Skip to main content
Annals of the American Thoracic Society logoLink to Annals of the American Thoracic Society
. 2014 Jan;11(Suppl 1):S21–S27. doi: 10.1513/AnnalsATS.201306-189MG

A Brave New World: The Lung Microbiota in an Era of Change

Leopoldo N Segal 1,, Martin J Blaser 1
PMCID: PMC3972973  PMID: 24437400

Abstract

The development of culture-independent techniques has revolutionized our understanding of how our human cells interact with the even greater number of microbial inhabitants of our bodies. As part of this revolution, data are increasingly challenging the old dogma that in health, the lung mucosa is sterile. To understand how the lung microbiome may play a role in human health, we identified five major questions for lung microbiome research: (1) Is the lung sterile? (2) Is there a unique core microbiome in the lung? (3) How dynamic are the microbial populations? (4) How do pulmonary immune responses affect microbiome composition? and (5) Are the lungs influenced by the intestinal immune responses to the gut microbiome? From birth, we are exposed to continuous microbial challenges that shape our microbiome. In our changing environment, perturbation of the gut microbiome affects both human health and disease. With widespread antibiotic use, the ancient microbes that formerly resided within us are being lost, for example, Helicobacter pylori in the stomach. Animal models show that antibiotic exposure in early life has developmental consequences. Considering the potential effects of this altered microbiome on pulmonary responses will be critical for future investigations.

Keywords: lung, microbiome, antibiotics, immune responses, inflammation


With the advances in culture-independent techniques that have occurred, there has been renewed interest in searching for microbes in the lung. New observations are challenging the old dogma that in health, the lung mucosa is sterile (1, 2). With the logarithmic improvements in sequencing technology and bioinformatics analysis, the study of the lung microbiome has received increasing interest. The changing research environment represents a new frontier to address important long-standing problems in human health.

Key Questions about the Lung Microbiome

1. Is the Lung Sterile?

For many years, this question did not seem relevant, given the failure of culture-dependent techniques to describe residential bacteria in the healthy lung. Among various factors, the low culturable bacterial burden in the lung, contamination with upper airway microbiota during sampling, and the difficulties in growing fastidious bacteria have contributed to the presumption that the lung is sterile (3, 4). With the use of culture-independent techniques, there is now substantial literature identifying the presence of bacteria products in the lower airways of normal individuals (58). Data from quantitative PCR for 16S ribosomal RNA (rRNA) genes confirm that the bacterial burden in the lower airways is less than in the upper airways. However, a diverse microbiome has been described in the lung of healthy subjects. A common finding in the lung is the presence of taxa often represented in the oral cavity (5, 811). Because sampling of the lower airways is usually performed by bronchoscopy, there is controversy as to whether these microbes represent bronchoscopic carryover of upper airway microbiota and/or microaspiration (Figure 1). The decrease in bacterial loads observed in subsequent samples has been suggested as supporting a significant carryover effect (5). However, this has not been a consistent finding in all studies, which suggests substantial technical heterogeneity (8). In contrast, it is well known that microaspiration is a common phenomenon among healthy persons, and that it is exacerbated in several respiratory disease states (1214). The finding of low bacterial burden in the normal lung supports the view that although the healthy lung has the ability to clear microaspirated microbes, a residual microbiota coexists in the lung mucosa. The ability to clear microorganisms also is commonly impaired in lung diseases (15, 16). Both carryover and microaspiration can variably contribute to the observed enrichment of the lower airway microbiome with upper airway taxa. Bronchoscopic techniques that are better able to minimize the contribution of carryover are needed. Further, considering the low bacterial burden in most lung samples, a high background microbiome signal commonly interferes with the interpretation of lung microbiome data (8). It is therefore expected that an unfavorable signal-to-noise ratio will commonly occur when trying to evaluate the lung microbiome of airways with low bacterial burden. New computational background subtraction techniques based on mathematical modeling, such as a neutral model, single-sided outlier test and SourceTracker, may account for carryover and have the potential to identify true lung taxa (7, 17, 18).

Figure 1.

Figure 1.

Key questions involving the lung microbiota.

One final dilemma in addressing this question concerns determining the viability of the lung microbiome. Classically, culture techniques have been used to address this question. Using 16S rRNA PCR techniques, current lung microbiome data in healthy lung may reflect only bacterial DNA fragments (59, 17, 19, 20). With the current data available and given the high sensitivity of powerful high-throughput sequencing technology, we may not be ready to answer this question. However, this limitation does not invalidate the relevance of determining the characteristics of a microbiome composed mainly of dead bacteria: the taxonomic composition may reflect a specific conglomerate of various microbial-associated molecular patterns (MAMPs) from nonviable bacteria that can elicit significant immunomodulatory effects on the host.

