Nihms 1610206

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

HHS Public Access

Author manuscript
Curr Opin Microbiol. Author manuscript; available in PMC 2021 August 01.
Author Manuscript

Published in final edited form as:


Curr Opin Microbiol. 2020 August ; 56: 24–29. doi:10.1016/j.mib.2020.06.001.

Perspectives in lung microbiome research


Imran Sulaiman1, Sheeja Schuster1, Leopoldo N. Segal1
1Division of Pulmonary, Critical Care, & Sleep Medicine, Department of Medicine, New York

University School of Medicine, NY

Abstract
Our understanding of the existence and role of the lung microbiome has grown at a slower pace
Author Manuscript

than other microbiome research areas. This is likely a consequence of the original dogma that the
lung was a sterile environment although there are other barriers that are worth discussing. Here we
will not be conducting an exhaustive review of the current literature on the lung microbiome but
rather we will focus on what we see as some important challenges that the field needs to face in
order to improve our mechanistic understanding of the lung microbiome and its role on human
health.

Keywords
microbiome

Lung microbiome research is at its infancy. While the gut microbiota is resilient and varies
Author Manuscript

broadly between different individuals [1], we are just now starting to appreciate that the
lungs are not sterile and in fact harbor a complex microbial landscape. Additionally, we now
know that the gut microbiome modulates the systemic immune tone contributing to the
differentiation of IL17 producing T cells and regulatory T cells [2], whereas the lung
microbiome field has so far only show associations between lung microbiota signatures and
host immune phenotype [3-5]. It is understood that gut dysbiosis affects the metabolic local
and systemic microenvironment [6,7] and contributes to the susceptibility to pathogens such
as Clostridium difficile [8], however we do not know whether lung microbiota dysbiosis
contributes to the acquisition of respiratory pathogens. Finally, while we are now uncovering
how the gut microbiota modulates the effectiveness of drugs such as immunomodulators for
cancer [9,10], we currently have not studied the effects of the lung microbiota on drugs such
Author Manuscript

Corresponding Author/ Address for Reprints: Leopoldo N. Segal, MD1 Leopoldo.Segal@nyumc.org, NYU School of Medicine 462
First Ave 7W54 New York, NY 10016, Tel: (212) 562-3752, Fax: (212) 263-7445.
Financial Disclosure: None
Declaration of interests
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to
influence the work reported in this paper.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our
customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of
the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered
which could affect the content, and all legal disclaimers that apply to the journal pertain.
Sulaiman et al. Page 2

as inhaled therapy for COPD, asthma or even on immunomodulators that have become the
Author Manuscript

front line therapy for non-small cell lung cancer.

In order to improve our mechanistic understanding of the lung microbiome we first need to
understand the reasons for its delay in development. Here we identify some of the key
obstacles in lung microbiome research that are important to consider.

Difficulty in sample collection


Analysis of the gut microbiome has concentrated on the use of stool samples [2,6-8,10,11], a
somewhat easily acquired specimen that is a direct product of transit through the entire
gastrointestinal tract. Although some have noted regional differences in the lower
gastrointestinal microbiota [12], the use of stool samples as a surrogate of the microbiome of
the gastrointestinal tract is widely accepted. In lung microbiome research, a large number of
Author Manuscript

investigations have concentrated on the use of sputum samples [9,13,14] given the
accessibility of these samples, whether this be spontaneous (easier among subjects with
respiratory conditions with high mucus production such as cystic fibrosis) or induced.
However, the heterogeneity of the microbial topography throughout the respiratory tract
complicates this approach. For example, the upper airway microbiota (including the dental
microbiome, oral microbiome and supraglottic microbiota) is invariantly colonized with
anaerobes that coexist at high concentration (similar to the ones measured in the lower
gastrointestinal tract) [15-17]. In contrast, the lower airway microbiota has significantly
reduced bacterial concentration and with clear differences in diversity when compared to the
upper airway microbiota (Figure 1A) [18-21]. Since expectorated sputum is the result of the
sum of secretions throughout the respiratory tract, multiple topographical sources can
contribute to microbes found in the microbial assessment of these samples. Thus, sputum
Author Manuscript

can be: 1) a perfect representation of the upper airways (including oral and dental); 2) a
mixture of upper and lower airway microbes; or 3) a perfect representation of the lower
airways. Given the significantly higher bacterial burden in the upper airways than in the
lower airways most evidence supports that the first two scenarios are the most likely. This is
further confirmed by two investigations that collected paired upper and lower airway
samples (one in a cohort of subjects with asthma and a second on patients with
bronchiectasis) and demonstrated that the microbial similarity between sputum and upper
airway samples was greater than the similarity between sputum and lower airway samples
[22,23]. If the microbes present in sputum are a variable mixture of microbes from the lower
and upper airways, it is unclear how to determine the relative contribution of those two
different topographical sources to the final collection of microbes observed in sputum.
Further, recent investigations have suggested that the method for sputum collection
Author Manuscript

