 3.34E-06 0.004704 SLC22A10 0.021988434 5.215854455 4.65E-06 0.006119 GUCY1B3 0.025796036 5.192145133 5.04E-06 0.006208 CEP55 0.020556219 5.095856166 6.94E-06 0.007431 GATM -0.051271784 -5.090310071 7.07E-06 0.007431 EFEMP2 -0.02109123 -5.082125381 7.26E-06 0.007431 CES1 -0.046758077 -5.06965904 7.57E-06 0.007431

(21)

Table 3. Top pathways linked with genes significantly associated with PRMEmph signature in bronchial brushings of party smokers and COPD patients

Name of the pathway Term_id Adj.P. Val

• Positively associated pathways

Cytokine-mediated signalling pathway GO:0019221 1.55E-07 Cellular response to cytokine stimulus GO:0071345 1.53829E-06

Response to cytokine GO:0034097 7.14025E-06

Defence response to virus GO:0051607 0.000202424

Response to virus GO:0009615 0.000341767

Defence response GO:0006952 0.001182

Immune response GO:0006955 0.002424

Immune system process GO:0002376 0.005083

Cellular response to type I interferon GO:0071357 0.007262 Type I interferon signalling pathway GO:0060337 0.007262

Response to type I interferon GO:0034340 0.00924

Negative regulation of multi-organism process GO:0043901 0.012274 Defence response to another organism GO:0098542 0.017913

Cornification GO:0070268 0.018896

Bile acid and bile salt transport GO:0015721 0.023652

Bile acid and bile salt transport GO:0015721 0.023652

Response to other organism GO:0051707 0.034265

Response to external biotic stimulus GO:0043207 0.034903

Immune effector process GO:0002252 0.040605

Response to biotic stimulus GO:0009607 0.043063

• Negatively associated pathways

Extracellular matrix GO:0031012 0.008048

Collagen-containing extracellular matrix GO:0062023 0.011341 Constitutive Signalling by NOTCH1 t(7;9)

(NOTCH1:M1580_K2555) Translocation Mutant

REAC:R-HSA-2660826 0.039409

Signalling by NOTCH1

t(7;9)(NOTCH1:M1580_K2555) Translocation Mutant

REAC:R-HSA-2660825 0.039409

(22)

Referenties

GERELATEERDE DOCUMENTEN

To understand the residential real estate development sector, the development process and the relation between the several markets of the real estate sector will be explained,

0% 10% 20% 30% 40% 50% 60% 70% 80% korfbal Handbal Zeilen/surfen Atletiek Floorbal Honkbal Boksen Karate/ kungfu Turnen/Gym Paardensport Volleybal stoei/ vechtspelen tafeltennis

Er was geen significant interactie-effect tussen type tweetaligheid en de covariaat op de synthesescore wanneer er werd gecontroleerd voor balans (p = 0.080) (zie Figuur 25),

Silva J, Mak W, Zvetkova I, Appanah R, Nesterova TB, Webster Z, Peters AHFM, Jenuwein T, Otte AP, Brockdorff N: Establishment of histone H3 methylation on the inactive X

Therefore this study focussed on how the type of language used in a message and the valence of the reactions of other community members can moderate the effect of the tie

repetitive floods in two coastal towns with different shares of houses in flood zones to explore a bottom-up response to increasing flood probability and severity of floods with

The increased use of computers and the Internet by children has focused attention on software for younger computer users and studying their search behaviour and interaction

ANNs 52 are mathematical models, inspired by bio- logical neural networks, which can be used in all three machine learning paradigms (i.e. supervised learning 53 , unsupervised