Abstract
Culture-independent study of the lower respiratory tract after lung transplantation has enabled an understanding of the microbiome – that is, the collection of bacteria, fungi, and viruses, and their respective gene complement – in this niche. The lung has unique features as a microbial environment, with balanced entry from the upper respiratory tract, clearance, and local replication. There are many pressures impacting the microbiome after transplantation, including donor allograft factors, recipient host factors such as underlying disease and ongoing exposure to the microbe-rich upper respiratory tract, and transplantation-related immunosuppression, antimicrobials, and post-surgical changes. To date, we understand that the lung microbiome after transplant is dysbiotic; that is, it has higher biomass and altered composition compared to a healthy lung. Emerging data suggest that specific microbiome features may be linked to host responses, both immune and non-immune, and clinical outcomes such as chronic lung allograft dysfunction (CLAD), but many questions remain. The goal of this review is to put into context our burgeoning understanding of the lung microbiome in the post-lung transplant patient, the interactions between microbiome and host, the role the microbiome may play in post-transplant complications, and critical outstanding research questions.
Introduction
Lung transplantation is the only therapy for many end-stage lung diseases, but lung allografts have the shortest survival of all solid-organ transplants1. Early challenges include primary graft dysfunction (PGD), acute cellular and antibody-mediated rejection (ACR, AMR), infections, and anastomotic complications, while the principal barrier to long-term allograft survival is chronic lung allograft dysfunction (CLAD), which leads to relentless graft failure1,2. Several lung infections increase the risk of later CLAD and azithromycin slows progression3,4 suggesting a role for host-microbiota interactions in CLAD pathogenesis.
The microbiome, defined as the collection of microbes and their genetic content in a distinct ecological location5,6, considers entire communities and not just individual organisms, employs statistical analysis of community characteristics, and enables identification of microbiome “types.” While the microbe-rich gastrointestinal tract was the initial focus of such work7, emerging studies of the lung microbiome in lung transplantation have demonstrated marked abnormalities compared to health, termed “dysbiosis”8–15. In the post-transplantation setting, however, the most relevant comparison is not to individuals without lung disease, but between post-transplant subjects with healthy allografts and those with dysfunction. This review addresses the unique post-transplantation lung microbiome, links to transplant outcomes, and host-microbiota cross-talk. In addition to the bacterial microbiome, which is the most-studied microbiome component, we address the less-studied communities of viruses and fungi.
Culture-Independent Sequencing
Microbial culture has been indispensable for understanding microbial pathogenesis but captures only a subset of the microbial landscape. Advances in sequencing technology (“next-generation” or “deep sequencing”) and computational methods have enabled a comprehensive, unbiased culture-independent approach16. Sequence-based approaches quantify the relative abundances of specific microbes in a sample and readily identify both potential pathogens and the vast commensal landscape. The most widely-used approach targets the bacterial 16S rRNA gene17–20. Here polymerase chain reaction (PCR) amplifies bacterial DNA regions that, when sequenced, infer bacterial taxonomy16,20. Targeted 16S rRNA gene analysis also captures archaea, which are scant in the lung and generally under-studied. Fungi (the “mycobiome”) are studied using analogous targeting of shared fungal regions, such as the 18S rRNA gene or internal transcribed spacer (ITS) sequence in the ribosomal gene locus.
In contrast, viruses have no such universally shared sequences so a complementary approach used is shotgun metagenomic sequencing, which sequences all the DNA (or RNA depending on sample preparation)21,22. Thus, shotgun sequencing can profile viruses (the “virome”) as well as fungi and bacteria. Another advantage to shotgun sequencing is that it also captures microbial gene content, which assesses the genetic potential for microbial functions (e.g., metabolism, virulence factors, antimicrobial resistance). However, large amounts of host relative to microbial DNA in lung samples increases the cost and complexity of analysis23; although with recent advances, shotgun metagenomics is enjoying broader use in lung microbiome research23–26. Table 1 lists selected key elements in microbiome investigation relevant to this review.
Table 1.
|
The Normal Lung Microbiome
Composition and source of the lung bacterial microbiome in health.
Traditionally, the lung below the glottis was thought to be sterile, but recent work has dispelled this dogma even in health27,28. This is perhaps not surprising, as the lung is contiguous with the microbe-rich upper respiratory tract (URT). However, the microbial biomass in the lung is orders of magnitude lower than the oropharynx. Therefore, stringent approaches are required to distinguish authentic signal from artifact derived from both oropharyngeal carryover during bronchoscopy and the background noise of microbial sequences from instruments and reagents22,29. Applying such approaches, we now know the healthy lung microbiota largely overlaps those from the URT and glottic region28. The most abundant taxa are from Firmicutes and Bacteroidetes phyla, with the most common genera Prevotella, Streptococcus, and Veillonella28,30,27,28,30,31.
The near-identical composition of these two niches that are physically contiguous but ecologically distinct indicates that the primary source of lung bacteria is the URT30,32–34. They are derived mainly by microaspiration known to occur in all healthy people27, balanced by clearance through mucociliary and innate immune mechanisms35–37. There are modest regional differences within the lung and slight divergence from upper respiratory communities, suggesting differential clearance and possibly limited local replication even in healthy people30. Indeed, a proportion of these lung-derived bacteria are viable and transcriptionally active38. Thus the healthy lung bacterial microbiome is low-biomass, transient, and dynamic, which differs from the gut and URT microbiomes and indicates distinct microbial ecology in the lung39. This balance is disrupted in diseases such as cystic fibrosis (CF), bronchiectasis and post-transplantation where local growth may become a primary factor for lung microbial composition.
The lung virome and mycobiome.
These components of the lung microbiome are less studied. Apart from acute infection with community-acquired viruses, sequence-based analysis has identified Anelloviridae as the most common and abundant component of the lung virome, along with bacteriophage, and more variable detection of other eukaryotic viruses including Herpesviruses and Papillomaviruses40,41. Anelloviridae are small, single-stranded DNA circular-genome viruses found in blood and many tissues, with nearly 100% prevalence in humans, but have not been associated with disease42.
The fungal constituents in health are also understudied and present unique challenges8,23,28,41,43–45. In addition to URT and background contamination, many samples have very low fungal biomass and low number of fungal taxa present46, fungal cell walls are difficult to lyse, there are biases in 18S and ITS primers, non-standardized nomenclature, and poor annotation of fungal databases41. The limited small studies in healthy lungs show low fungal biomass and inter-individual heterogeneity, with fungi from the Ascomycota phylum most common, followed less commonly by Basidiomycota. The main genera were Candida, Saccharomyces, Penicillium, Cladosporium, and occasionally other environmental taxa; Aspergillus and Pneumocystis have been inconsistently identified at low levels in health.
The lung bacterial microbiome and local immune tone.
In the healthy lung, aspirated bacteria encounter efficient mechanical and innate immune defenses which oppose colonization37,47. Nevertheless, despite the low biomass and dynamic nature of the lung bacterial microbiome, it impacts the local immune tone. In healthy humans, the presence of bacterial sequences in the lung that match those in the oropharynx correlates with higher basal Th17 lymphocytes when compared to people with little authentic lung bacterial content48. Thus, entry of URT-derived microbiota may regulate tonic immune profiles in the lung.
At the same time, it is well-established that the gut microbiome, with exponentially higher biomass, sets systemic immune tone, and it may also impact the lung immune milieu through metabolites that enter the systemic circulation, the so-called gut-lung axis49–51. In mice, both local lung and gut microbiota influenced lung immune profiles, but local lung microbial diversity and composition more strongly influenced pulmonary immune tone (referred to as the lung-lung axis)52. Thus, both the lung-lung and gut-lung axes contribute to the lung immune setpoint.
Establishment of the Post-Transplantation Lung Microbiome
Studies of the post-transplantation lung microbiome8–15,53,54 have uniformly found higher microbial biomass and dysbiosis55 characterized by lower alpha-diversity (a measure incorporating both the number of species and their evenness) and altered composition compared to healthy subjects. Dysbiosis likely results from donor-dependent, recipient-specific, and transplantation-related factors.
Pre-transplantation lung disease influences the post-transplantation microbiome.
Patients with CF or other suppurative lung diseases have distinct lung microbiomes8,10,56,57, which can impact the post-transplant microbiome. Patients with CF have upper respiratory mucosal and sinus pathology with persistent colonization, and despite bilateral transplantation, these microbes often repopulate the lung allograft56–58. The post-transplant bacterial microbiome in CF tends to be distinct from non-suppurative lung disease and, similar to pre-transplant profiles, often enriched in Proteobacteria, particularly Pseudomonas, though this is not universal8–10,57.
Non-suppurative lung conditions requiring transplantation have microbiota that deviate from non-diseased healthy individuals, including emphysema, interstitial pulmonary fibrosis, and other interstitial diseases59–62. Although less dramatic and lower microbial biomass than that seen in CF, pre-transplant lung dysbiosis may also impact the post-transplant microbiome, especially in cases of single lung transplant54.
As described above, the URT is the source of the lung microbiome in health, and microaspiration is especially frequent post-transplantation63,64. The URT is markedly dysbiotic in individuals with advanced lung disease both before and after transplantation65. Whether this results from the underlying lung disease, its treatment, or both, is unclear. Nevertheless, the markedly dysbiotic oropharyngeal microbiome that provides a source of lung microbes likely contributes to lung microbiota post-transplantation.
Donor factors may impact the post-transplant microbiome.
Donor lungs are selected based on the absence of significant pathology, but post-transplant donor-derived infections are well-known66. Donors are mechanical ventilated, and in this setting the lung microbiome is dysbiotic67–70. While not well-studied, donor microbiota persistence in the allograft may be a factor shaping the post-transplant lung microbiome10. This may be altered further by the use of peri-transplantation antibiotics, but the degree to which this happens is unknown. A striking example of donor dysbiosis involves the lung virome, which is dominated by Anelloviridae. Anelloviridae sequences in donor lungs were elevated 100-fold compared to healthy people71, and these elevated Anelloviridae persist post-transplantation (and disseminate from the allograft to recipient circulation)72.
Transplant-related factors impact the lung microbiome.
Multiple surgical factors alter post-transplant lung microbial defense73. Mainstem bronchial anastomoses and airway ischemia both may impair the mucociliary elevator, transplantation results in a lymphatic system that is not surgically reconnected, and vagal denervation leads to loss of afferent stimulation and dysfunctional cough reflex, impairing clearance.
Immunosuppression is required for allograft tolerance but increases the risk of both community-acquired and opportunistic infections74. It seems logical that immune suppression would impact the lung microbiome through modulation of immune clearance but this has not been studied directly and is a key area for investigation75.
Patients are given antimicrobials post-transplant for both prophylactic and therapeutic indications74,76. Antimicrobials profoundly alter the gut bacterial microbiome composition, decrease diversity, and increase the reservoir of antibiotic resistance genes77,78. Emerging findings suggest parallel effects may impact the lung79. In mice, ceftriaxone reduced the relative abundance of lung Proteobacteria (Gram-negative bacteria) and increased Firmicutes (Gram-positives)52. Chronic azithromycin is used to reduce exacerbations in obstructive lung diseases, and in lung transplantation for CLAD prevention. Azithromycin may involve both immunomodulatory and antibacterial mechanisms, but whether it works through modifying the lung microbiome is unknown and discordant effects been reported53,80,81. In post-transplant patients, azithromycin did not influence lung microbiome total bacterial burden, alpha-diversity, or high-abundance taxa, though differences in low abundance taxa such as reduced Enterobacteriaceae and Pseudomonas were seen at 1–2 years53.
The Lung Bacterial Microbiome After Transplantation
Microbiome dysbiosis post-transplantation.
Multiple studies have found elevated bacterial burden compared to healthy controls, ranging from 15-fold to 1000-fold higher8,9,15,53,54. Despite substantial methodological differences, studies indicate that a subset of post-transplant patients maintains a lung microbiome composition predominantly of oropharyngeal-type flora while most have a marked decrease in alpha-diversity accompanied an outgrowth of specific taxa8,14,15,53. Quantitative comparison of lung versus URT communities shows outgrowth in the lung of commensals and potential pathogens, such as Pseudomonas, Enterobacteriaceae, Staphylococcus, as well as anaerobes8. In longitudinal studies of BAL from routine care, URT flora usually dominated in the first year after transplant14,15. The most abundant phylum was Bacteroidetes, followed by Firmicutes, then Proteobacteria, and Actinobacteria14,15. A smaller proportion of samples exhibited outgrowth of Corynebacterium and potential pathogens such as Pseudomonas, Staphylococcus, and Stenotrophomonas. In CF, two main types of post-transplant lung bacterial communities have been described: one typical of oral bacteria (particularly Streptococcus and Veillonella), and one dominated by Pseudomonas10.
Enrichment of Proteobacteria following transplantation is common, notably Pseudomonas9,10,57,58,82. A study that carried out a more granular analysis revealed two species within the genus, P. aeruginosa and P. flourescens9. A higher relative abundance of P. aeruginosa was associated with lower diversity, more bacterial DNA, greater neutrophilia, and having symptoms at time of sampling. These culture-independent data add nuance to the culture-based associations between Pseudomonas aeruginosa and bronchiolitis obliterans syndrome (BOS; the most common phenotype of CLAD)11,82–85.
Thus, across studies, the post-transplant lung microbiome is higher biomass and deviates from healthy individuals, but there is a high degree of patient-specificity. While this finding precludes broad generalizations about any homogenous “post-transplant lung microbiome” composition, it is this very heterogeneity that suggests distinct outcomes may be linked to microbiome profiles. Furthermore, the term “dysbiosis” is generally used to connote divergence from health, but essentially all post-transplant patients’ lung microbiomes differ from non-transplant healthy people. Therefore, the key question is whether there is an optimal post-transplantation lung microbiome versus communities that associate with elevated risk of post-transplantation complications. Similarly, it is important to distinguish microbiomes characterized during “routine” surveillance bronchoscopy, versus sampling done for-cause such as symptoms or allograft dysfunction.
Spatial and temporal heterogeneity of the lung microbiome.
While the lung microbiome is often conceptualized as a homogenous entity, there is under-appreciated spatial heterogeneity30. In transplant patients, differences have been reported between proximal and distal samples12,86 and between native lungs and allografts in after single lung transplant for interstitial lung disease (ILD) (in individuals with lung function decline)54. This could result from regional differences in the entry, clearance, or local replication.
There are complex dynamics at the time of transplant and in the first post-transplant year66. Early on, donor microbiota persists in some subjects post-transplant, while in others, it is rapidly replaced10. In CF, Pseudomonas strains derived from recipient populated the allograft within days58,57. Far less is known about how pre-transplant native lung communities impact the post-transplant microbiome in non-CF patients66.
Interestingly, there is extensive change over time in bacterial composition at lower (more granular) taxonomic levels, but more stability at higher taxonomic levels. In a longitudinal study, only ~10% of operational taxonomic units (OTUs, the molecular surrogate for bacterial species) found at the final time point were present initially11. However, 46% of subjects exhibiting consistent phylum-level profiles over time. Furthermore, of those that had transitions in phylum-level profiles, about half went from dysbiosis (one phylum constituting >70% of the community) to no dysbiosis.
The bacterial microbiome and transplantation outcomes.
BOS, the more common obstructive form of CLAD, has had variable associations with the lung microbiome. An early study found that in CF, the presence of Pseudomonas species post-transplant appeared protective against BOS, particularly if the patient had Pseudomonas (identified by culture) pre-transplant10. In contrast, enrichment with oral taxa increased BOS incidence. This did not hold for non-CF subjects. Furthermore, decreased diversity over time within an individual increased BOS risk. These findings raised the hypothesis that the stability of microbiome pre-/post-transplant, rather than specific taxa, impacts BOS risk.
However, subsequent studies reported different microbiome correlations with BOS. A small, uncontrolled study found enrichment of Actinomyces and Xanthomonadaceae in BOS11. Adding further complexity, both time- and location-specificity also matter. A study using shotgun metagenomic sequencing from routine bronchoscopy samples before BOS onset found Actinobacteria-dominance (Propionibacterium and Corynebacterium) at three months post-transplant had lower rates of BOS, less inflammation in BAL, and lower rates of ACR13. However, this association did not hold for later time points, suggesting a time-dependent association with outcomes. In an examination of topographical features of the post-transplant lung microbiome, microbial and inflammatory profiles differed in proximal (early BAL return) and distal airways (late BAL return), and BOS was linked to specific taxa from three phyla (Proteobacteria, Bacteroidetes, Firmicutes) in distal but not proximal specimens12.
A recent study prospectively analyzed the microbiome from BAL from routine bronchoscopy one year after transplantation and looked at the predictors of subsequent CLAD87. They found that bacterial biomass was a strong predictor of CLAD development or death within 500 days of the sample collection. For the highest tertile of biomass there was a hazard ratio (HR) of 5.98 (10.56 in multivariable modeling) for the composite outcome and when analyzed continuously for each log10 increase in bacterial biomass there was a HR of 1.89 (2.49 in multivariable model). In their compositional cluster analysis they found subjects who had “pharyngeal” or “inflammation-associated” taxa (such as Pseudomonas) had the highest risk of CLAD or death. No specific taxon was independently associated with CLAD. Additionally, immunosuppression neither significantly impacted bacterial microbiome composition nor mediated the effect between the microbiome and outcome. Overall, this study that found that early microbiome ‘type’ was linked to subsequent adverse outcomes.
In summary, emerging data suggest a complex relationship between the lung bacterial microbiome and BOS. Unraveling this complexity must account for temporal, spatial, and patient-specific factors. In addition, cross-kingdom interactions with the virome and mycobiome, as well as an integration with the multifaceted host response after lung transplantation needs future study.
The Lung Virome and Mycobiome after Transplantation
Eukaryotic viruses.
Herpesviruses, particularly cytomegalovirus (CMV) and herpes simplex virus (HSV), are important pathogens after lung transplantation, and are targets of prophylactic antiviral regimens74. Community-acquired respiratory viruses (CARV’s), such as influenza, enteroviruses, and rhinoviruses, lead to acute morbidity and increased risk of later CLAD3,88,89. These viruses are typically identified by targeted PCR methods. While there is a burgeoning literature probing the human virome through sequence-based methods40,71,72,89,90, only a few focused on the lung and even fewer in lung transplantation.
The most abundant eukaryotic viruses in lung transplant BAL are of the Anelloviridae family--small circular DNA viruses (such as Torque Teno Virus; TTV)40,71,72,89,90. Despite their prevalence and abundance, Anelloviridae effects on the host are not well understood. However, they are under immune control, as blood levels increase in organ transplantation and AIDS91. Anelloviridae levels typically drop before and during episodes of ACR in solid organ transplantation, which has led to interest in potentially using Anelloviridae as a functional measure of therapeutic immunosuppression40,71,89,92.
In the lung, altered peri-transplantation dynamics of Anelloviridae has been associated with primary graft dysfunction (PGD)71. As noted earlier, donor lungs have elevated Anelloviridae levels before procurement. Perhaps counterintuitively, subjects with PGD had smaller increases in Anelloviridae levels across the peri-transplant period. This may reflect immune mechanisms that both inhibit viral replication and injure tissue, or early tissue injury itself limiting viral replication. Interestingly, donor Anelloviridae variants were transplanted along with the allograft and disseminated systemically in the recipient, while variants present in recipient blood at time of transplant often migrated into the new lung72. Lung Anelloviridae levels remain elevated after transplant and correlate with the degree of bacterial dysbiosis, indicating inter-kingdom covariation40,72. The consequences of persistent Anelloviridae elevations and allograft/recipient virome intermixing remain to be determined.
In addition to the DNA viral landscape, RNA viruses are of great interest88, and some reports have described a high frequency of persistent RNA CARVs post-transplant, including influenza A and B, parainfluenza, and rhinovirus89. No association between viruses and ACR was identified, though BOS was not investigated.
Bacteriophages.
Bacteriophages (or phages) are abundant in the lungs of transplant recipients, particularly phages that infect bacterial inhabitants of the human respiratory tract71. Phages can influence bacterial pathogenesis in the lung, particularly in CF93. Also, emerging data suggest that phages may directly interact with human cells and modulate innate immune function94. However, what role phages play post-transplantation is unknown. Phages have also garnered interest as tools to fight multi-drug resistant bacterial infections such as Pseudomonas and Mycobacterium species, including in lung transplant recipients95,96. Therefore, understanding the lung bacteriophage landscape may contribute to future therapies in lung transplantation.
Novel viruses in the respiratory tract.
A limitation in interrogating the virome by deep-sequencing is the incomplete annotation of viral databases. Indeed, a sizable portion of sequences in viral metagenomic studies cannot be annotated, termed viral “dark matter.” Sometimes low-quality or partial sequence alignments are found to unexpected non-human viruses, leaving the significance uncertain. Recently, sequences were identified in lung transplant specimens that partially aligned with a porcine circovirus97. Through sequential contig-building and alignment, those sequences were determined to be a novel viral family termed Redondoviridae (named for their small, circular DNA genome)97. This newly-identified virus was found in human BAL and oropharyngeal samples but rarely from other sites or species, and appears to be the second most common DNA virus in such specimens (after Anelloviridae). Future studies are needed to determine what role these (and potentially additional) newly-discovered members of the human virome have in lung pathogenesis, including post-transplantation.
The lung mycobiome.
Based on fungal culture, Aspergillus and Candida are commonly identified in lung samples after transplantation8,46. Aspergillus is associated with early anastomotic complications, as it grows in ischemic anastomotic tissue98. Several studies have linked Aspergillus infection or colonization to later CLAD99,100, although recent work found no association101. Invasive Candida infections are common after lung transplantation, usually involving bloodstream, pleural space, or surgical site74,102.
Sequence-based investigation of fungal microbiome (mycobiome) can employ targeted amplification or shotgun sequencing. The first post-transplantation metagenomic analysis found fungi were highly variable between patients in absolute quantity and composition8,46. Candida was the most prevalent taxa, and when abundant in the lung it was also abundant in the oropharynx. This contrasted with healthy controls, where Candida was prevalent in oral samples but scarce in lung. Aspergillus was seen in BAL, sometimes at high levels, while other subjects had Cryptococcus and environmental fungi. Additionally, there was significant inter-kingdom covariation between Candida and Rothia, Streptococcus, and Veillonella46.
Deep-sequencing was recently applied to microbial biofilms on endobronchial stents used to treat airway complications in a predominantly post-lung transplant cohort103. Candida spp. were the most common fungal taxa in biofilms, followed by Aspergillus, which was particularly prevalent in stents placed to treat anastomotic dehiscence. Fungi generally considered environmental taxa (Scedosporium, Stereum, Sarcinomyces) were often seen at high absolute abundances. There was also evidence for fungal-bacterial co-variation and a link between stent material and biofilm type.
However, there are challenges to mycobiome analysis including issues of quantitative variability between samples, low absolute fungal biomass in many patients, sequence contamination, biases in ITS primers and database limitations46, and further studies are need to address the lung mycobiome and to relationship to clinical outcomes.
Mechanisms of Lung Microbiome-Host Interactions and Injury
Both microbial pathogen-associated molecular patterns (PAMPs) sensed by host pathogen-recognition receptors (PRRs) and microbial metabolites mediate microbiome-host interactions37, and it is essential to understand these interactions after transplant (Figure 2).
Microbiome modulation of inflammatory responses.
When a potential respiratory pathogen is identified by culture following transplantation, it is challenging to distinguish infection from colonization. One study of individuals suspected to have infection found the BAL microbiome profiles combined with cytokine signatures added useful context to culture data, and suggested that the microbiome modulated inflammatory responses104. Specifically, clinically-diagnosed pneumonia had low alpha-diversity, enrichment of potential pathogens, and elevated inflammatory cytokines. In contrast, in colonization (culture positive without clinical signs of infection) there were more oral commensals and an adaptive immune profile characterized by regulatory T-cells (Treg), decreased IL-17A, and increased IL-2 and IP-10. The authors postulated that commensal anaerobes activate Tregs, which constrain the inflammatory response and limit progression to pneumonia. Another report linked the relative abundance of Pseudomonas in allografts with neutrophilic inflammation and levels of the neutrophil chemoattractant CXCL8 (IL-8), whereas the presence of oral taxa (Streptococcus, Prevotella, Veillonella, Actinomyces) negatively correlated with CXCL8, as did allograft alpha-diversity53. These findings suggest that diverse communities comprised of predominantly URT taxa are less likely to promote neutrophilic inflammation and pneumonia. The post-transplant lung microbiome has also been linked to myeloid-derived suppressor cells (MDSCs) in BAL, which are bone marrow cells that home to the lung and modulate T-cell responses12. Multiple MDSC subtypes were found in the post-transplant lung, including pro-inflammatory and immunosuppressive phenotypes, and the MDSC subtype correlated with microbiome composition.
Additional insight comes from comparison of allografts versus native lungs of single lung transplant recipients with underlying ILD54. Compared to native lungs with ILD, allografts had higher microbial biomass and increased Acinetobacter and Pseudomonas, along with inflammatory cytokines and elevated acetylated proline-glycine-proline (Ac-PGP), a collagen breakdown product that increases neutrophilic inflammation and vascular permeability. Metabolome profiles correlated in particular with allograft Proteobacteria, implicating microbial-derived small molecules as potential drivers of inflammation. Together, these findings suggest that the lung allograft microbiome regulates local immune responses48,52,105.
Remodeling and fibrosis.
CLAD pathogenesis involves remodeling and fibrosis. Studies from the SysCLAD Consortium identified links between the post-transplant bacterial microbiome and host-response gene expression profiles14,15. BAL samples were characterized as pro-inflammatory (elevated TNF and cyclooxygenase-2 (COX2)), pro-remodeling (elevated platelet-derived growth factor-D (PDGF-D) and tissue inhibitors of metalloproteinases-1/matrix metalloproteinase-12 (TIMP1/MMP-12) ratio), or intermediate profiles15. The pro-inflammatory profile correlated with increased Firmicutes and Proteobacteria, peaked 3–4 months post-transplant, and had low macrophage abundance and elevated neutrophils. In contrast, the remodeling profile had increased Bacteroidetes, peaked about 12 months, and had more macrophages and fewer neutrophils. In vitro stimulation of monocytic cell lines with bacteria from these phyla recapitulated the inflammatory versus remodeling profiles. Another study extended those findings to address a fibroproliferative phenotype14. It identified an “anabolic profile” in 20% of post-transplant BAL samples characterized by increased host thrombospondin-1 and PDGF-D, which correlated with Prevotella, Streptococcus, Veillonella, and Neisseria. Conversely, a “catabolic profile” seen in 12% of subjects showed upregulated MMPs, collagen-V1 A-2, fibronectin-1, insulin-like growth factor 1 (IGF1), along with reduced microbial diversity, Staphylococcus, Corynebacterium, Stenotrophomonas, and Haemophilus, and culture-positive infections. In co-culture of macrophage and fibroblast cell lines, stimulation with Staphylococcus or Pseudomonas led to increased MMPs, consistent with the catabolic profile, whereas Prevotella or Streptococcus led to more collagen and fibronectin deposition, like the anabolic profile. These reports link distinct microbiome profiles to host fibroproliferation responses relevant to CLAD and provide tractable in vitro model systems to dissect host-microbe interactions.
Conclusions and future directions
The post-transplantation lung microbiome is dysbiotic, with markedly higher bacterial, fungal and viral abundance and altered composition compared to health. Emerging data suggest that the composition of bacterial communities is associated with adverse outcomes such as CLAD, although inconsistencies between reports limit conclusions, and the mycobiome and virome remain under-studied. Critical issues in understanding the microbiome in relation to outcomes include the need to account for patient-specificity, spatial heterogeneity, time-dependence, indications for sampling, and methodological differences. While data so far do not provide an answer about what might be an optimal or “healthy” post-transplantation lung microbiome, studies have begun to suggest links between microbiome composition and inflammation, fibrosis, and remodeling mechanisms relevant to both ACR and CLAD. As an emerging field, these promising early findings need future validation in high-quality clinical studies to establish links to outcomes as well as basic investigations to dissect host-microbiome mechanisms, which offer the potential for novel diagnostics and therapeutics after lung transplantation.
Future work is needed to examine the generalizability of the microbiome “types” seen in previous studies and determine what microbiome-related metrics might be operationalized as biomarkers for adverse outcome risk stratification or to guide therapeutic decisions. A second key area is to better understand the mechanisms of cross-talk between microbiome and host by which microbial constituents impact allograft outcomes. A third area needing additional research is the understudied virome and mycobiome.
Finally, a key frontier is targeting the respiratory tract microbiome for therapeutic purposes after lung transplantation. Perhaps the first step would be understanding how the microbiome influences or perhaps mediates responses to treatment with macrolides, such as reduced neutrophilic inflammation, and whether distinct microbiome profiles may be responsible for the clinical heterogeneity in response4,109. Beyond that, therapeutic targeting of the microbiome might take the form of supplementing communities (a “probiotic” approach)106, selectively reducing pro-inflammatory taxa (such as in phage therapy)94,95, or using microbially-mediated small molecules107,108 that have beneficial host-effects.
Acknowledgments
This work was supported by NIH grants R01-HL113252 and R61/33-HL137063 to RGC, and JEM was supported by KL2-TR001879.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
REFERENCES
- 1.Chambers DC, Cherikh WS, Harhay MO, et al. The International Thoracic Organ Transplant Registry of the International Society for Heart and Lung Transplantation: Thirty-sixth adult lung and heart–lung transplantation Report—2019; Focus theme: Donor and recipient size match. J Hear Lung Transplant. 2019;38(10):1042–1055. doi: 10.1016/j.healun.2019.08.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Verleden GM, Glanville AR, Lease ED, et al. Chronic lung allograft dysfunction: Definition, diagnostic criteria, and approaches to treatment―A consensus report from the Pulmonary Council of the ISHLT. J Hear Lung Transplant. 2019;38(5):493–503. doi: 10.1016/j.healun.2019.03.009 [DOI] [PubMed] [Google Scholar]
- 3.Valentine VG, Gupta MR, Walker JE, et al. Effect of Etiology and Timing of Respiratory Tract Infections on Development of Bronchiolitis Obliterans Syndrome. J Hear Lung Transplant. 2009;28(2):163–169. doi: 10.1016/j.healun.2008.11.907 [DOI] [PubMed] [Google Scholar]
- 4.Corris PA, Ryan VA, Small T, et al. A randomised controlled trial of azithromycin therapy in bronchiolitis obliterans syndrome (BOS) post lung transplantation. Thorax. 2015;70(5):442–450. doi: 10.1136/thoraxjnl-2014-205998 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Marchesi JR, Ravel J. The vocabulary of microbiome research: a proposal. Microbiome. 2015;3(1). doi: 10.1186/s40168-015-0094-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Berg G, Rybakova D, Fischer D, et al. Microbiome definition re-visited: old concepts and new challenges. Microbiome. 2020;8(1):103. doi: 10.1186/s40168-020-00875-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Wilson KH, Blitchington RB. Human colonic biota studied by ribosomal DNA sequence analysis. Appl Environ Microbiol. 1996;62(7):2273–2278. doi: 10.1128/aem.62.7.2273-2278.1996 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Charlson ES, Diamond JM, Bittinger K, et al. Lung-enriched organisms and aberrant bacterial and fungal respiratory microbiota after lung transplant. Am J Respir Crit Care Med. 2012;186(6):536–545. doi: 10.1164/rccm.201204-0693OC [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Dickson RP, Erb-Downward JR, Freeman CM, et al. Changes in the Lung Microbiome following Lung Transplantation Include the Emergence of Two Distinct Pseudomonas Species with Distinct Clinical Associations. Davis IC, ed. PLoS One. 2014;9(5):e97214. doi: 10.1371/journal.pone.0097214 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Willner DL, Hugenholtz P, Yerkovich ST, et al. Reestablishment of Recipient-associated Microbiota in the Lung Allograft Is Linked to Reduced Risk of Bronchiolitis Obliterans Syndrome. Am J Respir Crit Care Med. 2013;187(6):640–647. doi: 10.1164/rccm.201209-1680OC [DOI] [PubMed] [Google Scholar]
- 11.Borewicz K, Pragman AA, Kim HB, Hertz M, Wendt C, Isaacson RE. Longitudinal analysis of the lung microbiome in lung transplantation. FEMS Microbiol Lett. 2013;339(1):57–65. doi: 10.1111/1574-6968.12053 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Sharma NS, Wille KM, Athira S, et al. Distal airway microbiome is associated with immunoregulatory myeloid cell responses in lung transplant recipients. J Hear Lung Transplant. 2018;37(2):206–216. doi: 10.1016/j.healun.2017.07.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Schott C, Weigt SS, Turturice BA, et al. Bronchiolitis obliterans syndrome susceptibility and the pulmonary microbiome. J Hear Lung Transplant. 2018;37(9):1131–1140. doi: 10.1016/j.healun.2018.04.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Mouraux S, Bernasconi E, Pattaroni C, et al. Airway microbiota signals anabolic and catabolic remodeling in the transplanted lung. J Allergy Clin Immunol. 2018;141(2):718–729.e7. doi: 10.1016/j.jaci.2017.06.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Bernasconi E, Pattaroni C, Koutsokera A, et al. Airway Microbiota Determines Innate Cell Inflammatory or Tissue Remodeling Profiles in Lung Transplantation. Am J Respir Crit Care Med. 2016;194(10):1252–1263. doi: 10.1164/rccm.201512-2424OC [DOI] [PubMed] [Google Scholar]
- 16.Consortium HMP, Huttenhower C, Human Microbiome Project Consortium. Structure, function and diversity of the healthy human microbiome. Nature. 2012;486(7402):207–214. doi: 10.1038/nature11234 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Olsen GJ, Woese CR. Ribosomal RNA: a key to phylogeny. FASEB J. Published online 1993. doi: 10.1096/fasebj.7.1.8422957 [DOI] [PubMed] [Google Scholar]
- 18.Lane DJ, Pace B, Olsen GJ, Stahl DA, Sogin ML, Pace NR. Rapid determination of 16S ribosomal RNA sequences for phylogenetic analyses. Proc Natl Acad Sci. 1985;82(20):6955–6959. doi: 10.1073/pnas.82.20.6955 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Pace NR. A molecular view of microbial diversity and the biosphere. Science (80- ). 1997;276(5313):734–740. https://www.ncbi.nlm.nih.gov/pubmed/9115194 [DOI] [PubMed] [Google Scholar]
- 20.Woese CR. Interpreting the universal phylogenetic tree. Proc Natl Acad Sci U S A. 2000;97(15):8392–8396. doi: 10.1073/pnas.97.15.8392 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Morgan XC, Huttenhower C. Chapter 12: Human microbiome analysis. PLoS Comput Biol. 2012;8(12):e1002808. doi: 10.1371/journal.pcbi.1002808 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Kim D, Hofstaedter CE, Zhao C, et al. Optimizing methods and dodging pitfalls in microbiome research. Microbiome. 2017;5(1):52. doi: 10.1186/s40168-017-0267-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Clarke EL, Lauder AP, Hofstaedter CE, et al. Microbial lineages in sarcoidosis a metagenomic analysis tailored for low-microbial content samples. Am J Respir Crit Care Med. 2018;197(2):225–234. doi: 10.1164/rccm.201705-0891OC [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Marotz CA, Sanders JG, Zuniga C, Zaramela LS, Knight R, Zengler K. Improving saliva shotgun metagenomics by chemical host DNA depletion. Microbiome. 2018;6(1):42. doi: 10.1186/s40168-018-0426-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Gu W, Crawford ED, O’Donovan BD, et al. Depletion of Abundant Sequences by Hybridization (DASH): using Cas9 to remove unwanted high-abundance species in sequencing libraries and molecular counting applications. Genome Biol. 2016;17(1):41. doi: 10.1186/s13059-016-0904-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Mac Aogáin M, Lau KJX, Cai Z, et al. Metagenomics Reveals a Core Macrolide Resistome Related to Microbiota in Chronic Respiratory Disease. Am J Respir Crit Care Med. 2020;202(3):433–447. doi: 10.1164/rccm.201911-2202OC [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Dickson RP, Erb-Downward JR, Freeman CM, et al. Bacterial Topography of the Healthy Human Lower Respiratory Tract. MBio. 2017;8(1):1–12. doi: 10.1128/mBio.02287-16 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Charlson ES, Bittinger K, Haas AR, et al. Topographical Continuity of Bacterial Populations in the Healthy Human Respiratory Tract. Am J Respir Crit Care Med. 2011;184(8):957–963. doi: 10.1164/rccm.201104-0655OC [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Salter SJ, Cox MJ, Turek EM, et al. Reagent and laboratory contamination can critically impact sequence-based microbiome analyses. BMC Biol. 2014;12:87. doi: 10.1186/s12915-014-0087-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Dickson RP, Erb-Downward JR, Freeman CM, et al. Spatial variation in the healthy human lung microbiome and the adapted island model of lung biogeography. Ann Am Thorac Soc. 2015;12(6):821–830. doi: 10.1513/AnnalsATS.201501-029OC [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Charlson ES, Bittinger K, Chen J, et al. Assessing Bacterial Populations in the Lung by Replicate Analysis of Samples from the Upper and Lower Respiratory Tracts. Highlander SK, ed. PLoS One. 2012;7(9):e42786. doi: 10.1371/journal.pone.0042786 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Glendinning L, Collie D, Wright S, Rutherford KMD, McLachlan G. Comparing microbiotas in the upper aerodigestive and lower respiratory tracts of lambs. Microbiome. 2017;5(1):145. doi: 10.1186/s40168-017-0364-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Bassis CM, Erb-Downward JR, Dickson RP, et al. Analysis of the Upper Respiratory Tract Microbiotas as the Source of the Lung and Gastric Microbiotas in Healthy Individuals. Ravel J, ed. MBio. 2015;6(2):e00037. doi: 10.1128/mBio.00037-15 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Ubags NDJJ Marsland BJ. Mechanistic insight into the function of the microbiome in lung diseases. Eur Respir J. 2017;50(3):1602467. doi: 10.1183/13993003.02467-2016 [DOI] [PubMed] [Google Scholar]
- 35.Thaiss CA, Zmora N, Levy M, Elinav E. The microbiome and innate immunity. Nature. 2016;535(7610):65–74. doi: 10.1038/nature18847 [DOI] [PubMed] [Google Scholar]
- 36.Cribbs SK, Crothers K, Morris A. Pathogenesis of HIV-Related Lung Disease: Immunity, Infection, and Inflammation. Physiol Rev. 2020;100(2):603–632. doi: 10.1152/physrev.00039.2018 [DOI] [PubMed] [Google Scholar]
- 37.Hakansson AP, Orihuela CJ, Bogaert D. Bacterial-host interactions: Physiology and pathophysiology of respiratory infection. Physiol Rev. 2018;98(2):781–811. doi: 10.1152/physrev.00040.2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Ren L, Zhang R, Rao J, et al. Transcriptionally Active Lung Microbiome and Its Association with Bacterial Biomass and Host Inflammatory Status. mSystems. 2018;3(5). doi: 10.1128/msystems.00199-18 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Dickson RP, Erb-Downward JR, Martinez FJ, Huffnagle GB. The Microbiome and the Respiratory Tract. Annu Rev Physiol. 2016;78(1):481–504. doi: 10.1146/annurev-physiol-021115-105238 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Young JC, Chehoud C, Bittinger K, et al. Viral metagenomics reveal blooms of anelloviruses in the respiratory tract of lung transplant recipients. Am J Transpl. 2015;15(1):200–209. doi: 10.1111/ajt.13031 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Tipton L, Ghedin E, Morris A. The lung mycobiome in the next-generation sequencing era. Virulence. 2017;8(3):334–341. doi: 10.1080/21505594.2016.1235671 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Spandole S, Cimponeriu D, Berca LM, Mihăescu G. Human anelloviruses: an update of molecular, epidemiological and clinical aspects. Arch Virol. 2015;160(4):893–908. doi: 10.1007/s00705-015-2363-9 [DOI] [PubMed] [Google Scholar]
- 43.Mac Aogáin M, Chandrasekaran R, Lim AYH, et al. Immunological corollary of the pulmonary mycobiome in bronchiectasis: the CAMEB study. Eur Respir J. 2018;52(1):1800766. doi: 10.1183/13993003.00766-2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Cui L, Lucht L, Tipton L, et al. Topographic Diversity of the Respiratory Tract Mycobiome and Alteration in HIV and Lung Disease. Am J Respir Crit Care Med. 2015;191(8):932–942. doi: 10.1164/rccm.201409-1583OC [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Tipton L, Müller CL, Kurtz ZD, et al. Fungi stabilize connectivity in the lung and skin microbial ecosystems. Microbiome. 2018;6(1):1–14. doi: 10.1186/s40168-017-0393-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Bittinger K, Charlson ES, Loy E, et al. Improved characterization of medically relevant fungi in the human respiratory tract using next-generation sequencing. Genome Biol. 2014;15(10):487. doi: 10.1186/s13059-014-0487-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Iwasaki A, Foxman EF, Molony RD. Early local immune defences in the respiratory tract. Nat Rev Immunol. 2017;17(1):7–20. doi: 10.1038/nri.2016.117 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Segal LN, Clemente JC, Tsay J-CJ, et al. Enrichment of the lung microbiome with oral taxa is associated with lung inflammation of a Th17 phenotype. Nat Microbiol. 2016;1(5):16031. doi: 10.1038/nmicrobiol.2016.31 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Dang AT, Marsland BJ. Microbes, metabolites, and the gut–lung axis. Mucosal Immunol. 2019;12(4):843–850. doi: 10.1038/s41385-019-0160-6 [DOI] [PubMed] [Google Scholar]
- 50.Barcik W, Boutin RCT, Sokolowska M, Finlay BB. The Role of Lung and Gut Microbiota in the Pathology of Asthma. Immunity. 2020;52(2):241–255. doi: 10.1016/j.immuni.2020.01.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Blacher E, Levy M, Tatirovsky E, Elinav E. Microbiome-Modulated Metabolites at the Interface of Host Immunity. J Immunol. 2017;198(2):572–580. doi: 10.4049/jimmunol.1601247 [DOI] [PubMed] [Google Scholar]
- 52.Dickson RP, Erb-Downward JR, Falkowski NR, Hunter EM, Ashley SL, Huffnagle GB. The Lung Microbiota of Healthy Mice Are Highly Variable, Cluster by Environment, and Reflect Variation in Baseline Lung Innate Immunity. Am J Respir Crit Care Med. 2018;198(4):497–508. doi: 10.1164/rccm.201711-2180OC [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Spence CD, Vanaudenaerde B, Einarsson GG, et al. Influence of azithromycin and allograft rejection on the post–lung transplant microbiota. J Hear Lung Transplant. 2020;39(2):176–183. doi: 10.1016/j.healun.2019.11.007 [DOI] [PubMed] [Google Scholar]
- 54.Sharma NS, Vestal G, Wille K, et al. Differences in airway microbiome and metabolome of single lung transplant recipients. Respir Res. 2020;21(1). doi: 10.1186/s12931-020-01367-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Petersen C, Round JL. Defining dysbiosis and its influence on host immunity and disease. Cell Microbiol. 2014;16(7):1024–1033. doi: 10.1111/cmi.12308 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Pletcher SD, Goldberg AN, Cope EK. Loss of Microbial Niche Specificity Between the Upper and Lower Airways in Patients With Cystic Fibrosis. Laryngoscope. 2019;129(3):544–550. doi: 10.1002/lary.27454 [DOI] [PubMed] [Google Scholar]
- 57.Syed SA, Whelan FJ, Waddell B, Rabin HR, Parkins MD, Surette MG. Reemergence of Lower-Airway Microbiota in Lung Transplant Patients with Cystic Fibrosis. Ann Am Thorac Soc. 2016;13(12):2132–2142. doi: 10.1513/AnnalsATS.201606-431OC [DOI] [PubMed] [Google Scholar]
- 58.Beaume M, Köhler T, Greub G, et al. Rapid adaptation drives invasion of airway donor microbiota by Pseudomonas after lung transplantation. Sci Rep. 2017;7:40309. doi: 10.1038/srep40309 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Yadava K, Pattaroni C, Sichelstiel AK, et al. Microbiota Promotes Chronic Pulmonary Inflammation by Enhancing IL-17A and Autoantibodies. Am J Respir Crit Care Med. 2016;193(9):975–987. doi: 10.1164/rccm.201504-0779OC [DOI] [PubMed] [Google Scholar]
- 60.Einarsson GG, Comer DM, McIlreavey L, et al. Community dynamics and the lower airway microbiota in stable chronic obstructive pulmonary disease, smokers and healthy non-smokers. Thorax. 2016;71(9):795–803. doi: 10.1136/thoraxjnl-2015-207235 [DOI] [PubMed] [Google Scholar]
- 61.Molyneaux PL, Willis-Owen SAG, Cox MJ, et al. Host–Microbial Interactions in Idiopathic Pulmonary Fibrosis. Am J Respir Crit Care Med. 2017;195(12):1640–1650. doi: 10.1164/rccm.201607-1408OC [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Invernizzi R, Wu BG, Barnett J, et al. The Respiratory Microbiome in Chronic Hypersensitivity Pneumonitis is Distinct from that of Idiopathic Pulmonary Fibrosis. Am J Respir Crit Care Med. Published online 2020. doi: 10.1164/rccm.202002-0460OC [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Wood RK. Esophageal Dysmotility, Gastro-esophageal Reflux Disease, and Lung Transplantation: What Is the Evidence? Curr Gastroenterol Rep. 2015;17(12):48. doi: 10.1007/s11894-015-0474-9 [DOI] [PubMed] [Google Scholar]
- 64.Baumann B, Byers S, Wasserman-Wincko T, et al. Postoperative Swallowing Assessment After Lung Transplantation. Ann Thorac Surg. 2017;104(1):308–312. doi: 10.1016/j.athoracsur.2017.01.080 [DOI] [PubMed] [Google Scholar]
- 65.Simon-Soro A, Sohn MB, McGinniss JE, et al. Upper Respiratory Dysbiosis with a Facultative-dominated Ecotype in Advanced Lung Disease and Dynamic Change after Lung Transplant. Ann Am Thorac Soc. 2019;16(11):1383–1391. doi: 10.1513/AnnalsATS.201904-299OC [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Snell G, Hiho S, Levvey B, Sullivan L, Westall G. Consequences of donor-derived passengers (pathogens, cells, biological molecules and proteins) on clinical outcomes. J Hear Lung Transplant. 2019;000:1–5. doi: 10.1016/j.healun.2019.06.019 [DOI] [PubMed] [Google Scholar]
- 67.Kelly BJ, Imai I, Bittinger K, et al. Composition and dynamics of the respiratory tract microbiome in intubated patients. Microbiome. 2016;4:1–13. doi: 10.1186/s40168-016-0151-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Dickson RP, Schultz MJ, Van Der Poll T, et al. Lung Microbiota Predict Clinical Outcomes in Critically Ill Patients. Am J Respir Crit Care Med. 2020;201:555–563. doi: 10.1164/rccm.201907-1487OC [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Kitsios GD, Fitch A, Manatakis DV., et al. Respiratory Microbiome Profiling for Etiologic Diagnosis of Pneumonia in Mechanically Ventilated Patients. Front Microbiol. 2018;9(July):1–15. doi: 10.3389/fmicb.2018.01413 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Langelier C, Kalantar KL, Moazed F, et al. Integrating host response and unbiased microbe detection for lower respiratory tract infection diagnosis in critically ill adults. Proc Natl Acad Sci U S A. Published online 2018. doi: 10.1073/pnas.1809700115 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Abbas AA, Diamond JM, Chehoud C, et al. The Perioperative Lung Transplant Virome: Torque Teno Viruses Are Elevated in Donor Lungs and Show Divergent Dynamics in Primary Graft Dysfunction. Am J Transpl. 2017;17(5):1313–1324. doi: 10.1111/ajt.14076 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Abbas AA, Young JC, Clarke EL, et al. Bidirectional transfer of Anelloviridae lineages between graft and host during lung transplantation. Am J Transpl. 2019;19(4):1086–1097. doi: 10.1111/ajt.15116 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Hayanga JWA, D’Cunha J. The surgical technique of bilateral sequential lung transplantation. J Thorac Dis. 2014;6(8):1063. doi: 10.3978/J.ISSN.2072-1439.2014.07.02 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Remund KF, Best M, Egan JJ. Infections relevant to lung transplantation. Proc Am Thorac Soc. 2009;6(1):94–100. doi: 10.1513/pats.200809-113GO [DOI] [PubMed] [Google Scholar]
- 75.De Vlaminck I, Khush KK, Strehl C, et al. XTemporal response of the human virome to immunosuppression and antiviral therapy. Cell. 2013;155(5):1178. doi: 10.1016/j.cell.2013.10.034 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Ahya VN, Diamond JM. Lung Transplantation. Med Clin North Am. 2019;103(3):425–433. doi: 10.1016/j.mcna.2018.12.003 [DOI] [PubMed] [Google Scholar]
- 77.Lanza VF, Baquero F, Martínez JL, et al. In-depth resistome analysis by targeted metagenomics. Microbiome. 2018;6(1):11. doi: 10.1186/s40168-017-0387-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Abeles SR, Jones MB, Santiago-Rodriguez TM, et al. Microbial diversity in individuals and their household contacts following typical antibiotic courses. Microbiome. 2016;4(1):39. doi: 10.1186/s40168-016-0187-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Vandeplassche E, Tavernier S, Coenye T, Crabbé A. Influence of the lung microbiome on antibiotic susceptibility of cystic fibrosis pathogens. Eur Respir Rev. 2019;28(152):190041. doi: 10.1183/16000617.0041-2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Segal LN, Clemente JC, Wu BG, et al. Randomised, double-blind, placebo-controlled trial with azithromycin selects for anti-inflammatory microbial metabolites in the emphysematous lung. Thorax. 2017;72(1):13–22. doi: 10.1136/thoraxjnl-2016-208599 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Taylor SL, Leong LEX, Mobegi FM, et al. Long-Term Azithromycin Reduces Haemophilus influenzae and Increases Antibiotic Resistance in Severe Asthma. Am J Respir Crit Care Med. Published online 2019. doi: 10.1164/rccm.201809-1739OC [DOI] [PubMed] [Google Scholar]
- 82.Botha P, Archer L, Anderson RL, et al. Pseudomonas aeruginosa colonization of the allograft after lung transplantation and the risk of bronchiolitis obliterans syndrome. Transplantation. 2008;85(5):771–774. doi: 10.1097/TP.0b013e31816651de [DOI] [PubMed] [Google Scholar]
