Features of the bronchial bacterial microbiome associated with atopy, asthma, and responsiveness to inhaled corticosteroid treatment

Features of the bronchial bacterial microbiome associated with atopy, asthma, and responsiveness to inhaled corticosteroid treatment

Accepted Manuscript Features of the bronchial bacterial microbiome associated with atopy, asthma and responsiveness to inhaled corticosteroid treatmen...

3MB Sizes 0 Downloads 98 Views

Accepted Manuscript Features of the bronchial bacterial microbiome associated with atopy, asthma and responsiveness to inhaled corticosteroid treatment Juliana Durack, PhD, Susan V. Lynch, PhD, Snehal Nariya, BS, Nirav R. Bhakta, MD, PhD, Avraham Beigelman, MD, Mario Castro, MD, Anne-Marie Dyer, MS, Elliot Israel, MD, Monica Kraft, MD, Richard J. Martin, MD, David T. Mauger, PhD, Sharon R. Rosenberg, MD, MS, Tonya Sharp-King, PhD, Steven R. White, MD, Prescott G. Woodruff, MD, MPH, Pedro C. Avila, MD, MPH, Loren C. Denlinger, MD, PhD, Fernando Holguin, MD, Stephen C. Lazarus, MD, Njira Lugogo, MD, Wendy C. Moore, MD, Stephen P. Peters, MD, Loretta Que, MD, Lewis J. Smith, MD, Christine A. Sorkness, PharmD, Michael Wechsler, MD, MMSc, Sally E. Wenzel, MD, Homer A. Boushey, MD, Yvonne J. Huang, MD PII:

S0091-6749(16)31283-0

DOI:

10.1016/j.jaci.2016.08.055

Reference:

YMAI 12438

To appear in:

Journal of Allergy and Clinical Immunology

Received Date: 21 March 2016 Revised Date:

2 August 2016

Accepted Date: 12 August 2016

Please cite this article as: Durack J, Lynch SV, Nariya S, Bhakta NR, Beigelman A, Castro M, Dyer AM, Israel E, Kraft M, Martin RJ, Mauger DT, Rosenberg SR, Sharp-King T, White SR, Woodruff PG, Avila PC, Denlinger LC, Holguin F, Lazarus SC, Lugogo N, Moore WC, Peters SP, Que L, Smith LJ, Sorkness CA, Wechsler M, Wenzel SE, Boushey HA, Huang YJ, for the National Heart, Lung and Blood Institute’s “AsthmaNet”, Features of the bronchial bacterial microbiome associated with atopy, asthma and responsiveness to inhaled corticosteroid treatment, Journal of Allergy and Clinical Immunology (2016), doi: 10.1016/j.jaci.2016.08.055. 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.

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIPT

1

Features of the bronchial bacterial microbiome associated with atopy, asthma

2

and responsiveness to inhaled corticosteroid treatment.

3 1

2

3

8

9

6

RI PT

Avraham Beigelman, MD4., Mario Castro, MD,4,5., Anne-Marie Dyer, MS6., Elliot Israel, MD7., Monica Kraft, MD ., Richard J. Martin, MD ., David T. Mauger, PhD ., Sharon R. Rosenberg, MD, 10

6

11

3

MS ., Tonya Sharp-King, PhD ., Steven R. White, MD ., Prescott G. Woodruff, MD, MPH ., 10

12

13

Pedro C. Avila, MD, MPH ., Loren C. Denlinger, MD, PhD ., Fernando Holguin, MD ., Stephen 3

14

15

15

C. Lazarus, MD ., Njira Lugogo, MD ., Wendy C. Moore, MD ., Stephen P. Peters, MD ., 14

10

12

Loretta Que, MD ., Lewis J. Smith, MD ., Christine A. Sorkness, PharmD ., Michael Wechsler, 13

3

SC

9

2

MD, MMSc ., Sally E. Wenzel, MD ., Homer A. Boushey, MD ., and Yvonne J. Huang, MD ., for the National Heart, Lung and Blood Institute’s “AsthmaNet”.

University of California, Dept. of Medicine, Division of Gastroenterology, San Francisco, CA; 2 University of Michigan, Division of Pulmonary & Critical Care Medicine, Ann Arbor, MI; 3 University of California, Dept. of Medicine, San Francisco, CA; 4 Washington University School of Medicine, Division of Pediatrics, St Louis, MO; 5 Washington University School of Medicine, Division of Pulmonary and Critical Care Medicine, St Louis, MO; 6 Penn State University, Department of Public Health Sciences, Hershey, PA; 7 Brigham & Women's Hospital, Dept. of Medicine, Boston, MA; 8University of Arizona, Health Sciences, Tucson, AZ; 9 National Jewish Hospital, Dept. of Medicine, Denver, CO; 10 Northwestern University, Dept. of Medicine, Chicago, IL; 11 University of Chicago, Dept. of Medicine, Chicago, IL; 12 University of Wisconsin-Madison, Dept. of Medicine, Madison, WI; 13 The University of Pittsburgh Asthma Institute at UPMC/UPSOM, Pittsburgh, PA; 14 Duke University School of Medicine, Duke Asthma, Allergy & Airway Center, Durham, NC; 15 Wake Forest School of Medicine, Winston-Salem, NC;

M AN U

1

TE D

Corresponding author: Homer A. Boushey,

Professor of Medicine, Div. of Pulmonary/Critical Care Medicine University of California, San Francisco 505 Parnassus Avenue M1292, Box 0130 San Francisco, CA 94143 Phone: 415-476-8019 Fax: 415-502-6235 Email: [email protected]

EP

36

1

Juliana Durack, PhD ., Susan V. Lynch, PhD ., Snehal Nariya, BS ., Nirav R. Bhakta, MD, PhD .,

Running title: Bronchial microbiome in asthma, atopy, and health.

AC C

4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35

1

ACCEPTED MANUSCRIPT

37

Abstract

38 Background: Compositional differences in bronchial bacterial microbiota have been

40

associated with asthma, but it remains unclear whether the findings are attributable to

41

asthma, to aeroallergen sensitization or to inhaled corticosteroid treatment.

RI PT

39

42

Objectives: To compare the bronchial bacterial microbiota in adults with steroid-naive

44

atopic asthma (AA), with atopy but no asthma (ANA), and non-atopic healthy subjects

45

(HC), and determine relationships of bronchial microbiota to phenotypic features of

46

asthma.

SC

43

47

Methods: Bacterial communities in protected bronchial brushings from 42 AA, 21 ANA,

49

and 21 HC subjects were profiled by 16S rRNA gene sequencing. Bacterial composition

50

and community-level functions inferred from sequence profiles were analyzed for

51

between-group differences. Associations with clinical and inflammatory variables were

52

examined, including markers of type 2-related inflammation and change in airway hyper-

53

responsiveness following six weeks of fluticasone treatment.

M AN U

48

54

Results: The bronchial microbiome differed significantly among the three groups.

56

Asthmatic subjects were uniquely enriched in members of the Haemophilus, Neisseria,

57

Fusobacterium, Porphyromonas and Sphingomonodaceae, and depleted in members of

58

the Mogibacteriaceae and Lactobacillales. Asthma-associated differences in predicted

59

bacterial functions included involvement of amino acid and short-chain fatty acid

60

metabolism pathways. Subjects with type 2-high asthma harbored significantly lower

61

bronchial bacterial burden. Distinct changes in specific microbiota members were seen

62

following fluticasone treatment. Steroid-responsiveness was linked to differences in

63

baseline compositional and functional features of the bacterial microbiome.

EP

AC C

64

TE D

55

65

Conclusion: Even in mild steroid-naive asthma subjects, differences in the bronchial

66

microbiome are associated with immunologic and clinical features of the disease. The

67

specific differences identified suggest possible microbiome targets for future approaches

68

to asthma treatment or prevention.

2

ACCEPTED MANUSCRIPT

70

Key messages: •

71 72

The bronchial bacterial microbiota of both mild atopic-asthma (steroid-naïve) and atopy-alone differ from that of healthy controls and also differ from each other.



Asthma is associated with enrichment in members of the Haemophilus,

RI PT

69

73

Neisseria, Fusobacterium, Porphyromonas and Sphingomonodaceae and with

74

depletion of Lactobacillus. •

The T2-high asthma phenotype is associated with low bronchial bacterial burden.

76



ICS-response is linked to a distinct bacterial community composition and functional profile prior to steroid exposure.

78

M AN U

77

SC

75

Capsule summary: Mild atopic asthma is associated with distinct differences in the

80

composition and inferred functional capacities of bronchial bacterial microbiota, which

81

further associate with type 2-low airway inflammation and with responsiveness to inhaled

82

corticosteroid treatment.

TE D

79

83

Key words: Asthma, atopy, microbiome, corticosteroids, 16S ribosomal RNA, bacteria,

85

Th2 inflammation, Three-gene mean, metabolic pathways, SCFAs.

86

EP

84

Abbreviations:

88 89 90 91 92 93 94 95 96 97 98 99

AA – Atopic asthmatic subjects ANA – Atopic non-asthmatic subjects HC – Healthy controls ICS – Inhaled corticosteroid ACQ – Asthma Control Questionnaire OW – Oral wash BB – Bronchial brush OTU – Operational taxonomic unit SCFAs – Short chain fatty acids TGM – Three-gene mean PICRUSt - Phylogenetic Reconstruction of Unobserved States KEGG - Kyoto Encyclopedia of Genes and Genomes

AC C

87

3

ACCEPTED MANUSCRIPT

Introduction:

101

Recent culture-independent studies have documented that the composition of

102

commensal lower respiratory tract bacteria (microbiota) differs between asthmatic and

103

healthy adults (1-6). Additionally, phenotypic features of asthma, such as measures of

104

airway hyper-responsiveness, asthma control, and transcriptional response to steroids,

105

correlate with patterns of bronchial microbiota composition (4, 5). Though different

106

studies have reported asthma-associated enrichment (higher relative abundance) of

107

different taxa [i.e. bacterial-derived 16S ribosomal RNA gene sequences that exhibit an

108

operator-defined level of sequence homology (typically 97%)], enrichment in members of

109

the phylum Proteobacteria is a repeating signature. Asthmatic subjects in most previous

110

studies were treated with inhaled corticosteroids (ICS), casting some uncertainty on

111

whether the findings reflect the effects of ICS treatment or of asthma itself. Similarly,

112

many asthmatic patients are atopic (7, 8), raising the question as to whether asthma-

113

associated differences in respiratory microbiota are related to underlying atopy, itself

114

associated with altered mucosal immune function (9, 10). Collectively, these

115

considerations indicate a need to elucidate differences in bronchial microbiota

116

associated with asthma versus atopy, and with important phenotypic features of this

117

disease, such as the level of T2-type inflammation and responsiveness to ICS treatment.

SC

M AN U

TE D

118

RI PT

100

Accordingly, we compared the bronchial bacterial microbiome among adults with mild

120

steroid-naive atopic asthma, with atopy without asthma, and healthy non-atopic non-

121

asthmatic controls. We hypothesized that specific compositional and functional

122

differences in bronchial microbiota are associated with asthma and with distinguishing

123

phenotypic features of the disease, including evidence of Th2 inflammation and

124

responsiveness to ICS treatment.

AC C

125

EP

119

4

ACCEPTED MANUSCRIPT

Methods:

127

Study Population and Sample Collection.

128

This study was conducted at nine sites in the NHLBI AsthmaNet, using a standardized

129

bronchoscopy protocol for sample collection. Of 186 adults screened, 84 subjects were

130

enrolled (Figure S1A-B): 42 atopic asthmatics (AA), 21 atopic non-asthmatics (ANA)

131

and 21 non-atopic healthy control subjects (HC). Atopy was defined by serologic

132

evidence (>0.35 kU/l) of sensitivity to ≥1 of 12 aeroallergens (specific IgE by

133

ImmunoCap; Thermo-Scientific; Table S1). Asthma was confirmed by airway hyper-

134

responsiveness (methacholine PC20 ≤8 mg/mL or FEV1 improvement ≥12% post-

135

albuterol). At enrollment, asthmatics had been clinically stable for three months, and had

136

an Asthma Control Questionnaire (ACQ) score of <1.5 (11) without the use of a

137

controller medication. Exclusion criteria included a history of smoking, respiratory

138

infection within six weeks or antibiotic use within 3 months of enrollment

139

(Supplementary Methods).

140

M AN U

SC

RI PT

126

Samples processed for microbiota analysis included oral wash (OW) (12), a saline flush

142

(10 mL) of the bronchoscope suction channel (“scope-flush”) and protected bronchial

143

brushings (BB; Figure S1C; Supplemental Methods). AA were further randomized in a

144

2:1 ratio to treatment with inhaled fluticasone propionate (250 mcg, GlaxoSmithKline) or

145

placebo twice daily for six weeks and re-assessed post-treatment. Each subject signed

146

informed consent approved by their center’s IRB; an NHLBI-appointed Data Safety

147

Monitoring Board (DSMB) oversaw the study conduct.

TE D

141

EP

148

Nucleic acid extraction and quantitation of 16S rRNA gene copy number

150

Nucleic acids from OW, and 3 BB were extracted as previously described (5, 12) using a

151

modified bead-beating protocol and the AllPrep kit (Qiagen). 16S rRNA gene copy

152

number was assessed by quantitative PCR using universal primers (Supplemental

153

Methods).

154

AC C

149

155

16S rRNA-based sequencing and raw data processing.

156

For BB and OW samples, variable region 4 (V4) of the 16S rRNA gene was amplified

157

using the primer combination 515F/806R (13, 14) and sequenced on the Illumina MiSeq

158

platform. Using chimera-checked and quality-filtered sequence reads (Supplemental

159

Methods), a 97% sequence homology cut-off was used to define bacterial taxa (also

5

ACCEPTED MANUSCRIPT

160

referred to as operational taxonomic units; OTUs) and classified using the Greengenes

161

database (15). A phylogenetic tree was built using FastTree (16) and used to compute

162

Faith’s Phylogenetic Diversity (17) of samples using an OTU table multiply-rarefied to

163

52,317 sequences per sample (Supplemental Methods).

RI PT

164 “Three-gene mean” bronchial epithelial signature of type 2 inflammation.

166

Expression levels of three bronchial epithelial genes (CLCA1, SERPINB2 and POSTN)

167

previously shown to be induced by IL-13 (18, 19), were measured using RNA extracted

168

in parallel with DNA in this study to calculate the “three-gene mean” (TGM) score for

169

each participant (20). Type 2 (T2) – high asthma was defined by TGM scores ≥1.117

170

(two standard deviations above the average TGM-score in HC), confirmed by

171

unsupervised cluster and principal component analysis.

M AN U

SC

165

172 Statistical analyses:

174

We applied principal coordinates analyses (PCoA) on an unweighted UniFrac distance

175

matrix and PERMANOVA (21) to identify the determinants of variation in bacterial

176

community beta-diversity. Linear mixed-effects model (LME) (22) was employed to

177

examine relationships between paired OW and BB microbiota; negative binomial (NB)

178

and zero-inflated negative binomial (ZINB) regression models (23-26) corrected for false

179

discovery (Benjamini-Hochberg, q-value <0.1) were used to identify OTUs differentially

180

abundant between subject groups and paired-samples, respectively. We applied

181

Phylogenetic Reconstruction of Unobserved States (PICRUSt) to predict functional

182

capacities of the microbiota (27), and NB regression to compare inferred functional

183

pathway predictions across groups. Procrustes analysis (13) was used to explore the

184

strength of relationships between paired samples in the AA group (Supplemental

185

Methods).

EP

AC C

186

TE D

173

6

ACCEPTED MANUSCRIPT

Results:

188

Study group characteristics.

189

AA had mild well-controlled disease, significantly higher serum total IgE and blood and

190

sputum eosinophil cell counts (Table 1, Figure S2A-C) than HC. Compared to the ANA,

191

the asthmatics had significantly higher serum IgE, were sensitized to more of the

192

aeroallergens tested (Table 1), were more likely to be sensitive to cat, dog, and mouse

193

(Table S1), and to report a history of allergic rhinitis and eczema (Table S2). However,

194

they did not exhibit significant differences in environmental exposures as assessed by

195

questionnaire (Table S3).

RI PT

187

SC

196

Bacterial microbiota in bronchial brushings are compositionally distinct from oral

198

wash.

199

To evaluate mucosa-associated bronchial microbiota, we focused on protected BB.

200

Sequence-based bacterial community analysis could be performed in the same

201

proportion (67%) of samples collected in each of the three groups (Figure S1D-E).

202

Samples that could not be sequenced had lower bacterial burden as indicated from 16S

203

rRNA (p<0.0001; Figure S2D), despite recovery of similar mammalian cell burden as

204

assessed by quantifying β-actin copy number (Supplemental Methods). Subjects with

205

insufficient 16S rRNA amplicon for microbiota profiling were younger [median 28 (22-37)

206

vs. 34 yrs. (25-44); p=0.03] but did not differ in any other characteristic measured.

TE D

207

M AN U

197

Potential oral contamination of BB was evaluated by comparing a random subset of 30

209

BB-paired scope-flushes. Bacterial burden of scope-flushes for BB with sufficient 16S

210

rRNA for sequencing was indistinguishable from those unable to be sequenced (p>0.1;

211

Figure S2E). Additionally, PCoA analysis of bacterial community composition in paired

212

OW and BB samples showed these two niches to be compositionally distinct (LME β

213

=0.31, p<0.0001; Figure 1A), with BB samples exhibiting lower phylogenetic diversity

214

(Faith’s index; p<0.0001; Figure 1B).

AC C

215

EP

208

216

Features of the bronchial bacterial microbiota and distinct combinations of

217

specific bacterial taxa are associated with atopic asthma or atopy alone.

218

Alpha-diversity indices, such as richness (number of observed taxa), Shannon’s

219

diversity, and evenness, as well as bacterial burden did not differ amongst the groups

220

(Figure S2F-I). However, phylogenetic diversity (Faith’s index) tended to be higher in AA

7

ACCEPTED MANUSCRIPT

compared with HC (p=0.06; Figure 1C). Since this index weights phylogenetic

222

relatedness of the bacteria detected, this observation suggests that the bronchial

223

airways of AA harbor more phylogenetically diverse bacterial communities. Although

224

phylogenetic diversity in the ANA also appeared to be greater than in HC (Figure 1C),

225

the difference fell short of significance. Overall, inter-subject bacterial community

226

composition (beta-diversity) was highly heterogeneous across subjects (Figure S3A).

227

This was significantly related to bacterial richness independent of the study group

228

(Figure S3B-E), but was not associated with any of the clinical measures or

229

environmental exposures evaluated in this study (Tables 1 and S1-3). Compositional

230

variability in bronchial bacterial microbiota was significantly greater within the asthmatic

231

group (weighted UniFrac distance F-test p<0.001; Figure S3F), indicating a greater

232

degree of bacterial community heterogeneity in this group.

M AN U

SC

RI PT

221

233 234

Following these initial assessments for differences in overall bacterial community

235

composition, we conducted OTU-level analyses to determine if the relative abundance of

236

specific taxa differed significantly across groups; 76 taxa were found to do so in the AA

237

vs HC comparison (Table S4). Phylum-level differences in AA (Figures 2A and S4A)

238

included significant enrichment for members of the Bacteroidetes (Prevotella),

239

Fusobacteria

240

(Haemophilus and Neisseria), the latter previously associated with more severe, ICS-

241

requiring asthma (2-6, 28). Conversely, asthmatic subjects showed reduced relative

242

abundance

243

(Actinobacillus), and Firmicutes (Lactobacillus). ANA subjects also demonstrated

244

significant differences in the relative abundance of 100 taxa compared to HC (Table

245

S5B). The taxa most relatively enriched in ANA included members of the Proteobacteria

246

(Aggregatibacter,

247

(Corynebacterium) phyla (Figures 2B and S4B); those depleted included members of

248

the phylum Bacteroidetes (Porphyromonas and Prevotella).

TE D

members

of

the

(Actinomyces)

Fusobacteria

and

(Leptotrichia),

Proteobacteria

Proteobacteria

EP

of

Haemophilus),

AC C

249

(Fusobacterium), Actinobacteria

Firmicutes

(Granulicatella)

and

Actinobacteria

250

We noted that the ANA shared 26% and 29% of taxa that were also relatively enriched

251

or depleted in the AA, when compared to HC. Despite these similarities, OTU-level

252

analysis identified 103 taxa that differed significantly between the AA and ANA groups

253

(Table S6; Figures 2C and S4C). To pinpoint bacteria discretely associated with

254

asthma, we evaluated which taxa among those that distinguished AA from HC, also

8

ACCEPTED MANUSCRIPT

255

distinguished AA from ANA (Figure 3). By this approach, asthma-associated taxa

256

included members of the Haemophilus (OTU4406393), Fusobacterium (OTU4405869;

257

OTU2438396),

258

Sphingomonodaceae (OTU8331815); while taxa negatively associated with asthma

259

included members of the Mogibacteriaceae (OTU4335578) and Lactobacillales

260

(OTU4480189; OTU4469032). The same approach also identified taxa specific to atopy-

261

alone, which included enrichment in Aggregatibacter (OTU4432431), Corynebacterium

262

(OTU1015518; OTU495067) and Prevotella (OTU4372058; OTU134265) (Figure 3). Of

263

the taxa identified as specifically associated with either asthma or atopy-alone, a subset

264

exhibited strong, significant correlations with features of atopy (IgE, blood and sputum

265

eosinophil counts); these associations were distinct in the two atopic groups. For

266

example, specific taxa uniquely enriched in AA, belonging to Sphingomonodaceae and

267

Fusobacteria, were positively associated with all three markers of atopy, while of those

268

specifically enriched in ANA, Sharpea and Prevotella (OTU1052181; OTU134265)

269

correlated with IgE and blood eosinophil counts, respectively (Figure 3). These data

270

indicate that while taxonomic overlap exists between AA and ANA subjects, discrete

271

bacterial enrichments characterize these groups, a subset of which are associated with

272

biomarkers of atopic disease.

