Next Article in Journal
A Mechanistic Theory of Development-Aging Continuity in Humans and Other Mammals
Next Article in Special Issue
Mechanistic Insights into the Link between Gut Dysbiosis and Major Depression: An Extensive Review
Previous Article in Journal
Artificial Intelligent Deep Learning Molecular Generative Modeling of Scaffold-Focused and Cannabinoid CB2 Target-Specific Small-Molecule Sublibraries
Previous Article in Special Issue
Role of Dietary Supplements and Probiotics in Modulating Microbiota and Bone Health: The Gut-Bone Axis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Targeting the Pulmonary Microbiota to Fight against Respiratory Diseases

Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China
*
Author to whom correspondence should be addressed.
Cells 2022, 11(5), 916; https://doi.org/10.3390/cells11050916
Submission received: 14 January 2022 / Revised: 26 February 2022 / Accepted: 1 March 2022 / Published: 7 March 2022
(This article belongs to the Special Issue Gut Microbiota in Nutrition and Health)

Abstract

:
The mucosal immune system of the respiratory tract possesses an effective “defense barrier” against the invading pathogenic microorganisms; therefore, the lungs of healthy organisms are considered to be sterile for a long time according to the strong pathogens-eliminating ability. The emergence of next-generation sequencing technology has accelerated the studies about the microbial communities and immune regulating functions of lung microbiota during the past two decades. The acquisition and maturation of respiratory microbiota during childhood are mainly determined by the birth mode, diet structure, environmental exposure and antibiotic usage. However, the formation and development of lung microbiota in early life might affect the occurrence of respiratory diseases throughout the whole life cycle. The interplay and crosstalk between the gut and lung can be realized by the direct exchange of microbial species through the lymph circulation, moreover, the bioactive metabolites produced by the gut microbiota and lung microbiota can be changed via blood circulation. Complicated interactions among the lung microbiota, the respiratory viruses, and the host immune system can regulate the immune homeostasis and affect the inflammatory response in the lung. Probiotics, prebiotics, functional foods and fecal microbiota transplantation can all be used to maintain the microbial homeostasis of intestinal microbiota and lung microbiota. Therefore, various kinds of interventions on manipulating the symbiotic microbiota might be explored as novel effective strategies to prevent and control respiratory diseases.

1. Introduction

Over the past two decades, accumulated studies on the interactions between lung microbiota and the host immune system have provided invaluable understanding about the immune modulating functions of the lung microbiome [1,2]. The lungs of healthy individuals have been considered to be sterile for a long time according to the classic respirology theory. However, the development of culture-independent sequencing technology has proved that there are abundant and diverse microbial communities in the respiratory tract and have intimate associations with the host’s health and disease [3,4,5]. The acquisition and maturation of the lung microbiome in the early life can be influenced by delivery mode, feeding practices, living environment, and other affecting factors [6]. The interactions among the lung microbiota, infected viruses, invading bacteria, and the host immune system can affect the susceptibility to lower respiratory tract infections and diseases [7]. Therefore, investigations on the microbial composition and the immune function of the lung microbiome may provide novel effective interventions and preventive measures to treat respiratory diseases.
The respiratory tract has a large exposed surface to execute the air-exchange function, while the abundance and diversity of the lung microbiome are obviously influenced by the contacting air environment [8]. Though the lungs of mammals are equipped with an effective antimicrobial defense system, the relatively lower microbial biomass of the pulmonary microbiota successfully colonized the respiratory tract and obtained tolerance to the host immune system [9,10]. Moreover, the stably harbored lung commensal bacteria can generate colonizing resistance and help to fight against the invading outer pathogens by producing various kinds of antimicrobial molecules. Therefore, the lung microbial ecosystem is maintained by the balance of transiently entered and selectively eliminated microorganisms [11]. The bi-directional cross-talk between the lung microbiome and the host immune system plays a fundamental role in keeping the lung immune homeostasis. On one side, the inhabited pulmonary microbiota can influence the maturation of the host immune system by producing numerous structural ligands and metabolites (such as lipopolysaccharide, peptidoglycan, and short-chain fatty acids). On the other side, the host’s innate and adaptive immune system can alter the lung microbiome by forming biophysical barriers, secreting immunoglobulin A (IgA), producing antimicrobial peptides, and recognizing the resident and viable microbes [12]. Therefore, many factors that cause lung microbiome dysbiosis can alter the pulmonary immune homeostasis and induce the occurrence and development of respiratory inflammation and diseases [13].
Increasing evidence has revealed that the disturbed balances of intestinal microbiota and lung microbiota had intimate correlations and can corporately cause respiratory diseases. The comprehensive and sophisticated interactions between intestinal microbiota and lung microbiota and their collective actions in modulating the pulmonary immune homeostasis might provide new therapeutic targets and manipulating strategies for clinical treatment of respiratory infections [14,15]. Research about the gut-lung axis demonstrated the depletion of the gut microbiota in C57BL/6 mice could induce lung bacterial dissemination, organ damage and enhance mice mortality during Streptococcus pneumoniae infections. However, the restored process of the gut microbiota by fecal microbiota transplantation (FMT) could reverse the survival rate of broad-spectrum antibiotics treated mice by regulating alveolar macrophage function and inflammation response [16,17]. Probiotics treatments targeting gut and lung microbiota could confer health benefits for the host during the chronic lung disease progression, and other interventions to protect the microbial ecosystem balance of the gastrointestinal tract and the respiratory tract could also be exploited to protect the respiratory immune system [18,19,20].
In this review, we mainly summarize the composition of respiratory microbiota and their potential functions related to the host immune system. Moreover, we also discuss the novel therapeutic approaches targeting the gut and lung microbiota to treat respiratory diseases.

2. The Origin of Pulmonary Microbiota and the Influencing Factors

The lungs of healthy individuals have previously been considered to be sterile because the traditional microbiology approaches usually cannot give out positive cultural results [21]. Progress on the culture-independent techniques has proved that the colonization of microbial community in respiratory tract began immediately after birth, for the reason that the bacterial DNA in the tracheal aspirates of healthy neonates could be detected 24 h after birth [22]. By comparing the microbial communities in oral wash, nasal swab, and bronchoalveolar lavage (BAL) from the healthy subjects, the bacterial communities of the lungs shared much higher similarity with those from the oral cavity, but were different from the nasal cavity. Therefore, the lung microbiota might quite possibly originate from the oral microbiota through the microaspiration, which usually occurred during the sleep process when the tone of the oral and pharyngeal muscles is diminished [23].
Initial microbial colonization in the respiratory tract is mainly impacted by delivery mode, antibiotic usage, dietary structure, environment exposure, and pathogenic infections (Figure 1) [24,25,26,27]. In early life, caesarean delivery patterns, increasing use of antibiotics, changes in food composition, and the contacting environmental microorganisms can all directly and indirectly impact the diversity and abundance of the lung microbiome [28,29,30]. Moreover, pathogenic infections induced by various kinds of viruses can also play important roles in shaping lung microbiota formation [31,32]. Comprehensive analysis of the environmental and lifestyle factors that influence the early colonization of the lung microbiome might provide preventative interventions and therapeutic strategies to treat respiratory diseases [33,34].

