Accepted Manuscript Neutrophils are a major source of the epithelial barrier disrupting cytokine Oncostatin M in mucosal airways disease Kathryn L. Pothoven, PhD, James E. Norton, MSc, Lydia A. Suh, BSc, Roderick G. Carter, BSc, Kathleen E. Harris, BSc, Assel Biyasheva, PhD, Kevin Welch, MD, Stephanie Shintani-Smith, MD, David B. Conley, MD, Mark C. Liu, MD, Atsushi Kato, PhD, Pedro C. Avila, MD, Qutayba Hamid, MD, PhD, Leslie C. Grammer, III, MD, Anju T. Peters, MD, Robert C. Kern, MD, Bruce K. Tan, MD, Robert P. Schleimer, PhD PII:
S0091-6749(16)31462-2
DOI:
10.1016/j.jaci.2016.10.039
Reference:
YMAI 12512
To appear in:
Journal of Allergy and Clinical Immunology
Received Date: 27 June 2016 Revised Date:
13 October 2016
Accepted Date: 19 October 2016
Please cite this article as: Pothoven KL, Norton JE, Suh LA, Carter RG, Harris KE, Biyasheva A, Welch K, Shintani-Smith S, Conley DB, Liu MC, Kato A, Avila PC, Hamid Q, Grammer III LC, Peters AT, Kern RC, Tan BK, Schleimer RP, Neutrophils are a major source of the epithelial barrier disrupting cytokine Oncostatin M in mucosal airways disease, Journal of Allergy and Clinical Immunology (2017), doi: 10.1016/j.jaci.2016.10.039. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
ACCEPTED MANUSCRIPT
1
Neutrophils are a major source of the epithelial barrier disrupting cytokine Oncostatin M
2
in mucosal airways disease
3 4 Kathryn L. Pothoven1 PhD, James E. Norton1 MSc, Lydia A. Suh1 BSc, Roderick G.
6
Carter1 BSc, Kathleen E. Harris1 BSc, Assel Biyasheva1 PhD, Kevin Welch2 MD,
7
Stephanie Shintani-Smith2 MD, David B. Conley2 MD, Mark C. Liu3 MD, Atsushi Kato1
8
PhD, Pedro C. Avila1 MD, Qutayba Hamid4 MD, PhD, Leslie C. Grammer III1 MD, Anju T.
9
Peters1 MD, Robert C. Kern2 MD, Bruce K. Tan2 MD, Robert P. Schleimer1,2 PhD
SC
RI PT
5
M AN U
10 11
1
12
Feinberg School of Medicine, Chicago, Ill
13
2
14
Chicago, Ill
15
3
16
Johns Hopkins Asthma and Allergy Center, Baltimore, Md
17
4
18
Research Institute, Montreal, Quebec, Canada
TE D
Divisions of Allergy and Clinical Immunology, Pulmonary and Critical Care Medicine,
Meakins-Christie Laboratories of McGill University and McGill University Health Center
EP
20
Department of Otolaryngology, Northwestern University Feinberg School of Medicine,
AC C
19
Division of Allergy-Immunology, Department of Medicine, Northwestern University
21
This study was supported in part by Grants R37HL068546, R01HL078860,
22
U19AI106683 and T32AI007476-16 from the NIH and by the
23
Ernest S. Bazley Foundation
24 25 26
ACCEPTED MANUSCRIPT
Capsule Summary:
28
In this study, we show that neutrophils are a major source of OSM, the epithelial barrier
29
disrupter, in chronic rhinosinusitis (CRS). GM-CSF, a known stimulus of OSM
30
production, was also elevated in nasal polyps and was detected at levels sufficient to
31
induce in vitro OSM production in peripheral blood neutrophils. OSM-producing
32
neutrophils were found in tissues from patients with severe asthma. Neutrophils may be
33
an important source of OSM resulting in impaired epithelial barrier function.
SC
RI PT
27
34 Clinical implications:
36
Neutrophils are a major source of OSM producing cells in CRS, and severe asthma. GM-
37
CSF was elevated in CRS, at levels sufficient to induce neutrophil derived OSM
38
production. Therapeutic targeting of OSM, neutrophils or GM-CSF may be beneficial in
39
mucosal disease through the restoration of epithelial barrier function.
M AN U
35
TE D
40 Keywords:
42
Oncostatin M, Epithelial barrier, Neutrophils, Chronic rhinosinusitis, Atopic asthma,
43
Granulocyte-Monocyte Colony Stimulating Factor
44
EP
41
Abbreviations:
46
ARG1: Arginase 1
47
BAL: Bronchoalveolar lavage
48
CRS: Chronic rhinosinusitis
49
CRSsNP: CRS without nasal polyps
50
CRSwNP: CRS with nasal polyps
51
ECP: Eosinophil cationic protein
52
FEV1: Forced Expiratory Volume in 1 Second
AC C
45
ACCEPTED MANUSCRIPT
FSTL1: Follistatin-like 1
54
GM-CSF: Granulocyte-Macrophage colony stimulating factor
55
HKSA: Heat killed staphylococcus aureus
56
NHBE: Normal human bronchial epithelial cells
57
MMR: Macrophage mannose receptor
58
PMN: Polymorphonuclear leukocyte/neutrophil
59
OSM: Oncostatin M
60
UT: Uncinate tissue
AC C
EP
TE D
M AN U
SC
RI PT
53
ACCEPTED MANUSCRIPT
61
Abstract:
62 Background: We have previously shown that Oncostatin M (OSM) is elevated in nasal
64
polyps of chronic rhinosinusitis (CRS) patients, as well as in bronchoalveolar lavage
65
(BAL) fluids after segmental allergen challenge in allergic asthmatics. We also showed in
66
vitro that physiological levels of OSM impair barrier function in differentiated airway
67
epithelium.
SC
RI PT
63
68
Objective: We sought to determine which hematopoietic or resident cell type(s) were the
70
source of the OSM expressed in mucosal airways disease.
M AN U
69
71
Methods: Paraffin-embedded NP sections were stained with fluorescence-labeled
73
specific antibodies against OSM, GM-CSF and hematopoietic cell specific markers. Live
74
cells were isolated from NP and matched blood samples for flow cytometric analysis.
75
Neutrophils were isolated from whole blood, cultured with the known OSM inducers GM-
76
CSF and FSTL1, and levels of OSM were measured in the supernatants. Bronchial
77
biopsy sections from controls, moderate asthmatics and severe asthmatics were stained
78
for OSM and neutrophil elastase.
EP
AC C
79
TE D
72
80
Results: OSM staining was observed in NP, showed co-localization with neutrophil
81
elastase (n=10), and did not co-localize with markers for eosinophils, macrophages, T
82
cells or B cells (n=3-5). Flow cytometric analysis of NP (n=9) showed that 5.1±2% of
83
CD45+ cells were OSM+, and of the OSM+ cells, 56±7% were CD16+Siglec8-, indicating
84
neutrophil lineage. Only .6±.4% of CD45+ events from matched blood samples (n=5)
85
were OSM+, suggesting that elevated OSM in CRS was locally stimulated and produced.
86
A majority of OSM+ neutrophils expressed Arginase 1 (72.5±12%), suggesting a N2
ACCEPTED MANUSCRIPT
phenotype. GM-CSF was elevated in nasal polyp tissue compared to control, and was
88
sufficient to induce OSM production (p<.001) in peripheral blood neutrophils in vitro.
89
OSM+ neutrophils were also observed at elevated levels in biopsies from patients with
90
severe asthma. Additionally, OSM protein was elevated in induced sputum from
91
asthmatic patients compared to controls (p<.05).
92
RI PT
87
Conclusions: Neutrophils are a major source of OSM producing cells in CRS and severe
94
asthma.
SC
93
M AN U
95 96 97 98 99
103 104 105 106 107 108 109 110 111 112
EP
102
AC C
101
TE D
100
ACCEPTED MANUSCRIPT
113
Introduction:
114 Epithelial barrier dysfunction has been shown to be important in the pathogenesis of
116
many diseases including asthma, atopic dermatitis, chronic rhinosinusitis (CRS), and
117
eosinophilic esophagitis (EoE) 1, 2, 3, 4, 5, 6, 7. Our laboratory has previously shown that the
118
cytokine Oncostatin M (OSM) was elevated in nasal polyps of chronic rhinosinusitis
119
(CRS) patients, in esophageal biopsies from eosinophilic esophagitis (EoE) patients, and
120
in BAL fluids after allergen challenge in allergic asthmatic patients 8. Physiological levels
121
of OSM were sufficient to induce barrier dysfunction in ex vivo cultured airway epithelium
122
as measured by decreased epithelial resistance, increased epithelial permeability, and
123
loss of tight junction structure. Additionally, levels of OSM in both nasal tissue from CRS
124
patients and bronchoalveolar lavage fluid (BAL) from allergen-challenged allergic
125
asthma patients correlated with markers of epithelial leak, suggesting that OSM may
126
mediate barrier dysfunction in vivo in mucosal disease 8. We hypothesized that OSM-
127
induced loss of barrier function could allow the entry of various allergens, pathogens and
128
environmental factors into the tissue where they could elicit a chronic immune response.
129
It is important now to identify the cell type responsible for OSM production in mucosal
130
disease in order to understand the mechanism of barrier disruption in eosinophilic
131
mucosal disease.
