Gut-lung Axis
11th Sep, 2019

This post is specifically for health practitioners. Create a free account

Alternatively, view general public info.

Learn more about vital.ly

Gut-lung axis Essentials 

Chronic lung disorders, such as asthma, chronic obstructive pulmonary disease (COPD), cystic fibrosis (CF) and idiopathic pulmonary fibrosis (IPF) are usually accompanied by intestinal symptoms which imply the existence of an immunological link between the gut and the lungs. This link is referred to as the gut-lung axis (1). The composition of the gut microbiota affects immune responses and development of inflammation, not only in the gastrointestinal tract but also in distal sites such as the lungs (2).

The existence of microbial organisms in the lungs, referred to as the lung microbiome, has recently been discovered leading to research into their effect on lung function and disease. Changes in gut microbiota could affect lung microbiota, and vice versa (3). Dysbiosis in gut and lung microbiota have been linked to increased incidence and severity of asthma, allergy, CF, COPD and respiratory infections (1,4,5).

A healthy gut microbiota maintains homeostatic local immune responses through the exposure of structural ligands (e.g. lipopolysaccharide) and secreted metabolites (e.g. short-chain fatty acids). Invading microorganisms and absorbed metabolites influence circulating lymphocytes and contribute to the regulation of the systemic immune responses. Gut microbiota dysbiosis results in altered microbiota-derived signals which lead to a changed immune response. In early life, when the immune system is still developing, this disturbance can substantially alter the way in which the immune system perceives its surroundings in later life, which can lead to the development of chronic inflammatory disorders in the gut and lung. In adulthood, dysbiosis of the gut microbiota (e.g. exposure to cigarette smoke) can cause systemic inflammation and an overgrowth of opportunistic pathogens which can lead to chronic inflammation in the lungs (5). Exacerbations of chronic lung diseases occur as an uncontrolled and inappropriate inflammatory response regulated in part by the gut microbiota (2).

The gut microbiome communicates with the lung via multiple bi-directional pathways (Figure 1) and commensal gut microbiota appear to be critical in lung defence against bacterial invasion (2,6).


Figure 1. Gut-lung communication (7) CC BY 4.0

Gut-lung axis Essentials - Fig 1

 

Research into the gut-lung axis is still in its infancy, but it may potentially serve as a new direction for lung disease treatment, with some of the existing probiotic strains showing beneficial effects on lung health (4,5). A better understanding of microbiome interactions with inflammation and other major risk factors for chronic lung disease such as host genetics and cigarette smoking (Figure 2) will aid in optimising current treatments and in managing chronic lung conditions.

 

Figure 2. Risk factors for chronic respiratory diseases and associated pathology with microbiome. Adapted from (8) CC BY-NC-ND 4.0

 

Gut-lung axis Essentials - Fig 2

 

Traditional Understanding

Historically, traditional culture-based studies and classic teaching indicated that normal lungs are free from bacteria, and this notion has persisted in contemporary medicine (1). Recent evidence has identified microbial organisms in the lungs in healthy individuals (the lung microbiome).

 

The lung microbiome

In humans, lung microbiota have been identified in healthy donors and in patients with chronic lung disease (1). The total number of bacteria in lung tissue is relatively small compared with that of gut microbiota. As with the gut microbiome, the lung microbiome is considered to be important to immunity by priming the immune system. Dysbiosis in the lung predisposes to the development of respiratory disease and may be relevant in the aetiology and/or progression of chronic lung diseases (1,9). It is not clear whether dysbiosis is a cause or a consequence of immune dysregulation and disease initiation or progression. Further study of the relationship between lung microbiota and respiratory disease might result in future manipulation of lung microbiota as a rational and promising therapy (1). It has been suggested that exacerbations of chronic lung disease are more likely due to episodes of dysbiosis of the microbial ecosystem of the respiratory tract rather than infection (2).