Porphyromonas

(OTU495451)

and

M AN U

SC

RI PT

(OTU1304),

TE D

273

Neisseria

We further explored whether predicted functions of the bronchial bacterial microbiome

275

differed among the three groups using PICRUSt (27), an algorithm that predicts bacterial

276

metagenomes in silico from 16S rRNA sequence. This analysis revealed significant

277

differences across groups (Figure 4). Predicted bacterial gene functions enriched in AA

278

included those involved in metabolism of amino acids and carbohydrates, especially of

279

short chain fatty acids (SCFAs) such as butanoate and propanoate (more commonly

280

known as butyrate and propionate, respectively). Also noteworthy was a relative

281

depletion in predicted bacterial functions involved in lipopolysaccharide biosynthesis

282

among asthma-associated bacterial communities.

AC C

283

EP

274

284

Type 2-high asthma is associated with reduced bronchial bacterial burden.

285

TGM scores were higher in AA (Figure 5A) and, consistent with prior reports (20, 29),

286

correlated positively with serum IgE (rSpearman=0.36, p<0.01), blood and sputum

287

eosinophil counts (rS=0.32 and rS=0.37, respectively, p≤0.01), and negatively with FEV1

288

(rS= –0.45, p<0.005) and PC20 (rS= –0.41, p<0.005). Ten of 40 AA had T2-high asthma

9

ACCEPTED MANUSCRIPT

and exhibited significantly higher ACQ scores, serum IgE, blood and sputum eosinophil

290

counts, and younger age, than T2-low asthmatics (Table S7). T2-high asthmatics

291

demonstrated significantly lower bronchial bacterial burden than T2-low asthmatics

292

(Figure 5C); bacterial burden also was negatively correlated with TGM scores across all

293

asthmatic subjects (rS= –0.43, p<0.01) independent of age (GLM p=0.03). Because of

294

their low bacterial burden BBs from only four T2-high asthmatics, could be sequenced

295

for profiling, an insufficient sample size to allow meaningful assessment of differences in

296

bacterial microbiota composition between T2-high and T2-low subjects.

RI PT

289

297

Inhaled corticosteroid-responsiveness is associated with distinct features of the

299

bronchial bacterial microbiota present before treatment.

300

We hypothesized that differences in baseline airway microbiota characteristics may be

301

associated with ICS-responsiveness, defined as ≥2-fold increase in PC20Mch after ICS-

302

treatment. Of the asthmatics included, 15 were classified as ICS-responders, 10 as non-

303

responders, and three were excluded as PC20 was not performed (Figure S5A-B).

304

Compared to non-responders, ICS-responders did not differ in PC20, serum IgE, blood

305

and sputum eosinophil counts, or bronchial burden at baseline but did have lower FEV1

306

and a trend toward a higher TGM score (p<0.08) (Table S8; Figure S5C), which

307

decreased following treatment (p<0.05; Figure S4D).

TE D

M AN U

SC

298

308

The baseline composition of profiled bronchial bacterial microbiota in ICS-responders

310

(n=10) differed from that in the non-responders (n=5; unweighted UniFrac PERMANOVA

311

R2=0.13, p=0.01), and was significantly more similar to that of HC (Figure 6A). Bacterial

312

families enriched at baseline in ICS non-responders included Microbacteriaceae,

313

Pasteurellaceae (e.g. asthma-associated Haemophilus OTU4406393) and several

314

others (Figure 6C and Table S9). Conversely, bacterial families enriched at baseline in

315

ICS-responders

316

Sphingomonodaceae. Compared with ICS-responders, the predicted functions of

317

bacterial communities of non-responders were enriched in xenobiotic biodegradation

318

pathways (Figure 6D and Table S10), implicating potential enhanced capacity for

319

synthetic chemical degradation.

AC C

EP

309

included

Streptococcaceae,

Fusobacteriaceae

and

320 321

Following six weeks of ICS vs. placebo treatment, we found no significant changes in

322

bacterial burden or phylogenetic diversity in the AA group (Figure S6A-B). Bacterial

10

ACCEPTED MANUSCRIPT

community analysis in paired samples before and after treatment was limited, as several

324

subjects did not have sufficient 16S rRNA to obtain sequence data at both time points.

325

Nonetheless, we reasoned that ICS exposure might have distinct effects on the

326

microbiotas of non-responders and ICS-responders (2) and explored ICS-induced

327

compositional changes in the latter, more predominant group. No significant differences

328

were seen in the magnitude of changes (beta-diversity assessed by unweighted Unifrac

329

distance) between paired samples (n=8 each in placebo or ICS-treated responders;

330

Figure S6C-E). At the taxon level, however, ICS treatment resulted in increased relative

331

abundance of Microbacteriaceae, Neisseria and Moraxella and depletion of a specific

332

Fusobacterium, which was not observed with the placebo treatment (Figure 6D, Tables

333

S11-12). We unfortunately could not analyze compositional changes in ICS-non-

334

responders, as only 2 subjects in this subgroup had sufficient 16S rRNA in both pre- and

335

post- treatment samples.

336

M AN U

SC

RI PT

323

Discussion:

338

Our findings show compositional and predicted functional differences in the bronchial

339

bacterial microbiomes of atopic asthmatic, atopic non-asthmatic, and healthy individuals.

340

An important implication of these findings is that control for allergic sensitization is

341

necessary in studies aimed at understanding differences in the respiratory microbiome

342

associated with asthma. Despite overlap in bacterial genera significantly associated with

343

both atopic groups, our study identified specific bacterial taxa whose relative enrichment

344

or depletion were discretely associated with asthma. Analyses based on metagenomic

345

inference further suggest that genes for pathways involved in the metabolism of short-

346

chain fatty acids and amino acids are enriched in the asthmatic bronchial microbiome.

347

We additionally observed that the bronchial microbiome among asthmatics at baseline

348

differed compositionally and functionally according to their responsiveness to treatment

349

with inhaled corticosteroids.

EP

AC C

350

TE D

337

351

Commonly reported microbial community metrics (e.g., richness, evenness, and burden)

352

did not differ significantly among our three groups. This is not unexpected due to the

353

broad characterization of microbial composition provided by such measures, the inter-

354

individual heterogeneity of microbiota composition found in all human niches, including

355

the lung (30), and the mild disease severity in our subjects. Considering the clinical

356

homogeneity of our asthmatic group, their heterogeneity in bronchial bacterial

11

ACCEPTED MANUSCRIPT

composition is striking. Moreover, this heterogeneity was predominantly observed

358

among T2-low asthma subjects, in whom bronchial bacterial burden was significantly

359

greater than in T2-high subjects. The trend towards higher bacterial phylogenetic

360

diversity in our mild asthmatic subjects suggests their lower airways are receptive to

361

colonization by a wider variety of bacteria. Although we could not pinpoint any specific

362

clinical features associated with this finding, additional contributing factors might include

363

differences in previous environmental exposures, in clearance of microorganisms, or in

364

other immune function parameters not identified here.

RI PT

357

365

Reasons for the low bronchial bacterial burden in T2-high asthma are unclear, but the

367

finding echoes a recent observation in severe asthma, of an inverse relationship

368

between bronchial bacterial burden and numbers of bronchial biopsy eosinophils (4). A

369

possible explanation is the bactericidal activity of more numerous airway eosinophils (31,

370

32) in T2-high airway inflammation. Given prior reports of inverse relationships between

371

bacterial and fungal richness (33, 34), the low bacterial burden in T2-high asthma could

372

also reflect increased fungal or bacteriophage burden. Indeed microbial interactions are

373

not limited to bacteria, as bacterial-viral interactions have been associated with asthma

374

exacerbations and risk for asthma development (35, 36). Such inter-kingdom

375

interactions (37) should be considered in future studies.

TE D

M AN U

SC

366

376

This study expands the list of bacterial groups previously associated with asthma, further

378

supporting the idea that alterations in microbial composition from a healthy state, is a

379

characteristic of asthma. Enrichment in certain Proteobacteria members (e.g.

380

Haemophilus and Neisseria) among our ICS-naïve asthmatics resembles prior findings

381

in ICS-using patients (2-6, 28). However, we found other taxa discretely associated with

382

asthma including Fusobacterium and Porphyromonas, two oral-associated anaerobes

383

capable of augmenting pathogenic behavior of opportunistic respiratory pathogens, such

384

as Pseudomonas aeruginosa (38). Additionally, both Fusobacterium (39) and

385

Haemophilus

386

Sphingomonodaceae represented another asthma-specific taxon whose abundance, like

387

that of Fusobacterium, correlated with sputum eosinophilia. This finding echoed the

388

reported

389

Sphingomonodaceae) in activating NKT-cells to produce T2-cytokines (41-43) and the

390

reported association of this bacterial family with bronchial reactivity (5).

AC C

EP

377

(40)

activity

induce

of

MUC5AC

expression

glycosphingolipids

(present

in

in

bronchial

the

cell

epithelial

cells.

membrane

of

12

ACCEPTED MANUSCRIPT

391 Asthma-related alterations in bronchial microbiota composition also involve relative

393

depletion in certain taxa, including members of the Lactobacillales. Numerous studies

394

have demonstrated protective effects of certain Lactobacillus strains against atopy by

395

various mechanisms including alteration of gastrointestinal (44) and respiratory tract

396

permeability (45, 46). Our finding that the predicted functions of asthma-associated

397

microbiota were relatively depleted in machinery for LPS biosynthesis suggests another

398

possibility, for continuous stimulation of airways with LPS has been shown to suppress

399

T2 immune activation (47). We propose, though, that any “pro-asthmatic” activity of a

400

bronchial microbiome is likely dependent on functional effects consequent to interactions

401

among many microbiota members in the airway microenvironment, rather than the

402

activity of any one species. Microbiome-related functions indicated by our analyses as

403

potentially enhanced in asthma include increased capacity for metabolism of butyrate

404

and propionate, SCFAs that maintain epithelial barrier function and immune tolerance in

405

the gut (48-50). We speculate that utilization of anti-inflammatory SCFAs by members

406

of the asthmatic airway microbiota may contribute to atopic asthma by reducing their

407

bioavailability and the consequent capacity to down-regulate host inflammatory

408

responses to aeroallergens and pathogens.

SC

M AN U

TE D

409

RI PT

392

Microbiome-related functions might also affect responsiveness to corticosteroid

411

treatment. We found that pre-treatment enrichment in an asthma-associated

412

Haemophilus, a genus with species previously shown to reduce response of BAL

413

macrophages to corticosteroids (2), was associated with diminished response to six

414

weeks of treatment with inhaled fluticasone. Analysis of the predicted metagenome of

415

pre-treatment bronchial microbiota present in ICS-non-responders also indicated

416

enhancement of xenobiotic degradation capacity, which we hypothesize may contribute

417

to their diminished response. In contrast, ICS-induced changes to the bronchial

418

microbiota in ICS-responsive asthmatics showed enrichment of previously reported

419

asthma-associated taxa such as Neisseria and Moraxella (2, 3, 6). Community shift

420

detected in response to lactose-containing placebo inhaler was not surprising, as an

421

influx of an additional sugar would be expected to alter the composition of microbial

422

communities in the airways, favoring those species with the metabolic capacity to utilize

423

such carbon sources. As importantly, these results emphasize the need to consider the

424

effects of repeated inhalation of particles, especially of an ICS, as a selective pressure

AC C

EP

410

13

ACCEPTED MANUSCRIPT

425

and nutritional source for airway microbiome members with the catalytic capacity to

426

degrade such xenobiotics.

427 Perturbations to the bronchial microbiota associated with atopy-alone is also a novel

429

finding of this study. ANA subjects were less sensitized than the AA subjects and

430

showed primarily low T2 inflammation of bronchial epithelium. While the greater severity

431

of allergy in the AA group cannot be ruled out as contributing to the difference in

432

bacterial signature observed between AA and ANA subjects, distinct taxa associations

433

with different markers of atopy and allergic inflammation were observed, suggesting that

434

distinct microbial interactions with the host immune system occur in these two patient

435

groups. These observations in ANA subjects support an association between airway

436

colonization and atopy-related altered mucosal immune functions (9, 10). Atopy-specific

437

taxa included members of the Pasteurellaceae (specifically Aggregatibacter and

438

Haemophilus). Members of Aggregatibacter are associated with periodontal disease

439

driven by high pro-inflammatory cytokines (e.g., TNF-a, IL1B, IL-6 and IL-8;) and

440

reduced levels of IL-10 (51, 52). It is tempting to speculate that airway enrichment of this

441

genus in ANA subjects could contribute to asthma protection through activation of the T1

442

arm of the immune system. We also highlight that specific taxa belonging to certain

443

genera (e.g., Prevotella and Haemophilus) were associated with atopy-only or asthma.

444

This underscores the likely importance of species- or strain-level functional differences in

445

microbial interactions related to disease status.

TE D

M AN U

SC

RI PT

428

446

The limitations of our study include exclusive analysis of bacterial communities, the

448

absence of non-atopic asthmatic subjects and the relatively narrow breadth of asthma

449

phenotype captured in this cohort. Moreover, our sample size was small in some

450

comparisons, particularly in assessing changes in the bronchial microbiota in paired

451

samples before and after ICS vs. placebo treatment. However, strengths include the

452

large number of subjects who were characterized and underwent invasive bronchoscopy

453

to collect samples for analysis. Our study protocol also attended carefully to procedural

454

and sampling methods to reduce contamination and analyze for possible contribution of

455

non-bronchial sources of bacterial DNA to the dataset.

AC C

EP

447

456 457

Conclusion:

14

ACCEPTED MANUSCRIPT

Our findings highlight the complexity of bacterial relationships to asthma in a background

459

of atopy, conditions that are associated with distinct alterations in the airway

460

microbiota. To achieve a comprehensive understanding of microbial factors involved in

461

the induction of, management of, or protection against asthma, there is an important

462

need to better understand functions collectively expressed by consortia of airway

463

microbes, which could have a profound influence on asthma.

464

RI PT

458

Acknowledgements:

466

We additionally would like to thank the following coordinators and staff, and to identify

467

the grants supporting the study: Robert Pedicini, BS1; Kathy Zheng, MPH1; Duanny

468

Alva, MPH2; Assel Biyasheva, PhD2; Jenny Hixon, BS, CCRC2; Lucius Robinson III,

469

BS, CMA, CCRC2; Mary Gill, RN, BSN3; James T. Good, MD3; Christena Kolakowski,

470

MS3; Allen Stevens, CCRC, NREMT3; E. Rand Sutherland, MD3; Julia Bach, RN4; Rich

471

Cornwell, MD4; Holly Eversoll, RN4; Tiffany Huard4; Keith Meyer, MD4; Barbara Miller,

472

RN4; Ann Sexton, MPH4; Michele Wolff, RN4; Merritt Fajt, MD5; Sherri Hill, BS5; Lisa

473

Lane, BS5; Russell Traister, MD5; Cathy Vitari, RN, BSN AE-C5; Vanessa Curtis, RRT6;

474

Brenda Patterson, RN, APN6; Cheryl Shelton, RN, BSN6; Kelly Norsworthy, BA, CPT7;

475

Kelsey Wollen, BA7; Eugene Bleeker, MD8; Christopher Barrios, MD8; Suzan Farris,

476

CCRP8; Jeffrey Krings, FNP8; Victor Ortega, MD8; Cheryl Wilmoth, CCRP8; Matthew

477

Bowman, BS9; Linda Engle, BS9; Jennifer Lucier, BS9; Aimee J. Merchlinski, MS9;

478

Kathryn Trasatt, BS9; Angela Updegrave9; Rachel Weber, BS9; Ronald R. Zimmerman,

479

Jr., MPA9. We also wish to thank Elizabeth F. Juniper of McMaster’s University,

480

Canada, for permitting our use of her Asthma Control Questionnaire, and Andrew

481

Manies of UCSF for assistance with manuscript preparation.

AC C

EP

TE D

M AN U

SC

465

15

ACCEPTED MANUSCRIPT

References: 1.

RI PT

2.

Denner DR, Sangwan N, Becker JB, Hogarth DK, Oldham J, Castillo J, Sperling AI, Solway J, Naureckas ET, Gilbert JA, White SR. 2015. Corticosteroid therapy and airflow obstruction influence the bronchial microbiome, which is distinct from that of bronchoalveolar lavage in asthmatic airways. Journal of Allergy and Clinical Immunology. Goleva E, Jackson LP, Kirk Harris J, Robertson CE, Sutherland ER, Hall CF, Good JTJ, Gelfand EW, Martin RJ, Leung D, Y. M. 2013. The Effects of Airway Microbiome on Corticosteroid Responsiveness in Asthma. American Journal of Respiratory and Critical Care Medicine 188:1193-1201. Hilty M, Burke C, Pedro H, Cardenas P, Bush A, Bossley C, Davies J, Ervine A, Poulter L, Pachter L, Moffatt MF, Cookson WOC. 2010. Disordered Microbial Communities in Asthmatic Airways. PLOSone 5:e8578. Huang YJ, Nariya S, Harris JM, Lynch SV, Choy DF, Arron JR, Boushey HA. 2015. The airway microbiome in patients with severe asthma: Associations with disease features and severity. The Journal of allergy and clinical immunology 136:874-884. Huang YJ, Nelson CE, Brodie EL, Desantis TZ, Baek MS, Liu J, Woyke T, Allgaier M, Bristow J, Wiener-Kronish JP, Sutherland ER, King TS, Icitovic N, Martin RJ, Calhoun WJ, Castro M, Denlinger LC, Dimango E, Kraft M, Peters SP, Wasserman SI, Wechsler ME, Boushey HA, Lynch SV. 2011. Airway microbiota and bronchial hyperresponsiveness in patients with suboptimally controlled asthma. The Journal of allergy and clinical immunology 127:372-381.e371-373. Marri PR, Stern DA, Wright AL, Billheimer D, Martinez FD. 2013. Asthma-associated differences in microbial composition of induced sputum. The Journal of allergy and clinical immunology 131:346-352.e341-343. Burrows B, Martinez FD, Halonen M, Barbee RA, Cline MG. 1989. Associations of asthma with serum IgE levels and skin-test reactivity to allergens. N Engl J Med 320:271277. Pearce N, Pekkanen J, Beasley R. 1999. How much asthma is really attributable to atopy? Thorax 54:268-272. Kariyawasam HH, Rotiroti G. 2013. Allergic rhinitis, chronic rhinosinusitis and asthma: unravelling a complex relationship. Current Opinion in Otolaryngology & Head and Neck Surgery 21:79-86. Tomazic PV, Birner-Gruenberger R, Leitnera A, Obrist B, Spoerkc S, Lang-Loidolt D. 2014. Nasal mucus proteomic changes reflect altered immune responses and epithelial permeability in patients with allergic rhinitis. . The Journal of allergy and clinical immunology 133:741-750. Juniper EF, O'Byrne PM, Guyatt GH, Ferrier PJ, King DR. 1999. Development and validation of a questionnaire to measure asthma control. Eur Respir J. 14:902-907. Iwai S, Fei M, Huang D, Fong S, Subramanian A, Grieco K, Lynch SV, Huang L. 2012. Oral and Airway Microbiota in HIV-Infected Pneumonia Patients. J. Clin. Microbiol. 50:2995-3002. Caporaso JG, Lauber CL, Walters WA, Berg-Lyons D, Huntley J, Fierer N, Owens SM, Betley J, Fraser L, Bauer M, Gormley N, Gilbert JA, Smith G, Knight R. 2012. Ultra-high-throughput microbial community analysis on the Illumina HiSeq and MiSeq platforms. The ISME Journal 6:1621-1624. Caporaso JG, Lauber CL, Walters WA, Berg-Lyons D, Lozupone CA, Turnbaugh PJ, Fierer N, Knight R. 2011. Global patterns of 16S rRNA diversity at a depth of millions of sequences per sample. Proceedings of the National Academy of Sciences of the United States of America 108 Suppl 1:4516-4522. DeSantis TZ, Hugenholtz P, Larsen N, Rojas M, Brodie EL, Keller K, Huber T, Dalevi D, Hu P, Andersen GL. 2006. Greengenes, a chimera-checked 16S rRNA gene database and workbench compatible with ARB. Applied and environmental microbiology 72:5069-5072.

3.

SC

4.

7.

8. 9.

10.

11. 12.

EP

6.

TE D

M AN U

5.

AC C

482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534

13.

14.

15.

16

ACCEPTED MANUSCRIPT

21. 22. 23. 24. 25.

26. 27.

28.

29.

30.

RI PT

20.

SC

19.

M AN U

18.

TE D

17.