SC
M AN U
TE D
EP
AC C
132
RI PT
115
133
Neutrophil-derived OSM has been shown to contribute to the pathogenesis of many
134
conditions including acute lung injury, asthma, breast cancer, and rheumatoid arthritis 9,
135
10, 11, 12
136
complement factor 5a, and thrombin have all been shown to induce OSM expression in
137
various cell types 13, 14, 15, 16, 17, only GM-CSF was sufficient to induce OSM in neutrophils
138
11, 18
. Although osteopontin, prostaglandin E2 (PGE2), follistatin-like 1 (FSTL1),
. Elbjeirami et al. showed that OSM was induced during neutrophil transendothelial
ACCEPTED MANUSCRIPT
migration in response to endothelial production of GM-CSF 18. Queen et al. showed that
140
co-culture of neutrophils with the breast cancer cell lines MDA-MB-231 and T-47D
141
induced OSM production that was lost when GM-CSF was blocked 11. Additionally, Miller
142
et al, have shown that follistatin like-1 (FSTL1) was necessary for OSM induction in a
143
mouse model of chronic asthma 16. Both GM-CSF and FSTL1 have been shown to be
144
elevated in airways disease 19, 20, 21, 22, and we hypothesized that one or both of these
145
cytokines may induce neutrophil derived OSM in CRS.
SC
RI PT
139
146
Recent studies have described subtypes of polarized neutrophils, the classical,
148
proinflammatory N1 neutrophil and the anti-inflammatory, or tumorigenic N2 neutrophil
149
23
150
M1 and M2 macrophage counterparts 23. N2 neutrophils specifically have been shown to
151
promote tumor growth and metastasis, as well as play an important role in repair
152
processes and the resolution of inflammation 24, 25, 26. N1 and N2 neutrophil polarization
153
has been best studied in mice, and N2 neutrophils have been shown to express the
154
macrophage mannose receptor (MMR), arginase 1 (ARG1), chitinase-like 3 (Ym1), IL-10
155
and TGFβ, which are also characteristic markers of M2 macrophages. Both M2
156
macrophages and N2 neutrophils have been associated with tissue repair mechanisms
157
25
158
cytokines and chemokines such as TNF, IL-1β, CCL3, CCL5, IL-6 and IL-12 25. Although
159
progress is being made, N2 neutrophils in humans are much less extensively studied,
160
and the markers used to define N2 neutrophils in mice may not all be relevant as
161
markers for human N2 neutrophils 27. Because N2 neutrophils and OSM have been
162
shown to be important for repair processes, we wanted to test the hypothesis that N2
163
neutrophils were responsible for OSM production in CRS. In this manuscript we show
164
that neutrophils are a major source of OSM producing cells in nasal polyps and
M AN U
147
EP
TE D
. These N1 and N2 neutrophils have been shown to be very similar in function to their
AC C
. Compared to N2, N1 neutrophils have been shown to express more proinflammatory
ACCEPTED MANUSCRIPT
165
neutrophils also express OSM in severe asthma. We also show that the OSM inducer,
166
granulocyte-macrophage colony-stimulating factor (GM-CSF), was elevated in nasal
167
polyps at levels sufficient to induce OSM production in cultured neutrophils.
RI PT
168 169 170
SC
171 172
M AN U
173 174 175 176 177
181 182 183 184 185 186 187 188 189 190
EP
180
AC C
179
TE D
178
ACCEPTED MANUSCRIPT
191
Methods:
192 Patients and tissue sample collection:
194
Control subjects and patients with CRS were recruited from the Allergy-Immunology and
195
Otolaryngology clinics of the Northwestern Medicine and the Northwestern Sinus Center
196
at Northwestern Medicine. Nasal polyps and peripheral blood were obtained during
197
routine functional endoscopic sinus surgery from patients with CRS, as defined by the
198
American Academy of Otolaryngology—Head and Neck Surgery Chronic Rhinosinusitis
199
Task Force 28. Patients with established immunodeficiency, pregnancy, coagulation
200
disorder, or cystic fibrosis were excluded from this study. Characteristics of subjects in
201
the study are shown in table E1. Control subjects did not have a history of sinonasal
202
inflammation, and tissue was collected during endoscopic skull-base tumor excisions,
203
and benign skull-base procedures including cerebrospinal fluid leak, encephalocele, and
204
petrous apex cholesterol granuloma. The tumors excised include: pituitary adenoma (5),
205
chordoma (4), meningioma (2), esthesioneuroblastoma (1), clival metastatic carcinoma
206
(1), inverted Schneiderian papilloma (1) and osteoma (1). Asthma and atopic status
207
were determined by the surgeon treating the patient. Segmental allergen challenge and
208
bronchoalveolar lavage (BAL) were performed in a patient cohort of allergic asthmatics,
209
as previously described 29, 30. Bronchial biopsies were obtained from asthmatics during
210
bronchoscopy at the Meakins-Christie Laboratories at McGill University Montreal,
211
Quebec with the approval of the respective Institutional Review Boards, which has been
212
previously described 31. Induced sputum was obtained in the Allergy-Immunology clinic
213
of Northwestern Medicine as previously described 32, 33.
214 215 216
AC C
EP
TE D
M AN U
SC
RI PT
193
ACCEPTED MANUSCRIPT
Immunofluorescent staining:
218
Nasal polyp specimens were processed for histology as previously described 34.
219
Sections were blocked with PBS containing 5% goat serum (Vector Laboratories,
220
Burlingame, Calif) and .5% Triton X-100 (Sigma Aldrich, St Louis, MO) for 1 hour at
221
room temperature (RT), then incubated with either rabbit anti-OSM (N-1, 1:400, Santa
222
Cruz Biotechnology, Dallas, TX), mouse anti-neutrophil elastase (NP57, 1:100, Dako
223
North America, Carpinteria, CA), mouse anti-CD68 (PG-M1, 1:50, Thermofisher), mouse
224
anti-ECP (EG2, 1:1000, Diagnostics Development, Uppsala, Sweden), mouse anti-
225
tryptase (AA1, 1:10000, Thermofisher), mouse anti-CD3 (SP7, 1:1, Thermofisher)
226
mouse anti-CD20 (L26, 1:250, Thermofisher) or rat anti-GM-CSF (BVD2-21C11, 1:100,
227
Abcam, Cambridge, MA) for 1 hour at RT. The sections were then incubated with
228
secondary antibody for 1 hour at RT with Alexa Fluor 488 goat anti-rabbit antibody,
229
Alexa Fluor goat anti-mouse 568, Alexa Fluor goat anti-rat 568, Alexa Fluor goat anti
230
rabbit 647, or Alexa Fluor goat anti-mouse 647(Thermofisher). Slides were mounted with
231
slowfade gold antifade reagent with DAPI counter stain (Thermofisher). To control for
232
background staining, additional slides were stained with the secondary antibody only.
233
Imaging was performed using a Nikon A1R confocal microscope using the 20x objective.
234
Images were processed using ImageJ software.
SC
M AN U
TE D
EP
AC C
235
RI PT
217
236
Flow Cytometric Analysis:
237
Live cells were isolated from nasal polyp tissues obtained during endoscopic sinus
238
surgery. Nasal polyps were incubated overnight on an oscillator in 1mg/mL collagenase I
239
(Worthington Biochemical Corporation, Lakewood, NJ) and 30ug/mL DNase
240
(Worthington Biochemical Corporation) in RPMI at 4 degrees in a gentleMACS tube.
241
Cells were then dissociated using a gentleMACS cell dissociator (Miltenyi Biotec, San
242
Diego, CA) and then passed through a 70µm cell strainer (BD Biosciences, San Jose,
ACCEPTED MANUSCRIPT
CA). Live blood cells were isolated from the buffy coat of centrifuged peripheral blood to
244
ensure that the granulocyte population would be present for analysis. For flow cytometry,
245
cells were first stained using the LIVE/DEAD Aqua dead cell staining kit (Thermofisher,
246
Waltham, MA), to facilitate gating only on live cells. Cells were then blocked using 10µL
247
of Fc blocking reagent for 15min (Miltenyi Biotec, San Diego, CA) and then cells were
248
stained at RT for 15min with one of 3 panels of antibodies listed in supplemental table
249
E2. Following cell surface staining, cells were then fixed and permeablized using the BD
250
cytofix/cytoperm kit for intracellular staining following manufacturer’s protocols (BD
251
Biosciences, San Jose, CA), and cells were stained intracellularly for 30min at 4°C. Cells
252
were analyzed on an LSRII flow cytometer with FACSDiva software. All analysis and
253
compensation were done using FlowJo software (Treestar, Ashland, OR). All flow
254
cytometric studies were prepared using the appropriate single stained control beads (BD
255
Biosciences, San Jose, CA and eBioscience, San Diego, CA) and fluorescence minus
256
one (FMO) controls.