There are many factors, such as anatomical injuries, pathological effects, physiological changes and immune system defects which could disrupt the lung microbiota and result in chronic lung diseases (1). The influence of gut microbiota on lung immunity has been recently explored. Changes in the gut microbiome have been linked with lung immunity and clinical outcomes in chronic respiratory diseases. Conversely, the lung microbiota also influences the gut microbiota through the bloodstream (2).

 

Latest Research

Asthma

Asthma is a chronic and multifactorial disease thought to be caused by a combination of genetic and environmental factors including air pollution and allergens.

The lung microbiome is altered during the development of allergic asthma and it has been suggested that both the gut and lung microbiota, as well as the local and systemic immune response, play a role in predisposing an individual to asthma (10).

Lung microbiome and asthma

  • The composition of lung microbiota in individuals with asthma is different from healthy controls (1). The underlying inflammation in atopic asthma appears to be related to the lung microbiota composition (2,11).
  • Treatment with inhaled corticosteroids is associated with changes in the airway inflammatory response to microbiota (12,13).

Gut microbiome and asthma

A correlation has been demonstrated between gut microbial diversity and asthma.

  • Several maternal factors are associated with childhood asthma risk (Figure 3).
  • Infant antibiotic use during the first year of life is the greatest predictor of childhood asthma (14,15).
  • Colonization with fewer bacteria species during the first month of life is associated with an asthma diagnosis at age 7 (16).
  • There appears to be a critical window of colonization that occurs during early infancy in which gut microbial communities shape immune maturation and confer susceptibility to allergic airway inflammation (17). Studies have examined gut microbiota maturation over the first year of life in infants at high risk for asthma, and whether it is modifiable by early-life Lactobacillus supplementation (18). Results showed that early-life gut microbial development is distinct, but plastic, offering a novel strategy for early-life preventive interventions.
  • Adults with long-term asthma were found to have a decreased abundance of Bifidobacteria (19).
  • Faecal microbial transplant therapy for asthma may represent a therapeutic approach in the foreseeable future (20).

 

Figure 3. Early life factors influencing gut microbial composition and risk of allergy and asthma (15,21,22,23,24).

 

The idea of primary prevention of asthma and allergy by manipulation of environmental microbial exposures seems a very promising approach. The first months of life, or even the prenatal period, seem to be the most sensitive for effective intervention. Identification of individuals with a high asthma risk at this early stage would be very important (25). A better understanding of microbiome-driven pathophysiology and inflammation, in conjunction with the interaction of major risk factors for asthma development, such as host genetics and tobacco smoke, would aid in optimizing current treatments and in managing this chronic lung condition (26).

The Hygiene Hypothesis and Asthma

An association between decreasing frequency of childhood infections and the increasing development of asthma and allergies has been recognized for years, giving rise to the hygiene hypothesis: that a decrease in infectious exposures early in life (due to improved hygiene, sanitation, cleaner homes and environments) results in deranged mucosal tolerance and increased autoimmune pathology. Early bacterial stimulation appears to reduce asthma susceptibility by helping the immune system develop lifelong tolerance to innocuous antigens (2). Hygiene-associated deviations in gut microflora composition may be the environmental factor underlying the development of atopy and asthma in genetically predisposed individuals (27).

Several cross-sectional studies in different countries have compared the prevalence of asthma, hay fever and allergic sensitization in children living in farming and non-farming environments. They showed that children born and raised in a farming environment were less prone to the development of atopic symptoms and asthma later in life. This protective effect was even stronger in adults that had remained in the farming environment. These observations support the hypothesis that exposure to a wide range of diverse microbial signals during the first few months of life has a major impact on susceptibility to the development of asthma (7).

 

Chronic Obstructive Pulmonary Disease

COPD is a type of obstructive lung disease characterized by long-term poor airflow. The gut and lung microbiota contribute to the pathogenesis and exacerbations of COPD. Studies indicate that bacterial colonization in the lung is associated with a chronic bronchitic phenotype with an increased risk of exacerbations and accelerated loss of lung function.

Once innate lung defence mechanisms are disrupted by an initiating factor (e.g. smoke exposure or air pollution) the microbiome in the airway is disrupted which drives inflammatory processes in COPD, causing progressive airflow obstruction and parenchymal damage (2).