Price MN, Dehal PS, Arkin AP. 2009. FastTree: Computing Large Minimum Evolution Trees with Profiles instead of a Distance Matrix. Molecular Biology and Evolution 26:1641-1650. Faith DP. 1992. Conservation evaluation and phylogenetic diversity. Biological Conservation 61:1-10. Woodruff PG, Boushey HA, Dolganov GM, Barker CS, Yang YH, Donnelly S, Ellwanger A, Sidhu SS, Dao-Pick TP, Pantoja C, Erle DJ, Yamamoto KR, Fahy JV. 2007. Genome-wide profiling identifies epithelial cell genes associated with asthma and with treatment response to corticosteroids. Proc Natl Acad Sci USA 104:15858-15868. Woodruff PG, Modrek B, Choy DF, Jia J, Abbas AR, Ellwanger A, Koth LL, Arron JR, Fahy JV. 2009. T-helper type 2-driven inflammation defines major subphenotypes of asthma. Am J Respir Crit Care Med. 180:388-395. Bhakta NR, Solberg OD, Nguyen CP, Nguyen CN, Arron JR, Fahy JV, Woodruff PG. 2013. A qPCR-based metric of Th2 airway inflammation in asthma. Clin. Transl. Allergy 3:24. Anderson MJ. 2001. A new method for non-parametric multivariate analysis of variance. Austral Ecology 26:32-46. Pinheiro JC. 2005. Linear Mixed Effects Models for Longitudinal Data, Encyclopedia of Biostatistics. John Wiley & Sons, Ltd. Gerber GK, Onderdonk AB, Bry L. 2012. Inferring dynamic signatures of microbes in complex host ecosystems. PLoS computational biology 8:e1002624. McMurdie PJ, Holmes S. 2014. Waste Not, Want Not: Why Rarefying Microbiome Data Is Inadmissible. PLoS computational biology 10:e1003531. Kassahun W, Neyens T, Molenberghs G, Faes C, Verbeke G. 2014. Marginalized multilevel hurdle and zero-inflated models for overdispersed and correlated count data with excess zeros. Statistics in medicine 33:4402-4419. Min Y, Agresti A. 2005. Random effect models for repeated measures of zero-inflated count data. Statistical Modelling 5:1-19. Langille MGI, Zaneveld J, Caporaso JG, McDonald D, Knights D, Reyes JA, Clemente JC, Burkepile DE, Vega Thurber RL, Knight R, Beiko RG, Huttenhower C. 2013. Predictive functional profiling of microbial communities using 16S rRNA marker gene sequences. Nat Biotech 31:814-821. Simpson JL, Daly J, Baines KJ, Yang IA, Upham JW, Reynolds PN, Hodge S, James AL, Hugenholtz P, Willner D, Gibson PG. 2015. Airway dysbiosis: Haemophilus influenzae and Tropheryma in poorly controlled asthma. The European respiratory journal. Peters MC, Mekonnen ZK, Yuan S, Bhakta NR, Woodruff PG, Fahy JV. 2014. Measures of gene expression in sputum cells can identify TH2-high and TH2-low subtypes of asthma. The Journal of allergy and clinical immunology 133:388-394. Ursell LK, Clemente JC, Rideoutb JR, Gevers D, Caporaso JG, Knight R. 2012. The interpersonal and intrapersonal diversity of human-associated microbiota in key body sites. J. Allergy Clin. Immunol. 129:1204–1208. Hogan SP, Waddell A, Fulkerson PC. 2013. Eosinophils in Infection and Intestinal Immunity. . Current Opinion in Gastroenterology 29:7–14. Travers J, Rothenberg ME. 2015. Eosinophils in mucosal immune responses. Mucosal Immunol 8:464-475. Dollive S, Chen Y-Y, Grunberg S, Bittinger K, Hoffmann C, Vandivier L, Cuff C, Lewis JD, Wu GD, Bushman FD. 2013. Fungi of the Murine Gut: Episodic Variation and Proliferation during Antibiotic Treatment. PLoS ONE 8:e71806. Peleg AY, Hogan DA, Mylonakis E. 2010. Medically important bacterial–fungal interactions. Nat Rev Micro 8:340-349. Kloepfer KM, Lee WM, Pappas TE, Kang TJ, Vrtis RF, Evans MD, Gangnon RE, Bochkov YA, Jackson DJ, Lemanske RF, Jr., Gern JE. 2014. Detection of pathogenic bacteria during rhinovirus infection is associated with increased respiratory symptoms and asthma exacerbations. The Journal of allergy and clinical immunology 133:13011307, 1307.e1301-1303.

EP

16.

AC C

535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590

31. 32. 33.

34. 35.

17

ACCEPTED MANUSCRIPT

41.

42.

43. 44. 45.

46.

47.

48.

RI PT

40.

SC

39.

M AN U

38.

TE D

37.

Teo Shu M, Mok D, Pham K, Kusel M, Serralha M, Troy N, Holt Barbara J, Hales Belinda J, Walker Michael L, Hollams E, Bochkov Yury A, Grindle K, Johnston Sebastian L, Gern James E, Sly Peter D, Holt Patrick G, Holt Kathryn E, Inouye M. The Infant Nasopharyngeal Microbiome Impacts Severity of Lower Respiratory Infection and Risk of Asthma Development. Cell Host & Microbe 17:704-715. Tarkka MT, Sarniguet A, Frey-Klett P. 2009. Inter-kingdom encounters: recent advances in molecular bacterium-fungus interactions. Current genetics 55:233-243. Pana Y, Tenga D, Burkea AC, Haasea EM, Scannapieco FA. 2008. Oral bacteria modulate invasion and induction of apoptosis in HEp-2 cells by Pseudomonas aeruginosa. Microbial Pathogenesis 46:73-79. Nagaoka K, Yanagihara K, Harada Y, Yamada K, Migiyama Y, Morinaga Y, Hasegawa H, Izumikawa K, Kakeya H, Nishimura M, Kohno S. 2013. Macrolides inhibit Fusobacterium nucleatum-induced MUC5AC production in human airway epithelial cells. Antimicrobial agents and chemotherapy 57:1844-1849. Araki N, Yanagihara K, Morinaga Y, Yamada K, Nakamura S, Yamada Y, Kohno S, Kamihira S. 2010. Azithromycin inhibits nontypeable Haemophilus influenzae-induced MUC5AC expression and secretion via inhibition of activator protein-1 in human airway epithelial cells. European journal of pharmacology 644:209-214. Akbari O, Stock P, Meyer E, Kronenberg M, Sidobre S, Nakayama T, Taniguchi M, Grusby MJ, DeKruyff RH, Umetsu DT. 2003. Essential role of NKT cells producing IL-4 and IL-13 in the development of allergen-induced airway hyperreactivity. Nat Med 9:582588. Kinjo Y, Wu D, Kim G, Xing GW, Poles MA, Ho DD, Tsuji M, Kawahara K, Wong CH, Kronenberg M. 2005. Recognition of bacterial glycosphingolipids by natural killer T cells. Nature 434:520-525. Zajonc DM, Girardi E. 2015. Recognition of Microbial Glycolipids by Natural Killer T Cells. Frontiers in immunology 6:400. Özdemir O. 2010. Various effects of different probiotic strains in allergic disorders: an update from laboratory and clinical data. Clin Exp Immunol 160:295-304. Abreu NA, Nagalingam NA, Song Y, Roediger FC, Pletcher SD, Goldberg AN, Lynch SV. 2012. Sinus microbiome diversity depletion and Corynebacterium tuberculostearicum enrichment mediates rhinosinusitis. Sci Transl Med. 12:151ra124. Fujimura KE, Demoor T, Rauch M, Faruqi AA, Jang S, Johnson CC, Boushey HA, Zoratti E, Ownby D, Lukacs NW, Lynch SV. 2014. House dust exposure mediates gut microbiome Lactobacillus enrichment and airway immune defense against allergens and virus infection. Proc Natl Acad Sci USA 111:805-810. Schuijs MJ, Willart MA, Vergote K, Gras D, Deswarte K, Ege MJ, Branco Madeira F, Beyaert R, van Loo G, Bracher F, von Mutius E, Chanez P, Lambrecht BN, Hammad H. 2015. Farm dust and endotoxin protect against allergy through A20 induction in lung epithelial cells. Science 349:1106-1110. Arrieta MC, Stiemsma LT, Dimitriu PA, Thorson L, Russell S, Yurist-Doutsch S, Kuzeljevic B, Gold MJ, Britton HM, Lefebvre DL, Subbarao P, Mandhane P, Becker A, McNagny KM, Sears MR, Kollmann T, Mohn WW, Turvey SE, Brett Finlay B. 2015. Early infancy microbial and metabolic alterations affect risk of childhood asthma. Science translational medicine 7:307ra152. Kelly CJ, Zheng L, Campbell EL, Saeedi B, Scholz CC, Bayless AJ, Wilson KE, Glover LE, Kominsky DJ, Magnuson A, Weir TL, Ehrentraut SF, Pickel C, Kuhn KA, Lanis JM, Nguyen V, Taylor CT, Colgan SP. 2015. Crosstalk between MicrobiotaDerived Short-Chain Fatty Acids and Intestinal Epithelial HIF Augments Tissue Barrier Function. Cell Host Microbe 17:662-671. Trompette A, Gollwitzer ES, Yadava K, Sichelstiel AK, Sprenger N, Ngom-Bru C, Blanchard C, Junt T, Nicod LP, Harris NL, Marsland BJ. 2014. Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat Med 20:159-166.

EP

36.

AC C

591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644

49.

50.

18

ACCEPTED MANUSCRIPT

51.

EP

TE D

M AN U

SC

RI PT

52.

Bostanci N, Akgül B, Tsakanika V, Allaker RP, Hughes FJ, McKay IJ. 2011. Effects of low-dose doxycycline on cytokine secretion in human monocytes stimulated with Aggregatibacter actinomycetemcomitans. Cytokine 56:656-661. Hirose M, Ishihara K, Saito A, Nakagawa T, Yamada S, Okuda K. 2001. Expression of cytokines and inducible nitric oxide synthase in inflamed gingival tissue. J Periodontol. 72:590-597.

AC C

645 646 647 648 649 650 651

19

ACCEPTED MANUSCRIPT

Table 1. Study cohort characteristics p-value#

28 (23 - 46)

NS

-

-

-

-

-

-

48%

43%

NS€

52%

71%

NS€

25 (21 - 28)

26 (22 - 28)

NS

100 (93 - 108) 102 (98 - 116) 5.0 (2.5 - 5.0)

<0.01&

<0.0001&

Age (yrs)

33 (25 - 41)

28 (24 - 44)

Age of asthma diagnosis (yrs)

9 (5 - 22)

-

Duration of Asthma (yrs)

21 (14 - 27)

-

ACQ Score*

0.3 (0 - 1.3)

-

% Male

45%

% White

60%

M AN U

26 (23 - 29)

Atopic, non-asthmatic (ANA) (n = 21)

RI PT

Atopic asthmatic (AA) (n = 42)**

BMI (kg/m2)

89 (76 - 97) 100 (84 - 106)

7.5 (5.0 - 14.3)

98 (89 - 109) 104 (97 - 109) 3.0 (-0.5 - 5.5)

Methacholine PC20

1.2 (0.3 - 3.2)

>32$

>32$

-

Blood eosinophils (absolute) Blood eosinophils (%)

200 (100 - 393) 3.3 (1.5 - 5.6) 55.5 (49.3 - 62.0) 54.6 (31.8 - 64.9) 0.4 (0.0 - 1.1)

100 (87 - 200) 2.0 (1.4 - 3.0) 56.9 (51.0 - 63.8) 39.5 (26.5 - 50.0) 0.0 (0.0 - 0.6)

100 (60 - 200) 1.4 (1.0 - 3.0)

<0.01

58.4 (52.7 63.5) 41.8 (29.2 76.5) 0.0 (0.0 - 0.4)

169.5 (56.3 - 321.3) 6 (2-7)

64.0 (22.0 - 164.5) 3 (2-5)

15.0 (5.0 31.0) -

TE D

FEV1 % predicted (pre-albuterol) FEV1 % predicted (post-albuterol) Change in FEV1%

Blood neutrophils (%)

Sputum neutrophils (%)

EP

Sputum eosinophils (%) Serum IgE (IU/mL)

¢

AC C

Number of positive sIgE

654 655 656 657 658 659 660 661 662

Healthy control (HC) (n = 21)

Variable

SC

652 653

NS&

<0.01¥ NS NS NS¥ <0.01&¥ <0.05&

All values are medians (IQR).*ACQ, Asthma Control Questionnaire ** Number of exacerbations requiring oral steroids in the past 5 years: zero exacerbations (38 subjects); one exacerbation (3 subjects); two $ exacerbations (one subject). Methacholine challenge was stopped at 32 mg/dL and PC20 for these subjects ¥ ¢ was censored. For between group statistical comparisons see Supplementary Figure S2A-C. Number of positive specific IgE (sIgE >0.35 kU/l) from a total of 12 aeroallergens tested by ImmunoCap assay (for breakdown of specific aeroallergens see Supplementary Table S1). Statistical significance was determined # € & using Kruskal-Wallis or Chi-square or Mann-Whitney test for AA vs. ANA comparison.

20

ACCEPTED MANUSCRIPT

Figure Legends:

664

Figure 1: (A) Principal coordinate analysis (unweighted UniFrac) shows compositional

665

dissimilarity between paired BB and OW samples (LME p<0.0001). (B) Phylogenetic

666

diversity (Faith’s index) in BB and paired OW samples (& Wilcoxon matched-pairs signed

667

rank test). (C) Phylogenetic diversity (Faith’s index) in BB samples for the three subject

668

groups (Welch’s corrected t-test).

RI PT

663

669

Figure 2: Bacterial taxa, significantly enriched or depleted in relative abundance (at

671

least 2 fold; NB regression, q<0.1) in (A) AA (n=28) compared to HC (n=13); (B) ANA

672

(n=15) compared to HC; (C) AA compared to ANA. The OTUs indicated represent the

673

most abundant representatives within the indicated genus.

SC

670

674

Figure 3: Mean difference in specific bacterial taxa between groups. Asthma-specific

676

taxa are similarly abundant in AA compared to HC and ANA (NB regression, q<0.1).

677

Atopy-only taxa are similarly abundant in ANA vs. both HC and AA. Taxa positively

678

correlated (rPearson≥0.5, q<0.1) with blood eosinophil counts (BEO), serum IgE (IgE) or

679

sputum eosinophil counts (SEO) in AA (*) or ANA (#) subjects.

M AN U

675

680

Figure 4. Mean difference in predicted KEGG orthologs (KOs) between groups. Asthma-

682

specific pathways are similarly abundant in AA compared to HC and ANA. Atopy-only

683

pathways are similarly abundant in ANA compared to HC and AA. Statistical significance

684

was determined using NB regression model corrected for false discovery rate (q<0.1).

EP

685

TE D

681

Figure 5. (A) AA displayed greater expression of epithelial genes induced by type 2

687

cytokines compared to non-asthmatic subjects. T2-high AA, with a TGM ≥1.117 (cut-off

688

value indicated by a dashed line) are colored in maroon red. (B) Significantly lower

689

bacterial burden was observed among T2-high asthma subjects. Statistical significance

690

was determined using Wilcoxon rank sum test.

691

AC C

686

692

Figure 6. (A) Mean distance to HC (unweighted UniFrac; Bonferroni-corrected t-test).

693

(B) Taxa significantly enriched or depleted in relative abundance (at least 2 fold; NB

694

regression, q<0.1) in ICS-responders vs. non-responders. (C) Predicted KEGG

695

pathways associated with xenobiotic biodegradation and metabolism in ICS-responders

21

ACCEPTED MANUSCRIPT

and non-responders. (D) Taxa, differentially expressed (at least 2-fold; ZINB regression,

697

q<0.1) in asthmatics following ICS or placebo treatment.

AC C

EP

TE D

M AN U

SC

RI PT

696

22

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

SUPPLEMENTAL FIGURES AND TABLES:

M AN U

SC

RI PT

Figure S1: Schematic diagrams summarizing the breakdown of participants enrolled in this study. (A) Of the 102 asthmatic adults screened for this study, 42 atopic asthmatics (AAs) were enrolled. These subjects underwent specimen collection at baseline (prior to any treatment) and were then randomized to receive inhaled placebo or 250 mcg of fluticasone (ICS) twice daily for six weeks. (B) Of the 84 atopic non-asthmatic (ANAs) and non-atopic non-asthmatic healthy control (HCs) subjects screened, 21 from each group were enrolled in this study. (C) Enrolled participants were seen at multiple visits (indicated by arrows) in nine participating study centers, during which they underwent spirometry (S), methacholine provocation (MP), sputum induction (IS) and bronchoscopy (B). Following baseline assessment (visits 1 and 2) asthmatic patients underwent additional assessment post six weeks twice-daily treatment with ICS or placebo inhaler (post-treatment assessment visits 3 and 4). (D) A schematic diagram of the total bronchial brush samples (BBs) collected during bronchoscopy at baseline (visit 2) indicating the breakdown of subjects with sufficient bacterial 16S rRNA for bacterial profile. The proportion of sequenced to not-sequenced BB samples did not vary by subject group or clinical study site (#Chi-square test; p>0.5). (E) Breakdown of BBs collected from asthmatics post-treatment (visit 4) showing the proportion of sequenced to not sequenced samples, which did not vary between type of inhaler used (&Fishers exact test; p>0.5).

EP

TE D

Figure S2: (A) Total serum IgE concentrations differentiate the three groups. (B, C) Atopic asthmatics show evidence of systemic eosinophilia. (D) Bacterial burden (normalized to ß-actin expression) in sequenced samples was significantly higher than in non-sequenced samples. (E) Bacterial burden in the second scope flush was indistinguishable between a random subset of sequenced and non-sequenced samples, while their paired brush 16S rRNA burden was significantly higher for the sequenced samples. (F) Bacterial burden (normalized to ß-actin expression) for sequenced samples did not significantly vary between study groups. (G) Bacterial richness did not significantly vary between study groups. (H) Bacterial diversity based on Shannon Index did not significantly vary between study groups. (I) Bacterial evenness based on Pielou’s Index did not significantly vary between study groups. Statistical significance was determined using Wilcoxon rank sum test, & Wilcoxon matched-pairs signed rank test or $Welch’s corrected t-test.

AC C

Figure S3. The bronchial microbiota of all groups is highly heterogeneous, but compositional variability is higher in the atopic asthmatic subjects. Principal coordinate analysis plots based on unweighted UniFrac distance, where bronchial brush samples (A) from all subjects (28 atopic asthmatics – AA; 15 atopic non asthmatics – ANA; 13 healthy controls – HC) are colored by group, which does not explain variation in community composition (PERMANOVA p>0.05); (B) from all subjects are colored by richness or (C) from AAs only colored by richness or (D) ANAs only colored by richness or (E) HC only colored by richness. Community richness (Observed species ranging from 219 to 1122 operational taxonomic units (OTUs), significantly explained variability in composition between samples in all groups (PERMANOVA, p≤0.01). (F) Distance box plots based on weighted UniFrac distance matrix show greater compositional variability in the AA compared to both ANAs and HC subjects (F-test p<0.05).

ACCEPTED MANUSCRIPT

RI PT

Figure S4: Summary of bacterial taxa at a phylum level, significantly enriched or depleted in relative abundance (at least 2 fold) in (A) atopic asthmatic (AAs) patients (n=28) compared to healthy control subjects (HC, n=13); (B) atopic non-asthmatic (ANAs) subjects (n=15) compared to HC subjects; (C) AAs patients compared to ANAs subjects. The number of operational taxonomic units (OTU) included in each phyla are indicated by #OTU. Statistical significance was determined using Negative binomial regression model corrected for false discovery rate (q<0.1).

M AN U

SC

Figure S5: (A) ICS-responders showed increase in the dose of methacholine in the PC20 challenge after 6 weeks of treatment with fluticasone, but not ICS non-responders or asthmatics taking a placebo inhaler. (B) ICS-responders showed at least one doubling in the dose of methacholine after 6 weeks of treatment with fluticasone. (C) ICS-responders exhibited slightly higher T2-type inflammation at baseline compared to ICS non-responders. Subjects identified as T2-high asthmatics are colored in maroon. (D) ICS-responders showed significant decline in the three-gene signature after treatment with fluticasone compared to both ICS non-responders and placebo controls. Statistical significance was determined using Wilcoxon rank sum test and & Wilcoxon matched-pairs rank sum test. Figure S6: (A) Treatment with ICS had no significant effect on the change in bacterial burden compared to placebo inhaler. Statistical significance was determined using Wilcoxon rank sum test. (B) Treatment with ICS had no significant effect on the change in Faith’s phylogenetic diversity compared to placebo inhaler. Statistical significance was determined using Wilcoxon rank sum test. (C) Procrustes analysis plot based on unweighted UniFrac distance of asthmatics taking placebo treatment (n = 8) shows no significant similarity in community composition between pre- and post-treatment paired samples (M = 0.635, p>0.05). (D) Procrustes analysis plot based on unweighted UniFrac distance shows no significant similarity in community composition between pre- and post-treatment paired samples (M = 0.504, p>0.05) in ICSresponders (n = 8). (E) Analysis of paired unweighted UniFrac distance shows no significant difference in the magnitude of the community shift identified with treatment using fluticasone or a lactose containing placebo inhaler. Statistical significance was determined using Wilcoxon rank sum test.

AC C

EP

TE D

2

2

ACCEPTED MANUSCRIPT

Table S1: Breakdown of sensitivities to specific aeroallergens in the atopic asthmatics and atopic non-asthmatic subjects. p-value#

Healthy controls (n = 21) -

NS NS <0.05 0.05 0.05 NS NS NS NS NS NS NS

RI PT

sIgE (Mite-1) sIgE (Mite-2) sIgE (Cat) sIgE (Dog) sIgE (Mouse) sIgE (Grass mix) sIgE (Cockroach) sIgE (Mold mix) sIgE (Tree mix 1) sIgE (Tree mix 2) sIgE (Weed) sIgE (Weed mix)

Atopic non-asthmatics (n = 21) 50% 60% 40% 32% 0% 37% 11% 16% 21% 21% 18% 29%

SC

Variable

Atopic asthmatics (n = 42) 56% 54% 68% 59% 17% 58% 28% 38% 42% 34% 34% 47%

#

M AN U

Percentage of positive specific IgE (sIgE >0.35 kU/l) as tested by ImmunoCap assay. Statistical significance was determined using Chi-square test.