3. The Diversity and Composition of Respiratory Microbiota

At birth, the acquisitions of infant microbial community in the mucosal surfaces is mainly determined by the microbes derived from the mother’s vagina, skin and intestinal tract [35,36]. In the subsequent early life period, the distribution, composition and development of respiratory microbiota are transiently diversified together with the maturation of the host immune system [37,38]. As the air-exchange area, the lung environmental condition is vastly different from other body sites, therefore the compositions and diversities of lung microbiota are mainly determined by the transient change of entering outer microorganisms and selective elimination of viable microorganisms. During the microbe–host crosstalk process, the host respiratory tract has developed a variety of selective strategies to maintain the balance of the microbial ecosystem [39].
Though the lung has a large surface to directly contact with the outer air environments, the pulmonary immune system is equipped with an effective antimicrobial and defensive system to fight against the invading foreign microorganisms. Therefore, the microbial biomass in the lung is remarkably lower than that in other body sites [7,40]. At the phylum level, the predominant microbial communities in the lungs are mainly composed of Proteobacteria, Firmicutes, Tenericutes, and Bacteroidetes. When analyzed at the genera level, the most common genera in the lungs of healthy individuals are mainly composed of Prevotella, Veillonella, Streptococcus and Pseudomonas. When compared with the adjacent sites, enhanced richness of Proteobacteria, Ralstonia and Haemophilus and decreased abundance of Prevotella-affiliated taxa are consistently observed, the unique compositions of lung microbiota are possibly related to the redox state and oxygen application of the lower respiratory tract [41,42,43]. The composition of lung microbiota in health and disease states differed apparently when the balance of host immune response is disturbed by viruses, allergens or genetic deficiency. Upon the conditions of cystic fibrosis (CF), chronic obstructive pulmonary disease (COPD), and other chronic lung diseases, the predominant genera were shifted to Pseudomonas, Streptococcus, Prevotella and Haemophilus. When compared with the healthy individuals, the richness of Bacteroidetes in the patients was significantly decreased [7]. Studies about the lung microbiota in patients with asthma indicated that the abundance of Proteobacteriae was increased which might be driven by the Haemophilus, Moraxella and Neisseria species [44]. Multiple studies demonstrated that gut dysbiosis was observed in patients with coronavirus disease 2019 (COVID-19), and the gut microbiota richness and composition of COVID-19 patients were quite different from those of healthy controls. After severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, the richness and diversity of gut microbiota were both significantly decreased, and microbial richness could not restore to normal levels even after 6-month recovery. However, the predominant microbial taxa of severely ill patients were characterized by Burkholderia cepacia complex, Staphylococcus epidermidis, or Mycoplasma spp. (including M. hominis and M. orale) [45,46,47].

4. Relations between the Gut Microbiota and Lung Microbiota Mediated by the Gut-Lung Axis

The “Zang-Fu” theory in traditional Chinese medicine describes that “lung and large intestineare interior-exteriorly related”, which demonstrates the close physiological and pathological connections between the gut and lung [48]. In fact, the microbiota–host communications can transmit multiple intestinal signals to different distal organs and contribute to host health and disease [49,50]. By comparing the microbial community structures between lung and intestine bacteria, evidence reveals that members of lung and intestine bacteria can directly exchange through the lymph circulation [51,52,53]. Additionally, gut microbiota can produce various kinds of bioactive metabolites (such as butyrate, p-cresol sulfate, and indoles) to impact the host immune response and energy homeostasis [54,55,56,57,58]. The innate lymphoid cells (ILCs) derived from intestinal lamina propria have important action on host defense and inflammatory responses. When the interleukin-25-induced group 2 innate lymphoid cells (ILC2) and interleukin-22 (IL-22)-producing group 3 innate lymphoid cells (ILC3) migrate to the lung, the host resistance to pneumonia and other inflammatory infections could be promoted [59,60,61,62]. Therefore, the gut microbiome plays an important role in regulating pulmonary immune function and health protection through signals transmitted by the gut–lung axis.
The regulating role of the lung microbiota on intestinal infectious diseases through the gut–lung axis should also not be neglected (Figure 2). Alteration of pulmonary microbiota is found to be able to modulate the microbial communities of gut and influence intestinal immunity and disorders [63]. Pulmonary infections caused by Mycobacterium tuberculosis can induce a distinct dysbiosis of the gut microbiota by decreasing the α-diversity; however, the anti-tuberculosis therapy can also cause a rapid and significant change in the diversity and composition of the gut microbiota [64,65,66,67,68,69]. Recently, the gut microbiota is considered to be a novel potential therapeutic target in treating tuberculosis. Supplementation of Lactobacillus could restore the dysregulated the gut microbiota and enhance the lung dendritic cells (DCs) function and subsequent T cell response to control tuberculosis [70]. Moreover, the diversity of the intestinal microbial community can be significantly disturbed in the Pneumocystis murina and Klebsiella pneumoniae caused respiratory infection [71,72]. The dysbiosis of gut microbiota and subsequent dysregulation of microbiota-related immunological processes could also be observed in patients with asthma, COPD, and other chronic respiratory diseases [73,74]. Therefore, precise treating approaches to modify the lung and gut microbiome might play important roles in the management and treatment of respiratory diseases.

5. The Lung Immune Homeostasis Shaped by Microbe-Host Interactions

The crosstalk between the pulmonary microbiota and host mucosal immune system plays a fundamental role in maintaining lung immune homeostasis [12]. Numerous factors that cause pulmonary microbiota dysbiosis could disturb the immune function and induce inflammation responses and airway diseases [34]. The “hygiene hypothesis” explained the critical relevance between the symbiotic microbiota alteration and the immune homeostasis dysregulation, because the modern lifestyle in industrialized societies altered the human microbial ecosystem and increased the occurring chances of infectious disease [75,76]. In early life, the formation and development of the commensal microbiota have critical impacts on the maturation of the immune system.
The microorganism-associated molecular patterns (such as lipopolysaccharide and flagellin) of commensal microbes can induce the production of secretory immunoglobulin A (sIgA) and establish the balance of immune recognition and immune tolerance [77]. During the process of pregnancy, the pattern of the immune system in the fetal environment is dominated by the Th2 phenotype. When the neonate’s symbiotic microbiome is acquired after birth, the polarization of lung naïve T cells begins to shift from Th2 to Th1 phenotype, and then the infant`s resistance to allergic diseases is enhanced [78,79,80]. Because the microbial compounds could induce the differentiation of regulatory T cells (Treg) and Th17 cells, the dysregulated lung microbiota by bleomycin treatment induce the production of interleukin-17B (IL-17B) and tumor necrosis factor-α (TNF-α) through Toll-like receptor-Myd88 adaptor signaling [81,82]. Therefore, manipulation of the infant microbial community could train the responses of the innate and adaptive immune system, and provide promising approaches to benefit life-long health [83,84].