Evidence indicates that bronchus protection is a consequence of antigen handling in the gut via T cell-dependent recruitment and activation of neutrophils and secretion of antibacterial factors (2).

The lung microbiotas in patients with mild and moderate COPD are similar to the lung microbiota in healthy controls. The change of lung microbiota may only be found in patients with advanced COPD (10).

 

Cystic Fibrosis

Cystic fibrosis (CF) is an inherited disorder that mainly affects the lungs and is characterised by a build-up of thick, intransient mucus linings of the digestive and respiratory mucosa. The syndrome of CF in the lungs displays a progressive development of bronchiectasis  and obstructive lung disease. The relationship between lung microbiota and the pathogenesis of CF is still controversial, however, the gut microbiota is considered as a link in CF.

  • Gut microbiota and their metabolites can influence host metabolism and immune function, which are heavily implicated in CF progression and comorbidity management (28). Individuals with CF display a significantly altered gut microbiota, including reduced microbial diversity compared to healthy controls (29).
  • Intakes of some dietary flavonoids may be associated with gut microbiota variations with potential consequences for metabolism, immune function, and inflammation, which are important in CF lung disease and co-morbidity management (28).
  • Limited preliminary studies have shown that oral probiotics may help reduce the frequency of pulmonary exacerbation and associated hospital admissions, as well as gut inflammation and discomfort in children and adults with CF (30,31,32,33,34).
  • The use of probiotics is rational and has been in practice since the 1980s, despite the paucity of supporting evidence and lack of specific guidelines (31).
  • Vitamin D status may be essential for the development of a healthy gut microbiome, particularly in conditions defined by chronic mucosal inflammation such as CF. It has been suggested that using high dose vitamin D may be used to favourably alter the aberrant mucosa seen in patients with CF. This may result in improved clinical outcomes in association with a low inflammatory environment that allows beneficial bacteria to outcompete opportunistic pathogens. Vitamin D also potentially plays a role in pulmonary function (35).

 

Tuberculosis

  • Tuberculosis (TB) outcomes can be affected by gut microbiota. Maintaining homeostasis of gut microbiota may be beneficial for recovery and prevention of recurrence of tuberculosis infection (36).
  • The cross-talk between the lung and gut microbiome, as well as innate and adaptive immune cells that may link these two mucosal sites, appear to be important in the prevention, pathogenesis, and treatment of TB (37).
  • Narrow-spectrum, first-line anti-TB drugs have dramatic effects on microbiome diversity and immunity, which persists even after the completion of TB drug therapy (38,39). The persistent dysbiosis that accompanies curative anti-TB treatment could contribute to post-treatment susceptibility to mycobacterial reinfection, as well as infection with other diseases linked with altered immune responses (40,41).

 

Experimental Studies

Recent experimental studies are summarised in Table 1.

 

Table 1. Recent experimental studies involving the gut-lung axis

Gut-lung axis Essentials - Table 1

 

 

