The human gut microbiome is a complex ecosystem consisting of over a trillion microorganisms. It has become an essential factor in the regulation of our immune system and overall physiological health. This detailed community of bacteria, archaea, viruses, and fungi not only carries out essential functions in the gastrointestinal tract but also considerably affects widespread processes throughout the body.1,2 The gut microbiome plays a critical role in various biochemical and metabolic pathways, improving nutrient absorption, assisting digestion, and enhancing metabolic activities.3 Also, these microorganisms are essential for immune regulation, serving as mediators that help calibrate the host’s immune response to environmental stimuli. Recent research has simplified the energetic interactions between the gut microbiome and widespread inflammatory responses. It has been shown that gut flora can activate and modulate these responses, which may have important repercussions on peripheral tissues.2,4 Meanwhile, host immune response can also affect the homeostasis of gut microbiota. Previous studies show that exposure mouse lungs to lipopolysaccharide (LPS) can trigger inflammation, leading to a substantial augmentation of gut microbiome.2 These findings imply that disruptions in gut microbiota may arise as a consequence of widespread inflammation, further complicating our understanding of host-microbe interactions.
Asthma is a prevalent chronic respiratory condition marked by limitations in airflow, increased bronchial responsiveness, excessive mucus production, and inflammation in the airways.5 It is estimated that approximately 334 million individuals are currently affected by this disease.6 A important portion of asthma attacks is attributed to an overabundance of Type 2 inflammation, and the contributions of immunoglobulin E (IgE) antibodies and T helper (Th) 2 cells have been investigated regarding their roles in the inflammatory processes.7 However, research into the interactions between the gut microbiome and airway immune responses has not received the same level of attention as the study of inflammatory mechanisms in asthma. Recently, there have been promising clinical outcomes related to using orally administered probiotics for repairing bronchial epithelium, which indicates potential adjunctive therapeutic benefits in asthma management.8 In addition, some experimental treatments designed to influence the interactions between the gut microbiome and airway immune responses have become potential options for asthma therapy. Evidence demonstrates that providing short-chain fatty acids (SCFAs), such as propionate, to neonatal mice lacking plasmacytoid dendritic cells (pDCs) can enhance the migration of monocyte-derived dendritic cells (moDCs) to the lungs, improve Sema4a signaling, and restore the response of Nrp-1+ regulatory T cells (Tregs), eventually leading to protection against severe viral bronchiolitis and subsequent asthma development.9 Thus, the modulation of the interaction between gut microbiota and airway immune responses may represent a critical area for future asthma treatment research. In this review, we will simplify the crosstalk between the gut microbiome and airway immune responses, along with their therapeutic implications for asthma.
Crosstalk Between Gut Microbiome and Airway Immune ResponsesGut Microbiome Distribution and Airway Immune ResponsesIn the gastrointestinal tract (GIT), basic composition of GIT microbiota is mainly composed of bacteria from the phyla Firmicutes (64%, mainly Gram-positive Clostridium, Bacillus, Lactobacillus, and Enterococcus species), and Bacteroidetes (23%, mostly Gram-negative Bacteroides and Prevotella species.10 Other microbial species include viruses, fungi, and even archaea.2 Studies indicate that the gut microbiome is a critical component of physiological balance, offering numerous benefits from the digestion of complex carbohydrates to the regulation of immune responses.11 Within this ecosystem, the composition and distribution of gut microbiota greatly impact host immune functions, suggesting that variations in these microbial populations may be associated with the pathophysiology of asthma and other allergic conditions. Among the various species present in the gut, Bacteroides fragilis is notable for its unique capacity to produce Polysaccharide A (PSA), which is essential for the differentiation of cluster of differentiation 4-positive (CD4+) T cells and the regulation of T helper cell responses. This interaction is essential in shaping the immune environment, as it encourages a balanced response between Th1 and Th2 cells.12–14 At the same time, the colonization of segmented filamentous bacteria (SFB) activates the ILC3/IL-22/SAA1/2 signaling pathway, which subsequently boosts the production of IL-17A by RORγt Th17 cells.14 Increased levels of IL-17A have been linked to a severe, neutrophilic inflammatory profile characteristic of certain asthma types.15 In addition, fungi within the gut microbiota may also impact asthma-related immune responses by supporting the generation of immune cells in the gastrointestinal tract.7 Mouse models have shown that fungal overgrowth can worsen Th2 cell-mediated airway inflammation after an airway challenge.2 Collectively, these studies propose that there exist interactions between different gut microbiota species and airway immune responses and play an indispensable role in asthma development and exacerbation.
Gut Microbiome Dysbiosis in Early-Life and Airway Immune ResponsesAccording to the “hygiene hypothesis”, appropriate exposure to particular microbiome constituents early in life is essential to stimulate the immune development and maturation, while their absence could heighten the risk of developing asthma and allergic diseases.16 Research indicates that the early microbial environment, influenced by commensal microbiota, plays a critical role in promoting immune maturation and establishing tolerance to environmental antigens.17 Early-life dysbiosis, often triggered by factors such as antibiotic use, can disrupt immune development and raise the likelihood of asthma and allergies.18 For example, research shows that administering antibiotics during the first six months of life is associated with a higher risk of developing allergic diseases.19,20 The period between infancy and early childhood is essential for the development of the immune system and the microbiome due to crosstalk at various levels.14 A major decline in the relative abundance of beneficial microbiota, including genera such as Lachnospira, Veillonella, Faecalibacterium, and Rothia, has been documented in children with a tendency toward asthma.21,22 One of the key factors is delivery through a Caesarean section (CS), which postpones the colonization of bacteria in the intestines. This method notably decreases the presence of beneficial bacteria, including Dolosigranulum and Corynebacterium, which are linked to health.23 And the presence of Clostridium in the neonatal gut shortly after a cesarean section is greatly associated with the development of recurrent wheeze (OR = 1.75; 95% CI 1.09 to 2.80) and allergic sensitization (OR = 1.54; 95% CI 1.02 to 2.31), suggesting a potential increase in asthma risk later in life.24,25
Short-Chain Fatty Acids and Airway Immune ResponsesMetabolites derived from microbes play an important role in asthma by influencing immune responses in the airways. The gut microbiota possesses an important metabolic capacity to change components derived from the host and dietary elements, such as lipids, carbohydrates, and proteins, into various metabolites that can either promote or hinder the development of the host’s mucosal immunity.26 Some of these metabolites, including SCFAs and secondary bile acids, exhibit antimicrobial properties, thereby providing protection against pathogenic bacteria and promoting immune homeostasis.27 SCFAs are primarily produced by Firmicutes in the gastrointestinal tract from dietary fibers or through the fermentation of nondigestible carbohydrates.28 As metabolites, SCFAs have been shown to modify cellular functions by influencing gene expression, chemotaxis, differentiation, proliferation, and apoptosis.29 The SCFA-sensing G-protein-coupled receptor GPR109A, which is activated by SCFAs in intestinal epithelial cells, colonic macrophages, and dendritic cells, can induce anti-inflammatory effects, promote the differentiation of regulatory T cells and IL-10-producing T cells, and support epithelial homeostasis.17,30,31 Therefore, it is likely that SCFAs have an impact on airway immune responses through these various mechanisms (Figure 1).
Figure 1 Molecular mechanism of gut microbiota and its metabolites in airway immunity of asthma (drawn by Figdraw, ID: TWAOO5cdc2). Short-chain fatty acid (SCFAs), including acetate, propionate, and butyrate, mainly produced by bacterial metabolism in the human gut after dietary fiber intake, inhibits HDAC function and enhances FOXP3 expression, thereby promoting Treg differentiation and IL-10 production via GPR43 and other pathways. Polysaccharide A (PSA) derived from Bacteroides fragilis, can also act on Tregs through TLR2 to promote Treg function by enhancing expression of IL-10. Adhesion of segmented filamentous bacteria (SFB) prompts intestinal epithelial cells (IECs) to release serum amyloid A (SAA), establishing a feedback loop with antigen-presenting cells (APCs) and type 3 innate lymphoid cells (ILC3) that facilitates interleukin-mediated differentiation of naive CD4+ T cells into Th17 cells. In Peyer’s patch, B cells generated increased immunoglobulin E (IgE) via a CD4 T-cell / IL-4-dependent mechanism. Immune-related cells and cytokines produced by these pathways travel through blood circulation to the lungs, affecting Th2-mediated responses in asthma. Th2 cells release IL-4, IL-13, and IL-5 in response to allergen stimulation, facilitating inflammation and tissue remodeling by encouraging the infiltration and activation of eosinophils and basophils, enhancing IgE production by B cells, and causing excessive mucus secretion.
Three primary SCFAs, acetate, propionate, and butyrate, are produced by gut bacteria in a molar ratio of 60:20:20, contingent upon the fiber content of the diet.22 Other SCFAs present in the gut, such as valerate, caproate, and isovalerate, are found in smaller quantities.32 Research has shown that the antimicrobial effect induced by SCFA butyrate is the strongest when compared to propionate, while such an effect has not been observed with acetate.33 Besides, the study conducted by Schulthess et al demonstrated that oral butyrate supplementation effectively limits the spread of pathogenic bacteria. Evidence indicates that children with the highest levels of propionate and butyrate (≥95th percentile) in their feces at one year of age exhibited considerably lower instances of atopic sensitization and reduced odds of developing asthma between the ages of 3 and 6 years.34 Similarly, propionate intake can diminish the effectiveness of newly recruited dendritic cells (DCs) and enhance the responses of effector Th2 cells, thereby providing protection against allergic airway inflammation in mice, which is associated with decreased levels of serum total IgE.35,36 When dietary supplementation containing butyrate, propionate, and acetate was administered, the physiological function of DCs in vancomycin-treated mice was modulated, correlating with weakened allergic immune responses in the lungs.35 The introduction of SCFAs in microbiota-gut-lung communication emphasizes the influence of metabolites mediated by changes in gut microbiota, which may contribute to health by regulating immune tolerance and inflammation.
Implications for Asthma Treatment StrategiesDietary Structure and Eating HabitsDiet is a fundamental necessity for our lives and is essential not only for enhancing health and supporting growth but also for influencing the diverse microbial communities within GIT. The sources, types, and quality of food can shape the gut microbiome by altering its composition and function, which in turn affects interactions between hosts and microbes.37 Our focus is on the role of fiber intake and healthy dietary patterns, which contribute to the prevention and treatment of asthma generation and development. Chronic inflammation of the airways is a prominent characteristic of asthma, and emerging evidence indicates that diet can influence this inflammatory process.38,39 Notably, previous studies have demonstrated a major correlation between the prevalence of asthma and the adoption of westernized dietary patterns.40 The Western diet is typically characterized by a higher consumption of animal products, with inadequate intake of fruits, vegetables, whole grains, and legumes. This dietary pattern has been shown to exacerbate airway inflammation by increasing the production of cytokines, which subsequently affects the development and manifestation of asthma.41,42 In contrast, a high intake of fruits and vegetables is associated with a reduction in pro-inflammatory cytokines and an increase in anti-inflammatory markers, thereby decreasing the risk of asthma exacerbation.41,43 Besides, another study indicated that a higher consumption of vegetables, legumes, fruits, nuts, cereals, and fish, along with a reduced intake of meat and poultry, is linked to lower odds of asthma attacks.44 Conversely, it has been demonstrated that a diet high in fat and low in fiber can influence the incidence of allergic diseases.45 This may contribute to the rising prevalence of allergic diseases, including asthma, allergic rhinitis, and other allergic conditions over recent decades.46
Dietary fiber, defined as edible carbohydrate polymers that resist digestion by endogenous enzymes, plays a critical role in regulating intestinal bacteria and maintaining immune homeostasis.37,46 Recent research has confirmed that the supplementation of dietary fiber enhances the proliferation of beneficial bacteria, such as Bifidobacterium and Lactobacillus, in healthy adults.45 As a result, certain types of dietary fibers can be categorized as prebiotics.47 The gut microbiota can effectively metabolize and use these fibers, leading to an increase in the concentration of circulating SCFAs.36 Some studies have indicated that dietary fiber may positively influence the balance of Th1/Th2 immunity, considerably inhibit inflammatory responses in allergic rhinitis and concurrent asthma and reduce airway inflammation by suppressing DC function.46,48 Both animal studies and clinical trials involving humans have demonstrated that early intake of dietary oligosaccharides can help prevent the onset of allergic asthma and other allergic conditions.48,49 Conversely, a long-term dietary pattern characterized by insufficient dietary fiber intake is strongly associated with an enhanced risk of airway allergic diseases, including asthma, due to a decrease in butyrate- and SCFA-producing bacteria.14,46 Consequently, appropriate amounts of dietary fiber can regulate intestinal bacteria, preserve immune homeostasis, and thus may be regarded as an effective therapeutic strategy for the treatment and prevention of asthma. Although it presents challenges, this healthy dietary approach should be advocated for the majority of asthma patients.
Prebiotics and Probiotics UsePrebiotics are characterized as specialized substrates that are selectively fermented by host microorganisms. They offer numerous health benefits, which include defense against pathogens, immune modulation, improved bowel function, positive metabolic effects, and enhanced satiety. Also, they play an essential role in maintaining the stability of microbiota48,50–53. Importantly, certain dietary fibers such as fructooligosaccharides, galactooligosaccharides, and inulin have been recognized as key prebiotic agents. These agents can notably influence the composition and functional capacities of probiotics while also favorably modifying the gut microbiota towards a more beneficial state.26,54 At the same time, the interactions between probiotics and prebiotics indicate a complex relationship in which probiotics exert their beneficial effects through well-documented molecular and cellular mechanisms. These mechanisms include the enhancement of innate immunity, reduction of inflammation induced by pathogens, and promotion of the maturation of the mucosal immune system. Probiotics achieve these beneficial effects by secreting various antimicrobial compounds, which include organic acids and SCFAs.55,56 A important study indicated that a four-week supplementation with synbiotics, which consisted of 90% short-chain galacto-oligosaccharides and 10% long-chain fructo-oligosaccharides, resulted in a notable decrease in Th2 cytokine production and an improvement in peak expiratory flow (PEF) among patients with allergic asthma.57 Therefore, it can be inferred that the strategic incorporation of prebiotics and probiotics could represent a feasible therapeutic approach for managing conditions marked by airway inflammation, thus necessitating further investigation into their synergistic effects within clinical environments.
Several studies have indicated that probiotics might are an alternative form of medication for asthma or as an adjunct to asthma therapy.58 Probiotics can greatly reduce disease severity primarily by modulating immune responses that are involved in allergic inflammation.59 Conversely, a lack of pathogen exposure during early childhood heightens the risk of developing allergic asthma, resulting in a shift in the immune response from a Th1 to a Th2 response pattern.54 Prior research has demonstrated that probiotics can affect respiratory immunity by influencing the production of interferons (IFNs), diminishing the synthesis of IL-4, IL-13, and IgE, redirecting the Th2 response toward a Th1 type, and collectively contributing to the mitigation of allergic predisposition and reactions.50,60 Also, both prebiotics and probiotics may hinder the activation of genes related to asthma through the PI3K/Akt and TLR4/NF-κB pathways, thereby decelerating the progression of asthma.54 Recently, some beneficial bacteria, including Lactobacillus, Bifidobacterium, Lachnospira, and Akkermansia, have demonstrated anti-asthmatic effects.61Lactobacillus and Bifidobacterium, considered traditional probiotics, are of particular interest.62 For instance, Lactobacillus exhibits strong immunomodulatory capabilities and improves gastrointestinal disorders.63 Meanwhile, the administration of Lactobacillus has been shown to alleviate asthma symptoms.64 It has also been established that treatment with plantarum 06CC2 in ovalbumin-sensitized mice leads to a reduction in the levels of histamine, total IgE, and ovalbumin-specific IgE in the serum, thereby greatly easing allergic symptoms.65 In a separate study, L. reuteri, a member of the Lactobacillus genus, modifies specific gut microbes and enhances butyrate production, which encourages regulatory T-cell proliferation and mitigates allergy-associated Th2 immune responses.66,67Bifidobacterium, another major probiotic, has been shown to alleviate respiratory symptoms and enhance quality of life.68,69 It was demonstrated that MRx0004, an important type of Bifidobacterium, can reduce the infiltration of neutrophils, eosinophils in mice with severe asthma.70 And Bifidobacterium infantis CGMCC313-2 has been found to greatly lower serum levels of IgE, IgG1, IL-4, and IL-13 in mice exhibiting allergic asthma.71 Comprehensively, prebiotics and probiotics may be a therapeutic strategy for allergic asthma, warranting further investigation into their mechanisms of action, optimal dosing, and safety, all of which could contribute to more rational treatment approaches for asthma.
Antibiotics UseThe introduction of antibiotics marks one of the most important developments in modern medicine, fundamentally changing the field of infectious disease management and leading to a major decrease in both morbidity and mortality related to bacterial infections, thereby protecting countless lives and improving overall public health outcomes.72,73 Nevertheless, new studies have started to reveal possible unintended consequences associated with antibiotic use, especially when administered during critical periods of early-life development, increased risk of developing early persistent asthma.74,75 Besides, the disruption of the gut microbiome caused by antibiotics is linked to various infectious and autoimmune diseases affecting the gastrointestinal system.76
Currently, the primary mechanisms through which antibiotics influence the occurrence of allergic diseases, including asthma, are as follows.77 To begin with, antibiotics may direct the immune system towards an allergic pathway by diminishing the severity and duration of infections.19 The treatment with antibiotics can result in a reduction of phylogenetic diversity, which may lead to the displacement of potential pathogens and a delay in the maturation of both the microbiome and the immune system, eventually impacting immune homeostasis.78 A recent investigation conducted by the STEPS study has shown that exposure to antibiotics before the age of one is linked to an increased vulnerability to asthma by the age of seven years.79 Secondly, it has been demonstrated that the Th2-skewed response induced by antibiotics may play an important role in the etiology of allergies.19 Evidence indicates that the administration of oral antibiotics can lead to enhanced serum IgE concentrations, exacerbating basophil-mediated Th2 inflammation by increasing the basophil population during asthma attacks in older children.80 Lastly, antibiotics may exert their effects by disturbing the gut microbiota.81 The perturbation of the gut microbiome induced by antibiotics may result in the depletion of SCFAs, which in turn can lead to hyperactivation of intestinal macrophages and expansion of pro-inflammatory Th cells, consequently increasing susceptibility to airway inflammation and infections.14 In addition, it has been demonstrated that exposure to antibiotics in early life can alter the composition and function of the microbiome in the lungs, leading to dysregulation of innate and adaptive immunity, thus assisting the development of asthma.77,79 A study conducted in Sweden, involving 493,785 children, has suggested a strong positive association between antibiotic use and the incidence and progression of asthma, particularly during the first six months of life.82 This evidence indicates that reducing the excessive consumption of antibiotics is highly advisable for the prevention of asthma in childhood.83
While there are no definitive guidelines for the early use of antibiotics, it is worthwhile to explore methods of managing early antibiotic administration to reduce the risk of asthma as an emerging research focus. What’s more, another study has shown that supplementation with SCFA butyrate may reverse the hypo-responsiveness of intestinal macrophages induced by antibiotics, support appropriate T cell functions, and avert immune dysfunction linked to antibiotics.84 Accordingly, aiming to restore macrophage homeostasis following antibiotic treatment could represent a novel and effective approach to preventing enduring immune dysfunction in patients with asthma.84
Breast FeedingEnvironmental exposures have gained increasing recognition as important factors contributing to the onset of allergic diseases, particularly through their interactions with various human host factors.85 Specifically, breastfeeding is recognized as one of the earliest environmental exposures associated with respiratory health, with numerous studies demonstrating its protective effects against the development of asthma during childhood.86,87 Research conducted by Dogaru et al indicates that exclusive breastfeeding for the initial six months of life considerably lowers the likelihood of developing asthma and other allergic conditions in later years.88 Besides, a systematic review by reinforces this claim, emphasizing the critical role of breastfeeding influencing the infant’s immune system and decreasing asthma incidence.89
The composition of human breast milk is detailed, comprising various factors that engage with the infant’s immune system and intestinal environment, including allergens, cytokines, immunoglobulins, polyunsaturated fatty acids, and chemokines.90 These factors have the potential to modify the gut microbiome and subsequent immune development in children, thereby affecting the risk of various respiratory infections.91 Besides, multiple studies indicate that weaning is linked to strong immune responses directed at the developing intestinal microbiota, thereby protecting against pathological imprinting through the induction of RORγt Tregs.92 The mucosal immune system around the time of weaning undergoes a major transformation from the neonatal to the adult state, which is considered an important phase in the maturation of the immune system following birth.93,94 In addition, another study has revealed that infants who ceased breastfeeding and began consuming solid foods within the first three months of life are more vulnerable to allergies, and respiratory and gastrointestinal infections.95 This raises important questions regarding the appropriate timing and manner in which the breastfeeding process should conclude. It has been suggested that an extended duration of breastfeeding can reduce the risk of developing allergic asthma across all age groups.96 Exclusive breastfeeding for a minimum of six months and partial breastfeeding for up to one year is highly recommended, as this may alleviate the incidence of respiratory infections during infancy.97,98 Based on these findings, breastfeeding is deemed to possess properties that may prevent asthma throughout an individual’s life.99 Nevertheless, other factors may partially influence the relationship between breastfeeding and allergic diseases, including the diets of both the mother and the infant, maternal microbiota, and exposure to exogenous allergens.90 Thus, further research into breastfeeding is essential to determine the optimal breastfeeding strategies for the prevention and treatment of asthma.
ConclusionTo summarize, the complex interaction between the host immune system and the gut microbiome stands out as an essential factor in maintaining immune homeostasis. This review has thoroughly examined a considerable amount of evidence that associates the gut microbiome with the development and regulation of airway immunity. It emphasizes the diverse mechanisms through which various gut microbial species and their metabolites influence immune responses within the airways. Importantly, SCFAs have been recognized as key contributors to the modulation of host immunity, playing an essential role in decreasing allergic reactions and alleviating airway inflammation.100 The therapeutic implications of this relationship are major, particularly considering asthma management. The findings indicate that dietary modifications and the encouragement of early breastfeeding practices may function as protective strategies against the emergence of asthma and other related conditions. Besides, the timing and administration of probiotics and antibiotics in early life have been correlated with the development of asthma, emphasizing the necessity for a careful approach to these interventions. Despite the encouraging insights provided by current research, a critical gap exists concerning the establishment of specific protocols for using probiotics and antibiotics during early childhood. The available literature lacks thorough clinical trials that define optimal strategies for these interventions, suggesting an urgent need for further studies.79,101 Future research should focus on clarifying the mechanisms that support the gut-lung axis and on creating evidence-based guidelines for microbiome-targeted interventions. Eventually, incorporating these findings into clinical practice could lead to innovative strategies for preventing and treating asthma and related respiratory conditions. By concentrating on the modulation of the microbiome during early life, it may be feasible to bolster immune resilience and lessen the impact of asthma, thereby enhancing public health outcomes. The ongoing investigation into the gut microbiome’s role in airway immunity offers substantial potential for advancing our comprehension of asthma pathogenesis and for formulating effective preventive measures.
Data Sharing StatementThe original contributions presented in the study are included in the article. Further inquiries can be directed to the corresponding author.
Ethics Approval and Consent to ParticipateEthical review and approval were not required for the study on human participants in accordance with the local legislation and institutional requirements.
AcknowledgmentsWe thank all the institutions affiliated with the authors for supporting this review.
Author ContributionsAll authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.
FundingThis study was supported by the Joint Foundation Established by Enterprises and the Basic and Applied Basic Research Foundation of Guangdong Province, China (Project Number:2022A1515220169), Natural Science Foundation of China (Project Number:81300012), and Appropriate Health Technology Promotion Project of Guangdong Province (Project Number:202006181142034974).
DisclosureThe authors have no conflicts of interest to declare.
References1. de Oliveira GLV, Oliveira CNS, Pinzan CF, de Salis LVV, Cardoso CRDB. Microbiota modulation of the gut-lung axis in COVID-19. Front Immunol. 2021;12:635471. doi:10.3389/fimmu.2021.635471
2. Barcik W, Boutin RCT, Sokolowska M, et al. The role of lung and gut microbiota in the pathology of asthma. Immunity. 2020;52(2):241–255. doi:10.1016/j.immuni.2020.01.007
3. Heintz-Buschart A, Wilmes P. Human gut microbiome: function matters. Trends Microbiol. 2018;26(7):563–574. doi:10.1016/j.tim.2017.11.002
4. Perler BK, Friedman ES, Wu GD. The role of the gut microbiota in the relationship between diet and human health. Annu Rev Physiol. 2023;85:449–468. doi:10.1146/annurev-physiol-031522-092054
5. Thomas D, Mcdonald VM, Pavord ID, et al. Asthma remission: what is it and how can it be achieved? Eur Respir J. 2022;60(5):2102583. doi:10.1183/13993003.02583-2021
6. Pavón-Romero GF, serrano-pérez NH, García-Sánchez L, et al. Neuroimmune Pathophysiology in Asthma. Front Cell Dev Biol. 2021;9:663535. doi:10.3389/fcell.2021.663535
7. Boutin RC, Petersen C, Woodward SE. Bacterial-fungal interactions in the neonatal gut influence asthma outcomes later in life. Elife. 2021;10.
8. Dwivedi M, Radichev I, Kemp EH. Alteration of immune-mechanisms by human microbiota and development and prevention of human diseases. J Immunol Res. 2017;2017:6985256. doi:10.1155/2017/6985256
9. Lynch JP, Werder RB, Loh Z. Plasmacytoid dendritic cells protect from viral bronchiolitis and asthma through semaphorin 4a-mediated T reg expansion. J Exp Med. 2018;215(2):537–557. doi:10.1084/jem.20170298
10. Lin T-L, Shu -C-C, Chen Y-M. Like cures like: pharmacological activity of anti-inflammatory lipopolysaccharides from gut microbiome. Front Pharmacol. 2020;11:554. doi:10.3389/fphar.2020.00554
11. Fan Y, Pedersen O. Gut microbiota in human metabolic health and disease. Nat Rev Microbiol. 2021;19(1):55–71. doi:10.1038/s41579-020-0433-9
12. Wosinska L, Cotter PD, O’sullivan O, et al. The potential impact of probiotics on the gut microbiome of athletes. Nutrients. 2019;11(10):2270. doi:10.3390/nu11102270
13. Frati F, Salvatori C, Incorvaia C, et al. The role of the microbiome in asthma: the gut⁻lung axis. Int J mol Sci. 2018;20(1):123. doi:10.3390/ijms20010123
14. Zheng D, Liwinski T, Elinav E. Interaction between microbiota and immunity in health and disease. Cell Res. 2020;30(6):492–506. doi:10.1038/s41422-020-0332-7
15. Ito T, Hirose K, Saku A, et al. IL-22 induces Reg3γ and inhibits allergic inflammation in house dust mite-induced asthma models. J Exp Med. 2017;214(10):3037–3050. doi:10.1084/jem.20162108
16. Chunxi L, Haiyue L, Yanxia L, et al. The gut microbiota and respiratory diseases: new evidence. J Immunol Res. 2020;2020:2340670. doi:10.1155/2020/2340670
17. Kemter AM, Nagler CR. Influences on allergic mechanisms through gut, lung, and skin microbiome exposures. J Clin Invest. 2019;129(4):1483–1492. doi:10.1172/JCI124610
18. Arrieta M-C, Stiemsma LT, Dimitriu PA. Early infancy microbial and metabolic alterations affect risk of childhood asthma. Sci Transl Med. 2015;7(307):307ra152. doi:10.1126/scitranslmed.aab2271
19. Dharmage SC, Lodge CJ, Lowe AJ, Allen KJ. Antibiotics and risk of asthma: a debate that is set to continue. Clin Exp Allergy. 2015;45(1):6–8. doi:10.1111/cea.12424
20. Mitre E, Susi A, Kropp LE, et al. Association between use of acid-suppressive medications and antibiotics during infancy and allergic diseases in early childhood. JAMA Pediatr. 2018;172(6):e180315. doi:10.1001/jamapediatrics.2018.0315
21. Le Doare K, Holder B, Bassett A, et al. Mother’s milk: a purposeful contribution to the development of the infant microbiota and immunity. Front Immunol. 2018;9:361. doi:10.3389/fimmu.2018.00361
22. Stiemsma LT, Turvey SE. Asthma and the microbiome: defining the critical window in early life. Allergy Asthma Clin Immunol. 2017;13:3. doi:10.1186/s13223-016-0173-6
23. Kim G, J B, Kim MJ, et al. Delayed establishment of gut microbiota in infants delivered by cesarean section. Front Microbiol. 2020;11:2099. doi:10.3389/fmicb.2020.02099
24. Shaterian N, Abdi F, Ghavidel N, et al. Role of cesarean section in the development of neonatal gut microbiota: a systematic review. Open Med. 2021;16(1):624–639. doi:10.1515/med-2021-0270
25. Penders J, Thijs C, Van Den Brandt PA, et al. Gut microbiota composition and development of atopic manifestations in infancy: the KOALA birth cohort study. Gut. 2007;56(5):661–667. doi:10.1136/gut.2006.100164
26. Campbell C, Kandalgaonkar MR, GOLONKA RM, et al. Crosstalk between gut microbiota and host immunity: impact on inflammation and immunotherapy. Biomedicines. 2023;11(2). doi:10.3390/biomedicines11020294
27. Rea MC, Clayton E, O’Connor PM. Antimicrobial activity of lacticin 3147 against clinical Clostridium difficile strains. J Med Microbiol. 2007;56(Pt 7):940–946. doi:10.1099/jmm.0.47085-0
28. Dupraz L, Magniez A, Rolhion N, et al. Gut microbiota-derived short-chain fatty acids regulate IL-17 production by mouse and human intestinal γδ T cells. Cell Rep. 2021;36(1):109332. doi:10.1016/j.celrep.2021.109332
29. Corrêa-oliveira R, FACHI JL, VIEIRA A, et al. Regulation of immune cell function by short-chain fatty acids. Clin Transl Immunology. 2016;5(4):e73. doi:10.1038/cti.2016.17
30. Bach Knudsen KE, Lærke HN, Hedemann MS. Impact of diet-modulated butyrate production on intestinal barrier function and inflammation. Nutrients. 2018;10(10):1499.
31. Singh N, Gurav A, Sivaprakasam S, et al. Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis [J]. Immunity. 2014;40(1):128–139. doi:10.1016/j.immuni.2013.12.007
32. Kayama H, Okumura R, Takeda K. Interaction between the microbiota, epithelia, and immune cells in the intestine. Annu Rev Immunol. 2020;38:23–48. doi:10.1146/annurev-immunol-070119-115104
33. Schulthess J, Pandey S, Capitani M, et al. The Short Chain Fatty Acid Butyrate Imprints an Antimicrobial Program in Macrophages. Immunity. 2019;50(2):432–45.e7. doi:10.1016/j.immuni.2018.12.018
34. 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:10.1111/all.13660
35. Wypych TP, Wickramasinghe LC, Marsland BJ. The influence of the microbiome on respiratory health. Nat Immunol. 2019;20(10):1279–1290. doi:10.1038/s41590-019-0451-9
36. 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:10.1038/nm.3444
37. Makki K, Deehan EC, Walter J, et al. The impact of dietary fiber on gut microbiota in host health and disease. Cell Host Microbe. 2018;23(6):705–715. doi:10.1016/j.chom.2018.05.012
38. Crespo A, Giner J, Torrejón M, et al. Clinical and inflammatory features of asthma with dissociation between fractional exhaled nitric oxide and eosinophils in induced sputum. J Asthma. 2016;53(5):459–464. doi:10.3109/02770903.2015.1116086
39. Berthon BS, Macdonald‐Wicks LK, Gibson PG, Wood LG. Investigation of the association between dietary intake, disease severity and airway inflammation in asthma. Respirology. 2013;18(3):447–454. doi:10.1111/resp.12015
40. Kim J-H, Ellwood PE, Asher MI. Diet and asthma: looking back, moving forward. Respir Res. 2009;10(1):49. doi:10.1186/1465-9921-10-49
41. Alwarith J, Kahleova H, Crosby L, et al. The role of nutrition in asthma prevention and treatment. Nutr Rev. 2020;78(11):928–938. doi:10.1093/nutrit/nuaa005
42. Kim HY, Lee HJ, Chang YJ. Interleukin-17-producing innate lymphoid cells and the NLRP3 inflammasome facilitate obesity-associated airway hyperreactivity. Nat Med. 2014;20(1):54–61. doi:10.1038/nm.3423
43. Seyedrezazadeh E, Moghaddam MP, Ansarin K, et al. Fruit and vegetable intake and risk of wheezing and asthma: a systematic review and meta-analysis. Nutr Rev. 2014;72(7):411–428. doi:10.1111/nure.12121
44. Rice JL, Romero KM, Galvez Davila RM. Association between adherence to the Mediterranean diet and asthma in Peruvian children. Lung. 2015;193(6):893–899. doi:10.1007/s00408-015-9792-9
45. Ting NL-N, Lau HC-H, Yu J. Cancer pharmacomicrobiomics: targeting microbiota to optimise cancer therapy outcomes. Gut. 2022;71(7):1412–1425. doi:10.1136/gutjnl-2021-326264
46. Zhang Z, L S, Pang W, et al. Dietary fiber intake regulates intestinal microflora and inhibits ovalbumin-induced allergic airway inflammation in a mouse model. PLoS One. 2016;11(2):e0147778. doi:10.1371/journal.pone.0147778
47. Holscher HD. Dietary fiber and prebiotics and the gastrointestinal microbiota. Gut Microbes. 2017;8(2):172–184. doi:10.1080/19490976.2017.1290756
48. Blanco-Pérez F, Steigerwald H, Schülke S, et al. The dietary fiber pectin: health benefits and potential for the treatment of allergies by modulation of gut microbiota. Curr Allergy Asthma Rep. 2021;21(10):43. doi:10.1007/s11882-021-01020-z
49. Kostadinova AI, Pablos-Tanarro A, Diks MA. Dietary intervention with β-lactoglobulin-derived peptides and a specific mixture of fructo-oligosaccharides and bifidobacterium breve m-16v facilitates the prevention of whey-induced allergy in mice by supporting a tolerance-prone immune environment. Front Immunol. 2017;8:1303. doi:10.3389/fimmu.2017.01303
50. Balta I, Butucel E, Mohylyuk V, et al. Novel insights into the role of probiotics in respiratory infections, allergies, cancer, and neurological abnormalities. Diseases. 2021;9(3):60. doi:10.3390/diseases9030060
51. Sanders ME, Merenstein DJ, Reid G, Gibson GR, Rastall RA. Probiotics and prebiotics in intestinal health and disease: from biology to the clinic. Nat Rev Gastroenterol Hepatol. 2019;16(10):605–616. doi:10.1038/s41575-019-0173-3
52. Gibson GR, Hutkins R, Sanders ME. Expert consensus document: the International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nat Rev Gastroenterol Hepatol. 2017;14(8):491–502. doi:10.1038/nrgastro.2017.75
53. Van Baarlen P, Wells JM, Kleerebezem M. Regulation of intestinal homeostasis and immunity with probiotic lactobacilli. Trends Immunol. 2013;34(5):208–215. doi:10.1016/j.it.2013.01.005
54. Wu Z, Mehrabi Nasab E, Arora P, Athari SS. Study effect of probiotics and prebiotics on treatment of OVA-LPS-induced of allergic asthma inflammation and pneumonia by regulating the TLR4/NF-kB signaling pathway. J Transl Med. 2022;20(1):130. doi:10.1186/s12967-022-03337-3
55. Compare D, Rocco A, Coccoli P, et al. Lactobacillus casei DG and its postbiotic reduce the inflammatory mucosal response: an ex-vivo organ culture model of post-infectious irritable bowel syndrome. BMC Gastroenterol. 2017;17(1):53. doi:10.1186/s12876-017-0605-x
56. Lopez-Santamarina A, Lamas A, Del Carmen Mondragón A, et al. Probiotic effects against virus infections: new weapons for an old war. Foods. 2021;10(1):130. doi:10.3390/foods10010130
57. Van De Pol MA, Lutter R, Smids BS, Weersink EJM, Van Der Zee JS. Synbiotics reduce allergen-induced T-helper 2 response and improve peak expiratory flow in allergic asthmatics. Allergy. 2011;66(1):39–47. doi:10.1111/j.1398-9995.2010.02454.x
58. Jin SW, Lee GH, Jang MJ, et al. Lactic acid bacteria ameliorate diesel exhaust particulate matter-exacerbated allergic inflammation in a murine model of asthma. Life. 2020;10(11):260.
59. Osborn DA, Sinn JK. Probiotics in infants for prevention of allergic disease and food hypersensitivity. Cochrane Database Syst Rev. 2007;4:Cd006475.
60. Cristofori F, Dargenio VN, Dargenio C, et al. Anti-inflammatory and immunomodulatory effects of probiotics in gut inflammation: a door to the body. Front Immunol. 2021;12:578386. doi:10.3389/fimmu.2021.578386
61. Jeong J, Lee HK. The role of CD4(+) T cells and microbiota in the pathogenesis of asthma. Int J mol Sci. 2021;22(21):11822. doi:10.3390/ijms222111822
62. Sanders ME, Benson A, Lebeer S, Merenstein DJ, Klaenhammer TR. Shared mechanisms among probiotic taxa: implications for general probiotic claims. Curr Opin Biotechnol. 2018;49:207–216. doi:10.1016/j.copbio.2017.09.007
63. Zhang Z, Lv J, Pan L, et al. Roles and applications of probiotic lactobacillus strains. Appl Microbiol Biotechnol. 2018;102(19):8135–8143. doi:10.1007/s00253-018-9217-9
64. Du T, Lei A, Zhang N, et al. The beneficial role of probiotic lactobacillus in respiratory diseases. Front Immunol. 2022;13:908010. doi:10.3389/fimmu.2022.908010
65. George Kerry R, Patra JK, Gouda S, Park Y, Shin H-S, Das G. Benefaction of probiotics for human health: a review. J Food Drug Anal. 2018;26(3):927–939. doi:10.1016/j.jfda.2018.01.002
66. Li L, Fang Z, Liu X, et al. Lactobacillus reuteri attenuated allergic inflammation induced by HDM in the mouse and modulated gut microbes. PLoS One. 2020;15(4):e0231865. doi:10.1371/journal.pone.0231865
67. Arrieta M-C, Arévalo A, Stiemsma L. Associations between infant fungal and bacterial dysbiosis and childhood atopic wheeze in a nonindustrialized setting. J Allergy Clin Immunol. 2018;142(2):424–34.e10. doi:10.1016/j.jaci.2017.08.041
68. Drago L, Cioffi L, Giuliano M, et al. The Probiotics in Pediatric Asthma Management (PROPAM) study in the primary care setting: a randomized, controlled, double-blind trial with ligilactobacillus salivarius LS01 (DSM 22775) and bifidobacterium breve B632 (DSM 24706). J Immunol Res. 2022;2022:3837418. doi:10.1155/2022/3837418
69. Miraglia Del Giudice M, Indolfi C, Capasso M, et al. Bifidobacterium mixture (B longum BB536, B infantis M-63, B breve M-16V) treatment in children with seasonal allergic rhinitis and intermittent asthma. Ital J Pediatr. 2017;43(1):25. doi:10.1186/s13052-017-0340-5
70. Raftis EJ, Delday MI, Cowie P. Bifidobacterium breve MRx0004 protects against airway inflammation in a severe asthma model by suppressing both neutrophil and eosinophil lung infiltration. Sci Rep. 2018;8(1):12024. doi:10.1038/s41598-018-30448-z
71. Liu M-Y, Yang Z-Y, Dai W-K. Protective effect of Bifidobacterium infantis CGMCC313-2 on ovalbumin-induced airway asthma and β-lactoglobulin-induced intestinal food allergy mouse models. World J Gastroenterol. 2017;23(12):2149–2158. doi:10.3748/wjg.v23.i12.2149
72. Cook MA, Wright GD. The past, present, and future of antibiotics. Sci Transl Med. 2022;14(657):eabo7793. doi:10.1126/scitranslmed.abo7793
73. Gould IM, Bal AM. New antibiotic agents in the pipeline and how they can help overcome microbial resistance. Virulence. 2013;4(2):185–191. doi:10.4161/viru.22507
74. Kayyal M, Javkar T, Firoz Mian M, et al. Sex dependent effects of post-natal penicillin on brain, behavior and immune regulation are prevented by concurrent probiotic treatment. Sci Rep. 2020;10(1):10318. doi:10.1038/s41598-020-67271-4
75. Lu Y, Wang Y, Wang J, et al. Early-life antibiotic exposure and childhood asthma trajectories: a national population-based birth cohort. Antibiotics. 2023;12(2):314. doi:10.3390/antibiotics12020314
76. Fishbein SRS, Mahmud B, Dantas G. Antibiotic perturbations to the gut microbiome. Nat Rev Microbiol. 2023;21(12):772–788. doi:10.1038/s41579-023-00933-y
77. Bentouhami H, Bungwa MK, CASAS L, et al. Asthma occurrence in children and early life systemic antibiotic use: an incidence density study. Allergy Asthma Clin Immunol. 2023;19(1):18. doi:10.1186/s13223-023-00773-8
78. Cortes L, Wopereis H, Tartiere A, et al. Metaproteomic and 16S rRNA gene sequencing analysis of the infant fecal microbiome. Int J mol Sci. 2019;20(6):1430. doi:10.3390/ijms20061430
79. Liu C, Makrinioti H, Saglani S, et al. Microbial dysbiosis and childhood asthma development: integrated role of the airway and gut microbiome, environmental exposures, and host metabolic and immune response. Front Immunol. 2022;13:1028209. doi:10.3389/fimmu.2022.1028209
80. Hill DA, Siracusa MC, Abt MC. Commensal bacteria-derived signals regulate basophil hematopoiesis and allergic inflammation. Nat Med. 2012;18(4):538–546. doi:10.1038/nm.2657
81. van Nimwegen FA, Penders J, Stobberingh EE. Mode and place of delivery, gastrointestinal microbiota, and their influence on asthma and atopy. J Allergy Clin Immunol. 2011;128(5):948–55.e1–3. doi:10.1016/j.jaci.2011.07.027
82. Wypych TP, Marsland BJ. Antibiotics as instigators of microbial dysbiosis: implications for asthma and allergy. Trends Immunol. 2018;39(9):697–711. doi:10.1016/j.it.2018.02.008
83. Khalkhali HR, Oshnouei S, Salarilak S, Rad MR, Karamyar M, Khashabi J. Effects of antibiotic consumption on children 2–8 years of age developing asthma. Epidemiol Health. 2014;36:e2014006. doi:10.4178/epih/e2014006
84. Scott NA, Andrusaite A, Andersen P. Antibiotics induce sustained dysregulation of intestinal T cell immunity by perturbing macrophage homeostasis. Sci Transl Med. 2018;10(464).
85. Murrison LB, Brandt EB, Myers JB, Hershey GKK. Environmental exposures and mechanisms in allergy and asthma development. J Clin Invest. 2019;129(4):1504–1515. doi:10.1172/JCI124612
86. Lee-Sarwar KA, Lasky-Su J, Kelly RS, Litonjua AA, Weiss ST. Gut microbial-derived metabolomics of asthma. Metabolites. 2020;10(3):97.
87. Lodge CJ, Tan DJ, Lau M. Breastfeeding and asthma and allergies: a systematic review and meta-analysis. Acta Paediatr. 2015;104(467):38–53. doi:10.1111/apa.13132
88. Dogaru CM, Nyffenegger D, Pescatore AM, Spycher BD, Kuehni CE. Breastfeeding and childhood asthma: systematic review and meta-analysis. Am J Epidemiol. 2014;179(10):1153–1167. doi:10.1093/aje/kwu072
89. Owora AH, Zhang Y. Childhood wheeze trajectory-specific risk factors: a systematic review and meta-analysis. Pediatr Allergy Immunol. 2021;32(1):34–50. doi:10.1111/pai.13313
90. Oddy WH. Breastfeeding, childhood asthma, and allergic disease. Ann Nutr Metab. 2017;70(Suppl 2):26–36. doi:10.1159/000457920
91. Guo J, Zhu W, Wang H, et al. Risk factors and prognosis of recurrent wheezing in Chinese young children: a prospective cohort study. Allergy Asthma Clin Immunol. 2019;15:38. doi:10.1186/s13223-019-0351-4
92. Donaldson GP, Ladinsky MS, Yu KB. Gut microbiota utilize immunoglobulin A for mucosal colonization. Science. 2018;360(6390):795–800. doi:10.1126/science.aaq0926
93. Torow N, Hornef MW. The timed pathway to homeostasis. Immunity. 2019;50(5):1127–1129. doi:10.1016/j.immuni.2019.04.012
94. Al Nabhani Z, Dulauroy S, Marques R, et al. A weaning reaction to microbiota is required for resistance to immunopathologies in the adult. Immunity. 2019;50(5):1276–88.e5. doi:10.1016/j.immuni.2019.02.014
95. Coutts A. Nutrition and the life cycle 2: infancy and weaning. Br J Nurs. 2000;9(21):2205–6,8,10passim. doi:10.12968/bjon.2000.9.21.5424
96. Greer FR, Sicherer SH, Burks A. The effects of early nutritional interventions on the development of atopic disease in infants and children: the role of maternal dietary restriction, breastfeeding, hydrolyzed formulas, and timing of introduction of allergenic complementary foods. Pediatrics. 2019;143(4):183–191.
97. Oddy WH. Breast feeding and respiratory morbidity in infancy: a birth cohort study. Arch Dis Child. 2003;88(3):224–228. doi:10.1136/adc.88.3.224
98. Miliku K, Azad MB. Breastfeeding and the developmental origins of asthma: current evidence, poss
Comments (0)