2. Is There a Core Microbiome?

Are the organisms found in the lung unique? One challenging observation is that the lower airway microbiome is more similar to the upper airway microbiome of the same individual than it is to the lung microbiome of a different person (17). Although part of this might be explained if significant carryover had occurred, most data suggest substantial diversity in healthy individuals (79, 19). Therefore, it is important to establish the degree of overlap between different individuals. Conceptually, the search for a shared core is consistent with the presumption that the lung environment imposes selection pressure on the microbial inhabitants in the lung. As an example, data have shown the presence of Tropheryma whipplei in the lung of normal subjects (6). Importantly, this microorganism was not found in upper airway samples when 500 sequences per sample were evaluated. This observation suggests that T. whipplei reaches the lung by either hematogenous spread or by microaspiration. In either circumstance, the data suggest that the lung is a true niche for T. whipplei. This tropism for the lung might explain why it is easier to find T. whipplei in the lower airways than in upper airway samples. The evidence that colonization of the lower airways with this microbe was enhanced in immunodeficiency further suggests a dynamic state for the host–microbiota interaction. We therefore postulate that two components of the microbiome may elicit synergistic host responses: one in which unique taxa find a special niche in the lung environment, and a second in which MAMPs exert an immunomodulatory role. These two components of the lung microbiome deserve further exploration.

3. What Are the Dynamics of the Lung Microbiome?

With the common use of antibiotics and antiinflammatory drugs, a significant impact on the lung microbiome can be expected. Disturbances of the lung microbial community can lead to either no change (resistance), an altered microbiota that may return to its original composition (resilience), a permanently altered microbiota that could be functionally similar (functional redundancy), or a microbiota permanently altered in both composition and function. These dynamic changes might be relevant for our understanding of pathogenesis in pulmonary health and disease. In the lung, microaspiration commonly is a repetitive phenomenon (12). As discussed previously, this might explain the frequent observation of oral taxa such as Prevotella and Veillonella in the lower airway microbiome. Such periodic exposure of the lower airways to upper airway microorganisms represents an important seeding mechanism that may influence microbial selection in the lower airways. As an example of this selection pressure, S. pneumoniae, a minor component of both the upper and lower airway microbiomes, causes more than half of all cases of community-acquired bacterial pneumonia. Further, dynamic changes in lower airway microbiota may reflect changes in bacterial tropism related to the development of lung injury, and/or immunosuppression states (such as in cystic fibrosis, bronchiectasis, alcoholism, or HIV infection). Several examples of dynamic changes in airway microbiota have been proposed to substantially affect disease progression. In chronic obstructive pulmonary disease, exacerbations are commonly caused by acquisition of new strains and associated with increased inflammation and decreased lung function (21). In cystic fibrosis, changes in bacterial diversity are associated with disease progression and consequent colonization with pathogens (22). Diversity of airway microbiota in cystic fibrosis has been found to decrease significantly with disease stage (22, 23). Also, the composition of the airway microbiota before an exacerbation event may be relevant to determine the microbial constituents involved in the exacerbations (24). Lower airway microbiota diversity (as measured by Shannon diversity indices) and a Pseudomonas-dominated microbiome preceding the exacerbation are predictive of larger changes in microbiome structure during exacerbations. These data indicate that a dynamic change from a “healthy” to an “unhealthy” airway microbiota leads to a susceptible microbial environment necessary for pathogen enrichment and host injury. Investigation of the dynamic changes in the lung microbiome will require prospective and longitudinal studies.

4. What Are the Mucosal Inflammatory Implications of the Lung Microbiome?

The relationships between our human cells and our inhabitant microorganisms are not accidental, but likely have been programmed over long periods of time (25). The role of the gut microbiota in shaping the mucosal immune system is well understood (26, 27). The composition of the intestinal microbiota regulates the balance between helper T type 17 cells and T-regulatory cells (Th17:Treg balance) in the small intestinal lamina propria (26). Compared with conventionally colonized animals, germ-free mice have defective development of immunity, with fewer CD4+ and CD8+ T cells (28, 29). However, this symbiosis between the gut microbiome and the intestinal mucosa relies on barrier mechanisms that exclude most bacteria, preventing invasion and leading to tolerance to the continuous exposure by bacterial products. Loss of these important mechanisms may lead to bacterial invasion and chronic inflammatory processes, such as inflammatory bowel disease (30). In the lung, little is known about the effects of the airway microbiome on immune maturation and the types of tolerance that occur in the lower airway, which may in fact be relevant to the symbiotic interactions. Data from two large European studies showed that specified environmental exposures could be protective against development of asthma and atopy (31, 32). The use of molecular techniques to evaluate specific patterns of exposure to farm-related bacteria or fungi in two cohorts (PARSIFAL: Prevention of Allergy Risk Factors for Sensitization in Children Related to Farming and Anthroposophic Lifestyle; and GABRIELA: Multidisciplinary Study to Identify the Genetic and Environmental Causes of Asthma in the European Community [GABRIEL] Advanced Study) has further characterized certain environmental microbiomes as protective against asthma (33). Increased environmental exposure to microbial products has been shown to be protective factor against allergy (34, 35). This microbial exposure occurs in early childhood and is determined by mode of birth delivery, farm habitation, or antibiotic use, all of which have been shown to be factors conferring protection or conversely increasing risk of asthma later in life (33, 3640). This has renewed interest in the “hygiene hypothesis,” which states that changing diets, improved sanitary conditions, and increased use of antibiotics prevent the exposure to microbes needed for adequate immune maturation in early life. This results in T-cell subset imbalances, such as Treg cell deficiencies, that may predispose to immune diseases.

In asymptomatic adults, enrichment of the lung microbiome with oral-characteristic taxa is associated with increased inflammatory cells and exhaled nitric oxide, suggesting that there are specific inflammatory responses to the airway microbiome (8). Although it might seem obvious that the microbiota plays a role in stimulating a host immune response, it remains unclear whether the observed associations are causal or not. Furthermore, it is likely that this is a bidirectional association and that the characteristics of the airway microbiota are determined by the host immune response as well. This is suggested by the observation that subjects with immunodeficiency have a lung microbiome enriched with T. whipplei and that there is subsequent reduction of the relative abundance of T. whipplei with antiretroviral therapy (6). Other potential roles of the airway microbiome may be related to metabolic processes (e.g., drug metabolism), control of surfactant production, or gas exchange, areas for which more investigation is needed.

5. Is There Cross-Talk between the Intestinal Microbiome–Mucosa Interaction and the Lung?

Analyses of the intestinal tract show that gut microbiota composition is relevant for the education of the immune system. Furthermore, there is increasing evidence that the gastrointestinal mucosa is the predominant site of microbiota–host interaction, and can play a role in the development of immune responses at distal mucosal sites. Several lines of investigation support the concept of cross-talk between the intestinal microbiome–mucosa interaction and the lung. In mice, disruption of the gastrointestinal microbiota leads to abnormal airway allergic responses (41, 42). In other mouse models, oral ingestion of various strains of Lactobacillus and ingestion of bacterial products modulate allergic pulmonary inflammation (4345). More importantly, there is evidence that children whose stomach is colonized with Helicobacter pylori are 40–60% less likely to have childhood-onset asthma than children who are not carriers (46). Similarly, reduced intestinal microbiome diversity in infants is associated with increased risk for allergic rhinitis and peripheral blood eosinophilia (47). Taken together, these data support the hypothesis that the gut microbiota shapes the systemic immune system, thereby affecting the lung mucosa. Alternatively, changes in gut microbiota might also reflect changes in the oropharyngeal microbiota, which may directly affect the lung microbiota and host immune response through microaspiration. Ultimately, both the gut and lung mucosa may function as a single system-wide organ and share the physiological function of immune surveillance and shaping of host responses. More investigation is needed to determine the immunological mechanisms that link these two mucosal sites.

Evidence That the Human Microbiome Is Changing, and Potential Consequences

Ancient, Vertical

Animals have had residential microbes colonizing them ever since animals first appeared on this planet, about 500 million years ago. These microbes live in specific niches in and on their hosts, and often are persistent for sizeable fractions of the host’s lifetime. Importantly, a large fraction of these residential microbes, which can be considered the normal microbiome of their host, can be vertically transmitted from mother to offspring (48). This has been true for countless millions of years, and includes all mammals, such as humans.

Onslaught

In the period since World War II, there have been sharp rises in the rates of many important diseases including asthma, type 1 diabetes mellitus, and obesity (49). We have focused our research efforts on obesity, because of the rising epidemic in the United States (Figure 2A) and other developed countries.

Figure 2.

Figure 2.

(A) Obesity trends in U.S. adults: evidence of changing physiology. Source: CDC behavioral risk factor surveillance system. Available from: http://www.cdc.gov/obesity/data/adult.html. (B) Outpatient antibiotic usage rates by region of the United States in 2010. Reproduced by permission from Reference 62.

Birth

The transfer of microbiota from mothers to human babies is facing unprecedented constraints. At the outset, it is fair to say that we do not know the magnitude of this problem, whether the constraints are large or trivial, but they are extensive. About half of all pregnant women in the United States are receiving antibiotics while they are pregnant. These can lead to extinctions in their microbiota just before the transfer to their offspring. Especially if received just before birth, for example, to prevent group B Streptococcus infection, the antibiotic also may be present in the baby’s blood and the mother’s milk. These would create further selection for organisms resistant to the antibiotic and change the environment in the baby for the receipt of the hand-off of organisms from both mother and environment. In addition, about one-third of all babies born in the United States are delivered via Caesarian section. In Brazil and urban China, the C-section rate approaches 50%, and even in developing countries, such as Ecuador and Iran, the rates exceed 40%. One consequence of C-section is that the baby no longer transits the vagina, and is not coated with the vaginal microbiota (50). In consequence, the initial microbiota in C-section and vaginally delivered babies at all sites—skin, mouth, and intestinal tract—differ immediately after birth (48). When and if the microbiota ever fully return to a normal developmental pathway is not yet known.

Data suggest that the ability to mount a balanced immune response, fundamental for managing healthy airways, is influenced by early exposure to pathogenic bacteria (51, 52). Upper airway colonization with Moraxella catarrhalis and Haemophilus influenzae in early life is associated with Th2 and Th17 inflammatory patterns that may recruit and activate eosinophils and neutrophils while counteracting Th1 responses (52). Further, respiratory syncytial virus infection early in life affects regulatory T-cell phenotypic plasticity, lowering their suppressive capacity and rendering the host more susceptible to asthma later in life (53). This airway exposure to pathogenic microbes in early life may contribute to the development of chronic inflammation and increased risk for asthma.

Failure to establish a healthy gut microbiota early in life also alters maturation of the immune system, promoting altered allergic responses in adulthood (54, 55). In a mouse model, oral vancomycin causes an exacerbated allergic response, whereas streptomycin does not, suggesting that microbial composition can influence allergic sensitization (56). In a murine model of allergic airway disease, germ-free mice exhibit more severe disease than conventionally housed controls, an effect that could be ameliorated by recolonization with conventional microbiota (57). Similar results were found when disturbance of the gut microbiome was induced by administration of antibiotics in drinking water or by an antibiotic–fungal microbiota combination (42, 58). Microbial colonization early in life regulates mucosal invariant natural killer T-cell tolerance and can impact immune responses at mucosal sites later in adulthood (59). Regulatory T-cell–mediated tolerance mechanisms may be disrupted when specific gut microbial populations, such as Clostridium species, have been lost or altered (27, 56). Therefore, gut colonization early in life plays an important role in guiding immune development and maintaining mucosal homeostasis throughout life (60, 61).

The onslaught on the inherited microbiota does not end at birth. Young children frequently receive antibiotics. It is clear that there is substantial overuse of antibiotics, especially for upper respiratory infections including otitis media. There have been some small improvements in reducing this problem; nevertheless, Centers for Disease Control and Prevention data (62) indicate that children are receiving more than one course of antibiotics on average in each of the first 2 years of life (Table 1). Extrapolating from the prevalence data in 2010 across the United States suggests that the average child may be receiving nearly 3 courses of antibiotics by the time they are 2, 11 courses by the time they are 10, and 17 courses by the time they are 20 years old. Such data are consistent with more limited studies in other developed countries (6365).

Table 1.

Cumulative outpatient antibiotic use, by age

Patient Age Group (yr) Number of prescriptions (millions) Prescriptions/1,000 People Average Number of Courses
During Period Cumulative
0–1 16.6 1,365 2.73 2.73
2–9 29 1,021 8.17 10.9
10–19 28.9 677 6.78 17.68
20–39 55.4 669 13.38 31.06
40–64 81.6 797 19.93 50.98
≥65 41.1 1,020
Total 258 833    

Adapted by permission from Reference 62.

In fact, antibiotic usage in the United States shows important regional differences (Figure 2B). In 2010, 258 million courses of antibiotics were prescribed to outpatients in the United States, representing 833 courses per 1,000 people. In the Northeast and Midwest, rates were similar to the national averages, but the rates in the West (638/1,000 people) were about 50% lower than in the South (936/1,000 people). Because the incidence and severity of bacterially induced infectious diseases of Southerners and Westerners probably do not differ by 50%, the differences in antibiotic use likely indicate differences in medical custom and practice. Interestingly, the 2010 maps on obesity prevalence and antibiotic use show striking parallels. Such observations raise the hypothesis that excess antibiotic usage is one of the drivers of the current obesity epidemic.

Consequences on the Farm

Support for this idea comes from observations on the farm, beginning in the 1940s. At that time, it was learned that feeding livestock with low doses of antibiotics (called subtherapeutic antibiotic treatment, which we abbreviate as STAT) promoted the growth of farm animals (66, 67). There were three important observations:

  • 1.

    STAT was effective across a broad range of livestock, from chickens to cows, representing a broad swath of vertebrate evolution.

  • 2.

    The effects were seen with a broad range of antibacterial agents, regardless of chemical structure, drug classification, mechanism of action, or targeted organisms. Antiviral and antifungal agents were not effective.

  • 3.

    The younger in life the treatment was begun, the greater the effect from STAT.

These last observations suggested the hypothesis that the STAT regimens were affecting the development of the animals. As such, we sought to address this question in a mouse model (68).

Mouse Models

Initial work focused on C57/BL6 mice. Providing a variety of antibiotics to mice in a STAT model showed that each regimen tested changed the development of adiposity in the mice, as well as early-life bone development (68). Studies of the microbiota showed a shift in composition as well as a change in gene abundances related to short-chain fatty acid synthesis, and there was evidence in the liver of widespread changes in expression of genes involved with intermediate carbohydrate metabolism and with lipid production and transport. On-going work has confirmed and extended the findings in terms of the windows of exposure, and the combined effects of dietary and antibiotic interventions. These studies provide a model system for exploring the role of early life antibiotic exposures on host development, and may provide clues about the etiology of the obesity epidemic.

Effects on the Lung

Is widespread antibiotic use having effects on the formation and dynamics of the lung microbiome? At this point, it is clear that we just do not know. Antibiotics could affect the lung microbiome either directly through their selective force, or indirectly through effects on the gut microbiome and its downstream effects on immunity. Our “other” genome, as the human microbiome has been considered, is facing an era of major changes, induced by lifestyle, diet, and antibiotic use. The use of culture-independent 16S rRNA and metagenomic approaches provides a new vision to our understanding of the lung as a unique mucosal niche exposed to a complex microbial environment. We are just learning about associations involving the microbiome of the gastrointestinal tract and the airways, as well as the immunological consequences of the human microbiome. The combination of modern changes in microbial environment and technological developments that allow understanding the human microbiome provides a unique opportunity to explore mechanisms of disease in this brave new world.

Footnotes

Supported by R01 DK090989; UH2 AR57506; the Diane Belfer Program for Human Microbial Ecology; the Knapp Family Foundation; and UL1 TR000038.

Author disclosures are available with the text of this article at www.atsjournals.org.

References

  • 1.Wilson R, Dowling RB, Jackson AD. The biology of bacterial colonization and invasion of the respiratory mucosa. Eur Respir J. 1996;9:1523–1530. doi: 10.1183/09031936.96.09071523. [DOI] [PubMed] [Google Scholar]
  • 2.Kahn FW, Jones JM. Diagnosing bacterial respiratory infection by bronchoalveolar lavage. J Infect Dis. 1987;155:862–869. doi: 10.1093/infdis/155.5.862. [DOI] [PubMed] [Google Scholar]
  • 3.Monso E, Ruiz J, Rosell A, Manterola J, Fiz J, Morera J, Ausina V. Bacterial infection in chronic obstructive pulmonary disease: a study of stable and exacerbated outpatients using the protected specimen brush. Am J Respir Crit Care Med. 1995;152:1316–1320. doi: 10.1164/ajrccm.152.4.7551388. [DOI] [PubMed] [Google Scholar]
  • 4.Goldstein EJ, Citron DM, Goldman PJ, Goldman RJ. National hospital survey of anaerobic culture and susceptibility methods: III. Anaerobe. 2008;14:68–72. doi: 10.1016/j.anaerobe.2008.01.001. [DOI] [PubMed] [Google Scholar]
  • 5.Charlson ES, Bittinger K, Haas AR, Fitzgerald AS, Frank I, Yadav A, Bushman FD, Collman RG. Topographical continuity of bacterial populations in the healthy human respiratory tract. Am J Respir Crit Care Med. 2011;184:957–963. doi: 10.1164/rccm.201104-0655OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Lozupone C, Cota-Gomez A, Palmer BE, Linderman DJ, Charlson ES, Sodergren E, Mitreva M, Abubucker S, Martin J, Yao G, et al. Widespread colonization of the lung by Tropheryma whipplei in HIV infection. Am J Respir Crit Care Med. 2013;187:1110–1117. doi: 10.1164/rccm.201211-2145OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Morris A, Beck JM, Schloss PD, Campbell TB, Crothers K, Curtis JL, Flores SC, Fontenot AP, Ghedin E, Huang L, et al. Comparison of the respiratory microbiome in healthy non-smokers and smokers. Am J Respir Crit Care Med. 2013;187:1067–1075. doi: 10.1164/rccm.201210-1913OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.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. doi: 10.1186/2049-2618-1-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Hilty M, Burke C, Pedro H, Cardenas P, Bush A, Bossley C, Davies J, Ervine A, Poulter L, Pachter L, et al. Disordered microbial communities in asthmatic airways. PLoS One. 2010;5:e8578. doi: 10.1371/journal.pone.0008578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Huang YJ, Nelson CE, Brodie EL, Desantis TZ, Baek MS, Liu J, Woyke T, Allgaier M, Bristow J, Wiener-Kronish JP, et al. Airway microbiota and bronchial hyperresponsiveness in patients with suboptimally controlled asthma J Allergy Clin Immunol 2011127372–381.e1–3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Segal LN, Methe BA, Nolan A, Hoshino Y, Rom WN, Dawson R, Bateman E, Weiden MD. HIV-1 and bacterial pneumonia in the era of antiretroviral therapy. Proc Am Thorac Soc. 2011;8:282–287. doi: 10.1513/pats.201006-044WR. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Gleeson K, Eggli DF, Maxwell SL. Quantitative aspiration during sleep in normal subjects. Chest. 1997;111:1266–1272. doi: 10.1378/chest.111.5.1266. [DOI] [PubMed] [Google Scholar]
  • 13.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. doi: 10.1111/j.1440-1843.2010.01875.x. [DOI] [PubMed] [Google Scholar]
  • 14.Rascon-Aguilar IE, Pamer M, Wludyka P, Cury J, Coultas D, Lambiase LR, Nahman NS, Vega KJ. Role of gastroesophageal reflux symptoms in exacerbations of COPD. Chest. 2006;130:1096–1101. doi: 10.1378/chest.130.4.1096. [DOI] [PubMed] [Google Scholar]
  • 15.Randell SH, Boucher RC. Effective mucus clearance is essential for respiratory health. Am J Respir Cell Mol Biol. 2006;35:20–28. doi: 10.1165/rcmb.2006-0082SF. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Taylor AE, Finney-Hayward TK, Quint JK, Thomas CM, Tudhope SJ, Wedzicha JA, Barnes PJ, Donnelly LE. Defective macrophage phagocytosis of bacteria in COPD. Eur Respir J. 2010;35:1039–1047. doi: 10.1183/09031936.00036709. [DOI] [PubMed] [Google Scholar]
  • 17.Charlson ES, Bittinger K, Chen J, Diamond JM, Li H, Collman RG, Bushman FD. Assessing bacterial populations in the lung by replicate analysis of samples from the upper and lower respiratory tracts. PLoS One. 2012;7:e42786. doi: 10.1371/journal.pone.0042786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Knights D, Kuczynski J, Charlson ES, Zaneveld J, Mozer MC, Collman RG, Bushman FD, Knight R, Kelley ST. Bayesian community-wide culture-independent microbial source tracking. Nat Methods. 2011;8:761–763. doi: 10.1038/nmeth.1650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Erb-Downward JR, Thompson DL, Han MK, Freeman CM, McCloskey L, Schmidt LA, Young VB, Toews GB, Curtis JL, Sundaram B, et al. Analysis of the lung microbiome in the “healthy” smoker and in COPD. PLoS One. 2011;6:e16384. doi: 10.1371/journal.pone.0016384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.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. doi: 10.1164/rccm.201111-2075OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Sethi S, Evans N, Grant BJ, Murphy TF. New strains of bacteria and exacerbations of chronic obstructive pulmonary disease. N Engl J Med. 2002;347:465–471. doi: 10.1056/NEJMoa012561. [DOI] [PubMed] [Google Scholar]
  • 22.Zhao J, Schloss PD, Kalikin LM, Carmody LA, Foster BK, Petrosino JF, Cavalcoli JD, VanDevanter DR, Murray S, Li JZ, et al. Decade-long bacterial community dynamics in cystic fibrosis airways. Proc Natl Acad Sci USA. 2012;109:5809–5814. doi: 10.1073/pnas.1120577109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Fodor AA, Klem ER, Gilpin DF, Elborn JS, Boucher RC, Tunney MM, Wolfgang MC. The adult cystic fibrosis airway microbiota is stable over time and infection type, and highly resilient to antibiotic treatment of exacerbations. PLoS One. 2012;7:e45001. doi: 10.1371/journal.pone.0045001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Carmody LA, Zhao J, Schloss PD, Petrosino JF, Murray S, Young VB, Li JZ, LiPuma JJ. Changes in cystic fibrosis airway microbiota at pulmonary exacerbation. Ann Am Thorac Soc. 2013;10:179–187. doi: 10.1513/AnnalsATS.201211-107OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Blaser MJ. Harnessing the power of the human microbiome. Proc Natl Acad Sci USA. 2010;107:6125–6126. doi: 10.1073/pnas.1002112107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Ivanov II, Frutos Rde L, Manel N, Yoshinaga K, Rifkin DB, Sartor RB, Finlay BB, Littman DR. Specific microbiota direct the differentiation of IL-17–producing T-helper cells in the mucosa of the small intestine. Cell Host Microbe. 2008;4:337–349. doi: 10.1016/j.chom.2008.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Atarashi K, Tanoue T, Shima T, Imaoka A, Kuwahara T, Momose Y, Cheng G, Yamasaki S, Saito T, Ohba Y, et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science. 2011;331:337–341. doi: 10.1126/science.1198469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Chung H, Pamp SJ, Hill JA, Surana NK, Edelman SM, Troy EB, Reading NC, Villablanca EJ, Wang S, Mora JR, et al. Gut immune maturation depends on colonization with a host-specific microbiota. Cell. 2012;149:1578–1593. doi: 10.1016/j.cell.2012.04.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Jiang HQ, Thurnheer MC, Zuercher AW, Boiko NV, Bos NA, Cebra JJ. Interactions of commensal gut microbes with subsets of B- and T-cells in the murine host. Vaccine. 2004;22:805–811. doi: 10.1016/j.vaccine.2003.11.022. [DOI] [PubMed] [Google Scholar]
  • 30.Duchmann R, Kaiser I, Hermann E, Mayet W, Ewe K, Meyer zum Buschenfelde KH. Tolerance exists towards resident intestinal flora but is broken in active inflammatory bowel disease (IBD) Clin Exp Immunol. 1995;102:448–455. doi: 10.1111/j.1365-2249.1995.tb03836.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Vartiainen E, Petays T, Haahtela T, Jousilahti P, Pekkanen J. Allergic diseases, skin prick test responses, and IgE levels in north Karelia, Finland, and the republic of Karelia, Russia. J Allergy Clin Immunol. 2002;109:643–648. doi: 10.1067/mai.2002.123307. [DOI] [PubMed] [Google Scholar]
  • 32.von Mutius E, Radon K.Living on a farm: impact on asthma induction and clinical course Immunol Allergy Clin North Am 200828631–647.ix–x [DOI] [PubMed] [Google Scholar]
  • 33.Ege MJ, Mayer M, Normand AC, Genuneit J, Cookson WO, Braun-Fahrlander C, Heederik D, Piarroux R, von Mutius E. Exposure to environmental microorganisms and childhood asthma. N Engl J Med. 2011;364:701–709. doi: 10.1056/NEJMoa1007302. [DOI] [PubMed] [Google Scholar]
  • 34.Ege MJ, Bieli C, Frei R, van Strien RT, Riedler J, Ublagger E, Schram-Bijkerk D, Brunekreef B, van Hage M, Scheynius A, et al. Prenatal farm exposure is related to the expression of receptors of the innate immunity and to atopic sensitization in school-age children. J Allergy Clin Immunol. 2006;117:817–823. doi: 10.1016/j.jaci.2005.12.1307. [DOI] [PubMed] [Google Scholar]
  • 35.Roduit C, Wohlgensinger J, Frei R, Bitter S, Bieli C, Loeliger S, Buchele G, Riedler J, Dalphin JC, Remes S, et al. Prenatal animal contact and gene expression of innate immunity receptors at birth are associated with atopic dermatitis J Allergy Clin Immunol 2011127179–185.185.e1 [DOI] [PubMed] [Google Scholar]
  • 36.van Nimwegen FA, Penders J, Stobberingh EE, Postma DS, Koppelman GH, Kerkhof M, Reijmerink NE, Dompeling E, van den Brandt PA, Ferreira I, Mommers M, et al. Mode and place of delivery, gastrointestinal microbiota, and their influence on asthma and atopy J Allergy Clin Immunol 2011128948–955.e1–3 [DOI] [PubMed] [Google Scholar]
  • 37.Bager P, Melbye M, Rostgaard K, Benn CS, Westergaard T. Mode of delivery and risk of allergic rhinitis and asthma. J Allergy Clin Immunol. 2003;111:51–56. doi: 10.1067/mai.2003.34. [DOI] [PubMed] [Google Scholar]
  • 38.Marra F, Marra CA, Richardson K, Lynd LD, Kozyrskyj A, Patrick DM, Bowie WR, Fitzgerald JM. Antibiotic use in children is associated with increased risk of asthma. Pediatrics. 2009;123:1003–1010. doi: 10.1542/peds.2008-1146. [DOI] [PubMed] [Google Scholar]
  • 39.Murk W, Risnes KR, Bracken MB. Prenatal or early-life exposure to antibiotics and risk of childhood asthma: a systematic review. Pediatrics. 2011;127:1125–1138. doi: 10.1542/peds.2010-2092. [DOI] [PubMed] [Google Scholar]
  • 40.Depner M, Ege MJ, Genuneit J, Pekkanen J, Roponen M, Hirvonen MR, Dalphin JC, Kaulek V, Krauss-Etschmann S, Riedler J, et al. Atopic sensitization in the first year of life. J Allergy Clin Immunol. 2013;131:781–788. doi: 10.1016/j.jaci.2012.11.048. [DOI] [PubMed] [Google Scholar]
  • 41.Noverr MC, Falkowski NR, McDonald RA, McKenzie AN, Huffnagle GB. Development of allergic airway disease in mice following antibiotic therapy and fungal microbiota increase: role of host genetics, antigen, and interleukin-13. Infect Immun. 2005;73:30–38. doi: 10.1128/IAI.73.1.30-38.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Noverr MC, Noggle RM, Toews GB, Huffnagle GB. Role of antibiotics and fungal microbiota in driving pulmonary allergic responses. Infect Immun. 2004;72:4996–5003. doi: 10.1128/IAI.72.9.4996-5003.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Forsythe P, Inman MD, Bienenstock J. Oral treatment with live Lactobacillus reuteri inhibits the allergic airway response in mice. Am J Respir Crit Care Med. 2007;175:561–569. doi: 10.1164/rccm.200606-821OC. [DOI] [PubMed] [Google Scholar]
  • 44.Ezendam J, van Loveren H. Lactobacillus casei shirota administered during lactation increases the duration of autoimmunity in rats and enhances lung inflammation in mice. Br J Nutr. 2008;99:83–90. doi: 10.1017/S0007114507803412. [DOI] [PubMed] [Google Scholar]
  • 45.Kitagaki K, Businga TR, Kline JN. Oral administration of CpG-ODNs suppresses antigen-induced asthma in mice. Clin Exp Immunol. 2006;143:249–259. doi: 10.1111/j.1365-2249.2005.03003.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Chen Y, Blaser MJ. Inverse associations of Helicobacter pylori with asthma and allergy. Arch Intern Med. 2007;167:821–827. doi: 10.1001/archinte.167.8.821. [DOI] [PubMed] [Google Scholar]
  • 47.Bisgaard H, Li N, Bonnelykke K, Chawes BL, Skov T, Paludan-Muller G, Stokholm J, Smith B, Krogfelt KA.Reduced diversity of the intestinal microbiota during infancy is associated with increased risk of allergic disease at school age J Allergy Clin Immunol 2011128646–652.e1–5 [DOI] [PubMed] [Google Scholar]
  • 48.Dominguez-Bello MG, Costello EK, Contreras M, Magris M, Hidalgo G, Fierer N, Knight R. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc Natl Acad Sci USA. 2010;107:11971–11975. doi: 10.1073/pnas.1002601107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Bach JF. The effect of infections on susceptibility to autoimmune and allergic diseases. N Engl J Med. 2002;347:911–920. doi: 10.1056/NEJMra020100. [DOI] [PubMed] [Google Scholar]
  • 50.Organisation for Economic Co-operation and Development (OECD)Caesarean sections. In: Health at a glance 2011: OECD indicators. Paris, France: OECD Publishing; 2011. Available from: http://dx.doi.org/10.1787/health_glance-2011-en [Google Scholar]
  • 51.Bisgaard H, Hermansen MN, Buchvald F, Loland L, Halkjaer LB, Bonnelykke K, Brasholt M, Heltberg A, Vissing NH, Thorsen SV, et al. Childhood asthma after bacterial colonization of the airway in neonates. N Engl J Med. 2007;357:1487–1495. doi: 10.1056/NEJMoa052632. [DOI] [PubMed] [Google Scholar]