Loading...
References
1Wang J, Li F, Tian Z. Role of microbiota on lung homeostasis and diseases. Sci China Life Sci. 2017;60(12):1407–15.
2He Y, Wen Q, Yao F, Xu D, Huang Y, Wang J. Gut-lung axis: The microbial contributions and clinical implications. Crit Rev Microbiol. 2017 Feb;43(1):81–95.
3Bingula R, Filaire M, Radosevic-Robin N, Berthon J-Y, Bernalier-Donadille A, Vasson M-P, et al. Characterisation of gut, lung, and upper airways microbiota in patients with non-small cell lung carcinoma: Study protocol for case-control observational trial. Medicine (Baltimore). 2018 Dec;97(50):e13676.
4Anand S, Mande SS. Diet, Microbiota and Gut-Lung Connection. Front Microbiol. 2018;9:2147.
5Budden KF, Gellatly SL, Wood DLA, Cooper MA, Morrison M, Hugenholtz P, et al. Emerging pathogenic links between microbiota and the gut-lung axis. Nat Rev Microbiol. 2017;15(1):55–63.
6McAleer JP, Kolls JK. Contributions of the intestinal microbiome in lung immunity. Eur J Immunol. 2018;48(1):39–49.
7Mathieu E, Escribano-Vazquez U, Descamps D, Cherbuy C, Langella P, Riffault S, et al. Paradigms of Lung Microbiota Functions in Health and Disease, Particularly, in Asthma. Front Physiol. 2018;9:1168.
8Shukla SD, Budden KF, Neal R, Hansbro PM. Microbiome effects on immunity, health and disease in the lung. Clin Transl Immunology. 2017 Mar;6(3):e133.
9Tamburini S, Clemente JC. Gut microbiota: Neonatal gut microbiota induces lung immunity against pneumonia. Nat Rev Gastroenterol Hepatol. 2017;14(5):263–4.
10Dickson RP, Erb-Downward JR, Martinez FJ, Huffnagle GB. The Microbiome and the Respiratory Tract. Annu Rev Physiol. 2016;78:481–504.
11Turturice BA, McGee HS, Oliver B, Baraket M, Nguyen BT, Ascoli C, et al. Atopic asthmatic immune phenotypes associated with airway microbiota and airway obstruction. PLoS One. 2017 Oct 20;12(10).
12Durack J, Lynch SV, Nariya S, Bhakta NR, Beigelman A, Castro M, et al. Features of the bronchial bacterial microbiome associated with atopy, asthma and responsiveness to inhaled corticosteroid treatment. J Allergy Clin Immunol. 2017 Jul;140(1):63–75.
13Ozturk AB, Turturice BA, Perkins DL, Finn PW. The Potential for Emerging Microbiome-Mediated Therapeutics in Asthma. Curr Allergy Asthma Rep. 2017 Aug 10;17(9):62.
14Korpela K, Salonen A, Virta LJ, Kekkonen RA, Forslund K, Bork P, et al. Intestinal microbiome is related to lifetime antibiotic use in Finnish pre-school children. Nat Commun. 2016 Jan 26;7.
15Wu P, Feldman AS, Rosas-Salazar C, James K, Escobar G, Gebretsadik T, et al. Relative Importance and Additive Effects of Maternal and Infant Risk Factors on Childhood Asthma. PLoS One. 2016 Mar 22;11(3).
16Abrahamsson 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 Jun;44(6):842–50.
17Ver Heul A, Planer J, Kau AL. The Human Microbiota and Asthma. Clin Rev Allergy Immunol. 2018 Nov 13;
18Durack J, Kimes NE, Lin DL, Rauch M, McKean M, McCauley K, et al. Delayed gut microbiota development in high-risk for asthma infants is temporarily modifiable by Lactobacillus supplementation. Nat Commun. 2018 Feb 16;9.
19Hevia A, Milani C, López P, Donado CD, Cuervo A, González S, et al. Allergic Patients with Long-Term Asthma Display Low Levels of Bifidobacterium adolescentis. PLoS One. 2016 Feb 3;11(2).
20Kang Y, Cai Y. Future prospect of faecal microbiota transplantation as a potential therapy in asthma. Allergol Immunopathol (Madr). 2018 Jun;46(3):307–9.
21Lapin B, Piorkowski J, Ownby D, Freels S, Chavez N, Hernandez E, et al. The Relationship between Prenatal Antibiotic Use and Asthma in At-Risk Children. Ann Allergy Asthma Immunol. 2015 Mar;114(3):203–7.
