Hidden Link: How Gut Health Influences Asthma and COPD Treatment Outcomes

Introduction
Chronic obstructive pulmonary disease (COPD) remains one of the leading causes of morbidity and mortality worldwide, currently affecting more than 400 million people. It is characterized by progressive inflammation, structural lung destruction, and persistent airflow limitation that significantly impair quality of life and reduce life expectancy. Asthma, another highly prevalent chronic respiratory condition, affects hundreds of millions globally, with approximately 60% to 80% of patients exhibiting allergic phenotypes. These phenotypes are marked by airway hyperresponsiveness, episodic dyspnea, wheezing, and chest tightness. Traditionally, both COPD and asthma have been approached as primarily pulmonary diseases, with treatments focused almost exclusively on airway pathology. However, emerging evidence highlights the importance of systemic influences, particularly those originating from the gastrointestinal tract, in shaping respiratory disease progression and outcomes.
The concept of the gut-lung axis has gained increasing attention as a framework for understanding this relationship. This bidirectional communication system links the intestinal microbiota and the respiratory tract through immune, inflammatory, and metabolic pathways. Disruption of gut microbial balance, known as dysbiosis, has been implicated in the pathophysiology of COPD. Alterations in gut microbiota composition can influence systemic inflammation, immune regulation, and even local pulmonary responses. Similarly, in asthma, diminished production of short-chain fatty acids (SCFAs), such as butyrate, propionate, and acetate, due to impaired microbial diversity, has been shown to reduce immune tolerance, promote airway remodeling, and exacerbate allergic inflammation. These findings suggest that gut health may directly impact asthma severity and long-term outcomes.
Diet plays a central role in maintaining microbial diversity and function. Diets low in fiber and rich in processed foods have been associated with impaired microbiome composition, heightened systemic inflammation, and worsened asthma control. In contrast, adherence to dietary patterns such as the Mediterranean diet, which emphasizes fruits, vegetables, legumes, whole grains, fish, and healthy fats, has been shown to restore microbial balance, enhance SCFA production, and reduce airway inflammation. Such dietary interventions not only improve respiratory symptoms but also contribute to broader systemic health benefits, reinforcing the value of nutritional strategies in managing asthma and COPD.
Beyond asthma and COPD, growing research also explores the interplay between the gut microbiome and pulmonary diseases with high mortality rates, including lung cancer. In 2020, lung cancer accounted for nearly 18% of all cancer-related deaths worldwide. The recognition that both gut and pulmonary microbiomes can influence carcinogenesis, tumor progression, and response to therapy underscores the far-reaching implications of the gut-lung axis for comprehensive respiratory care.
This review aims to synthesize current evidence on the role of gastrointestinal microbiota in respiratory health and disease. By examining how dysbiosis influences disease occurrence, progression, and prognosis, it underscores the clinical importance of targeting the gut-lung axis. The analysis highlights not only mechanistic insights into immune and inflammatory pathways but also practical therapeutic implications, including microbiome-targeted interventions, dietary modifications, and emerging probiotic or prebiotic therapies. Understanding these complex interconnections may ultimately transform treatment approaches for asthma, COPD, and other respiratory diseases, moving toward more holistic and effective strategies that integrate both pulmonary and systemic health.
Understanding the Gut-Lung Axis in Chronic Respiratory Diseases
The complex interrelationship between intestinal health and respiratory function stems from intricate physiological connections that affect disease progression in both systems. Recent research reveals that pulmonary diseases like asthma and COPD involve not just isolated lung dysfunction but also depend on interactions with distant organ systems, particularly the gut. This emerging field examines how gastrointestinal microbiota influence respiratory function through a sophisticated network known as the gut-lung axis.
Shared embryonic origin of gut and lung tissues
The respiratory and digestive systems, despite their anatomical separation in adults, share a common developmental origin from the primitive gut tube during embryogenesis. Specifically, both systems arise from the endoderm of the foregut, with the respiratory system developing from an endodermal diverticulum in the ventral foregut wall, while the esophagus forms from its dorsal wall. In avian embryos, this separation process occurs rapidly, taking approximately 10 hours to form two independent endodermal structures.
The molecular underpinnings of this shared development involve precise genetic regulation. Homeobox-containing transcription factors (Hox genes) control gut regionalization, with Hoxa3 and Hoxb4 specifically expressed in the foregut endoderm. Additionally, other critical developmental regulators include:
- Sonic Hedgehog (SHH) – expressed throughout the endodermal layer
- Sox2 – expressed in foregut endoderm
- Sox9 – expressed in midgut/hindgut endoderm
- Nkx2.1 – specifically expressed in the developing trachea
These shared embryonic origins and developmental pathways partly explain why both systems exhibit similarities in mucosal structure and immunological responses to environmental exposures.
Bidirectional signaling via immune and metabolic pathways
The gut-lung axis functions through sophisticated bidirectional communication networks involving immune cells, metabolites, and neural signaling. This crosstalk occurs through multiple pathways, creating a functional link between these anatomically distinct systems.