6. The Lung Microbiota: Potential Target to Prevent and Treat Pulmonary Infections

The microbial communities harbored in the lung can build a protective barrier against respiratory diseases, while the complicated interactions among the pulmonary microbiome, pathogenic virome, and host immunity act as critical roles in lung inflammation and immune responses [85,86]. The secondary bacterial infections often happen together with or after viral infections, and the infectious mechanism can be explained the dysregulated innate and acquired immune homeostasis and the pathological damage which are caused by the airway tract viruses. Conversely, persistent infection or colonization of pathogenic bacteria can also induce viral infections by increasing the expression of viral entry receptors [87,88,89]. Emerging evidence has proved that the sophisticated interactions between viruses and bacteria at multiple levels in the lower respiratory tract can influence the host phenotypic effects; therefore, investigations on the viral and bacterial co-infections help to explore novel effective approaches for preventing respiratory diseases [90,91,92].
Recent researches on the lung microbiome promote the understanding of the therapeutic target of commensal microbiota for various kinds of respiratory diseases. Numerous probiotics have been widely applied to treat infectious airway diseases for the beneficial role of regulating the host immunity and inhibiting the invasion of the pathogen [93]. Accumulated studies have demonstrated that oral or nasal administration of Lactobacillus and other probiotics could modulate the respiratory innate immune responses and promote health benefits against influenza virus, and several other lactic acid bacteria (LAB) strains were also reported to be able to stimulate the mucosal immune system and provide effective protection against Streptococcus pneumoniae infections [94,95,96,97]. The symbiotic microbiota can systematically impact the host respiratory system against Klebsiella pneumoniae infection and produce short-chain fatty acids (SCFAs) and subsequently activate the G protein-coupled receptors (GPCRs). The metabolic SCFAs could protect against syncytial virus (RSV) infection by involving and engagement of interferon-β (IFN-β) via the IFN-1 receptor (IFNAR) signaling [98,99,100]. The enhanced anti-viral function of alveolar macrophage was mainly derived by the up-regulation of IFN-β, and then the recovery of the lung pathology was also promoted [101,102]. According to the fact that the gut microbiota dysbiosis was involved in the magnitude of COVID-19 severity through modulating the host immune responses, targeted manipulation to restore the gut microbiota could be an important strategy to treat COVID-19 and speed up recovery [103,104,105]. Nutritional intervention may play a prominent role in establishing and regulating the compositions of intestinal and lung microbiome, therefore the applications of probiotics, prebiotics and functional foods can prevent or alleviate respiratory infections by directly inhibiting the growth of pathogens or indirectly modulating the host’s immune function (Figure 3) [106,107].

7. Conclusions

Recent research on the lung microbiome revealed its immune regulating functions and the protective role in fighting against respiratory diseases. The predominant members of the lung microbiome can comprise a microbial barrier to inhibit the colonization of invading pathogenic microorganisms, and the beneficial metabolites produced by the lung microbiome can enhance respiratory immunity and prevent the occurrence of respiratory diseases. Moreover, the intimate relations between intestinal microbiota and lung microbiota provide new therapeutic targets for clinical treatments of respiratory infections. Probiotics, prebiotics, functional foods and fecal microbiota transplantation can all be applied to maintain the microbial homeostasis of intestinal microbiota and lung microbiota. In all, various kinds of interventions targeting the symbiotic microbiota can be used as novel strategies to prevent and control the respiratory diseases.

