JCM Accepts, published online ahead of print on 21 May 2014 J. Clin. Microbiol. doi:10.1128/JCM.00035-14 Copyright © 2014, American Society for Microbiology. All Rights Reserved.



Airway Microbiome Dynamics in Exacerbations of Chronic Obstructive



Pulmonary Disease

3  4 

Yvonne J. Huang1, Sanjay Sethi2,3, Timothy Murphy3, Snehal Nariya1, Homer A.



Boushey1, Susan V. Lynch4

6  7 

1



University of California San Francisco



2

Division of Pulmonary/Critical Care/Allergy/Sleep Medicine; Department of Medicine,

Division of Pulmonary/Critical Care/Sleep Medicine; Veterans Western New York

10 

Health System, Buffalo, New York

11 

3

Department of Medicine, University at Buffalo State University of New York

12 

4

Division of Gastroenterology; Department of Medicine, University of California San

13 

Francisco

14  15 

Corresponding author:

16 

Yvonne J. Huang, MD

17 

505 Parnassus Avenue, Box 0130

18 

San Francisco, CA 94143-0130

19 

Email: [email protected]

20 

Tel: (415) 476-9456; Fax: (888) 786-7576

21  22  23 

24 

ABSTRACT

25 

Specific bacterial species are implicated in the pathogenesis of exacerbations of chronic

26 

obstructive pulmonary disease (COPD). However, recent studies of clinically stable

27 

COPD patients have demonstrated a greater diversity of airway microbiota, whose role in

28 

acute exacerbations is unclear. In this study temporal changes in the airway microbiome

29 

before, at the onset of, and after an acute exacerbation were examined in 60 sputum

30 

samples collected from subjects enrolled in a longitudinal study of bacterial infection in

31 

COPD. Microbiome composition and predicted functions were examined using 16S

32 

rRNA-based culture-independent profiling methods. Shifts in the abundance (>2-fold,

33 

p 4 weeks) from an individual patient

92 

(3).

93  94 

To investigate the exacerbation-related microbiome, subjects and samples were carefully

95 

selected for this study to include a diversity of exacerbations, those that were and were

96 

not associated with a new strain, and also treatment approaches commonly prescribed in

97 

clinical practice (antibiotics only, systemic corticosteroids only, or both). Five time

98 

points per subject were examined (Figure 1). The specimens spanned an exacerbation-

99 

related time period occurring in a given year between 1999 and 2005 (specific year

100 

depending on the subject). Pre- and post-exacerbation samples were those available

101 

closest to the exacerbation when the patients were at their baseline respiratory state and

102 

on their usual COPD treatment. All subjects were encouraged to seek treatment early for

103 

a suspected exacerbation, however a fixed duration of symptoms was not proscribed.

104 

However, all exacerbations samples included in this study were obtained prior to

105 

initiation of any specific treatment for the exacerbation. Exacerbation events were

106 

determined using pre-defined criteria described previously (3), involving evaluation of

107 

respiratory symptoms (dyspnea, cough, sputum production, viscosity and purulence) and

108 

their change from usual state. Pneumonia was ruled out by chest x-ray if suspected.

109 

Decisions about treatments were made by study physicians (Drs. Sethi and Murphy)

110 

based on symptoms (especially sputum purulence) and prior to the availability of sputum

111 

culture data from the concurrent visit. Antibiotics were prescribed if bacterial infection

112 

was thought to be a cause of the exacerbation, whereas steroids only were prescribed if a

113 

non-bacterial etiology was suspected.

5

114  115 

Sample preparations

116 

DNA was extracted utilizing methods previously described (15) and detailed further in

117 

the supplemental material. Briefly, universal 16S rRNA bacterial primers (Bact-27F,

118 

Bact-1492R) were used to generate amplicons which were processed for hybridization to

119 

G2 PhyloChip microarrays (Second Genome; South San Francisco, CA) by methods

120 

previously described (15, 16). Each sample was hybridized to an individual array,

121 

resulting in 60 microbiome profiles. Bacterial taxa were defined as organisms sharing ≥

122 

97% 16S rRNA gene sequence homology and classified according to Hugenholtz

123 

taxonomy in the 2011 iteration of the Greengenes database (17).

124  125 

Data analyses

126 

Raw array data were processed as previously described (18-20), including correction for

127 

background and noise, scaling of fluorescence intensities to spiked-in quantitative

128 

standards, followed by further data normalization based on the mean intensity of all

129 

profiles. Presence of taxa was determined using published methods (20, 21) with slightly

130 

modified scoring thresholds applied based on spiked-in quantitative standards. Further

131 

information is detailed in the supplemental material. The initial normalized dataset

132 

combined for all 60 samples included ~ 4,800 taxa, which was then filtered to include

133 

taxa present in at least 10% of samples, yielding a final dataset of ~3,300 taxa for

134 

analysis (available in Table S5). Log2-transformed intensities, which correlate with taxa

135 

relative abundance (22), were used in all primary analyses which were performed using R

136 

version 2.14.1.

6

137  138 

Community richness (number of taxa detected), evenness (relative distribution of taxa in

139 

a community) and diversity [Shannon and inverse Simpson indices, which consider both

140 

richness and evenness in the calculation, as well as Faith’s phylogenetic diversity which

141 

additionally incorporates phylogenetic information in this calculation (23)] were

142 

calculated using R packages vegan and picante (24, 25). Changes in these metrics were

143 

tested using repeated measures ANOVA (RM-ANOVA) and paired t-tests. Bray-Curtis

144 

and Unifrac distance measures (26, 27) were calculated and used in ordination analyses

145 

(e.g. non-metric multidimensional scaling, NMDS) to assess compositional differences

146 

and relationships to clinical factors, including the timing of pre- and post-exacerbation

147 

sample collection relative to exacerbation onset date, by distance matrix-based

148 

PERMANOVA (28). Taxon-level analyses to identify specific compositional changes

149 

between time points were performed using linear models based on moderated t-statistics

150 

and a paired test design in R package limma (29). Adjustment for multiple comparisons

151 

was applied using the Benjamini-Hochberg method or q-values (30). Changes of at least

152 

2-fold (log2 difference > 1.0; unadjusted p 2-fold. Overall, these

230 

observations suggest relative stability in airway microbiota composition during a

231 

clinically stable period prior to AECOPD in this cohort.

232  233 

At exacerbation, it was predominantly members of the Proteobacteria such as

234 

Moraxellaceae, Pasteurellaceae, Pseudomonadaceae, and Enterobacteriaceae that were

235 

enriched, when compared to Pre2 (Table 2). These enriched taxa included other

236 

potential respiratory pathogens in addition to those typically associated with AECOPD.

237 

In contrast, taxa that were decreased in abundance at exacerbation relative to Pre2, were

238 

predominantly Actinobacteria, Clostridia and Bacteroidia (Table 2). Actinobacteria

239 

comprise a large group of metabolically diverse organisms that are prolific producers of

240 

secondary metabolites, including those with antimicrobial activity (35), while clade IV

241 

and XIVA Clostridia are known inducers of anti-inflammatory T-regulatory cells (36). To

242 

speculate, these observations suggest the possibility that members of the microbiome that

243 

are depleted at the time of exacerbation may otherwise help maintain community and

244 

host immune homeostasis and potentially counteract detrimental effects of respiratory

245 

pathogenic species.

246  247 

The most dramatic changes in community composition were observed after treatment for

248 

exacerbation (Exac to Post1). This was supported by a significant difference in the mean

249 

Unifrac distance of communities between Exac and Post1 compared to that for

250 

communities between Pre2 and Exac, reflecting a significant change in phylogenetic

11

251 

make-up of the community (Fig. S1; p < 0.01). Among those taxa exhibiting significant

252 

alterations in relative abundance,, the vast majority decreased in abundance, were

253 

primarily members of the Proteobacteria, and included organisms both associated and not

254 

traditionally associated with COPD (p < 0.05, q-value < 0.12; Table S1).

255  256 

Striking differences, however, were seen in the effects of different treatments on

257 

community composition (Fig. 3B). Treatment with antibiotics alone reduced the

258 

abundance of many taxa (mostly Proteobacteria), a number of which exhibited further

259 

reductions in relative abundance at the second post-treatment time point (Post1 to Post2).

260 

The latter supports our data indicating a relationship between treatment type and

261 

community composition in post-treatment samples and provides evidence for the

262 

prolonged suppressive effects of antibiotics on some microbiota members. For subjects

263 

who received any antibiotics (either alone or with steroids), a similar pattern of reduction

264 

in microbiota members at Post1 was observed. However, in contrast to those treated with

265 

antibiotics-only, those treated with both antibiotics and steroids showed a significant

266 

increase in abundance of primarily Proteobacteria from Post1 to Post2. This was

267 

reflected also in an increase in phylogenetic diversity at Post2 compared to Post1 for this

268 

group (p < 0.01) and suggests that recovery of airway community diversity following

269 

antibiotic treatment could be influenced by concomitant corticosteroid treatment.

270  271 

In contrast to antibiotics, treatment with corticosteroids alone led predominantly to

272 

enrichment for many taxa, mainly Proteobacteria but also Bacteroidetes and Firmicutes

273 

members (Fig. 3B). Large increases were observed especially for Enterobacteriaceae (up

12

274 

to 16-fold), Lachnospiraceae, Burkholderiaceae, and Neisseriaceae (Table S2). This was

275 

mirrored by a significantly higher mean 16S rRNA copy number, a proxy for bacterial

276 

burden, in this group compared to the other treatment groups (Figure S2; RM-ANOVA p

277 

< 0.02; steroids only vs. Abx only group, p 0.5, BH-adjusted p < 0.05), e.g. Pasteurellaceae,

290 

Enterobacteriaceae, and Pseudomonadaceae (Figure 4). Most belong to the larger class

291 

of γ-Proteobacteria, as well as other classes of Proteobacteria. In contrast, taxa

292 

negatively correlated with H. influenzae were primarily members of more

293 

phylogenetically distant groups. Similar correlation patterns were observed for P.

294 

aeruginosa and M. catarrhalis (data not shown). These observations suggest that

295 

enrichment of phylogenetically related organisms in a pathogen-rich airway milieu may

13

296 

represent an important factor in microbiome dynamics associated with the pathogenesis

297 

of exacerbations.

298  299 

Quantitative PCR and sequencing validation studies

300 

Total 16S rRNA gene copy numbers ranged from 1.9 x 105 to 1.8 x 108 (Figure S2).

301 

While these did not differ significantly over time, type of treatment for exacerbation was

302 

a significant factor in observed differences (RM-ANOVA, p = 0.02) with the group

303 

treated with corticosteroids alone demonstrating higher mean total 16S rRNA copies

304 

(steroids-only vs. Abx-only; p 2-fold were

330 

used as PICRUSt inputs. The results predicted a number of KEGG gene orthologs (KOs;

331 

Kyoto Encyclopedia of Genes and Genomes, release 67.1) associated with known KEGG

332 

pathways, as enriched or depleted within the bacterial community at these time points

333 

(Figure 5). Predicted functions enriched at exacerbation included pathways involved in

334 

viral and bacterial infection (e.g. influenza A, S. aureus infection), as well as those

335 

involved in apoptosis and p53 signaling. Conversely, functions predicted to be depleted

336 

involved pathways associated with response to viral infection (e.g. RIG-I like receptor

337 

signaling), and pathways involved in flavonoid and steroid biosynthesis [known anti-

338 

inflammatory mediators (38)], as well as betalain and indole alkaloid production [known

339 

anti-microbial properties (39, 40)]. These observations suggest that loss of community

340 

functions associated with maintenance of microbial homeostasis and regulation of host

341 

immune response are associated with AECOPD. This is further supported by the

15

342 

observation that after treatment, reserve trends were observed. For example, predicted

343 

KOs that were enriched for after exacerbation included those involved in betalain, indole

344 

alkaloid, and flavonol biosynthesis pathways. In addition, KOs involved in macrolide

345 

biosynthesis also were predicted to be enriched post-treatment. Thus microbiome

346 

enrichment for taxa that encode these anti-inflammatory functional capacities may

347 

contribute to recovery from exacerbation.

348  349 

DISCUSSION

350 

In this study we extensively examined dynamics of the bacterial airway microbiome in

351 

the setting of AECOPD. In addition to assessment of compositional changes by multiple

352 

methods, we further explored the predicted functional capacities encoded by microbiota

353 

identified as potentially important based on observed compositional shifts. Thus this

354 

study represents the most comprehensive effort to date to evaluate the exacerbation-

355 

associated microbiome in COPD and serves as a foundation for approaches that could be

356 

applied to study larger numbers of patients. Collectively, our findings demonstrate that

357 

the COPD airway microbiome is rich in both bacterial species and functional capacity,

358 

and that identifiable changes in this microbiome are associated with the development,

359 

treatment, and resolution of AECOPD.

360  361 

An important limitation of our study is that the intensive microbiome analyses performed

362 

on sixty samples derived from only a small number of COPD patients. Offsetting this

363 

limitation is the longitudinal nature of the study, providing us with valid pre-exacerbation

364 

samples, as well as exacerbation samples prior to any specific treatment for exacerbation.

16

365 

Future studies involving larger patient numbers will be necessary, especially given the

366 

heterogeneity of COPD and interest in identifying best treatment approaches for different

367 

phenotypes (41). Despite the cohort size, we were able to capture some of the

368 

heterogeneity associated with exacerbations by intentionally including a proportionally

369 

even number of subjects treated for exacerbation using regimens prescribed in clinical

370 

practice. In this context, we were able to identify microbiome characteristics

371 

differentially associated with features of exacerbations in these subjects. Follow-up

372 

studies involving larger numbers of carefully phenotyped patients would be of interest.

373  374 

Several specific findings from our study are important to emphasize and have potential

375 

clinical implications to be further investigated. An important overall observation from a

376 

microbial community perspective is that many bacterial groups, not limited to typical

377 

COPD pathogens, were amongst those contributing to the most salient compositional

378 

changes observed. These dynamics, involving Proteobacteria taxa in addition to those

379 

representative of H. influenzae, M. catarrhalis and P. aeruginosa, suggest potentially

380 

important contributions of related microbiota to the etiopathogenesis of AECOPD.

381 

Independent analyses confirmed that the abundance of these typical COPD pathogens

382 

was highly correlated with the presence of related bacterial phylotypes. While array

383 

cross-hybridization at the taxon level is a possible factor in these analyses, it unlikely

384 

accounts for the number of distinct groups, characterized at family-level and higher,

385 

found to be correlated with these species. Our observations are consistent with previous

386 

demonstrations that organisms in an ecosystem can promote enrichment for related

387 

species in a ‘like will to like’ phenomenon, as observed for gut microbiota and

17

388 

susceptibility to pathogen colonization (37). Moreover, quorum-sensing mechanisms

389 

between organisms can influence bacterial pathogenicity or resistance to antimicrobials

390 

(42, 43). Overall, findings from this and other studies (44) indicate that the context of the

391 

microbial community is important to consider in efforts to better understand the

392 

pathogenesis of AECOPD.

393  394 

Another important observation is that inter-subject variability in microbial community

395 

changes at exacerbation were seen. Similar observations were made in a study using a

396 

human model of viral-induced COPD exacerbation (44). In our study, compositional

397 

changes at exacerbation in general did not involve very large shifts, although this was not

398 

true of all subjects. This is similar to what has been described in exacerbations of cystic

399 

fibrosis and non-CF bronchiectasis (45, 46). This suggests that AECOPD could also

400 

result from cumulative effects of smaller scale changes in community composition that,

401 

based on PICRUSt functional predictions, lead not only to an increase in pathogen-

402 

elicited inflammation but also loss of potentially key protective functions in the

403 

microbiome. Moreover, organisms do not need to be highly prevalent in a community to

404 

have important effects on disease pathogenesis, as has been shown for periodontitis (47).

405 

However, two of our subjects (#56 and #111) did exhibit a greater compositional shift at

406 

exacerbation based on NMDS analysis (data not shown). Although new bacterial strains

407 

were identified in these patients, others also with new strains detected did not

408 

demonstrate large compositional shifts at exacerbation. Collectively, these observations

409 

suggest there is likely inter-individual heterogeneity in the AECOPD-associated

410 

microbiome.

18

411  412 

Finally, our observations regarding the effects of COPD treatments on microbiota

413 

composition have potentially important clinical implications. Inhaled corticosteroids are

414 

commonly used in the management of COPD and other airway diseases, and our results

415 

suggest that ICS use may alter the microbiome. Whether this has implications for

416 

susceptibility to exacerbations or their outcome will require additional study. Also,

417 

treatments for exacerbation led to significant changes in community composition, some

418 

of which may last beyond the usual timeframe of clinical recovery from exacerbations.

419 

In particular, oral steroid therapy alone led to community enrichment for many

420 

microbiota. This suggests that steroids, even administered systemically, may promote

421 

microbial colonization in the airways. Overall, the long-term effects of such treatments

422 

on the microbiome in COPD, particularly with recurrent administrations, is unknown.

423  424 

Strengths of this study include the systematically collected temporal samples examined,

425 

encompassing key periods before, at, and after AECOPD. Furthermore, we extended

426 

upon compositional analyses to explore the functional capacity encoded by key

427 

microbiome members. Weaknesses include the small number of subjects, the focus on

428 

only bacteria in examining the microbiome, and also use of sputum as a representative

429 

specimen. Despite the potential for saliva admixture, sputum represents the only

430 

practical approach for repeated airway sampling of COPD patients, particularly during

431 

exacerbations when bronchoscopy may be unsafe. Important microbial insights have been

432 

gleaned from analysis of sputum in many studies of COPD, including several from this

433 

cohort (3, 4, 9, 44).

19

434  435 

In summary, results of this study indicate that the context of the airway microbiome

436 

needs to be considered in efforts to improve understanding of the pathogenesis of

437 

AECOPD. Given the heterogeneity of COPD, it will be important to study larger

438 

numbers of subjects to identify microbiome determinants associated with specific COPD

439 

phenotypes and associated exacerbations.

440  441 

ACKNOWLEDGEMENTS

442 

This work was supported by a University of California Tobacco-related Disease Research

443 

Program grant (17FT-0040) and a National Institutes of Health grant HL105572 to Y.J.H,

444 

and an American Lung Association award to S.V.L.

445  446 

The authors gratefully acknowledge Ali Faruqi for guidance in performing PICRUSt

447 

analysis of microarray-identified taxa, and to Emily Cope for technical assistance with

448 

Illumina MiSeq sequencing and guidance in the sequencing data analysis.

449  450  451  452  453  454  455  456 

20

457 

REFERENCES

458 

1. Global strategy for the diagnosis, management and prevention of chronic obstructive

459 

pulmonary disease. 2013. Available from http://www.goldcopd.org/. Accessed 1/6/2014.

460 

2. Sethi S, Murphy TF. 2008. Infection in the pathogenesis and course of chronic

461 

obstructive pulmonary disease. N. Engl. J. Med. 359:2355-2365.

462 

3. Sethi S, Evans N, Grant BJ, Murphy TF. 2002. New strains of bacteria and

463 

exacerbations of chronic obstructive pulmonary disease. N. Engl. J. Med. 347:465-471.

464 

4. Murphy TF, Brauer AL, Eschberger K, Lobbins P, Grove L, Cai X, Sethi S.

465 

Pseudomonas aeruginosa in chronic obstructive pulmonary disease. 2008. Am. J. Respir.

466 

Crit. Care Med. 177:853-860.

467 

5. Huang YJ, Kim E, Cox MJ, Brodie EL, Brown R, Wiener-Kronish JP, Lynch SV. 2010.

468 

A persistent and diverse airway microbiota present during chronic obstructive pulmonary

469 

disease exacerbations. OMICS. 14:9-59.

470 

6. Erb-Downward JR, Thompson DL, Han MK, Freeman CM, McCloskey L, Schmidt

471 

LA, Young VB, Toews GB, Curtis JL, Sundaram B, Martinez FJ, Huffnagle GB. 2011.

472 

Analysis of the lung microbiome in the "healthy" smoker and in COPD. PLoS One.

473 

6:e16384. doi:10.1371/journal.pone.0016384

474 

7. Pragman AA, Kim HB, Reilly CS, Wendt C, Isaacson RE. 2012. The lung microbiome

475 

in moderate and severe chronic obstructive pulmonary disease. PLoS One. 7:e47305.

476 

doi:10.1371/journal.pone.0047305.

477 

8. Cabrera-Rubio R, Garcia-Núñez M, Setó L, Antó JM, Moya A, Monsó E, Mira A.

478 

2012. Microbiome diversity in the bronchial tracts of patients with chronic obstructive

479 

pulmonary disease. J. Clin. Microbiol. 50:3562-8.

21

480 

9. Papi A, Bellettato CM, Braccioni F, Romagnoli M, Casolari P, Caramori G, Fabbri LM,

481 

Johnston SL. 2006. Infections and airway inflammation in chronic obstructive pulmonary

482 

disease severe exacerbations. Am. J. Respir. Crit. Care Med. 173:1114-1121.

483 

10. Rosell A, Monso E, Soler N, Torres F, Angrill J, Riise G, Zalacain R, Morera J,

484 

Torres A. 2005. Microbiologic determinants of exacerbation in chronic obstructive

485 

pulmonary disease. Arch. Intern. Med. 165:891-897.

486 

11. Sze MA, Dimitriu PA, Hayashi S, Elliott WM, McDonough JE, Gosselink JV,

487 

Cooper J, Sin DD, Mohn WW, Hogg JC. 2012. The lung tissue microbiome in chronic

488 

obstructive pulmonary disease. Am. J. Respir. Crit. Care Med. 185:1073-80.

489 

12. Duan K, Dammel C, Stein J, Rabin H, Surette MG. 2003. Modulation of

490 

Pseudomonas aeruginosa gene expression by host microflora through interspecies

491 

communication. Mol. Microbiol. 50:1477-1491.

492 

13. Endt K, Stecher B, Chaffron S, Slack E, Tchitchek N, Benecke A, Van Maele L,

493 

Sirard JC, Mueller AJ, Heikenwalder M, Macpherson AJ, Strugnell R, von Mering C,

494 

Hardt WD. 2010. The microbiota mediates pathogen clearance from the gut lumen after

495 

non-typhoidal Salmonella diarrhea. PLoS Pathog. 6:e1001097.

496 

doi:10.1371/journal.ppat.1001097.

497 

14. Sethi S, Wrona C, Eschberger K, Lobbins P, Cai X, Murphy TF. 2008. Inflammatory

498 

profile of new bacterial strain exacerbations of chronic obstructive pulmonary disease.

499 

Am. J. Respir. Crit. Care Med. 177:491-497.

500 

15. Huang YJ, Nelson CE, Brodie EL, Desantis TZ, Baek MS, Liu J, Woyke T, Allgaier

501 

M, Bristow J, Wiener-Kronish JP, Sutherland ER, King TS, Icitovic N, Martin RJ,

502 

Calhoun WJ, Castro M, Denlinger LC, Dimango E, Kraft M, Peters SP, Wasserman SI,

22

503 

Wechsler ME, Boushey HA, Lynch SV. 2011. Airway microbiota and bronchial

504 

hyperresponsiveness in patients with suboptimally controlled asthma. J. Allergy Clin.

505 

Immunol. 127:372-381.

506 

16. Flanagan JL, Brodie EL, Weng L, Lynch SV, Garcia O, Brown R, Hugenholtz P,

507 

DeSantis TZ, Andersen GL, Wiener-Kronish JP, Bristow J. 2007. Loss of bacterial

508 

diversity during antibiotic treatment of intubated patients colonized with Pseudomonas

509 

aeruginosa. J. Clin. Microbiol. 45:1954-1962.

510 

17. McDonald D, Price MN, Goodrich J, Nawrocki EP, DeSantis TZ, Probst A,

511 

Andersen GL, Knight R, Hugenholtz P. 2012. An improved Greengenes taxonomy with

512 

explicit ranks for ecological and evolutionary analyses of bacteria and archaea. ISME J.

513 

6:610-8.

514 

18. Brodie EL, Desantis TZ, Joyner DC, Baek SM, Larsen JT, Andersen GL, Hazen TC,

515 

Richardson PM, Herman DJ, Tokunaga TK, Wan JM, Firestone MK. 2006. Application

516 

of a high-density oligonucleotide microarray approach to study bacterial population

517 

dynamics during uranium reduction and reoxidation. Appl. Environ. Microbiol. 72:6288-

518 

6298.

519 

19. DeSantis TZ, Brodie EL, Moberg JP, Zubieta IX, Piceno YM, Andersen GL. 2007.

520 

High-density universal 16S rRNA microarray analysis reveals broader diversity than

521 

typical clone library when sampling the environment. Microb Ecol. 53:371-383.

522 

20. Hazen TC, Dubinsky EA, DeSantis TZ, Andersen GL, Piceno YM, Singh N, Jansson

523 

JK, Probst A, Borglin SE, Fortney JL, Stringfellow WT, Bill M, Conrad ME, Tom LM,

524 

Chavarria KL, Alusi TR, Lamendella R, Joyner DC, Spier C, Baelum J, Auer M, Zemla

23

525 

ML, Chakraborty R, Sonnenthal EL, D'haeseleer P, Holman HY, Osman S, Lu Z, Van

526 

Nostrand JD, Deng Y, Zhou J, Mason OU. 2010. Deep-sea oil plume enriches indigenous

527 

oil-degrading bacteria. Science. 330:204-8.

528 

21. Handley KM, Wrighton KC, Piceno YM, Andersen GL, DeSantis TZ, Williams KH,

529 

Wilkins MJ, N'Guessan AL, Peacock A, Bargar J, Long PE, Banfield JF. 2012. High-

530 

density PhyloChip profiling of stimulated aquifer microbial communities reveals a

531 

complex response to acetate amendment. FEMS Microbiol. Ecol. 81:188-204.

532 

22. DeSantis TZ, Stone CE, Murray SR, Moberg JP, Andersen GL. 2005. Rapid

533 

quantification and taxonomic classification of environmental DNA from both prokaryotic

534 

and eukaryotic origins using a microarray. FEMS Microbiol. Lett. 245:271-8.

535 

23. Faith DP. Conservation evaluation and phylogenetic diversity. 1992. Biol. Conserv.

536 

61:1–10.

537 

24. Oksanen J, Kindt R, Legendre P, O'Hara B, Simpson GL, Solymos P, Henry M,

538 

Stevens H, Wagner H. 2008.Vegan: Community ecology package. R package version

539 

1.16-1.

540 

25. Kembel SW, Cowan PD, Helmus MR, Cornwell WK, Morlon H, Ackerly DD,

541 

Blomberg SP, Webb CO. 2010. Picante: R tools for integrating phylogenies and ecology.

542 

Bioinformatics. 26:1463-1464.

543 

26. Gotelli N, Ellison A. 2004. A primer of ecological statistics. Sinauer Associates, Inc.

544 

Sunderland, MA.

545 

27. Lozupone C, Knight R. 2005. UniFrac: a new phylogenetic method for comparing

546 

microbial communities. Appl. Environ. Microbiol. 71: 8228–8235.

24

547 

28. Anderson, M.J. 2001. A new method for non-parametric multivariate analysis of

548 

variance. Austral Ecology. 26:32–46.

549 

29. Smyth, G. K. 2004. Linear models and empirical Bayes methods for assessing

550 

differential expression in microarray experiments. Statistical Applications in Genetics

551 

and Molecular Biology. 3(1):Article3.

552 

30. Storey JD, Tibshirani R. 2003. Statistical significance for genomewide studies. Proc.

553 

Natl. Acad. Sci. U S A. 100:9440-5.

554 

31. Sunagawa S, DeSantis TZ, Piceno YM, Brodie EL, DeSalvo MK, Voolstra CR, Weil

555 

E, Andersen GL, Medina M. 2009. Bacterial diversity and White Plague Disease-

556 

associated community changes in the Caribbean coral Montastraea faveolata. ISME J.

557 

3:512-21..

558 

32. Ivanov II, Atarashi K, Manel N, Brodie EL, Shima T, Karaoz U, Wei D, Goldfarb KC,

559 

Santee CA, Lynch SV, Tanoue T, Imaoka A, Itoh K, Takeda K, Umesaki Y, Honda K,

560 

Littman DR. 2009. Induction of intestinal Th17 cells by segmented filamentous bacteria.

561 

Cell. 139:485-98.

562 

33. Langille MG, Zaneveld J, Caporaso JG, McDonald D, Knights D, Reyes JA,

563 

Clemente JC, Burkepile DE, Vega Thurber RL, Knight R, Beiko RG, Huttenhower C.

564 

2013. Predictive functional profiling of microbial communities using 16S rRNA marker

565 

gene sequences. Nat. Biotechnol. 31:814-21.

566 

34. Pauwels RA, Buist SA, Calverley PM, Jenkins CR, Hurd SS. 2001. Global Strategy

567 

for the Diagnosis, Management, and Prevention of Chronic Obstructive Pulmonary

568 

Disease. Am. J. Respir. Crit. Care Med. 163: 1256-1276.

25

569 

35. Ventura M, Canchaya C, Tauch A, Chandra G, Fitzgerald GF, Chater KF, van

570 

Sinderen D. 2007. Genomics of Actinobacteria: tracing the evolutionary history of an

571 

ancient phylum. Microbiol. Mol. Biol. Rev. 71:495-548.

572 

36. Atarashi K, Tanoue T, Shima T, Imaoka A, Kuwahara T, Momose Y, Cheng G,

573 

Yamasaki S, Saito T, Ohba Y, Taniguchi T, Takeda K, Hori S,Ivanov II, Umesaki Y, Itoh

574 

K, Honda K. 2011. Induction of colonic regulatory T cells by indigenous Clostridium

575 

species. Science. 331(6015):337-41.

576 

37. Stecher B, Chaffron S, Kappeli R, Hapfelmeier S, Freedrich S, Weber TC, Kirundi J,

577 

Suar M, McCoy KD, von Mering C, Macpherson AJ, Hardt WD. 2010. Like will to like:

578 

Abundances of closely related species can predict susceptibility to intestinal colonization

579 

by pathogenic and commensal bacteria. PLoS Pathog. 6:e1000711.

580 

38. Cushnie TP, Lamb AJ. 2011. Recent advances in understanding the antibacterial

581 

properties of flavonoids. Int. J. Antimicrob. Agents. 38:99-107.

582 

39. Vulić JJ, Cebović TN, Canadanović VM, Cetković GS, Djilas SM, Canadanović-

583 

Brunet JM, Velićanski AS, Cvetković DD, Tumbas VT. 2013. Antiradical, antimicrobial

584 

and cytotoxic activities of commercial beetroot pomace. Food Funct. 4:713-721.

585 

40. Zoraghi R, Worrall L, See RH, Strangman W, Popplewell WL, Gong H, Samaai T,

586 

Swayze RD, Kaur S, Vuckovic M, Finlay BB, Brunham RC, McMaster WR, Davies-

587 

Coleman MT, Strynadka NC, Andersen RJ, Reiner NE. 2011. Methicillin-resistant

588 

Staphylococcus aureus (MRSA) pyruvate kinase as a target for bis-indole alkaloids with

589 

antibacterial activities. J. Biol. Chem. 286:44716-25.

590 

41. Miravitlles M, Soler-Cataluña JJ, Calle M, Soriano JB. 2013. Treatment of COPD by

591 

clinical phenotypes: putting old evidence into clinical practice. Eur Respir J. 41:1252-6.

26

592 

42. Armbruster CE, Hong W, Pang B, Weimer KE, Juneau RA, Turner J, Swords WE.

593 

2010. Indirect pathogenicity of Haemophilus influenzae and Moraxella catarrhalis in

594 

polymicrobial otitis media occurs via interspecies quorum signaling. MBio. 1:e00102-10..

595 

43. Sibley CD, Duan K, Fischer C, Parkins MD, Storey DG, Rabin HR, Surette MG.

596 

2008. Discerning the complexity of community interactions using a Drosophila model of

597 

polymicrobial infections. PLoS Pathog. 4:e1000184..

598 

44. Molyneaux PL, Mallia P, Cox MJ, Footitt J, Willis-Owen SA, Homola D, Trujillo-

599 

Torralbo MB, Elkin S, Kon OM, Cookson WO, Moffatt MF, Johnston SL. 2013.

600 

Outgrowth of the bacterial airway microbiome after rhinovirus exacerbation of chronic

601 

obstructive pulmonary disease. Am. J. Respir. Crit. Care Med. 188:1224-31.

602 

45. Zhao J, Schloss PD, Kalikin LM, Carmody LA, Foster BK, Petrosino JF, Cavalcoli

603 

JD, Vandevanter DR, Murray S, Li JZ, Young VB, Lipuma JJ. 2012. Decade-long

604 

bacterial community dynamics in cystic fibrosis airways. Proc Natl Acad Sci U S A.

605 

109:5809-5814.

606 

46. Tunney, MM, Einarsson, GG, Wei, L, Drain, M, Klem, ER, Cardwell, C, Ennis, M,

607 

Boucher, RC, Wolfgang, MC & Elborn, JS. 2013. Lung microbiota and bacterial

608 

abundance in patients with bronchiectasis when clinically stable and during exacerbation.

609 

Am. J. Respir. Crit. Care Med. 187:1118-1126.

610 

47. Hajishengallis G, Liang S, Payne MA, Hashim A, Jotwani R, Eskan MA, McIntosh

611 

ML, Alsam A, Kirkwood KL, Lambris JD, Darveau RP, Curtis MA. 2011. Low-

612 

abundance biofilm species orchestrates inflammatory periodontal disease through the

613 

commensal microbiota and complement. Cell Host Microbe. 10:497-506.

614 

27

Table 1. Clinical characteristics of subjects and exacerbations. All subjects were Caucasian male veterans. Age, lung function and smoking status information were collected during clinically stable state at parent study enrollment. Inhaled corticosteroid use status was current for analyzed time points. Subject ID

Age

FEV1 (%)

Smoking status (pack-yrs)

Inhaled steroid use

Clinical score at Exac

Sputum purulence at Exac

Sputum culture result from Exac sample

Pre-existing or new bacterial strain at Exac

Antibiotic for Exac

3

67

7

65

Systemic steroids for Exac

91

Ex (10)

Yes

20

Purulent

P. aeruginosa

New

Azithromycin

No

63

Ex (105)

No

18

Purulent

H. influenzae

New

Ofloxacin

No

19

51

63

Current (48)

Yes

19

Purulent

M. catarrhalis

New

Azithromycin

No

40

63

56

Current (112)

Yes

14

Mucoid

None

NA

None

Yes

46

71

48

Ex (125)

No

13

Mucopurulent

M. catarrhalis Klebsiella spp.

Pre-existing NA

None

Yes

48

64

46

Current (45)

Yes

15

Mucoid

None

NA

None

Yes

49

66

61

Ex (100)

Yes

24

Purulent

P. fluorescens

NA

Azithromycin

Yes

56

61

42

Current (135)

Yes

22

Purulent

H. influenzae

New

Ciprofloxacin

Yes

Mucoid

H. influenzae M. catarrhalis S. pneumoniae

Pre-existing Pre-existing Pre-existing

63 67

80 66

51 48

Ex (87) Current (50)

Yes No

13 23

None

Yes

Trimethoprim/ sulfamethoxazole

No

Purulent

S. pneumoniae

New Pre-existing New

Ofloxacin

Yes

New

Gatifloxacin

Yes

74

72

55

Ex (96)

Yes

21

Mucopurulent

H. influenzae, P. aeruginosa

111

68

38

Current (108)

Yes

23

Purulent

M. catarrhalis

Table 2. List of bacterial taxa that demonstrated the largest changes in relative abundance (> 2-fold, p-value < 0.05, q-value < 0.30), at the onset of exacerbation compared to the most recent pre-exacerbation time point (Pre2). The results are based on analysis of all subjects in the study (n = 12 subjects). Taxon (Class/Family/Genus)

prokMSA_ID for taxon representative species

γ-Proteobacteria/Moraxellaceae/Acinetobacter γ-Proteobacteria/Moraxellaceae/Perlucidibaca γ-Proteobacteria/Moraxellaceae/Acinetobacter γ-Proteobacteria/Moraxellaceae/Acinetobacter β-Proteobacteria/Anaplasmataceae/Ehrlichia γ-Proteobacteria/Pasteurellaceae/Actinobacillus α-Proteobacteria/Sphinogomonadaceae/Sphingomonas β-Proteobacteria/Comamonadaceae/Comamonas γ-Proteobacteria/Enterobacteriaceae γ-Proteobacteria/Pseudomonadaceeae/Pseudomonas Bacilli/Bacillaceae/unclassified ε-Proteobacteria/Campylobacteraceae/Campylobacter Bacteroidia/Rikenellaceae/unclassified Clostridia/Clostridiales Family XIII/Mogibacterium Actinobacteria/ Microbacteriaceae/Agromyces Clostridia/Lachnospiraceae/unclassified Actinobacteria/ACK-M1/unclassified Actinobacteria/Micrococcaceae/Rothia Actinobacteria/Nocardiaceae/Rhodococcus

60270 105696 106871 101304 6030 52835 89065 71872 10365 8434 89195 108770 101395 14167 12174 72977 96076 58305 85556

Fold-change (increase or decrease at exacerbation) +2.9 +2.7 +2.4 +2.4 +2.2 +2.1 +2.1 +2.0 +2.0 +2.0 -1.9 -1.9 -1.9 -1.8 -1.8 -1.8 -1.8 -1.8 -1.8

p-value

0.01 0.02 0.04, 0.006 0.02 0.04 0.01 0.03 0.04 0.01 0.04 0.03 0.01 0.002 0.02 0.005 0.01 0.03 0.04

1

Figure legends.

2 3

Figure 1. Time points of samples collected before, at the onset of, and after an acute COPD

4

exacerbation. Line coloring indicates the type of exacerbation treatment the subject received.

5 6

Figure 2. A. Non-metric multidimensional scaling (NMDS) ordination demonstrates that

7

samples from all subjects in the study segregate into distinct clusters (indicated by ellipses) based

8

on the type of treatment prescribed for exacerbation [antibiotics (Abx) only, oral steroids only, or

9

both; n = 4 subjects per treatment group; Bray-Curtis distance-based PERMANOVA, p < 0.05].

10

Ellipses represent 95% confidence interval for the standard error of the distances of samples in

11

each treatment group. B. NMDS plot of only post-exacerbation treatment samples from all

12

subjects demonstrates that the relationship between sample community composition and

13

treatment type remains strong, though not statistically significant (p = 0.08), when the analysis is

14

limited to only these samples. C. NMDS plot of pre-exacerbation samples alone from all subjects

15

shows that chronic use of inhaled corticosteroids may be associated with differences in bacterial

16

community composition (Bray-Curtis distance-based PERMANOVA, p = 0.08).

17 18

Figure 3. A. Volcano plots indicating taxa that are significantly increased (upper right quadrant)

19

or decreased (upper left quadrant) in pair-wise comparisons as shown, using moderated t-test

20

models (R package limma). Results shown are from analyses of all subjects. Dashed lines

21

indicate significant FDR-adjusted p-values and changes in relative abundance of at least 2-fold,

22

or log2 =1. Taxa exhibiting significant changes are colored by phylum-level classification as

23

shown. Note that in addition to highlighted taxa, many other microbiota members exhibit

24

smaller scale changes in abundance, which cumulatively may contribute importantly to

25

microbiome community function, B. Changes in taxa relative abundance from Exac to Post1 and

26

Post1 to Post2, segregated by type of exacerbation treatment (antibiotics only, systemic

27

corticosteroids only, or both; n = 4 subjects in each treatment group).

28 29

Figure 4. Correlations between the relative abundance of H. influenzae (a member of the

30

Pasteurellaceae/GammaProteobacteria) and that of all other identified taxa. The analysis was

31

performed using data from all 60 samples in this study. Significant positive and negative

32

correlations are shown (Pearson R > 0.5, Benjamini-Hochberg adjusted p < 0.05). Positive

33

correlations (red) occur predominantly with members of Pasteurellaceae or closely related

34

bacterial families and classes of Proteobacteria. Negative correlations (blue) occur with bacterial

35

families and classes that phylogenetically are more distant to H. influenzae/Pasteurellaeae. Tree

36

branches are color-coded by bacterial class (legend).

37 38

Figure 5. KEGG pathways associated with predicted gene functions (KOs) encoded by

39

metagenomes of bacterial taxa that either were increased (red) or decreased (blue) ≥ 2-fold in

40

relative abundance at the time of and after exacerbations. Among the microbiome-related

41

functions identified were those involved in promotion of inflammation by pathogens (*) and

42

production of anti-microbial and anti-inflammatory compounds (**), functions that were

43

differentially represented at exacerbation compared to post-exacerbation.

Exacerbation onset subj 111 subj 74 subj 56

Exacerbation treatment group

subj 49 subj 63

Antibiotics only Corticosteroids only Both antibiotics & corticosteroids

subj 48 subj 46 subj 40 subj 67 subj 19 subj 7 subj 3

-150

-100

-50

0

50

Days relative to exacerbation onset

Figure 1.

100

150

0.05

111Ex

Both

63Ex 56Ex

111Pre1 111Post1

46Post2 63Post2 63Pre1 63Post1

0.00

NMDS2

63Pre2

111Pre2

40Pre1

-0.05

Steroid only

46Pre2 7Post1 111Post2 48Pre1 67Post2 56Post1 7Pre1 49Post1 7Ex 74Pre1 48Ex 67Ex 74Post1 67Pre1 40Pre2 74Ex 48Post2 67Post1 74Post2 56Pre2 1 7Post2 48Post 46Post1 56Pre1 48Pre2 49Pre2 7Pre2 56Post2 74Pre2 46Ex 67Pre2 40Post2 46Pre1 49Pre1 49Post2 40Post1 40Ex 3Pre1 3Ex 3Post1 3Post2 49Ex19Pre2 19Post2 19Pre1 19Post1 3Pre2 Abx only 19Ex

-0.05

0.00 NMDS1

Figure 2A.

0.05

0.06 0.04

Steroid only

0.00

NMDS2

0.02

40Post1 40Post2 56Post2 48Post2 48Post1 49Post2 63Post1

49Post1 3Post1

-0.02

63Post2

7Post2 74Post2 19Post1 7Post1 46Post1 74Post1 67Post2

56Post1 67Post1

Both

111Post2 19Post2

111Post1

3Post2

Abx only

-0.06

-0.04

46Post2

-0.05

0.00

0.05 NMDS1

Figure 2B.

Figure 2C.

Clostridiaceae/Lachnospiraceae (Clostridia)

Classes (tree branches): Gammaproteobacteria Verrucomicrobia e Chlamydiae Aquificae gut clone group Catabacter Bacilli Fusobacteria Mollicutes Symbiobacteria Deltaproteobacteria Betaproteobacteria Alphaproteobacteria Acidobacteria NC10−2 Spirochaetes Sphingobacteria Bacteroidetes Flavobacteria Epsilonproteobacteria mgA−1 TM7−3 Cyanobacteria OP9 Actinobacteria

Bacillaceae

Staphylococcus spp.

Lactobacillus spp.

Streptococcaceae (Lactobacillales)

Enterobacteriaceae Helicobacter Campylobacter (Epsilonproteobacteria)

0.01

Pasteurellaceae (includes genus Haemophilus)

Abundance Higher

Lower Positive correlation Negative correlation

Deltaproteobacteria

Pseudomonadaceae Moraxellaceae Oxalobacteraceae (Betaproteobacteria)

Figure 4.

KEGG pathways related to biologic functions represented by predicted KOs Apoptosis Basal transcription factors Betalain biosynthesis** Biosynthesis of 12-,14-, and 16-membered macrolides** Circadian rhythm-plant Cytochrome P450 Epithelial cell signaling in H. pylori infection* Ether lipid metabolism Flavone and flavonol biosynthesis** Hypertrophic cardiomyopathy Indole alkaloid biosynthesis** Influenza A* p53 signaling pathway Renin-angiotensin system RIG-I-like receptor signaling pathway Staphylococcus aureus infection* Steroid biosynthesis Toxoplasmosis Transcription related proteins Viral myocarditis

Figure 5.

Pre2 to Exac

Exac to Post1

Airway microbiome dynamics in exacerbations of chronic obstructive pulmonary disease.

Specific bacterial species are implicated in the pathogenesis of exacerbations of chronic obstructive pulmonary disease (COPD). However, recent studie...
1MB Sizes 15 Downloads 6 Views