Skip to main content

REVIEW article

Front. Pediatr., 04 February 2020
Sec. Neonatology
Volume 7 - 2019 | https://doi.org/10.3389/fped.2019.00564

Predicting BPD: Lessons Learned From the Airway Microbiome of Preterm Infants

  • Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, United States

Bronchopulmonary dysplasia (BPD) is the chronic lung disease of prematurity with an operational definition, various different clinical phenotypes, and a complex, multifactorial etiology. Newer unbiased systems biology approaches have identified various “omic” factors associated with the pathogenesis and prediction of BPD. Recent microbi “omic” studies have discovered that airways of newborns harbor a low biomass but distinct microbiome signature as early as at the time of birth. This early airway microbiome may serve to prime the host immune system and may play a role in modulating the infant's future susceptibility to severe BPD development. Temporal changes are observed in airway microbiome of preterm infants from birth to the diagnosis of BPD, with an overall decrease in bacterial diversity, and development of a relative dysbiosis marked by increased Gammaproteobacteria and decreased Lactobacilli abundance. This review will summarize previous investigations of the airway microbiome in preterm infants, appraise the utility of using the airway microbiome to predict BPD development, discuss possible molecular mechanisms involved, and speculate on future microbiome-mediated therapeutics for BPD.

Introduction

Distinct microbial populations exist throughout the human body and have been the subject of investigation relating to human disease pathogenesis, susceptibility, and progression. The airway microbiome has been studied in the context of multiple pulmonary diseases including chronic obstructive pulmonary disease, asthma, and cystic fibrosis (14). Bronchopulmonary dysplasia (BPD), the most common chronic lung disease of prematurity, may result from lung injury due to a range of factors including infection, respiratory support, edema, and oxygen toxicity. Recent investigations have described differences in the airway microbiome of preterm infants that may influence susceptibility of BPD development, suggesting another parallel pathway contributory to abnormal lung development. In this manuscript, we review the potential origins of the airway microbiome, plausible covariates that influence the airway microbiome, investigations exploring associations between microbial communities and BPD susceptibility, potential mechanisms for microbiome-derived lung maldevelopment, and future microbiome-based targeted therapeutic approaches.

Origins of the Infant Airway Microbiome

How, when, and from what source(s) does the infant's airway microbiome originate? It is unclear when the airway microbiome is first established. Study of the in utero and early postnatal microbiome suggests the possibility of a fetal microbiome preceding birth. Ureaplasma and Mycoplasma have been reported invaders of the amniotic cavity associated with intraamniotic infection and preterm labor (5, 6) and have been associated with BPD development and thus therapeutically targeted (7, 8). A distinct placental microbiome has previously been described and most closely resembles the oral microbiome (9). More recent investigations of the placental microbiome report that the placental microbiome is of low abundance and low biomass (10, 11), but a very recent report from De Goffau et al. (12) refutes this claim. According to De Goffau's study, placenta does not harbor a microbiome but may contain potential pathogens. In healthy, term pregnancies, the amniotic cavity likely remains sterile until amniotic membrane rupture (13). Bacteria have also been detected from infant meconium samples (14) and cord blood (15), further suggesting that the fetal environment may not be sterile.

Our group conducted a study in which tracheal aspirate samples from infants were collected at birth or within 6 h of birth, and each sample was found to harbor a diverse microbiome signature (16). As majority of these samples were collected right at birth, most soon after cesarean sections, we speculate that airway microbiome signature at birth could have fetal origins. Previous studies of mother–infant dyads have identified similarities between the microbiome of the amniotic fluid and infant meconium highly abundant in Proteobacteria and Firmicutes, followed by an increase in diversity and later resemblance between the breast milk and infants' intestinal microbiome (17). The placental microbiome reported by prior investigations (9), largely composed of Proteobacteria and Firmicutes, also resembles that of the airway microbiome in infants at birth that our group has reported (16). However, studies of mother–infant dyads have not concurrently investigated the amniotic fluid, placenta, and airway microbiome at birth, and the actual origin of the early airway microbiome remains unknown.