Author Contributions

Writing—original draft, Z.L.; writing—editing, Y.L. and Q.S.; writing—review, J.W., B.L., Y.Q., K.L. and D.S.; conceptualization, Z.M. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Shanghai Rising-Star Program (No. 19QA1411200) and the Chinese National Natural Science Foundation Grant (31672606).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Wypych, T.P.; Wickramasinghe, L.C.; Marsland, B.J. The influence of the microbiome oan respiratory health. Nat. Immunol. 2019, 20, 1279–1290. [Google Scholar] [CrossRef] [PubMed]
  2. Moffatt, M.F.; Cookson, W.O. The lung microbiome in health and disease. Clin. Med. 2017, 17, 525–529. [Google Scholar] [CrossRef] [PubMed]
  3. Carney, S.M.; Clemente, J.C.; Cox, M.J.; Dickson, R.P.; Huang, Y.J.; Kitsios, G.D.; Kloepfer, K.M.; Leung, J.M.; LeVan, T.D.; Molyneaux, P.L.; et al. Methods in lung microbiome research. Am. J. Respir. Cell Mol. Biol. 2020, 62, 283–299. [Google Scholar] [CrossRef] [PubMed]
  4. Dickson, R.P. The microbiome and critical illness. Lancet Respir. Med. 2016, 4, 59–72. [Google Scholar] [CrossRef] [Green Version]
  5. O’Dwyer, D.N.; Ashley, S.L.; Gurczynski, S.J.; Xia, M.; Wilke, C.; Falkowski, N.R.; Norman, K.C.; Arnold, K.B.; Huffnagle, G.B.; Salisbury, M.L.; et al. Lung microbiota contribute to pulmonary inflammation and disease progression in pulmonary fibrosis. Am. J. Respir. Crit. Care. Med. 2019, 199, 1127–1138. [Google Scholar] [CrossRef]
  6. Unger, S.A.; Bogaert, D. The respiratory microbiome and respiratory infections. J. Infect. 2017, 74, S84–S88. [Google Scholar] [CrossRef]
  7. Dickson, R.P.; Erb-Downward, J.R.; Huffnagle, G.B. The role of the bacterial microbiome in lung disease. Expert Rev. Respir. Med. 2013, 7, 245–257. [Google Scholar] [CrossRef] [Green Version]
  8. Chung, K.F. Airway microbial dysbiosis in asthmatic patients: A target for prevention and treatment? J. Allergy Clin. Immunol. 2017, 139, 1071–1081. [Google Scholar] [CrossRef] [Green Version]
  9. Shi, H.L.; Lan, Y.H.; Hu, Z.C.; Yan, Z.N.; Liu, Z.Z.; Kadier, X.; Ma, L.; Yu, J.Y.; Liu, J. Microecology research: A new target for the prevention of asthma. Chin. Med. J. (Engl.) 2020, 133, 2712–2720. [Google Scholar] [CrossRef]
  10. Singanayagam, A.; Ritchie, A.I.; Johnston, S.L. Role of microbiome in the pathophysiology and disease course of asthma. Curr. Opin. Pulm. Med. 2017, 23, 41–47. [Google Scholar] [CrossRef]
  11. Budden, K.F.; Gellatly, S.L.; Wood, D.L.; Cooper, M.A.; Morrison, M.; Hugenholtz, P.; Hansbro, P.M. Emerging pathogenic links between microbiota and the gut-lung axis. Nat. Rev. Microbiol. 2017, 15, 55–63. [Google Scholar] [CrossRef] [PubMed]
  12. Yang, D.; Xing, Y.; Song, X.; Qian, Y. The impact of lung microbiota dysbiosis on inflammation. Immunology 2020, 159, 156–166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Sommariva, M.; Le Noci, V.; Bianchi, F.; Camelliti, S.; Balsari, A.; Tagliabue, E.; Sfondrini, L. The lung microbiota: Role in maintaining pulmonary immune homeostasis and its implications in cancer development and therapy. Cell. Mol. Life Sci. 2020, 77, 2739–2749. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Wang, B.; Yao, M.; Lv, L.; Ling, Z.; Li, L. The Human Microbiota in Health and Disease. Engineering 2017, 3, 71–82. [Google Scholar] [CrossRef]
  15. Wang, J.; Li, F.; Tian, Z. Role of microbiota on lung homeostasis and diseases. Sci. China Life Sci. 2017, 60, 1407–1415. [Google Scholar] [CrossRef]
  16. Schuijt, T.J.; Lankelma, J.M.; Scicluna, B.P.; de Sousa e Melo, F.; Roelofs, J.J.; de Boer, J.D.; Hoogendijk, A.J.; de Beer, R.; de Vos, A.; Belzer, C.; et al. The gut microbiota plays a protective role in the host defence against pneumococcal pneumonia. Gut 2016, 65, 575–583. [Google Scholar] [CrossRef] [Green Version]
  17. Dickson, R.P.; Cox, M.J. Gut microbiota and protection from pneumococcal pneumonia. Gut 2017, 66, 384. [Google Scholar] [CrossRef] [Green Version]
  18. Ranucci, G.; Buccigrossi, V.; De Freitas, M.B.; Guarino, A.; Giannattasio, A. Early-Life Intestine Microbiota and Lung Health in Children. J. Immunol. Res. 2017, 2017, 8–13. [Google Scholar] [CrossRef]
  19. He, Y.; Wen, Q.; Yao, F.; Xu, D.; Huang, Y.; Wang, J. Gut–lung axis: The microbial contributions and clinical implications. Crit. Rev. Microbiol. 2017, 43, 81–95. [Google Scholar] [CrossRef]
  20. Antunes, A.E.C.; Vinderola, G.; Xavier-Santos, D.; Sivieri, K. Potential contribution of beneficial microbes to face the COVID-19 pandemic. Food Res. Int. 2020, 136, 109577. [Google Scholar] [CrossRef]
  21. Faner, R.; Sibila, O.; Agustí, A.; Bernasconi, E.; Chalmers, J.D.; Huffnagle, G.B.; Manichanh, C.; Molyneaux, P.L.; Paredes, R.; Pérez Brocal, V.; et al. The microbiome in respiratory medicine: Current challenges and future perspectives. Eur. Respir. J. 2017, 49, 1602086. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Dickson, R.P.; Erb-Downward, J.R.; Martinez, F.J.; Huffnagle, G.B. The microbiome and the respiratory tract. Annu. Rev. Physiol. 2016, 78, 481–504. [Google Scholar] [CrossRef] [Green Version]
  23. Bassis, C.M.; Erb-Downward, J.R.; Dickson, R.P.; Freeman, C.M.; Schmidt, T.M.; Young, V.B.; Beck, J.M.; Curtis, J.L.; Huffnagle, G.B. Analysis of the upper respiratory tract microbiotas as the source of the lung and gastric microbiotas in healthy individuals. MBio 2015, 6, e00037-15. [Google Scholar] [CrossRef] [Green Version]
  24. Vendl, C.; Nelson, T.; Ferrari, B.; Thomas, T.; Rogers, T. Highly abundant core taxa in the blow within and across captive bottlenose dolphins provide evidence for a temporally stable airway microbiota. BMC Microbiol. 2021, 21, 20. [Google Scholar] [CrossRef] [PubMed]