SC
M AN U
TE D
257
RI PT
243
Neutrophil isolation and cell culture:
259
Neutrophils were isolated from peripheral blood using an EasySep direct human
260
neutrophil isolation kit (Stemcell, Vancouver, BC). Cells were then counted and cultured
261
at 107 cells/mL in RPMI supplemented with 5% fetal bovine serum and 1%
262
penicillin/streptomycin. Neutrophil cultures were treated with GM-CSF (1-25 ng/mL, R&D
263
Systems, Minneapolis, MN), FSTL1 (100-1000 ng/mL, R&D Systems), Interferon-γ
264
(IFNγ) (10 ng/mL, R&D Systems), lipopolysaccharide (LPS) (1µg/mL, Enzo Life
265
Sciences, Farmingdale, NY), Interleukin 4 (IL-4) (20 ng/mL, R&D Systems), Interleukin
266
13 (IL-13) (20 ng/mL, R&D Systems), Interleukin 25 (IL-25) (10-100ng/mL, R&D
267
Systems), Interleukin 33 (IL-33) (10-100 ng/mL, R&D Systems), thymic stromal
268
lymphopoietin (TSLP) (10-100 ng/mL, R&D Systems) or Leukotriene C4 (LTC4) (10-6-10-7
AC C
EP
258
ACCEPTED MANUSCRIPT
Cayman Chemical, Ann Arbor, MI) for 20 hours, cell culture supernatants were collected
270
for protein analysis and cell lysates were collected to isolate RNA. Plates were coated
271
with E-selectin, ICAM (50ng/well, Peprotech, Rocky Hill, NJ) or bovine serum albumin
272
(BSA) (50ng/well, Sigma Aldrich) in pH 8 tris buffered saline (TBS) overnight at 4
273
degrees. Neutrophils were seeded into the wells and the culture supernatants were
274
harvested at 20 hours.
RI PT
269
SC
275 NHBE cell culture and injury:
277
Normal human bronchial epithelial (NHBE) cells were cultured at air-liquid interface
278
(ALI), as previously described 8. As a model of injury, fully differentiated NHBE were
279
treated apically with .5mL of either PBS, or PBS containing 5x108 particles/mL of heat
280
killed staphylococcus aureus (HKSA). Following culture for 48h, cell supernatants were
281
collected for protein analysis.
TE D
282
M AN U
276
Protein quantification:
284
Oncostatin M protein was detected in neutrophil cell culture supernatants using a
285
DuoSet ELISA kit (R&D Systems, Minneapolis, MN). FSTL1 and GM-CSF protein were
286
detected in neutrophil cell culture supernatants and FSTL1 was detected in sinus tissue
287
extracts using DuoSet ELISA kits (R&D Systems). GM-CSF protein was detected in
288
sinus tissue lysates using a Human Magnetic Protein Luminex Assay (R&D Systems).
289
OSM protein was detected in sinus tissue lysates as previously described 8. Neutrophil
290
Elastase protein (Hycult Biotech, Plymouth Meeting, PA) and Eosinophilic Cationic
291
Protein (MBL International, Woburn, MA) were detected using ELISA, and sinus tissue
292
extracts were prepared as previously described 35.
293 294
AC C
EP
283
ACCEPTED MANUSCRIPT
295 Quantitative RT-PCR and microarray analysis:
297
RNA isolation from tissue samples was prepared as previously described 34. Single-
298
strand cDNA was synthesized from 0.5µg of total RNA with SuperScript II reverse
299
transcriptase and random primers (Invitrogen, Carlsbad, Calif). Quantitative real-time
300
RT-PCR was performed using an Applied Biosystems 7500 Sequence Detection System
301
(Applied Biosystems, Foster City, Calif) in 20µL reactions (10µL of 2x TaqMan Master
302
mix [Applied Biosystems], 400nmol/L of each primer and 200nmol/L of TaqMan probe
303
plus 10µg of cDNA). Primer and probe sets for OSM (Hs00968300_g1) ARG1 and
304
MRC1 (Hs00267207_m1) were purchased from Applied Biosystems. Primers
305
(Forward:GAAGGTGAAGGTCGGAGTC, Reverse: GAAGATGGTGATGGGATTTC) and
306
probe (6-VIC-CAAGCTTCCCGTTCTCAGCC-MGB) for GAPDH were synthesized by
307
IDT technologies (IDT Technologies, Coralville, IA) and used to detect the housekeeping
308
gene in all RT-PCR studies. Microarray analysis was performed, as previously described
309
36, 37
TE D
M AN U
SC
RI PT
296
. All microarray data have been deposited to gene expression omnibus: GSE36830.
310 Statistical Analysis:
312
All data were analyzed using GraphPad Prism 6 software (GraphPad Software, La Jolla,
313
Calif). All data are reported as mean ± SEM. Differences between groups were analyzed
314
using a non-parametric Kruskal-Wallis ANOVA or Mann-Whitney U test. Correlations
315
were assessed using the Spearman correlation. Significance: *p<0.05, **p<0.01,
316
***p<0.001, ****p<.0001.
317 318 319 320
AC C
EP
311
ACCEPTED MANUSCRIPT
321 322
Results:
323 Neutrophils are a major source of OSM in Nasal Polyps:
325
To determine which cell type was making OSM in CRS, we first obtained sections from
326
nasal polyps and used immunofluorescence and specific antibodies to stain for OSM
327
and markers of various cell types including CD3, CD20, tryptase, elastase, ECP, and
328
CD68. We did not observe co-localization of OSM with markers for macrophages (Figure
329
1A-B) or eosinophils (Figure 1C-D), while we observed some co-localization with a minor
330
population of mast cells (Figure 1E-F). We also did not observe co-localization of OSM
331
with T cells or B cells (data not shown). Elastase positive cells made up the majority of
332
the OSM positive cells, suggesting that neutrophils are an important source of OSM
333
(Figure 1G-H). Control images with no primary antibody for CD68, ECP, tryptase, and
334
elastase staining strategies are shown in Figure E1A-D. Further analysis of our
335
previously published microarray database (GEO: GSE36830) showed that OSM mRNA
336
expression correlated with the granulocyte marker CD16 (FCGR3) (r = .61, p<.01; Figure
337
E1E), and the neutrophil specific protease, cathepsin G (CTSG) (r = .56, p<.01; Figure
338
E1F) in nasal polyps and uncinate tissue from control, CRSsNP and CRSwNP patients,
339
suggesting a potential relationship between OSM expression and neutrophils
340
FCGR3 is not neutrophil specific, FCGR3 and CTSG correlated with each other in the
341
database (r =.56, p<.01; Figure E1G), suggesting that neutrophils are responsible for
342
much of the FCGR3 expression in sinonasal tissue. We also observed a positive
343
correlation between elastase and OSM using lysates from both nasal polyps, and
344
uncinates from control, CRSsNP, and CRSwNP patients (r = .65, p<.01; Figure E1H).
345 346
36
. While
AC C
EP
TE D
M AN U
SC
RI PT
324
ACCEPTED MANUSCRIPT
347 Flow cytometric analysis of OSM producing cells in nasal polyps:
349
To further validate the observation that neutrophils are a major source of OSM
350
producers in nasal polyps, we utilized flow cytometry. Live cells were isolated from nasal
351
polyps, and stained with CD45, OSM, CD16 and Siglec-8. Within the CD45 gate,
352
5.1±2% of the cells were OSM+, and the OSM+ cells were determined using the
353
appropriate FMO control. Of the OSM+ cells, 29.8±8% were excluded during analysis of
354
the data because the wavelengths of the fluorophores used to stain for CD16-APCH7
355
siglec 8-Alexa 647 were near each other and the rigorous compensation algorithm that
356
we used excluded them to ensure the accuracy of the data. The majority of the cells that
357
were excluded were single positive for either CD16 or siglec 8, suggesting they were
358
granulocytes. Within the OSM+ gate, we observed two main populations; the major
359
population was CD16+Siglec-8- (80.4±5%), which were neutrophils, and the minor
360
population was CD16+Siglec-8+ (14.1±5%), which were either mast cells, basophils, or
361
eosinophils. We did observe a small CD16-Siglec-8- population (5.5±3%) that was
362
present in less than half of the polyps we analyzed (Figure 2A-C). Additionally,
363
neutrophils were isolated from peripheral blood and nasal polyp tissue of five matched
364
CRSwNP patients, and OSM expression in blood neutrophils was only minimally
365
observed in one patient (.6±.4%), indicating that in vivo OSM induction was likely a
366
locally induced event (Figure 2D). The full gating strategy for this antibody panel,
367
including the OSM FMO control is available in Figure E2.
SC
M AN U
TE D
EP
AC C
368
RI PT
348
369
OSM was expressed in a minor population of mast cells:
370
To further identify the cells within the minor CD16+Siglec8+ population of OSM
371
producers, we stained live cells isolated from nasal polyps with CD45, OSM, CD16,
372
FcεRI, c-kit, and either EMR-1 or Siglec8. In the CD45+ gate, 4.6±2% events were
ACCEPTED MANUSCRIPT
OSM+. Within the OSM+, 15.6±10% were FcεRI+c-kit+, indicating mast cells, 3.7±1%
374
were FcεRI+c-kit-, indicating basophils, and of the FcεRI-c-kit- cells, only 2.3±1% were
375
emr1+ or siglec8+, which would indicate eosinophils, leaving the remaining 73.3±11% of
376
the cells neutrophils (Figure 3). The full gating scheme for this antibody panel is
377
available in Figure E3, and the sample chosen as the representative example was
378
selected to best show the gating strategy for basophils and mast cells.