What Factors Influence the Airway Microbiome?

Before discussion of the airway microbiome and BPD susceptibility, it is critical to consider other clinical covariates that may influence infants' airway microbiome. A common variate of investigation has been antibiotic exposure. Intrapartum antibiotics have been reported to influence the oral microbiome of infants, including a reduction in the abundance of Lactobacillus (18). However, given the high risk for infection in the preterm population, antibiotics are a frequent exposure making such analyses challenging. In a longitudinal study of the airway microbiome from tracheal aspirates in preterm infants, all infants were exposed to antibiotics within the first 21 postnatal days with a median duration of 12 exposure days (19). Other studies in which fewer infants were exposed to antibiotics did not report differences in bacterial diversity between exposed and unexposed infants (20). We have previously reported a similar airway microbiome at birth in infants, regardless of prenatal antibiotic exposure. Additionally, despite multiple courses of antibiotic exposure, we observed comparable dysbiotic changes among infants that developed BPD (16).

Given that cesarean delivery may preclude exposure to vaginal flora, the influence of delivery mode has been evaluated. Studies analyzing the gut microbiome have demonstrated longitudinal differences persisting for a year after birth in cesarean born infants, including a decrease in Bacteroides abundance (21). Our group has reported that the airway microbiome did not differ based on whether infants were born via vaginal or cesarean delivery mode (16). In another study, infants born by vaginal delivery had an airway microbiome representative of vaginal flora, while the airway microbiome of infants born via cesarean delivery resembled the skin flora (22).

Breast milk influences the maturation of the intestinal microbiome with little evidence analyzing the influence of breast milk exposure and the airway microbiome. A study of infant–mother dyads (N = 107) compared the microbiome of the infant stool, breast milk, and areolar skin. While these microbial communities were distinct, the infant gut microbiome more closely resembled the maternal breast milk and areolar microbiome compared to randomly chosen comparison groups (23). Whereas, the intestinal microbiome may not seem relevant to pulmonary disease, correlations between the gut microbiome and lung diseases of childhood including asthma and cystic fibrosis have previously been reported (24, 25). Interestingly, breastfed infants have more abundant Lactobacilli and less abundant Proteobacteria compared to formula-fed infants (21), a similar differentiating feature of BPD-resistant infants compared to BPD-susceptible infants (16). Additionally, a meta-analysis comparing human milk to formula milk found decreased odds of BPD development in human milk-exposed infants (OR 0.78; 95% CI 0.68–0.88); however, the quality of evidence was low (26). More evidence is needed relating human milk to the airway microbiome given the overlap in microbial composition observed in breast milk-fed infants and BPD-resistant infants.

The Airway Microbiome at Birth

Given the possibility that bacteria may be present in the fetal environment, multiple investigators have described the airway microbiome at birth. However, characterizing the airway microbiome of infants presents several challenges including inaccessibility of the lower airway, the requirement of an invasive sampling technique (typically endotracheal intubation), and a low biomass necessitating molecular techniques susceptible to contamination. Despite these challenges, bacterial DNA have been observed in tracheal aspirates collected at the time of intubation in preterm infants (20) with changes in diversity occurring after the third postnatal day (19). From these studies, Proteobacteria (Acinetobacter spp.) and Firmicutes were the predominant phyla and Staphylococcus, Ureaplasma parvum, and Ureaplasma urealyticum were the most frequently identified organisms. Other observational studies of tracheal aspirates in preterm infants have found a similar predominance of Staphylococcus and Ureaplasma (27, 28). In isolates taken at birth or within 6 h of birth, we have reported a low biomass airway microbiome in both term and preterm infants. Firmicutes and Proteobacteria were predominant bacterial phyla with additional detection of Actinobacteria, Bacteroidetes, Tenericutes, Fusobacterium, Cyanobacteria, and Verrucomicrobia (16).