  25. Hufnagl, K.; Pali-Schöll, I.; Roth-Walter, F.; Jensen-Jarolim, E. Dysbiosis of the gut and lung microbiome has a role in asthma. Semin. Immunopathol. 2020, 42, 75–93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Fedosenko, S.V.; Ogorodova, L.M.; Karnaushkina, M.A.; Kulikov, E.S.; Kirillova, N.A. The airways microbial community composition in healthy individuals and bronchial asthma patients. Vestn. Ross. Akad. Meditsinskikh Nauk. 2014, 69, 71. [Google Scholar] [CrossRef] [PubMed]
  27. Niederwerder, M.C. Role of the microbiome in swine respiratory disease. Vet. Microbiol. 2017, 209, 97–106. [Google Scholar] [CrossRef]
  28. De, S.; Binkowska, J.; Bogaert, D. Early life microbiota and respiratory tract infections. Cell Host Microbe 2020, 28, 223–232. [Google Scholar]
  29. Renz, H.; Skevaki, C. Early life microbial exposures and allergy risks: Opportunities for prevention. Nat. Rev. Immunol. 2020, 21, 177–191. [Google Scholar] [CrossRef]
  30. Dickson, R.P.; Morris, A. Macrolides, inflammation and the lung microbiome: Untangling the web of causality. Thorax 2017, 72, 10–12. [Google Scholar] [CrossRef] [Green Version]
  31. Ptaschinski, C.; Lukacs, N.W. Early Life Respiratory Syncytial Virus Infection and Asthmatic Responses. Immunol. Allergy Clin. N. Am. 2019, 39, 309–319. [Google Scholar] [CrossRef] [PubMed]
  32. Nguyen, L.D.; Viscogliosi, E.; Delhaes, L. The lung mycobiome: An emerging field of the human respiratory microbiome. Front. Microbiol. 2015, 6, 89. [Google Scholar] [CrossRef] [PubMed]
  33. Bosch, A.; de Steenhuijsen Piters, W.; van Houten, M.A.; Chu, M.; Biesbroek, G.; Kool, J.; Pernet, P.; de Groot, P.; Eijkemans, M.; Keijser, B.; et al. Maturation of the infant respiratory microbiota, environmental drivers, and health consequences. a prospective cohort study. Am. J. Respir. Crit. Care Med. 2017, 196, 1582–1590. [Google Scholar] [CrossRef]
  34. Lloyd, C.M.; Marsland, B.J. Lung homeostasis: Influence of age, microbes, and the immune system. Immunity 2017, 46, 549–561. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Dominguez-Bello, M.G.; Costello, E.K.; Contreras, M.; Magris, M.; Hidalgo, G.; Fierer, N.; Knight, R. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc. Natl. Acad. Sci. USA 2017, 107, 11971–11975. [Google Scholar] [CrossRef] [Green Version]
  36. Hurley, E.; Mullins, D.; Barrett, M.P.; O’Shea, C.A.; Kinirons, M.; Ryan, C.A.; Stanton, C.; Whelton, H.; Harris, H.; O’Toole, P.W.; et al. The microbiota of the mother at birth and its influence on the emerging infant oral microbiota from birth to 1 year of age: A cohort study. J. Oral Microbiol. 2019, 11, 1599652. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Younge, N.E.; Araújo-Pérez, F.; Brandon, D.; Seed, P.C. Early-life skin microbiota in hospitalized preterm and full-term infants. Microbiome 2018, 6, 98. [Google Scholar] [CrossRef] [Green Version]
  38. Singh, N.; Vats, A.; Sharma, A.; Arora, A.; Kumar, A. The development of lower respiratory tract microbiome in mice. Microbiome 2017, 5, 61. [Google Scholar] [CrossRef] [Green Version]
  39. Huffnagle, G.B.; Dickson, R.P.; Lukacs, N.W. The respiratory tract microbiome and lung inflammation: A two-way street. Mucosal Immunol. 2017, 10, 299–306. [Google Scholar] [CrossRef] [Green Version]
  40. Li, Z.; Wang, X.; Di, D.; Pan, R.; Gao, Y.; Xiao, C.; Li, B.; Wei, J.; Liu, K.; Qiu, Y.; et al. Comparative analysis of the pulmonary microbiome in healthy and diseased pigs. Mol. Genet. Genom. 2021, 296, 21–31. [Google Scholar] [CrossRef]
  41. Morris, A.; Beck, J.M.; Schloss, P.D.; Campbell, T.B.; Crothers, K.; Curtis, J.L.; Flores, S.C.; Fontenot, A.P.; Ghedin, E.; Huang, L.; et al. Comparison of the respiratory microbiome in healthy nonsmokers and smokers. Am. J. Respir. Crit. Care Med. 2013, 187, 1067–1075. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Beck, J.M.; Schloss, P.D.; Venkataraman, A.; Twigg, H., 3rd; Jablonski, K.A.; Bushman, F.D.; Campbell, T.B.; Charlson, E.S.; Collman, R.G.; Crothers, K.; et al. Multicenter comparison of lung and oral microbiomes of HIV-infected and HIV-uninfected individuals. Am. J. Respir. Crit. Care Med. 2015, 192, 1335–1344. [Google Scholar] [CrossRef] [PubMed]
  43. Goddard, A.F.; Staudinger, B.J.; Dowd, S.E.; Joshi-Datar, A.; Wolcott, R.D.; Aitken, M.L.; Fligner, C.L.; Singh, P.K. Direct sampling of cystic fibrosis lungs indicates that DNA-based analyses of upper-airway specimens can misrepresent lung microbiota. Proc. Natl. Acad. Sci. USA 2012, 109, 13769–13774. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Mathieu, E.; Escribano-Vazquez, U.; Descamps, D.; Cherbuy, C.; Langella, P.; Riffault, S.; Remot, A.; Thomas, M. Paradigms of lung microbiota functions in health and disease, particularly, in asthma. Front. Physiol. 2018, 9, 1168. [Google Scholar] [CrossRef] [PubMed]
  45. Chen, Y.; Gu, S.; Chen, Y.; Lu, H.; Shi, D.; Guo, J.; Wu, W.R.; Yang, Y.; Li, Y.; Xu, K.J.; et al. Six-month follow-up of gut microbiota richness in patients with COVID-19. Gut 2021, 2021, 324090. [Google Scholar] [CrossRef]
  46. Cao, J.; Wang, C.; Zhang, Y.; Lei, G.; Xu, K.; Zhao, N.; Lu, J.; Meng, F.; Yu, L.; Yan, J.; et al. Integrated gut virome and bacteriome dynamics in COVID-19 patients. Gut Microbes 2021, 13, 1887722. [Google Scholar] [CrossRef]
  47. Zhong, H.; Wang, Y.; Shi, Z.; Zhang, L.; Ren, H.; He, W.; Zhang, Z.; Zhu, A.; Zhao, J.; Xiao, F.; et al. Characterization of respiratory microbial dysbiosis in hospitalized COVID-19 patients. Cell Discov. 2021, 7, 23. [Google Scholar] [CrossRef]
  48. Lou, Z.; Zhao, H.; Lyu, G. Mechanism and intervention of mucosal immune regulation based on “lung and large intestine being interior-exteriorly related” theory of traditional chinese medicine. Zhejiang Da Xue Xue Bao Yi Xue Ban 2020, 49, 665–678. [Google Scholar]
  49. Schroeder, B.O.; Bäckhed, F. Signals from the gut microbiota to distant organs in physiology and disease. Nat. Med. 2016, 22, 1079–1089. [Google Scholar] [CrossRef]
  50. Feng, Q.; Chen, W.D.; Wang, Y.D. Gut microbiota: An integral moderator in health and disease. Front. Microbiol. 2018, 9, 151. [Google Scholar] [CrossRef]
  51. Chakradhar, S. A curious connection: Teasing apart the link between gut microbes and lung disease. Nat. Med. 2017, 23, 402–404. [Google Scholar] [CrossRef] [PubMed]
  52. Liu, T.H.; Zhang, C.Y.; Din, A.U.; Li, N.; Wang, Q.; Yu, J.Z.; Xu, Z.Y.; Li, C.X.; Zhang, X.M.; Yuan, J.L.; et al. Bacterial association and comparison between lung and intestine in rats. Biosci. Rep. 2020, 40, BSR20191570. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Ubags, N.; Marsland, B.J. Mechanistic insight into the function of the microbiome in lung diseases. Eur. Respir. J. 2017, 50, 1602467. [Google Scholar] [CrossRef] [PubMed]
  54. Spiljar, M.; Merkler, D.; Trajkovski, M. The immune system bridges the gut microbiota with systemic energy homeostasis: Focus on TLRs, mucosal barrier, and SCFAs. Front. Immunol. 2017, 8, 1353. [Google Scholar] [CrossRef] [Green Version]
  55. Wypych, T.P.; Pattaroni, C.; Perdijk, O.; Yap, C.; Trompette, A.; Anderson, D.; Creek, D.J.; Harris, N.L.; Marsland, B.J. Microbial metabolism of l-tyrosine protects against allergic airway inflammation. Nat. Immunol. 2021, 22, 279–286. [Google Scholar] [CrossRef]
  56. Midha, A.; Ebner, F.; Schlosser-Brandenburg, J.; Rausch, S.; Hartmann, S. Trilateral relationship: Ascaris, microbiota, and host cells. Trends Parasitol. 2021, 37, 251–262. [Google Scholar] [CrossRef]
  57. Depner, M.; Taft, D.H.; Kirjavainen, P.V.; Kalanetra, K.M.; Karvonen, A.M.; Peschel, S.; Schmausser-Hechfellner, E.; Roduit, C.; Frei, R.; Lauener, R.; et al. Maturation of the gut microbiome during the first year of life contributes to the protective farm effect on childhood asthma. Nat. Med. 2020, 26, 1766–1775. [Google Scholar] [CrossRef]
  58. Yin, L.; Li, X.X.; Ghosh, S.; Xie, C.; Chen, J.; Huang, H. Role of gut microbiota-derived metabolites on vascular calcification in CKD. J. Cell. Mol. Med. 2021, 25, 1332–1341. [Google Scholar] [CrossRef]
  59. Mjösberg, J.; Rao, A. Lung inflammation originating in the gut. Science 2018, 359, 36–37. [Google Scholar] [CrossRef]
  60. Huang, Y.; Mao, K.; Chen, X.; Sun, M.A.; Kawabe, T.; Li, W.; Usher, N.; Zhu, J.; Urban, J.F., Jr.; Paul, W.E.; et al. S1P-dependent interorgan trafficking of group 2 innate lymphoid cells supports host defense. Science 2018, 359, 114–119. [Google Scholar] [CrossRef] [Green Version]
  61. Gray, J.; Oehrle, K.; Worthen, G.; Alenghat, T.; Whitsett, J.; Deshmukh, H. Intestinal commensal bacteria mediate lung mucosal immunity and promote resistance of newborn mice to infection. Sci. Transl. Med. 2017, 9, eaaf9412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Tamburini, S.; Clemente, J.C. Gut microbiota: Neonatal gut microbiota induces lung immunity against pneumonia. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 263–264. [Google Scholar] [CrossRef] [PubMed]
  63. Dumas, A.; Bernard, L.; Poquet, Y.; Lugo-Villarino, G.; Neyrolles, O. The role of the lung microbiota and the gut–lung axis in respiratory infectious diseases. Cell Microbiol. 2018, 20, e12966. [Google Scholar] [CrossRef] [Green Version]
  64. Naidoo, C.C.; Nyawo, G.R.; Wu, B.G.; Walzl, G.; Warren, R.M.; Segal, L.N.; Theron, G. The microbiome and tuberculosis: State of the art, potential applications, and defining the clinical research agenda. Lancet Respir. Med. 2019, 7, 892–906. [Google Scholar] [CrossRef]
  65. Luo, M.; Liu, Y.; Wu, P.; Luo, D.X.; Sun, Q.; Zheng, H.; Hu, R.; Pandol, S.J.; Li, Q.F.; Han, Y.P.; et al. Alternation of gut microbiota in patients with pulmonary tuberculosis. Front. Physiol. 2017, 8, 882. [Google Scholar] [CrossRef] [PubMed]
  66. Namasivayam, S.; Sher, A.; Glickman, M.S.; Wipperman, M.F. The microbiome and tuberculosis: Early evidence for cross talk. mBio 2018, 9, e01420-18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Namasivayam, S.; Maiga, M.; Yuan, W.; Thovarai, V.; Costa, D.L.; Mittereder, L.R.; Wipperman, M.F.; Glickman, M.S.; Dzutsev, A.; Trinchieri, G.; et al. Longitudinal profiling reveals a persistent intestinal dysbiosis triggered by conventional anti-tuberculosis therapy. Microbiome 2017, 5, 71. [Google Scholar] [CrossRef]
  68. Khan, N.; Khan, N.; Mendonca, L.; Dhariwal, A.; Fontes, G.; Menzies, D.; Xia, J.; Divangahi, M.; King, I.L. Intestinal dysbiosis compromises alveolar macrophage immunity to Mycobacterium tuberculosis. Mucosal Immunol. 2019, 12, 772–783. [Google Scholar] [CrossRef]
  69. Hu, Y.; Yang, Q.; Liu, B.; Dong, J.; Sun, L.; Zhu, Y.; Su, H.; Yang, J.; Yang, F.; Chen, X.; et al. Gut microbiota associated with pulmonary tuberculosis and dysbiosis caused by anti-tuberculosis drugs. J. Infect. 2019, 78, 317–322. [Google Scholar] [CrossRef]
  70. Negi, S.; Pahari, S.; Bashir, H.; Agrewala, J.N. Gut microbiota regulates mincle mediated activation of lung dendritic cells to protect against mycobacterium tuberculosis. Front. Immunol. 2019, 10, 1142. [Google Scholar] [CrossRef] [Green Version]
  71. Samuelson, D.R.; Charles, T.P.; de la Rua, N.M.; Taylor, C.M.; Blanchard, E.E.; Luo, M.; Shellito, J.E.; Welsh, D.A. Analysis of the intestinal microbial community and inferred functional capacities during the host response to Pneumocystis pneumonia. Exp. Lung Res. 2016, 42, 425–439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Martin, R.M.; Cao, J.; Brisse, S.; Passet, V.; Wu, W.; Zhao, L.; Malani, P.N.; Rao, K.; Bachman, M.A. Molecular epidemiology of colonizing and infecting isolates of Klebsiella pneumoniae. mSphere 2016, 1, e00261-16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Barcik, W.; Boutin, R.; Sokolowska, M.; Finlay, B.B. The role of lung and gut microbiota in the pathology of asthma. Immunity 2020, 52, 241–255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Bowerman, K.L.; Rehman, S.F.; Vaughan, A.; Lachner, N.; Budden, K.F.; Kim, R.Y.; Wood, D.; Gellatly, S.L.; Shukla, S.D.; Wood, L.G.; et al. Disease-associated gut microbiome and metabolome changes in patients with chronic obstructive pulmonary disease. Nat. Commun. 2020, 11, 5886. [Google Scholar] [CrossRef] [PubMed]
  75. Stiemsma, L.; Reynolds, L.; Turvey, S.; Finlay, B. The hygiene hypothesis: Current perspectives and future therapies. ImmunoTargets Ther. 2015, 4, 143–157. [Google Scholar] [CrossRef] [Green Version]
  76. Sonnenburg, J.L.; Sonnenburg, E.D. Vulnerability of the industrialized microbiota. Science 2019, 366, eaaw9255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Ding, M.; Yang, B.; Ross, R.P.; Stanton, C.; Zhao, J.; Zhang, H.; Chen, W. Crosstalk between siga-coated bacteria in infant gut and early-life health. Trends Microbiol. 2021, 29, 725–735. [Google Scholar] [CrossRef]
  78. Bachus, H.; Kaur, K.; Papillion, A.M.; Marquez-Lago, T.T.; Yu, Z.; Ballesteros-Tato, A.; Matalon, S.; León, B. Impaired tumor-necrosis-factor-α-driven dendritic cell activation limits lipopolysaccharide-induced protection from allergic inflammation in infants. Immunity 2019, 50, 225–240. [Google Scholar] [CrossRef] [Green Version]
  79. Ohnmacht, C.; Park, J.H.; Cording, S.; Wing, J.B.; Atarashi, K.; Obata, Y.; Gaboriau-Routhiau, V.; Marques, R.; Dulauroy, S.; Fedoseeva, M.; et al. The microbiota regulates type 2 immunity through RORγt+ T cells. Science 2015, 349, 989–993. [Google Scholar] [CrossRef]
  80. Blaser, M.J.; Dominguez-Bello, M.G. The human microbiome before birth. Cell Host Microbe 2016, 20, 558–560. [Google Scholar] [CrossRef] [Green Version]
  81. Wu, B.G.; Sulaiman, I.; Tsay, J.J.; Perez, L.; Franca, B.; Li, Y.; Wang, J.; Gonzalez, A.N.; El-Ashmawy, M.; Carpenito, J.; et al. Episodic aspiration with oral commensals induces a MyD88-dependent, pulmonary th17 response that mitigates susceptibility to Streptococcus pneumoniae. Am. J. Respir. Crit. Care Med. 2020, 203, 1099–1111. [Google Scholar] [CrossRef] [PubMed]
  82. Yang, D.; Chen, X.; Wang, J.; Lou, Q.; Lou, Y.; Li, L.; Wang, H.; Chen, J.; Wu, M.; Song, X.; et al. Dysregulated Lung commensal bacteria drive interleukin-17b production to promote pulmonary fibrosis through their outer membrane vesicles. Immunity 2019, 50, 692–706. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Zhou, X.; Du, L.; Shi, R.; Chen, Z.; Zhou, Y.; Li, Z. Early-life food nutrition, microbiota maturation and immune development shape life-long health. Crit. Rev. Food Sci. Nutr. 2019, 59, S30–S38. [Google Scholar] [CrossRef] [PubMed]
  84. Wang, S.; Egan, M.; Ryan, C.A.; Boyaval, P.; Dempsey, E.M.; Ross, R.P.; Stanton, C. A good start in life is important—perinatal factors dictate early microbiota development and longer term maturation. FEMS Microbiol. Rev. 2020, 44, 763–781. [Google Scholar] [CrossRef] [PubMed]
  85. Chellappan, D.K.; Sze Ning, Q.L.; Su Min, S.K.; Bin, S.Y.; Chern, P.J.; Shi, T.P.; Ee Mei, S.W.; Yee, T.H.; Qi, O.J.; Thangavelu, L.; et al. Interactions between microbiome and lungs: Paving new paths for microbiome based bio-engineered drug delivery systems in chronic respiratory diseases. Chem. Biol. Interact. 2019, 310, 108732. [Google Scholar] [CrossRef] [PubMed]
  86. Clark, S.E. Commensal bacteria in the upper respiratory tract regulate susceptibility to infection. Curr. Opin. Immunol. 2020, 66, 42–49. [Google Scholar] [CrossRef]
  87. Hanada, S.; Pirzadeh, M.; Carver, K.Y.; Deng, J.C. Respiratory viral infection-induced microbiome alterations and secondary bacterial pneumonia. Front. Immunol. 2018, 9, 2640. [Google Scholar] [CrossRef] [Green Version]
  88. Pettigrew, M.M.; Gent, J.F.; Pyles, R.B.; Miller, A.L.; Nokso-Koivisto, J.; Chonmaitree, T. Viral-bacterial interactions and risk of acute otitis media complicating upper respiratory tract infection. J. Clin. Microbiol. 2011, 49, 3750–3755. [Google Scholar] [CrossRef] [Green Version]
  89. Bakaletz, L.O. Viral–bacterial co-infections in the respiratory tract. Curr. Opin. Microbiol. 2017, 35, 30–35. [Google Scholar] [CrossRef]
  90. Bellinghausen, C.; Rohde, G.G.; Savelkoul, P.H.; Wouters, E.F.; Stassen, F.R. Viral-bacterial interactions in the respiratory tract. J. Gen. Virol. 2016, 97, 3089–3102. [Google Scholar] [CrossRef] [Green Version]
  91. Brinker, P.; Fontaine, M.C.; Beukeboom, L.W.; Salles, J.F. Host, Symbionts, and the Microbiome: The Missing Tripartite Interaction. Trends Microbiol. 2019, 27, 480–488. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Kiedrowski, M.R.; Bomberger, J.M. Viral-bacterial co-infections in the cystic fibrosis respiratory tract. Front Immunol. 2018, 9, 3067. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Shahbazi, R.; Yasavoli-Sharahi, H.; Alsadi, N.; Ismail, N.; Matar, C. Probiotics in treatment of viral respiratory infections and neuroinflammatory disorders. Molecules 2020, 25, 4891. [Google Scholar] [CrossRef] [PubMed]
  94. Tonetti, F.R.; Islam, M.A.; Vizoso-Pinto, M.G.; Takahashi, H.; Kitazawa, H.; Villena, J. Nasal priming with immunobiotic lactobacilli improves the adaptive immune response against influenza virus. Int. Immunopharmacol. 2020, 78, 106115. [Google Scholar] [CrossRef] [PubMed]
  95. Bae, J.Y.; Kim, J.I.; Park, S.; Yoo, K.; Kim, I.H.; Joo, W.; Ryu, B.H.; Park, M.S.; Lee, I.; Park, M.S. Effects of Lactobacillus plantarum and leuconostoc mesenteroides probiotics on human seasonal and Avian Influenza Viruses. J. Microbiol. Biotechnol. 2018, 28, 893–901. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Zelaya, H.; Laiño, J.; Villena, J.; Alvarez, S.; Agüero, G. Lactobacillus rhamnosus CRL1505 beneficially modulates the immunocoagulative response after pneumococcal infection in immunocompromised malnourished mice. Can. J. Microbiol. 2013, 59, 684–693. [Google Scholar] [CrossRef]