RI PT
373
SC
379
GM-CSF was elevated in nasal polyps, and induces neutrophil derived OSM production
381
in vitro:
382
Since GM-CSF and FSTL-1 have been shown to be important inducers of OSM, our next
383
aim was to determine whether GM-CSF and/or FSTL1 may be playing a role in in vivo
384
induction of neutrophil derived OSM. We first wanted to determine whether GM-CSF or
385
FSTL1 were sufficient to induce OSM in neutrophils. We treated neutrophils isolated
386
from whole blood in vitro for 20 hours with GM-CSF and/or FSTL1 11, 16, 18. GM-CSF
387
alone induced OSM protein release into cell culture supernatants at 1, 5, and 25 ng/mL,
388
whereas FSTL1 alone at 100 or 1000ng/mL did not induce OSM in purified neutrophils;
389
the combination of 1 or 25 ng/mL GM-CSF with 1000ng/mL FSTL1 was not more active
390
than GM-CSF alone (Figure 4A). This data was analyzed using both mean and median
391
and the statistics were the same; the data represented in Figure 4A shows the mean
392
values. Analysis of OSM mRNA following culture showed that both GM-CSF- and
393
FSTL1-treated neutrophils expressed OSM transcripts, suggesting that neutrophils are
394
capable of de novo production of OSM, although levels of OSM were not elevated
395
compared to control. (Figure E4A). We next wanted to determine whether other type 2
396
mediators were sufficient to induce neutrophil derived OSM. Neutrophils were either left
397
unstimulated, or stimulated with GM-CSF (25ng/mL), IL-25 (10-100 ng/mL), IL-33 (10-
398
100 ng/mL), TSLP (10-100 ng/mL) or leukotriene C4 (10-6 – 10-7 M) Figure E4B). Only
AC C
EP
TE D
M AN U
380
ACCEPTED MANUSCRIPT
GM-CSF was sufficient to induce OSM in neutrophils. We also wanted to determine
400
whether neutrophil ligation with E-selectin and ICAM would induce neutrophil derived
401
OSM. We coated plates with E-selectin, ICAM, and BSA as a control, and then cultured
402
neutrophils for 20 hours in the coated wells. We did not observe any induction of OSM
403
with E-selectin or ICAM ligation (Figure E4C).
404
RI PT
399
OSM has been shown to be important for the early stages of epithelial repair, specifically
406
by inducing basal cell proliferation and migration 38. We next hypothesized that injured
407
epithelium may upregulate GM-CSF to induce OSM production from infiltrating
408
neutrophils, which would then promote the early stages of epithelial repair. To determine
409
whether GM-CSF could potentially be produced from injured epithelium, we injured
410
normal human bronchial epithelial (NHBE) cells that were grown at air-liquid interface,
411
and when they were fully differentiated they were treated with heat killed Staphlococcus
412
aureus (HKSA) as described 39. Exposure for 48 hr to HKSA at a concentration of 5 x 108
413
particles/mL induced GM-CSF secretion into cell culture supernatants (Figure 4B).
414
Although HKSA can activate epithelium by various pathways, these data support the
415
concept that injured epithelium is capable of producing GM-CSF, which has the potential
416
to induce OSM expression in neutrophils that migrate to the site of injury. Since GM-CSF
417
was sufficient to induce neutrophil-derived OSM secretion, we next hypothesized that
418
GM-CSF would be elevated in nasal polyps compared to control UT. To do this, we
419
measured GM-CSF and FSTL1 protein expression in nasal polyps and UT from CRS
420
patients and controls. GM-CSF was elevated in nasal polyps compared to control UT
421
while FSTL1 was not elevated, suggesting that GM-CSF could be responsible for
422
induction of neutrophil OSM expression in nasal polyps (Figure 4C, D).
423
AC C
EP
TE D
M AN U
SC
405
ACCEPTED MANUSCRIPT
To determine the cell types responsible for GM-CSF production in nasal polyps, we
425
stained nasal polyp sections (n=4) for OSM, neutrophil elastase, and GM-CSF (Figure
426
4E and Figure E4D-E). Surprisingly, the neutrophils that expressed OSM also expressed
427
GM-CSF. GM-CSF has been shown to protect neutrophils against apoptosis 40,
428
suggesting that neutrophils may have an autocrine pathway by which GM-CSF
429
expression prolongs their own survival and promotes expression of OSM. We did not
430
observe GM-CSF expression in the epithelium in nasal polyps.
SC
RI PT
424
431
M AN U
432
OSM producing neutrophils may have a non-classical phenotype:
434
We next tested whether OSM-producing neutrophils were phenotypically classical,
435
inflammatory N1 neutrophils or tissue repair-associated N2 neutrophils. We utilized flow
436
cytometry, and stained cells isolated from nasal polyps for CD45, OSM, CD16, IL-5R,
437
myeloperoxidase (MPO) and Arg1 (Figure E5A). No flow cytometry antibodies were
438
available for elastase, so we used anti-MPO instead. Of cells within the CD45+ gate,
439
3.7±1% were OSM+, and of the OSM+ cells, 56.0±8% were CD16+IL-5R- neutrophils. In
440
the neutrophil gate, we found two populations; MPOhiArg1+ (72.5±12%) and MPOloArg1-
441
(22.1±8%) (Figure 5A-B). The expression of Arg1 suggests that the majority of
442
neutrophils taken from polyp tissue in vivo were not classical N1 neutrophils. As
443
discussed above, GM-CSF and FSTL1 are two potential inducers of OSM in vivo. To
444
determine whether GM-CSF induced neutrophils to assume a non-classical phenotype,
445
we compared neutrophils treated in vitro with GM-CSF to neutrophils that were polarized
446
either toward an N1 phenotype using LPS and IFNγ or toward an N2 phenotype using
447
IL-4 and IL-13. As expected we saw increased levels of OSM protein released into the
448
culture supernatants in response to GM-CSF, but we did not see any increased OSM in
449
the supernatants in either the N1 or N2 polarizing conditions (Figure 5C). To determine
AC C
EP
TE D
433
ACCEPTED MANUSCRIPT
whether neutrophil phenotypes influence OSM expression, we polarized neutrophils to
451
N1 or N2 conditions, or left them unstimulated for 24 hours. Following polarization, the
452
neutrophils were either left unstimulated or stimulated with GM-CSF. Unactivated, N1
453
and N2 neutrophils had no difference in expression of OSM, suggesting that all
454
neutrophils are capable of OSM expression (data not shown). To further define the
455
phenotype of our GM-CSF and FSTL1 treated neutrophils, we analyzed mRNA
456
expression of the type 2 markers mannose receptor (MMR) and arginase-1 (Arg1).
457
Neither GM-CSF or FSTL1 or their combination caused any induction of Arg1
458
(FigureE5B). However, GM-CSF and FSTL1 were both sufficient to induce elevated
459
mRNA for another N2 marker, MRC1, suggesting that GM-CSF and FSTL1 may induce
460
neutrophils to assume elements of an N2-like phenotypic state (Figure 5D).
M AN U
SC
RI PT
450
461
Neutrophil expression of OSM in asthmatic patients:
463
To determine whether neutrophils were also a source of OSM in asthma, we stained
464
bronchial biopsies from control patients (n=4) as well as those with moderate (n=5) and
465
severe (n=6) asthma for OSM and neutrophil elastase (Figure 6A). We counted
466
neutrophils and OSM+ cells in each specimen, and the tabulated data are represented in
467
Table E3. We did not observe any OSM+ cells in the control biopsies, but we detected
468
OSM+ cells in 60% of biopsies from moderate asthmatics and 100% of biopsies from
469
severe asthmatics (Figure E6A). Of the neutrophils counted in each specimen, 0±0%
470
were OSM positive in control biopsies, 14.0±9.8% were positive in moderate asthmatic
471
biopsies, and 30.1±10.2% were OSM positive in severe asthmatic biopsies (Figure E6B).
472
Additionally, of the OSM+ cells counted, 0±0% were neutrophils in control biopsies,
473
35.0±21.8% were neutrophils in moderate asthmatic biopsies, and 52.1±15.9% were
474
neutrophils in severe asthmatic biopsies (Figure 6B). Neutrophil counts were normalized
475
per mm of basement membrane within the biopsy section, and no differences in the total
AC C
EP
TE D
462
ACCEPTED MANUSCRIPT
476
neutrophil counts/mm basement membrane were observed between controls and
477
asthmatics, suggesting an increased state of activation of neutrophils rather than
478
increased neutrophil numbers (Figure E6C).
RI PT
479 To further test whether OSM is produced in the airways of patients with asthma, we
481
evaluated a set of induced sputum samples from controls and asthmatic subjects. We
482
observed elevated levels of OSM in induced sputum from asthmatic patients compared
483
to controls (Figure 6C). Finally, we previously reported that segmental allergen
484
challenge in allergic asthmatic patients leads to increased levels of OSM protein in BAL
485
fluids 8. We reevaluated the previous data set to test the role of neutrophils in the
486
response and found that OSM levels correlated with total neutrophil counts, total
487
lymphocyte counts, and total cell counts in the BAL (Figure E6D-E, H), but not with total
488
eosinophil counts or total macrophage counts (Figure E6F-G). Additionally, we
489
previously reported that a concentration of 50pg/mL of GM-CSF was measured in the
490
BAL in a separate cohort of allergen challenged allergic asthmatics, suggesting that GM-
491
CSF may also induce neutrophil-derived OSM production in asthmatic patients 20.
492
Together, these data suggest that OSM is produced by neutrophils in both sinus and
493
lung tissue undergoing allergic inflammation and may play a role in epithelial barrier
494
dysfunction.
496 497 498 499 500 501
M AN U
TE D
EP
AC C
495
SC
480
ACCEPTED MANUSCRIPT
502 Discussion:
504
We have previously showed that OSM was elevated in nasal polyp tissue from CRS
505
patients, and that physiological levels of OSM are sufficient to induce profound loss of
506
barrier function in differentiated bronchial and nasal epithelial cells cultured at air-liquid
507
interface 8. We hypothesized that elevated OSM in CRS may mediate epithelial barrier
508
dysfunction in vivo. Nasal polyps have been shown to express robust type 2
509
inflammation 41, 42, 43, and we originally speculated that the OSM producers in nasal
510
polyps were likely to be eosinophils, M2 macrophages, or mast cells. In the present
511
manuscript, we found rather that neutrophils are a major population of cells expressing
512
OSM in CRS tissues (Figure 1-3). We also identified a minor population of OSM+ cells
513
that were siglec 8+, and these cells were found to be mast cells (Figure 2-3). Neutrophils
514
are classically thought to be a first line of defense in response to tissue injury 44, and
515
neutrophil-derived OSM has been shown to be important in the early stages of epithelial
516
repair when basal cells proliferate and cover a wound 45. Neutrophils are known to be
517
increased in CRSsNP, CRSwNP and in certain forms of asthma, and production of OSM
518
by neutrophils could participate in both ongoing epithelial repair as well as promote the
519
epithelial barrier dysfunction that is observed in these diseases. While we did not
520
investigate whether neutrophils were responsible for OSM production in CRSsNP, we
521
suspect that neutrophil-derived OSM may also play a role in barrier dysfunction in this
522
non-polypoid form of CRS. It is possible that under normal circumstances, neutrophil-
523
derived OSM production is transient, allowing basal cells to reinitiate contact inhibition,
524
stop proliferating, and then differentiate back into mature epithelium. However, chronic
525
expression of OSM by neutrophils in mucosal disease may prolong or even perpetuate
526
the state of the epithelial repair process (Figure 7). OSM has been shown to induce
AC C
EP
TE D
M AN U
SC
RI PT
503
ACCEPTED MANUSCRIPT
527
epithelial-mesenchymal transition (EMT) 46, which antagonizes epithelial differentiation,
528
and expression of EMT biomarkers is well established in both CRS and asthma 47.
529 Neutrophils are not classically associated with type 2 immune responses, and are
531
generally considered to be a type 1 immune response effector cell. However,
532
neutrophilic subsets of CRS and asthma have been described. While nasal polyps in
533
Western countries tend to be highly eosinophilic, a significant proportion of nasal polyps
534
from patients in Eastern Asian countries are neutrophilic 214, 215, 216. In unpublished
535
studies not shown, we found that nasal polyps, that are highly eosinophilic in our
536
Chicago cohort, nonetheless have significant levels of neutrophils and neutrophil
537
elastase. Increased neutrophilia within nasal polyps has been linked to decreased
538
responsiveness to corticosteroid treatment 217. Severe asthma is a subtype of asthma
539
that is often characterized by neutrophilia 218, 219. It is thought that neutrophilia in asthma
540
associates with more severe disease, again, because neutrophils are not particularly
541
responsive to corticosteroid treatment 220. However, little is known about the role of
542
neutrophils in eosinophilic mucosal disease outside of these known neutrophilic
543
subtypes of CRS and asthma. Our data suggest that neutrophils may be capable of
544
playing a pathogenic role in eosinophilic mucosal disease through the induction of
545
epithelial barrier dysfunction mediated by the concurrent production of OSM and GM-
546
CSF.
SC
M AN U
TE D
EP
AC C
547
RI PT
530
548
We present circumstantial evidence that the neutrophils responsible for OSM production
549
are of the N2 phenotype. Recent studies have shown that neutrophils can be polarized
550
into either classical N1 neutrophils, which are thought to have a more inflammatory
551
phenotype, and N2 neutrophils, which promote tissue repair and tumorigenesis and are
552
thought to have an anti-inflammatory or regulatory phenotype 23, 24, 25, 26. We showed that
ACCEPTED MANUSCRIPT
the majority of OSM+ neutrophils from nasal polyps expressed the type 2 marker Arg1
554
(Figure 5), indicating a phenotype resembling N2 neutrophils more than N1 neutrophils.
555
We also showed that both GM-CSF and FSTL1 induced in vitro-stimulated neutrophils to
556
express elevated MMR compared to unstimulated neutrophils and N1 polarized
557
neutrophils, also indicating that OSM producing neutrophils could be skewed toward the
558
N2 phenotype (Figure 5). Since both GM-CSF and FSTL1 have been shown to be
559
important inducers of OSM 11, 16, 18, we tested their ability to induce OSM in vitro in
560
cultured neutrophils; of these two stimuli, only GM-CSF induced OSM production.
561
However, since we did not see any induction of Arg1 in response to GM-CSF, another
562
factor may be necessary for full induction of the N2 phenotype. These studies implicate
563
GM-CSF as a potential inducer of OSM in neutrophils in vivo. Since circulating
564
neutrophils do not express OSM in patients, this induction likely takes place within nasal
565
polyp or lung tissue where these inducers can be found.
M AN U
SC
RI PT
553
TE D
566
Interestingly, immunofluorescent staining showed that GM-CSF localized to OSM-
568
producing neutrophils in nasal polyps, suggesting that OSM producing neutrophils may
569
induce their own OSM through autocrine production of GM-CSF (Figure 4E). There is
570
precedent for such a concept, as Wardlaw and colleagues showed that eosinophils
571
produce GM-CSF to prolong their own survival 48. In addition, Durand et al, have shown
572
that autoantibody ligation of FcγRIIIb (CD16) receptors on the surface of neutrophils
573
protects against apoptosis through the induction of autocrine GM-CSF 40. We have
574
previously shown elevated levels of autoantibodies in nasal polyps, suggesting that
575
elevated autoantibodies in nasal polyps may trigger GM-CSF expression in the
576
infiltrating neutrophils by a similar mechanism 49. Whether production of OSM by
577
neutrophils via an autocrine GM-CSF dependent pathway occurs in CRSwNP and
578
asthma is worthy of further investigation.
AC C
EP
567
ACCEPTED MANUSCRIPT
579 Importantly, we have also shown that OSM was elevated in induced sputum from
581
asthmatics compared to controls and that OSM+ cells were present in bronchial biopsies
582
from 100% of severe asthma patients, 60% of moderate asthma patients and none of
583
the healthy controls. These results suggest that the expression of OSM and its
584
association with barrier disruption applies to CRSwNP, asthma and, as shown in our
585
previous study, eosinophilic esophagitis. In the present study, 52% of the OSM+ cells
586
were neutrophils in severe asthmatics and 35% of the neutrophils were OSM+ in
587
moderate asthmatics, suggesting that neutrophil derived OSM plays a role in the
588
pathogenesis of asthma. OSM has previously been implicated in the induction of fibrosis
589
in a mouse model of asthma, though it was derived from macrophages 16, 50.
M AN U
SC
RI PT
580
590
Given that GM-CSF may promote prolonged survival in neutrophils as well as induce
592
OSM, therapeutic targeting of GM-CSF could potentially be beneficial in the treatment of
593
CRS and possibly other mucosal diseases like asthma and eosinophilic esophagitis.
594
Inhibition of GM-CSF could block neutrophil-derived OSM production, allowing
595
epithelium to enter the late phase of repair and differentiation, potentially resolving
596
chronic inflammation resulting from barrier dysfunction. Two strategies have been used
597
to inhibit GM-CSF in human trials, monoclonal antibodies against GM-CSF, and a
598
monoclonal antibody against GM-CSFRα. Clinical trials have been conducted with
599
monoclonal antibodies against GM-CSF (MOR103, KB003, and namilumab) in
600
rheumatoid arthritis, plaque psoriasis, and severe asthma. A phase II trial was
601
conducted using KB003 in asthmatic patients that were poorly controlled with long-acting
602
bronchodilators and either inhaled or oral corticosteroids. They did not observe improved
603
forced expiratory volume in 1 second (FEV1) in the entire patient cohort. However, when
604
they separated patients based on markers of poorly controlled asthma, peripheral blood
AC C
EP
TE D
591
ACCEPTED MANUSCRIPT
605
eosinophilia, low baseline FEV1, and high bronchodilator reversibility, they did detect an
606
improvement in FEV1 in response to treatment with KB003 51.
607 In summary, we have shown that neutrophils are a major source of OSM producing cells
609
in nasal polyps using both immunofluorescence and flow cytometric analyses. We have
610
also shown that GM-CSF is elevated in nasal polyps, and that physiological levels of
611
GM-CSF are sufficient to induce OSM production in cultured neutrophils. Interestingly,
612
GM-CSF is expressed by many of the same neutrophils that produce OSM in nasal
613
polyps, suggesting the presence of an autocrine activation. OSM-producing neutrophils
614
phenotypically resemble N2 neutrophils based on limited biomarker evaluation, but
615
further characterization of these cells will be required. OSM+ cells are also present in
616
biopsies from severe asthmatics, and approximately half of the OSM producing cells are
617
neutrophils, suggesting that neutrophil-derived OSM may also play a role in asthma.
618
Taken together, these studies further implicate OSM in barrier dysfunction in CRSwNP
619
as well as in asthma, and suggest that the OSM found in these diseases is produced by
620
neutrophils. Therapeutic intervention targeting either GM-CSF or OSM may thus be
621
beneficial in the treatment of CRS and asthma.
624 625 626 627 628 629 630
SC
M AN U
TE D
EP
623
AC C
622
RI PT
608
ACCEPTED MANUSCRIPT
Acknowledgements:
632
The authors wish to thank Dr. Bruce Bochner for the very kind gift of anti-siglec 8
633
antibody and would like to thank Ms. Jacqi Shaffer for the outstanding medical
634
illustration.
RI PT
631
635
This study was supported in part by Grants R37HL068546, R01HL078860, the Chronic
637
Rhinosinusitis Integrative Studies Program (CRISP - U19AI106683), and T32AI007476-
638
16 from the NIH, as well as by the Ernest S. Bazley Foundation.
SC
636
M AN U
639 640
Imaging work was performed at the Northwestern University Center for Advanced
641
Microscopy generously supported by NCI CCSG P30 CA060553 awarded to the Robert
642
H. Lurie Comprehensive Cancer Center.
EP AC C
644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668
TE D
643
ACCEPTED MANUSCRIPT
References:
Holgate, S.T., Lackie, P., Wilson, S., Roche, W. & Davies, D. Bronchial epithelium as a key regulator of airway allergen sensitization and remodeling in asthma. American journal of respiratory and critical care medicine 162, S113-117 (2000).
2.
Xiao, C. et al. Defective epithelial barrier function in asthma. The Journal of allergy and clinical immunology 128, 549-556 e541-512 (2011).
3.
Dejima, K., Randell, S.H., Stutts, M.J., Senior, B.A. & Boucher, R.C. Potential role of abnormal ion transport in the pathogenesis of chronic sinusitis. Archives of otolaryngology--head & neck surgery 132, 1352-1362 (2006).
4.
Bernstein, J.M., Gorfien, J., Noble, B. & Yankaskas, J.R. Nasal polyposis: immunohistochemistry and bioelectrical findings (a hypothesis for the development of nasal polyps). The Journal of allergy and clinical immunology 99, 165-175 (1997).
5.
Kern, R.C. et al. Perspectives on the etiology of chronic rhinosinusitis: an immune barrier hypothesis. American journal of rhinology 22, 549-559 (2008).
6.
Simon, D., Radonjic-Hosli, S., Straumann, A., Yousefi, S. & Simon, H.U. Active eosinophilic esophagitis is characterized by epithelial barrier defects and eosinophil extracellular trap formation. Allergy 70, 443-452 (2015).
7.
Zhang, N., Van Crombruggen, K., Gevaert, E. & Bachert, C. Barrier function of the nasal mucosa in health and type-2 biased airway diseases. Allergy 71, 295-307 (2016).
8.
Pothoven, K.L. et al. Oncostatin M promotes mucosal epithelial barrier dysfunction, and its expression is increased in patients with eosinophilic mucosal disease. The Journal of allergy and clinical immunology 136, 737-746 e734 (2015).
9.
EP
TE D
M AN U
SC
RI PT
1.
AC C
669 670 671 672 673 674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714
Grenier, A. et al. Oncostatin M production by blood and alveolar neutrophils during acute lung injury. Lab Invest 81, 133-141 (2001).
10.
Cross, A., Edwards, S.W., Bucknall, R.C. & Moots, R.J. Secretion of oncostatin M by neutrophils in rheumatoid arthritis. Arthritis Rheum 50, 1430-1436 (2004).
11.
Queen, M.M., Ryan, R.E., Holzer, R.G., Keller-Peck, C.R. & Jorcyk, C.L. Breast cancer cells stimulate neutrophils to produce oncostatin M: potential implications for tumor progression. Cancer Res 65, 8896-8904 (2005).
ACCEPTED MANUSCRIPT
Simpson, J.L., Baines, K.J., Boyle, M.J., Scott, R.J. & Gibson, P.G. Oncostatin M (OSM) is increased in asthma with incompletely reversible airflow obstruction. Experimental lung research 35, 781-794 (2009).
13.
Su, C.M. et al. Osteopontin Promotes Oncostatin M Production in Human Osteoblasts: Implication of Rheumatoid Arthritis Therapy. Journal of immunology 195, 3355-3364 (2015).
14.
Kastl, S.P. et al. In human macrophages the complement component C5a induces the expression of oncostatin M via AP-1 activation. Arterioscler Thromb Vasc Biol 28, 498-503 (2008).
15.
Repovic, P. & Benveniste, E.N. Prostaglandin E2 is a novel inducer of oncostatin-M expression in macrophages and microglia. The Journal of neuroscience : the official journal of the Society for Neuroscience 22, 53345343 (2002).
16.
Miller, M. et al. Fstl1 Promotes Asthmatic Airway Remodeling by Inducing Oncostatin M. Journal of immunology 195, 3546-3556 (2015).
17.
Kastl, S.P. et al. Thrombin induces the expression of oncostatin M via AP-1 activation in human macrophages: a link between coagulation and inflammation. Blood 114, 2812-2818 (2009).
18.
Elbjeirami, W.M., Donnachie, E.M., Burns, A.R. & Smith, C.W. Endotheliumderived GM-CSF influences expression of oncostatin M. Am J Physiol Cell Physiol 301, C947-953 (2011).
19.
Ohno, I. et al. Granulocyte/macrophage colony-stimulating factor (GM-CSF) gene expression by eosinophils in nasal polyposis. American journal of respiratory cell and molecular biology 5, 505-510 (1991).
20.
Kato, M. et al. Production of granulocyte/macrophage colony-stimulating factor in human airways during allergen-induced late-phase reactions in atopic subjects. Lymphokine Cytokine Res 11, 287-292 (1992).
EP
TE D
M AN U
SC
RI PT
12.
AC C
715 716 717 718 719 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 744 745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760
21.
Miller, M. et al. Segmental allergen challenge increases levels of airway follistatin-like 1 in patients with asthma. The Journal of allergy and clinical immunology (2016).
22.
Hamilos, D.L. et al. Chronic hyperplastic sinusitis: association of tissue eosinophilia with mRNA expression of granulocyte-macrophage colonystimulating factor and interleukin-3. The Journal of allergy and clinical immunology 92, 39-48 (1993).
ACCEPTED MANUSCRIPT
Fridlender, Z.G. et al. Polarization of tumor-associated neutrophil phenotype by TGF-beta: "N1" versus "N2" TAN. Cancer Cell 16, 183-194 (2009).
24.
Mishalian, I. et al. Tumor-associated neutrophils (TAN) develop protumorigenic properties during tumor progression. Cancer Immunol Immunother 62, 1745-1756 (2013).
25.
Ma, Y. et al. Temporal neutrophil polarization following myocardial infarction. Cardiovasc Res 110, 51-61 (2016).
26.
Cuartero, M.I. et al. N2 neutrophils, novel players in brain inflammation after stroke: modulation by the PPARgamma agonist rosiglitazone. Stroke 44, 3498-3508 (2013).
27.
Raes, G. et al. Arginase-1 and Ym1 are markers for murine, but not human, alternatively activated myeloid cells. Journal of immunology 174, 6561; author reply 6561-6562 (2005).
28.
Meltzer, E.O. et al. Rhinosinusitis: establishing definitions for clinical research and patient care. The Journal of allergy and clinical immunology 114, 155-212 (2004).
29.
Liu, M.C. et al. Evidence for elevated levels of histamine, prostaglandin D2, and other bronchoconstricting prostaglandins in the airways of subjects with mild asthma. The American review of respiratory disease 142, 126-132 (1990).
30.
Kato, A., Xiao, H., Chustz, R.T., Liu, M.C. & Schleimer, R.P. Local release of B cell-activating factor of the TNF family after segmental allergen challenge of allergic subjects. The Journal of allergy and clinical immunology 123, 369-375 (2009).
31.
Pepe, C. et al. Differences in airway remodeling between subjects with severe and moderate asthma. The Journal of allergy and clinical immunology 116, 544-549 (2005).
EP
TE D
M AN U
SC
RI PT
23.
AC C
761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780 781 782 783 784 785 786 787 788 789 790 791 792 793 794 795 796 797 798 799 800 801 802 803 804
32.
Woodruff, P.G. et al. Relationship between airway inflammation, hyperresponsiveness, and obstruction in asthma. The Journal of allergy and clinical immunology 108, 753-758 (2001).
33.
Fahy, J.V., Liu, J., Wong, H. & Boushey, H.A. Cellular and biochemical analysis of induced sputum from asthmatic and from healthy subjects. The American review of respiratory disease 147, 1126-1131 (1993).
ACCEPTED MANUSCRIPT
Kato, A. et al. Evidence of a role for B cell-activating factor of the TNF family in the pathogenesis of chronic rhinosinusitis with nasal polyps. The Journal of allergy and clinical immunology 121, 1385-1392, 1392 e1381-1382 (2008).
35.
Tieu, D.D. et al. Evidence for diminished levels of epithelial psoriasin and calprotectin in chronic rhinosinusitis. The Journal of allergy and clinical immunology 125, 667-675 (2010).
36.