- 83.Gregson AL, Wang X, Weigt SS, et al. Interaction between Pseudomonas and CXC Chemokines Increases Risk of Bronchiolitis Obliterans Syndrome and Death in Lung Transplantation. Am J Respir Crit Care Med. 2013;187(5):518–526. doi: 10.1164/rccm.201207-1228OC [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Vos R, Vanaudenaerde BM, Geudens N, Dupont LJ, Van Raemdonck DE, Verleden GM. Pseudomonal airway colonisation: risk factor for bronchiolitis obliterans syndrome after lung transplantation? Eur Respir J. 2008;31(5):1037–1045. doi: 10.1183/09031936.00128607 [DOI] [PubMed] [Google Scholar]
- 85.Moore CA, Pilewski JM, Venkataramanan R, et al. Effect of aerosolized antipseudomonals on Pseudomonas positivity and bronchiolitis obliterans syndrome after lung transplantation. Transpl Infect Dis. 2017;19(3):e12688. doi: 10.1111/tid.12688 [DOI] [PubMed] [Google Scholar]
- 86.Beaume M, Lazarevic V, Köhler T, et al. Microbial communities of conducting and respiratory zones of lung-transplanted patients. Front Microbiol. 2016;7(NOV):1749. doi: 10.3389/fmicb.2016.01749 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Combs MP, Wheeler DS, Luth JE, et al. Lung microbiota predict chronic rejection in healthy lung transplant recipients: a prospective cohort study. Lancet Respir Med. 2021;2600(20):1–12. doi: 10.1016/s2213-2600(20)30405-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Peghin M, Los-Arcos I, Hirsch HH, et al. Community-acquired Respiratory Viruses Are a Risk Factor for Chronic Lung Allograft Dysfunction. Clin Infect Dis. 2019;69(7):1192–1197. doi: 10.1093/cid/ciy1047 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Mitchell AB, Mourad B, Morgan LC, Oliver BGGG, Glanville AR. Transplanting the pulmonary virome: Dynamics of transient populations. J Hear Lung Transplant. 2018;37(9):1111–1118. doi: 10.1016/j.healun.2018.06.002 [DOI] [PubMed] [Google Scholar]
- 90.Segura-Wang M, Görzer I, Jaksch P, Puchhammer-Stöckl E. Temporal dynamics of the lung and plasma viromes in lung transplant recipients. Attoui H, ed. PLoS One. 2018;13(7):e0200428. doi: 10.1371/journal.pone.0200428 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Li L, Deng X, Linsuwanon P, et al. AIDS Alters the Commensal Plasma Virome. J Virol. 2013;87(19):10912–10915. doi: 10.1128/jvi.01839-13 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Frye BC, Bierbaum S, Falcone V, et al. Kinetics of Torque Teno Virus-DNA Plasma Load Predict Rejection in Lung Transplant Recipients. Transplantation. 2019;103(4):815–822. doi: 10.1097/TP.0000000000002436 [DOI] [PubMed] [Google Scholar]
- 93.Burgener EB, Sweere JM, Bach MS, et al. Filamentous bacteriophages are associated with chronic Pseudomonas lung infections and antibiotic resistance in cystic fibrosis. Sci Transl Med. 2019;11(488). doi: 10.1126/scitranslmed.aau9748 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Sweere JM, Van Belleghem JD, Ishak H, et al. Bacteriophage trigger antiviral immunity and prevent clearance of bacterial infection HHS Public Access. Science (80- ). 2019;363(6434). doi: 10.1126/science.aat9691 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Crunkhorn S. Phage therapy for Mycobacterium abscessus. Nat Rev Drug Discov. 2019;18(7):500. doi: 10.1038/d41573-019-00097-6 [DOI] [PubMed] [Google Scholar]
- 96.Aslam S, Courtwright AM, Koval C, et al. Early clinical experience of bacteriophage therapy in 3 lung transplant recipients. Am J Transplant. 2019;19(9):2631–2639. doi: 10.1111/ajt.15503 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Abbas AA, Taylor LJ, Dothard MI, et al. Redondoviridae, a Family of Small, Circular DNA Viruses of the Human Oro-Respiratory Tract Associated with Periodontitis and Critical Illness. Cell Host Microbe. 2019;25(5):719–729.e4. doi: 10.1016/j.chom.2019.04.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Crespo MM, McCarthy DP, Hopkins PM, et al. ISHLT Consensus Statement on adult and pediatric airway complications after lung transplantation: Definitions, grading system, and therapeutics. J Hear Lung Transplant. Published online 2018. doi: 10.1016/j.healun.2018.01.1309 [DOI] [PubMed] [Google Scholar]
- 99.Weigt SS, Elashoff RM, Huang C, et al. Aspergillus Colonization of the Lung Allograft Is a Risk Factor for Bronchiolitis Obliterans Syndrome. Am J Transpl. 2009;9(8):1903–1911. doi: 10.1111/j.1600-6143.2009.02635.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Weigt SS, Copeland CAF, Derhovanessian A, et al. Colonization with small conidia Aspergillus species is associated with bronchiolitis obliterans syndrome: a two-center validation study. Am J Transplant. 2013;13(4):919–927. doi: 10.1111/ajt.12131 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Law N, Hamandi B, Fegbeutel C, et al. Lack of association of Aspergillus colonization with the development of bronchiolitis obliterans syndrome in lung transplant recipients: An international cohort study. J Heart Lung Transplant. 2019;38(9):963–971. doi: 10.1016/j.healun.2019.06.007 [DOI] [PubMed] [Google Scholar]
- 102.Baker AW, Maziarz EK, Arnold CJ, et al. Invasive Fungal Infection After Lung Transplantation: Epidemiology in the Setting of Antifungal Prophylaxis. Clin Infect Dis. 2020;70(1):30. doi: 10.1093/CID/CIZ156 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.McGinniss JEJE, Imai I, Simon-Soro A, et al. Molecular analysis of the endobronchial stent microbial biofilm reveals bacterial communities that associate with stent material and frequent fungal constituents. Chaturvedi V, ed. PLoS One. 2019;14(5):e0217306. doi: 10.1371/journal.pone.0217306 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Shankar J, Nguyen MH, Crespo MM, et al. Looking Beyond Respiratory Cultures: Microbiome-Cytokine Signatures of Bacterial Pneumonia and Tracheobronchitis in Lung Transplant Recipients. Am J Transplant. 2016;16(6):1766–1778. doi: 10.1111/ajt.13676 [DOI] [PubMed] [Google Scholar]
- 105.O’Dwyer DN, Zhou X, Wilke CA, et al. Lung Dysbiosis, Inflammation, and Injury in Hematopoietic Cell Transplantation. Am J Respir Crit Care Med. 2018;198(10):1312–1321. doi: 10.1164/rccm.201712-2456OC [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Spacova I, Petrova MI, Fremau A, et al. Intranasal administration of probiotic Lactobacillus rhamnosus GG prevents birch pollen-induced allergic asthma in a murine model. Allergy. 2019;74(1):100–110. doi: 10.1111/all.13502 [DOI] [PubMed] [Google Scholar]
- 107.Martinez KB, Leone VA, Chang EB. Microbial Metabolites in Health and Disease: Navigating the Unknown in Search of Function. J Biol Chem. Published online 2017. doi: 10.1074/jbc.R116.752899 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Reisdorph NA, Cruickshank-Quinn C, Nkrumah-Elie Y, Reisdorph R. Application of metabolomics in lung research. In: Methods in Molecular Biology. Vol 1809. Humana Press Inc.; 2018:263–288. doi: 10.1007/978-1-4939-8570-8_17 [DOI] [PubMed] [Google Scholar]
- 109.Verleden SE, Spence C, Einarsson G, et al. The impact of azithromycin on the post lung transplant microbiome. J Hear Lung Transplant. 2018;37(4):S455. doi: 10.1016/j.healun.2018.01.1184 [DOI] [Google Scholar]