22Metsälä J, Lundqvist A, Virta LJ, Kaila M, Gissler M, Virtanen SM. Prenatal and post-natal exposure to antibiotics and risk of asthma in childhood. Clin Exp Allergy. 2015 Jan;45(1):137–45.
23Mulder B, Pouwels KB, Schuiling-Veninga CCM, Bos HJ, de Vries TW, Jick SS, et al. Antibiotic use during pregnancy and asthma in preschool children: the influence of confounding. Clin Exp Allergy. 2016;46(9):1214–26.
24Panzer AR, Lynch SV. Influence and effect of the human microbiome in allergy and asthma. Curr Opin Rheumatol. 2015 Jul;27(4):373–80.
25Sozańska B. Microbiome in the primary prevention of allergic diseases and bronchial asthma. Allergol Immunopathol (Madr). 2019 Feb;47(1):79–84.
26Frati F, Salvatori C, Incorvaia C, Bellucci A, Di Cara G, Marcucci F, et al. The Role of the Microbiome in Asthma: The Gut−Lung Axis. Int J Mol Sci. 2018 Dec 30;20(1).
27Mazmanian SK, Liu CH, Tzianabos AO, Kasper DL. An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune system. Cell. 2005 Jul 15;122(1):107–18.
28Li L, Somerset S. Associations between Flavonoid Intakes and Gut Microbiota in a Group of Adults with Cystic Fibrosis. Nutrients. 2018 Sep 7;10(9).
29Burke DG, Fouhy F, Harrison MJ, Rea MC, Cotter PD, O’Sullivan O, et al. The altered gut microbiota in adults with cystic fibrosis. BMC Microbiol. 2017 09;17(1):58.
30Bruzzese 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 Feb 19;9(2).
31del Campo R, Garriga M, Pérez-Aragón A, Guallarte P, Lamas A, Máiz L, et al. Improvement of digestive health and reduction in proteobacterial populations in the gut microbiota of cystic fibrosis patients using a Lactobacillus reuteri probiotic preparation: a double blind prospective study. J Cyst Fibros. 2014 Dec;13(6):716–22.
32Jafari S-A, Mehdizadeh-Hakkak A, Kianifar H-R, Hebrani P, Ahanchian H, Abbasnejad E. Effects of Probiotics on Quality of Life in Children with Cystic Fibrosis; A Randomized Controlled Trial. Iran J Pediatr. 2013 Dec;23(6):669–74.
33Van Biervliet S, Declercq D, Somerset S. Clinical effects of probiotics in cystic fibrosis patients: A systematic review. Clin Nutr ESPEN. 2017 Apr;18:37–43.
34Weiss B, Bujanover Y, Yahav Y, Vilozni D, Fireman E, Efrati O. Probiotic supplementation affects pulmonary exacerbations in patients with cystic fibrosis: a pilot study. Pediatr Pulmonol. 2010 Jun;45(6):536–40.
35Kanhere M, Chassaing B, Gewirtz AT, Tangpricha V. Role of vitamin D on gut microbiota in cystic fibrosis. J Steroid Biochem Mol Biol. 2018;175:82–7.
36Luo M, Liu Y, Wu P, Luo D-X, Sun Q, Zheng H, et al. Alternation of Gut Microbiota in Patients with Pulmonary Tuberculosis. Front Physiol. 2017;8:822.
37Gupta N, Kumar R, Agrawal B. New Players in Immunity to Tuberculosis: The Host Microbiome, Lung Epithelium, and Innate Immune Cells. Front Immunol. 2018;9:709.
38Hu Y, Yang Q, Liu B, Dong J, Sun L, Zhu Y, et al. Gut microbiota associated with pulmonary tuberculosis and dysbiosis caused by anti-tuberculosis drugs. J Infect. 2018 Aug 11;
39Namasivayam S, Maiga M, Yuan W, Thovarai V, Costa DL, Mittereder LR, et al. Longitudinal profiling reveals a persistent intestinal dysbiosis triggered by conventional anti-tuberculosis therapy. Microbiome. 2017 07;5(1):71.
40Hong B-Y, Maulén NP, Adami AJ, Granados H, Balcells ME, Cervantes J. Microbiome Changes during Tuberculosis and Antituberculous Therapy. Clin Microbiol Rev. 2016 Oct;29(4):914–25.