Seshadri, S. et al. Reduced expression of antimicrobial PLUNC proteins in nasal polyp tissues of patients with chronic rhinosinusitis. Allergy 67, 920928 (2012).
37.
Kato, A. et al. Dexamethasone and FK506 inhibit expression of distinct subsets of chemokines in human mast cells. Journal of immunology 182, 7233-7243 (2009).
38.
Beigel, F. et al. Oncostatin M mediates STAT3-dependent intestinal epithelial restitution via increased cell proliferation, decreased apoptosis and upregulation of SERPIN family members. PloS one 9, e93498 (2014).
39.
Homma, T. et al. Involvement of Toll-like receptor 2 and epidermal growth factor receptor signaling in epithelial expression of airway remodeling factors. American journal of respiratory cell and molecular biology 52, 471481 (2015).
40.
Durand, V., Renaudineau, Y., Pers, J.O., Youinou, P. & Jamin, C. Cross-linking of human FcgammaRIIIb induces the production of granulocyte colonystimulating factor and granulocyte-macrophage colony-stimulating factor by polymorphonuclear neutrophils. Journal of immunology 167, 3996-4007 (2001).
41.
Akdis, C.A. et al. Endotypes and phenotypes of chronic rhinosinusitis: a PRACTALL document of the European Academy of Allergy and Clinical Immunology and the American Academy of Allergy, Asthma & Immunology. The Journal of allergy and clinical immunology 131, 1479-1490 (2013).
EP
TE D
M AN U
SC
RI PT
34.
AC C
805 806 807 808 809 810 811 812 813 814 815 816 817 818 819 820 821 822 823 824 825 826 827 828 829 830 831 832 833 834 835 836 837 838 839 840 841 842 843 844 845 846 847 848 849
42.
Van Zele, T. et al. Differentiation of chronic sinus diseases by measurement of inflammatory mediators. Allergy 61, 1280-1289 (2006).
43.
Stevens, W.W. et al. Cytokines in Chronic Rhinosinusitis. Role in Eosinophilia and Aspirin-exacerbated Respiratory Disease. American journal of respiratory and critical care medicine 192, 682-694 (2015).
44.
Engelhardt, E. et al. Chemokines IL-8, GROalpha, MCP-1, IP-10, and Mig are sequentially and differentially expressed during phase-specific infiltration of
ACCEPTED MANUSCRIPT
890 891 892
leukocyte subsets in human wound healing. The American journal of pathology 153, 1849-1860 (1998). Goren, I. et al. Oncostatin M expression is functionally connected to neutrophils in the early inflammatory phase of skin repair: implications for normal and diabetes-impaired wounds. The Journal of investigative dermatology 126, 628-637 (2006).
46.
Nightingale, J. et al. Oncostatin M, a cytokine released by activated mononuclear cells, induces epithelial cell-myofibroblast transdifferentiation via Jak/Stat pathway activation. Journal of the American Society of Nephrology : JASN 15, 21-32 (2004).
47.
Schleimer, R.P. Immunopathogenesis of Chronic Rhinosinusitis and Nasal Polyposis. Ann Rev Path (2016).
48.
Anwar, A.R., Moqbel, R., Walsh, G.M., Kay, A.B. & Wardlaw, A.J. Adhesion to fibronectin prolongs eosinophil survival. The Journal of experimental medicine 177, 839-843 (1993).
49.
Tan, B.K. et al. Evidence for intranasal antinuclear autoantibodies in patients with chronic rhinosinusitis with nasal polyps. The Journal of allergy and clinical immunology 128, 1198-1206 e1191 (2011).
50.
Scaffidi, A.K. et al. Oncostatin M stimulates proliferation, induces collagen production and inhibits apoptosis of human lung fibroblasts. British journal of pharmacology 136, 793-801 (2002).
51.
Molfino, N.A. et al. Phase 2, randomised placebo-controlled trial to evaluate the efficacy and safety of an anti-GM-CSF antibody (KB003) in patients with inadequately controlled asthma. BMJ Open 6, e007709 (2016).
EP
TE D
M AN U
SC
RI PT
45.
AC C
850 851 852 853 854 855 856 857 858 859 860 861 862 863 864 865 866 867 868 869 870 871 872 873 874 875 876 877 878 879 880 881 882 883 884 885 886 887 888 889
ACCEPTED MANUSCRIPT
Figure 1: Oncostatin M was expressed in neutrophils.
894
Nasal polyp sections were stained for OSM in green and various cell type specific
895
markers as indicated in red; two representative examples from separate patients are
896
shown for each staining strategy. OSM did not co-localize with macrophages (A, B)
897
(n=3) or eosinophils (C,D) (n=5) and had minimal co-localization with mast cells (E,F)
898
(n=6). OSM did co-localize with neutrophils (G,H) (n=10).
SC
899
RI PT
893
Figure 2: Flow cytometric analysis showed that neutrophils are a major source of
901
OSM producing cells in nasal polyps.
902
(A) Within the CD45 gate, 5.1± 1.7% of cells were OSM+. (B) Within the OSM+ gate,
903
80.4±5% of cells were neutrophils (CD16+Sig8-) and 14.1±5% of cells were either
904
eosinophils, mast cells or basophils (CD16+Sig8+). (C) Quantification of the percentage
905
of OSM+ cells within the CD45+ gate, and the percentage of neutrophils (CD16+Sig8-),
906
and CD16+Sig8+ cells within the OSM+ gate, n=9. (D) In matched blood and nasal
907
polyps, .60± .36% of CD45+ cells in the blood were OSM+ while 6.3± 2.9% of CD45+
908
cells in nasal polyps were OSM+ (n=5, p< .05, Mann-Whitney U test), suggesting that
909
OSM was locally induced, n=5.
EP
TE D
M AN U
900
910
Figure 3: Flow cytometric analysis showed that mast cells were a minor source of
912
OSM producing cells in nasal polyps.
913
(A) Representative flow cytometric plot of the OSM+ gate. (B) Representative flow
914
cytometric plots of the c-kit-FcεRI- gate. (C) Quantification of the relative representation
915
of different cell types among the OSM+ cells. Mast cells (MC) were 15.7±9.6%,
916
basophils (Baso) were 3.7±1.5%, eosinophils (Eo) were 2.3±.9% and neutrophils (PMN)
917
were 73.3±10.6% within the OSM+ gate, n=4. OSM+ cells were 4.6±1.9% of the CD45+
918
cells.
AC C
911
ACCEPTED MANUSCRIPT
Figure 4: GM-CSF was elevated in nasal polyps and induced neutrophil derived
920
OSM.
921
(A) Blood neutrophils were stimulated with the indicated concentrations of GM-CSF,
922
FSTL1 or both and levels of OSM protein in the culture supernatant were measured;
923
(n=7-13, * p< .05, * p< .001, Kruskal-Wallis test). (B) Fully differentiated NHBE cells
924
were either unstimulated, or stimulated with HKSA, and levels of GM-CSF protein in the
925
cell culture supernatants were measured. HKSA treated NHBE released more GM-CSF,
926
21.3± 6.9pg/mL, than control NHBE, .87±.87 (n=4, p< .05, Mann-Whitney U test). (C)
927
GM-CSF protein, 2.6±.93pg/mg total protein, was elevated in nasal polyps compared to
928
control UT, .68±.42pg/mg total protein, (n=16-24, p< 05, Mann-Whitney U test). (D)
929
FSTL1 protein, .60±.47ng/mL, was elevated in nasal polyps compared to control UT,
930
.80±.33, (n=15-23, p=.31, Mann-Whitney U test). (E) OSM (alexa 488), GM-CSF (alexa
931
568) and neutrophil elastase (alexa 647) colocalized in nasal polyps (n=4).
M AN U
SC
RI PT
919
TE D
932
Figure 5: OSM producing neutrophils did not express a classical N1 phenotype.
934
(A) OSM+ neutrophils contained a distinct population of Arg1+MPOhi cells, representative
935
data. (B) 3.7±1.3% of cells in the CD45 gate were OSM+; within the OSM+ gate, 56.0±
936
8.2% of the cells were CD16+IL-5R- neutrophils. Within the neutrophil gate 72.56± 11.9%
937
of the cells were Arg1+MPOhi, and 22.1± 8.0% were Arg1-MPOlo, n=5. (C) Blood
938
neutrophils were either left untreated, or treated under N1 polarizing conditions
939
(LPS/IFNγ), N2 polarizing conditions (IL-4/IL-13) or with GM-CSF. GM-CSF treated
940
neutrophils secreted elevated levels of OSM into the cell culture supernatants, while N1
941
and N2 polarizing conditions did not induce OSM, (n=4-7, p< .01, Kruskal-Wallis test).
942
(D) Levels of MRC1 mRNA were elevated in neutrophils stimulated with GM-CSF either
943
alone or together with FSTL1, (n=3-7, p< .05, Kruskal-Wallis test).
944
AC C
EP
933
ACCEPTED MANUSCRIPT
Figure 6: OSM was elevated in asthmatic patients
946
(A) Bronchial biopsies from control patients, moderate asthmatics and severe asthmatics
947
were stained for OSM in green and neutrophil elastase in red, n=4-6. (B) Counts were
948
obtained of neutrophils and OSM+ cells; none of the control biopsies had any OSM+
949
cells, and of the OSM+ cells 35± 21.8% were neutrophils in moderate asthmatics, and
950
52.1± 15.9% were neutrophils in severe asthmatics. (C) Levels of OSM in the sputum of
951
asthmatic patients, 26.9± 9.5pg were elevated compared to control patients 3.9± 2.6pg,
952
(n=11-12, p< .05, Mann-Whitney U test).