Airway Microbial Signatures and BPD Development

Before the plausible contributions of the airway microbiome to BPD development are discussed, it is important to acknowledge that BPD is a not a concise disease, but rather consists of a range of clinical phenotypes resulting from heterogeneous prenatal and postnatal exposures. However, an underlying commonality to mechanisms of BPD development is pulmonary inflammation secondary to exposures such as mechanical ventilation, hyperoxia, pre- or postnatal infection, or fluid retention (29). These insults all occur to a developing, premature lung, thereby altering its developmental course. Nevertheless, differences in the airway microbiome between healthy and BPD susceptible preterm infants less likely represents an acute infection of the airway and more likely reflects respiratory dysbiosis, such as the predominance of Proteobacteria observed in disease states such as chronic obstructive pulmonary disease resulting in a dysregulated immune response (1, 2, 30).

A recent systematic review identified six studies that have evaluated the airway microbiome in preterm infants that developed BPD (31). The majority of included studies used amplification of the variable regions of 16S rRNA for bacterial sequencing (16, 19, 20, 27). In these studies, the airway microbiome was first described either at birth or in the early postnatal period and was subsequently characterized at different postnatal sampling intervals. The airway microbiome of infants developing BPD have been contrasted with different groups, including full-term matched controls and preterm infants that did not develop BPD. In comparing the airway microbiome at birth between infants that did and did not develop BPD, less bacterial diversity has been reported in infants that later developed BPD (20) and we have reported a more diverse microbiome in full-term post-menstrual age (PMA) matched infants at birth compared to infants that develop BPD (16). These differences in microbiome diversity may last into adulthood, as adult survivors with a history of BPD have a less diverse airway microbiome compared to preterm infants that did not develop BPD and healthy controls (32).

Studies analyzing microbial signatures and the outcome of BPD have found similar results but some institutional differences. We have reported that BPD susceptible infants have more abundant Proteobacteria and less abundant Firmicutes compared to PMA matched infants. These changes in microbial composition were also noted over time in infants developing BPD. Regarding individual bacteria at birth later associated with BPD development, Ureaplasma has been associated with an increased risk for developing BPD (27, 28). Chorioamnionitis has been cited as an independent risk factor for BPD for a very long time and we reported that preterm infants born to mothers with chorioamnionitis start off with decreased Lactobacilli in their airways at birth. Over time, a decrease in Acinetobacter abundance and increase in Staphylococcus and Klebsiella abundance has been reported in infants that develop BPD (20). However, these investigations of bacterial abundance and BPD susceptibility do not detail the mechanism for altered pulmonary development.

Recently, we conducted another study to using a software-based method to predict the metagenome of the tracheal aspirate microbiome from 16S rRNA sequencing data in extremely preterm infants (born at ≤ 28 weeks' gestation) and identified functional host ortholog genes that were differentially abundant in infants developing BPD compared to infants that did not develop BPD (33). We also identified metabolites and metabolic pathways that were differentially enriched in these samples by use of untargeted mass spectrometry and mummichog. The airway metabolome of infants developing BPD was enriched for metabolites involved in fatty acid activation and androgen and estrogen biosynthesis compared with infants that did not develop BPD. These findings suggest that in extremely preterm infants, the early airway microbiome may alter the metabolome, thereby modifying the risk of BPD. While the etiology of differential enrichment of sex steroid metabolic pathways remains unclear, previous studies have suggested a role for sexual dimorphism in BPD risk (33).

From ‘Biome to BPD: Mechanisms of Consideration

Proposed mechanisms by which the airway microbiome results in BPD development include immune system priming, oxidative stress, and metabolic dysregulation. The immunomodulatory role of the airway microbiome in human disease has been well-described (3436). Previous investigations have reported a decrease in inflammatory cytokines (e.g., tumor necrosis factor-α) in peripheral blood mononuclear cells following exposure to Lactobacillus-derived factors (37). Additionally, in in vitro studies of LPS-induced lung injury, inflammatory cytokines derived from alveolar macrophages were reduced upon exposure to Lactobacillus-derived factors (38). In vivo studies also reported less recruitment of inflammatory cells to the lung tissue with Lactobacillus administration (38). Instillation of Lactobacilli into adult wild-type mice lungs preserves alveolar architecture compared to germ-free mice controls (39). Nevertheless, neonatal germ-free mice have relatively protected alveolar structure in response to noxious stimuli such as hyperoxia (40). The probiotic use of Lactobacilli has already been demonstrated in the setting of enteropathogenic colonization and viral infection (41).

We have reported differences in exosome signatures between infants who do and do not develop BPD. Exosomes are vesicles produced by numerous cells and house molecular content, and may participate in cellular communication and pathogenic pathways, and may therefore provide clinical utility as a biomarker for disease. In a large prospective study in extremely preterm infants (born at <28 weeks' gestation), exosomes were longitudinally collected from tracheal aspirates and compared between infants that did and did not develop severe BPD (42). Infants developing severe BPD had 40 different micro RNA signatures compared to non-BPD infants. Upon analyzing these signatures in a validation cohort, low miR 876-3p expression had a sensitivity of 91.7% for severe BPD prediction (42). As previously discussed, Proteobacteria are more abundant in infants that develop severe BPD and using an in vivo, hyperoxia-based BPD mouse model, we compared the alveolar architecture of hyperoxia exposed mice to hyperoxia exposed mice also exposed to Proteobacterial LPS. Mice exposed to hyperoxia and Proteobacterial LPS had the most significant reduction in miR 876-3p expression supporting the role of miR 876-3p as a potential biomarker for Proteobacterial-induced BPD development and/or a therapeutic target (42). As miR 876-3p was found to be a protective miRNA in BPD pathogenesis, gain of function of miR 876-3p could be attempted in therapeutic studies of chronic lung injury. Additionally, further studies utilizing miR 876-3p to validate BPD prediction at other sites, to monitor therapeutic response to therapy, and/or predict other outcomes in extremely preterm infants (e.g., long term pulmonary morbidity) are needed. Interestingly based on bioinformatic platforms, one of the top predicted targets of miR 876-3p is androgen receptor (AR). We speculate that further study of miR 876-3p and its target AR has the potential to delineate the sex predilection seen in BPD.

Given that a dysbiotic airway microbiome has been associated with BPD development, we have recently investigated whether germ-free mice exhibit a BPD phenotype after exposure to hyperoxia (40). In these studies, pulmonary development was similar between germ-free and non-germ-free (NGF) mice under normoxic conditions. In a hyperoxic environment, both NGF and germ-free mice exhibited evidence of alveolar maldevelopment; however, more alveolarization, decreased cytokine levels, and improved lung function were observed in germ-free mice compared to NGF mice (40). These experiments suggest that while the airway microbiome may not be a necessary exposure for lung maldevelopment, it may have an additive or synergistic role.

Probiotics: A Plausible Therapeutic Option for BPD?

Several randomized trials have evaluated the effect of enteral probiotics and lung disease. In adults, probiotic exposure reduced ventilator-associated pneumonia compared to placebo in mechanically ventilated patients and reduced pulmonary exacerbations in patients with cystic fibrosis (43, 44). In a meta-analysis of probiotics in the context of allergic rhinitis, Lactobacillus-based probiotics were the probiotic used in the majority of studies and 22 randomized trials were identified. Probiotic-exposed patients had a reduction in symptom scores, an improvement in quality of life scores, and improved immunologic parameters compared to placebo (45). A randomized controlled trial of probiotics in children with asthma reported a reduction in asthma severity in probiotic exposed children compared to placebo and Lactobacillus-based probiotics may also reduce nosocomial infections in children (44, 46). Previous studies using enteral probiotics to reduce either necrotizing enterocolitis (NEC), a devastating gastrointestinal disease of prematurity, or late onset sepsis have not demonstrated a reduction in BPD (47). However, the influence of enteral probiotics on the airway microbiome has not been evaluated and randomized trials of aerosolized probiotics in preterm infants have never been conducted. Future studies are warranted to specifically target the lungs in animal models of respiratory diseases such as BPD, CF, and COPD. There may be a potential role of inhaled respiratory probiotics in the years to come.

Future Investigation

Although bacteria such as Lactobacilli may be lung protective, there have been no studies demonstrating this protective role in animal models of neonatal chronic lung disease. Once the benefits of reversing the respiratory dysbiosis by using respiratory probiotics are established in small- and large-animal models, randomized controlled trials in extremely preterm infants would be needed. Despite numerous studies of associations between the microbiome and BPD development, further investigation into how a dysbiotic microbiome mechanistically predisposes infants to BPD development is also warranted. As the relative abundance of microbiota results from various environmental pressures including nutrient availability, temperature, and oxygen, as well as other determinants of temporospatial patterns including rate of reproduction, bacterial elimination, and immigration (30), it is unclear how such variables influence and may explain differences in the airway microbiome between BPD-resistant and -susceptible infants. Moreover, it remains unclear as to whether the microbiome causes or is associated with BPD development. As the origin of the airway microbiome remains uncertain and may precede birth, microbial targets upstream of airway colonization have not yet been identified.

Author Contributions

All authors drafted, reviewed, edited, and finalized the manuscripts.

Funding

This work was supported by funds from the American Heart Association, National Institute of Health, and the University of Alabama at Birmingham.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

1. Millares L, Ferrari R, Gallego M, Garcia-Nuñez M, Pérez-Brocal V, Espasa M, et al. Bronchial microbiome of severe COPD patients colonised by Pseudomonas aeruginosa. Eur J Clin Microbiol Infect Dis. (2014) 33:1101–11. doi: 10.1007/s10096-013-2044-0

PubMed Abstract | CrossRef Full Text | Google Scholar

2. 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) 52:2813–23. doi: 10.1128/JCM.00035-14

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Huang YJ, Nelson CE, Brodie EL, Desantis TZ, Baek MS, Liu J, et al. Airway microbiota and bronchial hyperresponsiveness in patients with suboptimally controlled asthma. J Allergy Clin Immunol. (2011) 127:372–81.e1–3. doi: 10.1016/j.jaci.2010.10.048

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Hilty M, Burke C, Pedro H, Cardenas P, Bush A, Bossley C, et al. Disordered microbial communities in asthmatic airways. PLoS ONE. (2010) 5:e8578. doi: 10.1371/journal.pone.0008578

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Yoon BH, Romero R, Kim M, Kim EC, Kim T, Park JS, et al. Clinical implications of detection of Ureaplasma urealyticum in the amniotic cavity with the polymerase chain reaction. Am J Obstet Gynecol. (2000) 183:1130–7. doi: 10.1067/mob.2000.109036

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Perni SC, Vardhana S, Korneeva I, Tuttle SL, Paraskevas LR, Chasen ST, et al. Mycoplasma hominis and Ureaplasma urealyticum in midtrimester amniotic fluid: association with amniotic fluid cytokine levels and pregnancy outcome. Am J Obstet Gynecol. (2004) 191:1382–6. doi: 10.1016/j.ajog.2004.05.070

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Lowe J, Watkins WJ, Edwards MO, Spiller OB, Jacqz-Aigrain E, Kotecha SJ, et al. Association between pulmonary ureaplasma colonization and bronchopulmonary dysplasia in preterm infants: updated systematic review and meta-analysis. Pediatr infect Dis J. (2014) 33:697–702. doi: 10.1097/INF.0000000000000239

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Mabanta CG, Pryhuber GS, Weinberg GA, Phelps DL. Erythromycin for the prevention of chronic lung disease in intubated preterm infants at risk for, or colonized or infected with Ureaplasma urealyticum. Cochrane Database Syst Rev. (2003) 4:CD003744. doi: 10.1002/14651858.CD003744

CrossRef Full Text | Google Scholar

9. Aagaard K, Ma J, Antony KM, Ganu R, Petrosino J, Versalovic J. The placenta harbors a unique microbiome. Sci Transl Med. (2014) 6:237ra65. doi: 10.1126/scitranslmed.3008599

CrossRef Full Text | Google Scholar

10. Seferovic MD, Pace RM, Carroll M, Belfort B, Major AM, Chu DM, et al. Visualization of microbes by 16S in situ hybridization in term and preterm placentas without intraamniotic infection. Am J Obstet Gynecol. (2019) 221:146 e1–23. doi: 10.1016/j.ajog.2019.04.036

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Kuperman AA, Zimmerman A, Hamadia S, Ziv O, Gurevich V, Fichtman B, et al. Deep microbial analysis of multiple placentas shows no evidence for a placental microbiome. BJOG. (2019) 127:159–69. doi: 10.1111/1471-0528.15896

PubMed Abstract | CrossRef Full Text | Google Scholar

12. de Goffau MC, Lager S, Sovio U, Gaccioli F, Cook E, Peacock SJ, et al. Human placenta has no microbiome but can contain potential pathogens. Nature. (2019) 572:329–34. doi: 10.1038/s41586-019-1451-5

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Rehbinder EM, Lodrup Carlsen KC, Staff AC, Angell IL, Landrø L, Hilde K, et al. Is amniotic fluid of women with uncomplicated term pregnancies free of bacteria? Am J Obstet Gynecol. (2018) 219:289.e1–e12. doi: 10.1016/j.ajog.2018.05.028

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Hansen R, Scott KP, Khan S, Martin JC, Berry SH, Stevenson M, et al. First-pass meconium samples from healthy term vaginally-delivered neonates: an analysis of the microbiota. PLoS ONE. (2015) 10:e0133320. doi: 10.1371/journal.pone.0133320

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Jiménez E, Fernández L, Marín ML, Martín R, Odriozola JM, Nueno-Palop C, et al. Isolation of commensal bacteria from umbilical cord blood of healthy neonates born by cesarean section. Curr Microbiol. (2005) 51:270–4. doi: 10.1007/s00284-005-0020-3

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Lal CV, Travers C, Aghai ZH, Eipers P, Jilling T, Halloran B, et al. The airway microbiome at birth. Sci Rep. (2016) 6:31023. doi: 10.1038/srep31023

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Collado MC, Rautava S, Aakko J, Isolauri E, Salminen S. Human gut colonisation may be initiated in utero by distinct microbial communities in the placenta and amniotic fluid. Sci Rep. (2016) 6:23129. doi: 10.1038/srep23129

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Keski-Nisula L, Kyynäräinen HR, Kärkkäinen U, Karhukorpi J, Heinonen S, Pekkanen J. Maternal intrapartum antibiotics and decreased vertical transmission of Lactobacillus to neonates during birth. Acta Paediatr. (2013) 102:480–5. doi: 10.1111/apa.12186

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Mourani PM, Harris JK, Sontag MK, Robertson CE, Abman SH. Molecular identification of bacteria in tracheal aspirate fluid from mechanically ventilated preterm infants. PLoS ONE. (2011) 6:e25959. doi: 10.1371/journal.pone.0025959

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Lohmann P, Luna RA, Hollister EB, Devaraj S, Mistretta TA, Welty SE, et al. The airway microbiome of intubated premature infants: characteristics and changes that predict the development of bronchopulmonary dysplasia. Pediatr Res. (2014) 76:294–301. doi: 10.1038/pr.2014.85

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Bokulich NA, Chung J, Battaglia T, Henderson N, Jay M, Li H, et al. Antibiotics, birth mode, and diet shape microbiome maturation during early life. Sci Transl Med. (2016) 8:343ra82. doi: 10.1126/scitranslmed.aad7121

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Dominguez-Bello MG, Costello EK, Contreras M, Magris M, Hidalgo G, Fierer N, et al. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc Natl Acad Sci USA. (2010) 107:11971–5. doi: 10.1073/pnas.1002601107

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Pannaraj PS, Li F, Cerini C, Bender JM, Yang S, Rollie A, et al. Association between breast milk bacterial communities and establishment and development of the infant gut microbiome. JAMA Pediatr. (2017) 171:647–54. doi: 10.1001/jamapediatrics.2017.0378

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Abrahamsson TR, Jakobsson HE, Andersson AF, Björkstén B, Engstrand L, Jenmalm MC. Low gut microbiota diversity in early infancy precedes asthma at school age. Clin Exp Allergy. (2014) 44:842–50. doi: 10.1111/cea.12253

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Bruzzese E, Callegari ML, Raia V, Viscovo S, Scotto R, Ferrari S, et al. 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. doi: 10.1371/journal.pone.0087796

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Huang J, Zhang L, Tang J, Shi J, Qu Y, Xiong T, et al. Human milk as a protective factor for bronchopulmonary dysplasia: a systematic review and meta-analysis. Arch Dis Child Fetal Neonatal Ed. (2019) 104:F128–36. doi: 10.1136/archdischild-2017-314205

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Wagner BD, Sontag MK, Harris JK, Miller JI, Morrow L, Robertson CE, et al. Airway microbial community turnover differs by BPD severity in ventilated preterm infants. PLoS ONE. (2017) 12:e0170120. doi: 10.1371/journal.pone.0170120

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Payne MS, Goss KC, Connett GJ, Kollamparambil T, Legg JP, Thwaites R, et al. Molecular microbiological characterization of preterm neonates at risk of bronchopulmonary dysplasia. Pediatr Res. (2010) 67:412–8. doi: 10.1203/PDR.0b013e3181d026c3

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Balany J, Bhandari V. Understanding the impact of infection, inflammation, and their persistence in the pathogenesis of bronchopulmonary dysplasia. Front Med. (2015) 2:90. doi: 10.3389/fmed.2015.00090

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Dickson RP, Martinez FJ, Huffnagle GB. The role of the microbiome in exacerbations of chronic lung diseases. Lancet. (2014) 384:691–702. doi: 10.1016/S0140-6736(14)61136-3

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Pammi M, Lal CV, Wagner BD, Mourani PM, Lohmann P, Luna RA, et al. Airway microbiome and development of bronchopulmonary dysplasia in preterm infants: a systematic review. J Pediatr. (2019) 204:126–33.e2. doi: 10.1016/j.jpeds.2018.08.042

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Rofael SAD, McHugh TD, Troughton R, Beckmann J, Spratt D, Marlow N, et al. Airway microbiome in adult survivors of extremely preterm birth: the EPICure study. Eur Respir J. (2019) 53:1801225. doi: 10.1183/13993003.01225-2018

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Lal CV, Kandasamy J, Dolma K, Ramani M, Kumar R, Wilson L, et al. Early airway microbial metagenomic and metabolomic signatures are associated with development of severe bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol. (2018) 315:L810–5. doi: 10.1152/ajplung.00085.2018

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Brandtzaeg P. The role of humoral mucosal immunity in the induction and maintenance of chronic airway infections. Am J Respir Crit Care Med. (1995) 151:2081–6. doi: 10.1164/ajrccm.151.6.7767561

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Huang YJ, Erb-Downward JR, Dickson RP, Curtis JL, Huffnagle GB, Han MK. Understanding the role of the microbiome in chronic obstructive pulmonary disease: principles, challenges, and future directions. Transl Res. (2017) 179:71–83. doi: 10.1016/j.trsl.2016.06.007

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Cribbs SK, Beck JM. Microbiome in the pathogenesis of cystic fibrosis and lung transplant-related disease. Transl Res. (2017) 179:84–96. doi: 10.1016/j.trsl.2016.07.022

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Mechoud MA, Mateos MV, de Valdez GF, Villena J, Salvador GA, Rodriguez AV. Lactobacillus reuteri CRL1098 soluble factors modulate tumor necrosis factor alpha production in peripheral blood mononuclear cells: involvement of lipid rafts. Int Immunopharmacol. (2012) 14:446–53. doi: 10.1016/j.intimp.2012.08.020

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Griet M, Zelaya H, Mateos MV, Salva S, Juarez GE, de Valdez GF, et al. Soluble factors from Lactobacillus reuteri CRL1098 have anti-inflammatory effects in acute lung injury induced by lipopolysaccharide in mice. PLoS ONE. (2014) 9:e110027. doi: 10.1371/journal.pone.0110027

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Yun Y, Srinivas G, Kuenzel S, Linnenbrink M, Alnahas S, Bruce KD, et al. Environmentally determined differences in the murine lung microbiota and their relation to alveolar architecture. PLoS ONE. (2014) 9:e113466. doi: 10.1371/journal.pone.0113466

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Dolma K, Freeman AE, Rezonzew G, Payne GA, Xu X, Jilling T, et al. Effects of hyperoxia on alveolar and pulmonary vascular development in germ free mice. Am J Physiol Lung Cell Mol Physiol. (2019). doi: 10.1152/ajplung.00316.2019. [Epub ahead of print].

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Stecher B, Hardt WD. The role of microbiota in infectious disease. Trends Microbiol. (2008) 16:107–14. doi: 10.1016/j.tim.2007.12.008

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Lal CV, Olave N, Travers C, Rezonzew G, Dolma K, Simpson A, et al. Exosomal microRNA predicts and protects against severe bronchopulmonary dysplasia in extremely premature infants. JCI Insight. (2018) 3:93994. doi: 10.1172/jci.insight.93994

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Morrow LE, Kollef MH, Casale TB.Morrow LE, Kollef MH, Casale TB. Probiotic prophylaxis of ventilator-associated pneumonia: a blinded, randomized, controlled trial. Am J Respir Crit Care Med. (2010) 182:1058–64. doi: 10.1164/rccm.200912-1853OC

CrossRef Full Text | Google Scholar

44. Van Biervliet S, Declercq D, Somerset S. Clinical effects of probiotics in cystic fibrosis patients: a systematic review. Clin Nutr ESPEN. (2017) 18:37–43. doi: 10.1016/j.clnesp.2017.01.007

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Güvenç IA, Muluk NB, Mutlu FS, Eşki E, Altintoprak N, Oktemer T, et al. Do probiotics have a role in the treatment of allergic rhinitis? a comprehensive systematic review and meta-analysis. Am J Rhinol Allergy. (2016) 30:157–75. doi: 10.2500/ajra.2016.30.4354

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Hojsak I, Abdović S, Szajewska H, Milosević M, Krznarić Z, Kolacek S. Lactobacillus GG in the prevention of nosocomial gastrointestinal and respiratory tract infections. Pediatrics. (2010) 125:e1171–7. doi: 10.1542/peds.2009-2568

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Villamor-Martinez E, Pierro M, Cavallaro G, Mosca F, Kramer B, Villamor E. Probiotic supplementation in preterm infants does not affect the risk of bronchopulmonary dysplasia: a meta-analysis of randomized controlled trials. Nutrients. (2017) 9:E1197. doi: 10.3390/nu9111197

CrossRef Full Text | Google Scholar

Keywords: airway microbiome, bronchopulmonary dysplasia, probiotics, lung disease, prematurity

Citation: Gentle SJ and Lal CV (2020) Predicting BPD: Lessons Learned From the Airway Microbiome of Preterm Infants. Front. Pediatr. 7:564. doi: 10.3389/fped.2019.00564

Received: 03 November 2019; Accepted: 24 December 2019;
Published: 04 February 2020.

Edited by:

Karen Cecile Young, University of Miami, United States

Reviewed by:

Jonathan Michael Davis, Tufts University, United States
Venkatesh Sampath, Children's Mercy Hospital, United States

Copyright © 2020 Gentle and Lal. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Charitharth Vivek Lal, clal@peds.uab.edu

Download