41Wipperman MF, Fitzgerald DW, Juste MAJ, Taur Y, Namasivayam S, Sher A, et al. Antibiotic treatment for Tuberculosis induces a profound dysbiosis of the microbiome that persists long after therapy is completed. Sci Rep. 2017 Sep 7;7.
42Gray LEK, O’Hely M, Ranganathan S, Sly PD, Vuillermin P. The Maternal Diet, Gut Bacteria, and Bacterial Metabolites during Pregnancy Influence Offspring Asthma. Front Immunol. 2017 Mar 31;8.
43Schuijt TJ, Lankelma JM, Scicluna BP, de Sousa e Melo F, Roelofs JJTH, de Boer JD, et al. The gut microbiota plays a protective role in the host defence against pneumococcal pneumonia. Gut. 2016 Apr;65(4):575–83.
44Arrieta M-C, Stiemsma LT, Dimitriu PA, Thorson L, Russell S, Yurist-Doutsch S, et al. Early infancy microbial and metabolic alterations affect risk of childhood asthma. Sci Transl Med. 2015 Sep 30;7(307):307ra152.
45Kozakova H, Schwarzer M, Tuckova L, Srutkova D, Czarnowska E, Rosiak I, et al. Colonization of germ-free mice with a mixture of three lactobacillus strains enhances the integrity of gut mucosa and ameliorates allergic sensitization. Cell Mol Immunol. 2016 Mar;13(2):251–62.
46Kim HY, Lee HJ, Chang Y-J, Pichavant M, Shore SA, Fitzgerald KA, et al. IL-17 producing innate lymphoid cells and the NLRP3 inflammasome facilitate obesity-associated airway hyperreactivity. Nat Med. 2014 Jan;20(1):54–61.
47Verheijden KAT, Willemsen LEM, Braber S, Leusink-Muis T, Delsing DJM, Garssen J, et al. Dietary galacto-oligosaccharides prevent airway eosinophilia and hyperresponsiveness in a murine house dust mite-induced asthma model. Respir Res. 2015 Feb 7;16:17.
48Verheijden KAT, Willemsen LEM, Braber S, Leusink-Muis T, Jeurink PV, Garssen J, et al. The development of allergic inflammation in a murine house dust mite asthma model is suppressed by synbiotic mixtures of non-digestible oligosaccharides and Bifidobacterium breve M-16V. Eur J Nutr. 2016;55:1141–51.
49Maslowski KM, Vieira AT, Ng A, Kranich J, Sierro F, Yu D, et al. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature. 2009 Oct 29;461(7268):1282–6.
50Trompette A, Gollwitzer ES, Yadava K, Sichelstiel AK, Sprenger N, Ngom-Bru C, et al. Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat Med. 2014 Feb;20(2):159–66.
51Thorburn AN, McKenzie CI, Shen S, Stanley D, Macia L, Mason LJ, et al. Evidence that asthma is a developmental origin disease influenced by maternal diet and bacterial metabolites. Nat Commun. 2015 Jun 23;6:7320.
52Dumas A, Corral D, Colom A, Levillain F, Peixoto A, Hudrisier D, et al. The Host Microbiota Contributes to Early Protection Against Lung Colonization by Mycobacterium tuberculosis. Front Immunol. 2018;9:2656.
53Bradley KC, Finsterbusch K, Schnepf D, Crotta S, Llorian M, Davidson S, et al. Microbiota-Driven Tonic Interferon Signals in Lung Stromal Cells Protect from Influenza Virus Infection. Cell Reports. 2019 Jul;28(1):245-256.e4.
54Mortaz E, Adcock IM, Ricciardolo FLM, Varahram M, Jamaati H, Velayati AA, et al. Anti-Inflammatory Effects of Lactobacillus Rahmnosus and Bifidobacterium Breve on Cigarette Smoke Activated Human Macrophages. PLoS One. 2015 Aug 28;10(8).
55Barcik W, Pugin B, Brescó MS, Westermann P, Rinaldi A, Groeger D, et al. Bacterial secretion of histamine within the gut influences immune responses within the lung. Allergy. 2018 Dec 27;
56Barcik W, Pugin B, Westermann P, Perez NR, Ferstl R, Wawrzyniak M, et al. Histamine-secreting microbes are increased in the gut of adult asthma patients. Journal of Allergy and Clinical Immunology. 2016 Nov;138(5):1491-1494.e7.