Immune-mediated communication forms a central component of this axis. The gut microbiota significantly contributes to immune homeostasis by modulating the balance between pro-inflammatory Th17 and anti-inflammatory Treg cells. Moreover, these immune regulatory properties extend beyond the intestine to peripheral organs, including the lungs, through T cell migration. Researchers have observed that alterations in lung microbiota may reciprocally influence gut microbiota composition, underscoring the dynamic nature of this relationship.
Translocation of bacterial components represents another key mechanism in gut-lung communication. Studies have documented how impaired intestinal permeability allows gut microbes like Bacteroidetes and Enterobacteriaceae to migrate to the lungs of patients with acute respiratory distress syndrome. During this process, the mesenteric lymphatic system serves as an essential pathway through which bacterial fragments or metabolites (particularly short-chain fatty acids) can cross the intestinal barrier, reach systemic circulation, and modulate pulmonary immune responses.
Metabolic signaling provides yet another avenue for gut-lung interaction. Short-chain fatty acids (SCFAs), primarily produced through bacterial fermentation of dietary fibers, act as signaling molecules on antigen-presenting cells in the lungs, thereby attenuating inflammatory responses. These SCFAs can dampen allergic airway inflammation by promoting anti-inflammatory regulatory T cell differentiation. Furthermore, SCFAs influence immune cell development in bone marrow, representing another mechanism through which gut microbiota affect pulmonary immunity.
The bidirectional nature of this axis becomes apparent in cases of respiratory infection. For instance, influenza virus infection triggers increased proportions of Enterobacteriaceae while decreasing Lactobacilli and Lactococci in the gut. Similarly, lipopolysaccharide instillation in mouse lungs causes notable gut microbiota disturbances, demonstrating how lung conditions can affect intestinal microbial communities.
Neuroimmune interactions, particularly through the vagus nerve, constitute another pathway in gut-lung communication. This neural conduit allows intestinal stimuli to directly modulate pulmonary immune tone, creating yet another dimension to the gut-lung axis.
Microbiota Composition in Healthy vs Diseased States
Microbial communities within human gut and respiratory systems differ markedly between healthy individuals and those with chronic respiratory diseases. These differences offer critical insights into disease progression and potential therapeutic approaches for asthma and COPD management. Research into these distinct microbial signatures reveals how dysbiosis in one system might influence pathology in another through the gut-lung axis.
Dominant gut phyla: Firmicutes, Bacteroidetes, Actinobacteria
The human gut microbiota maintains a relatively consistent taxonomic structure despite individual variations. In healthy individuals, the gut harbors five major bacterial phyla with two dominant groups—Firmicutes and Bacteroidetes—together comprising approximately 90% of the total bacterial population. The remaining portion consists primarily of Actinobacteria, Proteobacteria, and Verrucomicrobia.
Looking at specific composition, the Firmicutes phylum contains over 200 different genera, including Lactobacillus, Bacillus, Clostridium, Enterococcus, and Ruminococcus, with Clostridium genera representing about 95% of this phylum. Conversely, Bacteroidetes primarily consists of Bacteroides and Prevotella genera. Though less abundant, Actinobacteria is mainly represented by the beneficial Bifidobacterium genus.
The ratio between Firmicutes and Bacteroidetes (F/B ratio) serves as an important health indicator. This ratio tends to increase with age, reaching its highest values in individuals aged 60-69 years. Research indicates this ratio shifts in various pathological conditions, including obesity, where higher Firmicutes proportions correlate with more efficient energy extraction from food.
Lung microbiota in COPD: Haemophilus, Moraxella, Streptococcus
Unlike gut microbiota, the respiratory microbiome shows greater spatial variation between individuals. In healthy lungs, four bacterial phyla—Firmicutes, Proteobacteria, Bacteroidetes, and Actinobacteria—constitute approximately 97% of all sequences. However, this balance shifts dramatically in COPD patients.
Studies consistently show increased colonization by pathogenic bacteria in COPD patients’ lungs. Particularly, Haemophilus influenzae, Moraxella catarrhalis, and Streptococcus pneumoniae dominate the airway microbiome in COPD, while Pseudomonas aeruginosa becomes prevalent in severe cases. Indeed, during COPD exacerbations, researchers have noted a microbial composition shift toward increased relative abundance of Proteobacteria (especially Moraxella) and decreased Firmicutes.
The dynamic nature of lung microbiota becomes evident when comparing stable COPD to acute exacerbations. In stable COPD, Firmicutes (31.63%) constitute the major phylum, followed by Bacteroidetes (28.94%) and Proteobacteria (19.68%). In contrast, during exacerbations, Proteobacteria (30.29%) become dominant, followed by Firmicutes (29.85%) and Bacteroidetes (14.02%). This bacterial shift correlates with clinical phenotypes, as patients with bacterial exacerbations show markedly decreased Firmicutes and increased Proteobacteria compared to those with eosinophilic exacerbations.
Reduced alpha-diversity in asthma and COPD patients
Alpha-diversity—a measure of microbial diversity within a single sample—appears altered in respiratory conditions. This metric encompasses both richness (number of different species) and evenness (distribution of these species). Multiple studies report trends toward lower alpha-diversity in disease cases compared to healthy controls in asthma, respiratory tract infections, influenza, respiratory allergies, and pneumonia.
For COPD specifically, the relationship between alpha-diversity and disease status shows some complexity. Studies reveal that the frequent exacerbator phenotype associates with lower sputum microbiome α-diversity (p=0.0031), a decrease that intensifies when sputum bacterial culture tests positive (p<0.001). Accordingly, older age also correlates with decreased sputum microbiome α-diversity (p=0.0030).
In bronchial asthma, decreased bacterial diversity correlates with disease severity. Certain bacteria appear associated with bronchial hyperactivity and obstruction, particularly Proteobacteria, Firmicutes (Streptococci), and Gammaproteobacteria, while respiratory commensals like Veillonella and Prevotella become less prevalent.
Hence, these microbial community disruptions not only serve as biomarkers for respiratory diseases but may actively participate in disease pathogenesis. The alteration in microbiota composition between healthy and diseased states provides valuable insights into the bidirectional communication along the gut-lung axis, offering potential avenues for novel therapeutic approaches targeting microbiota restoration.
How Gut Dysbiosis Alters COPD Progression
Recent investigations have uncovered compelling evidence that gut dysbiosis plays a critical role in COPD progression through multiple pathophysiological mechanisms. The interplay between intestinal microbiota alterations and respiratory pathology represents a vital factor in disease development beyond traditional lung-focused perspectives.
Increased gut permeability and systemic inflammation
Patients with COPD exhibit notable intestinal microbiome dysbiosis and increased intestinal permeability, often accompanied by inflammatory cell infiltration. This compromised intestinal barrier integrity manifests as enterocyte damage and intestinal hyperpermeability, indicating substantial functional alterations in the gastrointestinal tract.
Various factors contribute to this compromised gut integrity. Primarily, cigarette smoke exposure—a leading COPD risk factor—induces inflammatory responses that initially affect the lungs but subsequently “spill” into systemic circulation. This spillover effect triggers low-grade inflammatory responses throughout the intestine. Furthermore, the unmet metabolic oxygen demand (hypoxia) experienced by COPD patients during routine activities amplifies intestinal permeability and causes enterocyte damage.
Clinical evidence supports these observations. One study demonstrated that plasma zonulin, a marker of gut leakage, was elevated in COPD patients, with higher levels corresponding to more severe disease states. Additionally, research has documented that L-rhamnose/L-fructose urinary excretion ratios (another permeability indicator) increase substantially during acute COPD exacerbations compared to recovery periods.
Translocation of bacterial metabolites to lungs
Once intestinal barrier function becomes compromised, bacteria and their products can translocate from the gastrointestinal tract to extraintestinal sites, including mesenteric lymph nodes, bloodstream, and distant organs like the lungs. This bacterial translocation represents a key mechanism through which gut dysbiosis influences pulmonary pathology.
Exposure to cigarette smoke for 10 weeks (5 days weekly) has been shown to markedly increase bacterial translocation rates to mesenteric lymph nodes. Among the translocated products, lipopolysaccharide (LPS)—a major component of gram-negative bacteria—deserves particular attention as it increases gut barrier permeability. Interestingly, germ-free mice show relatively low LPS concentration in lungs, yet this reverses upon colonization with the gram-negative bacterium Bacteroides thetaiotaomicron, suggesting direct LPS translocation from gut to lungs.
Short-chain fatty acids (SCFAs) serve as critical mediators in gut-lung axis communication. Produced through bacterial fermentation of dietary fiber, SCFAs exhibit anti-inflammatory properties, help preserve colonic epithelial integrity, and regulate host energy balance. These gut-derived metabolites enter systemic circulation and subsequently:
- Prime myeloid cells in bone marrow that later migrate to lungs
- Activate peripheral immune cells that are recruited to lungs
- Regulate Treg cell differentiation through G protein-coupled receptors
Total SCFA levels appear much lower in patients with severe COPD (GOLD III-IV) than in healthy subjects, potentially contributing to diminished anti-inflammatory capacity.
Elevated Streptococcus parasanguinis_B in COPD patients
Multiple studies have identified specific bacterial shifts associated with COPD progression. Notably, researchers have observed increased abundance of several Streptococcus species, including S. parasanguinis_B and S. salivarius, in the intestines of COPD patients. This altered bacterial composition directly influences disease trajectory, as demonstrated through fecal microbiota transplantation experiments.
After receiving fecal transplants from COPD patients, mice exhibited elevated lung inflammation within 28 days. Furthermore, when mice underwent both fecal transplantation from COPD patients and biomass fuel smoke exposure for 20 weeks, they developed accelerated decline in lung function, severe emphysematous changes, airway remodeling, and mucus hypersecretion.
- parasanguinis_B appears particularly relevant as it possesses several virulence factors that may contribute to COPD pathogenesis. These include fibronectin/fibrinogen binding genes, collagen-binding surface proteins, and adhesin proteins that facilitate host cell adherence. Additionally, it contains oligopeptide-binding protein SarA-encoding genes that support colonization, potentially explaining its elevated presence in both intestinal and respiratory samples from COPD patients.
Through these interconnected mechanisms, gut dysbiosis emerges as a substantial contributor to COPD pathophysiology, offering novel targets for therapeutic intervention along the gut-lung axis.