(spontaneous vs. induced) will affect the results [24]. Some have proposed the exhaled
breath condensate (EBC) as a less invasive alternative for lung microbiome sampling
[25-28]. This involves condensing exhaled vapor into a liquid which is then sent for
sequencing, a much less invasive procedure when compared to bronchoscopy. However, in
an animal model the microbial composition of EBC samples was not representative of the
microbiota in the lower airways [29]. A comparison of bronchoscopy samples to EBC
samples in humans has not yet been performed. Thus, we currently do not have any accurate
surrogates for lung microbiome evaluation. This does not necessarily mean that sputum

Curr Opin Microbiol. Author manuscript; available in PMC 2021 August 01.
Sulaiman et al. Page 3

samples are without use in the evaluation of the lung microbiome, but it is our opinion that
Author Manuscript

investigators should be mindful about these limitations when planning their research. While
sputum has been used in culture-based approaches for decades. The focus of those studies
was primarily the detection of microbes with potential pathogenic role rather than a
comprehensive description of the microbial communities. Additionally, with current sputum
culture approaches we have accepted that the microbial yield from the lower airways is
variable as exemplified by the clinical need to have repeated sputum cultures in
Tuberculosis.

Low biomass samples


Most microbes in the human body are contained in the gastrointestinal tract. Many other
mucosae in topographical contact with the exterior world are also exposed to microbes but
the microbial biomass contained in those are significantly lower [30,31]. This imposes
Author Manuscript

several challenges for sample processing and data analysis. One such challenge is the
presence of bacterial DNA contamination from exogenous sources acquired through sample
collection and sample processing (Figure 1A). In a lower airway sample, these
“contaminant” bacterial DNA may be in higher concentration than the bacterial DNA
originating form “true” lung bacteria. Thus, it is expected that the amplification process and
subsequent sequencing will be biased towards these contaminants resulting in a large
proportion of the sequenced microbiota being dominated by them [32-34]. Importantly, the
nature of these contaminant bacterial DNA will depend on the method used. In addition,
further noise, in the sequencing output, could be expected from PCR error, index switching,
and chimera formation [35,36]. The downstream effects on the data will then be variable,
affecting diversity metrics and the relative abundance of the true bacteria present in the
lower airways. Different methods have been developed to identify a bacterial signal as
Author Manuscript

contaminant which are dependent on the quality and quantity of data on background samples
(e.g., DNA isolation negative controls, bronchoscopy or instrument controls). These
bioinformatic methods include abundance filtering, where a threshold is set to remove low
abundance bacteria (probably not useful in low biomass samples); removing all sequences
present at high prevalence in negative control samples (again a threshold can be set);
removing sequences based on an inverse correlation with bacterial DNA concentration; and
Bayesian methods. With these different methods, one could then remove sequences
identified as contaminants from the rest of the data set. However, given the compositional
nature (and thus not quantitative) of the sequencing data the effects on what’s left after
removal can be quite variable and difficult to predict or reproduce. In fact, a recent
consensus of lung microbiome experts have not recommend routine bioinformatic removal
of contaminants [37].
Author Manuscript

What do we know and why is it important to foster lung microbiome


research:
Even though we know now, with culture independent techniques, that the lungs are not
sterile, there are significant microbial ecological differences with other mucosae where the
presence of microbes has never been questioned. For example, in health, while the gut

Curr Opin Microbiol. Author manuscript; available in PMC 2021 August 01.
Sulaiman et al. Page 4

microbiota harbors a large microbial biomass that occupies the mucosal lumen, shows high
Author Manuscript

dynamics but also high resilience [38], the microbiota of the healthy lung behaves like a near
desert island. This is not surprising, since the main function of the lung is to allow for gas
exchange between our blood and the ambient air. Thus, the mucosal lumen of the airways
should be occupied by mostly air. It is now well accepted that the presence of microbes in
the lower airways of healthy lungs behave similar to an island with a tight balance of
microbial immigration (mostly from microaspiration of oral secretions) and elimination [39].
Further, the lung is not a single continuous longitudinal topographical system but rather a
branching series of parallel topographical systems that allows large amount of regional
microbial heterogeneity. Additionally, several conditions for bacterial growth vary within the
lung, such as blood flow, local pH, temperature and oxygen tension [40]. This, together with
individual patient factors such as inadequate airway clearance/subclinical aspiration or the
presence of chronic lung disease (such as COPD or bronchiectasis), can explain significant
Author Manuscript

heterogeneity within each subject and between subjects with similar diagnosis.

Despite these limitations, it is now clear that the lung microbiome affects the host immune
tone in the lower airways [41-43]. For example, several studies have shown that aspiration is
a common phenomenon [44,45], and the presence of oral commensals in the lower airways
(even in healthy lungs) is associated with a distinct metabolic profile and with an increased
immune tone characterized by IL17 producing T cells [46]. Multiple lines of investigations
support the effect of the lower airway microbiota on the host in multiple disease states such
as asthma, COPD, bronchiectasis, interstitial lung disease, lung cancer, etc [23,47-50].
However, most of these studies are based on cross-sectional human studies where
directionality of the associations is difficult to establish. To aid in understanding causality
better, there is a need for more longitudinal studies. Further, while we can support that the
lung microbiome affects the host immune tone, the molecular mechanisms are not clearly
Author Manuscript

established. Research into the metabolic profiling of the lower airway environment has
shown that the lower airways in fact contain multiple metabolites that could be end products
of microbial metabolism or else affected by microbial metabolic activity [51,52]. One
example of this is the identification of short chain fatty acids (SCFAs) in the lower airways,
which are end product of microbial fermentation done by anaerobic bacteria and have
significant immunomodulatory effects [53].

Next steps in lung microbiome research:


The lung microbiome field has been dominated by descriptive cross-sectional studies
focused on the “1,000 feet view” of the microbiome. It is becoming increasingly clear that it
is important to not only identify the presence of organisms in different environments but to
Author Manuscript

also understand the function of these organisms and how they may impact the host. How can
we accomplish these goals? Shotgun sequencing methods have been developed over the last
two decades [54-56]. The use of this untargeted sequencing approach of all microbial genes
provides information on both taxonomic composition (with increased resolution when
compared to 16S rRNA gene sequencing) as well as functional potential of different
microbes. While this technology is useful in better understanding how microbes function
there has been very few publications using this technology in analyzing the airway
microbiome, and in most cases focused on its use for taxonomy [57-61], primarily due to

Curr Opin Microbiol. Author manuscript; available in PMC 2021 August 01.
Sulaiman et al. Page 5

issues mentioned previous (low biomass and contamination). It is also important to note that
Author Manuscript

by studying genomic DNA we are not able to identify active microbial function nor whether
these sequences are from viable or non-viable microbes [62]. Another possible approach to
study microbial communities is through the use of metatranscriptomic RNA sequencing
approaches, which allow for characterization of microbial gene expression [62]. Using such
methods, we are able to ascertain which microbial genes are actively being transcribed,
therefore proving information on ongoing microbial function, and microbial viability (given
the short live nature of RNA). Even fewer investigations exists using metatranscriptomic
approaches in the lung and it is likely that the dynamics of these microbial signatures will
follow a different dynamic than DNA signatures (Figure 1B) [63,64]. Ultimately, microbial
functional molecules will need to be studied measuring proteins (metaproteomics) and
metabolites (metabolomics) both of which have rarely been used in lung microbiome
research [46,53].
Author Manuscript

While these novel methods will likely have significant impact on the lung microbiome field,
classical mechanistic investigations will be required to help progress the understanding of
the lower airway microbiome. These include the use of preclinical models [50,65,66],
longitudinal studies [67,68], and, ultimately, clinical trials modulating the microbial
environment.

Prior studies have provided a full state of the art review of lung microbiome [37,69]. In this
publication we provide a critical view of where the research field is, the major limitations
present and the strong belief that the search for mechanistic insights will depend on moving
on from descriptive, hypothesis free, cross sectional studies and focusing more on
hypothesis-driven, mechanistic studies.
Author Manuscript

Acknowledgments
Research support funding: R37 CA244775 (LNS, NIH/NCI); R01 HL125816 (SBK, LNS, NIH/NHLBI); PACT
grant (LNS, FNIH); Chest Foundation Grant (IS).

Financial support for the PACT project are 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.

References:
1. Healey GR, Murphy R, Brough L, Butts CA, Coad J: Interindividual variability in gut microbiota
and host response to dietary interventions. Nutr Rev 2017, 75:1059–1080. [PubMed: 29190368]
2. Douzandeh-Mobarrez B, Kariminik A: Gut Microbiota and IL-17A: Physiological and Pathological
Responses. Probiotics Antimicrob Proteins 2019, 11:1–10. [PubMed: 28921400]
Author Manuscript

3. Shukla SD, Budden KF, Neal R, Hansbro PM: Microbiome effects on immunity, health and disease
in the lung. Clin Transl Immunology 2017, 6:e133. [PubMed: 28435675]
4. Wypych TP, Wickramasinghe LC, Marsland BJ: The influence of the microbiome on respiratory
health. Nat Immunol 2019, 20:1279–1290. [PubMed: 31501577]
5. O'Dwyer DN, Dickson RP, Moore BB: The Lung Microbiome, Immunity, and the Pathogenesis of
Chronic Lung Disease. J Immunol 2016, 196:4839–4847. [PubMed: 27260767]

Curr Opin Microbiol. Author manuscript; available in PMC 2021 August 01.
Sulaiman et al. Page 6

6. Belizario JE, Faintuch J, Garay-Malpartida M: Gut Microbiome Dysbiosis and Immunometabolism:


New Frontiers for Treatment of Metabolic Diseases. Mediators Inflamm 2018, 2018:2037838.
Author Manuscript

[PubMed: 30622429]
7. Carding S, Verbeke K, Vipond DT, Corfe BM, Owen LJ: Dysbiosis of the gut microbiota in disease.
Microb Ecol Health Dis 2015, 26:26191. [PubMed: 25651997]
8. Antharam VC, Li EC, Ishmael A, Sharma A, Mai V, Rand KH, Wang GP: Intestinal dysbiosis and
depletion of butyrogenic bacteria in Clostridium difficile infection and nosocomial diarrhea. J Clin
Microbiol 2013, 51:2884–2892. [PubMed: 23804381]
9. Vivarelli S, Salemi R, Candido S, Falzone L, Santagati M, Stefani S, Torino F, Banna GL, Tonini G,
Libra M: Gut Microbiota and Cancer: From Pathogenesis to Therapy. Cancers (Basel) 2019, 11.
10. Ma W, Mao Q, Xia W, Dong G, Yu C, Jiang F: Gut Microbiota Shapes the Efficiency of Cancer
Therapy. Front Microbiol 2019, 10:1050. [PubMed: 31293523]
11. Sun MF, Shen YQ: Dysbiosis of gut microbiota and microbial metabolites in Parkinson's Disease.
Ageing Res Rev 2018, 45:53–61. [PubMed: 29705121]
12. Martinez-Guryn K, Leone V, Chang EB: Regional Diversity of the Gastrointestinal Microbiome.
Cell Host Microbe 2019, 26:314–324. [PubMed: 31513770]
Author Manuscript

13. Liu HY, Zhang SY, Yang WY, Su XF, He Y, Zhou HW, Su J: Oropharyngeal and Sputum
Microbiomes Are Similar Following Exacerbation of Chronic Obstructive Pulmonary Disease.
Front Microbiol 2017, 8:1163. [PubMed: 28690603]
14. Lopez Caro JC, Santibanez M, Garcia Rivero JL, Villanueva M, Sainz J, Gonzalez Astorqui P,
Hierro M, Rodriguez Porres M, Paras Bravo P, Mira A, et al.: Sputum Microbiome Dynamics in
Chronic Obstructive Pulmonary Disease Patients during an Exacerbation Event and Post-
Stabilization. Respiration 2019, 98:447–454. [PubMed: 31437842]
15. Ramakrishnan VR, Frank DN: Microbiome in patients with upper airway disease: Moving from
taxonomic findings to mechanisms and causality. J Allergy Clin Immunol 2018, 142:73–75.
[PubMed: 29803801]
16. Lee JT, Kim CM, Ramakrishnan V: Microbiome and disease in the upper airway. Curr Opin
Allergy Clin Immunol 2019, 19:1–6. [PubMed: 30543547]
17. Kumpitsch C, Koskinen K, Schopf V, Moissl-Eichinger C: The microbiome of the upper
respiratory tract in health and disease. BMC Biol 2019, 17:87. [PubMed: 31699101]
Author Manuscript

18. Li KJ, Chen ZL, Huang Y, Zhang R, Luan XQ, Lei TT, Chen L: Dysbiosis of lower respiratory
tract microbiome are associated with inflammation and microbial function variety. Respir Res
2019, 20:272. [PubMed: 31796027]
19. Mur LA, Huws SA, Cameron SJ, Lewis PD, Lewis KE: Lung cancer: a new frontier for
microbiome research and clinical translation. Ecancermedicalscience 2018, 12:866. [PubMed:
30263057]
20. Caverly LJ, LiPuma JJ: Good cop, bad cop: anaerobes in cystic fibrosis airways. Eur Respir J 2018,
52.
21. Layeghifard M, Li H, Wang PW, Donaldson SL, Coburn B, Clark ST, Caballero JD, Zhang Y,
Tullis DE, Yau YCW, et al.: Microbiome networks and change-point analysis reveal key
community changes associated with cystic fibrosis pulmonary exacerbations. NPJ Biofilms
Microbiomes 2019, 5:4. [PubMed: 30675371]
22. Durack J, Huang YJ, Nariya S, Christian LS, Mark Ansel K, Beigelman A, Castro M, Dyer AM,
Israel E, Kraft M, et al.: Bacterial biogeography of adult airways in atopic asthma. Microbiome
2018, 6:104. [PubMed: 29885665] *This study compared the microbiome in sputum with the one
Author Manuscript

present in oral, nasal and bronchial airway samples among a cohort of patients with asthma. In this
study, the authors demonstrated that the sputum microbiome resembles more the oral microbiota
than the lower airway microbiota.
23. Sulaiman I, Wu BG, Li Y, Scott AS, Malecha P, Scaglione B, Wang J, Basavaraj A, Chung S,
Bantis K, et al.: Evaluation of the airway microbiome in nontuberculous mycobacteria disease. Eur
Respir J 2018, 52.*This study evaluated the airway microbiota in a cohort of patients with
bronchiectasis. In a subgroup of subjects undergoing bronchoscopy they demonstrated that the
microbiota in sputum is more similar to the upper airway microbiota than it is to the lower airway
microbiota.

Curr Opin Microbiol. Author manuscript; available in PMC 2021 August 01.
Sulaiman et al. Page 7

24. Tangedal S, Aanerud M, Gronseth R, Drengenes C, Wiker HG, Bakke PS, Eagan TM: Comparing
microbiota profiles in induced and spontaneous sputum samples in COPD patients. Respir Res
Author Manuscript

2017, 18:164. [PubMed: 28851370]


25. Jackson AS, Sandrini A, Campbell C, Chow S, Thomas PS, Yates DH: Comparison of biomarkers
in exhaled breath condensate and bronchoalveolar lavage. Am J Respir Crit Care Med 2007,
175:222–227. [PubMed: 17110649]
26. Zakharkina T, Koczulla AR, Mardanova O, Hattesohl A, Bals R: Detection of microorganisms in
exhaled breath condensate during acute exacerbations of COPD. Respirology 2011, 16:932–938.
[PubMed: 21470340]
27. Carpagnano GE, Malerba M, Lacedonia D, Susca A, Logrieco A, Carone M, Cotugno G, Palmiotti
GA, Foschino-Barbaro MP: Analysis of the fungal microbiome in exhaled breath condensate of
patients with asthma. Allergy Asthma Proc 2016, 37:41–46. [PubMed: 27178886]
28. Xu Z, Shen F, Li X, Wu Y, Chen Q, Jie X, Yao M: Molecular and microscopic analysis of bacteria
and viruses in exhaled breath collected using a simple impaction and condensing method. PLoS
One 2012, 7:e41137. [PubMed: 22848436]
29. Glendinning L, Wright S, Tennant P, Gill AC, Collie D, McLachlan G: Microbiota in Exhaled
Author Manuscript

Breath Condensate and the Lung. Appl Environ Microbiol 2017, 83.
30. Karstens L, Asquith M, Caruso V, Rosenbaum JT, Fair DA, Braun J, Gregory WT, Nardos R,
McWeeney SK: Community profiling of the urinary microbiota: considerations for low-biomass
samples. Nat Rev Urol 2018, 15:735–749. [PubMed: 30315209]
31. Kong HH, Andersson B, Clavel T, Common JE, Jackson SA, Olson ND, Segre JA, Traidl-
Hoffmann C: Performing Skin Microbiome Research: A Method to the Madness. J Invest
Dermatol 2017, 137:561–568. [PubMed: 28063650]
32. Jervis-Bardy J, Leong LE, Marri S, Smith RJ, Choo JM, Smith-Vaughan HC, Nosworthy E, Morris
PS, O'Leary S, Rogers GB, et al.: Deriving accurate microbiota profiles from human samples with
low bacterial content through post-sequencing processing of Illumina MiSeq data. Microbiome
2015, 3:19. [PubMed: 25969736]
33. Lusk RW: Diverse and widespread contamination evident in the unmapped depths of high
throughput sequencing data. PLoS One 2014, 9:e110808. [PubMed: 25354084]
34. Glassing A, Dowd SE, Galandiuk S, Davis B, Chiodini RJ: Inherent bacterial DNA contamination
of extraction and sequencing reagents may affect interpretation of microbiota in low bacterial
Author Manuscript

biomass samples. Gut Pathog 2016, 8:24. [PubMed: 27239228]


35. Sze MA, Schloss PD: The Impact of DNA Polymerase and Number of Rounds of Amplification in
PCR on 16S rRNA Gene Sequence Data. mSphere 2019, 4.*This study identifies how technical
aspects of sequencing impacts the results obtained during microbiome analysis, specially
important when dealing with low biomass samples.
36. MacConaill LE, Burns RT, Nag A, Coleman HA, Slevin MK, Giorda K, Light M, Lai K, Jarosz M,
McNeill MS, et al.: Unique, dual-indexed sequencing adapters with UMIs effectively eliminate
index cross-talk and significantly improve sensitivity of massively parallel sequencing. BMC
Genomics 2018, 19:30. [PubMed: 29310587]
37. Carney SM, Clemente JC, Cox MJ, Dickson RP, Huang YJ, Kitsios GD, Kloepfer KM, Leung JM,
LeVan TD, Molyneaux PL, et al.: Methods in Lung Microbiome Research. Am J Respir Cell Mol
Biol 2019.*This is a consensus from many lung microbiome research groups that provides
guidelines about technical and research design.
38. Lozupone CA, Stombaugh JI, Gordon JI, Jansson JK, Knight R: Diversity, stability and resilience
Author Manuscript

of the human gut microbiota. Nature 2012, 489:220–230. [PubMed: 22972295]


39. Dickson RP, Erb-Downward JR, Freeman CM, McCloskey L, Beck JM, Huffnagle GB, Curtis JL:
Spatial Variation in the Healthy Human Lung Microbiome and the Adapted Island Model of Lung
Biogeography. Ann Am Thorac Soc 2015, 12:821–830. [PubMed: 25803243]
40. Man WH, de Steenhuijsen Piters WA, Bogaert D: The microbiota of the respiratory tract:
gatekeeper to respiratory health. Nat Rev Microbiol 2017, 15:259–270. [PubMed: 28316330]
41. Dickson RP, Erb-Downward JR, Falkowski NR, Hunter EM, Ashley SL, Huffnagle GB: The Lung
Microbiota of Healthy Mice are Highly Variable, Cluster by Environment, and Reflect Variation in
Baseline Lung Innate Immunity. Am J Respir Crit Care Med 2018.**In this investigation, studying

Curr Opin Microbiol. Author manuscript; available in PMC 2021 August 01.
Sulaiman et al. Page 8

the lung microbiome in mice from different cages and vendor the authors were able to identify
how the lung immune tone is tightly associated with the microbial signatures present in the lower
Author Manuscript

airways.
42. Segal LN, Alekseyenko AV, Clemente JC, Kulkarni R, Wu B, Chen H, Berger KI, Goldring RM,
Rom WN, Blaser MJ, et al.: Enrichment of lung microbiome with supraglottic taxa is associated
with increased pulmonary inflammation. Microbiome 2013, 1:19. [PubMed: 24450871]
43. Pattaroni C, Watzenboeck ML, Schneidegger S, Kieser S, Wong NC, Bernasconi E, Pernot J,
Mercier L, Knapp S, Nicod LP, et al.: Early-Life Formation of the Microbial and Immunological
Environment of the Human Airways. Cell Host Microbe 2018, 24:857–865 e854. [PubMed:
30503510] **One of the few investigations with lower airway samples in early life in humans. The
investigators demonstrate how changes in the lung microbiome are associated with lower airway
immune maturation.
44. Gleeson K, Eggli DF, Maxwell SL: Quantitative aspiration during sleep in normal subjects. Chest
1997, 111:1266–1272. [PubMed: 9149581]
45. Cvejic L, Harding R, Churchward T, Turton A, Finlay P, Massey D, Bardin PG, Guy P: Laryngeal
penetration and aspiration in individuals with stable COPD. Respirology 2011, 16:269–275.
Author Manuscript

[PubMed: 21054669]
46. Segal LN, Clemente JC, Tsay JC, Koralov SB, Keller BC, Wu BG, Li Y, Shen N, Ghedin E, Morris
A, et al.: Enrichment of the lung microbiome with oral taxa is associated with lung inflammation
of a Th17 phenotype. Nat Microbiol 2016, 1:16031. [PubMed: 27572644] **This investigation in
humans revealed how enrichemnt of the lower airway microbiota with oral commensals, a
signature commonly found in multiple investigation, is associated with the lower airway immune
tone.
47. Huang YJ, Nariya S, Harris JM, Lynch SV, Choy DF, Arron JR, Boushey H: The airway
microbiome in patients with severe asthma: Associations with disease features and severity. J
Allergy Clin Immunol 2015, 136:874–884. [PubMed: 26220531]
48. Sze MA, Dimitriu PA, Hayashi S, Elliott WM, McDonough JE, Gosselink JV, Cooper J, Sin DD,
Mohn WW, Hogg JC: The lung tissue microbiome in chronic obstructive pulmonary disease. Am J
Respir Crit Care Med 2012, 185:1073–1080. [PubMed: 22427533]
49. Tsay JJ, Wu BG, Badri MH, Clemente JC, Shen N, Meyn P, Li Y, Yie TA, Lhakhang T, Olsen E, et
al.: Airway Microbiota Is Associated with Upregulation of the PI3K Pathway in Lung Cancer. Am
Author Manuscript

J Respir Crit Care Med 2018, 198:1188–1198. [PubMed: 29864375] **This study performed in
subjects with lung cancer and controls identifies how a lower airway microbiota enriched with oral
commensals can lead to upregulation of host transcriptomic signatures known to be associated
with carcinogenesis.
50. O'Dwyer DN, Ashley SL, Gurczynski SJ, Xia M, Wilke C, Falkowski NR, Norman KC, Arnold
KB, Huffnagle GB, Salisbury ML, et al.: Lung Microbiota Contribute to Pulmonary Inflammation
and Disease Progression in Pulmonary Fibrosis. Am J Respir Crit Care Med 2019, 199:1127–
1138. [PubMed: 30789747] **In this work, the investigators confirms that increased bacterial load
is associated with poor prognosis in pulmonary fibrosis. Importantly, the addition of a preclinical
experimental model of pulmonary fibrosis provvides mechanistic insight for this association
identified in humans.
51. Demain AL: Microbial secondary metabolism: a new theoretical frontier for academia, a new
opportunity for industry. Ciba Found Symp 1992, 171:3–16; discussion 16-23. [PubMed:
1302184]
52. Morono Y, Wishart JR, Ito M, Ijiri A, Hoshino T, Torres M, Verba C, Terada T, Inagaki F, Colwell
Author Manuscript

FS: Microbial Metabolism and Community Dynamics in Hydraulic Fracturing Fluids Recovered
From Deep Hydrocarbon-Rich Shale. Front Microbiol 2019, 10:376. [PubMed: 30915042]
53. Segal LN, Clemente JC, Li Y, Ruan C, Cao J, Danckers M, Morris A, Tapyrik S, Wu BG, Diaz P, et
al.: Anaerobic Bacterial Fermentation Products Increase Tuberculosis Risk in Antiretroviral-Drug-
Treated HIV Patients. Cell Host Microbe 2017, 21:530–537 e534. [PubMed: 28366509]
54. Sharpton TJ: An introduction to the analysis of shotgun metagenomic data. Front Plant Sci 2014,
5:209. [PubMed: 24982662]
55. Quince C, Walker AW, Simpson JT, Loman NJ, Segata N: Shotgun metagenomics, from sampling
to analysis. Nat Biotechnol 2017, 35:833–844. [PubMed: 28898207]

Curr Opin Microbiol. Author manuscript; available in PMC 2021 August 01.
Sulaiman et al. Page 9

56. Couto N, Schuele L, Raangs EC, Machado MP, Mendes CI, Jesus TF, Chlebowicz M, Rosema S,
Ramirez M, Carrico JA, et al.: Critical steps in clinical shotgun metagenomics for the concomitant
Author Manuscript