  97. Villena, J.; Oliveira, M.L.; Ferreira, P.C.; Salva, S.; Alvarez, S. Lactic acid bacteria in the prevention of pneumococcal respiratory infection: Future opportunities and challenges. Int. Immunopharmacol. 2011, 11, 1633–1645. [Google Scholar] [CrossRef]
  98. Antunes, K.H.; Fachi, J.L.; de Paula, R.; da Silva, E.F.; Pral, L.P.; Dos Santos, A.Á.; Dias, G.; Vargas, J.E.; Puga, R.; Mayer, F.Q.; et al. Microbiota-derived acetate protects against respiratory syncytial virus infection through a GPR43-type 1 interferon response. Nat. Commun. 2019, 10, 3273. [Google Scholar] [CrossRef] [Green Version]
  99. Wu, T.; Li, H.; Su, C.; Xu, F.; Yang, G.; Sun, K.; Xu, M.; Lv, N.; Meng, B.; Liu, Y.; et al. Microbiota-derived short-chain fatty acids promote LAMTOR2-mediated immune responses in macrophages. mSystems 2020, 5, e00587-20. [Google Scholar] [CrossRef]
  100. Sencio, V.; Barthelemy, A.; Tavares, L.P.; Machado, M.G.; Soulard, D.; Cuinat, C.; Queiroz-Junior, C.M.; Noordine, M.L.; Salomé-Desnoulez, S.; Deryuter, L.; et al. Gut dysbiosis during influenza contributes to pulmonary pneumococcal superinfection through altered short-chain fatty acid production. Cell Rep. 2020, 30, 2934–2947. [Google Scholar] [CrossRef] [Green Version]
  101. Ji, J.J.; Sun, Q.M.; Nie, D.Y.; Wang, Q.; Zhang, H.; Qin, F.F.; Wang, Q.S.; Lu, S.F.; Pang, G.M.; Lu, Z.G. Probiotics protect against RSV infection by modulating the microbiota-alveolar-macrophage axis. Acta Pharmacol. Sin. 2021, 42, 1630–1641. [Google Scholar] [CrossRef] [PubMed]
  102. Yang, K.; Dong, W. Perspectives on probiotics and bronchopulmonary dysplasia. Front. Pediatr. 2020, 8, 570247. [Google Scholar] [CrossRef] [PubMed]
  103. Zuo, T.; Liu, Q.; Zhang, F.; Yeoh, Y.K.; Wan, Y.; Zhan, H.; Lui, G.; Chen, Z.; Li, A.; Cheung, C.P.; et al. Temporal landscape of human gut virome in SARS-CoV-2 infection and severity. Microbiome 2021, 9, 9. [Google Scholar] [CrossRef] [PubMed]
  104. Rajput, S.; Paliwal, D.; Naithani, M.; Kothari, A.; Meena, K.; Rana, S. COVID-19 and gut microbiota: A potential connection. Indian J. Clin. Biochem. 2021, 36, 266–277. [Google Scholar] [CrossRef] [PubMed]
  105. Dhar, D.; Mohanty, A. Gut microbiota and Covid-19—Possible link and implications. Virus Res. 2020, 285, 198018. [Google Scholar] [CrossRef] [PubMed]
  106. Piersigilli, F.; Grambezen, B.V.; Hocq, C.; Danhaive, O. Nutrients and microbiota in lung diseases of prematurity: The placenta-gut-lung triangle. Nutrients 2020, 12, 469. [Google Scholar] [CrossRef] [Green Version]
  107. Takahiro, Y.; Daisuke, T.; Koji, H. The diet-microbiota-metabolite axis regulates the host physiology. J. Biochem. 2016, 160, 1–10. [Google Scholar]
Figure 1. The acquisition and maturation of symbiotic microbiome. The colonization of symbiotic microbiome mainly impacted by delivery mode, antibiotic usage, dietary structure, environment exposure, pathogenic infections, and other affecting factors.
Figure 1. The acquisition and maturation of symbiotic microbiome. The colonization of symbiotic microbiome mainly impacted by delivery mode, antibiotic usage, dietary structure, environment exposure, pathogenic infections, and other affecting factors.
Cells 11 00916 g001
Figure 2. Cross-talk between the gut and lung through the gut-lung axis. Communications between the gut and lung can be realized by multiple signals transmitted from intestine to the lung. Firstly, certain microbial species of lung and intestine microbiota can directly exchange through the lymph circulation. Secondly, various kinds of bioactive metabolites produced by the gut microbiota and lung microbiota can be changed via blood circulation. Additionally, the innate lymphoid cells (ILC2 and3) can migrate from intestinal lamina propria to the lung.
Figure 2. Cross-talk between the gut and lung through the gut-lung axis. Communications between the gut and lung can be realized by multiple signals transmitted from intestine to the lung. Firstly, certain microbial species of lung and intestine microbiota can directly exchange through the lymph circulation. Secondly, various kinds of bioactive metabolites produced by the gut microbiota and lung microbiota can be changed via blood circulation. Additionally, the innate lymphoid cells (ILC2 and3) can migrate from intestinal lamina propria to the lung.
Cells 11 00916 g002
Figure 3. Strategies targeting the symbiotic microbiota to prevent and control respiratory diseases. Probiotics consumption, nutrient intervention, and fecal microbiota transplantation targeting the gut and lung microbiota could confer health benefits for respiratory diseases.
Figure 3. Strategies targeting the symbiotic microbiota to prevent and control respiratory diseases. Probiotics consumption, nutrient intervention, and fecal microbiota transplantation targeting the gut and lung microbiota could confer health benefits for respiratory diseases.
Cells 11 00916 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Li, Z.; Li, Y.; Sun, Q.; Wei, J.; Li, B.; Qiu, Y.; Liu, K.; Shao, D.; Ma, Z. Targeting the Pulmonary Microbiota to Fight against Respiratory Diseases. Cells 2022, 11, 916. https://doi.org/10.3390/cells11050916

AMA Style

Li Z, Li Y, Sun Q, Wei J, Li B, Qiu Y, Liu K, Shao D, Ma Z. Targeting the Pulmonary Microbiota to Fight against Respiratory Diseases. Cells. 2022; 11(5):916. https://doi.org/10.3390/cells11050916

Chicago/Turabian Style

Li, Zongjie, Yuhao Li, Qing Sun, Jianchao Wei, Beibei Li, Yafeng Qiu, Ke Liu, Donghua Shao, and Zhiyong Ma. 2022. "Targeting the Pulmonary Microbiota to Fight against Respiratory Diseases" Cells 11, no. 5: 916. https://doi.org/10.3390/cells11050916

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop