Abstract
Early life dietary patterns and timely maturation of mucosa‐associated microbial communities are important factors influencing immune development and for establishing robust immune tolerance networks. Microbial fermentation of dietary components in vivo generates a vast array of molecules, some of which are integral components of the molecular circuitry that regulates immune and metabolic functions. These in turn protect against aberrant inflammatory processes and promote effector immune responses that quickly eliminate pathogens. Multiple studies suggest that changes in dietary habits, altered microbiome composition, and microbial metabolism are associated with asthma risk and disease severity. While it remains unclear whether these microbiome alterations are a cause or consequence of dysregulated immune responses, there is significant potential for using diet in targeted manipulations of the gut microbiome and its metabolic functions in promoting immune health. In this article, we will summarize our knowledge to date on the role of dietary patterns and microbiome activities on immune responses within the airways. Given the malleability of the human microbiome, its integration into the immune system, and its responsiveness to diet, this makes it a highly attractive target for therapeutic and nutritional intervention in children with asthma.
Keywords: asthma, diet, inflammation, microbiome, microbiota
Key Message.
Diet–microbiome interactions in early life are emerging as key events that shape the infant's immune response to infectious and noninfectious encounters. Research on how specific dietary interventions impact the microbiota to generate immunoregulatory metabolites can be leveraged to develop more effective diets and molecules that improve human immune health.
1. INTRODUCTION
Recent decades have seen a rapid increase in chronic inflammatory disorders in children due to inappropriate or misdirected immune responses accompanied by insufficient development of immune regulatory networks. It is generally accepted that changes in environment, lifestyle, and dietary factors may play a role in the miseducation or deficient training of the immune system. 1 , 2 , 3 Epidemiological studies have suggested associations between the migration from traditional farming to urban environments, increase in processed food intake, lack of contact with animals, and increased exposure to epithelial barrier damaging agents with the increased incidence of asthma, atopic dermatitis and food allergy. 4 However, it is only relatively recently that the microbiota has been recognized as an important conduit for mediating environment and dietary effects on the regulation of immune tolerance networks.
Following birth, infants acquire microbes from their mother, other family members, animals, and their environmental exposures. 5 The complexity of the infant microbiome develops and matures during the first years of life and is supported by breastfeeding and the introduction of diverse complementary and solid foods. When microbiota acquisition and/or dietary supports are limited, this can lead to a delay in microbiota maturation. A shift away from traditional diets rich in plant‐based foods to highly processed foods is thought to be particularly important for negatively affecting microbiome diversity and composition, species‐specific characteristics, microbial metabolism, and immunological tolerance. 6 , 7 In fact, the American Gut Study showed that eating 30 plant‐based foods per week was associated with the highest levels of gut microbial diversity. 8 The consequences of altered microbiota composition or altered metabolic output include disrupted communication between the microbiota and the host immune system, which negatively influences immune homeostatic networks as the balance between immune tolerance and inflammation within tissues is regulated in part by the crosstalk between immune cells and the microbiota. 9 These mechanisms are important not only for protection against allergy and asthma but also for effective immune responses to pathogens such as SARS‐CoV‐2. 10 , 11 , 12 , 13 Microbial structural components (e.g., lipopolysaccharides, lipoteichoic acid, polysaccharide A, lipoproteins, peptidoglycans, and extracellular polysaccharides), genetic material (e.g., unmethylated CpG dinucleotides), microbe modified host factors (e.g., secondary bile acids), and metabolites derived from metabolism of dietary components (e.g., short‐chain fatty acids (SCFAs) and indoles) all impact immunological responses via a range of immune sensory systems including pattern recognition receptors (PRRs), G protein‐coupled receptors (GPCRs), aryl hydrocarbon receptor (AhR), nuclear hormone receptors such as the farnesoid X receptor (FXR) or can directly modulate gene expression through epigenetic mechanisms (Figure 1).
FIGURE 1.
Microbial factors impacting lung immune responses. Within the gut, bacterial metabolism of dietary components (e.g., fibers and amino acids) and host‐derived secretions (e.g., primary bile acids and mucins) generates metabolites including SCFAs, indoles and secondary bile acids that influence immune cells via GPCRs, aryl hydrocarbon receptor and nuclear receptors. Metabolites can be absorbed and have direct effects on distant organs such as the lungs. Cells exposed to bacterial factors in the gut can also migrate to the lung and exert their effects there. In addition to metabolites, bacterial structural components termed microbial‐associated molecular patterns can impact innate and adaptive processes via pattern recognition receptors (PRRs). Both microbes within the gut and the lungs signal via PRRs and can induce regulatory or effector cell lymphocytes.
Overall, changes in microbiota exposures and changes in dietary habits that occurred simultaneously may interact in unanticipated ways resulting in additive detrimental effects on the host immune system. Several studies have linked early life dietary and microbiota changes with an increased risk of asthma later in life, and these will be further discussed in this review.
2. INTERACTIONS BETWEEN INFANT DIET, MICROBIOME, AND ASTHMA
Overall dietary intake may influence allergic and asthma outcomes through intake of multiple nutrients with direct effects on the immune system (e.g., fish oils) and nutrients that have indirect effects on the immune system (e.g., fibers) via their utilization and metabolism by gut microbes. 14 , 15 However, nutrients do not exist in isolation, and it may be important to study nutrition (especially immunonutrition) using a whole diet approach to better understand the role of dietary intake on asthma outcomes. Whole diet approaches used in infancy and childhood in association with asthma outcomes include diet patterns that assess intake of advanced glycation end products (AGEs), diet indices such as the Mediterranean diet index and diet diversity. 14
A USA‐based study reported an association between increased AGEs intake in children 5–9 years of age and an increased odds of wheezing, wheeze‐disrupted sleep, and wheezing requiring prescription medication. 16 Despite the fact that this study was not conducted in children with allergic outcomes, it was postulated that increased intake of AGEs impacts immune inflammatory responses, disrupts the gut microbiome of children, and is associated with an increase in autoimmune diseases such as type I diabetes. 17 A number of studies have shown an increased prevalence of wheeze/asthma in children consuming a Western diet. 18 , 19 , 20 In contrast to these studies, the Dutch Generation R cohort did not find an association between a healthy dietary pattern in early life and a lower risk of atopic diseases, including asthma, in childhood. 21 , 22 However, unfortunately, the majority of studies comparing dietary effects on allergic outcomes in infants and young children to date do not include measures of microbiota composition or metabolic activity.
Three systematic reviews examining the Mediterranean diet in children and asthma outcomes have shown an inverse relationship between this index and reported wheeze and asthma. 23 , 24 , 25 While the Mediterranean diet studies in infants and children focused on asthma outcomes did not include microbiome measures, it is already known that increased fibers, fruit, and vegetable consumption during infancy is associated with an increase in gut microbial diversity. 15 Comparing Egyptian teenagers on a Mediterranean‐style diet to American teenagers consuming a Western diet, Egyptian children clustered to the Prevotella enterotype and American children clustered to the Bacteroides enterotype. In addition, higher levels of short‐chain fatty acids (SCFAs) were observed in Egyptian children, which was associated with increased abundance of microbial polysaccharide degradation‐encoding genes, and polysaccharide‐degrading genera. 26 A study comparing the gut microbiota of Bangladeshi and American teenagers also showed lower levels of Bacteroides and higher levels of Prevotella, Butyrivibrio, and Oscillospira, in the Bangladeshi children, associated with their non‐Western diet that is low in meat and sugar with higher intake of rice, bread, and lentils. The American children consuming Western diets had a higher abundance of Bacteroides. 27
Diet diversity in infancy and asthma/wheeze outcomes in childhood were reported by the PASTURE study and the Finnish Type I Diabetes Prediction and Prevention Study Prospective Cohort Study. 28 , 29 Roduit et al. showed in the PASTURE study that increased diet diversity in the first year of life was associated with reduced asthma by 6 years. For every additional food introduced, a 26% reduction in asthma risk was reported. Nwaru et al. also reported that reduced diet diversity at 12 months of life was associated with an increased risk of the development of asthma at age 5, as well as an increased risk of any wheeze. In the PASTURE cohort, a diet rich in fruit, vegetables, fish, and yogurt was associated with the highest levels of butyrate at 12 months, which correlated with a lower risk of asthma by 6 years of age. 30 As a proxy for diet diversity, increased protein, carbohydrate and fiber intake, as well as increased intake of family foods during the weaning period, has also been associated with an increase in gut microbiome diversity in infants. 15
Of all the dietary ingredients studied to date, fibers may be one of the most important for microbiota and immune health. Certain fibers, also termed microbiota‐accessible carbohydrates, are an essential food source for the microbiota in that they provide resources for microbial growth. They are central to food webs in the gut microbiota established through cross‐feeding, and reduced fiber intake has been shown to be associated with the loss of ancestral microbes. 31 Overall, species diversity and richness have been shown to be reduced by about one‐third in North Americans than Malawians or Amerindians, which might be due in part to differences in dietary fiber consumption. 32 A high fat/low fiber diet and obesity have been associated with alterations in gut microbiota composition and metabolic activity. The degradation of dietary fibers requires specific CAZymes (carbohydrate active enzymes), which are encoded in the genomes of specific bacterial strains. 33 , 34 While the human genome encodes potentially up to 17 glycoside hydrolases, thousands of gut microbiota genes that encode glycoside hydrolases, polysaccharide lyases, glycosyltransferases, and carbohydrate esterases have been described, demonstrating the indispensable role of the gut microbiota in fiber metabolism. 35 , 36 Members of the Bacteroidetes phylum (in particular Prevotella copri) seem to possess a greater number of CAZymes than other phyla, suggesting an increased capability to ferment a wider range of substrates. 37 , 38 As specific CAZyme gene clusters target discrete structures within dietary fibers, consequently specific subsets of microbes are supported by different types of dietary fibers, which highlights the potential for using selected fiber structures to achieve targeted functional, metabolic, and perhaps immunological outcomes. 39 , 40 Future studies will hopefully better classify different dietary fiber types on the basis of their specific immune functional properties, such as promotion of the epithelial barrier, induction of T regulatory cells, prevention of TH2 polarization, and inhibition of mast cell degranulation. Overall, fiber diversity may be more important immunologically than any single individual fiber type.
In one cross‐sectional observational study, an association between low dietary fiber intake and increased odds of reported asthma was noted among respondents on the National Health and Nutrition Examination survey (NHANES) survey in the USA. 41 They noted increased odds of asthma with lower fiber intake (lowest vs. highest reported quartile, OR, 1.4; 95% CI 1.0–1.8; p = .027) with significant interactions between fiber and both sex and race/ethnicity, in particular among women and non‐Hispanic white adults. Lowest quartile fiber intake was associated with increased odds of reported wheeze (OR, 1.3; 95% CI, 1.0–1.6; p = .018) and cough (OR, 1.7; 95% CI, 1.2–2.3; p = 0.002).
Several guidelines and systematic reviews have previously examined the role of both fiber and prebiotic supplementation with respect to allergy outcomes. We recently published an updated review of the role of dietary fiber in influencing allergic diseases, including asthma. 42 In summary, fibers are essential components of a healthy diet with multiple health benefits, and fiber intake has decreased at the same time as allergy rates have increased. There are a wide variety of fiber types, and specific fibers may contribute to maintain a tolerogenic mucosal environment and may protect against allergic disorders. However, the optimal prevention or treatment strategies involving fibers in humans have yet to be defined. One mechanism by which fiber impacts the immune system is dependent on microbial fermentation and secretion of bioactive metabolites. Thus, fiber supplementation alone may not be sufficient and simultaneous replacement of missing microbes may be required for optimal benefits to be observed. Overall, there is some evidence that a higher intake of dietary fiber (soluble or insoluble) may have potential protective effects on respiratory symptoms. However, the majority of the studies reviewed were small observational studies that did not clearly label the type of fiber ingested, which limits their interpretation and extrapolation. Further prospective intervention studies are needed to better define the effects of fiber consumption on respiratory outcomes. Prebiotic supplementation to asthma patients seems to have a positive effect on exercise‐induced bronchoconstriction and accompanying inflammation markers; however, as this was only a single intervention study with this design, an overall recommendation cannot be made.
3. ROLE OF THE GUT MICROBIOTA
The largest reservoir of microbes within the body is contained in the gastrointestinal tract, which encodes a genetic repertoire that is at least two orders of magnitude greater than the number of genes in the human genome. 43 The gut microbiome performs several essential functions including digestion of dietary components, synthesis of vitamins, education and regulation of the immune system, competitive exclusion of pathogens, removal of toxins and carcinogens, and support of intestinal barrier function. 44 These activities do not occur in isolation but involve interconnected nodes of microbial and host metabolic pathways, resulting in the generation of metabolites with immunoregulatory properties. In addition, immune cells and metabolites from the gut can directly or indirectly impact other organs, such as the lungs, via the circulatory, lymphatic, endocrine, and nervous systems. The “common mucosal immune system” was originally proposed by McDermott and Bienenstock in 1979, which links the mucosal sites and balances the close relationship between the host and mucosal‐associated microbiota. 45 , 46 Both the gut and the lung share features such as inducible immune follicles (Gut‐Associated Lymphoid Tissue (GALT) and Bronchus‐Associated Lymphoid tissue (BALT)), which coordinate many tasks including the secretion of IgA at the mucosal epithelium and T‐cell responses to luminal microbes. 47 Indeed, the importance of the gut microbiome in maintaining systemic homeostasis has now been recognized in other fields such as in metabolic diseases. 48
While the pathogenesis of asthma is complex with many contributing factors from genetic to environmental exposures, the exploration of the role of the gut microbiota in asthma has yielded important insights. Several studies have noted that patterns of gut colonization in early childhood can be associated with altered risk of airway disease later in life. One recent study identified a pathway whereby birth by cesarean section results in reduced infant Bacteroides and microbial sphingolipid production, which associates with an increased risk of asthma by 3 years of age. 49 Three gut microbiota composition states have been described for neonates, each of which was associated with a different relative risk of atopy and asthma later in life. 50 The highest risk group had lower levels of Bifidobacterium, Akkermansia, and Faecalibacterium. Arrieta et al. observed lower abundance of key bacterial genera (Faecalibacterium, Rothia, Lachnospiria and Veillonella (FLVR)) and reduced levels of acetate (a SCFA) at 3 months of age in Canadian infants that became atopic and developed wheeze by 1 year of age, with a significantly elevated risk of asthma by 3 years of age. 51 Interestingly, a similar study in an Ecuadorian cohort also revealed a microbiome signature at 3 months of age and reduced acetate levels that associated with risk of atopic wheeze later in life. 52 However, other than Veillonella species, different taxa (Streptococcus, Bacteroides, Ruminococcus, and Bifidobacterium) were associated with disease risk in children from Ecuador. This finding suggests that compositional differences may be less important than functional and metabolic outputs such as SCFAs.
The gut microbiota has not just been associated with disease risk but has also been shown to be altered in asthma patients with severe disease. A significant reduction in the family Verrucomicrobiaceae was observed in the gut microbiota of severe asthmatics, than patients with mild/moderate asthma, which was primarily due to reduced levels of Akkermansia muciniphila. 33 In experimental models, A. muciniphila protected mice from respiratory inflammatory responses to acute and chronic house dust mite extract exposure, associated with higher numbers of lung Tregs and reduced accumulation of the IL‐5‐dependent Siglec Fhigh eosinophils within lung tissue. This mechanism was MyD88 independent but did require viable bacterial cells, suggesting that heat sensitive factors or metabolites secreted in vivo were required for the A. muciniphila protective effects. In contrast to A. muciniphila, increased levels histamine secreting microbes, in particular Morganella morganii, were observed in the gut microbiota of severe asthma patients. 53 M. morganii has been previously implicated as one of the causative microbes for histamine fish poisoning (HFP), or scombroid food poisoning, which is an illness associated with consumption of high levels of histamine in fish, following the metabolism of histidine in the fish by bacteria during inappropriate handling during storage or processing. 54 , 55 Murine models clearly show that histamine secreted by microbes within the gut can have immunological effects in the gut and the lungs; however, it is unknown whether increased levels of histamine secretion by gut microbes can contribute to disease severity in asthma patients. 56 , 57 , 58 Indeed, given that specific microbes within the gut can also degrade histamine, perhaps it is the balance between microbial histamine generators and microbial histamine degraders that most significantly impacts the immune system and asthma symptoms. 59
4. ROLE OF THE RESPIRATORY MICROBIOTA
The role of the respiratory microbiota in asthma has been the subject of intense focus in recent years. The lung microbiota is now recognized to play a role in the pathogenesis of asthma, may influence asthma phenotypes, and can potentiate asthma exacerbations. Previously, it was thought the lungs were sterile, but the lung has its own distinct microbiota, just like the gut, and it has been found to be significantly different in nonasthma controls compared with those with respiratory diseases such as asthma. 60 , 61
The development of the lung microbiota begins early in life. Microbial dysbiosis, particularly at this early age, has been shown in multiple studies to increase a child's chances of later developing asthma, potentially by promoting Th2 inflammatory cascades. 62 , 63 Bisgaard et al demonstrated that colonization with Streptococcus pneumonia, Moraxella catarrhalis, and/or Haemophilus influenza sampled from hypopharyngeal aspirates from 1‐month‐old babies born to asthmatic mothers was significantly associated with persistent wheeze, hospitalization for wheeze and asthma diagnosis at age 5 years. 64 By contrast, increased abundance of nasopharyngeal Lactobacillus species during acute respiratory infection with respiratory syncytial virus (RSV) in infancy was associated with reduced risk of wheezing at 2 years of age. 65 Microbiota composition in children is also correlated with changes in asthma control and asthma severity. One study suggested that 15 bacterial genera and seven fungal genera showed a significant difference in overall abundance between severe asthma and nonasthma bronchoalveolar lavage samples (BALs), while another study found that Moraxella was the most commonly associated genus in the nasal airways of children with severe persistent asthma (average age of 11 years old in both studies). 66 , 67 Conversely, early childhood exposure to a diverse range of microbes, allergens, and environmental exposures such as having pets, living on a farm, and drinking unpasteurised milk can be protective against asthma, perhaps via effects on immune education and immune regulatory networks. 68 , 69 , 70 , 71 , 72 However, these exposures need to occur within a critical time window for this to occur successfully. There is also believed to be an interplay between the lung and the gut microbiome that can reduce the risk of allergic asthma later in life. 73
The composition of the lung microbiome in adults has also been found to be different when healthy volunteers and asthmatic patients have been compared. 61 Not only bacteria but also fungi and viruses can impact immune responses in the lungs. 74 , 75 These differences may contribute to airway hyperreactivity, mucous production, and the increased release of proinflammatory cytokines, which all contribute to asthma development. An increased level of pathogenic proteobacteria particularly Haemophilus influenza but also Streptococcus pneumonia and Moraxella catarrhalis and reduced levels of Bacteroidetes were observed in asthmatics when compared to samples from control volunteers. 61 Within the airways, the phylum Proteobacteria is often associated with worse asthma control, whereas Actinobacteria can correlate with improvement, or no change in asthma control. Mycoplasma pneumoniae, Chlamydophila pneumoniae, Neisseria Haemophilus, Campylobacter, and Leptotrichia species are also more often found in the airways of severe asthmatics or in corticosteroid‐resistant patients. 76 , 77 However, the respiratory microbiome of those with mild‐to‐moderate asthma is usually more similar to that of a patient with severe asthma than control nonasthma patients. 78
The mechanisms responsible for changes in the airway microbiota are not well‐understood, and in addition to medications, it is possible that the type of inflammatory response (i.e., eosinophil versus neutrophil), changes in host secretions (e.g., lipids), and cellular metabolism might influence microbial colonization and growth within the airways. Interestingly, neutrophilic exacerbations of asthma and chronic obstructive pulmonary disease (COPD) correlated with the presence of Proteobacteria in the sputum, whereas increased eosinophils in asthma patients, regardless of their BMI, was associated with an increased relative abundance of the genera Rothia, Dorea, Lautropia, and Haemophilus within BALs. 33 One study compared the bronchial brushings of nonasthma patients to those with asthma and atopy and found that in those with type‐2 inflammation, there was a lower bacterial burden when compared to other phenotypes. 79 Another study found that those with neutrophilic asthma were more likely to be colonized by Proteobacteria (Haemophilus and Moraxella), have an overall reduced microbial diversity and that this was usually related to a more severe asthma phenotype. 80 There is a previously shown association between airway neutrophilia, severe airway obstruction, and interleukin 18 (IL‐18), which may be related to differences in the lung microbiota. 81 Importantly, obesity has been associated with changes in the nasal and lung microbiota, even in obese individuals that do not have an asthma diagnosis. 33 It is unknown whether this difference in respiratory microbiota composition represents a pre‐asthma state, or whether obesity‐driven microbiota changes are independent of subsequent asthma risk.
Key to the role of the microbiota in the development of asthma is a cycle of inflammation, injury, repair, and remodeling that occurs in the airways. On the one hand, fungi, viruses, and bacteria within the airways can activate innate receptors such as Toll‐like receptor (TLR), RIG1‐like helicase, and NOD‐like receptor, which in turn activate the nuclear factor κB (NFκB) family of transcription factors and as a result induce hundreds of proinflammatory and host response genes. 82 , 83 The subsequent epithelial damage results in remodeling with airway thickening and a reduction in lung function, alterations in epithelial permeability and epithelial barrier with increased exposure to inflammatory molecules, allergens and an increased risk of infection that in turn can trigger an exacerbation. 21 , 84 On the other hand, the microbial composition and high endotoxin load of dust from the homes of Amish children was associated with enhanced induction of innate immune pathways and protection from asthma development, compared with genetically similar Hutterite children. 85 Several studies now support the concept that house dust samples from a traditional farming environment contain a diversity of microbes and microbial ligands that associate with increased nasal microbiome diversity of the same children who have lower risk of developing asthma. 68 , 86 , 87 It is currently unknown whether the protective effect of the dust‐associated microbes might be due to inhalation of multiple bacteria species and further colonization of the airways, or whether inhaled bacterial components that stimulate innate immune responses or bacterial‐derived metabolites may also play a role.
5. IMMUNE MECHANISMS MEDIATING MICROBIAL PROTECTIVE EFFECTS WITHIN THE LUNG
Murine models have been extensively used to decipher the role of the microbiota in the pathogenesis of airway inflammation. Neonatal mice were more susceptible to develop house dust mite‐induced allergic airway inflammation and airway hyperactivity than mature mice, which was associated with a shift from Gammaproteobacteria and Firmicutes toward a Bacteroidetes dominated microbiota and the development of PDL‐1‐dependent Helios‐Treg cells. 88 Germ‐free mice display significantly more pronounced type 2 inflammation when compared with conventionally colonized mice. Recolonization, especially early in life, can reverse many of these immunological defects. 89 Similarly, antibiotic treatment of neonatal mice leads to impaired maturation of Tregs, enhanced TH2 responses, and promotes proinflammatory colonic iNKT cells. 90 , 91 , 92 , 93 , 94 Conversely, specific bacterial strains, their components, or metabolites can successfully induce a variety of anti‐inflammatory responses in the gut and in the lung. For example, intranasal administration of an exopolysaccharide (EPS) from Bifidobacterium longum subsp. longum 35,624 was shown to protect against allergic airway disease in murine models, which was dependent on TLR2‐induced IL‐10 secretion. 95 , 96 Similarly, other microbial structural components such as lipopolysaccharides (LPS), polysaccharide A (PSA) and a Bifidobacterium‐derived cell wall extract can alter the allergic immune response and antiviral immune response in murine models. 97 , 98 , 99 A recent study by Lai et al on a murine COPD model showed that a commensal gut bacterial‐associated LPS, derived from Parabacteroides goldsteinii, was anti‐inflammatory. 100 The commensal‐derived LPS signaled via Toll‐like receptor 4 to reduce inflammatory signals, thereby enhancing epithelial barrier function and allowing for homeostatic metabolic processes to be restored.
In addition to bacterial structural components, multiple microbial metabolites influence respiratory inflammation. Products of amino acid metabolism, such as spermine, spermidine and tryptophan‐derived indoles, have been shown to reduce allergic inflammation in murine models. 101 , 102 Secondary bile acids (generated by microbial metabolism of primary bile acids) such as ursodeoxycholic acid have been shown to reduce eosinophilic airway inflammation via FXR signaling in dendritic cells. 103 Similarly, chenodeoxycholic acid attenuates allergic airway inflammation by inhibiting TH2 cytokine secretion. 104 One of the best‐studied immunoregulatory bacterial metabolites is the SCFAs butyrate, propionate, and acetate. 42 Fecal SCFA levels are low during the first 6 months of life, with acetate, propionate, and butyrate reaching stable levels in feces after 6, 8 or 10 months of age, respectively. 105 Short‐chain fatty acids exert effects on the host immune system via binding to G protein‐coupled receptors (GPCRs) such as GPR41, GPR43, and GPR109A, via epigenetic modifications that inhibit histone deacetylase (HDAC) activity, and most recently butyrate has been described as an aryl hydrocarbon receptor (AhR) ligand. 106 , 107 Short‐chain fatty acids are potent immunomodulators that promote IL‐10 secretion by dendritic cells and lymphocytes, influence Treg numbers and effectiveness, influence bone marrow hematopoiesis, reduce effector T‐cell activity, improve epithelial barrier, support IgA secretion by B lymphocytes, inhibit mast cell degranulation and modulate ILC activation. 108 , 109 , 110 , 111 , 112 , 113 , 114 , 115 Fiber consumption or SCFA administration in experimental models protects against colitis, inflammatory arthritis, respiratory syncytial virus infection, allergic airway inflammation, and food allergy. 30 , 116 , 117 Epigenetic mechanisms seem particularly important for the induction of T‐regulatory cells in the gut as butyrate enhances histone acetylation of the Foxp3 promoter, thereby driving Treg development. 118 , 119 Contrary to the beneficial effects of certain SCFAs, other bacterial‐derived metabolites such as 12,13‐ diHOME, an oxylipin, decrease the number of T regs via altering dendritic cell activation and reducing IL‐10 secretion. 120
6. CONCLUSIONS AND FUTURE PERSPECTIVES
Microbial‐derived factors are integral components of the molecular circuitry that regulate immune and metabolic functions required for host fitness and survival. Recent advances in culture‐based methods and sequencing technologies have revealed previously unappreciated complex communities of bacteria, fungi, and viruses that inhabit all body surfaces, including the respiratory tract. Many studies have now shown differences in the composition of bacterial communities in children that go on to develop asthma, or in those who already have asthma. However, the functional consequences of changes in the abundances of specific microbial taxa and their relevance to asthma disease mechanisms remain largely unknown. Subtle structural, species‐ or strain‐specific differences, even for genetically closely related microbes, may lead to distinct immunological outcomes. This lack of fundamental knowledge on microbiome–host functional interactions severely limits our ability to understand how shifts in microbiome community structure and function may trigger the onset and progression of diseases such as asthma over a lifetime.
Notwithstanding these limitations, microbiota analysis and interventions may be of use in several areas:
Endotyping—currently, a range of host factors are measured to define asthma disease endotypes. However, a more detailed and accurate endotyping of patients with asthma may be assisted by including an analysis of the composition and metabolic activity of an individual's respiratory and gut microbiota.
Risk assessments—there is significant potential for analysis of the microbiota to assist in the early prediction of later life asthma risk, and to help to identify those children who may develop the most severe forms of asthma. However, these studies must carefully include demographic, clinical, exposure and lifestyle factors as possible confounders in their analysis to correctly interpret their findings.
Response to therapy—microbiota profiling should be included in future clinical studies examining novel asthma medications. Responsiveness to glucocorticosteroids may be microbiota‐dependent, while the optimal choice of biologic may be heavily influenced by microbial factors. In other fields, such as cancer immunotherapy using checkpoint inhibitors, the importance of the microbiota in therapy success is well‐established. 121
Microbial therapies—specific microbes, and their metabolites, are being examined for their preventive and therapeutic effects, but given the explosion in knowledge regarding disease endotypes, it is possible that specific microbes will need to be carefully selected to mechanistically fit with specific disease endotypes and it is likely that one intervention will not work equally well for everyone with different types of asthma. In addition, the efficacy of existing therapeutics may be significantly improved by microbiota‐targeted interventions.
Dietary interactions—the metabolism of certain dietary factors (e.g., fibers) by microbes is required for the full immunological benefits of that dietary component to be realized. Studies examining nutritional interventions should include an analysis of the direct and indirect effects of the microbiome on their study endpoints. While there are already many unique challenges and difficulties in performing nutrition studies to provide evidence‐based recommendations, the inclusion of microbiota assessments may help identify those who require the intervention and those most likely to respond. In addition, knowledge of how specific dietary interventions impact the microbiota to generate immunoregulatory metabolites can be leveraged to develop more effective diets and molecules that improve human immune health.
In conclusion, it remains unclear whether and, if so, to what extent patterns of microbial dysbiosis actually drives rather than merely reflects associated patterns of immune reactivity within the airways. It is likely that differences in functional or metabolic outputs are probably more important immunologically, than taxonomic diversity alone. Of particular importance to understand is the potential relationship between timing and dietary‐microbiome effects on the immune system, especially in early life where there is a critical window of opportunity to influence the development of immune regulatory networks. Future research on diet–microbe–host interactions should be strongly encouraged as these discoveries will provide fundamental knowledge on the molecular communication networks that underpin life as a multicellular metacommunity and will progress our appreciation for the principle of biological diversity as a driver of physiological resilience and immune tolerance.
7.
PEER REVIEW
The peer review history for this article is available at https://publons.com/publon/10.1111/pai.13892.
ACKNOWLEDGMENT
Open access funding provided by IReL.
Lunjani N, Walsh L, Venter C, et al. Environmental influences on childhood asthma—The effect of diet and microbiome on asthma. Pediatr Allergy Immunol. 2022;33:e13892. doi: 10.1111/pai.13892
Editor: Ömer KALAYCI
REFERENCES
- 1. Alkotob SS, Cannedy C, Harter K, et al. Advances and novel developments in environmental influences on the development of atopic diseases. Allergy. 2020;75(12):3077‐3086. [DOI] [PubMed] [Google Scholar]
- 2. Lunjani N, Satitsuksanoa P, Lukasik Z, Sokolowska M, Eiwegger T, O'Mahony L. Recent developments and highlights in mechanisms of allergic diseases: microbiome. Allergy. 2018;73(12):2314‐2327. [DOI] [PubMed] [Google Scholar]
- 3. Armet AM, Deehan EC, O'Sullivan AF, et al. Rethinking healthy eating in light of the gut microbiome. Cell Host Microbe. 2022;30(6):764‐785. [DOI] [PubMed] [Google Scholar]
- 4. Akdis CA. Does the epithelial barrier hypothesis explain the increase in allergy, autoimmunity and other chronic conditions? Nat Rev Immunol. 2021;21(11):739‐751. [DOI] [PubMed] [Google Scholar]
- 5. Walter J, O'Mahony L. The importance of social networks‐an ecological and evolutionary framework to explain the role of microbes in the aetiology of allergy and asthma. Allergy. 2019;74(11):2248‐2251. [DOI] [PubMed] [Google Scholar]
- 6. Martínez Leo EE, Segura Campos MR. Effect of ultra‐processed diet on gut microbiota and thus its role in neurodegenerative diseases. Nutrition. 2020;71:110609. [DOI] [PubMed] [Google Scholar]
- 7. Sonnenburg ED, Smits SA, Tikhonov M, Higginbottom SK, Wingreen NS, Sonnenburg JL. Diet‐induced extinctions in the gut microbiota compound over generations. Nature. 2016;529(7585):212‐215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. McDonald D, Hyde E, Debelius JW, et al. American gut: an open platform for citizen science microbiome research. mSystems. 2018;3(3):e00031–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Forde B, Yao L, Shaha R, Murphy S, Lunjani N, O'Mahony L. Immunomodulation by foods and microbes: unravelling the molecular tango. Allergy. 2022. doi: 10.1111/all.15455. Online ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Albrich WC, Ghosh TS, Ahearn‐Ford S, et al. A high‐risk gut microbiota configuration associates with fatal hyperinflammatory immune and metabolic responses to SARS‐CoV‐2. Gut Microbes. 2022;14(1):2073131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Lunjani N, Albrich WC, Suh N, et al. Higher levels of bacterial DNA in serum associate with severe and fatal COVID‐19. Allergy. 2022;77:1312‐1314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Yeoh YK, Zuo T, Lui GC, et al. Gut microbiota composition reflects disease severity and dysfunctional immune responses in patients with COVID‐19. Gut. 2021;70:698‐706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Zuo T, Zhang F, Lui GCY, et al. Alterations in gut microbiota of patients with COVID‐19 during time of hospitalization. Gastroenterology. 2020;159:944‐955. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Venter C, Greenhawt M, Meyer RW, et al. EAACI position paper on diet diversity in pregnancy, infancy and childhood: novel concepts and implications for studies in allergy and asthma. Allergy. 2020;75(3):497‐523. doi: 10.1111/all.14051 [DOI] [PubMed] [Google Scholar]
- 15. Laursen MF, Andersen LB, Michaelsen KF, et al. Infant gut microbiota development is driven by transition to family foods independent of maternal obesity. mSphere. 2016;1(1):e00069‐15. doi: 10.1128/mSphere.00069-15 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Wang JG, Liu B, Kroll F, et al. Increased advanced glycation end product and meat consumption is associated with childhood wheeze: analysis of the national health and nutrition examination survey. Thorax. 2021;76(3):292‐294. doi: 10.1136/thoraxjnl-2020-216109 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Virtanen SM. Dietary factors in the development of type 1 diabetes. Pediatr Diabetes. 2016;17(Suppl 22):49‐55. doi: 10.1111/pedi.12341 [DOI] [PubMed] [Google Scholar]
- 18. de Cassia Ribeiro Silva R, Assis AM, Cruz AA, et al. Dietary patterns and wheezing in the midst of nutritional transition: a study in Brazil. Pediatr Allergy Immunol Pulmonol. 2013;26(1):18‐24. doi: 10.1089/ped.2012.0182 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Lee SC, Yang YH, Chuang SY, et al. Risk of asthma associated with energy‐dense but nutrient‐poor dietary pattern in Taiwanese children. Asia Pac J Clin Nutr. 2012;21(1):73‐81. [PubMed] [Google Scholar]
- 20. Ellwood P, Asher MI, Garcia‐Marcos L, et al. Do fast foods cause asthma, rhinoconjunctivitis and eczema? global findings from the international study of asthma and allergies in childhood (ISAAC) phase three. Thorax. 2013;68(4):351‐360. doi: 10.1136/thoraxjnl-2012-202285 [DOI] [PubMed] [Google Scholar]
- 21. Tromp II, Kiefte‐de Jong JC, de Vries JH, et al. Dietary patterns and respiratory symptoms in pre‐school children: the generation R study. Eur Respir J. 2012;40(3):681‐689. doi: 10.1183/09031936.00119111 [DOI] [PubMed] [Google Scholar]
- 22. Nguyen AN, Elbert NJ, Pasmans S, et al. Diet quality throughout early life in relation to allergic sensitization and atopic diseases in childhood. Nutrients. 2017;9(8):5. doi: 10.3390/nu9080841 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Nurmatov U, Devereux G, Sheikh A. Nutrients and foods for the primary prevention of asthma and allergy: systematic review and meta‐analysis. J Allergy Clin Immunol. 2011;127(3):724‐733.e1–30. doi: 10.1016/j.jaci.2010.11.001 [DOI] [PubMed] [Google Scholar]
- 24. Papamichael MM, Itsiopoulos C, Susanto NH, Erbas B. Does adherence to the mediterranean dietary pattern reduce asthma symptoms in children? a systematic review of observational studies. Public Health Nutr. 2017;20(15):2722‐2734. doi: 10.1017/S1368980017001823 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Lv N, Xiao L, Ma J. Dietary pattern and asthma: a systematic review and meta‐analysis. J Asthma Allergy. 2014;7:105‐121. doi: 10.2147/JAA.S49960 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Shankar V, Gouda M, Moncivaiz J, et al. Differences in gut metabolites and microbial composition and functions between Egyptian and U.S. children are consistent with their diets. mSystems. 2017;2(1):e00169‐16. doi: 10.1128/mSystems.00169-16 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Lin A, Bik EM, Costello EK, et al. Distinct distal gut microbiome diversity and composition in healthy children from Bangladesh and the United States. PLoS One. 2013;8(1):e53838. doi: 10.1371/journal.pone.0053838 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Roduit C, Frei R, Depner M, et al. Increased food diversity in the first year of life is inversely associated with allergic diseases. J Allergy Clin Immunol. 2014;133(4):1056‐1064. doi: 10.1016/j.jaci.2013.12.1044 [DOI] [PubMed] [Google Scholar]
- 29. Nwaru BI, Takkinen HM, Kaila M, et al. Food diversity in infancy and the risk of childhood asthma and allergies. J Allergy Clin Immunol. 2014;133(4):1084‐1091. doi: 10.1016/j.jaci.2013.12.1069 [DOI] [PubMed] [Google Scholar]
- 30. Roduit C, Frei R, Ferstl R, et al. High levels of butyrate and propionate in early life are associated with protection against atopy. Allergy. 2019;74(4):799‐809. [DOI] [PubMed] [Google Scholar]
- 31. Sonnenburg ED, Smits SA, Tikhonov M, Higginbottom SK, Wingreen NS, Sonnenburg JL. Diet‐induced extinctions in the gut microbiota compound over generations. Nature. 2016;529(7585):212‐215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Yatsunenko T, Rey FE, Manary MJ, et al. Human gut microbiome viewed across age and geography. Nature. 2012;486(7402):222‐227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Michalovich D, Rodriguez‐Perez N, Smolinska S, et al. Obesity and disease severity magnify disturbed microbiome‐immune interactions in asthma patients. Nat Commun. 2019;10(1):5711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. O'Keefe SJ, Li JV, Lahti L. Fat, fiber and cancer risk in African Americans and rural Africans. Nat Commun. 2015;28(6):6342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. el Kaoutari A, Armougom F, Gordon JI, Raoult D, Henrissat B. The abundance and variety of carbohydrate‐active enzymes in the human gut microbiota. Nat Rev Microbiol. 2013;11(7):497‐504. [DOI] [PubMed] [Google Scholar]
- 36. Rowland I, Gibson G, Heinken A, et al. Gut microbiota functions: metabolism of nutrients and other food components. Eur J Nutr. 2018;57(1):1‐24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Cantarel BL, Lombard V, Henrissat B. Complex carbohydrate utilization by the healthy human microbiome. PLoS One. 2012;7:e28742. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. de Filippis F, Pasolli E, Tett A, et al. Distinct genetic and functional traits of human intestinal prevotella copri strains are associated with different habitual diets. Cell Host Microbe. 2019;25:444‐453. [DOI] [PubMed] [Google Scholar]
- 39. Deehan EC, Yang C, Perez‐Muñoz ME, et al. Precision microbiome modulation with discrete dietary fiber structures directs short‐chain fatty acid production. Cell Host Microbe. 2020;27(3):389‐404. [DOI] [PubMed] [Google Scholar]
- 40. Hamaker BR, Tuncil YE. A perspective on the complexity of dietary fiber structures and their potential effect on the gut microbiota. J Mol Biol. 2014;426:3838‐3850. [DOI] [PubMed] [Google Scholar]
- 41. Saeed MA, Gribben KC, Alam M, Lyden ER, Hanson CK, LeVan TD. Association of dietary fiber on asthma, respiratory symptoms, and inflammation in the adult national health and nutrition examination survey population. Ann Am Thorac Soc. 2020;17(9):1062‐1068. [DOI] [PubMed] [Google Scholar]
- 42. Venter C, Meyer RW, Greenhawt M, et al. Role of dietary fiber in promoting immune health‐an EAACI position paper. Allergy. 2022;77:3185‐3198. doi: 10.1111/all.15430 [DOI] [PubMed] [Google Scholar]
- 43. Miyauchi E, Shimokawa C, Steimle A, Desai MS, Ohno H. The impact of the gut microbiome on extra‐intestinal autoimmune diseases. Nat Rev Immunol. 2022;1‐15. doi: 10.1038/s41577-022-00727-y. Online ahead of print. [DOI] [PubMed] [Google Scholar]
- 44. Wilmes P, Martin‐Gallausiaux C, Ostaszewski M, et al. The gut microbiome molecular complex in human health and disease. Cell Host Microbe. 2022;30(9):1201‐1206. [DOI] [PubMed] [Google Scholar]
- 45. McDermott MR, Bienenstock J. Evidence for a common mucosal immunologic system. I. Migration of B immunoblasts into intestinal, respiratory, and genital tissues. J Immunol. 1979;122:1892‐1898. [PubMed] [Google Scholar]
- 46. Gill N, Wlodarska M, Finlay BB. The future of mucosal immunology: studying an integrated system‐wide organ. Nat Immunol. 2010;11:558‐560. [DOI] [PubMed] [Google Scholar]
- 47. Barcik W, Boutin RCT, Sokolowska M, Finlay BB. The role of lung and gut microbiota in the pathology of asthma. Immunity. 2020;52:241‐255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Fan Y, Pedersen O. Gut microbiota in human metabolic health and disease. Nat Rev Microbiol. 2021;19:55‐71. [DOI] [PubMed] [Google Scholar]
- 49. Lee‐Sarwar KA, Chen Y‐C, Chen YY, et al. The maternal prenatal and offspring early‐life gut microbiome of childhood asthma phenotypes. Allergy. 2022. doi: 10.1111/all.15516. Online ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Fujimura KE, Sitarik AR, Havstad S, et al. Neonatal gut microbiota associates with childhood multisensitized atopy and T cell differentiation. Nat Med. 2016;22:1187‐1191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Arrieta M‐C, Stiemsma LT, Dimitriu PA, et al. Early infancy microbial and metabolic alterations affect risk of childhood asthma. Sci Transl Med. 2015;7:307ra152. [DOI] [PubMed] [Google Scholar]
- 52. Arrieta M‐C, Arévalo A, Stiemsma L, et al. Associations between infant fungal and bacterial dysbiosis and childhood atopic wheeze in a nonindustrialized setting. J Allergy Clin Immunol. 2018;142:424‐434.e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Barcik W, Pugin B, Westermann P, et al. Histamine‐secreting microbes are increased in the gut of adult asthma patients. J Allergy Clin Immunol. 2016;138:1491‐1494. [DOI] [PubMed] [Google Scholar]
- 54. Colombo FM, Cattaneo P, Confalonieri E, Bernardi C. Histamine food poisonings: a systematic review and meta‐analysis. Crit Rev Food Sci Nutr. 2016;28:1‐21. [DOI] [PubMed] [Google Scholar]
- 55. Feng C, Teuber S, Gershwin ME. Histamine (Scombroid) fish poisoning: a comprehensive review. Clin Rev Allergy Immunol. 2016;50:64‐69. [DOI] [PubMed] [Google Scholar]
- 56. Frei R, Ferstl R, Konieczna P, et al. Histamine receptor 2 modifies dendritic cell responses to microbial ligands. J Allergy Clin Immunol. 2013;132:194‐204. [DOI] [PubMed] [Google Scholar]
- 57. Ferstl R, Frei R, Schiavi E, et al. Histamine receptor 2 is a key influence in immune responses to intestinal histamine‐secreting microbes. J Allergy Clin Immunol. 2014;134:744‐746. [DOI] [PubMed] [Google Scholar]
- 58. Barcik W, Pugin B, Brescó MS, et al. Bacterial secretion of histamine within the gut influences immune responses within the lung. Allergy. 2019;74(5):899‐909. [DOI] [PubMed] [Google Scholar]
- 59. Pugin B, Barcik W, Westermann P, et al. A wide diversity of bacteria from the human gut produces and degrades biogenic amines. Microb Ecol Health Dis. 2017;28:1353881. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Trujillo J, Lunjani N, Ryan D, O'Mahony L. Microbiome‐immune interactions and relationship to asthma severity. J Allergy Clin Immunol. 2022;149(2):533‐534. [DOI] [PubMed] [Google Scholar]
- 61. Hilty M, Burke C, Pedro H, et al. Disordered microbial communities in asthmatic airways. PLoS One. 2010;5(1):e8578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Ngoc PL, Gold DR, Tzianabos AO, Weiss ST, Celedón JC. Cytokines, allergy, and asthma. Curr Opin Allergy Clin Immunol. 2005;5:161‐166. [DOI] [PubMed] [Google Scholar]
- 63. Russell SL, Gold MJ, Willing BP, Thorson L, McNagny KM, Finlay BB. Perinatal antibiotic treatment affects murine microbiota, immune responses and allergic asthma. Gut Microbes. 2013;4:158‐164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Bisgaard H, Hermansen MN, Buchvald F, et al. Childhood asthma after bacterial colonization of the airway in neonates. N Engl J Med. 2007;357(15):1487‐1495. [DOI] [PubMed] [Google Scholar]
- 65. Rosas‐Salazar C, Shilts MH, Tovchigrechko A, et al. Nasopharyngeal lactobacillus is associated with a reduced risk of childhood wheezing illnesses following acute respiratory syncytial virus infection in infancy. J Allergy Clin Immunol. 2018;142:1447‐1456.e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Goldman DL, Chen Z, Shankar V, Tyberg M, Vicencio A, Burk R. Lower airway microbiota and mycobiota in children with severe asthma. J Allergy Clin Immunol. 2018;141:808‐811. [DOI] [PubMed] [Google Scholar]
- 67. Chun Y, Do A, Grishina G, et al. Integrative study of the upper and lower airway microbiome and transcriptome in asthma. JCI Insight. 2020;5:e133707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Ege MJ, Mayer M, Normand AC, et al. Exposure to environmental microorganisms and childhood asthma. N Engl J Med. 2011;364:701‐709. [DOI] [PubMed] [Google Scholar]
- 69. Heederik D, von Mutius E. Does diversity of environmental microbial exposure matter for the occurrence of allergy and asthma? J Allergy Clin Immunol. 2012;130:44‐50. [DOI] [PubMed] [Google Scholar]
- 70. Riedler J, Braun‐Fahrlander C, Eder W, et al. Exposure to farming in early life and development of asthma and allergy: a cross‐sectional survey. Lancet. 2001;358:1129‐1133. [DOI] [PubMed] [Google Scholar]
- 71. Waser M, Michels KB, Bieli C, et al. Inverse association of farm milk consumption with asthma and allergy in rural and suburban populations across Europe. Clin Exp Allergy. 2007;37:661‐670. [DOI] [PubMed] [Google Scholar]
- 72. Ownby DR, Johnson CC, Peterson EL. Exposure to dogs and cats in the first year of life and risk of allergic sensitization at 6 to 7 years of age. JAMA. 2002;288:963‐972. [DOI] [PubMed] [Google Scholar]
- 73. Marsland BJ, Gollwitzer ES. Host‐microorganism interactions in lung diseases. Nat Rev Immunol. 2014;14(12):827‐835. [DOI] [PubMed] [Google Scholar]
- 74. Sullivan A, Hunt EB, Ward C, et al. The presence of aspergillus fumigatus in asthmatic airways is not clearly related to clinical disease severity. Allergy. 2020;75(5):1146‐1154. [DOI] [PubMed] [Google Scholar]
- 75. Freer G, Maggi F, Pifferi M, di Cicco ME, Peroni DG, Pistello M. The Virome and its major component, Anellovirus, a convoluted system molding human immune defenses and possibly affecting the development of asthma and respiratory diseases in childhood. Front Microbiol. 2008;9:686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Abdel‐Aziz MI, Brinkman P, Vijverberg SJH, et al. Sputum microbiome profiles identify severe asthma phenotypes of relative stability at 12 to 18 months. J Allergy Clin Immunol. 2021;147(1):123‐134. [DOI] [PubMed] [Google Scholar]
- 77. Sokolowska M, Frei R, Lunjani N, Akdis CA, O'Mahony L. Microbiome and asthma. Dermatol Res Pract. 2018;4:1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Marri PR, Stern DA, Wright AL, Billheimer D, Martinez FD. Asthma‐associated differences in microbial composition of induced sputum. J Allergy Clin Immunol. 2013;131(2):346‐352.e1–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Durack J, Lynch SV, Nariya S, et al. Features of the bronchial bacterial microbiome associated with atopy, asthma and responsiveness to inhaled corticosteroid treatment. J Allergy Clin Immunol. 2017;140(1):63‐75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Taylor SL, Leong LEX, Choo JM, et al. Inflammatory phenotypes in patients with severe asthma are associated with distinct airway microbiology. J Allergy Clin Immunol. 2018;141(1):94‐103.e15. [DOI] [PubMed] [Google Scholar]
- 81. Wood LG, Simpson JL, Hansbro PM, Gibson PG. Potentially pathogenic bacteria cultured from the sputum of stable asthmatics are associated with increased 8‐isoprostane and airway neutrophilia. Free Radic Res. 2009;44:146‐154. [DOI] [PubMed] [Google Scholar]
- 82. Wark PA, Johnston SL, Moric I, Simpson JL, Hensley MJ, Gibson PG. Neutrophil degranulation and cell lysis is associated with clinical severity in virus‐induced asthma. Eur Respir J. 2002;19:68‐75. [DOI] [PubMed] [Google Scholar]
- 83. O'Neill LA, Bowie AG. The family of five: TIR domain‐containing adaptors in toll‐like receptor signalling. Nat Rev Immunol. 2007;7:353‐364. [DOI] [PubMed] [Google Scholar]
- 84. Murray CS, Simpson A, Custovic A. Allergens, viruses, and asthma exacerbations. Proc Am Thorac Soc. 2004;1:99‐104. [DOI] [PubMed] [Google Scholar]
- 85. Stein MM, Hrusch CL, Gozdz J, et al. Innate immunity and asthma risk in Amish and Hutterite farm children. N Engl J Med. 2016;375:411‐421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Birzele LT, Depner M, Ege MJ, et al. Environmental and mucosal microbiota and their role in childhood asthma. Allergy. 2017;72:109‐119. [DOI] [PubMed] [Google Scholar]
- 87. Depner M, Ege MJ, Cox MJ, et al. Bacterial microbiota of the upper respiratory tract and childhood asthma. J Allergy Clin Immunol. 2017;139:826‐834. [DOI] [PubMed] [Google Scholar]
- 88. Gollwitzer ES, Saglani S, Trompette A, et al. Lung microbiota promotes tolerance to allergens in neonates via PD‐L1. Nat Med. 2014;20:642‐647. [DOI] [PubMed] [Google Scholar]
- 89. Herbst T, Sichelstiel A, Schar C, et al. Dysregulation of allergic airway inflammation in the absence of microbial colonization. Am J Respir Crit Care Med. 2011;184:198‐205. [DOI] [PubMed] [Google Scholar]
- 90. Wypych TP, Marsland BJ. Antibiotics as instigators of microbial Dysbiosis: implications for asthma and allergy. Trends Immunol. 2018;39:697‐711. [DOI] [PubMed] [Google Scholar]