Table S2: Responses to Prior clinical conditions questionnaire for atopic participants. Questions about Prior conditions

Atopic Asthmatics (n=42)

Atopic Nonasthmatics (n=21)

#

AC C

EP

TE D

Have you ever had eczema/allergic dermatitis? Yes 31% 5% No 69% 95% Have you ever had allergic rhinitis (hay fever)? Yes 53% 24% No 48% 76% Do you have chronic or recurrent sinusitis (treated with antibiotics and/or surgery)? Yes 7% 0% No 93% 100% Have you ever had pneumonia? Yes 29% 10% No 71% 91% Have you been diagnosed with sleep disordered breathing (sleep apnea)? Yes 5% 0% No 95% 100% Do you have gastroesophageal reflux disease (GERD)? Yes 10% 5% No 91% 95%

p-value#

0.02

0.06

NS

0.11

NS

NS

Statistical significance was determined using Chi-square test, where NS = p≥0.2.

3

ACCEPTED MANUSCRIPT

Table S3: Responses to Environmental exposure questionnaire for atopic participants. Atopic Asthmatics (n = 42)

Environmental exposure questions

Atopic Nonasthmatics (n = 21)

AC C

EP

TE D

M AN U

SC

RI PT

Do you live within a mile of a farm? Yes 17% 10% No 83% 91% Do you live within a mile of a major highway? Yes 67% 48% No 33% 52% Does the home you live in have a yard? Yes 69% 57% No 31% 43% Have you been around animals outside your home at least 2 days/week in the past 3 months? Yes 36% 29% No 64% 71% Are you frequently exposed (2 or more days/week) to tobacco smoke outside of your home? Yes 17% 10% No 83% 91% Has there been any mold or mildew, on any surfaces inside your house in the past 12 months? Yes 35% 35% No 65% 65% Does your household have any pets? Yes 33% 43% No 67% 57% In general, and on a regular basis, are you exposed to cats? Yes 29% 43% No 71% 57% In general, and on a regular basis, are you exposed to dogs? Yes 50% 43% No 50% 57% In general, and on a regular basis, are you exposed to other furry pets? Yes 7% 5% No 93% 95% In general, and on a regular basis, are you exposed to birds? Yes 7% 5% No 93% 95% In general, and on a regular basis, are you exposed to farm animals? Yes 5% 0% No 95% 100% Do you ever see cockroaches in your house? Yes 14% 19% No 86% 81%

p-value#

Do you ever see rodents or rodent droppings in your house? Yes 14% No 86% Do you ever see cockroaches OR rodents/rodent droppings in your house? Yes 29% No 71% #

NS

0.18

NS

NS

NS

NS

NS

NS

NS

NS

NS

NS

NS

19% 81%

NS

33% 67%

NS

Statistical significance was determined using Chi-square test, where NS = p≥0.2.

4

ACCEPTED MANUSCRIPT

Table S4: Specific bacterial taxa that differ in relative abundance between atopic asthmatic (AA) and healthy control (HC) subjects. #

AA 356.8

#

HC 61.5

AA-HC 295.3

q-value

Phylum

Class

0.0610

Bacteroidetes

Bacteroidia

Order Bacteroidales

Proteobacteria

Gammaproteobacteria

Pasteurellales

Fusobacteria

Fusobacteriia

Fusobacteriales

Proteobacteria

Betaproteobacteria

Neisseriales

Family Paraprevotellaceae

Genus Prevotella

Species tannerae

Pasteurellaceae

Haemophilus

-

Fusobacteriaceae

Fusobacterium

-

Neisseriaceae

Neisseria

-

Fusobacteriaceae

Fusobacterium

-

RI PT

OTU ID 2714267

243.8

1.0

242.8

2438396

192.5

1.7

190.8

4440404

176.8

0.2

176.7

3.00E106 1.66E-21

4405869

196.0

47.4

148.6

0.0347

Fusobacteria

Fusobacteriia

Fusobacteriales

100.2

0.0759

Actinobacteria

Actinobacteria

Actinomycetales

Actinomycetaceae

Actinomyces

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

-

Bacteroidales

Prevotellaceae

Prevotella

-

Spirochaetales

Spirochaetaceae

Treponema

-

Actinomycetales

Actinomycetaceae

Actinomyces

-

Clostridiales

Veillonellaceae

Veillonella

dispar

Clostridiales

Mogibacteriaceae

Anaerovorax

-

Bacteroidales

Prevotellaceae

Prevotella

-

Bacteroidales

Prevotellaceae

Prevotella

-

137.9

37.8

240049

48.2

0.8

47.3

0.0011

1541939

47.9

2.0

45.9

0.0011

Bacteroidetes

Bacteroidia

71146

43.4

1.4

42.0

0.0231

Spirochaetes

Spirochaetes

4448211

32.6

6.5

26.1

0.0476

Actinobacteria

Actinobacteria

Firmicutes

Clostridia

Firmicutes

Clostridia Bacteroidia

31.8

6.8

24.9

4468500

24.4

2.5

21.9

1.02E-34

642465

21.4

0.3

21.1

0.0041

Bacteroidetes

70671

87.4

67.5

19.9

2.66E-09

Bacteroidetes Spirochaetes

Bacteroidia

TE D

4388775

0.0297

M AN U

4296424

SC

4406393

0.0041

Spirochaetes

25.5

7.4

18.1

41911

17.7

0.2

17.4

0.0027

Bacteroidetes

Spirochaetales

Spirochaetaceae

Treponema

-

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

-

4297695

16.1

2.5

13.6

0.0667

Proteobacteria

Epsilonproteobacteria

Campylobacterales

Campylobacteraceae

Campylobacter

-

495451

11.3

0.4

10.9

4.53E-12

Bacteroidetes

Bacteroidia

Bacteroidales

Porphyromonadaceae

Porphyromonas

-

250288

14.1

5.4

8.8

5.95E-12

Firmicutes

Clostridia

Clostridiales

Peptostreptococcaceae

-

-

Fusobacteria

Fusobacteriia

EP

73875

4.02E-28

9.8

1.1

8.7

Fusobacteriales

Leptotrichiaceae

Leptotrichia

-

4307228

9.1

0.8

8.3

0.0550

Spirochaetes

Spirochaetes

Spirochaetales

Spirochaetaceae

Treponema

-

1398

8.2

0.4

7.8

0.0277

Bacteroidetes

Bacteroidia

Bacteroidales

Paraprevotellaceae

Prevotella

-

949816

9.2

1.7

7.5

0.0622

Firmicutes

Bacilli

Lactobacillales

Streptococcaceae

Streptococcus

-

Proteobacteria

Betaproteobacteria

AC C

1033013

6.48E-14

4480775

8.4

1.0

7.4

0.0231

Neisseriales

Neisseriaceae

Kingella

-

4331815

7.5

0.5

7.0

3.60E-10

Proteobacteria

Alphaproteobacteria

Sphingomonadales

Sphingomonadaceae

-

-

3859

11.6

4.9

6.7

1.42E-08

Spirochaetes

Spirochaetes

Spirochaetales

Spirochaetaceae

Treponema

amylovorum

855912

6.6

1.2

5.5

3.46E-09

Fusobacteria

Fusobacteriia

Fusobacteriales

Fusobacteriaceae

Fusobacterium

-

5

ACCEPTED MANUSCRIPT

#

AA

HC

#

AA-HC

q-value

Phylum

Class

Order

Family

Genus

Species

Proteobacteria

Alphaproteobacteria

Sphingomonadales

Sphingomonadaceae

Sphingopyxis

alaskensis

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

-

Firmicutes

Bacilli

Lactobacillales

Lactobacillaceae

Lactobacillus

-

Porphyromonadaceae

Porphyromonas

-

Prevotellaceae

Prevotella

-

Mogibacteriaceae

-

-

Prevotellaceae

Prevotella

copri

267852

5.9

0.6

5.3

1.96E-08

1033687

5.1

0.3

4.8

0.0914

292057

6.3

1.5

4.7

1.07E-07

328

4.9

0.4

4.5

0.0498

Bacteroidetes

Bacteroidia

Bacteroidales

Bacteroidetes

Bacteroidia

Bacteroidales

RI PT

OTU ID

4.5

0.1

4.4

4093791

14.5

10.7

3.8

0.0469

Firmicutes

Clostridia

Clostridiales

4410166

3.1

0.1

3.0

0.0073

Bacteroidetes

Bacteroidia

Bacteroidales

1304

3.0

0.1

2.9

0.0082

Proteobacteria

Betaproteobacteria

Neisseriales

Neisseriaceae

Neisseria

subflava

2377731

3.2

0.5

2.6

0.0002

Firmicutes

Clostridia

Clostridiales

Veillonellaceae

Selenomonas

-

Fusobacteria

Fusobacteriia

Fusobacteriales

0.8

2.3

2.7

0.4

2.3

0.0009

Firmicutes

Bacilli

29566

2.3

0.1

2.2

0.0200

Fusobacteria

Fusobacteriia

1062051

2.6

0.8

1.8

0.0083

Actinobacteria

Actinobacteria

Bacteroidetes

Cytophagia

Leptotrichiaceae

Leptotrichia

-

Lactobacillales

Lactobacillaceae

Lactobacillus

iners

Fusobacteriales

Leptotrichiaceae

Sneathia

-

Actinomycetales

Corynebacteriaceae

Corynebacterium

-

M AN U

3.1

130864

2.4

0.6

1.8

Cytophagales

Cytophagaceae

-

-

1135830

2.6

1.1

1.5

0.0603

Proteobacteria

Betaproteobacteria

Burkholderiales

Comamonadaceae

-

-

4375000

0.5

1.5

-0.9

0.0713

Proteobacteria

Gammaproteobacteria

Enterobacteriales

Enterobacteriaceae

-

-

165489

0.5

1.5

-1.0

0.0333

Proteobacteria

Gammaproteobacteria

Enterobacteriales

Enterobacteriaceae

-

-

-1.0

0.0108

Actinobacteria

Actinobacteria

Actinomycetales

Micrococcaceae

Kocuria

palustris

Firmicutes

Bacilli

Lactobacillales

Streptococcaceae

Streptococcus

-

Gammaproteobacteria

Pasteurellales

Pasteurellaceae

Haemophilus

-

Fusobacteriia

Fusobacteriales

Leptotrichiaceae

-

-

Proteobacteria

Gammaproteobacteria

Enterobacteriales

Enterobacteriaceae

-

-

Proteobacteria

Gammaproteobacteria

Enterobacteriales

Enterobacteriaceae

-

-

Firmicutes

Bacilli

Lactobacillales

Streptococcaceae

Streptococcus

-

160203

0.3

1.3

0.5

1.8

-1.3

4469627

0.9

2.2

-1.3

0.0204

Proteobacteria

4335450

1.4

3.3

-1.9

0.0041

Fusobacteria

51646

0.4

2.6

-2.2

2.96E-06

EP

538185

0.0165

TE D

1116069

0.0072

AC C

832

0.0010

SC

851668

0.0017

3799784

0.9

3.2

-2.3

1.43E-05

521996

0.3

2.8

-2.6

0.0252

710275

0.8

3.5

-2.6

8.49E-07

Proteobacteria

Gammaproteobacteria

Pseudomonadales

Moraxellaceae

Acinetobacter

-

849642

0.2

4.0

-3.8

1.67E-09

Proteobacteria

Betaproteobacteria

Burkholderiales

Comamonadaceae

-

-

Proteobacteria

Betaproteobacteria

4417996

1.4

5.5

-4.1

2.22E-10

Burkholderiales

Comamonadaceae

-

-

399903

2.4

6.7

-4.3

1.20E-08

Spirochaetes

Spirochaetes

Spirochaetales

Spirochaetaceae

Treponema

-

242070

1.7

6.2

-4.4

1.53E-10

Proteobacteria

Gammaproteobacteria

Pseudomonadales

Pseudomonadaceae

Pseudomonas

-

6

ACCEPTED MANUSCRIPT

OTU ID

HC

#

AA-HC

q-value

Phylum

Class

Order

Family

Genus

Species

Bacteroidetes

Bacteroidia

1.9

7.3

-5.5

Bacteroidales

Prevotellaceae

Prevotella

-

114510

2.8

11.6

-8.9

6.38E-23

Proteobacteria

Gammaproteobacteria

Enterobacteriales

Enterobacteriaceae

-

-

100791

2.7

12.2

-9.6

1.98E-25

Proteobacteria

Betaproteobacteria

Burkholderiales

Comamonadaceae

-

-

4333897

3.6

15.9

-12.3

3.51E-32

Proteobacteria

Gammaproteobacteria

Enterobacteriales

Enterobacteriaceae

-

-

Firmicutes

Clostridia

Clostridiales

-

-

-

Prevotellaceae

Prevotella

-

Methylobacteriaceae

Methylobacterium

-

RI PT

851961

1.38E-13

3.8

17.7

-13.9

74407

1.3

16.9

-15.6

1.69E-44

Bacteroidetes

Bacteroidia

Bacteroidales

4396717

2.5

25.6

-23.1

8.72E-68

Proteobacteria

Alphaproteobacteria

Rhizobiales

4469032

21.9

45.3

-23.4

2.20E-34

Firmicutes

Bacilli

Lactobacillales

-

-

-

70628

0.4

24.3

-24.0

2.55E-37

Fusobacteria

Fusobacteriia

Fusobacteriales

Leptotrichiaceae

Leptotrichia

-

Bacteroidetes

Bacteroidia

Bacteroidales

55854

0.2

24.9

-24.7

0.0610

98258

0.7

26.7

-26.0

0.0186

Cyanobacteria

4C0d-2

269930

22.8

81.1

-58.3

Proteobacteria

684

1.6

61.8

-60.3

8.82E139 0.0001

114813

1.0

66.9

-66.0

4404577

372.8

441.8

-69.0

4480189

10.0

98.2

-88.2

4335578

221.3

332.8

4404220

336.1

4400260

2.3

Prevotella

-

-

-

-

Gammaproteobacteria

Pseudomonadales

Pseudomonadaceae

-

-

Firmicutes

Bacilli

Lactobacillales

-

-

-

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

-

Firmicutes

Clostridia

452.3

-116.2

5.61E-69

Proteobacteria

123.2

-120.9

0.0333

-235.4

0

mean relative abundance

Fusobacteria

Peptostreptococcaceae

Peptostreptococcus

-

Lactobacillales

Lactobacillaceae

Lactobacillus

zeae

Clostridia

Clostridiales

Mogibacteriaceae

-

-

Gammaproteobacteria

Pasteurellales

Pasteurellaceae

Actinobacillus

-

Fusobacteriia

Fusobacteriales

Leptotrichiaceae

Leptotrichia

-

Bacteroidales

Prevotellaceae

Prevotella

-

EP

246.5

Firmicutes

Clostridiales

Bacilli

TE D

Firmicutes

-111.6

1.23E259 6.19E-92

M AN U

Prevotellaceae

MLE1-12

Bacteroidetes

Bacteroidia

AC C

11.1

6.31E102 4.03E-23

SC

4477971

1.85E-37

4307006 #

#

AA

7

ACCEPTED MANUSCRIPT

Table S5: Specific bacterial taxa that differ in relative abundance between atopic non-asthmatic (ANA) and healthy control (HC) subjects. HC

