HHS Public Access Author manuscript Author Manuscript

J Immunol. Author manuscript; available in PMC 2017 June 15. Published in final edited form as: J Immunol. 2016 June 15; 196(12): 4839–4847. doi:10.4049/jimmunol.1600279.

The Lung Microbiome, Immunity and the Pathogenesis of Chronic Lung Disease1 David N. O’Dwyer*, Robert P. Dickson*, and Bethany B. Moore*,† *Department

of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan Health System, Ann Arbor, MI, USA

Author Manuscript

†Department

of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor,

MI, USA

Abstract

Author Manuscript

The development of culture-independent techniques for microbiological analysis has uncovered the previously unappreciated complexity of the bacterial microbiome at various anatomic sites. The microbiome of the lung has relatively less bacterial biomass when compared to the lower gastrointestinal tract yet displays considerable diversity. The composition of the lung microbiome is determined by elimination, immigration and relative growth within its communities. Chronic lung disease alters these factors. Many forms of chronic lung disease demonstrate exacerbations that drive disease progression and are poorly understood. Mounting evidence supports ways in which microbiota dysbiosis can influence host defense and immunity, and in turn may contribute to disease exacerbations. Thus, the key to understanding the pathogenesis of chronic lung disease may reside in deciphering the complex interactions between the host, pathogen and resident microbiota during stable disease and exacerbations. In this brief review we discuss new insights into these labyrinthine relationships.

The microbiome shapes immunity

Author Manuscript

The microbiome is defined as the “ecological community of commensal, symbiotic and pathogenic organisms that share our body space” (1). Most studies of host and microbiome interaction in the human have focused almost exclusively on bacteria, biotic factors and the host. These complex communities of microbiota that inhabit environments such as the lung, skin or gut are now appreciated for their role in maintaining organ, tissue and immune homeostasis. One striking example is the early observation that germ-free mice have absent/ impaired secondary lymphoid architecture with resulting loss of lymphoid cells (2). Additionally, commensal microbiota can have both systemic and site-specific autonomous immune effects. For example, Staphylococcus epidermidis colonization of the skin promotes CD4 cell IFN-γ production which protects against infection with the parasite Leishmania major. In contrast, colonization of the gut with S. epidermidis had no effect (3). In other

1This work was supported in part by NIH Grant HL115618 (to B.B.M), NIH Grant AI117229 (to B.B.M), NIH Grant HL130641 (to R.P.D) and UL1TR000433 (to R.P.D) Corresponding author: Bethany B. Moore, 4053 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, ; Email: [email protected], Office phone: 734-647-8378, Office fax: 734-615-2331

O’Dwyer et al.

Page 2

Author Manuscript Author Manuscript

situations, it is well established that alterations of the gut microbiome can influence immune responses at distal sites. Antibiotic treatment, which disrupts gut microbiota accompanied by increases in fungal colonization, can greatly exaggerate the allergic response to intranasal challenge with the mold spore Aspergillus fumigatus. With antibiotic treatment, mice showed increased levels of eosinophils, mast cells, IL-5, IL-13, IFN-γ, IgE, and mucussecreting cells (4). More recently, modulation of gut microbiota through the use of probiotics has been shown to increase the frequency of B cells expressing IgA in the colon and lymph nodes, likely secondary to increased lymph node T follicular helper (Tfh) cells and IL-23expressing dendritic cells (5), all changes that are likely to improve host defense at mucosal sites or response to vaccination. Conversely, antibiotic treatment can limit development of Tfh cells (6). The response of pathogen recognition receptors (PRRs) of the innate immune response in the lung is well described. We know that defective components of the innate response can predispose to overwhelming infection and mortality and in some cases reduce injury from pathogens (7–9). The relationship between resident microbiota and this flagship innate response and the subsequent adaptive immune response in the lung is poorly understood. With this backdrop, we have chosen to explore what is known about the potential role of the microbiota (both gut and lung) and host interaction in regulating the pathogenesis of several important lung diseases. Origins of lung microbiome and debunking lung sterility

Author Manuscript

The lung is an organ constantly exposed to microbiota either through inhalation or subclinical micro aspiration from birth. Historically, medical texts allude to a sterile lung environment, and this dogma has persisted in contemporary medicine. In the last decade, a revolution of sorts has taken place in our understanding of how the lung and microbiota interact and exist. This revolution stems from new knowledge that the lung is not sterile (10) and in fact, harbors an abundance of diverse interacting microbiota. As mentioned above, the gut microbiome modulates host mucosal defense (11, 12); however, there is a paucity of information regarding the potential role of lung microbiota to regulate immunity and homeostasis.

Author Manuscript

The lung is not sterile, contrary to centuries of dogma asserting the same. Throughout the 1900’s this inference was re-enforced by respiratory culture-based protocols that sought only to identify clinically significant pathogens and by a spurious conclusion that upper respiratory tract microbes cultured from the lung represented contamination (10). The lung is a warm environment exposed to 7000 liters of diverse microbe-ridden air every day (13). Microbes adapt and exist in hostile environments like polar ice sheets of Antarctica and hot sulfur springs of Japan, yet the belief the human lung existed in a sterile state was “cultivated” in the medical literature for decades (14, 15). The upper respiratory tract and oropharynx, where microbes are found in abundance (16), is in direct communication with the lung, and subclinical aspiration of oropharyngeal contents occurs universally in humans (17, 18). Thus, the lung is subject to a constant level of microbe immigration and elimination through host mucosal defense and mucociliary clearance. Modern studies employing sequence-based bacterial identification techniques support the presence of a consistentlydetected diverse bacterial signal in the lung of healthy humans (19–22). Therefore, the notion of lung sterility has been debunked.

J Immunol. Author manuscript; available in PMC 2017 June 15.

O’Dwyer et al.

Page 3

Author Manuscript

This new understanding was animated by the discovery of culture-independent techniques for bacterial identification. The most commonly used approach involves high-throughput sequencing of amplicons of the 16s rRNA gene, a highly conserved locus in the bacterial genome. Sequences are then categorized and classified according to publically available prior knowledge taxonomic databases to allow for measures of total and relative abundance and diversity. The first application of culture-independent techniques was undertaken in a cohort of healthy controls and asthma patients. Hilty et al reported healthy airways contain bacteria similar, but distinct, from the upper respiratory tract and airways of asthma patients were enriched with Proteobacteria phylum (20).

Author Manuscript

Sampling the lung for microbiome sequencing is technically challenging given the relatively low biomass. Furthermore, sampling lower airways by bronchoscopy requires passage of the instrument through the oral or nasal route. This course allows for a theoretical risk of pharyngeal contamination of samples. Importantly, the mouth and nose microbiota are markedly different and studies have not identified any detectable influence on the reported microbiota based on scope insertion site (22, 23). In addition, if pharyngeal carryover from bronchoscope insertion site was heavily influencing the reported microbiota, then serial dilutions of samples should result in dilution of bacterial communities and signal. Several studies have established that this is not the case (19, 24). The evidence thus supports minimal contamination from pharyngeal sources acquired by bronchoscopic methods. Methodology and limitations in studies of the microbiota

Author Manuscript Author Manuscript

Accurate and relevant studies of the microbiome require consideration of several principles. Representative samples must be acquired from the distal airways, and as discussed above, the possibility of contamination from other niche microbiota must be addressed. Furthermore, contamination can occur during sample processing; use of “no template” controls is essential in low-biomass studies to assess for the effects of reagent contamination. The extraction of nucleic acid requires lysis of species, some of whom are more susceptible to cell disruption than others. This discrepancy can lead to overrepresentation of some species over others. Further steps that may alter the accuracy and reliability of acquired data include the generation of appropriate PCR primers, data normalization, the choice of reference database, and divergent measures of diversity (25). The use of 16s rRNA sequencing remains a pillar step in the sample processing, however, 16s rRNA sequencing may not be able to differentiate between species with varying immunogenicity and pathogenicity (26). The microbiome is subject to a number of factors that are known to change its composition including age, diet, ethnicity and study design in humans requires careful management of these potential influences (27–29). However, in tandem, researchers are developing novel and ingenious methods to limit any possible error in microbiome studies. For example, as an alternate process to 16s rRNA sequencing and primer choice, metagenomic data is being generated through the sequencing of all DNA from a sample (shotgun sequencing) which may be even more informative (30). Culturebased techniques remain highly relevant as complements to culture-independent techniques; in determination of viability, in speciation, and in microbial phenotyping.

J Immunol. Author manuscript; available in PMC 2017 June 15.

O’Dwyer et al.