M AN U
953
SC
RI PT
945
Figure 7: Proposed mechanism of OSM mediated barrier dysfunction in mucosal
955
disease
956
Under normal circumstances, when epithelium is injured, neutrophils are recruited to the
957
site of injury and potentially, transiently make OSM to promote the early stages of repair.
958
Due to transient OSM expression, once the epithelial cells become contact inhibited they
959
are able to enter the later stages of repair and redifferentiate back into functional
960
epithelium. However, under pathogenic conditions we hypothesize that neutrophils are
961
recruited to the injury site, and once at the site of injury the neutrophils are converted
962
into an alternative phenotype, potentially N2, that makes both OSM and GM-CSF. The
963
GM-CSF alone is sufficient to induce production of OSM in neutrophils, and will also
964
contribute to long-term survival of OSM producing neutrophils that may prevent late
965
stage repair, causing a long-term state of barrier dysfunction.
967 968 969
EP
AC C
966
TE D
954
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
Table E1: Patient Demographics
Age (y), median (range)
n = 33 (15F/18M) 45.5 (2273)
n = 24 (14F/10M)
CRSwNP n = 89 (36F/53M)
48 (19-75)
48 (18-69)
Y 5 1
N 15 32
U 13 0
Y 10 4
N 10 20
(p=.0142) (p=.2659)
1 3
30 18
2 1
1 0
23 24
Immunofluorescence Age (y), median (range) Neutrophil culture Age (y), median (range)
n = 13 (3F/10M) 37 (22-63) n = 20 (12F/8M) 58 (25-73)
AC C
EP
TE D
Protein lysates Age (y), median (range)
CRSwNP polyp
U 4 0
Y 53 43
N 29 45
U 7 1
0 0
18 10
68 78
2 1
SC
Chisquare (p<.0001) (p<.0001)
M AN U
Atopy Asthma Peri operative systemic steroids Antibiotics Methodology used Flow cytometry Age (y), median (range)
CRSsNP
RI PT
Total no. of subjects
Control
n = 6 (3F/3M) 53 (25-75)
n = 18 (11F/7M) 46 (19-67)
n=16(8F/8M) 44(18-69) n = 21 (8F/13M) 45 (25-66)
n = 8 (2F/6M) 36 (28-65) n = 20 (7F/13M) 48 (28-69)
n = 24 (11F/13M) 54 (25-69)
Dilution 1:10 1:40 1:40 1:10 A10 1:20 MHE-18 1:100 104D2 1:50 17022 1:10 HI30 1:40 3G8 1:40 26815 1:20 MPO455-8E6 1:20 658922 1:20
SC
Clone 17022 HI30 3G8
AC C
EP
TE D
M AN U
Table E2: Flow Cytometry Antibodies Antibody Figure 2 and 3: anti-human OSM-PE anti-human CD45-V450 anti-human CD16-APC-H7 anti-human Siglec 8-Alexa 647 anti-human EMR1-Alexa 647 anti-human FCεR1-Pe-Cy7 anti-human c-kit-Alexa 488 Figure 5: anti-human OSM-PE anti-human CD45-V450 anti-human CD16-APC-H7 anti-human IL-5R-APC anti-human MPO-FITC anti-human Arg1-Alexa 700
RI PT
ACCEPTED MANUSCRIPT
Manufacturer R&D systems, Minneapolis, MN BD Biosciences, San Jose, CA BD Biosciences, San Jose, CA Gift from Dr. Bruce Bochner AbD Serotec, Raleigh, NC BioLegend, San Diego, CA Biolegend, San Diego, CA R&D systems, Minneapolis, MN BD Biosciences, San Jose, CA BD Biosciences, San Jose, CA R&D systems, Minneapolis, MN ebioscience, San Diego, CA R&D systems, Minneapolis, MN
RI PT
ACCEPTED MANUSCRIPT
5
2
7
1
10
9
6 6
0 4
2 6
2 2
39
19
25
9
0 20 7 8 15 23 11 22 0
%PMN PMN/mm %OSM+ that are basement OSM+ membrane PMN
M AN U
3
TE D
20
60
15
35.27
10
14.29
14.69
0 66.67
0 66.67
9.85 8.86
25
6
76
48.72
52.25
9
0
100
36
17.93
0
0
0
0
0
0
10
10
0
100
50
31.97
2 0 4 0 0 0 0
2 0 1 0 0 0 0
0 0 75 0 0 0 0
0 0 20 0 0 0 0
8.41 13.24 22.44 43.08 16.41 39.15 0
AC C
Patient abb 0120908 Y abb 0151108 Y abb 0308207 Y abb0235206 Y abb 0120709 Y abb 0305807 Y Moderate abb asthma 0167606 N abb 0144408 Y abb 0240008 Y abb0200412 N abb0164406 Y Control abb349911 N abb0150705 N abb0160706 N abb0165706 N
EP
Patient groups Severe asthma
OSM+ nonTotal OSM+ OSM PMN PMN OSM+ PMN Y/N count count count count
SC
Table E3: Bronchial biopsy cell counts
0 0 3 0 0 0 0
ACCEPTED MANUSCRIPT
Figure E1: Control images from OSM staining with CD68 (A), ECP (B), tryptase (C), and elastase (D). (E) OSM expression levels correlated with the granulocyte marker FCGR3 and (F) with the
RI PT
neutrophil specific protease CTSG in the microarray dataset. (G) FCGR3 correlated with CTSG in the microarray. (H) OSM protein correlated with elastase protein in lysates from both nasal
SC
polyps and uncinates from CRS patients and controls.
Figure E2:
M AN U
Gating strategy for identification of OSM+ cells. Cells were first gated on forward and side scatter, and doublets (not shown) and dead cells were gated out. Cells were then gated on the CD45+ population, and then subsequently gated on the OSM+ population. The OSM+ population was then plotted based on CD16 and siglec 8 expression, and the CD16+Sig8- and CD16+Sig8+
Figure E3:
TE D
populations were quantified.
Contribution of mast cells, eosinophils and basophils to the OSM+ cell population. Cells were
EP
first gated on forward and side scatter, and doublets (not shown) and dead cells were gated out. Cells were then gated on the CD45+ population, and then gated on the OSM+ population. The
AC C
OSM+ population was then plotted on c-kit and FcεRI expression, and the mast cell, ckit+FcεRI+, and basophil c-kit-FcεRI+, populations were quantified. The c-kit-FcεRI- population was then plotted on CD16 and either EMR1 or Siglec 8 to discriminate neutrophils (CD16+EMR1/Siglec8-) from eosinophils, (CD16+EMR1/Siglec 8+).
Figure E4: (A) Blood neutrophils were isolated and either left unstimulated, or were stimulated with GMCSF, FSTL1 or both. There was a trend toward both GM-CSF and FSTL1 to elevate OSM
ACCEPTED MANUSCRIPT
mRNA compared to unstimulated controls. (B) Neutrophils were either left unstimulated or stimulated with 25ng/mL of GM-CSF or the indicated concentrations of IL-25, IL-33, TSLP or leukotriene C4 for 20h (n=5-6). Only GM-CSF was sufficient to induce OSM. (C) E-selectin and
RI PT
ICAM ligation did not induce neutrophil derived OSM (n=5-6). (D) OSM (alexa 488) and GMCSF (alexa 568) colocalized in nasal polyps, (n=4). (E) GM-CSF (alexa 568) and neutrophil
SC
elastase (alexa 647) colocalized in nasal polyps, (n=4).
Figure E5:
M AN U
Identification of putative N1 and N2 cells among the OSM+ neutrophils. (A) Cells were first gated on forward and side scatter, and doublets and dead cells were gated out. Cells were then gated on the CD45+ population, and then gated on the OSM+ population. The OSM+ population was then plotted on CD16 and IL-5R to discriminate neutrophils (CD16+IL-5R-) and eosinophils (CD16+IL-5R+). Cells were then gated on the neutrophil population and plotted on MPO and
TE D
Arg1, and the Arg1+MPOhi, and Arg1+MPOlo populations were quantified. (B) Levels of ARG1 mRNA were not elevated in neutrophils stimulated with GM-CSF either alone or together with
Figure E6:
EP
FSTL1 compared to control.
AC C
Bronchial biopsy sections from healthy controls and asthma patients were stained for OSM and neutrophil elastase (A): none of the control biopsies had any OSM+ cells, 60% of moderate asthmatic biopsies and 100% of severe asthmatic biopsies had OSM+ cells. (B) Of the neutrophils counted, 0±0% were OSM+ in controls, 14.0±9.8% were positive in moderate asthmatics, and 30.1±10.1% were positive in severe asthmatics. (C) No differences were seen in the number of neutrophils/mm of basement membrane. Levels of OSM in BAL from segmental allergen challenge of allergic asthmatics correlated with (D) total neutrophil count, (E)
ACCEPTED MANUSCRIPT
total lymphocyte count, and (H) total cell count, but not with (F) total eosinophil count or (G) total
AC C
EP
TE D
M AN U
SC
RI PT
macrophage count.
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT