Skip to main content

Main menu

  • Home
  • Current issue
  • ERJ Early View
  • Past issues
  • ERS Guidelines
  • Authors/reviewers
    • Instructions for authors
    • Submit a manuscript
    • Open access
    • COVID-19 submission information
    • Peer reviewer login
  • Alerts
  • Subscriptions
  • ERS Publications
    • European Respiratory Journal
    • ERJ Open Research
    • European Respiratory Review
    • Breathe
    • ERS Books
    • ERS publications home

User menu

  • Log in
  • Subscribe
  • Contact Us
  • My Cart
  • Log out

Search

  • Advanced search
  • ERS Publications
    • European Respiratory Journal
    • ERJ Open Research
    • European Respiratory Review
    • Breathe
    • ERS Books
    • ERS publications home

Login

European Respiratory Society

Advanced Search

  • Home
  • Current issue
  • ERJ Early View
  • Past issues
  • ERS Guidelines
  • Authors/reviewers
    • Instructions for authors
    • Submit a manuscript
    • Open access
    • COVID-19 submission information
    • Peer reviewer login
  • Alerts
  • Subscriptions

Functional lower airways genomic profiling of the microbiome to capture active microbial metabolism

Imran Sulaiman, Benjamin G. Wu, Yonghua Li, Jun-Chieh Tsay, Maya Sauthoff, Adrienne S. Scott, Kun Ji, Sergei B. Koralov, Michael Weiden, Jose C. Clemente, Drew Jones, Yvonne J. Huang, Kathleen A. Stringer, Lingdi Zhang, Adam Geber, Stephanie Banakis, Laura Tipton, Elodie Ghedin, Leopoldo N. Segal
European Respiratory Journal 2021 58: 2003434; DOI: 10.1183/13993003.03434-2020
Imran Sulaiman
1Division of Pulmonary, Critical Care, and Sleep Medicine, Dept of Medicine, New York University School of Medicine, New York, NY, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Imran Sulaiman
Benjamin G. Wu
1Division of Pulmonary, Critical Care, and Sleep Medicine, Dept of Medicine, New York University School of Medicine, New York, NY, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Yonghua Li
1Division of Pulmonary, Critical Care, and Sleep Medicine, Dept of Medicine, New York University School of Medicine, New York, NY, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jun-Chieh Tsay
1Division of Pulmonary, Critical Care, and Sleep Medicine, Dept of Medicine, New York University School of Medicine, New York, NY, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Maya Sauthoff
1Division of Pulmonary, Critical Care, and Sleep Medicine, Dept of Medicine, New York University School of Medicine, New York, NY, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Adrienne S. Scott
1Division of Pulmonary, Critical Care, and Sleep Medicine, Dept of Medicine, New York University School of Medicine, New York, NY, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Kun Ji
1Division of Pulmonary, Critical Care, and Sleep Medicine, Dept of Medicine, New York University School of Medicine, New York, NY, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Sergei B. Koralov
2Dept of Pathology, New York University School of Medicine, New York, NY, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Michael Weiden
1Division of Pulmonary, Critical Care, and Sleep Medicine, Dept of Medicine, New York University School of Medicine, New York, NY, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jose C. Clemente
3Dept of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
4Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Drew Jones
5Dept of Biochemistry and Molecular Pharmacology and Dept of Radiation Oncology, New York University School of Medicine, New York, NY, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Drew Jones
Yvonne J. Huang
6Division of Pulmonary and Critical Care Medicine, Dept of Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Kathleen A. Stringer
7Dept of Clinical Pharmacy, College of Pharmacy, and Division of Pulmonary and Critical Care Medicine, Dept of Medicine, School of Medicine, University of Michigan, Ann Arbor, MI, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Kathleen A. Stringer
Lingdi Zhang
8Center for Genomics and Systems Biology, Dept of Biology, New York University, New York, NY, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Adam Geber
8Center for Genomics and Systems Biology, Dept of Biology, New York University, New York, NY, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Adam Geber
Stephanie Banakis
8Center for Genomics and Systems Biology, Dept of Biology, New York University, New York, NY, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Laura Tipton
8Center for Genomics and Systems Biology, Dept of Biology, New York University, New York, NY, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Elodie Ghedin
8Center for Genomics and Systems Biology, Dept of Biology, New York University, New York, NY, USA
9Dept of Epidemiology, School of Global Public Health, New York University, New York, NY, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Leopoldo N. Segal
1Division of Pulmonary, Critical Care, and Sleep Medicine, Dept of Medicine, New York University School of Medicine, New York, NY, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Leopoldo N. Segal
  • For correspondence: Leopoldo.Segal@nyumc.org
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Background Microbiome studies of the lower airways based on bacterial 16S rRNA gene sequencing assess microbial community structure but can only infer functional characteristics. Microbial products, such as short-chain fatty acids (SCFAs), in the lower airways have significant impact on the host's immune tone. Thus, functional approaches to the analyses of the microbiome are necessary.

Methods Here we used upper and lower airway samples from a research bronchoscopy smoker cohort. In addition, we validated our results in an experimental mouse model. We extended our microbiota characterisation beyond 16S rRNA gene sequencing with the use of whole-genome shotgun (WGS) and RNA metatranscriptome sequencing. SCFAs were also measured in lower airway samples and correlated with each of the sequencing datasets. In the mouse model, 16S rRNA gene and RNA metatranscriptome sequencing were performed.

Results Functional evaluations of the lower airway microbiota using inferred metagenome, WGS and metatranscriptome data were dissimilar. Comparison with measured levels of SCFAs shows that the inferred metagenome from the 16S rRNA gene sequencing data was poorly correlated, while better correlations were noted when SCFA levels were compared with WGS and metatranscriptome data. Modelling lower airway aspiration with oral commensals in a mouse model showed that the metatranscriptome most efficiently captures transient active microbial metabolism, which was overestimated by 16S rRNA gene sequencing.

Conclusions Functional characterisation of the lower airway microbiota through metatranscriptome data identifies metabolically active organisms capable of producing metabolites with immunomodulatory capacity, such as SCFAs.

Abstract

This study shows that both whole-genome shotgun and RNA metatranscriptome sequencing can be done on lower airway samples and can provide valuable information on bacterial function https://bit.ly/3hNmZfi

Introduction

Characterisation of the lower airway microbiota by 16S rRNA gene sequencing has revealed that the lower airways are frequently enriched with oral commensals in healthy subjects [1–7], most likely due to microaspiration. The presence of oral commensals in the lower airways has also been identified in multiple pulmonary diseases such as cystic fibrosis, bronchiectasis, chronic obstructive pulmonary disease and lung cancer [7–11]. However, the viability of organisms identified in lower airway samples using targeted gene sequencing is uncertain, and most investigations have been limited to just taxonomic description of the lower airway microbiota and its association with host phenotypes [1, 2, 7, 12–14]. Whole-genome shotgun (WGS) and RNA metatranscriptome sequencing can directly capture gene content and active transcription, respectively. These techniques have the potential to provide a more precise functional assessment of the lower airway microbiome [15–17]. Due to the limited microbial biomass in the lower airways, these methods are challenging and have not yet been fully evaluated in comparison with standard microbial profiling and inferred functional content based on 16S rRNA gene sequencing.

Functionally active microbes can produce microbial products of relevance to the host and may modify host functions [13, 14, 18]. For example, short-chain fatty acids (SCFAs) cannot be produced by mammalian cells (with the exception of acetate), but are produced by facultative and obligate anaerobes under hypoxic conditions [19–24]. SCFAs produced by the gut microbiota induce regulatory T-cells that modify asthma, inflammatory bowel disease and cancer [19–24]. These SCFAs have also been identified in the lower airways and, with 16S rRNA gene sequencing, our group has shown that their presence is associated with enrichment of the lower airway microbiota with oral commensals [13]. To better characterise functional aspects of the lower airway microbiome, we explored the use of WGS and RNA metatranscriptome approaches to uncover active microbial metabolism of immunologically relevant metabolites, such as SCFAs.

Methods

Participants and samples

For this study samples were used from 21 healthy participants who were recruited for research bronchoscopy as part of our ongoing Chronic Obstructive Pulmonary Disease and Smoker Control cohort. All participants signed informed consent and the protocol was approved by the New York University and Bellevue Hospital Center (New York, NY, USA) institutional review boards (approval S14-01546). Further details are provided in the supplementary material.

Sample processing

DNA was extracted from all samples using the QIAamp DNA Mini Kit spin column protocol (Qiagen, Germantown, MD, USA). RNA extraction was carried out with the miRNeasy Micro Kit (Qiagen). Bacterial burden was measured by droplet digital PCR (ddPCR). All samples had high-throughput sequencing of bacterial 16S rRNA gene amplicons, and WGS and RNA metatranscriptome sequencing. Sequence data were filtered for bacteria only. Additionally, to identify active bacterial metabolism, SCFAs were measured by mass spectrometry. Further details are provided in the supplementary material.

Mouse experiment

20 female C57BL/6J mice (8–14 weeks of age, 18–22 g) were used (Jackson Research Laboratories, Bar Harbor, ME, USA). Three mice were inoculated with PBS, while the remaining 17 mice were inoculated with a mixture of human oral commensals consisting of Prevotella melaninogenica, Streptococcus mitis and Veillonella parvula. Bronchoalveolar lavage (BAL) samples were sent for 16S rRNA gene sequencing and RNA metatranscriptome sequencing. New York University Institutional Animal Care and Use approved the animal studies (approval S16-00032). Further details are provided in the supplementary material.

Statistical analysis

For association with discrete factors, we used nonparametric tests (Mann–Whitney or Kruskal–Wallis ANOVA). We used the vegan package in R to construct principal coordinate analysis based on Bray–Curtis distances [25, 26]. To cluster microbiome communities into exclusive “metacommunities” we used a Dirichlet Multinomial Mixture (DMM) model [27, 28]. To evaluate differences between groups within each sequence data type, we evaluated differential expression with DESeq2 [29] with a false discovery rate (FDR) <0.05 [30]. All data is publicly available in the Sequence Read Archive under accession numbers PRJNA603592, PRJNA573853 and PRJNA603675. All codes utilised for the analysis included in this article are available at: www.github.com/segalmicrobiomelab/functional_microbiomics. Further details are provided in the supplementary material.

Results

We recruited 21 healthy smokers for this study; lower airway samples from two subjects did not yield an adequate cDNA library for metatranscriptome sequencing and were excluded from the analysis, leaving a study cohort of 19 subjects (table 1). 16S rRNA gene sequencing characterised the microbiota present in background (“BKG”) controls, upper airway (“UA”) and lower airway (“BAL”) samples. Hierarchical clustering of the most abundant taxa shows that the microbiota in UA and BKG samples are differentially contained within the two dominant clusters (figure 1a). Some BAL samples were more similar to the UA samples, composed of taxa commonly identified as oral commensals such as Veillonella, Prevotella and Streptococcus, whereas other samples were more similar to BKG samples dominated by taxa such as Methylobacterium, Actinobacillus and Lactobacillus. We confirmed by DMM that BAL samples clustered into two distinct groups (figure 1b). Samples that clustered with UA samples were enriched with supraglottic predominant taxa (BAL.16S.SPT), while samples that clustered with BKG samples were enriched with background predominant taxa (BAL.16S.BPT) [1, 2]. This last cluster represents a group of samples with lower microbial biomass and with a taxonomic composition where most of the identified taxa likely come from methodological contamination and not from “true” lower airway microbes. Significant differences between all sample types were determined by both α-diversity (Shannon Index; figure 1c) and β-diversity (Bray–Curtis distance; figure 1d); these microbial community metrics further supported qualifying BAL.16S.SPT samples as more similar to UA samples. The median bacterial load, as determined by ddPCR, was around 1000-fold higher for UA samples and 10-fold higher for BAL samples compared with BKG samples (figure 1e). However, three of the BAL samples clearly had higher bacterial burden, with levels similar to those found in UA samples; they were all identified as BAL.16S.SPT based on taxonomic composition (figure 1e). To explore functional aspects using the 16S rRNA gene sequence data, we inferred metagenomic composition by PICRUSt [31]. Comparison of the inferred metagenome between BAL.16S.SPT and BAL.16S.BPT samples suggested that there should be several KEGG (Kyoto Encyclopedia of Genes and Genomes) and associated functional pathways differentially expressed between these two clusters (figure 1f and data 1 file in the supplementary material). While this analysis infers microbial function, it is possible to directly measure microbial function through the use of other new next-generation sequencing methods and metabolomics.

View this table:
  • View inline
  • View popup
TABLE 1

Baseline characteristics of the study population

FIGURE 1
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 1

16S rRNA gene, whole-genome shotgun (WGS) and RNA sequencing: background (BKG), upper airway (UA) and bronchoalveolar (BAL) samples were collected via bronchoscopy, and 16S rRNA gene, WGS and RNA sequencing was performed. SPT: supraglottic predominant taxa; BPT: background predominant taxa; DMM: Dirichlet Multinomial Mixture. a) A heatmap based on the Bray–Curtis distance for 16S rRNA gene sequencing illustrates the top taxa for all samples. Hierarchical clustering showed two clear clusters: one cluster with BKG samples and BAL samples similar to BKG (BPT), and one cluster with UA samples and BAL samples similar to UA (SPT). b) DMM showed two clusters had the best model fit for the 16S rRNA gene sequencing. c) α-diversity (Shannon Index) showed significant (Wilcoxon) difference between all samples, and the lowest diversity in UA samples and among BAL samples that clustered to BAL.16S.SPT by DMM. Box plots represent median and interquartile range; whiskers represent the 5–95th percentile. d) β-diversity (Bray–Curtis) also indicates a significant (PERMANOVA) difference between all samples for 16S rRNA gene sequencing. e) Bacterial load measured by droplet digital PCR showed the highest levels in UA samples (Kruskal–Wallis). BAL samples also had higher levels when compared with BKG samples. Median values are indicated. f) The inferred metagenome was assessed using PICRUSt, highlighting several significantly enriched pathways (coloured in red). The dashed line indicates a false discovery rate cut-off of 0.05. g) β-diversity (Bray–Curtis) for WGS showed a significant (PERMANOVA) difference between all samples, with UA samples separate from BKG and BAL.BPT samples. Three BAL.SPT samples clustered with UA samples. h) β-diversity (Bray–Curtis) for RNA showed a significant (PERMANOVA) difference between all sample types. Two BAL.SPT samples clustered with UA samples. i) z-transformed Bray–Curtis distance between BAL samples and paired UA samples showed clear separation of BAL.16S.BPT and BAL.16S.SPT samples in 16S rRNA gene sequencing. This separation was not as clear in WGS and RNA sequencing.

Evaluation of the lower airway metagenome and metatranscriptome

To further characterise functional aspects of the airway microbiota we profiled the metagenome by WGS sequencing and the metatranscriptome by RNA sequencing. For this analysis, all BAL and UA samples were used, while only two BKG samples had RNA sequencing libraries that passed quality control. Importantly, rarefaction analysis for the WGS and RNA data showed plateauing of the curves at a lower depth than the one accomplished in this investigation (supplementary figure S2). Within the WGS and RNA sequence data, UA and BKG samples were significantly different from each other, based on α- and β-diversity (figure 1g and h and supplementary figure S1a and b), similar to the 16S rRNA gene sequence data, while BAL samples were similar to either the UA or BKG samples. Using each Bray–Curtis distance matrix for 16S rRNA gene sequence, WGS and RNA metatranscriptome data, we compared the paired distances between BAL and UA samples. 16S rRNA gene data indicated a clear separation of what we identified as BAL.16S.SPT and BAL.16S.BPT, but this distinction was lost in WGS and RNA metatranscriptome data (figure 1i). Importantly, β-diversity analyses on both WGS and RNA sequencing data showed that all BAL samples identified as BAL.16S.BPT remained similar to BKG samples. However, among BAL samples identified as BAL.16S.SPT, both WGS and RNA sequencing identified a subset of samples that clearly showed greater similarity to UA samples, while others showed greater similarity to BKG samples (figure 1g and h). Interestingly, two out of three BAL samples that had the greatest similarities with UA samples in WGS data also had the greatest similarities with UA samples in the RNA metatranscriptome data (figure 1i).

Taxonomic signature differences between sequencing data types

To evaluate similarities of taxonomic annotation at a global level we used PROCRUSTES with Monte Carlo simulation. While there was high correlation in β-diversity between WGS and RNA taxonomic assignment (p=0.001), there was no significant correlation when 16S rRNA gene sequencing data were compared with WGS or RNA data (supplementary figure S3).

Based on 16S rRNA gene sequencing, several taxa were significantly enriched (FDR <0.05) in samples identified as BAL.SPT compared with BAL.BPT (figure 2a and data 2 file in the supplementary material), with a number of known oral commensals such as Prevotella, Veillonella and Streptococcus enriched in BAL.SPT.

Further differences identified in taxonomic signatures between sequencing data types are discussed in the supplementary material.

FIGURE 2
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 2

Taxonomic annotation of all three sequencing data types. BAL: bronchoalveolar lavage; SPT: supraglottic predominant taxa; BPT: background predominant taxa; WGS: whole-genome shotgun. DESeq2 analysis of taxonomic annotation (at the genus level) between BAL.16S.SPT versus BAL.16S.BPT samples (false discovery rate <0.05) was performed on a) 16S rRNA gene sequencing data, c) WGS data and e) RNA metatranscriptome data. Circle size is representative of relative abundance. b, d, f) Gene set enrichment analysis was used to compare the taxonomic signatures identified as distinctly enriched in BAL.16S.SPT versus BAL.16S.BPT samples across the different sequencing platforms.

Functional overlap and differences across sequencing data types

Using gene set enrichment analysis to compare the functional annotations across the sequencing data types, we identified significant overlap between the data obtained (>1000 overlapping KEGG Orthologies (KOs) for each comparison) (figure 3a). In order to compare the differentially enriched pathways identified (with DESeq2) between BAL.16S.SPT and BAL.16S.BPT, we overlapped the fold change of the functional pathways (summarised to Level 3 of annotation). We identified some concordance in the directionality of the fold change, most identified as enriched in BAL.16S.SPT (figure 3b). For example, fatty acid biosynthesis as well as purine and pyrimidine metabolism were significantly enriched in all three sequence datasets. However, the presence of statistical significance (identified in figure 3b by the use of coloured symbols) differed by the method used. Furthermore, other functional pathways showed discordant directionality. For example, genes belonging to the fatty acid metabolism pathway appeared to be significantly depleted in BAL.16S.SPT samples by the inferred metagenome but significantly enriched in WGS and nonsignificantly enriched in RNA sequencing data (figure 3b). Since several genes annotated to fatty acid biosynthesis and fatty acid metabolism are part of the production of SCFAs (end-products of microbial metabolism associated with enrichment of the lower airway microbiota with oral anaerobes [13]), we measured the levels of these products directly using mass spectrometry.

FIGURE 3
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 3

Functional annotation of all three sequencing data types: 16S rRNA gene, whole-genome shotgun (WGS) and RNA metatranscriptome. MAPK: mitogen-activated protein kinase; CoA: coenzyme A; ABC: ATP-binding cassette; BAL: bronchoalveolar lavage; SPT: supraglottic predominant taxa; BPT: background predominant taxa; KO: KEGG Orthology. a) Gene set enrichment analysis comparing functional signatures identified across the different sequence data types as distinctly enriched in BAL.16S.SPT versus BAL.16S.BPT samples based on KO annotation (differential enrichment performed based on DESeq2 analysis). b) KOs were summarised to associated pathways; differential expression between BAL.16S.SPT and BAL.16S.BPT displayed as circles for 16S rRNA gene, diamonds for WGS and squares for RNA data. Coloured symbols indicate statistical significance (DESeq2; p<0.05) for each sequence data type and symbol size is relative to the amount of KOs contributing to that pathway. Two pathways highlighted in red include fatty acid biosynthesis, which shows concordance of directionality between the three sequence data types, and fatty acid metabolism, which shows discordance.

Further differences identified in functional signatures between sequencing data types are discussed in the supplementary material.

SCFA levels are different in the upper and lower airways

Initially, SCFA levels in ex vivo cultures were analysed (supplementary material). We then evaluated SCFA levels in the 19 UA and BAL samples and four BKG samples. The levels of four out of seven SCFAs were significantly higher in UA samples when compared with BKG samples: acetate, propionate, isovalerate and butyrate (figure 4). However, the levels of three other SCFAs measured were similar when comparing UA with BKG samples: hexanoate, valerate and octanoate (data not shown). These data suggest that some SCFAs are not produced by oral commensals or that their measurement lacks a dynamic range above BKG samples. Among BAL samples, there were three SCFAs with levels statistically higher than BKG samples: acetate, propionate and isovalerate (all of which were exponentially higher in UA samples). However, three BAL samples identified as BAL.16S.SPT had significantly higher levels of these three SCFAs (figure 4). These concentrations are comparable to those we have previously published as measurable in the lower airways of a separate cohort and found to be correlated with a regulatory T-cell phenotype, i.e. blunted interleukin-17 and interferon-γ response [13]. The remaining BAL samples had similar levels to BKG samples (regardless of their 16S.SPT/16S.BPT grouping). Importantly, the levels of these SCFAs in BAL samples did not correlate with levels in UA samples (p-values nonsignificant for all comparisons) (supplementary figure S7), suggesting that microaspiration of upper airway secretions containing these metabolites was not the main source of SCFAs in the lung. To further test whether levels of SCFAs are dependent on lower airway microbial metabolism, we correlated the findings from the different genomic datasets with the measured SCFAs.

FIGURE 4
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 4

Concentrations of short-chain fatty acids (SCFAs) in bronchoscopy samples: a panel of SCFAs were measured and compared (Kruskal–Wallis) in background (BKG), upper airway (UA) and bronchoalveolar lavage (BAL) samples by gas chromatography-mass spectroscopy. SPT: supraglottic predominant taxa; BPT: background predominant taxa. SCFAs were derived from the linear phase of the standard curve leading to the following cut-off values (dotted line): a) 1 µM for acetate, b) 0.6 µM for propionate, c) 0.01 µM for isovalerate and d) 1 µM for butyrate. ns: nonsignificant.

The RNA metatranscriptome correlates with measured SCFAs in the lower airways

At a global compositional level (β-diversity), the levels of these four SCFAs were not statistically significantly associated with the 16S rRNA gene sequencing data, but three out of four were statistically significantly associated with the WGS and RNA metatranscriptome data (figure 5a). This is likely to be driven by the three BAL samples with high levels of measured SCFAs identified by mass spectrometry (figure 4), as this correlation was not seen with BAL.BPT samples. Furthermore, rarefying the three datasets led to no significant change in the correlations.

FIGURE 5
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 5

Diversity correlations with short-chain fatty acid (SCFA) measurements. WGS: whole-genome shotgun; BKG: background; UA: upper airway; BAL: bronchoalveolar lavage; BPT: background predominant taxa; SPT: supraglottic predominant taxa; KO: KEGG Orthology. a) Levels of SCFAs with acetate, propionate, isovalerate and butyrate were tested (PERMANOVA) against β-diversity distribution of data from all three sequencing techniques in BAL samples. Coloured bars indicate a statistically significant association. b–d) Relative abundance of three KOs with direct annotation to measured SCFAs was compared across sample types: K01738 (acetate), K00925 (propionate) and K01034 (butyrate) with b) 16S rRNA gene, c) WGS and d) RNA metatranscriptome sequencing. Box plots represent median and interquartile range; whiskers represent the 5–95th percentile. e) RNA metatranscriptome taxonomic annotation for these three SCFA-associated KOs in UA, BAL.RNA.SPT, BAL.RNA.BPT and BKG samples are represented. Each circle represents a different sample type and colours indicate a different taxonomic annotation.

Since DMM clustering on 16S rRNA gene sequencing data did not distinguish BAL samples with high and low SCFAs levels, we performed DMM analysis of the WGS and RNA metatranscriptome data (supplementary methods), again identifying two separate clusters among BAL samples.

As validation, we focused on three KOs with direct SCFA annotation: K01738 for acetate, K00925 for propionate and K01034 for butyrate. KO enrichment differences could be identified between sample types with the RNA metatranscriptome data, but not with the inferred metagenome (16S rRNA data) or WGS data (figure 5b–d). KOs in the RNA metatranscriptome data were significantly elevated in UA samples and at very low levels in BKG samples. Importantly, the BAL samples identified in DMM clustering as being compositionally similar to UA samples had significantly higher levels of these KOs when compared with the remaining BAL samples which clustered with BKG samples (p<0.03 for all comparisons) (figure 5d). We then evaluated the taxonomic annotation available for these three KOs in the RNA metatranscriptome data and noted that the taxonomic source for these genes was predominantly oral commensals, e.g. Streptococcus and Veillonella (figure 5e). Thus, the RNA metatranscriptome has better resolution (when compared with 16S rRNA gene sequencing) for the identification of enriched genes involved in the metabolism of SCFAs present in oral anaerobes, and supports the presence of viable (RNA measurable) and metabolically active (metabolites elevated above background) bacteria in the lower airways.

By identifying potential contaminants (coming from DNA/RNA present in the bronchoscope or added through sample processing) within each sequencing method using the decontam package [32] (supplementary results), we ensured that these microbial patterns identified were related to signals present in the lower airways.

The RNA signature is lost earlier than the DNA signature in a mouse model of aspiration

It is possible that the improved resolution of RNA metatranscriptome data in the identification of active microbial metabolism in the lower airways is due to differences in DNA and RNA clearance over time. To evaluate the stability and functional dynamics of aspirated oral commensals in the lower airways, we used a mouse model. For this, mice were inoculated with a mixture of human oral commensals consisting of Prevotella, Streptococcus and Veillonella co-housed with a PBS control group. Mice were sacrificed at 1 h, 4 h, 1 day, 3 days and 7 days post-inoculation (figure 6a), and BAL samples were sent for 16S rRNA gene and RNA metatranscriptome sequencing. β-diversity analysis on 16S rRNA gene sequencing data shows that BAL samples remain similar to the mixture of human oral commensals for at least 1 day and become more similar to PBS by day 3 with a concordant decrease in the relative abundance of oral commensals (figure 6b–d). However, in the analysis based on the RNA metatranscriptome, BAL samples remain similar to the mixture of human oral commensals until the 4-h time-point and become more similar to PBS by day 1, with a concordant rapid loss of the RNA signal from oral commensals (figure 6e–g). These data support that discrepancies between these sequencing data can be time dependent and likely reflect the loss of viable (and transcriptionally active) microbes.

FIGURE 6
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 6

Mouse experiment with 16S rRNA gene and RNA metatranscriptome sequencing. MOC: mixture of human oral commensals. a) Schematic of the experiment. Mice (n=17) were inoculated with a mixture of Prevotella, Streptococcus and Veillonella (MOC) and sacrificed at specific time intervals: 1 h, 4 h, 1 day, 3 days and 7 days. Bronchoalveolar lavage samples were analysed by b–d) 16S rRNA gene and e–g) RNA metatranscriptome sequencing: b, e) principle coordinate analysis was performed with Bray–Curtis distances by time-point, c, f) mean intergroup distance between sample time-point and PBS was calculated, and d, g) relative abundance for taxa annotated to Prevotella, Streptococcus and Veillonella was calculated for each time-point.

Discussion

Functional characterisation of the lower airway microbiota has been attempted in a limited number of studies. In most of these, the inferred metagenome was used [2, 33]. Few have attempted metagenomic analyses [15]. The purpose of this study was to evaluate different sequence data types in the evaluation of the functional microbiome of the lower airways and to use the measurement of SCFAs as an independent biological outcome, i.e. a direct measure of bacterial metabolism. Our analysis showed that, among lower airway samples with enrichment of oral commensals, determined by taxonomic assignment of 16S rRNA gene sequencing, the use of WGS or RNA metatranscriptomic sequencing provides a distinct representation of functional aspects of the lower airway microbiota. Importantly, by pairing our sequence data with SCFA measurement, we showed that in lower airway samples with oral commensal enrichment, based on 16S rRNA gene sequencing, there is a subset with evidence of active microbial metabolism indicative of viability of the lower airway microbiota. This active microbial metabolism in the lower airways has been shown to influence lower airway immunity [34]. Further support for dissimilarity between 16S rRNA gene and RNA metatranscriptomic sequencing is provided with a mouse model of aspiration of oral commensals, demonstrating time-dependent differences likely related to loss of metabolically active microbes as the lower airways clear them.

With the introduction of next-generation sequencing, we have discovered complex microbial communities within several different mucosae [35–38]. For each of these environments, the microbial–host interplay has an impact on mucosal homeostasis in health and disease [37–42]. Within the lower airways, several studies have shown that complex microbial communities significantly impact the mucosal host immune tone [1, 2, 14, 43, 44]. For example, we have previously shown that lower airway enrichment with oral commensals leads to an increased lower airway inflammatory tone, characterised by a T-helper 17-like inflammatory phenotype [2, 45]. Thus, it is increasingly important to describe these environments beyond just the presence/absence of bacteria and to look at the functional implications of these bacteria. A common technique used to evaluate bacterial function is to infer the metagenome from 16S rRNA gene sequencing data. Major concerns associated with this approach are the poor strain resolution of 16S rRNA gene sequencing and the dependence on existing reference databases of annotated microbes, which can bias the results. Direct measurement of microbial genes can be accomplished by WGS and RNA metatranscriptome sequencing. In this study, we used all three methods to evaluate taxonomic and functional signatures in the lower airways. As previously described, we identified a subset of subjects that had a lower airway microbiota enriched with oral commensals such as Prevotella, Veillonella and Streptococcus [2]. Enrichment with oral commensals in a subset of samples that were identified as BAL.SPT based on 16S rRNA gene sequencing was also found in WGS and RNA sequencing data but, importantly, not in all of them. This enrichment with supraglottic taxa in the lower airways and its impact on host immune tone also has implications in disease states, as we have previously shown [2]. Thus, the information we glean from each of these data types is variable and potentially important when combined in a multi-omic approach. In addition, we have shown performing multi-omic analysis on lower airways samples is a feasible approach that provides deeper insight into the lower airway micro-environment.

As validation for such an approach, we focused on SCFAs. Several publications have identified SCFAs as the products of bacterial metabolism [46, 47]; the role these metabolites play in disease has been extensively studied in the gastrointestinal microbiota [20, 21, 23, 24], and is thought to be beneficial in inflammatory bowel disease and bowel cancer [48–51]. Within the lower airways, we have described that levels of these metabolites are associated with oral commensal enrichment (as defined by 16S rRNA gene sequencing) and have significant immunomodulatory effects on T-cells [13]. In our prior investigations we also noted that not all subjects that had enrichment of the lower airway microbiota with oral commensals had elevated levels of SCFAs [13]. This suggests that functional characterisation of the lower airway microbiota cannot be fully determined based solely on 16S rRNA gene sequencing data. We therefore integrated our WGS and RNA data with the measurement of SCFAs in UA, BAL and BKG samples. As expected, SCFAs were highest in UA samples, consistent with the presence of oral anaerobes in these high biomass samples. Within BAL samples, a small subset of samples had detectable concentrations of acetate, propionate and isovalerate at levels similar to the UA samples, identified as BAL.SPT samples based on 16S rRNA gene sequencing. Other BAL samples also identified as BAL.SPT based on 16S rRNA gene sequencing had low/below the limit of detection SCFA levels that were comparable with BAL.BPT and BKG samples. In contrast, the RNA metatranscriptome showed better sample type differentiation concordant with detected levels of SCFAs. In a recent report evaluating samples from 13 subjects, SCFA levels did not correlate with bacterial burden in BAL samples [52]. In the current investigation we identified associations between SCFAs and the lower airway RNA metatranscriptome, suggesting that it is this active microbial translation that can be associated with levels of SCFAs. Importantly, taxonomic evaluation of these KOs identified that the bacteria expressing these genes were oral commensals such as Streptococcus, Prevotella and Veillonella. Thus, these data suggest that in these samples, although oral commensals might have reached the lower airways and left traces of their genomic DNA, these bacteria have been cleared and are neither transcriptionally active nor capable of producing SCFAs at the time of sampling. This is further supported with a pre-clinical model of aspiration where mice were exposed to a mixture of human oral commensals and sacrificed at different time-points post-exposure, showing rapid loss of an RNA metatranscriptome signal from these microbes and longer persistence of a 16S rRNA gene signal. Considering the known immunomodulatory effects of SCFAs and other microbial metabolites, both possibly beneficial and detrimental depending on the different human conditions, improved understanding of the value of different sequencing methods will be key to gain functional insights of the lower airway microbiome.

There are several limitations to this study. First, in our analysis we did not remove any potential contaminants, which we found as the predominant taxa identified in many of the lower airway samples. Removing taxa identified as contaminant is frequently done in many microbiome studies hoping to remove contamination. However, there are many sources of noise, including DNA contamination from the reagents/bronchoscope and stochastic sequencing noise [53, 54]. Furthermore, in low biomass samples background removal can be quite variable within different sequencing datasets and its effect on the resulting new dataset is unclear. A recent guideline on lung microbiome research has not recommended background removal [55], so our analysis was limited to just identifying possible contaminants. Importantly, none of the oral commensals associated with active microbial metabolism were identified as background contaminants. In addition, our sample size was small and further validation will require a larger cohort. The lack of good correlation between 16S rRNA gene sequencing and WGS data may seem quite surprising since both are based on a similar DNA template. However, similarity between two genomic datasets will be dependent on 1) their ability to detect “true” bacterial signals present in a biological sample (a well-recognised challenge in low biomass samples), and 2) the background contamination due to differences in library preparation and sequencing techniques [53, 54]. We also recognise that this approach could impose a significant increase in sequencing cost compared with traditional 16S rRNA gene sequencing. However, improved accuracy in identifying active microbial metabolism in the lower airways can potentially lead to novel mechanistic insights about microbial metabolites with significant potential effects on the host, such as SCFAs. Future investigations should focus on determining the value of this improved accuracy by evaluating the potential implications for the host immune tone, an undertaking that should be designed with a larger cohort. Thus, we acknowledge that in the current investigation we did not attempt to evaluate host factors. Instead, we focused on functional evaluation of the lower airway microbiota using SCFAs as proof of concept. It is important to note that we are already facing an increase in literature suggesting that “near-bedside” metagenomics is feasible (both technically and computationally), and has potential clinical implications in terms of rapid detection of pathogens when compared with culture-based approaches and ability to detect resistant genes [56, 57]. In this setting, our data support that RNA sequencing could provide a better resolution of what microbial functions are active at a given time and may therefore contribute to the development of more targeted therapy. We also acknowledge that concentrations of acetate, which are not specific to microbes, can be influenced by the host and the environment, including water. For our assay, we used freshly opened high-performance liquid chromatography-grade water which did not have detectable acetate above that in the BKG samples. Dilution of BAL can affect the levels of SCFAs, but should not affect compositional data such as metagenome/metatranscriptome data. Future investigations may consider estimating BAL dilution factors, noting that there is still controversy in the literature about the accuracy and the best method for this [58, 59]. Also, variability between sequence data type may be due to differences in measured targets (target amplicon versus WGS versus RNA) as well as technical differences. For example, it is expected that deeper sequence depth will be needed to characterise the whole genome than to characterise taxonomic composition based on 16S rRNA gene sequencing and infer the metagenome using that data. The low bacterial biomass of the lower airway environment represents a critical challenge for the evaluation of the lower airway microbiome, both taxonomically and functionally. It is likely that this is an exponentially bigger problem for the WGS metagenome and RNA metatranscriptome. Although we aimed to achieve more sequence depth for our WGS metagenome and RNA metatranscriptome data, it is not surprising that differences may be more difficult to assess and that there is a greater level of background intrusion in these samples using these methods. However, the correlation of these data with SCFA levels suggests a more accurate functional evaluation can be achieved with WGS metagenome and RNA metatranscriptome data than with 16S rRNA gene sequencing data. Finally, the analyses presented here focused on fatty acid metabolism as a surrogate for bacterial activity. The highest level of precision and differential functional expression for lower airway samples identified by using RNA metatranscriptome data suggests that this might be a preferable functional method. However, it is likely that other microbial functional pathways may be important to study in health and disease, and future investigations should focus on experimental approaches to expand the observations made as proof of concept here.

In summary, the evaluation of the lower airway microbiome with 16S rRNA gene sequencing is limited in assessing bacterial function and therefore in assessing the potential impact on disease/host. The use of functional microbiome approaches that measure bacterial genes (WGS) and bacterial transcripts (RNA metatranscriptome) provides evidence of viable and active bacterial metabolism in the lower airways, and will likely define subgroups of lower airway microbiota with different implications for the host.

Supplementary material

Supplementary Material

Please note: supplementary material is not edited by the Editorial Office, and is uploaded as it has been supplied by the author.

Supplementary methods and results ERJ-03434-2020.Supplement

Supplementary figures ERJ-03434-2020.Figures

Supplementary data file 1 ERJ-03434-2020.Supp_Data_1

Supplementary data file 2 ERJ-03434-2020.Supp_Data_2

Supplementary data file 3 ERJ-03434-2020.Supp_Data_3

Supplementary data file 4 ERJ-03434-2020.Supp_Data_4

Shareable PDF

Supplementary Material

This one-page PDF can be shared freely online.

Shareable PDF ERJ-03434-2020.Shareable

Footnotes

  • Data availability: All data is publicly available in the Sequence Read Archive under accession numbers PRJNA603592, PRJNA573853 and PRJNA603675. All codes utilised for the analysis included in this article are available at: www.github.com/segalmicrobiomelab/functional_microbiomics

  • Author contributions: Conception and design: I. Sulaiman and L.N. Segal. Acquisition of data: B.G. Wu, Y. Li, A.S. Scott, K. Ji, A. Geber, S. Banakis, E. Ghedin, I. Sulaiman and L.N. Segal. Analysis and interpretation of data: B.G. Wu, J-C. Tsay, M. Sauthoff, M. Weiden, J.C. Clemente, D. Jones, Y.J. Huang, K.A. Stringer, L. Zhang, L. Tipton, E. Ghedin, I. Sulaiman and L.N. Segal. Drafting or revising the manuscript: I. Sulaiman, B.G. Wu, Y. Li, M. Sauthoff, A.S. Scott, J-C. Tsay, S.B. Koralov, M. Sauthoff, M. Weiden, J.C. Clemente, D. Jones, Y.J. Huang, K.A. Stringer, E. Ghedin and L.N. Segal. Final approval of the manuscript: I. Sulaiman and L.N. Segal.

  • This article has supplementary material available from erj.ersjournals.com This article has an editorial commentary: https://doi.org/10.1183/13993003.00321-2021

  • Conflict of interest: I. Sulaiman has nothing to disclose.

  • Conflict of interest: B.G. Wu has nothing to disclose.

  • Conflict of interest: Y. Li has nothing to disclose.

  • Conflict of interest: J-C. Tsay has nothing to disclose.

  • Conflict of interest: M. Sauthoff has nothing to disclose.

  • Conflict of interest: A.S. Scott has nothing to disclose.

  • Conflict of interest: K. Ji has nothing to disclose.

  • Conflict of interest: S.B. Koralov has nothing to disclose.

  • Conflict of interest: M. Weiden has nothing to disclose.

  • Conflict of interest: J.C. Clemente has nothing to disclose.

  • Conflict of interest: D. Jones has nothing to disclose.

  • Conflict of interest: Y.J. Huang has nothing to disclose.

  • Conflict of interest: K.A. Stringer has nothing to disclose.

  • Conflict of interest: L. Zhang has nothing to disclose.

  • Conflict of interest: A. Geber has nothing to disclose.

  • Conflict of interest: S. Banakis has nothing to disclose.

  • Conflict of interest: L. Tipton has nothing to disclose.

  • Conflict of interest: E. Ghedin has nothing to disclose.

  • Conflict of interest: L.N. Segal has nothing to disclose.

  • Support statement: R37 CA244775 (L.N. Segal, National Institutes of Health/National Cancer Institute), R01 HL125816 (S.B. Koralov and L.N. Segal, National Institutes of Health/National Heart, Lung, and Blood Institute), R01 GM111400 (K.A. Stringer, National Institutes of Health/National Institute of General Medical Sciences), Stony Wold (I. Sulaiman), Chest Foundation Grant (I. Sulaiman), Flight Attendant Medical Research Institute (B.G. Wu), PACT grant (L.N. Segal, Foundation for the National Institutes of Health (FHIH)), R01AI129958 (Y.J. Huang). Financial support for the PACT project is made possible through funding support provided to the FNIH by AbbVie Inc., Amgen Inc., Boehringer Ingelheim Pharma GmbH & Co. KG, Bristol-Myers Squibb, Celgene Corporation, Genentech Inc., Gilead, GlaxoSmithKline plc, Janssen Pharmaceutical Companies of Johnson & Johnson, Novartis Institutes for Biomedical Research, Pfizer Inc. and Sanofi. Funding information for this article has been deposited with the Crossref Funder Registry.

  • Received October 16, 2020.
  • Accepted December 19, 2020.
  • Copyright ©The authors 2021. For reproduction rights and permissions contact permissions{at}ersnet.org
https://www.ersjournals.com/user-licence

References

  1. ↵
    1. Segal LN,
    2. Alekseyenko AV,
    3. Clemente JC, et al.
    Enrichment of lung microbiome with supraglottic taxa is associated with increased pulmonary inflammation. Microbiome 2013; 1: 19. doi:10.1186/2049-2618-1-19
    OpenUrlCrossRefPubMed
  2. ↵
    1. Segal LN,
    2. Clemente JC,
    3. Tsay JC, et al.
    Enrichment of the lung microbiome with oral taxa is associated with lung inflammation of a Th17 phenotype. Nat Microbiol 2016; 1: 16031. doi:10.1038/nmicrobiol.2016.31
    OpenUrl
    1. Beck JM,
    2. Schloss PD,
    3. Venkataraman A, et al.
    Multicenter comparison of lung and oral microbiomes of HIV-infected and HIV-uninfected individuals. Am J Respir Crit Care Med 2015; 192: 1335–1344. doi:10.1164/rccm.201501-0128OC
    OpenUrlCrossRef
    1. Huffnagle GB,
    2. Dickson RP,
    3. Lukacs NW
    . The respiratory tract microbiome and lung inflammation: a two-way street. Mucosal Immunol 2017; 10: 299–306. doi:10.1038/mi.2016.108
    OpenUrlCrossRef
    1. Dickson RP,
    2. Erb-Downward JR,
    3. Martinez FJ, et al.
    The microbiome and the respiratory tract. Annu Rev Physiol 2016; 78: 481–504. doi:10.1146/annurev-physiol-021115-105238
    OpenUrlCrossRefPubMed
    1. Lozupone C,
    2. Cota-Gomez A,
    3. Palmer BE, et al.
    Widespread colonization of the lung by Tropheryma whipplei in HIV infection. Am J Respir Crit Care Med 2013; 187: 1110–1117. doi:10.1164/rccm.201211-2145OC
    OpenUrlCrossRefPubMed
  3. ↵
    1. Molyneaux PL,
    2. Willis-Owen SAG,
    3. Cox MJ, et al.
    Host–microbial interactions in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2017; 195: 1640–1650. doi:10.1164/rccm.201607-1408OC
    OpenUrlCrossRefPubMed
    1. Sulaiman I,
    2. Wu BG,
    3. Li Y, et al.
    Evaluation of the airway microbiome in nontuberculous mycobacteria disease. Eur Respir J 2018; 52: 1800810. doi:10.1183/13993003.00810-2018
    OpenUrlAbstract/FREE Full Text
    1. Tsay JJ,
    2. Wu BG,
    3. Badri MH, et al.
    Airway microbiota is associated with upregulation of the PI3K pathway in lung cancer. Am J Respir Crit Care Med 2018; 198: 1188–1198. doi:10.1164/rccm.201710-2118OC
    OpenUrl
    1. Erb-Downward JR,
    2. Thompson DL,
    3. Han MK, et al.
    Analysis of the lung microbiome in the “healthy” smoker and in COPD. PLoS One 2011; 6: e16384. doi:10.1371/journal.pone.0016384
    OpenUrlCrossRefPubMed
  4. ↵
    1. Sze MA,
    2. Dimitriu PA,
    3. Hayashi S, et al.
    The lung tissue microbiome in chronic obstructive pulmonary disease. Am J Respir Crit Care Med 2012; 185: 1073–1080. doi:10.1164/rccm.201111-2075OC
    OpenUrlCrossRefPubMedWeb of Science
  5. ↵
    1. Durack J,
    2. Huang YJ,
    3. Nariya S, et al.
    Bacterial biogeography of adult airways in atopic asthma. Microbiome 2018; 6: 104. doi:10.1186/s40168-018-0487-3
    OpenUrl
  6. ↵
    1. Segal LN,
    2. Clemente JC,
    3. Li Y, et al.
    Anaerobic bacterial fermentation products increase tuberculosis risk in antiretroviral-drug-treated HIV patients. Cell Host Microbe 2017; 21: 530–537. doi:10.1016/j.chom.2017.03.003
    OpenUrlCrossRefPubMed
  7. ↵
    1. Segal LN,
    2. Clemente JC,
    3. Wu BG, et al.
    Randomised, double-blind, placebo-controlled trial with azithromycin selects for anti-inflammatory microbial metabolites in the emphysematous lung. Thorax 2017; 72: 13–22. doi:10.1136/thoraxjnl-2016-208599
    OpenUrlAbstract/FREE Full Text
  8. ↵
    1. Clarke EL,
    2. Lauder AP,
    3. Hofstaedter CE, et al.
    Microbial lineages in sarcoidosis. A metagenomic analysis tailored for low-microbial content samples. Am J Respir Crit Care Med 2018; 197: 225–234. doi:10.1164/rccm.201705-0891OC
    OpenUrl
    1. Liu N,
    2. Kan J,
    3. Cao W, et al.
    Metagenomic next-generation sequencing diagnosis of peripheral pulmonary infectious lesions through virtual navigation, radial EBUS, ultrathin bronchoscopy, and ROSE. J Int Med Res 2019; 47: 4878–4885. doi:10.1177/0300060519866953
    OpenUrl
  9. ↵
    1. Qi C,
    2. Hountras P,
    3. Pickens CO, et al.
    Detection of respiratory pathogens in clinical samples using metagenomic shotgun sequencing. J Med Microbiol 2019; 68: 996–1002. doi:10.1099/jmm.0.000968
    OpenUrlCrossRef
  10. ↵
    1. Cribbs SK,
    2. Uppal K,
    3. Li S, et al.
    Correlation of the lung microbiota with metabolic profiles in bronchoalveolar lavage fluid in HIV infection. Microbiome 2016; 4: 3. doi:10.1186/s40168-016-0147-4
    OpenUrlPubMed
  11. ↵
    1. Trompette A,
    2. Gollwitzer ES,
    3. Yadava K, et al.
    Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat Med 2014; 20: 159–166. doi:10.1038/nm.3444
    OpenUrlCrossRefPubMed
  12. ↵
    1. Chambers ES,
    2. Preston T,
    3. Frost G, et al.
    Role of gut microbiota-generated short-chain fatty acids in metabolic and cardiovascular health. Curr Nutr Rep 2018; 7: 198–206. doi:10.1007/s13668-018-0248-8
    OpenUrlCrossRefPubMed
  13. ↵
    1. Katsidzira L,
    2. Ocvirk S,
    3. Wilson A, et al.
    Differences in fecal gut microbiota, short-chain fatty acids and bile acids link colorectal cancer risk to dietary changes associated with urbanization among Zimbabweans. Nutr Cancer 2019; 71: 1313–1324. doi:10.1080/01635581.2019.1602659
    OpenUrl
    1. Ohigashi S,
    2. Sudo K,
    3. Kobayashi D, et al.
    Changes of the intestinal microbiota, short chain fatty acids, and fecal pH in patients with colorectal cancer. Dig Dis Sci 2013; 58: 1717–1726. doi:10.1007/s10620-012-2526-4
    OpenUrlCrossRefPubMed
  14. ↵
    1. Parada Venegas D,
    2. De la Fuente MK,
    3. Landskron G, et al.
    Short chain fatty acids (SCFAs)-mediated gut epithelial and immune regulation and its relevance for inflammatory bowel diseases. Front Immunol 2019; 10: 277. doi:10.3389/fimmu.2019.00277
    OpenUrlPubMed
  15. ↵
    1. Sanna S,
    2. van Zuydam NR,
    3. Mahajan A, et al.
    Causal relationships among the gut microbiome, short-chain fatty acids and metabolic diseases. Nat Genet 2019; 51: 600–605. doi:10.1038/s41588-019-0350-x
    OpenUrlCrossRefPubMed
  16. ↵
    1. Dray S,
    2. Dufour AB
    . The ade4 package: implementing the duality diagram for ecologists. J Stat Softw 2007; 22: 1–20. doi:10.18637/jss.v022.i04
    OpenUrlCrossRef
  17. ↵
    1. Lozupone C,
    2. Lladser ME,
    3. Knights D, et al.
    UniFrac: an effective distance metric for microbial community comparison. ISME J 2011; 5: 169–172. doi:10.1038/ismej.2010.133
    OpenUrlCrossRefPubMedWeb of Science
  18. ↵
    1. Holmes I,
    2. Harris K,
    3. Quince C
    . Dirichlet multinomial mixtures: generative models for microbial metagenomics. PLoS One 2012; 7: e30126. doi:10.1371/journal.pone.0030126
    OpenUrlCrossRefPubMed
  19. ↵
    1. Morgan M
    . DirichletMultinomial: Dirichlet-Multinomial Mixture Model Machine Learning for Microbiome Data. R package version 1.20.0. 2017. https://bioconductor.org/packages/release/bioc/html/DirichletMultinomial.html Date last accessed: 9 January 2021.
  20. ↵
    1. Love MI,
    2. Huber W,
    3. Anders S
    . Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol 2014; 15: 550. doi:10.1186/s13059-014-0550-8
    OpenUrlCrossRefPubMed
  21. ↵
    1. Reiner A,
    2. Yekutieli D,
    3. Benjamini Y
    . Identifying differentially expressed genes using false discovery rate controlling procedures. Bioinformatics 2003; 19: 368–375. doi:10.1093/bioinformatics/btf877
    OpenUrlCrossRefPubMedWeb of Science
  22. ↵
    1. Langille MG,
    2. Zaneveld J,
    3. Caporaso JG, et al.
    Predictive functional profiling of microbial communities using 16S rRNA marker gene sequences. Nat Biotechnol 2013; 31: 814–821. doi:10.1038/nbt.2676
    OpenUrlCrossRefPubMed
  23. ↵
    1. Davis NM,
    2. Proctor DM,
    3. Holmes SP, et al.
    Simple statistical identification and removal of contaminant sequences in marker-gene and metagenomics data. Microbiome 2018; 6: 226. doi:10.1186/s40168-018-0605-2
    OpenUrlCrossRefPubMed
  24. ↵
    1. Huang YJ,
    2. Nariya S,
    3. Harris JM, et al.
    The airway microbiome in patients with severe asthma: associations with disease features and severity. J Allergy Clin Immunol 2015; 136: 874–884. doi:10.1016/j.jaci.2015.05.044
    OpenUrlCrossRefPubMed
  25. ↵
    1. McAleer JP,
    2. Kolls JK
    . Contributions of the intestinal microbiome in lung immunity. Eur J Immunol 2018; 48: 39–49. doi:10.1002/eji.201646721
    OpenUrlCrossRef
  26. ↵
    1. Byrd AL,
    2. Belkaid Y,
    3. Segre JA
    . The human skin microbiome. Nat Rev Microbiol 2018; 16: 143–155. doi:10.1038/nrmicro.2017.157
    OpenUrlCrossRefPubMed
    1. Ma B,
    2. Forney LJ,
    3. Ravel J
    . Vaginal microbiome: rethinking health and disease. Annu Rev Microbiol 2012; 66: 371–389. doi:10.1146/annurev-micro-092611-150157
    OpenUrlCrossRefPubMedWeb of Science
  27. ↵
    1. O'Dwyer DN,
    2. Dickson RP,
    3. Moore BB
    . The lung microbiome, immunity, and the pathogenesis of chronic lung disease. J Immunol 2016; 196: 4839–4847. doi:10.4049/jimmunol.1600279
    OpenUrlAbstract/FREE Full Text
  28. ↵
    1. Shreiner AB,
    2. Kao JY,
    3. Young VB
    . The gut microbiome in health and in disease. Curr Opin Gastroenterol 2015; 31: 69–75. doi:10.1097/MOG.0000000000000139
    OpenUrlCrossRefPubMed
    1. Beri K
    . Skin microbiome & host immunity: applications in regenerative cosmetics & transdermal drug delivery. Future Sci OA 2018; 4: FSO302. doi:10.4155/fsoa-2017-0117
    OpenUrl
    1. Bull MJ,
    2. Plummer NT
    . Part 1: the human gut microbiome in health and disease. Integr Med 2014; 13: 17–22.
    OpenUrl
    1. Lazar V,
    2. Ditu, LM,
    3. Pircalabioru GG, et al.
    Aspects of gut microbiota and immune system interactions in infectious diseases, immunopathology, and cancer. Front Immunol 2018; 9: 1830. doi:10.3389/fimmu.2018.01830
    OpenUrl
  29. ↵
    1. Lozupone CA
    . Unraveling interactions between the microbiome and the host immune system to decipher mechanisms of disease. mSystems 2018; 3: e00183-17. doi:10.1128/mSystems.00183-17
    OpenUrlAbstract/FREE Full Text
  30. ↵
    1. Segal LN,
    2. Rom WN,
    3. Weiden MD
    . Lung microbiome for clinicians. New discoveries about bugs in healthy and diseased lungs. Ann Am Thorac Soc 2014; 11: 108–116. doi:10.1513/AnnalsATS.201310-339FR
    OpenUrlCrossRefPubMed
  31. ↵
    1. Shenoy MK,
    2. Iwai S,
    3. Lin DL, et al.
    Immune response and mortality risk relate to distinct lung microbiomes in patients with HIV and pneumonia. Am J Respir Crit Care Med 2017; 195: 104–114. doi:10.1164/rccm.201603-0523OC
    OpenUrlCrossRef
  32. ↵
    1. Wu BG,
    2. Sulaiman I,
    3. Tsay JJ, et al
    . Episodic aspiration with oral commensals induces a MyD88-dependent, pulmonary T-helper cell type 17 response that mitigates susceptibility to Streptococcus pneumoniae. Am J Respir Crit Care Med 2021; 203: 1099–1111. doi:10.1164/rccm.202005-1596OC
    OpenUrl
  33. ↵
    1. Roediger WE
    . Role of anaerobic bacteria in the metabolic welfare of the colonic mucosa in man. Gut 1980; 21: 793–798. doi:10.1136/gut.21.9.793
    OpenUrlAbstract/FREE Full Text
  34. ↵
    1. Eftimiadi C,
    2. Buzzi E,
    3. Tonetti M, et al.
    Short-chain fatty acids produced by anaerobic bacteria alter the physiological responses of human neutrophils to chemotactic peptide. J Infect 1987; 14: 43–53. doi:10.1016/s0163-4453(87)90808-5
    OpenUrlCrossRefPubMed
  35. ↵
    1. Gomes SD,
    2. Oliveira CS,
    3. Azevedo-Silva J, et al.
    The role of diet related short-chain fatty acids in colorectal cancer metabolism and survival: prevention and therapeutic implications. Curr Med Chem 2020; 27: 4087–4108. doi:10.2174/0929867325666180530102050
    OpenUrl
    1. Hinnebusch BF,
    2. Meng S,
    3. Wu JT, et al.
    The effects of short-chain fatty acids on human colon cancer cell phenotype are associated with histone hyperacetylation. J Nutr 2002; 132: 1012–1017. doi:10.1093/jn/132.5.1012
    OpenUrlAbstract/FREE Full Text
    1. Scheppach W,
    2. Bartram HP,
    3. Richter F
    . Role of short-chain fatty acids in the prevention of colorectal cancer. Eur J Cancer 1995; 31A: 1077–1080. doi:10.1016/0959-8049(95)00165-f
    OpenUrlCrossRef
  36. ↵
    1. Silva JPB,
    2. Navegantes-Lima KC,
    3. Oliveira ALB, et al.
    Protective mechanisms of butyrate on inflammatory bowel disease. Curr Pharm Des 2018; 24: 4154–4166. doi:10.2174/1381612824666181001153605
    OpenUrl
  37. ↵
    1. Yue M,
    2. Kim JH,
    3. Evans CR, et al.
    Measurement of short-chain fatty acids in respiratory samples: keep your assay above the water line. Am J Respir Crit Care Med 2020; 202: 610–612. doi:10.1164/rccm.201909-1840LE
    OpenUrl
  38. ↵
    1. Erb-Downward JR,
    2. Falkowski NR,
    3. D'Souza JC, et al.
    Critical relevance of stochastic effects on low-bacterial-biomass 16S rRNA gene analysis. mBio 2020; 11: e00258-20. doi:10.1128/mBio.00258-20
    OpenUrlAbstract/FREE Full Text
  39. ↵
    1. Salter SJ,
    2. Cox MJ,
    3. Turek EM, et al.
    Reagent and laboratory contamination can critically impact sequence-based microbiome analyses. BMC Biol 2014; 12: 87. doi:10.1186/s12915-014-0087-z
    OpenUrlCrossRefPubMed
  40. ↵
    1. Carney SM,
    2. Clemente JC,
    3. Cox MJ, et al.
    Methods in lung microbiome research. Am J Respir Cell Mol Biol 2020; 62: 283–299. doi:10.1165/rcmb.2019-0273TR
    OpenUrlCrossRefPubMed
  41. ↵
    1. Charalampous T,
    2. Kay GL,
    3. Richardson H, et al.
    Nanopore metagenomics enables rapid clinical diagnosis of bacterial lower respiratory infection. Nat Biotechnol 2019; 37: 783–792. doi:10.1038/s41587-019-0156-5
    OpenUrlCrossRefPubMed
  42. ↵
    1. Pendleton KM,
    2. Erb-Downward JR,
    3. Bao Y, et al.
    Rapid pathogen identification in bacterial pneumonia using real-time metagenomics. Am J Respir Crit Care Med 2017; 196: 1610–1612. doi:10.1164/rccm.201703-0537LE
    OpenUrlCrossRef
  43. ↵
    1. Marcy TW,
    2. Merrill WW,
    3. Rankin JA, et al.
    Limitations of using urea to quantify epithelial lining fluid recovered by bronchoalveolar lavage. Am Rev Respir Dis 1987; 135: 1276–1280. doi:10.1164/arrd.1987.135.6.1276
    OpenUrlCrossRefPubMedWeb of Science
  44. ↵
    1. Rennard SI,
    2. Basset G,
    3. Lecossier D, et al.
    Estimation of volume of epithelial lining fluid recovered by lavage using urea as marker of dilution. J Appl Physiol 1986; 60: 532–538. doi:10.1152/jappl.1986.60.2.532
    OpenUrlCrossRefPubMedWeb of Science
PreviousNext
Back to top
View this article with LENS
Vol 58 Issue 1 Table of Contents
European Respiratory Journal: 58 (1)
  • Table of Contents
  • Index by author
Email

Thank you for your interest in spreading the word on European Respiratory Society .

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Functional lower airways genomic profiling of the microbiome to capture active microbial metabolism
(Your Name) has sent you a message from European Respiratory Society
(Your Name) thought you would like to see the European Respiratory Society web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Print
Citation Tools
Functional lower airways genomic profiling of the microbiome to capture active microbial metabolism
Imran Sulaiman, Benjamin G. Wu, Yonghua Li, Jun-Chieh Tsay, Maya Sauthoff, Adrienne S. Scott, Kun Ji, Sergei B. Koralov, Michael Weiden, Jose C. Clemente, Drew Jones, Yvonne J. Huang, Kathleen A. Stringer, Lingdi Zhang, Adam Geber, Stephanie Banakis, Laura Tipton, Elodie Ghedin, Leopoldo N. Segal
European Respiratory Journal Jul 2021, 58 (1) 2003434; DOI: 10.1183/13993003.03434-2020

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero

Share
Functional lower airways genomic profiling of the microbiome to capture active microbial metabolism
Imran Sulaiman, Benjamin G. Wu, Yonghua Li, Jun-Chieh Tsay, Maya Sauthoff, Adrienne S. Scott, Kun Ji, Sergei B. Koralov, Michael Weiden, Jose C. Clemente, Drew Jones, Yvonne J. Huang, Kathleen A. Stringer, Lingdi Zhang, Adam Geber, Stephanie Banakis, Laura Tipton, Elodie Ghedin, Leopoldo N. Segal
European Respiratory Journal Jul 2021, 58 (1) 2003434; DOI: 10.1183/13993003.03434-2020
Reddit logo Technorati logo Twitter logo Connotea logo Facebook logo Mendeley logo
Full Text (PDF)

Jump To

  • Article
    • Abstract
    • Abstract
    • Introduction
    • Methods
    • Results
    • Discussion
    • Supplementary material
    • Shareable PDF
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Subjects

  • Respiratory infections and tuberculosis
  • Tweet Widget
  • Facebook Like
  • Google Plus One

More in this TOC Section

Original Research Articles

  • Dupilumab and aspirin tolerance in NSAID-exacerbated respiratory disease
  • GPR183 antagonism reduces macrophage infiltration in influenza and SARS-CoV-2
  • Ecleralimab blocks responses to allergen in mild asthma
Show more Original Research Articles

Pulmonary infection

  • Prognostic factors of community-acquired staphylococcal pneumonia
  • COVID-19, pulmonary function and quality of life
  • Effect of different airway protective masks on cardiopulmonary parameters
Show more Pulmonary infection

Related Articles

Navigate

  • Home
  • Current issue
  • Archive

About the ERJ

  • Journal information
  • Editorial board
  • Press
  • Permissions and reprints
  • Advertising

The European Respiratory Society

  • Society home
  • myERS
  • Privacy policy
  • Accessibility

ERS publications

  • European Respiratory Journal
  • ERJ Open Research
  • European Respiratory Review
  • Breathe
  • ERS books online
  • ERS Bookshop

Help

  • Feedback

For authors

  • Instructions for authors
  • Publication ethics and malpractice
  • Submit a manuscript

For readers

  • Alerts
  • Subjects
  • Podcasts
  • RSS

Subscriptions

  • Accessing the ERS publications

Contact us

European Respiratory Society
442 Glossop Road
Sheffield S10 2PX
United Kingdom
Tel: +44 114 2672860
Email: journals@ersnet.org

ISSN

Print ISSN:  0903-1936
Online ISSN: 1399-3003

Copyright © 2023 by the European Respiratory Society