Page 4

Microbiome development and composition in healthy lung

Author Manuscript Author Manuscript Author Manuscript

The human microbiota inhabits several organs and is primarily colonized by 6 phyla: Firmicutes, Bacteroidetes, Proteobacteria, Actinobacteria, Fusobacteria and Cyanobacteria (31–34). Murine studies have demonstrated bacterial load in the lungs increases over the first 2 weeks of life, and the phyla of organisms found in the lung shifted from Gammaproteobacteria and Firmicutes towards Bacteriodetes (35). Such developmental changes in the microbiota were associated with accumulation of a PD-L1-dependent T regulatory cell population that could promote tolerance to allergen challenge (35). These data suggest acquisition of a lung microbiome is an important early life event necessary to protect the lung from injurious responses to inhaled antigens. In humans, studies have largely focused on gut microbiota and shown that newborns acquire microbiota that resembles their mother quickly and in a manner specific to the method of delivery. Dominguez-Bello et al reported infants born by vaginal delivery acquired bacterial communities resembling their mother’s vaginal microbiota, dominated by Lactobacillus, Prevotella, or Sneathia species. Infants born by cesarean section acquired skin-predominant Staphylococcus, Corynebacterium, and Propionibacterium species (36). These communities were undifferentiated across multiple body habitats in the infants, in contrast to the diverse communities evident in the mothers. No studies to date have examined the dynamic changes that may occur in the lower respiratory tract microbiota as childhood progresses. However, it is likely from studies of the upper respiratory tract and intestinal microbiota that these bacterial communities are dynamic (37, 38). There is relatively low bacterial biomass in the human lung. Bacterial loads from bronchoalveolar lavage have reported ranges from 4.5 to 8.25 log copies per/ml (39, 40). Further analysis of lung tissue samples demonstrates some 10 – 100 bacterial cells per 1000 human cells (41). The healthy lung has been studied using culture-independent techniques and the predominant phyla are Bacteroidetes and Firmicutes (21, 24). While individuals exhibit some spatial variation in the microbiota of their respiratory tract, intra-subject variation is significantly less than that of inter-subject variation (39). Dynamic changes in lung microbiome in health and disease

Author Manuscript

The lung is a relatively low nutrient resource compared to the intestinal tract for supporting microbiome development. Furthermore, physiological conditions are regionally variable even in healthy lungs. Conditions that affect bacterial proliferation include: oxygen tension, blood flow, local pH, temperature, effector inflammatory cell disposition, and epithelial cell architecture (42, 43). Coupled to this variable biogeography of the lung microbiome are the factors that influence microbe immigration and elimination from the lower respiratory tract. Taken together, these factors determine the dynamic change of the microbial ecosystem of the lung. Lung disease alters the population dynamics through effects on immigration/elimination and the local conditions of the microbial ecosystem of the lung (Table 1). Chronic lung disease in many forms, results in heterogeneous architectural distortion of the lung: upper lobe predominant destruction of the terminal bronchioles in emphysema and lower lobe predominant distortion of the parenchyma by honeycombing in Idiopathic Pulmonary Fibrosis (IPF) (44). Changes in the viscosity of the mucus and pH occur in Cystic Fibrosis

J Immunol. Author manuscript; available in PMC 2017 June 15.

O’Dwyer et al.

Page 5

Author Manuscript

(CF) (45). The resultant changes in oxygen tension, ventilation, perfusion, inflammatory cells and other local factors exert pressure on population dynamics. Immigration of microbiota from the upper respiratory to the lower respiratory tract is primarily promoted by subclinical aspiration which occurs in both health and disease and overt clinical infection occurs when local defense is blunted or overwhelmed (17, 18). Chronic lung disease is commonly associated with gastroesophageal reflux which may result in elevated volumes of micro aspiration (46, 47). Elimination is achieved by cough and mucociliary clearance. Host inflammatory cells are responsible for eradication of pathogens and the type and number of effector cells are associated with certain features of the microbiome. In a comparison of inflammatory cells and microbiota detected in bronchoalveolar lavage (BAL) fluid, Segal et al demonstrated increased community abundance of Prevotella and Veillonella (common anaerobic oral commensals) associated with higher levels of lymphocyte and neutrophil inflammation (24). Therefore the lung microbiome has a potential role to play in the pathogenesis of chronic lung disease through both the ability of lung microbiota to modulate local inflammatory responses and the influence of chronic lung diseases on the lung microbiome in turn.

Author Manuscript Author Manuscript

Chronic lung diseases include asthma, Chronic Obstructive Pulmonary Disease (COPD), CF and IPF. Interestingly, these diseases are all characterized by natural histories that are punctuated by periods of acute exacerbations. Exacerbations are characterized by acute worsening of pulmonary symptoms and a decline in pulmonary function. Such exacerbations are responsible for significant mortality and morbidity. The onset of exacerbation may herald accelerated disease progression and many patients fail to return to their previous functional and physiological baseline (48). Studies of these events may reveal key data that re-animate our current understanding of the pathogenesis of chronic lung disease, and it is likely that these exacerbations are accompanied or induced by microbiota dysbiosis. COPD

Author Manuscript

The relationship between COPD exacerbation and acute bacterial infection of the airway remains disputed. Potential pathogens cultured from sputum during COPD exacerbations are less frequently cultured during periods of clinical stability (49). Sethi et al identified similar culture densities of Haemophilus influenza, and lower densities of Moraxella catarrhalis and Streptococcus pneumoniae in sputum collected during acute exacerbations compared to samples during clinical stability (50). The use of antibiotic therapy in COPD exacerbations also lacks clarity. Recent work reported a clear role in reducing the rate of treatment failure for severe disease in hospitalized patients, but the role is unclear for mild to moderate disease (51). Culture-independent techniques have identified a diverse and abundant pulmonary microbiota in exacerbations from a variety of sampling types (41, 52–54) and exacerbations are definitively associated with changes in respiratory microbiota and airway inflammation. Millares et al analyzed sputum from COPD patients during exacerbations with paired sampling from periods of clinical stability and found increases in the relative abundance of bacteria associated with exacerbations, namely Haemophilus, Pseudomonas and Moraxella (52). Huang et al reported an alteration in community content towards the Proteobacteria phylum during COPD exacerbations, including non-typical COPD pathogens (53). Additionally, the influence of viral exposure may trigger COPD exacerbations (55) but

J Immunol. Author manuscript; available in PMC 2017 June 15.

O’Dwyer et al.

Page 6

Author Manuscript

the relationship between viral infection, microbiome composition and host defense is poorly understood. Patients experimentally infected with rhinovirus develop clinical features of COPD exacerbation and culprit viruses have been isolated in respiratory samples from 36– 56% of patients with exacerbations versus 6–19% of patients at clinical baseline (56–59). Interestingly, Molyneaux et al compared sputum in COPD patients at baseline and during exacerbations and noted that sputum acquired post-viral exposure demonstrated a shift towards the Proteobacteria phylum (60), a potential explanation for the increased presence of Pseudomonas spp noted in COPD exacerbations (52). Sequencing-based studies of tissue from COPD patients have demonstrated an increase in the Firmicutes community in severe disease (GOLD stage 4) attributable to an increase in the Lactobacillus genus (41). Phagocytosis of Lactobacillus spp by human macrophages reduces the effects of cigarette smoke-related inflammation, potentially suggesting these species are beneficial modifiers of smoking-related lung disease (61). Animal models of RSV infection have demonstrated that the anti-viral response within the lung mucosa can be modulated by the administration of Lactobacillus rhamnosus species prior to infection (62). Thus, changes in the microbiome may represent an adaptation to try to protect the lung from respiratory viral infection. However, we speculate that, as suggested above, once pathogenic infection does occur, these beneficial changes may be lost.

Author Manuscript

Asthma

Author Manuscript Author Manuscript

Studies of airway microbiota in asthma have established that composition is altered when compared to controls. Hilty et al identified asthma patients with more frequent Proteobacteria (particularly Haemophilus) in the bronchial tree compared to controls. The authors also noted a decrease in Bacteroidetes, especially Prevotella species, in asthmatic airways (20). Studies of asthma severity identified similar altered microbiomes with a predominance of Proteobacteria, and the observation that the airway microbiome of asthmatic patients was more diverse that non-asthmatic controls (63). Huang et al reported an association between bronchial hyper-responsiveness and community diversity and composition, secondary to an increased abundance of Proteobacteria (64). Bronchial hyperresponsiveness is accentuated during exacerbation and is an accurate predictor of future exacerbations (65). Alterations to the microbiome appear similar in both mild and severe disease and are specifically associated with features of the disease. No studies have analyzed the airway microbiome in asthma exacerbations. However, an estimated 80% of asthma exacerbations are associated with viral infection (66, 67). The host microbiome interaction may be crucial in the development of asthma. Ege et al have demonstrated that children with broad microbial exposures (i.e. traditional farming) were protected from asthma and atopy in childhood (68). Further studies reported an association with high fiber diet and a reduced risk of asthma (69). The proposed mechanism was related, in part, to an altered immune response. Mice fed a low fiber diet exhibited reduced levels of short chain fatty acids (SCFA) with increased allergic inflammation whereas mice with a high fiber diet had elevated levels of SCFA and were protected against allergic inflammation. SCFA propionate treatment of mice resulted in the generation of macrophages and dendritic cells with enhanced phagocytic properties but an attenuated capacity to initiate T helper type 2 response, a crucial component of allergic inflammation (69). Furthermore, studies have suggested resident microbiota may promote Th17-dependent neutrophil inflammation in a

J Immunol. Author manuscript; available in PMC 2017 June 15.

O’Dwyer et al.

Page 7

Author Manuscript

murine model of ovalbumin-induced asthma (70). Similarly, an experimental model of allergic airway inflammation is exacerbated by the administration of antibiotics during early life. This correlated with a reduction in microbial load and diversity (71). IPF

Author Manuscript

IPF is a chronic fatal remodeling disease of the lung parenchyma of unknown etiology (72). The natural history of IPF is characterized by exacerbations that contribute greatly to disease related morbidity and mortality. Recent work has highlighted a potential role for both viral and bacterial infection in the pathogenesis of IPF (73–77). Unlike with asthma or COPD, the current definition of acute exacerbations of IPF excludes active infectious pathogens (78). Disease progression in IPF is characterized by an alteration in the microbiome with a relative increased abundance of Streptococcus and Staphylococcus taxonomic groups (79). This has particular relevance given recent work identifying pneumolysin, a Pneumococcus produced toxin, that mediates fibrotic progression in animal models via injury of the alveolar epithelium (73). Further study by Molyneaux et al describes an increased bacterial burden in the BAL fluid of IPF patients compared to controls using culture-independent techniques. These communities were enriched with Haemophilus, Streptococcus, Neisseria and Veillonella (76). The greater the bacterial burden in these patients, the greater the independent association with IPF disease progression. Recent trials of trimethoprimsulphamethtoxazole have demonstrated benefit with improved Medical Research Council dyspnea scores, quality of life and even all-cause mortality (HR 0.21; 95% CI 0.06 – 0.78: p=0.02)(80). This further supports a role for bacterial burden in disease progression. Aspects of host defense and innate immunity also have putative roles in IPF disease progression (81– 86). We have previously suggested defective toll-like receptor (TLR) 3 signaling promotes IPF disease progression (81). The L412F polymorphism (rs3775291) of TLR3 results in a functional defect in primary lung fibroblasts from IPF patients. This defect leads to aberrant inflammation and blunted interferon responses to TLR3 activation by synthetic dsRNA [and likely pathogen-associated molecular patterns (PAMPs) although this was not directly examined]. Genotyping studies of two independent cohorts of IPF patients confirmed an association between this polymorphism with increased mortality and functional decline. However, the interaction between the pulmonary microbiome, host defense, acute infection and IPF disease progression remains unclear. Ultimately, the questions of whether chronic lung disease promotes microbiome alterations or microbiome changes modify chronic lung disease remains to be answered.

Author Manuscript CF

Author Manuscript

The manifestations of CF in the lung involve the progressive development of bronchiectasis and obstructive lung disease. Central to disease progression are exacerbations of CF bronchiectasis which are responsible for significant mortality, morbidity and accelerated disease progression (87). Exacerbations of disease are attributed to infection by specific pathogens that are cultured from sputum during exacerbations and clinical stability. These pathogens commonly include Staphylococcus aureus and Pseudomonas aeruginosa (88). The evidence to support the use of antibiotics directed against these pathogens is sparse. The bacterial density of sputum is not altered during CF exacerbations when antibiotics are administered (89). Clinical trials have not reported an association between the clinical J Immunol. Author manuscript; available in PMC 2017 June 15.

O’Dwyer et al.

Page 8

Author Manuscript Author Manuscript

response during antibiotic therapy and the in vitro susceptibility of the cultured bacteria to the administered antibiotics (90, 91). Indeed, culture independent analysis has primed a revision of our long held understanding of the bacterial pathogenesis of CF lung disease. Studies have consistently reported that CF exacerbations are not associated with increased bacterial density or attenuated diversity (92–94). However, evidence would support a loss of diversity with increasing age and disease severity, which was strongly associated with cumulative antibiotic exposure (95, 96). The emergence of new pathogens may have implications for our understanding of the microbiome and lung disease interaction in CF. Non-tuberculosis mycobacterium (NTM), in particular Abscessus, are associated with increased mortality and morbidity in CF (97–99). Guidelines for the management of Abscessus have been published and long term treatment with broad spectrum antibiotics is required in these cases (100). The consequences for the lung microbiome for this treatment remain unknown. This is of particular relevance given the limited treatment benefit for Abscessus infection in CF (100). The relationship between the microbiome, antibiotic exposure, exacerbation and ultimate disease progression will thus require further careful study in CF. Evidence and implications for a gut-lung axis and the regulation of host defense for chronic lung disease exacerbations

Author Manuscript Author Manuscript

Exactly how microbiota may regulate innate immunity in health and disease is an area of active investigation, and very little is known about how the lung microbiota may specifically regulate lung immunity or the development of bronchial-associated lymphoid tissue. There is growing appreciation for the fact that the gut commensal microbiota is an important regulator of the innate immune system (101, 102). The bacterial biomass of the intestine dwarfs the relative mass seen in the lung (103). In healthy adults the intestine microbiota consists predominantly of 3 phyla: Bacteroides, Prevotella and Ruminococcus (104). There is evidence to support a crucial early period during life where intestinal microbiome development is important for the regulation of an appropriate immune response in the lung. CF and asthma are examples of chronic lung disease where disease course and susceptibility are influenced by shifts in the composition of the gut microbiota (105, 106). Furthermore, in the absence of normal gut biota, the host is more susceptible to pulmonary infections including Listeria monocytogenes (107), Klebsiella pneumoniae (108) and viruses (109). This raises the interesting possibility that exacerbations of chronic lung disease may arise from impaired innate and adaptive immune function secondary to alterations in the host gut microbiota. As mentioned above, patients with progressive IPF show evidence of enhanced burden of Streptococcus and Staphylococcus species in the lung, and previous studies have shown that the ability of neutrophils from microbiota-depleted mice to kill S. pneumoniae and S. aureus are reduced (110). Currently it is not known whether the accumulation of these species in the lung correlates with loss of gut microbial communities during IPF disease progression, but this is an interesting area for future study (Figure 1). Support for such a concept comes from recent work showing that bacterial activation of NLR receptors in the gut leads to enhanced production of reactive oxygen species within alveolar macrophages, the sentinel innate immune cell within the lung (101); implying that conditions associated with loss of gut bacterial homeostasis (e.g. antibiotic use) could provide a window of opportunity for lung immunity to be impaired. In COPD, exacerbations can occur due to

J Immunol. Author manuscript; available in PMC 2017 June 15.

O’Dwyer et al.

Page 9

Author Manuscript

viral infections (60, 111), and resulting pathogenesis could be the result of dysbiosis leading to altered airway microbiota and disproportionate inflammation. While loss of gut commensal signaling may impair lung innate immunity in this disorder, cigarette smoke directly and indirectly contributes to impaired innate immunity in the lung via alterations in ciliary function, mucus, innate immune cell phagocytosis and via direct enhancement of bacterial virulence (e.g. enhanced biofilm formation) [reviewed in (112)]. These changes could impact the ability of respiratory pathogens to exacerbate COPD.

Author Manuscript

Given the propensity for viruses to precipitate lung disease exacerbations, it is interesting to note the potential impact of respiratory viral infection on the intestinal microbiota. Wang et al reported that influenza infection may lead to alterations in intestinal microbiota with a reduction in Lactobacillus and Lactococci and an outgrowth of Enterbacteriaceae. As noted above, this may lead to a loss of beneficial microbiota for smoking-related disease. The authors demonstrated that these shifts in intestinal microbiota were not secondary to lytic influenza gut infection. This injury was mediated by Th17 cells and IL-17 neutralization resulted in attenuated injury (113). In addition, antibiotic-mediated depletion of intestinal microbiota led to attenuated intestinal injury. Interestingly, this study also highlighted the importance of an effector T cell that arose in the lung post-infection then migrated to the small intestine to provide IFN-γ to alter the gut microbiome. Ultimately the alterations in the gut microbiota stimulated epithelial-derived IL-15 to promote the Th17 response. It is possible that IL-17 responses arising in the gut may further impact lung disease (114). IL-17 is involved in the elimination of certain pathogens (115) and is implicated in the pathogenesis of several pulmonary pathologies including asthma, Sarcoidosis, Obliterative Bronchiolitis, CF and bone marrow transplant related pneumonitis(116–120).

Author Manuscript Author Manuscript

IL-17 may also play a central role in the dynamic change that occurs within the pulmonary microbiota of COPD. Yadava et al report the impact of experimental change on the lung microbiota in an emphysema animal model. Specific pathogen free and axenic mice were challenged with lipopolysaccharide (LPS)/elastase for 4 weeks. Microbiota diversity and abundance was decreased in the LPS/elastase model with an abundance of Pseudomonas, Lactobacillus and a depletion of Prevotella. Loss of bacterial load was associated with attenuated IL-17 production. The intranasal application of microbiota enriched fluid to axenic mice enhanced IL-17 production. The neutralization of IL-17 in mice harboring microbiota led to dampened inflammation and reduced disease burden (121). Several studies have implicated IL-17 in hepatic fibrosis, and certain experimental models of pulmonary fibrosis are IL-17A dependent (120, 122, 123). Furthermore, studies examining the development of intestinal fibrosis have reported an association with alterations in the microbiota and Th17 responses (124). The intestine is a known source of Th17 cells through binding of segmented filamentous bacteria (SFB) to intestinal epithelial cells (125). The case may be similar in the lung. Gauguet et al have demonstrated that intestinal SFB have the ability to promote pulmonary innate immunity through the induction of IL-17 to provide resistance to S. aureus pneumonia in animal models (126). This constitutes further evidence to support a gut-lung microbiome axis that may be pivotal in modulating the innate immune response of the lung. However, direct evidence of a gut-lung axis promoting exacerbation of chronic lung disease is limited to experimental data to date and requires further study.

J Immunol. Author manuscript; available in PMC 2017 June 15.

O’Dwyer et al.

Page 10

Author Manuscript

Conclusions

Author Manuscript

In conclusion, growing evidence suggests alterations in the lung and/or gut microbiota characterize chronic lung diseases and may allow for exacerbations caused by endogenous microbiota alterations or susceptibility to new infection (Figure 1). We speculate that impairment in lung innate immunity caused by microbial dysbiosis may promote susceptibility of the host to infections that can exacerbate chronic lung diseases. Furthermore, shifts in cytokine profiles mediated by changes in the microbiota may also promote epithelial injury and fibrotic outcomes. Overall, there appears to be a vital crosstalk between the gut and lung mucosa and the microbial communities within. The device through which this cross talk may be achieved remains unknown. Possible instruments include translocation of gut microbiota (including potential pathogens) through blood or lymphatics, modulation of circulating or lung-resident effector immune and inflammatory cells or alterations in systemic cytokine profiles. These results highlight the need for careful future human studies that will characterize not only the lung, but also the gut microbiota during periods of disease stability vs. exacerbations. In addition, murine models may allow us to interrogate the PRRs and cytokine signaling pathways that promote exacerbations.

References

Author Manuscript Author Manuscript

1. Lederberg J, McCray AT. ‘Ome sweet ‘omics - A genealogical treasury of words. Scientist. 2001; 15:8–8. 2. Bauer H, Horowitz RE, Levenson SM, Popper H. The response of the lymphatic tissue to the microbial flora. Studies on germfree mice. Am J Pathol. 1963; 42:471–483. [PubMed: 13966929] 3. Naik S, Bouladoux N, Wilhelm C, Molloy MJ, Salcedo R, Kastenmuller W, Deming C, Quinones M, Koo L, Conlan S, Spencer S, Hall JA, Dzutsev A, Kong H, Campbell DJ, Trinchieri G, Segre JA, Belkaid Y. Compartmentalized control of skin immunity by resident commensals. Science. 2012; 337:1115–1119. [PubMed: 22837383] 4. 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. [PubMed: 15321991] 5. Manuzak JA, Hensley-McBain T, Zevin AS, Miller C, Cubas R, Agricola B, Gile J, Richert-Spuhler L, Patilea G, Estes JD, Langevin S, Reeves RK, Haddad EK, Klatt NR. Enhancement of Microbiota in Healthy Macaques Results in Beneficial Modulation of Mucosal and Systemic Immune Function. J Immunol. 2016 6. Block KE, Zheng Z, Dent AL, Kee BL, Huang H. Gut Microbiota Regulates K/BxN Autoimmune Arthritis through Follicular Helper T but Not Th17 Cells. J Immunol. 2016; 196:1550–1557. [PubMed: 26783341] 7. Ishizaki Y, Takemoto M, Kira R, Kusuhara K, Torisu H, Sakai Y, Sanefuji M, Yukaya N, Hara T. Association of toll-like receptor 3 gene polymorphism with subacute sclerosing panencephalitis. J Neurovirol. 2008; 14:486–491. [PubMed: 19016379] 8. Netea MG, Wijmenga C, O’Neill LA. Genetic variation in Toll-like receptors and disease susceptibility. Nat Immunol. 2012; 13:535–542. [PubMed: 22610250] 9. Khor CC, Chapman SJ, Vannberg FO, Dunne A, Murphy C, Ling EY, Frodsham AJ, Walley AJ, Kyrieleis O, Khan A, Aucan C, Segal S, Moore CE, Knox K, Campbell SJ, Lienhardt C, Scott A, Aaby P, Sow OY, Grignani RT, Sillah J, Sirugo G, Peshu N, Williams TN, Maitland K, Davies RJ, Kwiatkowski DP, Day NP, Yala D, Crook DW, Marsh K, Berkley JA, O’Neill LA, Hill AV. A Mal functional variant is associated with protection against invasive pneumococcal disease, bacteremia, malaria and tuberculosis. Nat Genet. 2007; 39:523–528. [PubMed: 17322885] 10. Dickson RP, Erb-Downward JR, Martinez FJ, Huffnagle GB. The Microbiome and the Respiratory Tract. Annu Rev Physiol. 2016; 78:481–504. [PubMed: 26527186]

J Immunol. Author manuscript; available in PMC 2017 June 15.

O’Dwyer et al.

Page 11

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

11. Caballero S, Pamer EG. Microbiota-mediated inflammation and antimicrobial defense in the intestine. Annu Rev Immunol. 2015; 33:227–256. [PubMed: 25581310] 12. Martin C, Burgel PR, Lepage P, Andrejak C, de Blic J, Bourdin A, Brouard J, Chanez P, Dalphin JC, Deslee G, Deschildre A, Gosset P, Touqui L, Dusser D. Host-microbe interactions in distal airways: relevance to chronic airway diseases. Eur Respir Rev. 2015; 24:78–91. [PubMed: 25726559] 13. Winslow CE. A New Method of Enumerating Bacteria in Air. Science. 1908; 28:28–31. [PubMed: 17834254] 14. Thomas DN, Dieckmann GS. Antarctic Sea ice--a habitat for extremophiles. Science. 2002; 295:641–644. [PubMed: 11809961] 15. Yamamoto H, Hiraishi A, Kato K, Chiura HX, Maki Y, Shimizu A. Phylogenetic evidence for the existence of novel thermophilic bacteria in hot spring sulfur-turf microbial mats in Japan. Appl Environ Microbiol. 1998; 64:1680–1687. [PubMed: 9572936] 16. Wilson MT, Hamilos DL. The nasal and sinus microbiome in health and disease. Curr Allergy Asthma Rep. 2014; 14:485. [PubMed: 25342392] 17. Gleeson K, Eggli DF, Maxwell SL. Quantitative aspiration during sleep in normal subjects. Chest. 1997; 111:1266–1272. [PubMed: 9149581] 18. Huxley EJ, Viroslav J, Gray WR, Pierce AK. Pharyngeal aspiration in normal adults and patients with depressed consciousness. Am J Med. 1978; 64:564–568. [PubMed: 645722] 19. 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] 20. Hilty M, Burke C, Pedro H, Cardenas P, Bush A, Bossley C, Davies J, Ervine A, Poulter L, Pachter L, Moffatt MF, Cookson WO. Disordered microbial communities in asthmatic airways. PLoS One. 2010; 5:e8578. [PubMed: 20052417] 21. Morris A, Beck JM, Schloss PD, Campbell TB, Crothers K, Curtis JL, Flores SC, Fontenot AP, Ghedin E, Huang L, Jablonski K, Kleerup E, Lynch SV, Sodergren E, Twigg H, Young VB, Bassis CM, Venkataraman A, Schmidt TM, Weinstock GM, Lung HIVMP. Comparison of the respiratory microbiome in healthy nonsmokers and smokers. Am J Respir Crit Care Med. 2013; 187:1067– 1075. [PubMed: 23491408] 22. Bassis CM, Erb-Downward JR, Dickson RP, Freeman CM, Schmidt TM, Young VB, Beck JM, Curtis JL, Huffnagle GB. Analysis of the upper respiratory tract microbiotas as the source of the lung and gastric microbiotas in healthy individuals. MBio. 2015; 6:e00037. [PubMed: 25736890] 23. Dickson RP, Erb-Downward JR, Freeman CM, Walker N, Scales BS, Beck JM, Martinez FJ, Curtis JL, Lama VN, Huffnagle GB. Changes in the lung microbiome following lung transplantation include the emergence of two distinct Pseudomonas species with distinct clinical associations. PLoS One. 2014; 9:e97214. [PubMed: 24831685] 24. Segal LN, Alekseyenko AV, Clemente JC, Kulkarni R, Wu B, Gao Z, Chen H, Berger KI, Goldring RM, Rom WN, Blaser MJ, Weiden MD. Enrichment of lung microbiome with supraglottic taxa is associated with increased pulmonary inflammation. Microbiome. 2013; 1:19. [PubMed: 24450871] 25. Rogers GB, Shaw D, Marsh RL, Carroll MP, Serisier DJ, Bruce KD. Respiratory microbiota: addressing clinical questions, informing clinical practice. Thorax. 2015; 70:74–81. [PubMed: 25035125] 26. Zhu B, Xiao D, Zhang H, Zhang Y, Gao Y, Xu L, Lv J, Wang Y, Zhang J, Shao Z. MALDI-TOF MS distinctly differentiates nontypable Haemophilus influenzae from Haemophilus haemolyticus. PLoS One. 2013; 8:e56139. [PubMed: 23457514] 27. O’Toole PW I, Jeffery B. Gut microbiota and aging. Science. 2015; 350:1214–1215. [PubMed: 26785481] 28. David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, Ling AV, Devlin AS, Varma Y, Fischbach MA, Biddinger SB, Dutton RJ, Turnbaugh PJ. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014; 505:559–563. [PubMed: 24336217]

J Immunol. Author manuscript; available in PMC 2017 June 15.

O’Dwyer et al.

Page 12

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

29. Mason MR, Nagaraja HN, Camerlengo T, Joshi V, Kumar PS. Deep sequencing identifies ethnicity-specific bacterial signatures in the oral microbiome. PLoS One. 2013; 8:e77287. [PubMed: 24194878] 30. Shah N, Tang H, Doak TG, Ye Y. Comparing bacterial communities inferred from 16S rRNA gene sequencing and shotgun metagenomics. Pac Symp Biocomput. 2011:165–176. [PubMed: 21121044] 31. Eckburg PB, Bik EM, Bernstein CN, Purdom E, Dethlefsen L, Sargent M, Gill SR, Nelson KE, Relman DA. Diversity of the human intestinal microbial flora. Science. 2005; 308:1635–1638. [PubMed: 15831718] 32. Grice EA, Segre JA. The skin microbiome. Nat Rev Microbiol. 2011; 9:244–253. [PubMed: 21407241] 33. Frank DN, Feazel LM, Bessesen MT, Price CS, Janoff EN, Pace NR. The human nasal microbiota and Staphylococcus aureus carriage. PLoS One. 2010; 5:e10598. [PubMed: 20498722] 34. Kim TK, Thomas SM, Ho M, Sharma S, Reich CI, Frank JA, Yeater KM, Biggs DR, Nakamura N, Stumpf R, Leigh SR, Tapping RI, Blanke SR, Slauch JM, Gaskins HR, Weisbaum JS, Olsen GJ, Hoyer LL, Wilson BA. Heterogeneity of vaginal microbial communities within individuals. J Clin Microbiol. 2009; 47:1181–1189. [PubMed: 19158255] 35. Gollwitzer ES, Saglani S, Trompette A, Yadava K, Sherburn R, McCoy KD, Nicod LP, Lloyd CM, Marsland BJ. Lung microbiota promotes tolerance to allergens in neonates via PD-L1. Nat Med. 2014; 20:642–647. [PubMed: 24813249] 36. 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 U S A. 2010; 107:11971–11975. [PubMed: 20566857] 37. Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG, Contreras M, Magris M, Hidalgo G, Baldassano RN, Anokhin AP, Heath AC, Warner B, Reeder J, Kuczynski J, Caporaso JG, Lozupone CA, Lauber C, Clemente JC, Knights D, Knight R, Gordon JI. Human gut microbiome viewed across age and geography. Nature. 2012; 486:222–227. [PubMed: 22699611] 38. Biesbroek G, Tsivtsivadze E, Sanders EA, Montijn R, Veenhoven RH, Keijser BJ, Bogaert D. Early respiratory microbiota composition determines bacterial succession patterns and respiratory health in children. Am J Respir Crit Care Med. 2014; 190:1283–1292. [PubMed: 25329446] 39. Erb-Downward JR, Thompson DL, Han MK, Freeman CM, McCloskey L, Schmidt LA, Young VB, Toews GB, Curtis JL, Sundaram B, Martinez FJ, Huffnagle GB. Analysis of the lung microbiome in the “healthy” smoker and in COPD. PLoS One. 2011; 6:e16384. [PubMed: 21364979] 40. Charlson ES, Diamond JM, Bittinger K, Fitzgerald AS, Yadav A, Haas AR, Bushman FD, Collman RG. Lung-enriched organisms and aberrant bacterial and fungal respiratory microbiota after lung transplant. Am J Respir Crit Care Med. 2012; 186:536–545. [PubMed: 22798321] 41. 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] 42. Ingenito EP, Solway J, McFadden ER Jr, Pichurko B, Bowman HF, Michaels D, Drazen JM. Indirect assessment of mucosal surface temperatures in the airways: theory and tests. J Appl Physiol (1985). 1987; 63:2075–2083. [PubMed: 3693240] 43. West JB. Regional differences in the lung. Chest. 1978; 74:426–437. [PubMed: 699656] 44. Nishimura K, Kitaichi M, Izumi T, Nagai S, Kanaoka M, Itoh H. Usual interstitial pneumonia: histologic correlation with high-resolution CT. Radiology. 1992; 182:337–342. [PubMed: 1732946] 45. Collawn JF, Matalon S. CFTR and lung homeostasis. Am J Physiol Lung Cell Mol Physiol. 2014; 307:L917–923. [PubMed: 25381027] 46. Raghu G, Freudenberger TD, Yang S, Curtis JR, Spada C, Hayes J, Sillery JK, Pope CE 2nd, Pellegrini CA. High prevalence of abnormal acid gastro-oesophageal reflux in idiopathic pulmonary fibrosis. Eur Respir J. 2006; 27:136–142. [PubMed: 16387946] 47. Robinson NB, DiMango E. Prevalence of gastroesophageal reflux in cystic fibrosis and implications for lung disease. Ann Am Thorac Soc. 2014; 11:964–968. [PubMed: 24964289]

J Immunol. Author manuscript; available in PMC 2017 June 15.

O’Dwyer et al.

Page 13

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

48. Ryerson CJ, Cottin V, Brown KK, Collard HR. Acute exacerbation of idiopathic pulmonary fibrosis: shifting the paradigm. Eur Respir J. 2015; 46:512–520. [PubMed: 26232481] 49. 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. [PubMed: 7551388] 50. Sethi S, Sethi R, Eschberger K, Lobbins P, Cai X, Grant BJ, Murphy TF. Airway bacterial concentrations and exacerbations of chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2007; 176:356–361. [PubMed: 17478618] 51. Vollenweider DJ, Jarrett H, Steurer-Stey CA, Garcia-Aymerich J, Puhan MA. Antibiotics for exacerbations of chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2012; 12:CD010257. [PubMed: 23235687] 52. Millares L, Ferrari R, Gallego M, Garcia-Nunez M, Perez-Brocal V, Espasa M, Pomares X, Monton C, Moya A, Monso E. Bronchial microbiome of severe COPD patients colonised by Pseudomonas aeruginosa. Eur J Clin Microbiol Infect Dis. 2014; 33:1101–1111. [PubMed: 24449346] 53. Huang YJ, Sethi S, Murphy T, Nariya S, Boushey HA, Lynch SV. Airway microbiome dynamics in exacerbations of chronic obstructive pulmonary disease. J Clin Microbiol. 2014; 52:2813–2823. [PubMed: 24850358] 54. Pragman AA, Kim HB, Reilly CS, Wendt C, Isaacson RE. The lung microbiome in moderate and severe chronic obstructive pulmonary disease. PLoS One. 2012; 7:e47305. [PubMed: 23071781] 55. Hewitt R, Farne H, Ritchie A, Luke E, Johnston SL, Mallia P. The role of viral infections in exacerbations of chronic obstructive pulmonary disease and asthma. Ther Adv Respir Dis. 2015; doi: 10.1177/1753465815618113 56. Papi A, Bellettato CM, Braccioni F, Romagnoli M, Casolari P, Caramori G, Fabbri LM, Johnston SL. Infections and airway inflammation in chronic obstructive pulmonary disease severe exacerbations. Am J Respir Crit Care Med. 2006; 173:1114–1121. [PubMed: 16484677] 57. Rohde G, Wiethege A, Borg I, Kauth M, Bauer TT, Gillissen A, Bufe A, Schultze-Werninghaus G. Respiratory viruses in exacerbations of chronic obstructive pulmonary disease requiring hospitalisation: a case-control study. Thorax. 2003; 58:37–42. [PubMed: 12511718] 58. Mallia P, Message SD, Gielen V, Contoli M, Gray K, Kebadze T, Aniscenko J, Laza-Stanca V, Edwards MR, Slater L, Papi A, Stanciu LA, Kon OM, Johnson M, Johnston SL. Experimental rhinovirus infection as a human model of chronic obstructive pulmonary disease exacerbation. Am J Respir Crit Care Med. 2011; 183:734–742. [PubMed: 20889904] 59. Seemungal T, Harper-Owen R, Bhowmik A, Moric I, Sanderson G, Message S, Maccallum P, Meade TW, Jeffries DJ, Johnston SL, Wedzicha JA. Respiratory viruses, symptoms, and inflammatory markers in acute exacerbations and stable chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2001; 164:1618–1623. [PubMed: 11719299] 60. Molyneaux PL, Mallia P, Cox MJ, Footitt J, Willis-Owen SA, Homola D, Trujillo-Torralbo MB, Elkin S, Kon OM, Cookson WO, Moffatt MF, Johnston SL. Outgrowth of the bacterial airway microbiome after rhinovirus exacerbation of chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2013; 188:1224–1231. [PubMed: 23992479] 61. Mortaz E I, Adcock M, Ricciardolo FL, Varahram M, Jamaati H, Velayati AA, Folkerts G, Garssen J. Anti-Inflammatory Effects of Lactobacillus Rahmnosus and Bifidobacterium Breve on Cigarette Smoke Activated Human Macrophages. PLoS One. 2015; 10:e0136455. [PubMed: 26317628] 62. Tomosada Y, Chiba E, Zelaya H, Takahashi T, Tsukida K, Kitazawa H, Alvarez S, Villena J. Nasally administered Lactobacillus rhamnosus strains differentially modulate respiratory antiviral immune responses and induce protection against respiratory syncytial virus infection. BMC Immunol. 2013; 14:40. [PubMed: 23947615] 63. Marri PR, Stern DA, Wright AL, Billheimer D, Martinez FD. Asthma-associated differences in microbial composition of induced sputum. J Allergy Clin Immunol. 2013; 131:346–352. e341– 343. [PubMed: 23265859] 64. Huang YJ, Nelson CE, Brodie EL, Desantis TZ, Baek MS, Liu J, Woyke T, Allgaier M, Bristow J, Wiener-Kronish JP, Sutherland ER, King TS, Icitovic N, Martin RJ, Calhoun WJ, Castro M, Denlinger LC, Dimango E, Kraft M, Peters SP, Wasserman SI, Wechsler ME, Boushey HA, Lynch

J Immunol. Author manuscript; available in PMC 2017 June 15.

O’Dwyer et al.

Page 14

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

SVL. National Heart, and N. Blood Institute’s Asthma Clinical Research. Airway microbiota and bronchial hyperresponsiveness in patients with suboptimally controlled asthma. J Allergy Clin Immunol. 2011; 127:372–381. e371–373. [PubMed: 21194740] 65. Gonem S, Umar I, Burke D, Desai D, Corkill S, Owers-Bradley J, Brightling CE, Siddiqui S. Airway impedance entropy and exacerbations in severe asthma. Eur Respir J. 2012; 40:1156– 1163. [PubMed: 22408208] 66. Nicholson KG, Kent J, Ireland DC. Respiratory viruses and exacerbations of asthma in adults. BMJ. 1993; 307:982–986. [PubMed: 8241910] 67. Johnston SL, Pattemore PK, Sanderson G, Smith S, Lampe F, Josephs L, Symington P, O’Toole S, Myint SH, Tyrrell DA, et al. Community study of role of viral infections in exacerbations of asthma in 9–11 year old children. BMJ. 1995; 310:1225–1229. [PubMed: 7767192] 68. 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. [PubMed: 21345099] 69. Trompette A, Gollwitzer ES, Yadava K, Sichelstiel AK, Sprenger N, Ngom-Bru C, Blanchard C, Junt T, Nicod LP, Harris NL, Marsland BJ. Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat Med. 2014; 20:159–166. [PubMed: 24390308] 70. Lemaire MM, Dumoutier L, Warnier G, Uyttenhove C, Van Snick J, de Heusch M, Stevens M, Renauld JC. Dual TCR expression biases lung inflammation in DO11.10 transgenic mice and promotes neutrophilia via microbiota-induced Th17 differentiation. J Immunol. 2011; 187:3530– 3537. [PubMed: 21859957] 71. Russell SL, Gold MJ, Willing BP, Thorson L, McNagny KM, Finlay BB. Perinatal antibiotic treatment affects murine microbiota, immune responses and allergic asthma. Gut Microbes. 2013; 4:158–164. [PubMed: 23333861] 72. Raghu G, Rochwerg B, Zhang Y, Garcia CA, Azuma A, Behr J, Brozek JL, Collard HR, Cunningham W, Homma S, Johkoh T, Martinez FJ, Myers J, Protzko SL, Richeldi L, Rind D, Selman M, Theodore A, Wells AU, Hoogsteden H, Schunemann HJ. S. American Thoracic, s. European Respiratory, S. Japanese Respiratory, and A. Latin American Thoracic. An Official ATS/ERS/JRS/ALAT Clinical Practice Guideline: Treatment of Idiopathic Pulmonary Fibrosis. An Update of the 2011 Clinical Practice Guideline. Am J Respir Crit Care Med. 2015; 192:e3–19. [PubMed: 26177183] 73. Knippenberg S, Ueberberg B, Maus R, Bohling J, Ding N, Tort Tarres M, Hoymann HG, Jonigk D, Izykowski N, Paton JC, Ogunniyi AD, Lindig S, Bauer M, Welte T, Seeger W, Guenther A, Sisson TH, Gauldie J, Kolb M, Maus UA. Streptococcus pneumoniae triggers progression of pulmonary fibrosis through pneumolysin. Thorax. 2015; 70:636–646. [PubMed: 25964315] 74. Lasithiotaki I, Antoniou KM, Vlahava VM, Karagiannis K, Spandidos DA, Siafakas NM, Sourvinos G. Detection of herpes simplex virus type-1 in patients with fibrotic lung diseases. PLoS One. 2011; 6:e27800. [PubMed: 22205929] 75. Pulkkinen V, Bruce S, Rintahaka J, Hodgson U, Laitinen T, Alenius H, Kinnula VL, Myllarniemi M, Matikainen S, Kere J. ELMOD2, a candidate gene for idiopathic pulmonary fibrosis, regulates antiviral responses. FASEB J. 2010; 24:1167–1177. [PubMed: 19966137] 76. Molyneaux PL, Cox MJ, Willis-Owen SA, Mallia P, Russell KE, Russell AM, Murphy E, Johnston SL, Schwartz DA, Wells AU, Cookson WO, Maher TM, Moffatt MF. The role of bacteria in the pathogenesis and progression of idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 2014; 190:906–913. [PubMed: 25184687] 77. Folcik VA, Garofalo M, Coleman J, Donegan JJ, Rabbani E, Suster S, Nuovo A, Magro CM, Di Leva G, Nuovo GJ. Idiopathic pulmonary fibrosis is strongly associated with productive infection by herpesvirus saimiri. Mod Pathol. 2014; 27:851–862. [PubMed: 24232864] 78. Collard HR, Moore BB, Flaherty KR, Brown KK, Kaner RJ, King TE Jr, Lasky JA, Loyd JE, Noth I, Olman MA, Raghu G, Roman J, Ryu JH, Zisman DA, Hunninghake GW, Colby TV, Egan JJ, Hansell DM, Johkoh T, Kaminski N, Kim DS, Kondoh Y, Lynch DA, Muller-Quernheim J, Myers JL, Nicholson AG, Selman M, Toews GB, Wells AU, Martinez FJ. I. Idiopathic Pulmonary Fibrosis Clinical Research Network. Acute exacerbations of idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 2007; 176:636–643. [PubMed: 17585107]

J Immunol. Author manuscript; available in PMC 2017 June 15.

O’Dwyer et al.

Page 15

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

79. Han MK, Zhou Y, Murray S, Tayob N, Noth I, Lama VN, Moore BB, White ES, Flaherty KR, Huffnagle GB, Martinez FJ, Investigators C. Lung microbiome and disease progression in idiopathic pulmonary fibrosis: an analysis of the COMET study. Lancet Respir Med. 2014; 2:548– 556. [PubMed: 24767767] 80. Varney VA, Parnell HM, Salisbury DT, Ratnatheepan S, Tayar RB. A double blind randomised placebo controlled pilot study of oral co-trimoxazole in advanced fibrotic lung disease. Pulm Pharmacol Ther. 2008; 21:178–187. [PubMed: 17500020] 81. O’Dwyer DN, Armstrong ME, Trujillo G, Cooke G, Keane MP, Fallon PG, Simpson AJ, Millar AB, McGrath EE, Whyte MK, Hirani N, Hogaboam CM, Donnelly SC. The Toll-like receptor 3 L412F polymorphism and disease progression in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 2013; 188:1442–1450. [PubMed: 24070541] 82. Trujillo G, Meneghin A, Flaherty KR, Sholl LM, Myers JL, Kazerooni EA, Gross BH, Oak SR, Coelho AL, Evanoff H, Day E, Toews GB, Joshi AD, Schaller MA, Waters B, Jarai G, Westwick J, Kunkel SL, Martinez FJ, Hogaboam CM. TLR9 differentiates rapidly from slowly progressing forms of idiopathic pulmonary fibrosis. Sci Transl Med. 2010; 2:57ra82. 83. Lafyatis R, Farina A. New insights into the mechanisms of innate immune receptor signalling in fibrosis. Open Rheumatol J. 2012; 6:72–79. [PubMed: 22802904] 84. Margaritopoulos GA, Antoniou KM, Karagiannis K, Samara KD, Lasithiotaki I, Vassalou E, Lymbouridou R, Koutala H, Siafakas NM. Investigation of Toll-like receptors in the pathogenesis of fibrotic and granulomatous disorders: a bronchoalveolar lavage study. Fibrogenesis Tissue Repair. 2010; 3:20. [PubMed: 20937083] 85. Papanikolaou IC, Boki KA, Giamarellos-Bourboulis EJ, Kotsaki A, Kagouridis K, Karagiannidis N, Polychronopoulos VS. Innate immunity alterations in idiopathic interstitial pneumonias and rheumatoid arthritis-associated interstitial lung diseases. Immunol Lett. 2015; 163:179–186. [PubMed: 25540922] 86. Samara KD, Antoniou KM, Karagiannis K, Margaritopoulos G, Lasithiotaki I, Koutala E, Siafakas NM. Expression profiles of Toll-like receptors in non-small cell lung cancer and idiopathic pulmonary fibrosis. Int J Oncol. 2012; 40:1397–1404. [PubMed: 22344343] 87. Stenbit AE, Flume PA. Pulmonary exacerbations in cystic fibrosis. Curr Opin Pulm Med. 2011; 17:442–447. [PubMed: 21881509] 88. Ramsey BW. Management of pulmonary disease in patients with cystic fibrosis. N Engl J Med. 1996; 335:179–188. [PubMed: 8657217] 89. Gold R, Carpenter S, Heurter H, Corey M, Levison H. Randomized trial of ceftazidime versus placebo in the management of acute respiratory exacerbations in patients with cystic fibrosis. J Pediatr. 1987; 111:907–913. [PubMed: 3316565] 90. Hurley MN, Ariff AH, Bertenshaw C, Bhatt J, Smyth AR. Results of antibiotic susceptibility testing do not influence clinical outcome in children with cystic fibrosis. J Cyst Fibros. 2012; 11:288–292. [PubMed: 22436723] 91. Smith AL, Fiel SB, Mayer-Hamblett N, Ramsey B, Burns JL. Susceptibility testing of Pseudomonas aeruginosa isolates and clinical response to parenteral antibiotic administration: lack of association in cystic fibrosis. Chest. 2003; 123:1495–1502. [PubMed: 12740266] 92. Price KE, Hampton TH, Gifford AH, Dolben EL, Hogan DA, Morrison HG, Sogin ML, O’Toole GA. Unique microbial communities persist in individual cystic fibrosis patients throughout a clinical exacerbation. Microbiome. 2013; 1:27. [PubMed: 24451123] 93. 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. [PubMed: 23802813] 94. Stressmann FA, Rogers GB, Marsh P, Lilley AK, Daniels TW, Carroll MP, Hoffman LR, Jones G, Allen CE, Patel N, Forbes B, Tuck A, Bruce KD. Does bacterial density in cystic fibrosis sputum increase prior to pulmonary exacerbation? J Cyst Fibros. 2011; 10:357–365. [PubMed: 21664196] 95. Cox MJ, Allgaier M, Taylor B, Baek MS, Huang YJ, Daly RA, Karaoz U, Andersen GL, Brown R, Fujimura KE, Wu B, Tran D, Koff J, Kleinhenz ME, Nielson D, Brodie EL, Lynch SV. Airway microbiota and pathogen abundance in age-stratified cystic fibrosis patients. PLoS One. 2010; 5:e11044. [PubMed: 20585638]

J Immunol. Author manuscript; available in PMC 2017 June 15.

O’Dwyer et al.

Page 16

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

96. Zhao J, Schloss PD, Kalikin LM, Carmody LA, Foster BK, Petrosino JF, Cavalcoli JD, VanDevanter DR, Murray S, Li JZ, Young VB, LiPuma JJ. Decade-long bacterial community dynamics in cystic fibrosis airways. Proc Natl Acad Sci U S A. 2012; 109:5809–5814. [PubMed: 22451929] 97. Aitken ML, Limaye A, Pottinger P, Whimbey E, Goss CH, Tonelli MR, Cangelosi GA, Dirac MA, Olivier KN, Brown-Elliott BA, McNulty S, Wallace RJ Jr. Respiratory outbreak of Mycobacterium abscessus subspecies massiliense in a lung transplant and cystic fibrosis center. Am J Respir Crit Care Med. 2012; 185:231–232. [PubMed: 22246710] 98. Esther CR Jr, Esserman DA, Gilligan P, Kerr A, Noone PG. Chronic Mycobacterium abscessus infection and lung function decline in cystic fibrosis. J Cyst Fibros. 2010; 9:117–123. [PubMed: 20071249] 99. Sanguinetti M, Ardito F, Fiscarelli E, La Sorda M, D’Argenio P, Ricciotti G, Fadda G. Fatal pulmonary infection due to multidrug-resistant Mycobacterium abscessus in a patient with cystic fibrosis. J Clin Microbiol. 2001; 39:816–819. [PubMed: 11158161] 100. Floto RA, Olivier KN, Saiman L, Daley CL, Herrmann JL, Nick JA, Noone PG, Bilton D, Corris P, Gibson RL, Hempstead SE, Koetz K, Sabadosa KA, Sermet-Gaudelus I, Smyth AR, van Ingen J, Wallace RJ, Winthrop KL, Marshall BC, Haworth CS. US Cystic Fibrosis Foundation and European Cystic Fibrosis Society consensus recommendations for the management of nontuberculous mycobacteria in individuals with cystic fibrosis. Thorax. 2016; 71(Suppl 1):i1–i22. [PubMed: 26666259] 101. Clarke TB. Early innate immunity to bacterial infection in the lung is regulated systemically by the commensal microbiota via nod-like receptor ligands. Infect Immun. 2014; 82:4596–4606. [PubMed: 25135683] 102. Sassone-Corsi M, Raffatellu M. No vacancy: how beneficial microbes cooperate with immunity to provide colonization resistance to pathogens. J Immunol. 2015; 194:4081–4087. [PubMed: 25888704] 103. Savage DC. Microbial ecology of the gastrointestinal tract. Annu Rev Microbiol. 1977; 31:107– 133. [PubMed: 334036] 104. Arumugam M, Raes J, Pelletier E, Le Paslier D, Yamada T, Mende DR, Fernandes GR, Tap J, Bruls T, Batto JM, Bertalan M, Borruel N, Casellas F, Fernandez L, Gautier L, Hansen T, Hattori M, Hayashi T, Kleerebezem M, Kurokawa K, Leclerc M, Levenez F, Manichanh C, Nielsen HB, Nielsen T, Pons N, Poulain J, Qin J, Sicheritz-Ponten T, Tims S, Torrents D, Ugarte E, Zoetendal EG, Wang J, Guarner F, Pedersen O, de Vos WM, Brunak S, Dore J, Meta HITC, Antolin M, Artiguenave F, Blottiere HM, Almeida M, Brechot C, Cara C, Chervaux C, Cultrone A, Delorme CA, Denariaz G, Dervyn R, Foerstner KU, Friss C, van de Guchte M, Guedon E, Haimet F, Huber W, van Hylckama-Vlieg J, Jamet A, Juste C, Kaci G, Knol J, Lakhdari O, Layec S, Le Roux K, Maguin E, Merieux A, Melo Minardi R, M’Rini C, Muller J, Oozeer R, Parkhill J, Renault P, Rescigno M, Sanchez N, Sunagawa S, Torrejon A, Turner K, Vandemeulebrouck G, Varela E, Winogradsky Y, Zeller G, Weissenbach J, Ehrlich SD, Bork P. Enterotypes of the human gut microbiome. Nature. 2011; 473:174–180. [PubMed: 21508958] 105. Abrahamsson TR, Jakobsson HE, Andersson AF, Bjorksten B, Engstrand L, Jenmalm MC. Low diversity of the gut microbiota in infants with atopic eczema. J Allergy Clin Immunol. 2012; 129:434–440. 440 e431–432. [PubMed: 22153774] 106. Bruzzese E, Callegari ML, Raia V, Viscovo S, Scotto R, Ferrari S, Morelli L, Buccigrossi V, Lo Vecchio A, Ruberto E, Guarino A. Disrupted intestinal microbiota and intestinal inflammation in children with cystic fibrosis and its restoration with Lactobacillus GG: a randomised clinical trial. PLoS One. 2014; 9:e87796. [PubMed: 24586292] 107. Inagaki H, Suzuki T, Nomoto K, Yoshikai Y. Increased susceptibility to primary infection with Listeria monocytogenes in germfree mice may be due to lack of accumulation of L-selectin+ CD44+ T cells in sites of inflammation. Infect Immun. 1996; 64:3280–3287. [PubMed: 8757865] 108. Fagundes CT, Amaral FA, Vieira AT, Soares AC, Pinho V, Nicoli JR, Vieira LQ, Teixeira MM, Souza DG. Transient TLR activation restores inflammatory response and ability to control pulmonary bacterial infection in germfree mice. J Immunol. 2012; 188:1411–1420. [PubMed: 22210917]

J Immunol. Author manuscript; available in PMC 2017 June 15.

O’Dwyer et al.

Page 17

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

109. Abt MC, Osborne LC, Monticelli LA, Doering TA, Alenghat T, Sonnenberg GF, Paley MA, Antenus M, Williams KL, Erikson J, Wherry EJ, Artis D. Commensal bacteria calibrate the activation threshold of innate antiviral immunity. Immunity. 2012; 37:158–170. [PubMed: 22705104] 110. Clarke TB, Davis KM, Lysenko ES, Zhou AY, Yu Y, Weiser JN. Recognition of peptidoglycan from the microbiota by Nod1 enhances systemic innate immunity. Nat Med. 2010; 16:228–231. [PubMed: 20081863] 111. MacDonald M, Korman T, King P, Hamza K, Bardin P. Exacerbation phenotyping in chronic obstructive pulmonary disease. Respirology. 2013; 18:1280–1281. [PubMed: 24164687] 112. Jaspers I. Cigarette smoke effects on innate immune mechanisms in the nasal mucosa. Potential effects on the microbiome. Ann Am Thorac Soc. 2014; 11(Suppl 1):S38–42. [PubMed: 24437404] 113. Wang J, Li F, Wei H, Lian ZX, Sun R, Tian Z. Respiratory influenza virus infection induces intestinal immune injury via microbiota-mediated Th17 cell-dependent inflammation. J Exp Med. 2014; 211:2397–2410. [PubMed: 25366965] 114. Chewning JH, Weaver CT. Development and survival of Th17 cells within the intestines: the influence of microbiome- and diet-derived signals. J Immunol. 2014; 193:4769–4777. [PubMed: 25381358] 115. Tan HL, Rosenthal M. IL-17 in lung disease: friend or foe? Thorax. 2013; 68:788–790. [PubMed: 23604380] 116. Kudo M, Melton AC, Chen C, Engler MB, Huang KE, Ren X, Wang Y, Bernstein X, Li JT, Atabai K, Huang X, Sheppard D. IL-17A produced by alphabeta T cells drives airway hyperresponsiveness in mice and enhances mouse and human airway smooth muscle contraction. Nat Med. 2012; 18:547–554. [PubMed: 22388091] 117. Tan HL, Regamey N, Brown S, Bush A, Lloyd CM, Davies JC. The Th17 pathway in cystic fibrosis lung disease. Am J Respir Crit Care Med. 2011; 184:252–258. [PubMed: 21474644] 118. Facco M, Cabrelle A, Teramo A, Olivieri V, Gnoato M, Teolato S, Ave E, Gattazzo C, Fadini GP, Calabrese F, Semenzato G, Agostini C. Sarcoidosis is a Th1/Th17 multisystem disorder. Thorax. 2011; 66:144–150. [PubMed: 21139119] 119. Vanaudenaerde BM, De Vleeschauwer SI, Vos R, Meyts I, Bullens DM, Reynders V, Wuyts WA, Van Raemdonck DE, Dupont LJ, Verleden GM. The role of the IL23/IL17 axis in bronchiolitis obliterans syndrome after lung transplantation. Am J Transplant. 2008; 8:1911–1920. [PubMed: 18786233] 120. Zhou X, Loomis-King H, Gurczynski SJ, Wilke CA, Konopka KE, Ptaschinski C, Coomes SM, Iwakura Y, van Dyk LF, Lukacs NW, Moore BB. Bone marrow transplantation alters lung antigen-presenting cells to promote T17 response and the development of pneumonitis and fibrosis following gammaherpesvirus infection. Mucosal Immunol. 2015; doi: 10.1038/mi. 2015.85 121. Yadava K, Pattaroni C, Sichelstiel AK, Trompette A, Gollwitzer ES, Salami O, von Garnier C, Nicod LP, Marsland BJ. Microbiota Promotes Chronic Pulmonary Inflammation by Enhancing IL-17A and Autoantibodies. Am J Respir Crit Care Med. 2015; doi: 10.1164/rccm. 201504-0779OC 122. Tan Z, Qian X, Jiang R, Liu Q, Wang Y, Chen C, Wang X, Ryffel B, Sun B. IL-17A plays a critical role in the pathogenesis of liver fibrosis through hepatic stellate cell activation. J Immunol. 2013; 191:1835–1844. [PubMed: 23842754] 123. Wilson MS, Madala SK, Ramalingam TR, Gochuico BR, Rosas IO, Cheever AW, Wynn TA. Bleomycin and IL-1beta-mediated pulmonary fibrosis is IL-17A dependent. J Exp Med. 2010; 207:535–552. [PubMed: 20176803] 124. Ray S, De Salvo C, Pizarro TT. Central role of IL-17/Th17 immune responses and the gut microbiota in the pathogenesis of intestinal fibrosis. Curr Opin Gastroenterol. 2014; 30:531–538. [PubMed: 25255234] 125. Atarashi K, Tanoue T, Ando M, Kamada N, Nagano Y, Narushima S, Suda W, Imaoka A, Setoyama H, Nagamori T, Ishikawa E, Shima T, Hara T, Kado S, Jinnohara T, Ohno H, Kondo T, Toyooka K, Watanabe E, Yokoyama S, Tokoro S, Mori H, Noguchi Y, Morita H, Ivanov,

J Immunol. Author manuscript; available in PMC 2017 June 15.

O’Dwyer et al.

Page 18

Author Manuscript

Sugiyama T, Nunez G, Camp JG, Hattori M, Umesaki Y, Honda K. Th17 Cell Induction by Adhesion of Microbes to Intestinal Epithelial Cells. Cell. 2015; 163:367–380. [PubMed: 26411289] 126. Gauguet S, D’Ortona S, Ahnger-Pier K, Duan B, Surana NK, Lu R, Cywes-Bentley C, Gadjeva M, Shan Q, Priebe GP, Pier GB. Intestinal Microbiota of Mice Influences Resistance to Staphylococcus aureus Pneumonia. Infect Immun. 2015; 83:4003–4014. [PubMed: 26216419]

Author Manuscript Author Manuscript Author Manuscript J Immunol. Author manuscript; available in PMC 2017 June 15.

O’Dwyer et al.

Page 19

Author Manuscript Author Manuscript Author Manuscript

Figure 1. Proposed regulation of disease exacerbation by the gut-lung axis

Author Manuscript

During normal homeostasis, the lung microbiome is primarily characterized by low biomass, but prominent diversity in microbial species. In contrast, the healthy gut microbiome is characterized by high diversity and high biomass. In homeostasis, the gut microbiome helps shape development of lymphoid architecture and appropriate immune responsiveness. Loss of gut diversity (e.g. as a result of viral infection or antibiotic use) may cause dysregulation of IL-17 or bacterial killing mechanisms systemically; potentially leading to impaired alveolar macrophage function and the overgrowth of selective organisms with pathogenic potential that may result in disease exacerbation. Alternatively, some forms of chronic lung disease exacerbations may be due to translocation and/or expansion of bacterial contents from the gut. Direct insults to the lung (e.g. viral infection or aspiration) may exacerbate disease in part via their effects on the lung or gut microbiota. Alterations in systemic cytokines (e.g. Th2 or Th17 induction) may promote pathologic fibrotic remodeling as well.

J Immunol. Author manuscript; available in PMC 2017 June 15.

O’Dwyer et al.

Page 20

Table 1

Author Manuscript

Factors that influence the lung microbiota during acute and chronic disease Architectural: •

Airway obliteration (lung transplant, IPF)



Terminal bronchiole destruction (COPD)



Honeycombing and fibrosis (IPF)



Impaired mucociliary clearance (COPD, asthma)

Immunologic:

Author Manuscript



Innate immune cell impairment



Altered PRR signaling



Release of anti-microbial peptides



Apoptosis/Autophagy



Inflammation



Cytokine alterations

Microbiologic: •

Overgrowth of limited bacterial species (IPF, CF)



Antibiotic use (esp. in CF)



Lytic viral infection (COPD, asthma)



Latent viral infection (IPF?)



Biofilm formation (CF, COPD)

Pathologic: •

Osmotic changes (CF)



Thickened mucus (CF)

Author Manuscript



Damaged cilia (COPD)



Changes in oxygen tension, ventilation and perfusion (IPF, COPD, CF, asthma)



Micro aspiration (IPF)

Author Manuscript J Immunol. Author manuscript; available in PMC 2017 June 15.

The Lung Microbiome, Immunity, and the Pathogenesis of Chronic Lung Disease.

The development of culture-independent techniques for microbiological analysis has uncovered the previously unappreciated complexity of the bacterial ...
436KB Sizes 6 Downloads 9 Views