  • 52.Folsgaard NV, Schjorring S, Chawes BL, Rasmussen MA, Krogfelt KA, Brix S, Bisgaard H. Pathogenic bacteria colonizing the airways in asymptomatic neonates stimulates topical inflammatory mediator release. Am J Respir Crit Care Med. 2013;187:589–595. doi: 10.1164/rccm.201207-1297OC. [DOI] [PubMed] [Google Scholar]
  • 53.Krishnamoorthy N, Khare A, Oriss TB, Raundhal M, Morse C, Yarlagadda M, Wenzel SE, Moore ML, Peebles RS, Jr, Ray A, et al. Early infection with respiratory syncytial virus impairs regulatory T cell function and increases susceptibility to allergic asthma. Nat Med. 2012;18:1525–1530. doi: 10.1038/nm.2896. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Noverr MC, Huffnagle GB. The “microflora hypothesis” of allergic diseases. Clin Exp Allergy. 2005;35:1511–1520. doi: 10.1111/j.1365-2222.2005.02379.x. [DOI] [PubMed] [Google Scholar]
  • 55.Blaser MJ, Falkow S. What are the consequences of the disappearing human microbiota? Nat Rev Microbiol. 2009;7:887–894. doi: 10.1038/nrmicro2245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Russell SL, Gold MJ, Hartmann M, Willing BP, Thorson L, Wlodarska M, Gill N, Blanchet MR, Mohn WW, McNagny KM, et al. Early life antibiotic-driven changes in microbiota enhance susceptibility to allergic asthma. EMBO Rep. 2012;13:440–447. doi: 10.1038/embor.2012.32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Herbst T, Sichelstiel A, Schar C, Yadava K, Burki K, Cahenzli J, McCoy K, Marsland BJ, Harris NL. Dysregulation of allergic airway inflammation in the absence of microbial colonization. Am J Respir Crit Care Med. 2011;184:198–205. doi: 10.1164/rccm.201010-1574OC. [DOI] [PubMed] [Google Scholar]
  • 58.Hill DA, Siracusa MC, Abt MC, Kim BS, Kobuley D, Kubo M, Kambayashi T, Larosa DF, Renner ED, Orange JS, et al. Commensal bacteria–derived signals regulate basophil hematopoiesis and allergic inflammation. Nat Med. 2012;18:538–546. doi: 10.1038/nm.2657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Olszak T, An D, Zeissig S, Vera MP, Richter J, Franke A, Glickman JN, Siebert R, Baron RM, Kasper DL, et al. Microbial exposure during early life has persistent effects on natural killer T cell function. Science. 2012;336:489–493. doi: 10.1126/science.1219328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Renz H, Brandtzaeg P, Hornef M. The impact of perinatal immune development on mucosal homeostasis and chronic inflammation. Nat Rev Immunol. 2012;12:9–23. doi: 10.1038/nri3112. [DOI] [PubMed] [Google Scholar]
  • 61.Josefowicz SZ, Niec RE, Kim HY, Treuting P, Chinen T, Zheng Y, Umetsu DT, Rudensky AY. Extrathymically generated regulatory T cells control mucosal Th2 inflammation. Nature. 2012;482:395–399. doi: 10.1038/nature10772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Hicks LA, Taylor TH, Jr, Hunkler RJ. US outpatient antibiotic prescribing, 2010. N Engl J Med. 2013;368:1461–1462. doi: 10.1056/NEJMc1212055. [DOI] [PubMed] [Google Scholar]
  • 63.Wickens K, Pearce N, Crane J, Beasley R. Antibiotic use in early childhood and the development of asthma. Clin Exp Allergy. 1999;29:766–771. doi: 10.1046/j.1365-2222.1999.00536.x. [DOI] [PubMed] [Google Scholar]
  • 64.Arason VA, Kristinsson KG, Sigurdsson JA, Stefansdottir G, Molstad S, Gudmundsson S. Do antimicrobials increase the carriage rate of penicillin resistant pneumococci in children? Cross sectional prevalence study. BMJ. 1996;313:387–391. doi: 10.1136/bmj.313.7054.387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Harbarth S, Albrich W, Brun-Buisson C. Outpatient antibiotic use and prevalence of antibiotic-resistant pneumococci in France and Germany: a sociocultural perspective. Emerg Infect Dis. 2002;8:1460–1467. doi: 10.3201/eid0812.010533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Butaye P, Devriese LA, Haesebrouck F. Antimicrobial growth promoters used in animal feed: effects of less well known antibiotics on gram-positive bacteria. Clin Microbiol Rev. 2003;16:175–188. doi: 10.1128/CMR.16.2.175-188.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Ozawa E. Studies on growth promotion by antibiotics. I. Effects of chlortetracycline on growth. J Antibiot (Tokyo) 1955;8:205–211. [PubMed] [Google Scholar]
  • 68.Cho I, Yamanishi S, Cox L, Methe BA, Zavadil J, Li K, Gao Z, Mahana D, Raju K, Teitler I, et al. Antibiotics in early life alter the murine colonic microbiome and adiposity. Nature. 2012;488:621–626. doi: 10.1038/nature11400. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Annals of the American Thoracic Society are provided here courtesy of American Thoracic Society

RESOURCES