Asthma and Gut Health: A Hidden Connection
Emerging research has established a compelling link between intestinal microbiota composition and asthma development, offering fresh perspectives on this chronic respiratory condition. This relationship extends beyond simple correlation, suggesting fundamental mechanisms through which gut health influences respiratory outcomes.
Does gut health affect asthma severity?
Current evidence strongly supports that gut microbiota composition affects asthma severity through multiple pathways. Children with asthma exhibit markedly different gut bacterial profiles compared to healthy counterparts. Specifically, genera like Dialister, Faecalibacterium, and Roseburia (all belonging to Firmicutes phylum) appear substantially reduced in asthmatic children. These alterations may increase susceptibility to viral infections and contribute to asthma development.
Furthermore, intestinal metabolites produced by certain bacteria play crucial roles in asthma pathophysiology. Studies reveal that:
- Butyrate levels in stool are decreased in children with asthma and negatively correlate with serum IgE levels
- Reduced butyrate production compromises intestinal epithelial barrier function, potentially promoting allergen uptake in allergic asthma
- Microbial diversity at age 1 year strongly predicts asthma development by age 5, with distinct microbial signatures in children who later develop asthma
Alongside diversity changes, relative abundance of specific bacterial families demonstrates clinical relevance. For instance, the relative abundance of Veillonellaceae positively correlates with sputum eosinophilia percentage in non-severe asthma patients (r=0.579, P=0.030), connecting gut microbiota directly to inflammatory biomarkers.
Role of early-life microbial colonization
The first hundred days of life represent a critical timeframe during which the immune system exhibits greatest plasticity. Throughout this period, proper microbial colonization establishes immunological tolerance, whereas dysbiosis might predispose children to asthma.
Hypopharyngeal colonization status in neonates serves as a predictive marker for asthma development. Colonization with specific bacteria—S. pneumoniae, H. influenzae, M. catarrhalis, or combinations thereof—in asymptomatic neonates at 1 month increases wheeze risk by factors of two to four. More precisely, hazard ratios reveal escalating risks: 1.65 for first wheezy episode, 2.40 for persistent wheeze development, 2.99 for acute severe exacerbations, and 3.85 for hospitalization due to wheeze.
By age 5, asthma prevalence reached 33% in colonized children versus only 10% in non-colonized children (odds ratio, 4.57). Therefore, the population attributable risk of asthma associated with colonization was calculated at 4.6%.
Impact of antibiotics on asthma risk
Antibiotic administration, particularly in early life, consistently demonstrates association with increased asthma risk. A recent large-scale study involving over 1 million children found that antibiotic exposure before age 2 was associated with higher asthma risk (adjusted hazard ratio 1.24). Notably, this relationship demonstrates clear dose-dependency—receiving 5 or more courses versus 1-2 courses was linked to substantially higher asthma risk (aHR 1.52).
The mechanisms underlying this relationship involve disruption of the developing microbiome. Antibiotics provoke delays in microbiota maturation and deplete beneficial bacteria such as Lachnospiraceae—producers of short-chain fatty acids crucial for immune maturation. Consequently, epidemiological studies have confirmed associations between antibiotic consumption in the first year and asthma development at school age.
Recent research indicates that even maternal antibiotic use during pregnancy affects offspring risk. Children whose mothers used antibiotics prenatally or shortly thereafter showed increased childhood asthma risk. Even more concerning, this association persisted when examining siblings with different antibiotic exposures, suggesting a causal rather than merely correlative relationship.
Immune Modulation Through the Gut-Lung Axis
The molecular communication between gut and lungs orchestrates intricate immunological responses that affect respiratory disease outcomes. This bidirectional exchange involves specialized metabolites, immune cell programming, and inflammatory pathways that collectively shape pulmonary pathology in conditions like asthma and COPD.
SCFAs and Treg cell differentiation
Short-chain fatty acids (SCFAs)—primarily acetate, propionate, and butyrate—act as pivotal mediators in the gut-lung axis, regulating immune responses through multiple mechanisms. These bacterial fermentation products exhibit profound effects on T cell differentiation, promoting the development of regulatory T (Treg) cells essential for immune tolerance. To clarify, propionate and butyrate stimulate Treg cell differentiation from CD4+ T cells by upregulating Foxp3 gene transcription through histone acetylation. This process occurs independently of G protein-coupled receptors (GPCRs) like GPR41 and GPR43, instead relying on histone deacetylase (HDAC) inhibition.
Beyond Treg induction, SCFAs demonstrate versatile immunomodulatory capabilities:
- Enhance differentiation of monocytes into anti-inflammatory macrophages, providing protection against viral infections
- Activate the mTOR-S6K pathway necessary for T cell differentiation and cytokine expression
- Induce IL-10 expression in leukocytes, thereby suppressing inflammatory responses
Experimental evidence confirms that butyrate blocks NF-κB activation through its anti-inflammatory properties, ultimately elevating IL-10 levels while inhibiting pro-inflammatory cytokines like IL-12 and IFN-γ in dendritic cells. Furthermore, high-fiber diets increase circulating SCFA levels, protecting lungs from allergic inflammation, whereas low SCFA levels correlate with increased allergic airway disease.
NLRP3 inflammasome activation in COPD
The NLRP3 inflammasome represents a specialized inflammatory signaling platform that governs the maturation and secretion of IL-1-like cytokines through caspase-1-dependent proteolytic processing. In essence, this multiprotein complex consists of three components: the sensor NLRP3 protein, adaptor ASC (apoptosis-associated speck-like protein), and effector pro-caspase-1.
NLRP3 inflammasome activation typically follows a two-step process:
- Priming: Recognition of pathogen-associated molecular patterns by Toll-like receptors triggers NF-κB signaling, increasing transcription of NLRP3 protein, pro-IL-1β, and pro-IL-18
- Assembly: The inflammasome components assemble, with ASC binding NLRP3 to pro-caspase-1, leading to caspase-1 activation and subsequent processing of pro-IL-1β and pro-IL-18 into mature forms
Clinical studies reveal that NLRP3 expression is substantially higher in COPD patients compared to those without COPD (p=0.018). Interestingly, smoking appears to trigger NLRP3-mediated inflammation in stable COPD patients, with more pronounced increases observed in active smoking COPD groups versus non-smoking controls. In vitro and in vivo research demonstrates that NLRP3 knockdown in COPD models reduces IL-18, IL-1β, neutrophil, macrophage, and lymphocyte levels, alongside diminishing lung inflammation.
Cytokine signaling and airway inflammation
Cytokines serve as crucial immunomodulatory proteins in allergic airway inflammation, orchestrating complex interactions between innate and adaptive immune cells. While T-helper 2 (Th2) cells were traditionally considered the main drivers of allergic airway inflammation, recent studies highlight contributions from other helper T cells and their associated cytokines.
Several specialized T cell subsets play important roles:
- Th17 cells contribute to allergic rhinitis, asthma, and chronic rhinosinusitis with nasal polyps
- Th9 cells, producing IL-9, influence allergic reactions
- Th22 cells, secreting IL-22, IL-13, and TNF-α, affect airway inflammation
- Th25 cells, via IL-25 production, help initiate allergic responses
Epithelial cell-derived cytokines—including thymic stromal lymphopoietin, IL-33, IL-31, and IL-25—prove critical for initiating allergic reactions and inducing airway inflammation. Throughout the gut-lung axis, these inflammatory mediators, along with microbial metabolites like lipopolysaccharide, can travel via blood and lymphatic systems from intestines to lungs, impacting respiratory health. For instance, gut-derived LPS can reach lungs and cause acute lung injury through TLR4/NF-κB activation in alveolar macrophages.
Overall, the gut-lung axis provides a framework for understanding how intestinal health influences respiratory outcomes through intricate immune pathways, offering potential therapeutic targets for addressing both asthma and COPD through dietary and microbiome-based interventions.
Nutritional Interventions for Microbiota Restoration
Dietary interventions offer promising approaches for manipulating the gut-lung axis to improve outcomes in chronic respiratory diseases. As diet shapes microbial communities throughout the body, carefully selected nutritional strategies can restore microbial balance and potentially alleviate respiratory symptoms.
High-fiber diets and SCFA production
Fiber-rich diets modify both lung and gut microbiota composition by altering the ratio of Firmicutes to Bacteroidetes, simultaneously providing protection against allergic inflammation through increased circulating short-chain fatty acids (SCFAs). These microbial metabolites—primarily propionate, acetate, and butyrate—result from bacterial fermentation of undigested soluble dietary fibers and serve as critical immunomodulatory compounds. SCFAs exert their anti-inflammatory effects by activating G protein-coupled receptors (GPR41, GPR43, GPR109A) and inhibiting histone deacetylases.
At a cellular level, butyrate promotes intestinal barrier integrity through GPR109A-mediated NLRP3 inflammasome activation in intestinal epithelial cells. Furthermore, butyrate signals through GPR109A on colonic macrophages and dendritic cells to induce IL-10 production, an anti-inflammatory cytokine essential for immune homeostasis.
Clinical evidence supports high-fiber dietary interventions for respiratory conditions. A UK cohort study of 2,942 adults found that a “prudent” dietary pattern rich in fruits, vegetables, oily fish, and wholemeal cereals correlated with protection against impaired lung function and COPD, with particular benefits observed in male smokers.
Vitamin D and folate in immune regulation
Beyond macronutrients, micronutrients play vital roles in maintaining respiratory health. Vitamin D receptors appear throughout the immune system, making this nutrient crucial for regulating both innate and adaptive immunity. Vitamin D enhances neutrophil killing capacity in patients with bacterial respiratory infections while simultaneously lowering pro-inflammatory cytokine production.
Through its immunomodulatory functions, vitamin D increases antiviral responses of respiratory epithelial cells during infections. Additionally, vitamin D induces expression of antimicrobial peptide genes including cathelicidins and defensins, which disrupt pathogen integrity and cause apoptosis—actions that partly explain vitamin D’s “antibiotic” properties.
Western diet vs Mediterranean diet effects
The increasing prevalence of Western diets—characterized by high intake of saturated fats, refined sugars, and processed foods with low fiber content—coincides with rising rates of inflammatory and autoimmune diseases, including asthma. In contrast, the Mediterranean diet (MD) delivers complex carbohydrates, polyunsaturated fatty acids with anti-inflammatory properties, and bioactive compounds with antioxidant capabilities.
Remarkably, adherence to Mediterranean dietary patterns correlates with microbiota eubiosis reestablishment, as evidenced by increased Bacteroidetes and beneficial Clostridium groups alongside decreased Proteobacteria and Bacillaceae phyla. This dietary approach provides a balanced array of micronutrients including vitamins and minerals that help prevent malnutrition and immunodeficiencies.
Current scientific literature consistently highlights the superiority of plant-based and Mediterranean diets over Western diets in promoting gut health and preventing non-communicable diseases. Specifically, Mediterranean diets provide polyunsaturated fatty acids that, like SCFAs, exhibit anti-inflammatory and cardioprotective properties.
Probiotics and Fecal Transplantation in COPD and Asthma
Beyond conventional therapies, microbiome-based interventions offer promising approaches for respiratory conditions. Mounting evidence indicates that manipulating the gut microbiome through probiotics and fecal transplantation may yield therapeutic benefits for both asthma and COPD, underscoring the clinical relevance of the gut-lung axis.
Lactobacillus rhamnosus in murine COPD models
Research on Lactobacillus rhamnosus (Lr) in cigarette smoke-induced COPD mouse models reveals remarkable anti-inflammatory effects. Lr treatment administered before COPD induction and maintained throughout the study attenuated airway and lung parenchymal inflammation by reducing inflammatory cell infiltration and pro-inflammatory cytokines. Mice receiving Lr showed decreased levels of IL-1β, IL-6, TNF-α, KC, IL-17, and TGF-β in bronchoalveolar lavage fluid compared to untreated COPD mice.
At the tissue level, Lr administration:
- Reduced peribronchial inflammation and alveolar enlargement
- Decreased collagen deposition and elastic fiber destruction
- Maintained expression of MMP-9 and MMP-12 genes at levels comparable to control animals
These effects occurred alongside downregulation of TLR2, TLR4, TLR9, STAT3, and NF-κB expression in lung tissue, while increasing anti-inflammatory IL-10 levels and TIMP1/2 expression. Similar results were observed in human bronchial epithelial cells exposed to cigarette smoke extract, with Lr treatment inhibiting pro-inflammatory cytokine production.
Fecal microbiota transplantation: current evidence
Fecal microbiota transplantation (FMT) represents another emerging intervention for respiratory conditions. Initially recommended for recurrent Clostridium difficile infections, FMT has expanded into treating respiratory diseases. In mouse models, FMT alleviated hallmark features of COPD including inflammation, alveolar destruction, and impaired lung function. Furthermore, these protective effects were additive to smoking cessation.
For asthma specifically, research demonstrates that FMT improved airway inflammation in OVA-induced rat models and corrected intestinal short-chain fatty acid disorders. Rats receiving FMT showed remarkably improved lung function, decreased inflammatory cell content in peripheral blood and BALF, improved lung tissue pathology, and reduced collagen fiber deposition by a standardized mean difference of -2.25.
Challenges in clinical translation
Nevertheless, translating these promising preclinical results to human applications presents substantial hurdles. Currently, efforts to directly manipulate the lung microbiome without altering gut microbiota remain limited. This limitation stems from inadequate understanding of microbiome perturbation in lungs following antibiotic therapy and related pathologies.
Additional challenges include determining optimal bacterial strains, as the efficacy of different probiotics varies considerably. While recent meta-analysis showed probiotics improved asthma control test scores with an odds ratio of 1.18, researchers acknowledge insufficient evidence regarding specific strains. Consequently, as investigations progress, advanced techniques for sampling and manipulating lung-specific microbiomes will be essential for developing targeted intervention strategies.
Environmental and Lifestyle Factors Affecting the Gut-Lung Axis
Environmental factors fundamentally shape both gut and respiratory microbiomes, creating a complex interplay that influences chronic lung disease progression. Lifestyle choices and medical treatments often affect this delicate balance, potentially exacerbating respiratory conditions through alterations in the gut-lung axis.
Cigarette smoke and microbial diversity
Cigarette smoking stands as the primary risk factor for COPD development, with approximately 80% of COPD patients being current or former smokers. Beyond its direct pulmonary effects, smoking profoundly alters microbial communities throughout the body. In the gut, smoking reduces bacterial diversity while shifting composition toward increased Bacteroidetes and decreased Firmicutes phyla. Upon smoking cessation, this pattern gradually reverses, with Firmicutes and Actinobacteria increasing while Proteobacteria and Bacteroidetes decrease. Interestingly, chronic smoke exposure in mice creates similar dysbiosis patterns as observed in stage 4 COPD patients.
Malnutrition and SCFA depletion
Approximately 20% of COPD patients experience weight loss and protein-calorie malnutrition, negatively impacting mortality outcomes. Throughout aging, malnutrition becomes increasingly common, creating vulnerability to respiratory infections. This nutritional insufficiency extends to microbiota health, as inadequate fermentable fiber intake leads to decreased production of short-chain fatty acids (SCFAs). Given that SCFAs reduce inflammatory responses while enhancing pathogen defense mechanisms, their depletion through malnutrition amplifies pulmonary immune responses through the gut-liver-lung axis.
Steroid and antibiotic use in chronic lung disease
Although essential for managing respiratory exacerbations, medications themselves affect microbiota composition. Corticosteroid treatment generally increases respiratory microbiome diversity, while antibiotics initially decrease sputum bacterial diversity before it returns to normal approximately 14 days post-treatment. Strikingly, gut bacteria abundance increases throughout this period without affecting diversity. Animal studies demonstrate that antibiotic administration worsens smoking-induced emphysema by depleting beneficial SCFA-producing bacteria like Lactobacillus, Akkermansia, and Ruminococcus.
Conclusion 
The emerging understanding of the gut-lung axis reveals a fascinating bidirectional relationship between intestinal and respiratory health. Evidence throughout this review demonstrates how gut dysbiosis contributes substantially to both asthma and COPD pathogenesis through multiple interconnected mechanisms. These mechanisms include increased intestinal permeability, bacterial translocation, altered metabolite production, and systemic inflammation that ultimately affect pulmonary function and disease progression.
Microbial communities serve as central players in this relationship. Healthy individuals typically maintain balanced gut microbiota dominated by Firmicutes and Bacteroidetes, whereas COPD patients show distinctive shifts toward pathogenic bacteria such as Haemophilus, Moraxella, and Streptococcus in their airways. Furthermore, reduced alpha-diversity consistently appears in both asthmatic and COPD patients, highlighting dysbiosis as a key feature of respiratory disease states.
Dietary patterns undoubtedly shape these microbial communities. High-fiber diets promote beneficial bacteria that produce short-chain fatty acids, which subsequently modulate immune responses through Treg cell differentiation and anti-inflammatory pathways. The Mediterranean diet, rich in polyphenols and polyunsaturated fatty acids, contrasts sharply with Western dietary patterns that often exacerbate inflammatory responses through altered gut microbiota composition.
Early-life microbial colonization deserves special attention because it establishes immunological tolerance patterns that persist throughout life. Antibiotic exposure during critical developmental windows disrupts this delicate balance, potentially predisposing individuals to asthma later in life. This relationship appears dose-dependent, with multiple antibiotic courses carrying greater risk than limited exposure.
Therapeutic approaches targeting the gut-lung axis present promising avenues for clinical intervention. Probiotics such as Lactobacillus rhamnosus have demonstrated anti-inflammatory effects in COPD models, while fecal microbiota transplantation shows potential for restoring beneficial microbial communities. Though challenges remain in clinical translation, these microbiome-based interventions may eventually complement traditional respiratory therapies.
Environmental factors also profoundly affect the gut-lung axis. Cigarette smoke alters microbial diversity throughout the body, while malnutrition depletes essential microbial metabolites necessary for immune regulation. Even medications used to treat respiratory diseases—particularly antibiotics and corticosteroids—modify microbiota composition, sometimes with counterproductive effects.
The intricate connections between gut health and respiratory outcomes outlined in this review emphasize the need for a more holistic approach to treating chronic lung diseases. Rather than focusing exclusively on the lungs, clinicians might achieve better outcomes by addressing intestinal health through dietary modifications, careful antibiotic stewardship, and potentially microbiome-targeted therapies. After all, respiratory health may begin not in the lungs but in the gut—a paradigm shift that could transform how practitioners approach asthma and COPD management.
Key Takeaways
The gut-lung axis reveals how intestinal health directly impacts respiratory disease outcomes, offering new therapeutic pathways for asthma and COPD management.
- Gut dysbiosis drives respiratory inflammation: Compromised intestinal barrier allows bacterial metabolites to reach lungs, triggering systemic inflammation that worsens COPD and asthma symptoms.
- Early antibiotic exposure increases asthma risk: Children receiving antibiotics before age 2 show 24% higher asthma risk, with dose-dependent effects reaching 52% for multiple courses.
- High-fiber diets protect against respiratory disease: Fiber-rich foods boost beneficial bacteria that produce anti-inflammatory compounds, reducing allergic airway inflammation and improving lung function.
- Specific bacterial signatures predict disease severity: COPD patients show increased Haemophilus, Moraxella, and Streptococcus in airways, while reduced microbial diversity correlates with worse outcomes.
- Probiotics offer therapeutic potential: Lactobacillus rhamnosus reduces lung inflammation in COPD models, while fecal transplantation shows promise for restoring beneficial microbial balance.
Understanding this gut-lung connection empowers clinicians to adopt holistic treatment approaches that address intestinal health alongside traditional respiratory therapies, potentially improving patient outcomes through dietary interventions and careful antibiotic stewardship.
Frequently Asked Questions:
FAQs
Q1. How does gut health influence respiratory conditions like asthma and COPD? Gut health impacts respiratory conditions through the gut-lung axis. Gut dysbiosis can lead to increased intestinal permeability, allowing bacterial metabolites to reach the lungs and trigger systemic inflammation. This inflammation can worsen symptoms of asthma and COPD. Additionally, the gut microbiome plays a role in immune regulation, which affects respiratory health.
Q2. Can dietary changes improve outcomes for patients with asthma or COPD? Yes, dietary changes can potentially improve outcomes for patients with asthma or COPD. High-fiber diets promote beneficial gut bacteria that produce anti-inflammatory compounds like short-chain fatty acids. These compounds can help reduce airway inflammation and improve lung function. The Mediterranean diet, rich in polyphenols and polyunsaturated fatty acids, has shown particular promise in supporting respiratory health.
Q3. Are probiotics effective in treating respiratory conditions? Probiotics show promise in treating respiratory conditions, though more research is needed. Studies on Lactobacillus rhamnosus in COPD mouse models have demonstrated anti-inflammatory effects and reduced airway inflammation. However, the effectiveness of probiotics can vary depending on the specific strains used, and more clinical trials are needed to establish their efficacy in humans.
Q4. How do early-life factors affect the risk of developing asthma? Early-life factors significantly influence asthma risk. Antibiotic exposure in the first two years of life has been associated with a 24% higher risk of asthma, with multiple courses increasing the risk further. Additionally, the initial colonization of the gut microbiome in infancy plays a crucial role in developing immune tolerance, which can affect asthma susceptibility later in life.
Q5. What is the connection between cigarette smoking and gut microbiota in COPD? Cigarette smoking, the primary risk factor for COPD, significantly alters gut microbiota composition. It reduces bacterial diversity and shifts the balance of bacterial phyla, typically increasing Bacteroidetes and decreasing Firmicutes. These changes in gut microbiota can contribute to systemic inflammation and exacerbate COPD symptoms. Interestingly, smoking cessation can gradually reverse some of these microbial changes.
References:
[1] – https://www.news-medical.net/news/20240920/How-plant-based-Mediterranean-and-Western-diets-affect-gut-microbiota-and-disease.aspx
[2] – https://www.frontiersin.org/journals/microbiology/articles/
10.3389/fmicb.2023.1219942/full
[3] – https://pubmed.ncbi.nlm.nih.gov/25479317/
[4] – https://www.nejm.org/doi/full/10.1056/NEJMoa052632
[5] – https://www.cidrap.umn.edu/antimicrobial-stewardship/large-study-ties-early-life-antibiotic-exposure-higher-risk-asthma
[6] – https://www.nature.com/articles/mi201444
[7] – https://pmc.ncbi.nlm.nih.gov/articles/PMC11703846/
[8] – https://pmc.ncbi.nlm.nih.gov/articles/PMC8159117/
[9] – https://gut.bmj.com/content/73/5/751
[10] – https://www.sciencedirect.com/science/article/pii/S1043276024003291
[11] – https://www.tandfonline.com/doi/full/10.1080/19490976.2024.2414805
[12] – https://www.tandfonline.com/doi/full/10.1080/15412555.2021.2019208
[13] – https://pmc.ncbi.nlm.nih.gov/articles/PMC10580882/
[14] – https://pmc.ncbi.nlm.nih.gov/articles/PMC10221978/
[15] – https://pmc.ncbi.nlm.nih.gov/articles/PMC9012580/
[16] – https://journals.asm.org/doi/10.1128/genomea.00541-15
[17] – https://www.nature.com/articles/s41467-017-02573-2
[18] – https://jtd.amegroups.org/article/view/54245/html
[19] – https://pmc.ncbi.nlm.nih.gov/articles/PMC9921812/
[20] – https://www.frontiersin.org/journals/medicine/articles/10.3389/
fmed.2021.662262/full
[21] – https://sph.rutgers.edu/news/frequent-use-antibiotics-infants-and-young-children-may-increase-risk-asthma-allergies-and
[22] – https://pmc.ncbi.nlm.nih.gov/articles/PMC10993281/
[23] – https://biosignaling.biomedcentral.com/articles/10.1186/s12964-023-01219-9
[24] – https://pmc.ncbi.nlm.nih.gov/articles/PMC5669850/
[25] – https://www.nature.com/articles/s41598-022-11164-1
[26] – https://bmcpulmmed.biomedcentral.com/articles/10.1186/s12890-024-03435-6
[27] – https://gutpathogens.biomedcentral.com/articles/10.1186/s13099-025-00718-z
[28] – https://www.frontiersin.org/journals/nutrition/articles/10.3389/fnut.2021.652410/full
[29] – https://pmc.ncbi.nlm.nih.gov/articles/PMC7927055/
[30] – https://pmc.ncbi.nlm.nih.gov/articles/PMC7182195/
[31] – https://pubmed.ncbi.nlm.nih.gov/39775370/
[32] – https://www.frontiersin.org/journals/cellular-and-infection-microbiology/articles/10.3389/fcimb.2024.1411222/full
[33] – https://onlinelibrary.wiley.com/doi/10.1111/resp.13759
[34] – https://www.physiciansweekly.com/post/understanding-how-probiotics-help-patients-with-asthma-2
[35] – https://pmc.ncbi.nlm.nih.gov/articles/PMC8245763/
[36] – https://portlandpress.com/clinsci/article/136/18/1371/231866/Smoking-induced-microbial-dysbiosis-in-health-and
[37] – https://pmc.ncbi.nlm.nih.gov/articles/PMC11088139/
[38] – https://www.sciencedirect.com/science/article/pii/S1568163720303706
[39] – https://pmc.ncbi.nlm.nih.gov/articles/PMC7988087/
[40] – https://www.nature.com/articles/s12276-023-01099-6