ArticlePDF Available

Comparative analysis of the alveolar microbiome in COPD, ECOPD, Sarcoidosis, and ILD patients to identify respiratory illnesses specific microbial signatures

Authors:

Abstract and Figures

Studying respiratory illness-specific microbial signatures and their interaction with other micro-residents could provide a better understanding of lung microbial ecology. Each respiratory illness has a specific disease etiology, however, so far no study has revealed disease—specific microbial markers. The present study was designed to determine disease-specific microbial features and their interactions with other residents in chronic obstructive pulmonary diseases (stable and exacerbated), sarcoidosis, and interstitial lung diseases. Broncho-alveolar lavage samples (n = 43) were analyzed by SSU rRNA gene sequencing to study the alveolar microbiome in these diseases. A predominance of Proteobacteria followed by Firmicutes, Bacteroidetes, Actinobacteria, and Fusobacteria was observed in all the disease subsets. Shannon diversity was significantly higher in stable COPD when compared to exacerbated chronic obstructive pulmonary disease (ECOPD) (p = 0.0061), and ILD patient samples (p = 0.037). The lung microbiome of the patients with stable COPD was more diverse in comparison to ECOPD and ILD patients (p < 0.001). Lefse analysis identified 40 disease—differentiating microbial features (LDA score (log10) > 4). Species network analysis indicated a significant correlation (p < 0.05) of diseases specific microbial signature with other lung microbiome members. The current study strengthens the proposed hypothesis that each respiratory illness has unique microbial signatures. These microbial signatures could be used as diagnostic markers to differentiate among various respiratory illnesses.
Content may be subject to copyright.

Scientic Reports | (2021) 11:3963 | 
www.nature.com/scientificreports
Comparative analysis
of the alveolar microbiome
in COPD, ECOPD, Sarcoidosis,
and ILD patients to identify
respiratory illnesses specic
microbial signatures
Shashank Gupta1,6,7, Malini Shari2,7, Gaura Chaturvedi3,4,7, Agrima Sharma2,
Nitin Goel5, Monika Yadav6, Martin S. Mortensen1, Søren J. Sørensen1, Mitali Mukerji3 &
Nar Singh Chauhan6*
Studying respiratory illness-specic microbial signatures and their interaction with other micro-
residents could provide a better understanding of lung microbial ecology. Each respiratory illness
has a specic disease etiology, however, so far no study has revealed disease—specic microbial
markers. The present study was designed to determine disease-specic microbial features and their
interactions with other residents in chronic obstructive pulmonary diseases (stable and exacerbated),
sarcoidosis, and interstitial lung diseases. Broncho-alveolar lavage samples (n = 43) were analyzed by
SSU rRNA gene sequencing to study the alveolar microbiome in these diseases. A predominance of
Proteobacteria followed by Firmicutes, Bacteroidetes, Actinobacteria, and Fusobacteria was observed
in all the disease subsets. Shannon diversity was signicantly higher in stable COPD when compared
to exacerbated chronic obstructive pulmonary disease (ECOPD) (p = 0.0061), and ILD patient samples
(p = 0.037). The lung microbiome of the patients with stable COPD was more diverse in comparison
to ECOPD and ILD patients (p < 0.001). Lefse analysis identied 40 disease—dierentiating microbial
features (LDA score (log10) > 4). Species network analysis indicated a signicant correlation (p < 0.05)
of diseases specic microbial signature with other lung microbiome members. The current study
strengthens the proposed hypothesis that each respiratory illness has unique microbial signatures.
These microbial signatures could be used as diagnostic markers to dierentiate among various
respiratory illnesses.
Chronic obstructive pulmonary disease (COPD), interstitial lung diseases (ILD), sarcoidosis are dynamic, debili-
tating lung diseases with multiple comorbidities that aect millions of people worldwide13. COPD is character-
ized by persistent respiratory symptoms and airow limitations due to airway and/or alveolar abnormalities4.
Infections can further weaken the airway function and lead to the exacerbations of COPD5. ILD is a heteroge-
neous group of respiratory disorders presenting with dyspnea, cough, and/or impaired pulmonary function6.
Radiologic and histopathologic evaluation of the lungs shows patterns of inammation and brosis among ILD
patients7,8.
OPEN
          
Denmark.            
India.            
            
     Department of Pulmonary Medicine, Vallabhbhai Patel Chest
   Department of Biochemistry, Maharshi Dayanand University,
   
        
Chaturvedi. *email: nschauhan@mdurohtak.ac.in
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Vol:.(1234567890)
Scientic Reports | (2021) 11:3963 | 
www.nature.com/scientificreports/
ese pathophysiological disorders alter lung physiology and could induce lung microbial dysbiosis9. Studies
have been initiated to dene lung microbiome composition in health and disease subsets to identify microbial
markers for disease prognosis and timely therapeutic interventions1013. Assessment of the temporal and spatial
organization of lung microbes14 and the disease-associated key microbes have also been reported15,16. Several
studies have indicated lung microbial dysbiosis during theonset of various pathophysiological diseases when
compared to healthy controls11,1422. For instance, an abundance of Streptococcus, Corynebacterium, Alloiococ-
cus, Prevotella, Veillonella, Rothia, Porphyromonas, and Moraxella were associated with COPD patients14,15,17.
Haemophilus, Pseudomonas, and Moraxella microbial groups are reported to be enriched in the lung microbiome
during the onset of exacerbated COPD14,15,17,18. Similarly, an abundance of Veillonella, Megasphaera, Streptococcus,
Prevotella, Acidovorax was observed in the lung microbiome of patients with lung cancer19. Lung microbiome of
the asthma patients showed enrichment of Haemophilus, Moraxella, Neisseria, Streptococcus, and Staphylococcus
microbial species20. Streptococcus, Prevotella, Veillonella, Rothia, Actinomyces, Gemella, Granulicatella, Fusobac-
terium, Neisseria, and Atopobium species are abundant in the lung microbiome of the Cystic brosis patients21.
e lung microbiome of the sarcoidosis patients has the enrichment of Atopobium and Fusobacterium species22.
ese studies have indicated lung microbial dysbiosis during the onset of various pathophysiological dis-
orders. Despite varied etiology, dierent pathophysiological conditions showed enrichment of almost simi-
lar microbial groups in each disease subset. Streptococcus, Prevotella, Veillonella, Rothia, and Moraxella are
over-represented in the lung microbiome of patients with COPD, cystic brosis, asthma, and lung cancer1422.
Similarly, Atopobium and Fusobacterium are found enriched within the lung microbiome of patients with cystic
brosis, sarcoidosis, and ILD21,22. ese overlapping results limit the applicability of this information to develop
respiratory illness-specic molecular diagnostics. We hypothesized that patients with stable COPD, ECOPD
(exacerbated COPD), sarcoidosis, and other ILDs have varied disease etiology and each disease could have a
unique lung microbiome prole. e current study was designed to explore the composition and distribution of
microbial phylotypes in the disturbed physiological states of the lungs. A comparative lung microbiome analysis
between diseases, instead of comparison with healthy individuals could help to identify respiratory illness-specic
microbial markers that can be used for diseases-specic diagnosis. is attempt is a rst of its kind to conduct
an alveolar lung microbiome comparison among disease subsets.
Results
Quality of the sequencing dataset. Fourteen patients with stable COPD, thirteen patients with exac-
erbations of COPD, eight patients with ILD, and eight patients with sarcoidosis were enrolled in this study
(Table1, Supplementary TablesS1, S2, S3). A total of 1,282,459 raw reads were passed through the quality lter
and chimera detection resulting in 772,133 (mean per sample: 17,956 ± 1651) high quality and non-chimeric
reads. Based on dada2, amplicons were clustered into 2329 amplicon sequence variants (ASVs). e coverage of
our sequencing was assessed by rarefaction curves (Supplementary Fig.S1). ASV tables were rareed to 4,351
reads per sample to remove the sequencing biases and represent 2162 ASVs across the 43 samples.
Alveolar microbiome composition. Microbial diversity analysis among dierent disease groups iden-
tied the prevalence of 19 bacterial phyla representing 120 families and 286 genera. Proteobacteria held an
overwhelming predominance with an average relative abundance of 58.67%, followed by Firmicutes (20.6%),
Bacteroidetes (15.11%), Actinobacteria (3.13%), and Fusobacteria (1.1%). e remaining 14 phyla were only
observed in a fraction of the samples with a combined average abundance of less than 1% (Fig.1). Proteobacte-
ria were abundant in ILD patients with a relative abundance of 65.71% compared to exacerbated COPD, stable
COPD patients, and sarcoidosis patients, accounting for 56.54%, 59.68%, and 52.77%, respectively. In contrast,
Firmicutes were abundant in stable COPD and ECOPD patients, (17.43% and 23.89% of the relative abundance),
compared to ILD and sarcoidosis patients (Supplementary TableS4). At the family level, we observed the dif-
ference between the disease groups (Table2 & Supplementary TableS5). e most abundant genera in the four
disease groups were visualized in a heat map (Fig.2). A range of genera showed relatively lower abundance but
had high prevalence. ese included Serratia, Prevotella, Streptococcus, Reyranella, Escherichia-Shigella, Neis-
seria, and Ralstonia (Fig.2). Escherichia-Shigella, Haemophilus, Pseudomonas, and Serratia showed high vari-
ability among the four disease groups. Serratia was the most common genus in all groups except stable COPD.
Escherichia-Shigella was also consistent among COPD groups along with Pseudomonas. Enterobacter was dif-
ferentially abundant in stable COPD patients while Klebsiella and Staphylococcus were abundantin ILD patients.
Table 1. Characteristics of the participants in this study. Data are presented as percentage value or mean ± SD
as appropriate. BME: Biomass Exposure. *Data not available.
ECOPD (n = 13) Stable COPD (n = 14) Sarcoidosis (n = 8) ILD (n = 8)
Age, years 63.6 ± 5.79 53.6 ± 14.3 44.5 ± 13.1 53.5 ± 10.9
Sex (% male) 76.9 100 62.5 37.5
Smoker (n) 8 10 (3NA*) 1 (1NA*) 2
BME 5 (2NA*) 0 (3NA*) 3 (1NA*) 2
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Vol.:(0123456789)
Scientic Reports | (2021) 11:3963 | 
www.nature.com/scientificreports/
Alpha diversity among disease groups. We found no signicant dierence in observed richness
between the diseases (Fig. 3A; p = 0.099, Kruskal–Wallis test). However, using Shannon diversity index, we
observed statistically signicant dierences between diseases (Fig.3B; p = 0.001, Kruskal–Wallis test) with post
hoc tests revealing higher diversity in stable COPD compared to ILD (p = 0.037), and sarcoidosis (p = 0.004)
respectively; Mann–Whitney test, Bonferroni adjustment).
Beta diversity among disease groups. Bray–Curtis based PCA plots were analyzed at the genus level
to understand the community ordination (Fig.4). is approach revealed extensive overlap in membership
between the bacterial communities of the ECOPD, stable COPD, ILD, and sarcoidosis disease groups. e rst
two principal components accounted for 29.5% of variance explained, but we did not observe clear clustering.
e PERMANOVA test was used to assess how much the overall variation could be explained in groups, indicat-
ing no notable separation among the groups (p = 0.0610).
Microbial taxa associated with disease groups. LEfSe identied 40 discriminative features, out of
which, thirty taxa were discriminative for stable COPD patients, four taxa for ILD patients, and three taxa
for ECOPD and sarcoidosis patients (Fig.5). Taxa belonging to Firmicutes were signicantly more abundant
(p < 0.05) among ECOPD patients. Proteobacteria were more abundant among ILD patients, while Actinobac-
teria and Proteobacteria were signicantly more abundant (p < 0.05) in sarcoidosis patients. On the other hand,
Chlamydiae along with Firmicutes and Proteobacteria were signicantly more abundant (p < 0.05) in EC OPD
patient’s lungs (Supplementary TableS6). In ECOPD patients, the microbiome was characterized by a prepon-
derance of Streptococcus (LDA score [log10] > 4), whereas in the stable COPD patients, there was a preponder-
ance of Actinobacillus (LDA score [log10] > 4). However, ILD patient`s microbiome showed a very high abun-
dance of Haemophilus (LDA score [log10] > 4), while Corynebacterium was abundant in the sarcoidosis patient`s
(LDA score [log10] > 3).
Figure1. Microbiota composition at phylum level in each disease group. Stacked bar plot showing the mean
relative abundance, at phylum level, for each disease. Phyla with a mean relative abundance below 1% for all
diseases were excluded from the plot.
Table 2. e ve families most commonly identied from each disease groups and their percentage.
Family Exacerbation
COPD Family ILD Family Stable COPD Family Sarcoidosis
Enterobacteriaceae 19.53 Enterobacteriaceae 22.98 Pasteurellaceae 12.61 Prevotellaceae 13.43
Streptococcaceae 11.69 Pasteurellaceae 15.36 Reyranellaceae 11.90 Burkholderiaceae 12.09
Prevotellaceae 8.02 Prevotellaceae 8.30 Prevotellaceae 10.92 Streptococcaceae 11.34
Pseudomona-
daceae 7.96 Staphylococcaceae 7.77 Burkholderiaceae 10.10 Enterobacteriaceae 10.14
Pasteurellaceae 7.81 Streptococcaceae 7.34 Enterobacteriaceae 9.61 Reyranellaceae 8.62
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Vol:.(1234567890)
Scientic Reports | (2021) 11:3963 | 
www.nature.com/scientificreports/
Figure2. Heat map showing 50 most abundant genera in the four groups of samples. Columns represent the
groups and rows the genera and their relative abundance. e color key represents the relative abundance of
each genus.
Figure3. Observed richness (A) and Shannon diversity index (B). Comparing two groups using Mann–
Whitney test; ^comparing two or more groups using Kruskal–Wallis test. p < 0.05 denotes statistical signicance.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Vol.:(0123456789)
Scientic Reports | (2021) 11:3963 | 
www.nature.com/scientificreports/
Functional annotation of the lung microbiome. Predicated phenotypes based on taxonomic classi-
cation indicated that the majority of microbes were mesophilic (> 65%), gram-negative (> 74%) Bacillus (> 62%).
A majority of the lung microbes were generally considered to be associated with humans (> 60%), while the
remaining microbes were not commonly associated with a specic environment (> 30%). e majority of the
identied lung microbes predict a higher potential for the onset of various human disorders. e percentage of
Figure4. Principal component analysis. Dots represent samples and color represents dierent disease groups.
First two principal components (PC) explained 29.5% of the variance.
Figure5. LDA shows distinct lung microbiome composition associated with ECOPD, stable COPD, ILD and
sarcoidosis. LDA scores as calculated by LEfSe of taxa dierentially abundant in dierent disease group. Only
taxa with LDA scores of more than three and p value < 0.05 are shown here.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Vol:.(1234567890)
Scientic Reports | (2021) 11:3963 | 
www.nature.com/scientificreports/
such microbes was higher in the samples from sarcoidosis and other ILD groups (> 71–85%) as compared with
the COPD group (62–70%). e majority of the microbes from COPD lung were found to play a signicant role
in ammonia oxidation, sulfur metabolism, and complex carbohydrate catabolism. Lung microbes in sarcoidosis
and other ILD groups seemed to play a signicant metabolic role in polyphenol metabolism and dehalogenation
reactions in addition to the function carried out by COPD inherent microbes.
Core microbiome and its association with diseases. Common members of a microbial commu-
nity oen perform moderate functioning of the host-microbial symbiotic system. We estimated a high degree
of similarity between the core microbiome for each of the four diseases. Ten ASVs were shared among all of
them; these belonged to the genera Reyranella, Ochrobactrum, Mesorhizobium, Ralstonia, Achromobacter, Pseu-
domonas, Streptococcus, Granulicatella, and two unclassied genera belonging to Xanthobacteraceae. Moreover,
there were 11 ASVs unique to patients with sarcoidosis, three to ECOPD, 12 ASVs to stable COPD, and only two
ASVs to ILD (Supplementary TableS7, Supplementary Fig.S3).
To gain insight into the interaction between bacterial species in the lung microbiome, we performed a spe-
cies network analysis (only correlations with an absolute value of 0.60, p < 0.05). Examination of the microbial
network revealed that Xanthobacteraceae were highly connected with multiple other ASVs among all the disease
groups (Fig.6).
In ECOPD patients Escherichia-Shigella (Fig.6A) positively correlated with Subdoligranulum, Catenibacte-
rium, and negatively correlated with Chryseobacterium and Prevotella. Conversely, one of the most abundant
genera i.e., Streptococcus showed a negative correlation with Dialister, Ralstonia, and Christensenellaceae R-7
groups. However, the rest of the most abundant genera i.e., Serratia, Haemophilus, and Pseudomonas did not
correlate with any other genera. In the stable COPD subjects (Fig.6B), the most abundant genus was Reyranella,
which was negatively correlated with Haemophilus and Streptococcus whereas positively correlated with seven
other genera that belong to Achromobacter, Chryseobacterium, Mesorhizobium, Elizabethkingia, Sphingobacte-
rium, Ralstonia, Xanthobacteraceae family. e other two most abundant genera were Escherichia-Shigella and
Figure6. Bacterial co-existence and co-exclusion relationships with ASVs and dierent diseases. Each node
represents ASVs. Each edge represents a signicant correlation colored by co-existence (orange) or co-exclusion
relationships (blue).e size of the node corresponds to its degree of connectivity, while edge lengths are
arbitrary. ECOPD (A), stable COPD (B), sarcoidosis (C), and ILD (D).
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Vol.:(0123456789)
Scientic Reports | (2021) 11:3963 | 
www.nature.com/scientificreports/
Haemophilus, both of which belong to the Proteobacteria phylum. Escherichia-Shigella showed a strong positive
correlation with Dialister and Agathobacter, whereas it was negatively correlated with Elizabethkingia. On the
other hand, Haemophilus showed no correlation with any other genera.
e sarcoidosis disease group (Fig.6C) had the most negative connections with other members of the micro-
biota. Most abundant genera belonged to Serratia, Prevotella, Streptococcus, Reyranella, and Ralstonia. Except
for the Serratia, which did not show any correlation, all other dominant genera showed a strong correlation
with other genera in the group. Prevotella and Streptococcus genera were mostly negatively correlated, whereas
Reyranella and Ralstonia showed a positive correlation.
In the ILD subjects (Fig.6D), the most abundant genus, Hemophilus, did not show any correlation with others.
Moreover, Streptococcus sp. showed correlation with 11 out of 14 genera in this group. It was found to be posi-
tively correlated with three genera and negatively correlated with eight dierent genera. Despite the abundance
of the Klebsiella genera, it was positively correlated only with Pseudomonas aeruginosa and negatively correlated
with Mesorhizobium sp.
Discussion
e present results conrm earlier reports that the human respiratory tract contains a diverse microbiome23. Lung
microbiome composition is inuenced by the onset of respiratory illness, as well as with the usage of steroids,
aerosols, and antibiotics24,25. During airway diseases, lung microbiome can exacerbate the diseases, leading to
increased levels of morbidity and mortality15. Using the NGS platform, diverse non-cultivable bacteria were found
in the respiratory tract10. is study has explored the alveolar microbiome of the patients with COPD (stable and
exacerbation), ILD, and sarcoidosis to understand the similarities & dierences between the disease-associated
microbial phylotypes. We have accessed the composition, diversity, and core microbiome for each disease and
identied which aspects are related to lung diseases in general and which are diseases specic. Additionally, to
our knowledge, this is the rst alveolar microbiome report among the Indian population.
It was found that the microbial members of the bronchial microbiome do not change signicantly in
COPD patients26,27. However, the predominance of Proteobacteria in the present study is in line with previous
studies14,15,17,18. We observed a higher abundance of Firmicutes in stable COPD and ECOPD subjects compared
to ILD and sarcoidosis groups. We found a higher alpha diversity in the stable COPD group in comparison to
the other groups. A similar observation was found in previous studies14,28. ree taxa show a signicant abun-
dance in ECOPD patients- Streptococcus, Faecalibacterium, and Coprococcus. Streptococcus is the most widely
recognized microbe found in COPD patients14,17. Faecalibacterium is a common resident of the human gut, while
Coprococcus is usually found in the sputum, however, what role the latter two play in humans in the human lung
is still unexplored.
Few eorts have been made to explore lung microbiome in the patients with sarcoidosis and other types
of ILD23,29 and these studies were unable to dierentiate the lung microbiome structure among these disease
subtypes23. We were able to identify dierences in their microbial composition. We found an increase in Act-
inobacteria and a decrease in Proteobacteria in sarcoidosis patients as compared to ILD subjects. e increased
relative abundance of Streptococcus and Staphylococcus has been reported to contribute to disease progression
in idiopathic pulmonary brosis29. Similarly, we also observed a higher relative abundance of Streptococcus and
Staphylococcus in the ILD groups, as well as an increased alpha diversity in sarcoidosis compared to ILD patients.
Besides, PCA showed dierences in microbial variation between COPD (stable and exacerbation), ILD, and sar-
coidosis patients. Moreover, current data show a signicantly higher abundance of taxa belonging to the genera
Haemophilus, Stenotrophomonas, and Enterobacteriaceae family in the ILD group, whereas Corynebacterium and
Neisseria are more abundant in the sarcoidosis group. However, Haemophilus, known as pathogenic-bacteria, is
usually observed in COPD patients30.
ILD groups have enriched unique microbial groups. Moreover, we also observed a signicantly higher abun-
dance of Haemophilus in stable COPD groups. ese deviations could be seen as a possible outcome of diverse
ethnicity, as commonly observed in other human microbiome studies31,32.
When we compared the core microbiome of COPD (stable and exacerbation), ILD, and sarcoidosis patients,
we observed that eleven taxa were shared among these disease groups. is supports the idea that many features
shared between microbiota dier compositionally between these disease groups. Additionally, the core alveolar
microbiome in respiratory illness was found altered as compared to that of a healthy lung microbiome. Core lung
microbiome of a healthy individual harbors nine microbial genera33,34 of which only Pseudomonas, Streptococ-
cus, Prevotella, and Sphingobacterium were shared within the studied core microbiomes. is result supports
the hypothesis that the microbiome biotransformation may lead to the onset of pathophysiological disorders35.
Furthermore, in the genus-level abundance network analysis, Xanthobacteraceae were highly connected with
multiple other nodes between all the disease groups, indicating it as a keystone microbial taxon. Most of the
genera in stable COPD, ECOPD, ILD, and sarcoidosis patients showed both positive and negative correlation;
however, ECOPD and stable COPD patients showed a more positive correlation. We also noticed many potential
clinically relevant taxa such as Streptococcus sp. (observed in all diseased groups), Haemophilus sp. (observed
in stable COPD patients), Escherichia-Shigella sp. (observed in stable and ECOPD patients), and Pseudomonas
aeruginosa (observed in stable COPD and ILD patients), show correlation with other taxa1422. However, cor-
relations between taxa are not proof of functional relationships between members of the community. erefore,
further studies are required to focus on the functional role of such taxa found within these communities.
e present study has used very stringent inclusion criteria for patients screening. ough it has allowed
the identication of unbiased disease-specic samples, but also reduced the number of samples (~ 50 fold) for
the downstream analysis. Due to the limitation of samples, the current study is slightly underpowered and
higher numbers of samples are required to statistically strengthen the proposed claims. However, this pilot
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Vol:.(1234567890)
Scientic Reports | (2021) 11:3963 | 
www.nature.com/scientificreports/
study provides preliminary evidence in support of the hypothesis that there are diseases specic dierences in
the microbiomes of COPD (stable and exacerbation), ILD, and sarcoidosis patients.
Moreover, our study adds further insights into the microbial composition of the lung microbiota of Indian
patients suering from COPD, ILD, or sarcoidosis. Each disease subtype has dierential microbial phylotypes that
correlate with the abundance prole of other microbial taxa to possibly remodel the lung microbiome structure.
is study enhances our understanding of lung microbial ecology in various respiratory illnesses. Identied
microbial signatures could be utilized as prognostic markers for respiratory disease diagnosis and therapeutics.
Methods
Patient recruitment and Broncho-Alveolar Lavage (BAL) sample collection. is study was
approved by the institutional human ethics committee, Vallabhbhai Patel Chest Institute, University of Delhi,
Delhi, India. Adult patients with stable COPD, both male and female with a history of smoking (> 10 pack-years)
and/or biomass fuel exposure (> 10years) attending Vallabhbhai Patel Chest Institute were invited to participate
in the study. Patients classied as having exacerbated COPD if they presented within increased cough or sputum
production. All patients suspected of ILD underwent clinical evaluation including detailed history and exami-
nation. e diagnosis of ILD was made based on the American oracic Society/European Respiratory Society
International Multidisciplinary Consensus Classication of Idiopathic Interstitial Pneumonia 2001 guidelines36.
Similarly, the diagnosis of sarcoidosis was made based on the compatible clinical, radiological, laboratory, and
where available, histopathological parameters, as per the joint statement of the American oracic Society, the
European Respiratory Society, and the World Association of Sarcoidosis, and Other Granulomatous Disorders
(ATS/ERS/WASOG), and also the simultaneous exclusion of any other cause of the granulomatous disorder37.
Since Bronchoscopy was used as a diagnostic criterion, patients were excluded if they have taken antibiotics or
steroids prior to their inclusion.
Aer providing written informed consent, the patients underwent bronchoscopy as per the British oracic
Society (BTS) guidelines for bronchoscopy 2013. is study included bronchoalveolar lavage collected from
ECOPD (n = 13), stable COPD (n = 14), ILD (n = 8), and sarcoidosis (n = 8) patients, as well as saline buer passed
through a bronchoscope to be used as negative control.
Metagenomic DNA isolation from BAL samples and 16S rRNA gene sequencing. Each BAL
sample (1.5ml) was centrifuged at 13,000rpm for 1min to collect the bacterial pellet. e bacterial pellet was
processed with the alkaline lysis method38. Metagenomic DNA quantication was performed with Qubit 2.0
using high sensitivity DNA quantication kit (Invitrogen, USA). All samples were diluted to a DNA concentra-
tion of 25ngμl-1. e V3-V4 region of the bacterial 16S rRNA gene was amplied using gene-specic primer
sequences (Fwd 5-TCG TCG GCA GCG TCA GAT GTG TAT AAG AGA CAG CCT ACGGGNGGC WGC AG-3` and
Rev 5-GTC TCG TGG GCT CGG AGA TGT GTA TAA GAG ACA GGA CTACHVGGG TAT CTA ATCC-3`)39. Nex-
tera XT Index kit (Illumina, USA) was used to index each sample during library preparation following Illumina
technology workow document (www.suppo rt.illum ina.com). e indexed 16S rRNA amplicons were pooled in
equimolar concentration followed by paired-end sequencing on the Illumina MiSeq platform using paired-end
MiSeq 600 cycle V3 sequencing Kit following manufacturer instructions39.
Sequence and statistical analyses. Primers were removed from the MiSeq demultiplex FASTQ using
“cutadapt"40. Further, reads were analyzed by the QIIME2 pipeline41 through dada242 to infer the presence and
relative abundance of amplicon sequence variants (ASVs) across the samples. Based on data-derived rates of
Illumina sequencing errors, dada2 estimated an abundance distribution of distinct ASVs, which may dier by
only a single nucleotide. Using read quality scores for the dataset, forward and reverse reads were truncated at
270bp and 200bp, followed by trimming the 5`-end till 6bp for both forward and reverse reads, respectively;
other quality parameters used dada2 default values. Taxonomy was assigned using a pre-trained Naïve Bayes
classier (Silva database, release 132)43. e rarefaction curves (Supplementary Fig.S1) show the observed rich-
ness and Shannon diversity. To avoid the bias due to sampling depth, we rareed our dataset to 4351 high-quality
sequences per sample (90% of the minimum sample reads) using an in-house script. e function rarees each
sample 100 times, calculates the mean and standard deviation for observed richness and Shannon diversity
index, and returns the ASV counts for the iteration with the lowest mean Bray–Curtis distance among the 100
iterations.
All downstream analyses were performed on this rareed ASVs table unless otherwise mentioned. We used
two diversity indices i.e., observed richness, the number of taxa present in a sample at a particular taxonomic
level, and Shannon diversity index, a composite measure of both species richness and evenness. Alpha and beta
diversity was calculated using phyloseq v1.20.044 and visualized with ggplot2 v2.2.1in R v3.4.1.45. Comparison
of community richness and diversity between the four disease groups was assessed by the Kruskal–Wallis test,
with post hoc tests, performed using the Mann–Whitney test with Bonferroni adjustment applied. Signicance
testing between the disease groups for beta diversity was assessed using the PERMANOVA (permutational
multivariate analysis of variance). LEfSe was used to identify the microbiological markers associated with stable
COPD, ECOPD, ILD, and sarcoidosis disease groups by linear discriminate analysis (LDA) eect size of 3, and
for multiclass analysis one-against-all option was used with default parameters46.
Functional annotation of the lung microbiome. e taxonomically aliated OTU table was used to
annotate physiological functions and the lifestyle of human lung microbes associated with various disease sub-
sets with the METAGENassist server47.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Vol.:(0123456789)
Scientic Reports | (2021) 11:3963 | 
www.nature.com/scientificreports/
Core microbiota and bacterial co-occurrence. Considering the variable nature of metagenomic com-
positional data, we performed further analysis only for conserved taxa. Towards this, we estimated core micro-
biota within the samples with a presence in at least 50% of the samples within each disease group on the non-
rareed data. We examined co-occurrence patterns using network analysis on the core microbiota using Sparse
Correlations for Compositional data algorithm (SparCC) with a bootstrap procedure repeated 100 times48. Co-
occurrence was considered robust when the correlations (either positive or negative) were both ≥ 0.6 and corre-
lation coecients with two-tailed p values smaller than 0.05. e correlation was imported into Cytoscape v3.6.0
to build the co-occurrence network, where each node represents taxa and the edges between the nodes represent
the correlation coecients between taxa49.
Ethics approval and consent to participate. is study was carried out by following the recommenda-
tions of the Indian Council of Medical Research, India guidelines for biomedical research, with written informed
consent from all subjects. All subjects gave written informed consent under the Declaration of Helsinki. e
protocol was approved by the Institutional human ethics committee, Vallabhbhai Patel Chest Institute, Univer-
sity of Delhi, Delhi, India.
Consent for publication. e manuscript has been read and approved for submission by the named
authors.
Data availability
Sequence data generated in this study have been deposited at NCBI with an SRA submission ID SUB4935309
and Bio project accession ID PRJNA512576.
Received: 16 July 2020; Accepted: 1 February 2021
References
1. Schwarzkopf, L., Witt, S., Waelscher, J., Polke, M. & Kreuter, M. Associations between comorbidities, their treatment and survival
in patients with interstitial lung diseases—a claims data analysis. Respir. Res. 19, 73. https ://doi.org/10.1186/s1293 1-018-0769-0
(2018).
2. May, S. M. & Li, J. T. C. Burden of chronic obstructive pulmonary disease: Healthcare costs and beyond. Allergy Asthma. Proc. 36,
4–10. https ://doi.org/10.2500/aap.2015.36.3812 (2015).
3. Gerke, A. K., Judson, M. A., Cozier, Y. C., Culver, D. A. & Koth, L. L. Disease burden and variability in sarcoidosis. Ann. Am.
orac. Soc. 14, S421–S428. https ://doi.org/10.1513/Annal sATS.20170 7-564OT (2017).
4. Papi, A. et al. Infections and airway inammation in chronic obstructive pulmonary disease severe exacerbations. Am. J. Respir.
Crit. Care. Med. 173, 1114–1121. https ://doi.org/10.1164/rccm.20050 6-859OC (2006).
5. Sethi, S. & Murphy, T. F. Bacterial infection in chronic obstructive pulmonary disease in 2000: A state of the art review. Clin.
Microbiol. Rev. 14, 336–363. https ://doi.org/10.1128/CMR.14.2.336-363.2001 (2001).
6. Salisbury, M. L., Han, M. K., Dickson, R. P. & Molyneaux, P. L. e microbiome in interstitial lung disease: From pathogenesis to
treatment target. Curr. Opin. Pulm. Med. 23, 404–410. https ://doi.org/10.1097/MCP.00000 00000 00039 9 (2017).
7. Segal, L. N., Rom, W. N. & Weiden, M. D. Lung microbiome for clinicians. New discoveries about bugs in healthy and diseased
lungs. Ann. Am. orac. Soc. 11, 108–116. https ://doi.org/10.1513/Annal sATS.20131 0-339FR (2014).
8. Travis, W. D. et al. An ocial American oracic Society/European Respiratory Society statement: Update of the international
multidisciplinary classication of the idiopathic interstitial pneumonias. Am. J. Respir. Crit. Care. Med. 188, 733–748. https ://doi.
org/10.1164/rccm.20130 8-1483S T (2013).
9. Pragman, A. A. et al. Chronic obstructive pulmonary disease upper airway microbiota alpha diversity is associated with exacerba-
tion phenotype: A case-control observational study. Respir. Res. 20, 114. https ://doi.org/10.1186/s1293 1-019-1080-4 (2019).
10. Aho, V. T. E. et al.e microbiome of the human lower airways: A next generation sequencing perspective. World Allergy Organ
J. 8, 1–13. https ://doi.org/10.1186/s4041 3-015-0074-z (2015).
11. Kiley, J. P. Advancing respiratory research. Chest 140, 497–501. https ://doi.org/10.1378/chest .11-0774 (2011).
12. Dickson, R. P. et al. Spatial variation in the healthy human lung microbiome and the adapted island model of lung biogeography.
Ann. Am. orac. Soc. 12, 821–830. https ://doi.org/10.1513/Annal sATS.20150 1-029OC (2015).
13. Dickson, R. P., Martinez, F. J. & Hunagle, G. B. e role of the microbiome in exacerbations of chronic lung diseases. Lancet 384,
691–702. https ://doi.org/10.1016/S0140 -6736(14)61136 -3 (2014).
14. Pragman, A. A., Kim, H. B., Reilly, C. S., Wendt, C. & Isaacson, R. E. e lung microbiome in moderate and severe chronic obstruc-
tive pulmonary disease. PLoS ONE 7, e47305. https ://doi.org/10.1371/journ al.pone.00473 05 (2012).
15. O’Dwyer, D. N., Dickson, R. P. & Moore, B. B. e lung microbiome, immunity, and the pathogenesis of chronic lung disease. J
Immunol. 196, 4839–4847. https ://doi.org/10.4049/jimmu nol.16002 79 (2016).
16. Sibley, C. D. et al. Culture enriched molecular proling of the cystic brosis airway microbiome. PLoS ONE 6, e22702. https ://doi.
org/10.1371/journ al.pone.00227 02 (2011).
17. Pragman, A. A. et al. e lung tissue microbiota of mild and moderate chronic obstructive pulmonary disease. Microbiome. 6, 7.
https ://doi.org/10.1186/s4016 8-017-0381-4 (2018).
18. Millares, L. et al. Bronchial microbiome of severe COPD patients colonised by Pseudomonas aeruginosa. Eur. J. Clin. Microbiol.
Infect. Dis. 33, 1101–1111. https ://doi.org/10.1007/s1009 6-013-2044-0 (2014).
19. Ramírez-Labrada, A. G. et al. e inuence of lung microbiota on lung carcinogenesis, immunity, and immunotherapy. Trends.
Cancer. 6, 86–97. https ://doi.org/10.1016/j.treca n.2019.12.007 (2020).
20. Hilt y, M. et al. Disordered microbial communities in asthmatic airways. PLoS ONE 5, e8578. https ://doi.org/10.1371/journ
al.pone.00085 78 (2010).
21. Surette, M. G. e cystic brosis lung microbiome. Ann. Am. orac. Soc. 1, S61–S65. https ://doi.org/10.1513/Annal sATS.20130
6-159MG (2014).
22. Zimmermann, A. et al. Atopobium and Fusobacterium as novel candidates for sarcoidosis-associated microbiota. Eur. Re spir. J. 50,
1600746. https ://doi.org/10.1183/13993 003.00746 -2016 (2017).
23. B ecker, A. et al. e composition of the pulmonary microbiota in sarcoidosis—an observational study. Respir. Res. 20, 46. https ://
doi.org/10.1186/s1293 1-019-1013-2 (2019).
Content courtesy of Springer Nature, terms of use apply. Rights reserved

Vol:.(1234567890)
Scientic Reports | (2021) 11:3963 | 
www.nature.com/scientificreports/
24. Le, N. V. et al. Modulation of pulmonary microbiota by antibiotic or probiotic aerosol therapy: A strategy to promote immuno-
surveillance against lung metastases. Cell R ep. 24, 3528–3538. https ://doi.org/10.1016/j.celre p.2018.08.090 (2018).
25. Contoli, M. et al. Long-term eects of inhaled corticosteroids on sputum bacterial and viral loads in COPD. Eur. Respir. J. 50,
1700451. https ://doi.org/10.1183/13993 003.00451 -2017 (2017).
26. Huang, Y. J. et al. Airway microbiome dynamics in exacerbations of chronic obstructive pulmonary disease. J. Clin. Microbiol. 52,
2813–2823. https ://doi.org/10.1128/JCM.00035 -14 (2014).
27. Molyneaux, P. L. et al. Outgrowth of the bacterial airway microbiome aer rhinovirus exacerbation of chronic obstructive pulmo-
nary disease. Am. J. Respir. Crit. Care. Med. 188, 1224–1231. https ://doi.org/10.1164/rccm.20130 2-0341O C (2013).
28. Sze, M. A. et al. e lung tissue microbiome in chronic obstructive pulmonary disease. Am. J. Respir. Crit. Care. Med. 185,
1073–1080. https ://doi.org/10.1164/rccm.20111 1-2075O C (2012).
29. Han, M. K. et al. Lung microbiome and disease progression in idiopathic pulmonary brosis: An analysis of the COMET study.
Lancet. Respir. Med. 2, 548–556. https ://doi.org/10.1016/S2213 -2600(14)70069 -4 (2014).
30. Osman, K. L. et al. Patients with chronic obstructive pulmonary disease harbour a variation of Haemophilus species. Sci. Rep. 8,
14734. https ://doi.org/10.1038/s4159 8-018-32973 -3 (2018).
31. Chauhan, N. S. et al. Western Indian rural gut microbial diversity in extreme Prakriti endo-phenotypes reveals signature microbes.
Front. Microbiol. 9, 118. https ://doi.org/10.3389/fmicb .2018.00118 (2019).
32. Verma, M. K. et al. Functional metagenomics identies novel genes ABCTPP, TMSRP1 and TLSRP1 among human gut enterotypes.
Sci. Rep. 8, 1397. https ://doi.org/10.1038/s4159 8-018-19862 -5 (2019).
33. Beck, J. M., Young, V. B. & Hunagle, G. B. e microbiome of the lung. Transl. Res. 160, 258–266. https ://doi.org/10.1016/j.
trsl.2012.02.005 (2012).
34. Erb-Downward, J. R. et al. Analysis of the lung microbiome in the “healthy” smoker and in COPD. PLoS ONE 6, e16384. https ://
doi.org/10.1371/journ al.pone.00163 84 (2011).
35. Duvallet, C., Gibbons, S. M., Gurry, T., Irizarry, R. A. & Alm, E. J. Meta-analysis of gut microbiome studies identies disease-specic
and shared responses. Nat. Commun. 8, 1784. https ://doi.org/10.1038/s4146 7-017-01973 -8 (2017).
36. e American oracic Society/European Respiratory Society International. Multidisciplinary consensus classication of the
idiopathic interstitial pneumonias. Am. J. Respir. Crit. Care Med. 165, 277–304 (2002).
37. American oracic Society, European Respiratory Society, World Association of sarcoidosis and Other Granulomatous Disorders.
Statement on sarcoidosis. Am. J. Respir. Crit. Care Med. 160, 736–755 (1999).
38. Kumar, J. et al. An improved methodology to overcome key issues associated with the methods of human fecal metagenomic DNA
extraction. Genom. Proteom. Bioinf. 14, 371–378. https ://doi.org/10.1016/j.gpb.2016.06.002 (2016).
39. Klindworth, A. et al. Evaluation of general 16S ribosomal RNA gene PCR primers for classical and next-generation sequencing-
based diversity studies. Nucleic. Acids. Res. 7, e1. https ://doi.org/10.1093/nar/gks80 8 (2013).
40. Martin, M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet J. 17, 1. https ://doi.org/10.14806
/ej.17.1.200 (2011).
41. B olyen, E. et al. Reproducible, interactive, scalable, and extensible microbiome data science using QIIME 2. Nat. Biotechnol. 37,
852–857. https ://doi.org/10.1038/s4158 7-019-0209-9 (2020).
42. Callahan, B. J. et al. DADA2: High-resolution sample inference from Illumina amplicon data. Nat. Methods 13, 581–583. https ://
doi.org/10.1038/nmeth .3869 (2016).
43. Quast, C. et al. e SILVA ribosomal RNA gene database project: Improved data processing and web-based tools. Nucleic. Acids.
Res. 41, D590–D596. https ://doi.org/10.1093/nar/gks12 19 (2013).
44. McMurdie, P. J. & Holmes, S. Phyloseq: An R package for reproducible interactive analysis and graphics of microbiome census
data. PLoS ONE 8, e61217. https ://doi.org/10.1371/journ al.pone.00612 17 (2013).
45. Wickham, H. gg plot2, Elegant Graphics for Data Analysis (Springer, New York, 2016). https ://doi.org/10.1007/978-3-319-24277 -4.
46. S egata, N. et al. Metagenomic biomarker discovery and explanation. Genome. Biol. 12, R60. https ://doi.org/10.1186/gb-2011-12-
6-r60 (2011).
47. Arndt, D. et al. METAGENassist: A comprehensive web server for comparative metagenomics. Nucleic. Acids. Res. 40, W88-95.
https ://doi.org/10.1093/nar/gks49 7 (2012).
48. Friedman, J. & Alm, E. J. Inferring correlation networks from genomic survey data. PLoS Comput. Biol. 8, e1002687. https ://doi.
org/10.1371/journ al.pcbi.10026 87 (2012).
49. Otasek, D., Morris, J. H., Bouças, J., Pico, A. R. & Demchak, B. Cytoscape automation: Empowering workow-based network
analysis. Genome Biol. 20, 185. https ://doi.org/10.1186/s1305 9-019-1758-4 (2019).
Acknowledgements
We hereby acknowledge the sequencing facility support from CSIR-IGIB and UGC fellowship to GC.
Author contributions
N.G., M.S., and A.S. performed sample recruitment and DNA isolation. G.C., M.Y., M.M., and A.S. performed
sequencing of samples. S.G., N.S.C., S.J.S., and M.S.M. interpreted the data. S.G., M.S., M.M., and N.S.C. wrote
the manuscript. is project was conceived and designed by M.S. and N.S.C. All the authors have read, revised,
and approved the manuscript.
Funding
e current study was funded by Department of Biotechnology grant vide BT/PR10801/MED/29/826/2014.
Competing interests
e authors declare no competing interests.
Additional information
Supplementary Information e online version contains supplementary material available at https ://doi.
org/10.1038/s4159 8-021-83524 -2.
Correspondence and requests for materials should be addressed to N.S.C.
Reprints and permissions information is available at www.nature.com/reprints.
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and
institutional aliations.
Content courtesy of Springer Nature, terms of use apply. Rights reserved

Vol.:(0123456789)
Scientic Reports | (2021) 11:3963 | 
www.nature.com/scientificreports/
Open Access is article is licensed under a Creative Commons Attribution 4.0 International
License, which permits use, sharing, adaptation, distribution and reproduction in any medium or
format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the
Creative Commons licence, and indicate if changes were made. e images or other third party material in this
article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the
material. If material is not included in the article’s Creative Commons licence and your intended use is not
permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from
the copyright holder. To view a copy of this licence, visit http://creat iveco mmons .org/licen ses/by/4.0/.
© e Author(s) 2021
Content courtesy of Springer Nature, terms of use apply. Rights reserved
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... The blood microbiome is now being recognized as potentially affected by various systemic and inflammatory diseases, as microbial components and metabolites were identified in the blood and can directly interact with the immune system [5]. There is a growing body of evidence suggesting that alterations in microbiome, or dysbiosis, could play a role in sarcoidosis [6,7]. Dysbiosis may influence sarcoidosis development through several mechanisms, including immune dysregulation, metabolic shifts, or increased permeability of mucosal barriers that allow the translocation of bacteria or bacterial products into the bloodstream [8][9][10]. ...
... Currentlyin sarcoidosis research, bronchoalveolar lavage (BAL) is the primary sample type used for comparing microbiome composition in patients with sarcoidosis and control subjects [6,7,33]. Table 1. ...
... To our knowledge, two studies have specifically addressed the characterization of blood microbiota in healthy adults:a study conducted by Paise et al., in 2016 [35], and another by Panaiotov et al., in 2021 [9]. Both investigations reported similar findings regarding [6,7,33,34]. Notably, the presence of potentially pathogenic microbial species such as Mycobacterium and Neisseria spp., as well as commensals associated with sarcoidosis like Atopobium [7] and Cutibacterium, have been documented. ...
Article
Full-text available
Background: Sarcoidosis is a chronic inflammatory disease that can affect multiple organs. The aetiology of sarcoidosis is not fully understood, but there is increasing evidence that the microbiome may play a role. The blood microbiome is a collection of microorganisms that live in the bloodstream. It is a complex and dynamic community that is influenced by a variety of factors, including the host’s lifestyle and pathology. Recent studies have shown that people with sarcoidosis have alterations in their blood microbiome. These alterations include changes in the diversity, richness, and evenness of the microbial community. The abundance measures by which the blood microbiome diversity may detect instances of dysbiosis related to sarcoidosis aetiology. It should be clearly distinguished from microbiome changes related to unspecific inflammation or sepsis. However, the available evidence suggests that the microbiome may be a promising target for therapeutic interventions. Aim: The primary goal of this review was to assess and compare the existing metrics of microbiome composition and diversity as established by metagenomic analyses. Additionally, we aim to elucidate the potential causal relationship between these measures, the phenomenon of blood microbiome dysbiosis and the pathogenesis of sarcoidosis. Conclusion: In the present review, we investigated alpha diversity measures as characteristics of microbiome communities, examining their potential as indicators of dysbiosis, and the probablemechanisms of microbiome participation. A descriptive qualitative comparison was conducted between lung microbiome data of sarcoidosis patients and blood microbiome data of healthy adults. This comparison elucidates common taxa between the two microbiomes and identifies taxa potentially involved in sarcoidosis.
... Granuloma is a particular type of inflammatory response that involves the formation of a mass of immune cells known as granulocytes, which form a protective barrier around a foreign substance or body [3]. Sarcoidosis is a granulomatous multisystem inflammatory disease and has both immunological and genetic components [4] however, recent studies on bronchoalveolar lavage (BAL) indicate that the disease may also be linked to lung microbiota alterations [5,6] or antigen-detected pathogens as a risk factor [7]. It is hypothesized that the etiology of sarcoidosis is associated with an autoimmune reaction, genetic predisposition, bacterial infection, or host-microbiome dysbiosis [8]. ...
... Microbial dysbiosis can be marked by microbial species or genera that are either enriched or depleted due to disease. Recent metagenome analyses of BAL fluid [5,6,15] and tissue biopsy [12] have identified genera linked to microbial dysbiosis in lung disorders. The studies by Zimmermann et al. and Gupta et al. have provided valuable insights into the lung microbiome, studying BAL samples, associated with sarcoidosis [5,6]. ...
... Recent metagenome analyses of BAL fluid [5,6,15] and tissue biopsy [12] have identified genera linked to microbial dysbiosis in lung disorders. The studies by Zimmermann et al. and Gupta et al. have provided valuable insights into the lung microbiome, studying BAL samples, associated with sarcoidosis [5,6]. Zimmermann et al. identified Atopobium and Fusobacterium as novel candidates for sarcoidosis-associated microbiota in the lung of the patients. ...
Article
Full-text available
Introduction Single microbial pathogens or host-microbiome dysbiosis are the causes of lung diseases with suspected infectious etiology. Metagenome sequencing provides an overview of the microbiome content. Due to the rarity of most granulomatous lung diseases collecting large systematic datasets is challenging. Thus, single-patient data often can only be summarized visually. Objective To increase the information gain from a single-case metagenome analysis we suggest a quantitative and qualitative approach. Results The 16S metagenomic results of 7 patients with pulmonary sarcoidosis were compared with those of 22 healthy individuals. From lysed blood, total microbial DNA was extracted and sequenced. Cleaned data reads were identified taxonomically using Kraken 2 software. Individual metagenomic data were visualized with a Sankey diagram, Krona chart, and a heat-map. We identified five genera that were exclusively present or significantly enhanced in patients with sarcoidosis - Veillonella, Prevotella, Cutibacterium, Corynebacterium, and Streptococcus. Conclusions Our approach can characterize the blood microbiome composition and diversity in rare diseases at an individual level. Investigation of the blood microbiome in patients with granulomatous lung diseases of unknown etiology, such as sarcoidosis could enhance our comprehension of their origin and pathogenesis and potentially uncover novel personalized therapeutics. Keywords: Lung diseases, Sarcoidosis, Microbiome, Metagenome analysis, Visualization, Sankey diagram
... A pathogen invasion or gut microbiota dysbiosis evokes healthy cross-talk. It induces a host inflammatory response [10], leading to the onset of various illnesses and pathologies, including obesity, inflammatory bowel disease (IBD), autoimmune diseases, allergies, respiratory diseases, and neurological conditions [11,12]. Host-microbiota interactions were a prime target for overcoming gut dysbiosis-derived human illnesses [13]. ...
... Several studies have shown that a variety of microorganisms reside in the upper and lower respiratory tracts (LRTs), and the composition of these microbial communities can be altered in different respiratory disease states (6). The LRT micro-environment can be associated with lung disease including ILDs (7,8). Previous studies have shown that the lung microbiome of patients with idiopathic pulmonary fibrosis (IPF), a type of ILD, differs from that of healthy individuals (9). ...
Article
Full-text available
Background Interstitial lung disease (ILD) is a common complication of idiopathic inflammatory myopathy (IIM), which is one of the connective tissue diseases (CTD). It can lead to poor prognosis and increased mortality. However, the distribution and role of the lower respiratory tract (LRT) microbiome in patients with IIM-ILD remains unclear. This study aimed to investigate the microbial diversity and community differences in bronchoalveolar lavage fluid (BALF) in patients with IIM-ILD. Methods From 28 June 2021 to 26 December 2023, 51 individual BALF samples were enrolled, consisting of 20 patients with IIM-ILD, 16 patients with other CTD-ILD (including 8 patients with SLE and 8 with RA) and 15 patients with CAP. The structure and function of microbiota in BALF were identified by metagenomic next-generation sequencing (mNGS). Results The community evenness of LRT microbiota within the IIM-ILD group was marginally lower compared to the other CTD-ILD and CAP groups. Nonetheless, there were no noticeable differences. The species community structure was similar among the three groups, based on the Bray-Curtis distance between the samples. At the level of genus, the IIM-ILD group displayed a considerably higher abundance of Pseudomonas and Corynebacterium in comparison to the CAP group (p < 0.01, p < 0.05). At the species level, we found that the relative abundance of Pseudomonas aeruginosa increased significantly in the IIM-ILD group compared to the CAP group (p < 0.05). Additionally, the relative abundance of Prevotella pallens was significantly higher in other CTD-ILD groups compared to that in the IIM-ILD group (p < 0.05). Of all the clinical indicators examined in the correlation analysis, ferritin level demonstrated the strongest association with LRT flora, followed by Serum interleukin-6 level (p < 0.05). Conclusion Our research has identified particular LRT microorganisms that were found to be altered in the IIM-ILD group and were significantly associated with immune function and inflammatory markers in patients. The lower respiratory tract microbiota has potential in the diagnosis and treatment of IIM-ILD.
... Alpha-diversity has previously been reported to be decreased in airway samples from patients with COPD compared with controls, and to be further lowered with disease progression 13 14 20 and during COPD exacerbation. 23 In our study, we demonstrate that while evenness was reduced, both richness and PD were sustained in the lower airways of patients with COPD compared with healthy controls. The lower airway microbiota is proposed to be constantly modulated by the addition of bacteria through inhalation and micro-aspiration, local replication and survival, and removal through mucociliary clearance, cough, and immune responses. ...
Article
Full-text available
Background The lower airway microbiota in patients with chronic obstructive pulmonary disease (COPD) are likely altered compared with the microbiota in healthy individuals. Information on how the microbiota is affected by smoking, use of inhaled corticosteroids (ICS) and COPD severity is still scarce. Methods In the MicroCOPD Study, participant characteristics were obtained through standardised questionnaires and clinical measurements at a single centre from 2012 to 2015. Protected bronchoalveolar lavage samples from 97 patients with COPD and 97 controls were paired-end sequenced with the Illumina MiSeq System. Data were analysed in QIIME 2 and R. Results Alpha-diversity was lower in patients with COPD than controls (Pielou evenness: COPD=0.76, control=0.80, p=0.004; Shannon entropy: COPD=3.98, control=4.34, p=0.01). Beta-diversity differed with smoking only in the COPD cohort (weighted UniFrac: permutational analysis of variance R ² =0.04, p=0.03). Nine genera were differentially abundant between COPD and controls. Genera enriched in COPD belonged to the Firmicutes phylum. Pack years were linked to differential abundance of taxa in controls only (ANCOM-BC (Analysis of Compositions of Microbiomes with Bias Correction) log-fold difference/q-values: Haemophilus −0.05/0.048; Lachnoanaerobaculum −0.04/0.03). Oribacterium was absent in smoking patients with COPD compared with non-smoking patients (ANCOM-BC log-fold difference/q-values: −1.46/0.03). We found no associations between the microbiota and COPD severity or ICS. Conclusion The lower airway microbiota is equal in richness in patients with COPD to controls, but less even. Genera from the Firmicutes phylum thrive particularly in COPD airways. Smoking has different effects on diversity and taxonomic abundance in patients with COPD compared with controls. COPD severity and ICS use were not linked to the lower airway microbiota.
Article
Full-text available
Background Increasing evidence indicates the microbial ecology of chronic obstructive pulmonary disease (COPD) is intricately associated with the disease’s status and severity, and distinct microbial ecological variations exist between COPD and healthy control (HC). This systematic review and meta-analysis aimed to summarize microbial diversity indices and taxa relative abundance of oral, airway, and intestine microbiota of different stages of COPD and HC to make comparisons. Methods A comprehensive systematic literature search was conducted in PubMed, Embase, the Web of Science, and the Cochrane Library databases to identify relevant English articles on the oral, airway, and intestine microbiota in COPD published between 2003 and 8 May 2023. Information on microbial diversity indices and taxa relative abundance of oral, airway, and intestine microbiota was collected for comparison between different stages of COPD and HC. Results A total of 20 studies were included in this review, involving a total of 337 HC participants, 511 COPD patients, and 154 AECOPD patients. We observed that no significant differences in alpha diversity between the participant groups, but beta diversity was significantly different in half of the included studies. Compared to HC, Prevotella, Streptococcus, Actinomyces, and Veillonella of oral microbiota in SCOPD were reduced at the genus level. Most studies supported that Haemophilus, Lactobacillus, and Pseudomonas were increased, but Veillonella, Prevotella, Actinomyces, Porphyromonas, and Atopobium were decreased at the genus level in the airway microbiota of SCOPD. However, the abundance of Haemophilus, Lactobacillus and Pseudomonas genera exhibited an increase, whereas Actinomyces and Porphyromonas showed a decrease in the airway microbiota of AECOPD compared to HC. And Lachnospira of intestine microbiota in SCOPD was reduced at the genus level. Conclusion The majority of published research findings supported that COPD exhibited decreased alpha diversity compared to HC. However, our meta-analysis does not confirm it. In order to further investigate the characteristics and mechanisms of microbiome in the oral-airway- intestine axis of COPD patients, larger-scale and more rigorous studies are needed. Systematic review registration PROSPERO (https://www.crd.york.ac.uk/prospero/), identifier CRD42023418726.
Article
Full-text available
Sarcoidosis is a chronic granulomatous disorder characterized by unknown etiology, undetermined mechanisms, and non-specific therapies except TNF blockade. To improve our understanding of the pathogenicity and to predict the outcomes of the disease, the identification of new biomarkers and molecular endotypes is sorely needed. In this study, we systematically evaluate the biomarkers identified through Omics and non-Omics approaches in sarcoidosis. Most of the currently documented biomarkers for sarcoidosis are mainly identified through conventional “one-for-all” non-Omics targeted studies. Although the application of machine learning algorithms to identify biomarkers and endotypes from unbiased comprehensive Omics studies is still in its infancy, a series of biomarkers, overwhelmingly for diagnosis to differentiate sarcoidosis from healthy controls have been reported. In view of the fact that current biomarker profiles in sarcoidosis are scarce, fragmented and mostly not validated, there is an urgent need to identify novel sarcoidosis biomarkers and molecular endotypes using more advanced Omics approaches to facilitate disease diagnosis and prognosis, resolve disease heterogeneity, and facilitate personalized medicine.
Article
Full-text available
The lungs were long thought to be sterile until technical advances uncovered the presence of the lung microbial community. The microbiome of healthy lungs is mainly derived from the upper respiratory tract (URT) microbiome but also has its own characteristic flora. The selection mechanisms in the lung, including clearance by coughing, pulmonary macrophages, the oscillation of respiratory cilia, and bacterial inhibition by alveolar surfactant, keep the microbiome transient and mobile, which is different from the microbiome in other organs. The pulmonary bacteriome has been intensively studied recently, but relatively little research has focused on the mycobiome and virome. This up-to-date review retrospectively summarizes the lung microbiome’s history, composition, and function. We focus on the interaction of the lung microbiome with the oropharynx and gut microbiome and emphasize the role it plays in the innate and adaptive immune responses. More importantly, we focus on multiple respiratory diseases, including asthma, chronic obstructive pulmonary disease (COPD), fibrosis, bronchiectasis, and pneumonia. The impact of the lung microbiome on coronavirus disease 2019 (COVID-19) and lung cancer has also been comprehensively studied. Furthermore, by summarizing the therapeutic potential of the lung microbiome in lung diseases and examining the shortcomings of the field, we propose an outlook of the direction of lung microbiome research.
Article
Full-text available
Pulmonary sarcoidosis is a complex inflammatory disease characterized by granulomas in the lung tissue, leading to breathing difficulties and chest pain. Its etiology remains not fully understood, with factors such as allergies, autoimmune responses and genetics playing a role. This study explores the potential of blood microbiome dysbiosis, defined as an imbalance in the microbial ecosystem, as a missing piece of the puzzle in understanding the etiology of the disease. Our objective was to apply a decision-tree supervised machine learning hierarchical model to distinguish potential patterns of microbiome dysbiosis in blood samples from patients with pulmonary sarcoidosis as compared to healthy age-matched controls. Blood microbiome analysis, being individually-specific and stable, offers a unique perspective. Utilizing 16S rRNA gene amplicon sequencing, we analyzed the blood microbiome composition characterized by non-normally distributed and sparse data. Because of the rarity of the disease in Bulgaria, we studied a relatively small patient group, n = 7. The findings were compared to 21 healthy age-matched controls. Bioinformatics and statistical analysis play a pivotal role in microbiome analysis, especially when discerning associations between taxonomic composition and disorders such as pulmonary sarcoidosis. By analyzing the microbial diversity, we identified alterations in the blood microbiome composition between healthy individuals and those with sarcoidosis, which potentially may trigger the disease. Advanced machine learning techniques provided additional power to the analysis, that might be overlooked by the usual group statistics, confirming the differentiation of the diversity within the studied microbiome.
Article
Full-text available
Microbiota have emerged as key modulators of both the carcinogenic process and the immune response against cancer cells, and, thus, it seems to influence the efficacy of immunotherapy. While most studies have focused on analyzing the influence of gut microbiota, its composition substantially differs from that in the lung. Here, we describe how microbial life in the lungs is associated with host immune status in the lungs and, thus, how the identification of the microbial populations in the lower respiratory tract rather than in the gut might be key to understanding the lung carcinogenic process and to predict the efficacy of different treatments. Understanding the influence of lung microbiota on host immunity may identify new therapeutic targets and help to design new immunotherapy approaches to treat lung cancer.
Article
Full-text available
Cytoscape is one of the most successful network biology analysis and visualization tools, but because of its interactive nature, its role in creating reproducible, scalable, and novel workflows has been limited. We describe Cytoscape Automation (CA), which marries Cytoscape to highly productive workflow systems, for example, Python/R in Jupyter/RStudio. We expose over 270 Cytoscape core functions and 34 Cytoscape apps as REST-callable functions with standardized JSON interfaces backed by Swagger documentation. Independent projects to create and publish Python/R native CA interface libraries have reached an advanced stage, and a number of automation workflows are already published.
Article
Full-text available
Background: Chronic obstructive pulmonary disease (COPD) frequent exacerbators (FE) suffer increased morbidity and mortality compared to infrequent exacerbators (IE). The association between the oral and sputum microbiota and exacerbation phenotype is not well defined. The objective of this study was to determine key features that differentiate the oral and sputum microbiota of FEs from the microbiota of IEs during periods of clinical stability. Methods: We recruited 11 FE and 11 IE who had not used antibiotics or systemic corticosteroids in the last 1 month. Subjects provided oral wash and sputum samples, which underwent 16S V4 MiSeq sequencing and qPCR of 16S rRNA. Data were analyzed using Dada2 and R. Results: FE and IE were similar in terms of age, FEV1 percent predicted (FEV1pp), pack-years of tobacco exposure, and St. George's Respiratory Questionnaire score. 16S copy numbers were significantly greater in sputum vs. oral wash (p = 0.01), but phenotype was not associated with copy number. Shannon diversity was significantly greater in oral samples compared to sputum (p = 0.001), and IE samples were more diverse than FE samples (p < 0.001). Sputum samples from FE had more Haemophilus and Moraxella compared to IE sputum samples, due to dominance of these COPD-associated taxa in three FE sputum samples. Amplicon sequencing variant (ASV)-level analysis of sputum samples revealed one ASV (Actinomyces) was significantly more abundant in IE vs. FE sputum (padj = 0.048, Wilcoxon rank-sum test), and this persisted after controlling for FEV1pp. Principal coordinate analysis using Bray-Curtis distance with PERMANOVA analyses demonstrated clustering by anatomic site, phenotype, inhaled corticosteroid use, current tobacco use, COPD severity, and last professional dental cleaning. Conclusions: FE have less diverse oral and sputum microbiota than IE. Actinomyces was significantly more abundant in IE sputum than FE sputum. The oral and sputum microbiota of COPD subjects cluster based on multiple clinical factors, including exacerbation phenotype. Even during periods of clinical stability, the frequent exacerbator phenotype is associated with decreased alpha diversity, beta-diversity clustering, and changes in taxonomic abundance.
Article
Full-text available
Background Sarcoidosis is a systemic disease of unknown etiology. The disease mechanisms are largely speculative and may include the role microbial patterns that initiate and drive an underlying immune process. The aim of this study was to characterize the microbiota of the lung of patients with sarcoidosis and compare its composition and diversity with the results from patients with other interstitial lung disease (ILD) and historic healthy controls. Methods Patients (sarcoidosis, n = 31; interstitial lung disease, n = 19) were recruited within the PULMOHOM study, a prospective cohort study to characterize inflammatory processes in pulmonary diseases. Bronchoscopy of the middle lobe or the lingula was performed and the recovered fluid was immediately sent for analysis of the pulmonary microbiota by 16sRNA gene sequencing. Subsequent bioinformatic analysis was performed to compare the groups. Results There were no significant differences between patients with sarcoidosis or other ILDs with regard to microbiome composition and diversity. In addition, the abundance of the genera Atopobium, Fusobacterium, Mycobacterium or Propionibacterium were not different between the two groups. There were no gross differences to historical healthy controls. Conclusion The analysis of the pulmonary microbiota based on 16sRNA gene sequencing did not show a significant dysbiosis in patients with sarcoidosis as compared to other ILD patients. These data do not exclude a microbiological component in the pathogenesis of sarcoidosis. Electronic supplementary material The online version of this article (10.1186/s12931-019-1013-2) contains supplementary material, which is available to authorized users.
Article
Full-text available
H. haemolyticus is often misidentified as NTHi due to their close phylogenetic relationship. Differentiating between the two is important for correct identification and appropriate treatment of infective organism and to ensure any role of H. haemolyticus in disease is not being overlooked. Speciation however is not completely reliable by culture and PCR methods due to the loss of haemolysis by H. haemolyticus and the heterogeneity of NTHi. Haemophilus isolates from COPD as part of the AERIS study (ClinicalTrials - NCT01360398) were speciated by analysing sequence data for the presence of molecular markers. Further investigation into the genomic relationship was carried out using average nucleotide identity and phylogeny of allelic and genome alignments. Only 6.3% were identified as H. haemolyticus. Multiple in silico methods were able to distinguish H. haemolyticus from NTHi. However, no single gene target was found to be 100% accurate. A group of omp2 negative NTHi were observed to be phylogenetically divergent from H. haemolyticus and remaining NTHi. The presence of an atypical group from a geographically and disease limited set of isolates supports the theory that the heterogeneity of NTHi may provide a genetic continuum between NTHi and H. haemolyticus.
Article
Full-text available
Background: Interstitial lung diseases (ILDs) are associated with a high burden of disease. However, data on the prognostic impact of comorbidities and comorbidity-related pharmaceutical treatments in patients with various ILDs remain sparse. Methods: Using longitudinal claims data from a German Statutory Health Insurance Fund, we assessed comorbidity in ILD subtypes and associated drug treatments. Baseline comorbidity was assessed via the Elixhauser Comorbidity Index that was amended by ILD-relevant conditions. Drug treatment was assessed on the substance level using the ATC-codes of drugs prescribed at the time of ILD diagnosis. Subsequently, the comorbid conditions (main analysis) and pharmaceutical substances (secondary analysis) with a meaningful association to survival were identified for the complete ILD cohort and within the subtype strata. For this, we applied multivariate Cox models using a LASSO selection process and visualized the findings within comorbidomes. Results: In the 36,821 patients with ILDs, chronic obstructive pulmonary disease (COPD), arterial hypertension, and ischaemic heart disease (IHD) were the most prevalent comorbidities. The majority of patients with cardiovascular diseases received pharmaceutical treatment, while, in other relevant comorbidities, treatment quotas were low (COPD 46%, gastro-oesophageal reflux disease 65%). Comorbidities had a clinically meaningful detrimental effect on survival that tended to be more pronounced in the case of untreated conditions (e.g. hazard ratios for treated IHD 0.97 vs. 1.33 for untreated IHD). Moreover, comorbidity impact varied substantially between distinct subtypes. Conclusions: Our analyses suggest that comorbid conditions and their treatment profile significantly affect mortality in various ILDs. Therefore, comprehensive comorbidity assessment and management remains important in any ILD.
Article
Full-text available
Heterogeneity amidst healthy individuals at genomic level is being widely acknowledged. This, in turn, is modulated by differential response to environmental cues and treatment regimens, necessitating the need for stratified/personalized therapy. We intend to understand the molecular determinants of Ayurvedic way (ancient Indian system of medicine) of endo-phenotyping individuals into distinct constitution types termed “Prakriti,” which forms the basis of personalized treatment. In this study, we explored and analyzed the healthy human gut microbiome structure within three predominant Prakriti groups from a genetically homogenous cohort to discover differentially abundant taxa, using 16S rRNA gene based microbial community profiling. We found Bacteroidetes and Firmicutes as major gut microbial components in varying composition, albeit with similar trend across Prakriti. Multiple species of the core microbiome showed differential abundance within Prakriti types, with gender specific signature taxons. Our study reveals that despite overall uniform composition of gut microbial community, healthy individuals belonging to different Prakriti groups have enrichment of specific bacteria. It highlights the importance of Prakriti based endo-phenotypes to explain the variability amongst healthy individuals in gut microbial flora that have important consequences for an individual's health, disease and treatment.
Article
Full-text available
Every niche in the biosphere is touched by the seemingly endless capacity of microbes to transform the world around them by adapting swiftly and flexibly to the environmental changes, likewise the gastrointestinal tract is no exception. The ability to cope with rapid changes in external osmolarity is an important aspect of gut microbes for their survival and colonization. Identification of these survival mechanisms is a pivotal step towards understanding genomic suitability of a symbiont for successful human gut colonization. Here we highlight our recent work applying functional metagenomics to study human gut microbiome to identify candidate genes responsible for the salt stress tolerance. A plasmid borne metagenomic library of Bacteroidetes enriched human fecal metagenomic DNA led to identification of unique salt osmotolerance clones SR6 and SR7. Subsequent gene analysis combined with functional studies revealed that TLSRP1 within pSR7 and TMSRP1 and ABCTPP of pSR6 are the active loci responsible for osmotolerance through an energy dependent mechanism. Our study elucidates the novel genetic machinery involved in bestowing osmotolerance in Prevotella and Bacteroidetes, the predominant microbial groups in a North Indian population. This study unravels an alternative method for imparting ionic stress tolerance, which may be prevalent in the human gut microbiome.
Article
Pulmonary immunological tolerance to inhaled particulates might create a permissive milieu for lung metastasis. Lung microbiota contribute to pulmonary tolerance; here, we explored whether its manipulation via antibiotic or probiotic aerosolization favors immune response against melanoma metastasis. In lungs of vancomycin/neomycin-aerosolized mice, a decrease in bacterial load was associated with reduced regulatory T cells and enhanced T cell and NK cell activation that paralleled a significant reduction of melanoma B16 lung metastases. Reduction of metastases also occurred in lungs transplanted with bacterial isolates from antibiotic-treated lungs. Aerosolized Lactobacillus rhamnosus strongly promoted immunity against B16 lung metastases as well. Furthermore, probiotics or antibiotics improved chemotherapy activity against advanced B16 metastases. Thus, we identify a role for lung microbiota in metastasis and show that its targeting via aerosolization is a therapy that can prevent metastases and enhance responses to chemotherapy.