detection and typing of microbial pathogens. Sci Rep 2018, 8:13767. [PubMed: 30213965]
57. Clarke EL, Lauder AP, Hofstaedter CE, Hwang Y, Fitzgerald AS, Imai I, Biernat W, Rekawiecki B,
Majewska H, Dubaniewicz A, 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.
[PubMed: 28846439] *This is an investigation involving multiple sample types to evaluate the
lung microbiota in sarcoidosis. Importantly for this review, this is one of the few papers involving
whole genome shotgun sequencing in lower airway samples.
58. Cameron SJS, Lewis KE, Huws SA, Hegarty MJ, Lewis PD, Pachebat JA, Mur LAJ: A pilot study
using metagenomic sequencing of the sputum microbiome suggests potential bacterial biomarkers
for lung cancer. PLoS One 2017, 12:e0177062. [PubMed: 28542458]
59. Feigelman R, Kahlert CR, Baty F, Rassouli F, Kleiner RL, Kohler P, Brutsche MH, von Mering C:
Sputum DNA sequencing in cystic fibrosis: non-invasive access to the lung microbiome and to
pathogen details. Microbiome 2017, 5:20. [PubMed: 28187782]
60. Lim YW, Evangelista JS 3rd, Schmieder R, Bailey B, Haynes M, Furlan M, Maughan H, Edwards
Author Manuscript

R, Rohwer F, Conrad D: Clinical insights from metagenomic analysis of sputum samples from
patients with cystic fibrosis. J Clin Microbiol 2014, 52:425–437. [PubMed: 24478471]
61. Moran Losada P, Chouvarine P, Dorda M, Hedtfeld S, Mielke S, Schulz A, Wiehlmann L, Tummler
B: The cystic fibrosis lower airways microbial metagenome. ERJ Open Res 2016, 2.
62. Bashiardes S, Zilberman-Schapira G, Elinav E: Use of Metatranscriptomics in Microbiome
Research. Bioinform Biol Insights 2016, 10:19–25. [PubMed: 27127406]
63. Lee SW, Kuan CS, Wu LS, Weng JT: Metagenome and Metatranscriptome Profiling of Moderate
and Severe COPD Sputum in Taiwanese Han Males. PLoS One 2016, 11:e0159066. [PubMed:
27428540]
64. Ren L, Zhang R, Rao J, Xiao Y, Zhang Z, Yang B, Cao D, Zhong H, Ning P, Shang Y, et al.:
Transcriptionally Active Lung Microbiome and Its Association with Bacterial Biomass and Host
Inflammatory Status. mSystems 2018, 3.*This work is one of the few papers that have attempted
to perform metatranscriptome of lower airway samples, something that can be expected to be quite
challenging with low biomass samples.
65. Gollwitzer ES, Saglani S, Trompette A, Yadava K, Sherburn R, McCoy KD, Nicod LP, Lloyd CM,
Author Manuscript

Marsland BJ: Lung microbiota promotes tolerance to allergens in neonates via PD-L1. Nat Med
2014, 20:642–647. [PubMed: 24813249]
66. Dickson RP, Singer BH, Newstead MW, Falkowski NR, Erb-Downward JR, Standiford TJ,
Huffnagle GB: Enrichment of the lung microbiome with gut bacteria in sepsis and the acute
respiratory distress syndrome. Nat Microbiol 2016, 1:16113. [PubMed: 27670109]
67. Segal LN, Clemente JC, Wu BG, Wikoff WR, Gao Z, Li Y, Ko JP, Rom WN, Blaser MJ, Weiden
MD: Randomised, double-blind, placebo-controlled trial with azithromycin selects for anti-
inflammatory microbial metabolites in the emphysematous lung. Thorax 2017, 72:13–22.
[PubMed: 27486204] *One of the few investigations where research bronchoscopies were repeated
over time to evaluate the stability of the lung microbiota and the effects of azithromycin.
68. Twigg HL 3rd, Knox KS, Zhou J, Crothers KA, Nelson DE, Toh E, Day RB, Lin H, Gao X, Dong
Q, et al.: Effect of Advanced HIV Infection on the Respiratory Microbiome. Am J Respir Crit Care
Med 2016, 194:226–235. [PubMed: 26835554]
69. Dickson RP, Erb-Downward JR, Martinez FJ, Huffnagle GB: The Microbiome and the Respiratory
Author Manuscript

Tract. Annu Rev Physiol 2016, 78:481–504. [PubMed: 26527186]

Curr Opin Microbiol. Author manuscript; available in PMC 2021 August 01.
Sulaiman et al. Page 10
Author Manuscript
Author Manuscript

Figure 1: Technical challenges for lung microbiome research using culture independent
Author Manuscript

techniques.
A. Variable biomass through the respiratory tree affect the composition and signal to noise
ratio of each type of sample. B. While bacterial clearance in the lower airways is a well-
established physiological event the dynamics of the microbial molecular signatures need to
be studied.
Author Manuscript

Curr Opin Microbiol. Author manuscript; available in PMC 2021 August 01.

You might also like