#

ANAHC

ANA

4432431

963.7

55.5

908.3

828676

315.3

0.1

q-value

Phylum

Class

Order

0.0029

Proteobacteria

Gammaproteobacteria

Pasteurellales

315.3

0.0036

Fusobacteria

Fusobacteriia

Fusobacteriales

0.0302

Proteobacteria

Betaproteobacteria

Neisseriales

Family

Genus

Species

Pasteurellaceae

Aggregatibacter

segnis

Fusobacteriaceae

Fusobacterium

-

Neisseriaceae

-

-

Carnobacteriaceae

Granulicatella

-

RI PT

#

OTU ID

292.5

12.1

280.4

218.4

18.0

200.4

0.0143

Firmicutes

Bacilli

Lactobacillales

4306587

158.2

11.7

146.5

0.0758

Firmicutes

Clostridia

Clostridiales

Veillonellaceae

Selenomonas

-

134265

119.2

7.2

112.0

2.37E-152

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

-

4296424

141.9

37.8

104.1

0.0613

Actinobacteria

Actinobacteria

Actinomycetales

Actinomycetaceae

Actinomyces

-

Actinomycetales

Corynebacteriaceae

Corynebacterium

-

Bacteroidales

Prevotellaceae

Prevotella

melaninogenica

Bacteroidales

Porphyromonadaceae

Porphyromonas

-

M AN U

SC

978067 4483015

103.4

3.6

99.8

1.74E-111

Actinobacteria

Actinobacteria

105.2

5.6

99.6

0.0007

Bacteroidetes

Bacteroidia

4459993

102.7

4.5

98.2

0.0854

Bacteroidetes

Bacteroidia

976838

98.5

29.9

68.5

5.39E-95

Proteobacteria

Epsilonproteobacteria

Campylobacterales

Helicobacteraceae

Flexispira

2466322

65.7

0.2

65.6

0.0092

Firmicutes

Clostridia

Clostridiales

Peptostreptococcaceae

-

-

4388775

70.5

6.8

63.7

0.0047

Firmicutes

Clostridia

Clostridiales

Veillonellaceae

Veillonella

dispar

1015518

55.8

1.8

54.0

4.86E-58

Actinobacteria

Actinobacteria

Actinomycetales

Corynebacteriaceae

Corynebacterium

-

4469627

55.3

2.2

53.0

8.15E-63

Proteobacteria

Gammaproteobacteria

Pasteurellales

Pasteurellaceae

Haemophilus

-

4297222

47.9

4.3

43.6

1.36E-65

Firmicutes

Bacilli

Lactobacillales

Streptococcaceae

Streptococcus

-

995

44.7

2.5

42.2

0.0242

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

melaninogenica

1053321

75.5

36.6

38.9

3.63E-38

Proteobacteria

Gammaproteobacteria

Pseudomonadales

Moraxellaceae

Moraxella

-

4364176

47.9

11.3

36.6

1.91E-55

Firmicutes

Erysipelotrichi

Erysipelotrichales

Erysipelotrichaceae

Sharpea

-

267852

33.8

0.6

33.2

1.47E-27

AC C

EP

TE D

495067 4372058

Proteobacteria

Alphaproteobacteria

Sphingomonadales

Sphingomonadaceae

Sphingopyxis

alaskensis

4419634

33.3

2.3

31.0

0.0302

Fusobacteria

Fusobacteriia

Fusobacteriales

Leptotrichiaceae

Leptotrichia

-

1102

33.7

3.8

29.9

0.0583

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

nanceiensis

575

28.0

0.5

27.5

0.0014

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

melaninogenica

2438396

28.2

1.7

26.5

4.60E-36

Fusobacteria

Fusobacteriia

Fusobacteriales

Fusobacteriaceae

Fusobacterium

-

1044611

24.1

0.8

23.3

0.0434

Firmicutes

Clostridia

Clostridiales

Peptostreptococcaceae

-

-

4353757

24.0

1.3

22.7

0.0043

Proteobacteria

Gammaproteobacteria

Pasteurellales

Pasteurellaceae

Aggregatibacter

-

8

ACCEPTED MANUSCRIPT

787709

#

ANA

24.9

HC

#

ANAHC

q-value

Phylum

Class

Order

Family

Genus

Species

Actinomycetaceae

Actinomyces

-

Porphyromonadaceae

3.7

21.2

0.0192

Actinobacteria

Actinobacteria

Actinomycetales

0.0132

Bacteroidetes

Bacteroidia

Bacteroidales

4412630

22.1

1.3

20.8

876114

24.1

3.8

20.3

2.50E-32

Fusobacteria

Fusobacteriia

Fusobacteriales

71146

18.1

1.4

16.7

0.0714

Spirochaetes

Spirochaetes

Spirochaetales

4333897

32.1

15.9

16.2

1.12E-15

Proteobacteria

Gammaproteobacteria

Enterobacteriales

3859

20.6

4.9

15.7

9.42E-24

Spirochaetes

Spirochaetes

Spirochaetales

Porphyromonas

-

Leptotrichiaceae

-

-

Spirochaetaceae

Treponema

-

Enterobacteriaceae

-

-

Spirochaetaceae

Treponema

amylovorum

RI PT

OTU ID

14.7

1.9

12.8

2.17E-20

Proteobacteria

Betaproteobacteria

Neisseriales

Neisseriaceae

Kingella

-

18.9

7.0

11.9

4.85E-15

Proteobacteria

Betaproteobacteria

Neisseriales

Neisseriaceae

Neisseria

-

89

11.4

0.1

11.3

1.40E-05

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

-

1124080

11.5

1.0

10.5

7.55E-16

Bacteroidetes

Bacteroidia

Bacteroidales

-

-

-

M AN U

SC

63117 4373910

161677

10.4

0.3

10.1

1.07E-10

Proteobacteria

Gammaproteobacteria

Pseudomonadales

Moraxellaceae

Acinetobacter

-

4468500

12.4

2.5

9.9

1.12E-15

Firmicutes

Clostridia

Clostridiales

Mogibacteriaceae

Anaerovorax

-

124932

10.9

1.5

9.4

4.42E-15

Spirochaetes

Spirochaetes

Spirochaetales

Spirochaetaceae

Treponema

-

3931

10.7

2.1

8.6

1.20E-13

Spirochaetes

Spirochaetes

Spirochaetales

Spirochaetaceae

Treponema

-

1.11E-12

Proteobacteria

Gammaproteobacteria

Pasteurellales

Pasteurellaceae

Haemophilus

influenzae

Dethiosulfovibrionaceae

TG5

-

9.1

1.2

7.9

10.2

3.3

6.9

1.93E-09

Synergistetes

Synergistia

Synergistales

41911

6.6

0.2

6.4

0.0562

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

-

114510

17.7

11.6

6.1

0.0007

Proteobacteria

Gammaproteobacteria

Enterobacteriales

Enterobacteriaceae

-

-

4309323

6.8

1.0

5.8

0.0164

Proteobacteria

Gammaproteobacteria

Pasteurellales

Pasteurellaceae

Haemophilus

-

EP

TE D

956702 4460505

5.5

0.1

5.5

0.0005

Proteobacteria

Betaproteobacteria

Burkholderiales

Oxalobacteraceae

-

-

11.3

6.0

5.3

7.30E-05

Bacteroidetes

Bacteroidia

Bacteroidales

-

-

-

4351304

5.9

0.5

5.3

3.12E-08

Actinobacteria

Actinobacteria

Actinomycetales

Actinomycetaceae

Actinomyces

-

1052181

6.8

1.9

4.9

3.64E-07

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

nanceiensis

4374322

5.8

0.9

4.9

6.14E-08

Proteobacteria

Gammaproteobacteria

Pseudomonadales

Moraxellaceae

Acinetobacter

-

4419380

5.6

1.1

4.5

2.81E-07

Firmicutes

Erysipelotrichi

Erysipelotrichales

Erysipelotrichaceae

Bulleidia

p-1630-c5

4331815

4.8

0.5

4.3

7.69E-07

Proteobacteria

Alphaproteobacteria

Sphingomonadales

Sphingomonadaceae

-

-

517

4.3

0.2

4.1

0.0462

Bacteroidetes

Bacteroidia

Bacteroidales

Porphyromonadaceae

Porphyromonas

-

544313

4.3

0.3

4.0

7.71E-06

Proteobacteria

Gammaproteobacteria

Pseudomonadales

Pseudomonadaceae

Pseudomonas

-

328

4.0

0.4

3.6

0.0478

Bacteroidetes

Bacteroidia

Bacteroidales

Porphyromonadaceae

Porphyromonas

-

AC C

520222 4313608

9

ACCEPTED MANUSCRIPT

3799784

#

ANA

6.5

HC

#

ANAHC

q-value

Phylum

Class

Order

Family

Genus

Species

Enterobacteriaceae

-

-

Comamonadaceae

3.2

3.2

0.0031

Proteobacteria

Gammaproteobacteria

Enterobacteriales

0.0007

Proteobacteria

Betaproteobacteria

Burkholderiales

325027

5.1

2.0

3.1

43339

7.0

4.4

2.6

0.0562

Firmicutes

Clostridia

Clostridiales

2119418

2.7

0.1

2.6

0.0075

Proteobacteria

Gammaproteobacteria

Enterobacteriales

3600504

2.5

0.5

2.0

0.0031

Bacteroidetes

Bacteroidia

Bacteroidales

165489

3.4

1.5

1.9

0.0241

Proteobacteria

Gammaproteobacteria

Enterobacteriales

4454619

2.3

0.8

1.5

0.0353

Firmicutes

Clostridia

Clostridiales

4412991

2.0

0.5

1.5

0.0247

Proteobacteria

Betaproteobacteria

Neisseriales

4432889

2.1

0.6

1.5

0.0302

Actinobacteria

Actinobacteria

Actinomycetales

495451

1.5

0.4

1.2

0.0613

Bacteroidetes

Bacteroidia

Comamonas

-

Mogibacteriaceae

-

-

Enterobacteriaceae

-

-

Bacteroidaceae

Bacteroides

-

Enterobacteriaceae

-

-

Veillonellaceae

Selenomonas

-

Neisseriaceae

-

-

Intrasporangiaceae

-

-

Bacteroidales

Porphyromonadaceae

Porphyromonas

-

M AN U

SC

RI PT

OTU ID

51646

1.2

2.6

-1.4

0.0854

Proteobacteria

Gammaproteobacteria

Enterobacteriales

Enterobacteriaceae

-

-

1010329

0.9

2.6

-1.7

0.0173

Bacteroidetes

Flavobacteriia

Flavobacteriales

Flavobacteriaceae

Capnocytophaga

-

4426165

0.9

2.8

-1.9

0.0055

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

-

1595

1.4

3.6

-2.2

0.0051

Bacteroidetes

Bacteroidia

Bacteroidales

Porphyromonadaceae

Porphyromonas

-

8.94E-04

Fusobacteria

Fusobacteriia

Fusobacteriales

Fusobacteriaceae

Fusobacterium

-

Actinomycetaceae

Actinomyces

-

1.0

3.4

-2.4

0.2

3.0

-2.8

0.0001

Actinobacteria

Actinobacteria

Actinomycetales

4401957

1.9

6.2

-4.2

0.0920

Proteobacteria

Epsilonproteobacteria

Campylobacterales

Campylobacteraceae

Campylobacter

-

851961

0.3

7.3

-7.0

4.95E-10

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

-

245523

9.7

17.8

-8.0

3.05E-07

Bacteroidetes

Bacteroidia

Bacteroidales

-

-

-

EP

TE D

941024 4306356

0.1

8.2

-8.1

0.0908

Fusobacteria

Fusobacteriia

Fusobacteriales

Leptotrichiaceae

Leptotrichia

-

6.3

17.7

-11.4

1.35E-15

Firmicutes

Clostridia

Clostridiales

-

-

-

100791

0.1

12.2

-12.1

5.90E-09

Proteobacteria

Betaproteobacteria

Burkholderiales

Comamonadaceae

-

-

4443201

12.3

29.4

-17.1

9.85E-21

Bacteroidetes

Bacteroidia

Bacteroidales

Porphyromonadaceae

Tannerella

-

4396717

8.2

25.6

-17.4

1.91E-25

Proteobacteria

Alphaproteobacteria

Rhizobiales

Methylobacteriaceae

Methylobacterium

-

579608

0.1

18.7

-18.6

1.03E-10

Firmicutes

Bacilli

Lactobacillales

Streptococcaceae

Streptococcus

-

615020

15.9

36.7

-20.8

3.31E-24

Tenericutes

Mollicutes

Mycoplasmatales

Mycoplasmataceae

Mycoplasma

-

4344371

35.3

57.7

-22.4

1.86E-16

Proteobacteria

Alphaproteobacteria

Sphingomonadales

Sphingomonadaceae

Sphingomonas

-

70628

0.2

24.3

-24.1

4.42E-15

Fusobacteria

Fusobacteriia

Fusobacteriales

Leptotrichiaceae

Leptotrichia

-

4297119

3.7

39.8

-36.2

0.0968

Fusobacteria

Fusobacteriia

Fusobacteriales

Leptotrichiaceae

Leptotrichia

-

AC C

923032 4477971

10

ACCEPTED MANUSCRIPT

OTU ID

#

ANA

1048420

q-value

Phylum

Class

Order

Family

Genus

Species

36.8

-36.8

7.58E-09

Firmicutes

Bacilli

Lactobacillales

Streptococcaceae

Streptococcus

-

60.8

98.2

-37.4

1.28E-26

Firmicutes

Bacilli

Lactobacillales

Lactobacillaceae

7.0

46.4

-39.4

0.0149

Firmicutes

Bacilli

Lactobacillales

28118

0.1

52.9

-52.8

0.0029

Firmicutes

Clostridia

Clostridiales

684

1.9

61.8

-59.9

0.0103

Firmicutes

Bacilli

Lactobacillales

4376637

8.1

73.5

-65.4

0.0785

Spirochaetes

Spirochaetes

Spirochaetales

269930

0.1

81.1

-80.9

0.0065

Proteobacteria

Gammaproteobacteria

557974

10.1

96.8

-86.8

0.0812

Proteobacteria

Gammaproteobacteria

69782

1.7

122.8

-121.0

6.05E-101

Proteobacteria

Epsilonproteobacteria

3581175

3.0

133.8

-130.8

1.90E-137

Firmicutes

Clostridia

93.5

260.4

-166.9

7.86E-226

Bacteroidetes

Bacteroidia

4331006

30.5

205.6

-175.1

0

Bacteroidetes

Bacteroidia

4377418

215.8

413.2

-197.4

4.13E-185

Firmicutes

Clostridia

4307006

3.5

246.5

-242.9

5.60E-204

Bacteroidetes

Bacteroidia

2613485

4.7

428.8

-424.2

0

Bacteroidetes

Bacteroidia

252843 8.1 450.7 -442.6 mean relative abundance

0

Fusobacteria

Fusobacteriia

zeae

Streptococcus

-

Veillonellaceae

Selenomonas

-

-

-

-

Spirochaetaceae

Treponema

-

Pseudomonadales

Pseudomonadaceae

-

-

Pseudomonadales

Pseudomonadaceae

Pseudomonas

-

Campylobacterales

Helicobacteraceae

Helicobacter

-

Clostridiales

Veillonellaceae

-

-

Bacteroidales

Prevotellaceae

Prevotella

-

Bacteroidales

Prevotellaceae

Prevotella

-

Clostridiales

Tissierellaceae

Parvimonas

-

Bacteroidales

Prevotellaceae

Prevotella

-

Bacteroidales

Porphyromonadaceae

Porphyromonas

-

Fusobacteriales

Leptotrichiaceae

Leptotrichia

-

EP AC C

#

TE D

1004910

Lactobacillus

Streptococcaceae

RI PT

4480189

ANAHC

SC

0.1

#

M AN U

123320

HC

11

ACCEPTED MANUSCRIPT

Table S6: Specific bacterial taxa which differ in relative abundance between atopic asthmatic (AA) and atopic non-asthmatic (ANA) subjects. OTU ID

#

AA

#

AA-ANA

Class

Order

Family

Genus

Species

503.7

8.1

495.6

0.0255

Fusobacteria

Fusobacteriia

Fusobacteriales

Leptotrichiaceae

Leptotrichia

-

4307790

518.8

119.1

399.7

0.0255

Firmicutes

Clostridia

Clostridiales

Veillonellaceae

Selenomonas

-

4338372

363.6

86.0

277.6

0.0132

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

-

4406393

243.8

1.8

242.0

0.0014

Proteobacteria

Gammaproteobacteria

Pasteurellales

Pasteurellaceae

Haemophilus

-

2438396

192.5

28.2

164.3

0

Fusobacteria

Fusobacteriia

Fusobacteriales

Fusobacteriaceae

Fusobacterium

-

4405869

196.0

57.4

138.6

0.0711

Fusobacteria

Fusobacteriia

Fusobacteriales

Fusobacteriaceae

Fusobacterium

-

4393181

141.3

3.4

137.9

0.0004

Tenericutes

Mollicutes

Mycoplasmatales

Mycoplasmataceae

Mycoplasma

-

4382476

126.9

7.0

119.9

0.0014

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

-

SC

q-value

RI PT

Phylum

252843

M AN U

ANA

4297119

93.6

3.7

89.9

0.0014

Fusobacteria

Fusobacteriia

Fusobacteriales

Leptotrichiaceae

Leptotrichia

-

525942

94.6

16.9

77.6

0.0194

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

melaninogenica

4426165

59.0

0.9

58.1

0.0373

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

-

4318284

60.2

6.1

54.1

0.0599

Firmicutes

Clostridia

Clostridiales

Veillonellaceae

Dialister

-

923032

31.0

0.1

30.9

0.0255

Fusobacteria

Fusobacteriia

269930

22.8

0.1

22.6

0.0256

Proteobacteria

4452538

24.3

4.5

19.7

0.0077

Fusobacteria

4307309

19.5

7.5

12.0

1.93E-18

Fusobacteria

4468500

24.4

12.4

12.0

1.75E-14

Firmicutes

495451

11.3

1.5

9.7

1.92E-18

Bacteroidetes

4346863

10.9

1.2

9.7

0.0094

Bacteroidetes

245523

18.9

9.7

9.1

6.69E-11

69782

10.4

1.7

8.7

1.25E-16

Leptotrichiaceae

Leptotrichia

-

Pseudomonadales

Pseudomonadaceae

-

-

Fusobacteriia

Fusobacteriales

Fusobacteriaceae

Fusobacterium

-

Fusobacteriia

Fusobacteriales

Fusobacteriaceae

Fusobacterium

-

Clostridia

Clostridiales

Mogibacteriaceae

Anaerovorax

-

Bacteroidia

Bacteroidales

Porphyromonadaceae

Porphyromonas

-

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

-

Bacteroidetes

Bacteroidia

Bacteroidales

-

-

-

Proteobacteria

Epsilonproteobacteria

Campylobacterales

Helicobacteraceae

Helicobacter

-

AC C

EP

TE D

Fusobacteriales

Gammaproteobacteria

28118

7.7

0.1

7.5

0.0255

Firmicutes

Clostridia

Clostridiales

Veillonellaceae

Selenomonas

-

1580

7.1

0.6

6.5

0.0808

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

melaninogenica

4426163

7.7

1.6

6.1

0.0425

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

-

1033687

5.1

0.3

4.8

0.0689

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

-

92316

6.0

1.5

4.5

3.52E-08

Firmicutes

Clostridia

Clostridiales

Lachnospiraceae

-

-

62513

4.4

0.1

4.3

3.52E-05

Firmicutes

Clostridia

Clostridiales

Lachnospiraceae

Moryella

-

12

ACCEPTED MANUSCRIPT

OTU ID

#

AA

#

ANA

AA-ANA

q-value

Phylum

Class

Order

Family

Genus

Species

Firmicutes

Bacilli

Lactobacillales

Streptococcaceae

Streptococcus

-

Flavobacteriaceae

Capnocytophaga

-

Neisseriaceae

Neisseria

subflava

4.6

0.9

3.7

989074

3.4

0.1

3.2

0.0002

Bacteroidetes

Flavobacteriia

Flavobacteriales

1304

3.0

0.1

2.9

0.0044

Proteobacteria

Betaproteobacteria

Neisseriales

4331815

7.5

4.8

2.7

0.0212

Proteobacteria

Alphaproteobacteria

Sphingomonadales

Sphingomonadaceae

-

-

100791

2.7

0.1

2.5

0.0009

Proteobacteria

Betaproteobacteria

Burkholderiales

Comamonadaceae

-

-

55917

3.6

1.1

2.5

0.0004

Bacteroidetes

Bacteroidia

Bacteroidales

-

-

-

831

2.5

0.3

2.2

0.0897

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

-

2377731

3.2

1.1

2.0

0.0029

Firmicutes

Clostridia

Clostridiales

Veillonellaceae

Selenomonas

-

1465

2.1

0.1

2.0

0.0033

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

intermedia

4353264

3.8

1.8

2.0

0.0135

Proteobacteria

Alphaproteobacteria

Caulobacterales

Caulobacteraceae

-

-

4414261

2.1

0.5

1.6

0.0044

Firmicutes

Bacilli

Lactobacillales

Lactobacillaceae

Lactobacillus

-

1066621

2.2

0.6

1.6

0.0925

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

melaninogenica

1188

1.4

0.1

1.4

0.0449

Fusobacteria

Fusobacteriia

Fusobacteriales

Leptotrichiaceae

Leptotrichia

-

104313

1.2

0.3

0.9

0.0696

Proteobacteria

Gammaproteobacteria

Pseudomonadales

Pseudomonadaceae

Pseudomonas

-

SC

M AN U

0.0787

RI PT

523025

0.6

1.7

-1.1

0.0190

Firmicutes

Bacilli

Bacillales

Exiguobacteraceae

-

-

0.1

1.3

-1.2

0.0894

Proteobacteria

Gammaproteobacteria

Pasteurellales

Pasteurellaceae

Aggregatibacter

segnis

57758

0.5

1.7

-1.2

0.0047

Bacteroidetes

Flavobacteriia

Flavobacteriales

Flavobacteriaceae

Flavobacterium

columnare

4483174

0.7

2.0

-1.3

0.0092

Fusobacteria

Fusobacteriia

Fusobacteriales

Leptotrichiaceae

Leptotrichia

-

TE D

540269 4423236

0.4

2.1

-1.7

8.43E-05

Actinobacteria

Actinobacteria

Actinomycetales

Gordoniaceae

Gordonia

-

1.5

3.3

-1.8

0.0043

Cyanobacteria

Chloroplast

Streptophyta

-

-

-

4375000

0.5

2.7

-2.2

2.70E-06

Proteobacteria

Gammaproteobacteria

Enterobacteriales

Enterobacteriaceae

-

-

470724

0.4

3.8

-3.4

2.37E-10

Proteobacteria

Betaproteobacteria

Neisseriales

Neisseriaceae

Neisseria

-

3.65E-11

Betaproteobacteria

Burkholderiales

Comamonadaceae

Hydrogenophaga

-

Streptococcaceae

Streptococcus

-

AC C

EP

671258 3359884

339015

0.3

4.1

-3.8

669486

0.8

5.3

-4.4

2.19E-13

Firmicutes

Bacilli

Lactobacillales

1052181

1.8

6.8

-5.0

4.58E-13

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

nanceiensis

4335450

1.4

6.5

-5.1

2.75E-14

Fusobacteria

Fusobacteriia

Fusobacteriales

Leptotrichiaceae

-

-

4343627

0.3

5.7

-5.4

1.07E-14

Bacteroidetes

Bacteroidia

Bacteroidales

Bacteroidaceae

Bacteroides

fragilis

Proteobacteria

903686

2.8

8.3

-5.5

0.0689

Proteobacteria

Gammaproteobacteria

Pasteurellales

Pasteurellaceae

Aggregatibacter

segnis

4440670

19.8

25.3

-5.5

0.0055

Firmicutes

Clostridia

Clostridiales

Veillonellaceae

Veillonella

-

13

ACCEPTED MANUSCRIPT

OTU ID

#

AA

#

ANA

AA-ANA

q-value

Phylum

Class

Order

Family

Genus

Species

Proteobacteria

Gammaproteobacteria

Enterobacteriales

Enterobacteriaceae

-

-

Pasteurellaceae

Haemophilus

-

0.9

6.5

-5.6

4309323

1.1

6.8

-5.7

0.0588

Proteobacteria

Gammaproteobacteria

Pasteurellales

242070

1.7

8.2

-6.5

2.34E-18

Proteobacteria

Gammaproteobacteria

Pseudomonadales

Pseudomonadaceae

Pseudomonas

-

521996

0.3

7.1

-6.9

0.0007

Firmicutes

Bacilli

Lactobacillales

Streptococcaceae

Streptococcus

-

571873

0.4

7.6

-7.2

1.32E-18

Proteobacteria

Betaproteobacteria

Burkholderiales

Comamonadaceae

-

-

Spirochaetaceae

Treponema

-

Micrococcaceae

-

-

6.52E-17

RI PT

3799784

1.4

8.9

-7.5

1.36E-22

Spirochaetes

Spirochaetes

Spirochaetales

0.3

8.1

-7.8

6.36E-19

Actinobacteria

Actinobacteria

Actinomycetales

898611

1.0

9.3

-8.3

3.23E-25

Firmicutes

Clostridia

Clostridiales

Lachnospiraceae

Butyrivibrio

-

4428313

6.5

15.3

-8.8

3.16E-16

Firmicutes

Bacilli

Lactobacillales

Lactobacillaceae

Lactobacillus

-

SC

95168 543491

11.6

20.6

-9.0

3.10E-11

Spirochaetes

Spirochaetes

Spirochaetales

Spirochaetaceae

Treponema

amylovorum

0.1

9.1

-9.0

0.0615

Proteobacteria

Gammaproteobacteria

Pasteurellales

Pasteurellaceae

Haemophilus

influenzae

4303688

4.1

14.5

-10.4

8.09E-26

Proteobacteria

Betaproteobacteria

Burkholderiales

Burkholderiaceae

Lautropia

-

561636

0.1

10.9

-10.8

0.0232

Firmicutes

Bacilli

Lactobacillales

Streptococcaceae

Streptococcus

-

63117

1.9

14.7

-12.9

3.41E-39

Proteobacteria

Betaproteobacteria

Neisseriales

Neisseriaceae

Kingella

-

4386920

0.0

15.0

-15.0

0.0255

Proteobacteria

Gammaproteobacteria

Pseudomonadales

Pseudomonadaceae

Pseudomonas

-

2510538

1.6

17.5

-15.9

0.0254

Firmicutes

Bacilli

Lactobacillales

Carnobacteriaceae

-

-

4430639

7.5

24.6

-17.1

0.0254

Proteobacteria

Gammaproteobacteria

Pasteurellales

Pasteurellaceae

Haemophilus

parainfluenzae

353459

0.0

17.4

-17.4

0.087

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

copri

4443200

0.8

19.3

-18.5

8.79E-45

Bacteroidetes

Flavobacteriia

Flavobacteriales

Flavobacteriaceae

Capnocytophaga

ochracea

4412630

2.0

22.1

-20.1

0.0271

Bacteroidetes

Bacteroidia

Bacteroidales

Porphyromonadaceae

Porphyromonas

-

171157

8.6

31.3

-22.7

1.72E-57

Proteobacteria

Epsilonproteobacteria

Campylobacterales

Helicobacteraceae

Flexispira

-

259631

0.0

23.0

-23.0

0.0686

Firmicutes

Clostridia

Clostridiales

Lachnospiraceae

Ruminococcus

gnavus

0.0043

Fusobacteriia

Fusobacteriales

Fusobacteriaceae

Fusobacterium

-

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

melaninogenica

AC C

EP

TE D

M AN U

3859 956702

245

0.2

25.1

-25.0

575

2.5

28.0

-25.5

0.032

4301566

10.6

37.3

-26.7

0.0172

Firmicutes

Bacilli

Lactobacillales

Streptococcaceae

Streptococcus

-

886735

21.0

47.9

-26.9

1.18E-47

Firmicutes

Bacilli

Lactobacillales

Aerococcaceae

Alloiococcus

-

267852

5.9

33.8

-27.9

1.99E-82

Proteobacteria

Alphaproteobacteria

Sphingomonadales

Sphingomonadaceae

Sphingopyxis

alaskensis

4469032

21.9

50.2

-28.3

1.74E-50

Firmicutes

Bacilli

Lactobacillales

-

-

-

995

4.0

44.7

-40.6

0.010

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

melaninogenica

Fusobacteria

14

ACCEPTED MANUSCRIPT

OTU ID

#

ANA

AA-ANA

q-value

Phylum

Class

Order

Family

Genus

Species

2.03E-123

Firmicutes

Bacilli

Lactobacillales

Streptococcaceae

Streptococcus

-

Erysipelotrichaceae

Sharpea

-

Lactobacillaceae

Lactobacillus

zeae

7.1

47.9

-40.9

4364176

1.3

47.9

-46.6

1.99E-97

Firmicutes

Erysipelotrichi

Erysipelotrichales

4480189

10.0

60.8

-50.8

1.45E-151

Firmicutes

Bacilli

Lactobacillales

1015518

3.1

55.8

-52.7

5.83E-143

Actinobacteria

Actinobacteria

Actinomycetales

Corynebacteriaceae

Corynebacterium

-

4466150

5.5

63.3

-57.9

5.08E-05

Proteobacteria

Gammaproteobacteria

Pasteurellales

Pasteurellaceae

Haemophilus

-

4337755

30.6

94.8

-64.2

0.0255

Firmicutes

Bacilli

Gemellales

1925094

8.2

75.5

-67.4

5.19E-204

Actinobacteria

Actinobacteria

Actinomycetales

260397

3.7

77.7

-74.0

2.51E-191

Firmicutes

Erysipelotrichi

Erysipelotrichales

4372058

17.6

105.2

-87.6

0.026

Bacteroidetes

Bacteroidia

Bacteroidales

2.41E-263 1.94E-70

4459993

3.5

102.7

-99.1

0.0010

495067

1.8

103.4

-101.7

134265

1.0

119.2

4306587

12.7

4400869

10.9

891034

31.5

-

-

Micrococcaceae

-

-

Erysipelotrichaceae

Allobaculum

-

Prevotellaceae

Prevotella

melaninogenica

SC

-91.2 -92.2

Proteobacteria

Epsilonproteobacteria

Campylobacterales

Helicobacteraceae

Flexispira

-

Firmicutes

Clostridia

Clostridiales

Mogibacteriaceae

-

-

Bacteroidetes

Bacteroidia

Bacteroidales

Porphyromonadaceae

Porphyromonas

-

2.20E-171

Actinobacteria

Actinobacteria

Actinomycetales

Corynebacteriaceae

Corynebacterium

-

-118.2

9.40E-137

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

-

158.2

-145.5

0.0156

Firmicutes

Clostridia

Clostridiales

Veillonellaceae

Selenomonas

-

167.7

-156.8

0.0960

SR1

-

-

-

-

-

270.0

-238.5

0.0806

Proteobacteria

Gammaproteobacteria

Pasteurellales

Pasteurellaceae

Aggregatibacter

-

-872.4

0.0006

Proteobacteria

Gammaproteobacteria

Pasteurellales

Pasteurellaceae

Aggregatibacter

segnis

4432431 91.3 963.7 mean relative abundance

M AN U

98.5 313.5

TE D

7.3 221.3

Gemellaceae

EP

976838 4335578

RI PT

4297222

AC C

#

#

AA

15

ACCEPTED MANUSCRIPT

0.3 (0.0 - 1.0) 0.0 (0.0 - 0.2) -0.2 (-0.6 - 0.0) 121.5 (49.0 - 231.3) 6 (3-7) 27% 39%

<0.05 NS NS <0.05 € NS € NS € NS

SC

<0.05 NS NS <0.05 € NS € NS NS NS NS <0.05 NS NS NS NS NS NS <0.01 <0.01 <0.001 NS NS NS NS

M AN U

1.3 (0.4 - 3.8) 0.0 (0.0 - 0.4) 0.0 (-0.7 - 0.1) 323.5 (184.3 - 560.5) 5 (2-10) 40% 60%

p-value#

TE D

Sputum eosinophils (% Baseline) Sputum eosinophils (% post-ICS) Change in sputum eosinophils Serum IgE (IU/mL) Number of positive sIgE¢ % Eczema/Allergic dermatitis§ % Allergic rhinitis§

T2-low asthmatics (n = 30) 37 (27-43) 9 (5-22) 23 (14-29) 0.5 (0.2 - 0.7) 47% 60% 26 (23 - 29) 86 (76 - 96) 105 (83 - 105) 7 (5 - 12) 95 (90 - 106) 103 (98 - 115) 1 (-6 - 4) 0 (-5 - 6) 1.3 (0.4 - 2.7) 1.9 (0.3 - 5.8) 0.3 (-0.2 - 1.5) 130 (100 - 293) 2.7 (1.3 - 4.2) 56.1 (49.7 - 62.0) 55.8 (32.5 - 65.2) 49.4 (25.1 - 60.3) -9.6 (-18-1.8)

EP

Age (yrs) Age of Asthma diagnosis (yrs) Duration of Asthma (yrs) ACQ Score (Baseline)* % Male % White BMI FEV1 % pre-Ab (Baseline) FEV1 % post-Ab (Baseline) Change in FEV % (Baseline) FEV1 % pre-Ab (post-ICS) FEV1 % post-Ab (post-ICS) Change in FEV % pre-Ab Change in FEV % post-Ab PC20 (Baseline) PC20 (post ICS) Change in PC20 Blood eosinophils (absolute) Blood eosinophils (%) Blood neutrophils (%) Sputum neutrophils (% Baseline) Sputum neutrophils (% post-ICS) Change in sputum neutrophils

T2-high asthmatics (n = 10) 28 (24-33) 9 (5-24) 18 (9-23) 1.0 (0.8 - 1.3) 50% 60% 26 (23 - 30) 91 (68 - 110) 102 (93 - 114) 15 (7 - 25) 88 (79 - 97) 98 (91 - 105) 3 (-4 - 13) 1 (-3 - 5) 0.9 (0.2 - 3.8) 6.1 (1.6 - 12.4) 4.9 (1.2 - 11.5) 400 (245 - 600) 6.9 (4.6 - 10.3) 53.5 (43.7 - 60.5) 53.6 (33.9 - 64.9) 49.5 (32.8 - 59.9) 0.0 (-24.9 - 16.0)

AC C

Variable

RI PT

Table S7: Clinical and immunological comparison of T2-high compared to T2-low asthma patients.

¢

All values are medians (IQR). *ACQ, Asthma Control Questionnaire. Number of positive specific IgE (sIgE >0.35 kU/l) from a total of 12 aeroallergens tested by § # ImmunoCap assay. Subjects with a positive history of allergic conditions specified (self-reported). Statistical significance was determined using Mann-Whitney or € Fisher’s exact test.

16

ACCEPTED MANUSCRIPT

Table S8: Clinical and immunological comparison of ICS-responders compared to ICS non-responders. p-value#

RI PT

NS NS NS NS NS € NS € NS NS <0.05 NS <0.05 NS NS NS NS NS NS NS NS <0.01 NS NS NS NS NS NS NS NS NS NS

M AN U

SC

ICS non-responders (n = 10) 29 (20-39) 5 (2-9) 21 (13-26) 0.6 (0.1 - 1.0) 0.2 (0.0 - 0.7) 40% 70% 26 (24 - 28) 99 (94 - 110) 105 (100 - 114) 6 (5 - 7) 95 (90 - 106) 103 (98 - 115) 10 (5 - 14) 1 (-6 - 4) 0 (-5 - 6) 0.4 (-5.3 – 3.3) 1.9 (0.3 - 4.1) 1.9 (0.3 - 5.8) 0.3 (-0.2 - 1.5) 155 (100 - 315) 3.0 (1.0 - 5.1) 58.0 (50.4 - 64.3) 56.1 (35.0 - 66.7) 49.4 (25.1 - 60.3) -9.6 (-18-1.8) 0.4 (0.0 - 0.7) 0.0 (0.0 - 0.2) -0.2 (-0.6 - 0.0) 188.0 (66.8 - 310.8)

TE D

AC C

Age (yrs) Age of Asthma diagnosis (yrs) Duration of Asthma (yrs) ACQ Score (Baseline)* ACQ Score (post ICS)* % Male % White BMI FEV1 % pre-Ab (Baseline) FEV1 % post-Ab (Baseline) Change in FEV with Ab % (Baseline) FEV1 % pre-Ab (post-ICS) FEV1 % post-Ab (post-ICS) Change in FEV % with Ab (post-ICS) Change in FEV % pre-Ab Change in FEV % post-Ab % Change from baseline in pre-Alb FEV1 PC20 (Baseline) PC20 (post ICS) Change in PC20 Blood eosinophils (absolute) Blood eosinophils (%) Blood neutrophils (%) Sputum neutrophils (% Baseline) Sputum neutrophils (% post-ICS) Change in sputum neutrophils Sputum eosinophils (% Baseline) Sputum eosinophils (% post-ICS) Change in sputum eosinophils Serum IgE (IU/mL)

ICS responders (n = 15) 32 (27-43) 8 (5-22) 25 (18-27) 0.8 (0.2 - 1.2) 0.3 (0.0 - 0.5) 40% 47% 26 (23 - 29) 86 (71 - 95) 101 (85 - 105) 12 (8 - 15) 88 (79 - 97) 98 (91 - 105) 12 (5 - 14) 3 (-4 - 13) 1 (-3 - 5) 3.2 (-4.7 – 11.7) 0.8 (0.3 - 1.2) 6.1 (1.6 - 12.4) 4.9 (1.2 - 11.5) 200 (100 - 400) 3.8 (1.3 - 6.7) 53.3 (49.8 - 61.0) 51.5 (25.6 - 64.2) 49.5 (32.8 - 59.9) 0.0 (-24.9 - 16.0) 0.2 (0.0 - 1.0) 0.0 (0.0 - 0.4) 0.0 (-0.7 - 0.1) 153.0 (93.0 - 261.0)

EP

Variable

#



All values are medians (IQR). *ACQ, Asthma Control Questionnaire. Statistical significance was determined using Mann-Whitney or Fisher’s exact test.

Table S9: Specific taxa differentially expressed in ICS non-responders (ICS non-R) compared to ICS responders (ICS-R) at baseline.

18

ACCEPTED MANUSCRIPT

OTU ID

ICS # non-R

#

ICS-R

ICS nonR- R

q-value

Phylum

Class

Order

Family

Genus

Species

9157.6

0.5

9157.1

1.05E-104

Actinobacteria

Actinobacteria

Actinomycetales

Microbacteriaceae

-

-

1111.8

0.7

1111.1

1.29E-82

Proteobacteria

Gammaproteobacteria

Pasteurellales

Pasteurellaceae

Haemophilus

-

4459021

221

7.1

213.9

0.0658

Bacteroidetes

Bacteroidia

Bacteroidales

Porphyromonadaceae

Tannerella

-

4465803

210.2

3.7

206.5

1.06E-126

Bacteroidetes

Bacteroidia

Bacteroidales

Porphyromonadaceae

Porphyromonas

-

RI PT

12297 4406393

164

3.8

160.2

7.45E-112

Fusobacteria

Fusobacteriia

Fusobacteriales

Leptotrichiaceae

Leptotrichia

-

154

8.7

145.3

3.64E-140

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

-

2654263

116.4

4.5

111.9

0.0129

Proteobacteria

Gammaproteobacteria

Pasteurellales

Pasteurellaceae

Haemophilus

parainfluenzae

4437399

137.4

58.4

79

2.55E-50

Spirochaetes

Spirochaetes

Spirochaetales

Spirochaetaceae

Treponema

-

4404220

410.2

333.7

76.5

5.54E-12

Proteobacteria

Gammaproteobacteria

Pasteurellales

Pasteurellaceae

Actinobacillus

-

14920

60.6

0.2

60.4

2.48E-14

Firmicutes

Bacilli

Gemellales

Gemellaceae

Gemella

-

73875

84.6

26.9

57.7

4.06E-47

Spirochaetes

Spirochaetes

Spirochaetales

Spirochaetaceae

Treponema

-

71146

108.6

52.6

56

1.06E-30

Spirochaetes

Spirochaetes

Spirochaetales

Spirochaetaceae

Treponema

-

2613485

60.8

6.3

54.5

2.20E-58

Bacteroidetes

Bacteroidia

Bacteroidales

Porphyromonadaceae

Porphyromonas

-

Flavobacteriia

M AN U

SC

923032 4426165

54.4

0.6

53.8

4.08E-26

Bacteroidetes

Flavobacteriales

Flavobacteriaceae

Capnocytophaga

ochracea

548730

114.4

62.3

52.1

3.75E-24

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

-

949816

37.4

2.6

34.8

0.0389

Firmicutes

Bacilli

Lactobacillales

Streptococcaceae

Streptococcus

-

4376637

82.6

47.8

34.8

1.24E-14

Spirochaetes

Spirochaetes

Spirochaetales

Spirochaetaceae

Treponema

-

968873

33.8

0.1

33.7

1.63E-07

Bacteroidetes

Flavobacteriia

325027

33.6

1.4

32.2

0.0454

4459265

42.8

12.9

29.9

1013316

29.8

0.9

28.9

4480189

27.2

0.4

26.8

4468500

58.2

36.4

21.8

4446973

21.8

0.1

21.7

4313722

20.4

0.4

20

4449458

23.6

5.6

4422512

19.6

886735

20.2

TE D

4419376

Weeksellaceae

-

-

Betaproteobacteria

Burkholderiales

Comamonadaceae

Comamonas

-

2.28E-25

Bacteroidetes

Bacteroidia

Bacteroidales

Paraprevotellaceae

Prevotella

-

8.09E-23

Firmicutes

Clostridia

Clostridiales

Lachnospiraceae

-

-

3.05E-15

Firmicutes

Bacilli

Lactobacillales

Lactobacillaceae

Lactobacillus

zeae

6.44E-08

AC C

EP

Flavobacteriales

Proteobacteria

Firmicutes

Clostridia

Clostridiales

Mogibacteriaceae

Anaerovorax

-

0.0716

Bacteroidetes

Bacteroidia

Bacteroidales

Porphyromonadaceae

Paludibacter

-

0.0059

Fusobacteria

Fusobacteriia

Fusobacteriales

Fusobacteriaceae

Fusobacterium

-

18

0.0545

Proteobacteria

Gammaproteobacteria

Pseudomonadales

Moraxellaceae

Acinetobacter

-

2.1

17.5

5.90E-19

Tenericutes

Mollicutes

Mycoplasmatales

Mycoplasmataceae

Mycoplasma

-

3.6

16.6

2.32E-17

Firmicutes

Bacilli

Lactobacillales

Aerococcaceae

Alloiococcus

-

19

ACCEPTED MANUSCRIPT

OTU ID

ICS # non-R

ICS nonR- R

#

ICS-R

q-value

Phylum

Class

Order

Family

Genus

Species

17.6

1.3

16.3

6.34E-17

Actinobacteria

Actinobacteria

Actinomycetales

Actinomycetaceae

Actinomyces

-

4329788

33.2

19.3

13.9

6.48E-06

Firmicutes

Clostridia

Clostridiales

-

-

-

269930

43.2

32.5

10.7

0.0181

1157

11

1.1

9.9

8.65E-11

4299087

8.2

0.3

7.9

1609

7.6

0.3

1076969

11.8

4306773

9.8

4329957

5.8

137183

RI PT

46

Gammaproteobacteria

Pseudomonadales

Pseudomonadaceae

-

-

Bacilli

Lactobacillales

Streptococcaceae

Streptococcus

-

0.0389

Bacteroidetes

Flavobacteriia

Flavobacteriales

Flavobacteriaceae

Capnocytophaga

ochracea

7.3

1.60E-06

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

melaninogenica

4.5

7.3

2.31E-05

Firmicutes

Bacilli

Lactobacillales

Streptococcaceae

Streptococcus

-

3.3

6.5

2.74E-05

Fusobacteria

Fusobacteriia

Fusobacteriales

Leptotrichiaceae

Leptotrichia

-

0.2

5.6

8.21E-05

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

-

6.8

1.4

5.4

1.37E-05

Actinobacteria

Actinobacteria

Bifidobacteriales

Bifidobacteriaceae

-

-

886264

2.6

0.6

2

0.0398

Actinobacteria

Actinobacteria

Actinomycetales

Micrococcaceae

Rothia

mucilaginosa

M AN U

SC

Proteobacteria Firmicutes

0.8

3.6

-2.8

0.0550

Firmicutes

Clostridia

Clostridiales

Mogibacteriaceae

-

-

0.6

3.6

-3

0.0390

Fusobacteria

Fusobacteriia

Fusobacteriales

Fusobacteriaceae

Fusobacterium

-

968954

0.6

4.2

-3.6

0.0389

Firmicutes

Bacilli

Lactobacillales

Streptococcaceae

Streptococcus

-

1015518

1.2

4.9

-3.7

0.0176

Actinobacteria

Actinobacteria

Actinomycetales

Corynebacteriaceae

Corynebacterium

-

TE D

43339 645055

2.2

6

-3.8

0.0325

Actinobacteria

Actinobacteria

Actinomycetales

Actinomycetaceae

Actinomyces

-

4399781

1.2

5.1

-3.9

0.0130

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

-

989074

1.2

6.4

-5.2

0.0016

Bacteroidetes

Flavobacteriia

Flavobacteriales

Flavobacteriaceae

Capnocytophaga

-

1580

2.2

7.6

-5.4

0.0022

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

melaninogenica

0.0545

Firmicutes

Bacilli

Lactobacillales

Streptococcaceae

Streptococcus

-

Clostridia

Clostridiales

Lachnospiraceae

-

-

591583

0.2

5.6

-5.4

4341205

3.8

9.4

-5.6

965500

2.4

8.9

-6.5

254888

3.6

10.5

-6.9

1935279

0.8

9.1

-8.3

851935

0.4

9

-8.6

0.0916

610111

3.4

13.1

-9.7

3.71E-06

851824

1.2

12.1

-10.9

7.99E-07

124932

1.8

12.8

-11

3.19E-07

4306587

0.2

11.5

-11.3

0.0325

0.0056

EP

4306356

Firmicutes

Firmicutes

Bacilli

Lactobacillales

Streptococcaceae

Streptococcus

-

0.0005

Proteobacteria

Betaproteobacteria

Burkholderiales

Comamonadaceae

-

-

0.0125

Firmicutes

Bacilli

Lactobacillales

Streptococcaceae

Streptococcus

-

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

-

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

-

Firmicutes

Erysipelotrichi

Erysipelotrichales

Erysipelotrichaceae

Bulleidia

-

Spirochaetes

Spirochaetes

Spirochaetales

Spirochaetaceae

Treponema

-

Firmicutes

Clostridia

Clostridiales

Veillonellaceae

Selenomonas

-

AC C 0.0004

20

ACCEPTED MANUSCRIPT

OTU ID

#

ICS-R

ICS nonR- R

q-value

Phylum

Class

Order

Family

Genus

Species

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

-

Fusobacteria

Fusobacteriia

Fusobacteriales

Leptotrichiaceae

Leptotrichia

-

6.4

18.2

-11.8

1.15E-06

4297119

0.2

15

-14.8

0.0131

4373910

0.8

16

-15.2

8.52E-08

Proteobacteria

Betaproteobacteria

Neisseriales

Neisseriaceae

Neisseria

-

4440670

2.6

19.9

-17.3

3.30E-11

Firmicutes

Clostridia

Clostridiales

Veillonellaceae

Veillonella

-

1044611

1.6

23.9

-22.3

1.61E-12

Firmicutes

Clostridia

Clostridiales

Peptostreptococcaceae

-

-

537098

125.6

150.9

-25.3

0.0020

Firmicutes

Clostridia

Clostridiales

Lachnospiraceae

-

-

0.0155

Bacteroidetes

Bacteroidia

Bacteroidales

Paraprevotellaceae

Prevotella

-

Firmicutes

Clostridia

Clostridiales

Lachnospiraceae

Oribacterium

-

Actinobacteria

Actinobacteria

Actinomycetales

Micrococcaceae

Rothia

dentocariosa

RI PT

22951

1.4

28.5

-27.1

25.8

57.8

-32

3.94E-15

4466006

5.2

46.9

-41.7

0.0863

4313608

12.8

56.4

-43.6

1.16E-27

Bacteroidetes

Bacteroidia

Bacteroidales

-

-

-

245523

0.2

47.7

-47.5

1.07E-06

Bacteroidetes

Bacteroidia

Bacteroidales

-

-

-

M AN U

SC

86 4310395

0.2

52.1

-51.9

0.0233

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

-

21.2

98.8

-77.6

1.93E-49

Fusobacteria

Fusobacteriia

Fusobacteriales

Fusobacteriaceae

Fusobacterium

-

4330849

6.6

92.4

-85.8

2.08E-48

SR1

-

-

-

-

-

4344371

2

94.1

-92.1

4.52E-32

Proteobacteria

Alphaproteobacteria

Sphingomonadales

Sphingomonadaceae

Sphingomonas

-

1053321

17

110.2

-93.2

5.59E-60

Proteobacteria

Gammaproteobacteria

Pseudomonadales

Moraxellaceae

Moraxella

-

4335578

117.4

236

-118.6

6.17E-50

Firmicutes

Clostridia

Clostridiales

Mogibacteriaceae

-

-

4404577

253.4

388.7

-135.3

3.41E-38

Firmicutes

Clostridia

Clostridiales

Peptostreptococcaceae

Peptostreptococcus

-

4455767

56.8

206.1

-149.3

0.0597

Firmicutes

Bacilli

Lactobacillales

Streptococcaceae

Streptococcus

-

2.22E-99

Fusobacteriia

Fusobacteriales

Leptotrichiaceae

Leptotrichia

-

Bacteroidia

Bacteroidales

Paraprevotellaceae

Prevotella

tannerae

54.4

215.3

-160.9

0.8

165.9

-165.1

4308793

35.2

270.2

-235

4363066

9.8

333.7

-323.9

2469654

41.2

399.4

-358.2

4439603 1030 2727.2 mean relative abundance

-1697.2

0.0006

EP

252843 4448497

TE D

642465 2438396

Fusobacteria

Bacteroidetes

0.0389

Fusobacteria

Fusobacteriia

Fusobacteriales

Fusobacteriaceae

Fusobacterium

-

0.0568

Proteobacteria

Gammaproteobacteria

Pasteurellales

Pasteurellaceae

Aggregatibacter

-

Bacteroidetes

Bacteroidia

Bacteroidales

Paraprevotellaceae

Prevotella

-

Firmicutes

Bacilli

Lactobacillales

Streptococcaceae

Streptococcus

-

AC C

#

ICS # non-R

1.68E-219 0.0127

Table S10: Predicted KEGG pathways enriched in ICS non-responders compared to ICS responders at baseline.

21

ACCEPTED MANUSCRIPT

ICS nonresponders

ICSresponders

ICS non-responders responders

KEGG Super-pathways

KEGG pathways

6133

90535

Amino Acid Metabolism

Alanine, aspartate and glutamate metabolism

163683

182438

Amino Acid Metabolism

Amino acid related enzymes

67400

48235

19165

Amino Acid Metabolism

171954

78691

93263

Amino Acid Metabolism

231976

121154

110822

Amino Acid Metabolism

54604

16710

37894

Amino Acid Metabolism

46299

268

46031

Amino Acid Metabolism

159693

49782

109911

Amino Acid Metabolism

44

12

32

Amino Acid Metabolism

RI PT

96668 346121

Arginine and proline metabolism Cysteine and methionine metabolism Glycine, serine and threonine metabolism

Phenylalanine metabolism Phenylalanine, tyrosine and tryptophan biosynthesis Tryptophan metabolism

20667

31017

Amino Acid Metabolism

Tyrosine metabolism

205019

112275

Amino Acid Metabolism

Valine, leucine and isoleucine biosynthesis

276968

119360

157608

Amino Acid Metabolism

Valine, leucine and isoleucine degradation

46198

546

45652

Biosynthesis of Other Secondary Metabolites

Novobiocin biosynthesis

4388

18964

-14576

Biosynthesis of Other Secondary Metabolites

Penicillin and cephalosporin biosynthesis

9285

36231

-26946

Biosynthesis of Other Secondary Metabolites

Phenylpropanoid biosynthesis

149876

160184

-10308

Carbohydrate Metabolism

Amino sugar and nucleotide sugar metabolism

93433

4694

88739

Carbohydrate Metabolism

Ascorbate and aldarate metabolism

413978

211294

202684

Carbohydrate Metabolism

Butanoate metabolism

151818

133288

18530

Carbohydrate Metabolism

C5-Branched dibasic acid metabolism

510250

246694

263556

Carbohydrate Metabolism

Citrate cycle (TCA cycle)

91488

115716

-24228

Carbohydrate Metabolism

Fructose and mannose metabolism

26877

108183

-81306

Carbohydrate Metabolism

Galactose metabolism

542521

327948

214573

Carbohydrate Metabolism

Glycolysis / Gluconeogenesis

83375

82142

1233

Carbohydrate Metabolism

Inositol phosphate metabolism

116722

70965

45757

Carbohydrate Metabolism

Pentose and glucuronate interconversions

309090

182603

126487

Carbohydrate Metabolism

Pentose phosphate pathway

96668

6133

90535

Carbohydrate Metabolism

Propanoate metabolism

455675

229990

225685

Carbohydrate Metabolism

Pyruvate metabolism

AC C

TE D

51684 317294

EP

M AN U

SC

Lysine degradation

22

ACCEPTED MANUSCRIPT

ICS nonresponders

ICSresponders

ICS non-responders responders

KEGG Super-pathways

KEGG pathways

64173

-49729

Carbohydrate Metabolism

Starch and sucrose metabolism

42064

67886

Cell Motility

Bacterial motility proteins

62488

39219

23269

Cellular Processes and Signaling

2

57

-55

Cellular Processes and Signaling

57202

31502

25700

Cellular Processes and Signaling

29671

112498

-82827

Cellular Processes and Signaling

79239

102277

-23038

Cellular Processes and Signaling

46318

902

45416

Cellular Processes and Signaling

101147

21454

79693

Cellular Processes and Signaling

RI PT

14444 109950

Electron transfer carriers Germination

Inorganic ion transport and metabolism

M AN U

SC

Membrane and intracellular structural molecules Other ion-coupled transporters Other transporters Signal transduction mechanisms

37

155

-118

Cellular Processes and Signaling

Sporulation

85232

82785

2447

Energy Metabolism

Carbon fixation in photosynthetic organisms

48202

4851

43351

Energy Metabolism

64518

40288

24230

Energy Metabolism

Carbon fixation pathways in prokaryotes

38894

22677

Energy Metabolism

9573

364303

Energy Metabolism

1540

420

1120

Energy Metabolism

Photosynthesis

396

108

288

Energy Metabolism

Photosynthesis - antenna proteins

54942

11504

43438

Energy Metabolism

Photosynthesis proteins

332715

262004

70711

Enzyme Families

Peptidases

63873

37500

26373

Enzyme Families

Protein kinases

188379

99634

88745

Folding, Sorting and Degradation

Protein export

67735

42062

25673

Folding, Sorting and Degradation

RNA degradation

103067

21437

81630

Folding, Sorting and Degradation

Sulfur relay system

48173

2656

45517

Genetic Information Processing

Protein folding and associated processing

120672

63480

57192

Genetic Information Processing

Replication, recombination and repair proteins

166228

69731

96497

Genetic Information Processing

Translation proteins

10512

43144

-32632

Glycan Biosynthesis and Metabolism

Glycosaminoglycan degradation

62858

48757

14101

Glycan Biosynthesis and Metabolism

Glycosyltransferases

AC C

TE D

61571 373876

EP

Methane metabolism Nitrogen metabolism Oxidative phosphorylation

23

ACCEPTED MANUSCRIPT

ICS nonresponders

ICSresponders

ICS non-responders responders

KEGG Super-pathways

KEGG pathways

22647

-17127

Glycan Biosynthesis and Metabolism

Lipopolysaccharide biosynthesis proteins

126883

64237

Glycan Biosynthesis and Metabolism

Peptidoglycan biosynthesis

50687

12248

38439

Lipid Metabolism

51658

20661

30997

Lipid Metabolism

44

12

32

Lipid Metabolism

-136

Lipid Metabolism

12

13

Lipid Metabolism

1036463

799357

237106

Membrane Transport

188379

99634

88745

Membrane Transport

42102

177864

-135762

Membrane Transport

480439

264215

216224

Membrane Transport

1233749

1031431

202318

Membrane Transport

7503

30877

-23374

Metabolism

Fatty acid metabolism Glycerolipid metabolism Lipid biosynthesis proteins

SC

140

Fatty acid biosynthesis

M AN U

4 25

RI PT

5520 191120

Steroid biosynthesis ABC transporters Bacterial secretion system Phosphotransferase system (PTS)

Secretion system Transporters Amino acid metabolism

12

32

Metabolism

15503

38017

Metabolism

70507

69838

669

Metabolism

Energy metabolism

88

24

64

Metabolism

Lipid metabolism

3010

11274

-8264

Metabolism

Metabolism of cofactors and vitamins

Biosynthesis and biodegradation of secondary metabolites Carbohydrate metabolism

21

32

Metabolism

Nucleotide metabolism

248847

66227

Metabolism

Others

127273

82607

44666

Metabolism of Cofactors and Vitamins

Folate biosynthesis

121296

59194

62102

Metabolism of Cofactors and Vitamins

Lipoic acid metabolism

22645

Metabolism of Cofactors and Vitamins

Nicotinate and nicotinamide metabolism

46360

Metabolism of Cofactors and Vitamins

One carbon pool by folate

41407

Metabolism of Cofactors and Vitamins

Pantothenate and CoA biosynthesis

288859

Metabolism of Cofactors and Vitamins

Porphyrin and chlorophyll metabolism

AC C

53 315074

EP

TE D

44 53520

61527

38882

128513

82153

209922

168515

448003

159144

51658

20661

30997

Metabolism of Cofactors and Vitamins

Retinol metabolism

404632

189043

215589

Metabolism of Cofactors and Vitamins

Riboflavin metabolism

24

ACCEPTED MANUSCRIPT

ICS nonresponders

ICSresponders

ICS non-responders responders

KEGG Super-pathways

KEGG pathways

642

346

Metabolism of Cofactors and Vitamins

Thiamine metabolism

15689

38121

Metabolism of Cofactors and Vitamins

Ubiquinone and other terpenoid-quinone biosynthesis

96668

6133

90535

Metabolism of Other Amino Acids

9285

36231

-26946

Metabolism of Other Amino Acids

244909

142175

102734

Metabolism of Other Amino Acids

RI PT

988 53810

beta-Alanine metabolism Cyanoamino acid metabolism

602

1129

Metabolism of Other Amino Acids

57393

11496

Metabolism of Other Amino Acids

53634

15641

37993

Metabolism of Terpenoids and Polyketides

Biosynthesis of siderophore group nonribosomal peptides

195

50

145

Metabolism of Terpenoids and Polyketides

Carotenoid biosynthesis

319432

117227

202205

Metabolism of Terpenoids and Polyketides

Prenyltransferases

368339

126713

241626

Metabolism of Terpenoids and Polyketides

Terpenoid backbone biosynthesis

409304

360629

48675

Nucleotide Metabolism

Purine metabolism

170310

80320

89990

Nucleotide Metabolism

Pyrimidine metabolism

306381

206945

99436

Replication and Repair

Base excision repair

306381

206945

99436

Replication and Repair

DNA repair and recombination proteins

44

12

32

Signal Transduction

Calcium signaling pathway

180412

87533

92879

Signal Transduction

Two-component system

10512

43144

-32632

Signaling Molecules and Interaction

Bacterial toxins

257807

183480

74327

Transcription

Transcription factors

642256

442093

200163

Translation

Aminoacyl-tRNA biosynthesis

270199

206858

63341

Translation

Ribosome Biogenesis

54816

18151

36665

Translation

Ribosome biogenesis in eukaryotes

2 146

26

6

51658

20661

51658 51685

M AN U

TE D

AC C

79 578

Phosphonate and phosphinate metabolism

SC

1731 68889

EP

Glutathione metabolism

Selenocompound metabolism

77

Xenobiotics Biodegradation and Metabolism

Aminobenzoate degradation

432

Xenobiotics Biodegradation and Metabolism

Benzoate degradation

20

Xenobiotics Biodegradation and Metabolism

Caprolactam degradation

30997

Xenobiotics Biodegradation and Metabolism

Chloroalkane and chloroalkene degradation

20661

30997

Xenobiotics Biodegradation and Metabolism

Drug metabolism - cytochrome P450

21169

30516

Xenobiotics Biodegradation and Metabolism

Metabolism of xenobiotics by cytochrome P450

25

ACCEPTED MANUSCRIPT

ICS nonresponders

ICSresponders

ICS non-responders responders

KEGG Super-pathways

KEGG pathways

20661

30997

Xenobiotics Biodegradation and Metabolism

Naphthalene degradation

121

27

94

Xenobiotics Biodegradation and Metabolism

Polycyclic aromatic hydrocarbon degradation

26

6

20

Xenobiotics Biodegradation and Metabolism

26

6

20

Xenobiotics Biodegradation and Metabolism

RI PT

51658

Toluene degradation Xylene degradation

#

Post - Pre

195.3

0.5

194.8

241.0

71.6

4401186 4404731

Pre-

q-value

Phylum

Class

Order

Family

Genus

Species

0.0013

Proteobacteria

Betaproteobacteria

Neisseriales

Neisseriaceae

Eikenella

-

169.4

3.27E-140

Actinobacteria Proteobacteria

Actinobacteria

Actinomycetales

Corynebacteriaceae

Corynebacterium

-

Betaproteobacteria

Burkholderiales

Comamonadaceae

Delftia

-

Proteobacteria

Gammaproteobacteria

Pseudomonadales

Pseudomonadaceae

-

-

Tenericutes

Mollicutes

Mycoplasmatales

Mycoplasmataceae

Mycoplasma

-

Proteobacteria

Betaproteobacteria

Neisseriales

Neisseriaceae

Eikenella

-

4469492

183.6

16.4

167.3

0.0009

269930

129.9

12.0

117.9

0.0051

615020

39.4

0.9

38.5

7.57E-31

87506

31.3

4.1

27.1

0.0904

4366487

23.4

2.6

20.8

0.0951

Bacteroidetes Proteobacteria

M AN U

Post-

Flavobacteriia

Flavobacteriales

Flavobacteriaceae

Capnocytophaga

-

Gammaproteobacteria

Pseudomonadales

Pseudomonadaceae

Pseudomonas

-

Bacteroidia

Bacteroidales

Porphyromonadaceae

Porphyromonas

-

Bacteroidia

Bacteroidales

-

-

-

TE D

#

OTU ID

SC

Table S11: Specific taxa differentially expressed in placebo treated asthmatics post-treatment compared to baseline (pre-treatment) visit.

16.8

0.8

16.0

495451

19.5

10.6

8.9

0.0009

Bacteroidetes

245523

14.0

6.3

7.8

0.0004

Bacteroidetes

63117

10.1

2.6

7.5

0.0010

Proteobacteria

Betaproteobacteria

Neisseriales

Neisseriaceae

Kingella

-

Proteobacteria

Alphaproteobacteria

Rhodospirillales

Acetobacteraceae

Acidocella

-

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

nanceiensis

EP

1566691

0.0040

7.6

0.6

7.0

1052181

7.8

2.9

4.9

1.47E-09

763967

5.0

0.1

4.9

0.0658

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

-

4305935

5.9

1.4

4.5

0.0066

Bacteroidetes

Bacteroidia

Bacteroidales

Porphyromonadaceae

Paludibacter

-

399903

5.4

0.9

4.5

0.0010

Spirochaetes

Spirochaetes

Spirochaetales

Spirochaetaceae

Treponema

-

937813

4.5

0.3

4.3

0.0241

Firmicutes

Clostridia

Clostridiales

Tissierellaceae

Anaerococcus

-

458

4.9

0.8

4.1

0.0099

Proteobacteria

Gammaproteobacteria

Pasteurellales

Pasteurellaceae

Aggregatibacter

-

886735

6.0

2.1

3.9

0.0154

Firmicutes

Bacilli

Lactobacillales

Aerococcaceae

Alloiococcus

-

676367

3.8

0.3

3.5

0.0423

Firmicutes

Clostridia

Clostridiales

Veillonellaceae

Schwartzia

-

AC C

240252

2.40E-05

26

ACCEPTED MANUSCRIPT

OTU ID

#

Post-

Pre-

#

Post - Pre

q-value

Phylum

Class

Order

Family

Genus

Species

Betaproteobacteria

Neisseriales

Neisseriaceae

-

-

839

3.4

0.3

3.1

0.0013

Proteobacteria

4365143

3.1

0.1

3.0

0.0792

Actinobacteria

Actinobacteria

Actinomycetales

Corynebacteriaceae

Corynebacterium

-

Firmicutes

Clostridia

Clostridiales

4.4

1.8

2.6

Lachnospiraceae

Oribacterium

-

4312347

2.4

0.6

1.8

0.0350

Proteobacteria

Gammaproteobacteria

Cardiobacteriales

Cardiobacteriaceae

Cardiobacterium

-

92721

1.9

0.6

1.3

0.0029

Actinobacteria

Actinobacteria

Actinomycetales

Actinomycetaceae

Actinomyces

-

887744

1.9

0.6

1.3

0.0404

Bacteroidetes

Flavobacteriia

Flavobacteriales

Weeksellaceae

-

-

Cyanobacteria

Chloroplast

Streptophyta

-

RI PT

686900

0.0059

1.8

1.6

0.1

-

-

844535

0.8

2.0

-1.3

0.0350

Proteobacteria

Alphaproteobacteria

Caulobacterales

Caulobacteraceae

Asticcacaulis

-

2758

0.4

2.1

-1.8

0.0137

Fusobacteria

Fusobacteriia

Fusobacteriales

Leptotrichiaceae

Leptotrichia

-

299830

0.8

3.0

-2.3

0.0904

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

copri

506

0.3

3.3

-3.0

0.0668

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

-

Proteobacteria

Alphaproteobacteria

Caulobacterales

Caulobacteraceae

Asticcacaulis

-

Bacteroidales

Prevotellaceae

Prevotella

melaninogenica

Bacteroidales

Prevotellaceae

Prevotella

melaninogenica

M AN U

SC

4420570

0.0902

0.3

3.4

-3.1

1066621

1.0

4.3

-3.3

0.0951

Bacteroidetes

Bacteroidia

425

0.3

3.6

-3.4

0.0409

Bacteroidetes

Bacteroidia

1536

0.5

4.3

-3.8

0.0423

Firmicutes

Bacilli

Gemellales

-

-

-

Fusobacteria

Fusobacteriia

Fusobacteriales

Leptotrichiaceae

Leptotrichia

-

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

melaninogenica

BD1-5

-

-

-

-

943

0.3

4.0

-3.8

0.0608

1643

0.4

4.8

-4.4

0.0404

4457085

0.5

5.0

-4.5

2.30E-05

301

0.1

4.6

-4.5

0.0792

Bacteroidetes

-4.6

0.0340

Bacteroidetes

0.1

4.8

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

-

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

-

Proteobacteria

Gammaproteobacteria

Pseudomonadales

Moraxellaceae

Acinetobacter

-

Firmicutes

Clostridia

Clostridiales

Peptostreptococcaceae

-

-

Proteobacteria

Gammaproteobacteria

Pseudomonadales

Pseudomonadaceae

Pseudomonas

-

EP

68416

GN02

TE D

551109

0.0658

0.3

6.0

-5.8

250288

5.8

11.5

-5.8

6.95E-12

133961

0.5

7.3

-6.8

9.90E-05

105

0.8

8.4

-7.6

0.0608

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

-

Proteobacteria

Gammaproteobacteria

Enterobacteriales

Enterobacteriaceae

-

-

Bacteroidetes

Bacteroidia

Bacteroidales

Paraprevotellaceae

Prevotella

-

Actinobacteria

Actinobacteria

Actinomycetales

Actinomycetaceae

Actinomyces

-

Firmicutes

Clostridia

Clostridiales

Mogibacteriaceae

-

-

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

-

AC C

3530625

0.0071

274365

0.3

9.5

-9.3

0.0010

4459265

2.3

11.6

-9.4

1.49E-23

734

2.4

13.8

-11.4

0.0404

43339

4.5

24.9

-20.4

4.64E-11

-63.3

0.0552

594026

2.6

65.9

27

ACCEPTED MANUSCRIPT

#

mean relative abundance

#

Post-

#

Pre-

Post Pre

q-value

Phylum

Class

Order

Family

Genus

Species

5944.5

0.6

5943.9

1.08E-09

Actinobacteria

Actinobacteria

Actinomycetales

Microbacteriaceae

-

-

233.9

46.3

187.6

0.0071

Proteobacteria

Betaproteobacteria

Neisseriales

Neisseriaceae

Neisseria

-

855912

34.5

1.4

33.1

8.01E-23

Fusobacteria

Fusobacteriia

Fusobacteriales

Fusobacteriaceae

Fusobacterium

-

246528

103.4

73.0

30.4

1.67E-08

Proteobacteria

Gammaproteobacteria

Pseudomonadales

Moraxellaceae

Moraxella

-

3931

17.8

1.8

16.0

3.35E-14

Spirochaetes

Spirochaetes

Spirochaetales

Spirochaetaceae

Treponema

-

3860

13.5

0.8

12.8

1.13E-09

Spirochaetes

Spirochaetes

Spirochaetales

Spirochaetaceae

Treponema

amylovorum

930422

13.0

1.9

11.1

4.45E-10

Proteobacteria

Gammaproteobacteria

Pseudomonadales

Moraxellaceae

Moraxella

-

114813

11.3

3.3

8.0

2.30E-06

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

-

4472050

7.3

2.0

5.3

1.51E-14

Actinobacteria

Actinobacteria

Actinomycetales

Actinomycetaceae

-

-

Mycoplasmatales

Mycoplasmataceae

Mycoplasma

-

Erysipelotrichales

Erysipelotrichaceae

Sharpea

-

Clostridia

Clostridiales

Veillonellaceae

Dialister

-

BD1-5

-

-

-

-

Bacteroidia

Bacteroidales

-

-

-

Actinobacteria

Bifidobacteriales

Bifidobacteriaceae

Scardovia

-

5.6

1.4

4.3

5.83E-08

Tenericutes

Mollicutes

2.4

1.5

0.9

0.0821

Firmicutes

Erysipelotrichi

1105876

1.3

2.1

-0.9

0.0922

Firmicutes

148695

0.8

1.8

-1.0

0.0848

GN02

0.3

1.5

-1.3

0.0520

Bacteroidetes

5.3

6.6

-1.4

0.0020

Actinobacteria

1465

1.3

3.5

-2.3

1.34E-06

62513

0.9

3.4

-2.5

EP

23709 4411875

TE D

773109 4364176

SC

12297 4318672

M AN U

OTU ID

RI PT

Table S12: Specific taxa differentially expressed in ICS treated asthmatics post-treatment compared to baseline (pre-treatment) visit.

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

intermedia

0.0699

Firmicutes

Clostridia

Clostridiales

Lachnospiraceae

Moryella

-

0.0539

Actinobacteria

Actinobacteria

Actinomycetales

Corynebacteriaceae

Corynebacterium

-

Neisseriaceae

Kingella

-

0.8

3.5

-2.8

1.0

3.9

-2.9

0.0358

Proteobacteria

Betaproteobacteria

Neisseriales

645055

1.4

4.5

-3.1

0.0293

Fusobacteria

Fusobacteriia

Fusobacteriales

Fusobacteriaceae

Fusobacterium

-

43339

1.0

4.5

-3.5

0.0159

Firmicutes

Clostridia

Clostridiales

Mogibacteriaceae

-

-

422455

0.8

4.8

-4.0

0.0020

Tenericutes

Mollicutes

Acholeplasmatales

Acholeplasmataceae

Acholeplasma

-

AC C

495067 4380570

1580

1.4

5.6

-4.3

0.0111

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

melaninogenica

399903

2.4

7.5

-5.1

0.0009

Spirochaetes

Spirochaetes

Spirochaetales

Spirochaetaceae

Treponema

-

4004098

1.8

7.6

-5.9

5.69E-05

Tenericutes

RF3

ML615J-28

-

-

-

28

ACCEPTED MANUSCRIPT

3.3

9.3

-6.0

5.11E-12

Firmicutes

Bacilli

Lactobacillales

Lactobacillaceae

Lactobacillus

iners

92316

1.3

9.6

-8.4

0.0006

Firmicutes

Clostridia

Clostridiales

Lachnospiraceae

-

-

4426165

2.3

10.9

-8.6

1.25E-07

Bacteroidetes

Bacteroidia

Bacteroidales

Prevotellaceae

Prevotella

-

1.9

10.9

-9.0

4.04E-08

Firmicutes

Clostridia

Clostridiales

10.4

29.9

-19.5

3.10E-14

Firmicutes

Clostridia

Clostridiales

-

-

-

Peptostreptococcaceae

-

-

4421864

5.1

26.3

-21.1

0.0597

Firmicutes

Clostridia

Clostridiales

Veillonellaceae

Selenomonas

-

398192

0.3

23.1

-22.9

2.93E-06

Bacteroidetes

Bacteroidia

Bacteroidales

Paraprevotellaceae

Prevotella

-

4440670

1.5

24.9

-23.4

1.74E-18

Firmicutes

Clostridia

Clostridiales

Veillonella

-

4318284

3.9

69.1

-65.3

0.0359

Firmicutes

Clostridia

Clostridiales

Veillonellaceae

Dialister

-

2438396 12.5 123.5 mean relative abundance

-111.0

1.93E-102

Fusobacteria

Fusobacteriia

Fusobacteriales

Fusobacteriaceae

Fusobacterium

-

RI PT

893048 1044611

EP

TE D

M AN U

SC

Veillonellaceae

AC C

#

130864

29

ACCEPTED MANUSCRIPT

SUPPLEMENTAL METHODS:

SC

RI PT

Study Population and Sample Collection: Of 186 adults screened for eligibility at nine “AsthmaNet” clinical centers, 84 subjects were enrolled (Figure S1A-C). Each participating clinical center (Brigham and Women’s Hospital, Boston, MA; Northwestern University, Chicago, IL; National Jewish Health, Denver, CO; University of Wisconsin, Madison, WI; University of Pittsburgh, Pittsburgh, PA; Washington University, St. Louis, MO; University of California San Francisco, San Francisco, CA; Duke University, Durham, NC; Duke University, Durham, NC and Wake Forest University, WinstonSalem, NC) enrolled a median of 10 (IQR of 8-11) subjects of whom 50% (IQR of 39-64) were asthmatic. The study center was not a significant contributor to any of the differences in bacterial community composition reported in this study. Each subject signed informed consent approved by the center’s IRB, and an NHLBI-appointed Data Safety Monitoring Board (DSMB) oversaw the study conduct.

TE D

M AN U

Adult subjects with mild atopic asthma (AA; n=42) were enrolled who had demonstrated serologic evidence of sensitivity (>0.35 kU/l) to at least one of 12 aeroallergens (Table S1) identified using ImmunoCAP specific IgE test (Thermo scientific); and airway hyper-responsiveness (methacholine PC20 ≤8 mg/mL), or bronchodilator reversibility (FEV1 improvement ≥12% in response to albuterol). Asthmatic subjects had stable asthma for preceding 3 months, an Asthma Control Questionnaire (ACQ) score of <1.5 (1) and no use of a controller medication (such as an inhaled corticosteroid) in the preceding 6 months. Atopic non-asthmatic subjects (ANA; n=21) had sensitivity to at least one aeroallergen but no evidence of airway hyper-responsiveness, (PC20 >16 mg/mL) or bronchodilator reversibility, and no history of chronic sinusitis (for responses to additional prior clinical conditions and environmental exposure for AA and ANA subjects see Tables S2-S3). Healthy control subjects (HC; n=21) had no history of atopic symptoms, negative serologic tests for all aeroallergens tested, and no history of chronic respiratory or other disease. Exclusion criteria for all subjects included smoking, symptoms of respiratory tract infection (including acute sinusitis and bronchitis) in the previous 6 weeks, and antibiotic use in the previous 3 months.

AC C

EP

All subjects underwent spirometry, blood sampling, and sputum induction at visit 1 (Figure S1C). At visit 2, prior to bronchoscopy, oral wash samples (OW) consisting of a tongue scraping followed by oral rinse and gargle with sterile saline (10 mL), were collected as previously described (2). To evaluate for instrument carryover of oral bacterial DNA contamination during sample collection, a 10mL flush of sterile saline through the suction channel of the bronchoscope (“scope flush”) was collected following application of topical anesthesia to the vocal cords when the instrument was withdrawn before proceeding to collection of 4 protected bronchial brush (BB) samples as described previously (3). Asthmatic subjects were subsequently randomized in a 2:1 ratio to treatment with inhaled fluticasone propionate (250 mcg; GlaxoSmithKline) or placebo (lactose alone) from a dry powder inhaler twice daily for six weeks. Repeat assessments were then performed (visits 3 and 4). All samples intended for microbial analysis were stored in RNAlater (Ambion, Inc. Austin, TX) at -80°C until processing. Nucleic acid extraction: For the focus of this study, nucleic acids from OW and 3 BB’s were extracted as previously described (2, 3) using a bead-beating protocol and the AllPrep kit (Qiagen, CA), to purify DNA and RNA in parallel. Reagent controls including 10mL aliquots of the lidocaine and sterile saline used during bronchoscopy, and from “scope flush” samples were extracted for DNA and tested by quantitative PCR (Q-PCR) using universal bacterial primers (see below) for bacterial contamination, with all reagents demonstrating no evidence of significant bacterial contamination

30

ACCEPTED MANUSCRIPT

(<35 16S rRNA gene copy number). Each extraction batch also included a blank control; these went through all the steps in preparation for 16S rRNA sequencing (as described below) and were sequenced. DNA and RNA were quantified on a NanoDrop2000 (ThermoFisher, CA).

SC

RI PT

Quantitation of 16S rRNA and β-actin copy number: 16S rRNA gene copy number was assessed by quantitative PCR (Q-PCR) using the 16S rRNA universal primers P891F, P1033R and TaqMan UniProbe as previously described (4) in triplicate using TaqMan Universal Master Mix (Life Technologies). Mammalian β-actin copy numbers were evaluated using primers bActin-F (5’- CCTGGCACCCAGCACAAT-3’) and bActin-R (5’GCCGATCCACACGGAGTACT-3’) and bActin-TMP probe (5-FAM/ TCAAGATCATTGCTCCTCCTGAGCGC/3BHQ) (5). Total 16S rRNA and β-actin copy number was calculated against a standard curve of known 16SrRNA or β-actin copy numbers (1x102 – 1x108). Regression coefficients for all standard curves were > 0.98.

TE D

M AN U

16S rRNA-based airway and oral microbiota profiling using Illumina MiSeq: The variable region 4 (V4) of the 16S rRNA gene was amplified using 10 ng/l of OW and 100 ng/l of BB DNA template and 515F/806R primer combination as previously described (6, 7). Those samples without visible evidence of 16s rRNA PCR product on the first screen underwent repeat PCR screening with a bacterial-DNA control to rule out presence of sample-specific PCR inhibition. Amplicons were purified using SPRI beads (Beckman Coulter) or in the presence of multiple bands, gel-extracted with a Qiagen Gel Extraction kit (Qiagen) per manufacturer protocol, analyzed on Bioanalyzer (Aligent), quantified using the Qubit HS dsDNA kit (Invitrogen). Samples with amplicons <10ng were not sequenced. Blank control DNA extracts were also amplified, bead purified and included in each MiSeq run. Samples with sufficient 16S rRNA amplicon were pooled at 50 ng per sample, with 30-35 samples and a blank control. The barcoded, pooled library was quantified using the Qubit HS dsDNA kit (Invitrogen), denatured and 5pM was loaded onto the Illumina MiSeq cartridge (V3) in combination with a 15% (v/v) of denatured 12.5pM PhiX for sequencing.

AC C

EP

Paired-end sequences were combined using FLASh version 1.2.7 (8). Sequence analysis was performed using the Quantitative Insights into Microbial Ecology (QIIME) pipeline version 1.8.0 (9). Raw sequences were de-multiplexed by barcode and quality filtered by removing low quality sequences. Sequences with three or more consecutive bases with a Q score < 30 were truncated and discarded if the length was less than 75% of the original 250 bp read length. Sequences were aligned using PyNAST (10) and operational taxonomic units (OTUs) were picked at 97% sequence identity using uclust against the latest release of the Greengenes database (13_5) (11, 12). Reads that failed to hit the reference sequence collection were retained and clustered de novo. PyNAST-aligned sequences were chimera checked using ChimeraSlayer and putative chimeras as well as OTUs identified in negative controls or where not from kingdom Bacteria were removed from the OTU table. A phylogenetic tree was built using FastTree (13) and used to compute Faith’s Phylogenetic Diversity on OTU table multiply rarefied 100 times to 52,317 sequences per sample. Statistical analysis: All statistical analysis was performed as indicated in QIIME (9), R environment or using PRISM software. Welch’s corrected t-test, Kruskal-Wallis or Wilcoxon rank sum test and Chi-square or Fisher’s exact test where appropriate were used to determine significant differences in metadata between study groups conducted in PRISM. Ordination was visualized using Principle Coordinates Analysis (PCoA) on unweighted UniFrac distance matrix and plotted using Emperor in QIIME (14). Significant differences in beta-diversity between paired OW and BB samples were calculated on unweighted UniFrac PC1 coordinates (as a response variable) using Linear mixed

31

ACCEPTED MANUSCRIPT

M AN U

SC

RI PT

effects (LME) model ((15) using lmerTest package in R). PERMANOVA ((16); vegan package function Adonis in R) was used to evaluate significant differences in beta-diversity between groups of independent samples. Negative binomial (NB) regression model (17, 18) was used to identify specific OTUs (present in at least 3 samples) that differed in relative abundance between groups of subjects. Only OTUs having a false discovery (FDR) corrected using Benjamini Hochberg method, q-value of <0.1, were retained and declared as significantly differentially enriched taxa. Pearson correlations were performed to determine relationships between relative abundance of OTUs and study variables with q-value correction for false discovery. Phylogenetic tree constructed with taxa of interest in QIIME were annotated using Interactive tree of Life (19). Metagenome prediction from normalized 16S rRNA representative sequences was performed using PICRUSt (20). NB regression model with Benjamini Hochberg FDR correction for multiple comparisons was used to determine Kyoto Encyclopedia of Genes and Genomes (KEGG) gene orthologs (KO’s) that were significantly enriched in each group of interest. Zero-inflated negative binomial (ZINB) model (21,22) corrected for false discovery (Benjamini Hochberg, q-value of <0.1) was used to identify specific OTUs in paired samples before and after treatment with ICS or placebo inhaler. Procrustes analysis (23) based on unweighted UniFrac distance was used to compare community composition between paired samples in each treatment group.

1

TE D

ACKNOWLEGEMENTS: We additionally would like to thank the following coordinators and staff, and to identify the grants supporting the study: Robert Pedicini, BS1; Kathy Zheng, MPH1; Duanny Alva, MPH2; Assel Biyasheva, PhD2; Jenny Hixon, BS, CCRC2; Lucius Robinson III, BS, CMA, CCRC2; Mary Gill, RN, BSN3; James T. Good, MD3; Christena Kolakowski, MS3; Allen Stevens, CCRC, NREMT3; E. Rand Sutherland, MD3; Julia Bach, RN4; Rich Cornwell, MD4; Holly Eversoll, RN4; Tiffany Huard4; Keith Meyer, MD4; Barbara Miller, RN4; Ann Sexton, MPH4; Michele Wolff, RN4; Merritt Fajt, MD5; Sherri Hill, BS5; Lisa Lane, BS5; Russell Traister, MD5; Cathy Vitari, RN, BSN AE-C5; Vanessa Curtis, RRT6; Brenda Patterson, RN, APN6; Cheryl Shelton, RN, BSN6; Kelly Norsworthy, BA, CPT7; Kelsey Wollen, BA7; Eugene Bleeker, MD8; Christopher Barrios, MD8; Suzan Farris, CCRP8; Jeffrey Krings, FNP8; Victor Ortega, MD8; Cheryl Wilmoth, CCRP8; Matthew Bowman, BS9; Linda Engle, BS9; Jennifer Lucier, BS9; Aimee J. Merchlinski, MS9; Kathryn Trasatt, BS9; Angela Updegrave9; Rachel Weber, BS9; Ronald R. Zimmerman, Jr., MPA9 2

AC C

EP

Brigham & Women’s Hospital, Boston, MA (Grant: HL098102); Northwestern University, Chicago, IL (Grant: 3 4 HL098096); National Jewish Health; Denver, CO (Grant: HL098075 & TR001082); University of Wisconsin Madison, 5 6 Madison, WI (Grant: HL098090); University of Pittsburgh, Pittsburgh, PA (Grant: HL098177); Washington University, 7 St. Louis, MO (Grant: HL098098 & TR000448); University of California San Francisco, San Francisco, CA (Grant: 8 9 HL098107 & HL105572); Wake Forest University, Winston-Salem, NC (Grant: HL098103 & TR000454); Pennsylvania State University, Hershey, PA (Grant: HL098115)

32

ACCEPTED MANUSCRIPT

SUPPLEMENTAL MATERIAL REFERENCES:

6.

7.

8. 9.

10. 11.

12. 13. 14. 15. 16. 17. 18. 19. 20.

RI PT

SC

5.

M AN U

4.

TE D

3.

EP

2.

Juniper EF, O'Byrne PM, Guyatt GH, Ferrier PJ, King DR. 1999. Development and validation of a questionnaire to measure asthma control. Eur Respir J. 14:902-907. Iwai S, Fei M, Huang D, Fong S, Subramanian A, Grieco K, Lynch SV, Huang L. 2012. Oral and Airway Microbiota in HIV-Infected Pneumonia Patients. J. Clin. Microbiol. 50:2995-3002. Huang YJ, Nelson CE, Brodie EL, Desantis TZ, Baek MS, Liu J, Woyke T, Allgaier M, Bristow J, Wiener-Kronish JP, Sutherland ER, King TS, Icitovic N, Martin RJ, Calhoun WJ, Castro M, Denlinger LC, Dimango E, Kraft M, Peters SP, Wasserman SI, Wechsler ME, Boushey HA, Lynch SV. 2011. Airway microbiota and bronchial hyperresponsiveness in patients with suboptimally controlled asthma. J. Allergy Clin. Immunol. 127:372-381.e371-373. Iwai S, Huang D, Fong S, Jarlsberg LG, Worodria W, Yoo S, Cattamanchi A, Davis JL, Kaswabuli S, Segal M, Huang L, Lynch SV. 2014. The Lung Microbiome of Ugandan HIV-Infected Pneumonia Patients Is Compositionally and Functionally Distinct from That of San Franciscan Patients. PLOSone 9:e95726. Bhakta NR, Solberg OD, Nguyen CP, Nguyen CN, Arron JR, Fahy JV, Woodruff PG. 2013. A qPCR-based metric of Th2 airway inflammation in asthma. Clin. Transl. Allergy 3:24. Caporaso JG, Lauber CL, Walters WA, Berg-Lyons D, Huntley J, Fierer N, Owens SM, Betley J, Fraser L, Bauer M, Gormley N, Gilbert JA, Smith G, Knight R. 2012. Ultra-high-throughput microbial community analysis on the Illumina HiSeq and MiSeq platforms. The ISME Journal 6:1621-1624. Caporaso JG, Lauber CL, Walters WA, Berg-Lyons D, Lozupone CA, Turnbaugh PJ, Fierer N, Knight R. 2011. Global patterns of 16S rRNA diversity at a depth of millions of sequences per sample. Proceedings of the National Academy of Sciences of the United States of America 108 Suppl 1:4516-4522. Magoc T, Salzberg SL. 2011. FLASH: fast length adjustment of short reads to improve genome assemblies. Bioinformatics 27:2957-2963. Caporaso JG, Kuczynski J, Stombaugh J, Bittinger K, Bushman FD, Costello EK, Fierer N, Pena AG, Goodrich JK, Gordon JI, Huttley GA, Kelley ST, Knights D, Koenig JE, Ley RE, Lozupone CA, McDonald D, Muegge BD, Pirrung M, Reeder J, Sevinsky JR, Turnbaugh PJ, Walters WA, Widmann J, Yatsunenko T, Zaneveld J, Knight R. 2010. QIIME allows analysis of high-throughput community sequencing data. Nature methods 7:335-336. Caporaso JG, Bittinger K, Bushman FD, DeSantis TZ, Andersen GL, Knight R. 2010. PyNAST: a flexible tool for aligning sequences to a template alignment. Bioinformatics 26:266-267. DeSantis TZ, Hugenholtz P, Larsen N, Rojas M, Brodie EL, Keller K, Huber T, Dalevi D, Hu P, Andersen GL. 2006. Greengenes, a chimera-checked 16S rRNA gene database and workbench compatible with ARB. Applied and environmental microbiology 72:5069-5072. Edgar RC. 2010. Search and clustering orders of magnitude faster than BLAST. Bioinformatics 26:2460-2461. Price MN, Dehal PS, Arkin AP. 2009. FastTree: Computing Large Minimum Evolution Trees with Profiles instead of a Distance Matrix. Molecular Biology and Evolution 26:1641-1650. Vazquez-Baeza Y, Pirrung M, Gonzalez A, Knight R. 2013. EMPeror: a tool for visualizing highthroughput microbial community data. GigaScience 2:16. Pinheiro JC. 2005. Linear Mixed Effects Models for Longitudinal Data, Encyclopedia of Biostatistics. John Wiley & Sons, Ltd. Anderson MJ. 2001. A new method for non-parametric multivariate analysis of variance. Austral Ecology 26:32-46. Gerber GK, Onderdonk AB, Bry L. 2012. Inferring dynamic signatures of microbes in complex host ecosystems. PLoS computational biology 8:e1002624. McMurdie PJ, Holmes S. 2014. Waste Not, Want Not: Why Rarefying Microbiome Data Is Inadmissible. PLoS computational biology 10:e1003531. Letunic I, Bork P. 2007. Interactive Tree Of Life (iTOL): an online tool for phylogenetic tree display and annotation. Bioinformatics 23:127-128. Langille MGI, Zaneveld J, Caporaso JG, McDonald D, Knights D, Reyes JA, Clemente JC, Burkepile DE, Vega Thurber RL, Knight R, Beiko RG, Huttenhower C. 2013. Predictive functional

AC C

1.

33

ACCEPTED MANUSCRIPT

RI PT

SC M AN U TE D

23

EP

22.

AC C

21.

profiling of microbial communities using 16S rRNA marker gene sequences. Nat Biotech 31:814821. Kassahun W, Neyens T, Molenberghs G, Faes C, Verbeke G. 2014. Marginalized multilevel hurdle and zero-inflated models for overdispersed and correlated count data with excess zeros. Statistics in medicine 33:4402-4419. Min Y, Agresti A. 2005. Random effect models for repeated measures of zero-inflated count data. Statistical Modelling 5:1-19. Caporaso JG, Lauber CL, Walters WA, Berg-Lyons D, Huntley J, Fierer N, Owens SM, Betley J, Fraser L, Bauer M, Gormley N, Gilbert JA, Smith G, Knight R. 2012. Ultra-high-throughput microbial community analysis on the Illumina HiSeq and MiSeq platforms. ISME J 6: 1621-1624.

34

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