- 91. Fujimura KE, Lynch SV. Microbiota in allergy and asthma and the emerging relationship with the gut microbiome. Cell Host Microbe. 2015;17:592‐602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Sudo N, Yu XN, Aiba Y, et al. An oral introduction of intestinal bacteria prevents the development of a long‐term Th2‐skewed immunological memory induced by neonatal antibiotic treatment in mice. Clin Exp Allergy. 2002;32:1112‐1116. [DOI] [PubMed] [Google Scholar]
- 93. Burrello C, Garavaglia F, Cribiu FM, et al. Short‐term oral antibiotics treatment promotes inflammatory activation of colonic invariant natural killer T and conventional CD4(+) T cells. Front Med (Lausanne). 2018;5:21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Hill DA, Siracusa MC, Abt MC, et al. Commensal bacteria‐derived signals regulate basophil hematopoiesis and allergic inflammation. Nat Med. 2012;18:538‐546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Schiavi E, Gleinser M, Molloy E, et al. The surface‐associated exopolysaccharide of Bifidobacterium longum 35624 plays an essential role in dampening host Proinflammatory responses and repressing local TH17 responses. Appl Environ Microbiol. 2016;82:7185‐7196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Schiavi E, Plattner S, Rodriguez‐Perez N, et al. Exopolysaccharide from Bifidobacterium longum subsp. longum 35624 modulates murine allergic airway responses. Benef Microbes. 2018;9(5):1‐14. [DOI] [PubMed] [Google Scholar]
- 97. Budden KF, Gellatly SL, Wood DLA, et al. Emerging pathogenic links between microbiota and the gut‐lung axis. Nat Rev Microbiol. 2017;15:55‐63. [DOI] [PubMed] [Google Scholar]
- 98. Lin T‐L, Shu C‐C, Chen Y‐M, et al. Like cures like: pharmacological activity of anti‐inflammatory lipopolysaccharides from gut microbiome. Front Pharmacol. 2020;11:554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Groeger D, Schiavi E, Grant R, et al. Intranasal Bifidobacterium longum protects against viral‐induced lung inflammation and injury in a murine model of lethal influenza infection. EBioMedicine. 2020;60:102981. doi: 10.1016/j.ebiom.2020.102981 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Lai H‐C, Lin T‐L, Chen T‐W, et al. Gut microbiota modulates COPD pathogenesis: role of anti‐inflammatory parabacteroides goldsteinii lipopolysaccharide. Gut. 2022;71:309‐321. [DOI] [PubMed] [Google Scholar]
- 101. Wawrzyniak M, Groeger D, Frei R, et al. Spermidine and spermine exert protective effects within the lung. Pharmacol Res Perspect. 2021;9(4):e00837. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Zelante T, Puccetti M, Giovagnoli S, Romani L. Regulation of host physiology and immunity by microbial indole‐3‐aldehyde. Curr Opin Immunol. 2021;70:27‐32. [DOI] [PubMed] [Google Scholar]
- 103. Willart MAM, van Nimwegen M, Grefhorst A, et al. Ursodeoxycholic acid suppresses eosinophilic airway inflammation by inhibiting the function of dendritic cells through the nuclear farnesoid X receptor. Allergy. 2012;67:1501‐1510. [DOI] [PubMed] [Google Scholar]
- 104. Shaik FB, Panati K, Narasimha VR, Narala VR. Chenodeoxycholic acid attenuates ovalbumin‐induced airway inflammation in murine model of asthma by inhibiting the T(H)2 cytokines. Biochem Biophys Res Commun. 2015;463(4):600‐605. [DOI] [PubMed] [Google Scholar]
- 105. Tsukuda N, Yahagi K, Hara T, et al. Key bacterial taxa and metabolic pathways affecting gut short‐chain fatty acid profiles in early life. ISME J. 2021;15:2574‐2590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Koh A, de Vadder F, Kovatcheva‐Datchary P, Bäckhed F. From dietary fiber to host physiology: short‐chain fatty acids as key bacterial metabolites. Cell. 2016;165(6):1332‐1345. [DOI] [PubMed] [Google Scholar]
- 107. Marinelli L, Martin‐Gallausiaux C, Bourhis JM, Béguet‐Crespel F, Blottière HM, Lapaque N. Identification of the novel role of butyrate as AhR ligand in human intestinal epithelial cells. Sci Rep. 2019;9(1):643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Trompette A, Gollwitzer ES, Yadava K, et al. Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat Med. 2014;20(2):159‐166. [DOI] [PubMed] [Google Scholar]
- 109. Antunes KH, Fachi JL, de Paula R, et al. Microbiota‐derived acetate protects against respiratory syncytial virus infection through a GPR43‐type 1 interferon response. Nat Commun. 2019;10(1):3273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Zheng L, Kelly CJ, Battista KD, et al. Microbial‐derived butyrate promotes epithelial barrier function through IL‐10 receptor‐dependent repression of Claudin‐2. J Immunol. 2017;199(8):2976‐2984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Thio CL, Chi PY, Lai AC, Chang YJ. Regulation of type 2 innate lymphoid cell‐dependent airway hyperreactivity by butyrate. J Allergy Clin Immunol. 2018;142(6):1867‐1883. [DOI] [PubMed] [Google Scholar]
- 112. Folkerts J, Redegeld F, Folkerts G, et al. Butyrate inhibits human mast cell activation via epigenetic regulation of FcεRI‐mediated signaling. Allergy. 2020;75(8):1966‐1978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. O'Mahony L. Short chain fatty acids modulate mast cell activation. Allergy. 2020;75(8):1848‐1849. [DOI] [PubMed] [Google Scholar]
- 114. Sepahi A, Liu Q, Friesen L, Kim CH. Dietary fiber metabolites regulate innate lymphoid cell responses. Mucosal Immunol. 2021;14(2):317‐330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Macia L, Tan J, Vieira AT, et al. Metabolite‐sensing receptors GPR43 and GPR109A facilitate dietary fibre‐induced gut homeostasis through regulation of the inflammasome. Nat Commun. 2015;6:6734. [DOI] [PubMed] [Google Scholar]
- 116. Tan J, McKenzie C, Vuillermin PJ, et al. Dietary fiber and bacterial SCFA enhance oral tolerance and protect against food allergy through diverse cellular pathways. Cell Rep. 2016;15(12):2809‐2824. [DOI] [PubMed] [Google Scholar]
- 117. Paparo L, Nocerino R, Ciaglia E, et al. Butyrate as bioactive human milk protective component against food allergy. Allergy. 2021;76(5):1398‐1415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Arpaia N, Campbell C, Fan X, et al. Metabolites produced by commensal bacteria promote peripheral regulatory T‐cell generation. Nature. 2013;504(7480):451‐455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Furusawa Y, Obata Y, Fukuda S, et al. Commensal microbe‐derived butyrate induces the differentiation of colonic regulatory T cells. Nature. 2013;504(7480):446‐450. [DOI] [PubMed] [Google Scholar]
- 120. Levan SR, Stamnes KA, Lin DL, et al. Elevated faecal 12,13‐diHOME concentration in neonates at high risk for asthma is produced by gut bacteria and impedes immune tolerance. Nat Microbiol. 2019;4:1851‐1861. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Zhou CB, Zhou YL, Fang JY. Gut microbiota in cancer immune response and immunotherapy. Trends Cancer. 2021;7:647‐660. [DOI] [PubMed] [Google Scholar]