Skip to main content

MINI REVIEW article

Front. Immunol., 21 November 2023
Sec. Nutritional Immunology

Alveolar macrophage modulation via the gut–lung axis in lung diseases

  • Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China

Several studies have demonstrated great potential implications for the gut–lung axis in lung disease etiology and treatment. The gut environment can be influenced by diet, metabolites, microbiotal composition, primary diseases, and medical interventions. These changes modulate the functions of alveolar macrophages (AMs) to shape the pulmonary immune response, which greatly impacts lung health. The immune modulation of AMs is implicated in the pathogenesis of various lung diseases. However, the mechanism of the gut–lung axis in lung diseases has not yet been determined. This mini-review aimed to shed light on the critical nature of communication between the gut and AMs during the development of pulmonary infection, injury, allergy, and malignancy. A better understanding of their crosstalk may provide new insights into future therapeutic strategies targeting the gut–AM interaction.

1 Introduction

The gut–lung axis is a highly complex pathway. During the occurrence and development of lung diseases, a normal gut environment or dietary patterns helps build the necessary pulmonary immunity, and metabolites are directly or indirectly involved in immune modulation. Conversely, gut dysbiosis and inappropriate treatment usually induce aggravation (13). However, the cellular mechanism behind the gut–lung axis is unclear. It is well-known that alveolar macrophages (AMs), being highly specialized tissue-resident macrophages, are the key players in innate immunity and maintaining lung homeostasis; moreover, their functions change profoundly in various lung diseases. The origin, phenotype, and function of AMs and their role in phagocytosis and immune response have recently been elucidated (4, 5). This mini-review aimed to reveal the role of AMs in gut-lung interactions. The structural, metabolic, and cellular changes in this “target cell” mediated by the gut may help further understand the pathophysiology of lung diseases, derive new insight to improve the efficacy of some current treatments, and develop novel therapies based on the gut–lung axis concept.

2 AMs and lung diseases: modulation based on gut–lung axis mechanisms

2.1 Pulmonary infection

In the past few years, extensive studies have examined the role of the gut–lung axis in bacterial and viral infections. We hereby review how gut–lung axis-mediated AM modulation shapes the host immune defense against pulmonary infections as well as the crosstalk between this pathway and current therapies.

2.1.1 Bacterial pneumonia

Pneumonia causes more deaths than any other infectious diseases worldwide, especially since the coronavirus disease 2019 (COVID-19) pandemic (6). Streptococcus pneumoniae is the most common cause of community-acquired pneumonia (7). The gut microbiota (GM) contains pathogen-associated molecular patterns, as regulators of innate immunity, it modulates the metabolic pathways of AMs, and its depletion would weaken AM responsiveness toward bacterial virulence factors (8). Acetate, a metabolite produced by GM, enhances the phagocytic activity of AMs against S. pneumoniae in a nitric oxide (NO)-dependent manner (9). Even in the face of the more lethal K. pneumoniae pneumonia, short-chain fatty acids (SCFAs) can promote AMs to eliminate K. pneumoniae through LAMtor2-dependent signaling pathways (10). It is important to note that, although K. pneumoniae infection causes profound changes in GM, supplementation with multiple SCFAs can salvage impaired AM function and improve outcomes (11). In addition, for pneumonia caused by Streptococcus suis and melioidosis, GM has a positive role in promoting AM-mediated immune responses (12, 13). Overall, necessary GM and its metabolites affect lung subjects to infection, showing protective effects by arming AMs. Moreover, colonization of S. pneumoniae in human nasopharynx has a positive effect in enhancing AMs responsiveness to bacterial pathogens. This not only involves aspirated S. pneumoniae as an immune stimulus, but may also involve indirect stimulation from systemic reactions, the so-called gut-lung axis (14).

Although the discovery of antituberculosis therapy (ATT) that kills Mycobacterium tuberculosis (Mtb) brings hope to patients with tuberculosis, an increasing susceptibility of individuals who received medication toward reinfection or reactivation of Mtb raises doubts about whether widely used ATT has an off-target effect (15). A recent study reported that prior to Mtb infection, the use of isoniazid and pyrazinamide, rather than rifampicin, has a unique impact on GM, which in turn prevents the development of pulmonary immunity against Mtb (16). This is probably because of the impaired metabolism of AMs; an internal cellular environment that allows Mtb to grow is formed (17). Moreover, vaccination is an effective way to stop tuberculosis from spreading and the important role of AMs has been revealed, but the mechanism remains unclearly understood (18). A study has revealed a novel method by which subcutaneous Bacille Calmette–Guérin vaccination triggers immune alerts and forms immune memory of AMs in distal lung tissues, surprisingly this process is driven by changes in GM, and the induction of memory AMs is independent of circulating monocytes (19). This finding is contrary to the widely held view that the generation of immune memory is divided according to pathogen exposure site (20). However, this GM-mediated pathway will enhance our understanding of host defense mechanisms in distal organs. Notably, most of the current human vaccines are inoculated through the skin, including the COVID-19 vaccine (21).

Pseudomonas aeruginosa pneumonia is severe and fatal infection in hospitalized patients, especially those in intensive care units. Their prognosis depends on the use of antibiotics (Abx) as well as the pathogen’s virulence and the host’s immune response (22). P. aeruginosa ventilator-associated pneumonia mainly occurs in critically ill patients receiving overtime mechanical ventilation (MV) (23). Prolonged Abx use is usually necessary to reduce the recurrence rate; on the other hand, Abx treatment reduces the expression of Reg3β and TLR4 in the gut, which reduces the phagocytic activity of AMs against P. aeruginosa (24, 25). It should be noted that MV itself also reduces the pulmonary defense by increasing reactive oxygen species production of AMs; thus, the early intravascular injection of SCFAs, a derivative of GM, based on the gut–lung axis may help offset the negative effects (26). Unnecessary, prolonged, or broad-spectrum Abx exposure will induce intestinal barrier dysfunction, thereby weakening the phagocytosis of AMs. The oral supplementation of dead bacteria is conducive to reversing the compromised lung defense (27). Although probiotics may be more effective at activating the phagocytic capacity of AMs compared to dead bacteria, Abx treatment may inevitably affect the role of probiotics, and further research is needed to verify their relationship.

Pulmonary infections are more deadly for some high-risk people. Experiencing major surgery with severe tissue trauma tends to be immunosuppressive, especially for critically ill patients. This adverse effect can be improved by dexmedetomidine, which regulates GM to rescue the impaired phagocytic activity of AMs and reduce M2 polarization of macrophages (28). Moreover, influenza can induce GM disturbance, thereby reducing SCFA production and weakening the bactericidal activity of AMs, which is more likely to cause concurrent bacterial pneumonia. Such a superinfection could further lead to excessive mortality of influenza in these patients (29). Pulmonary infections are always fatal for patients with advanced HIV infection and concomitant GM and macrophage dysfunction. The alveolar mucosa, an important HIV pool, was a previously ignored mediator for continuous pneumonia (30, 31). However, the role of AMs in HIV–pneumonia patients remains unclear. Despite heterogeneity, many concurrent local or systemic diseases tend to impair lung defense function by affecting the gut environment, and an increased understanding of the gut–AM axis may have broader implications.

Neonatal pneumonia is the most common cause of infection leading to infant death. Although perinatal Abx use is crucial, it may increase the likelihood of life-threatening pneumonia later in infancy (32, 33). This is probably due to AM dysfunction caused by destruction of the GM balance. Interestingly, despite later fecal microbiota transplantation, some of the immune functions associated with chemotaxis and tissue repair could not be restored (34). In early life, to prepare for initial environmental exposure, macrophage precursors occupy the alveolar niche and become tissue-resident AMs (TRAMs) (35). Therefore, we must verify whether GM dysregulation impacts the developmental trajectory of AMs. Moreover, perinatal feeding practices and the delivery mode also greatly influence the neonatal GM; further research is needed to describe their role in AM origin (36, 37).

Previous studies have shown that the enhanced bacterial killing activity of AMs is mediated by Toll-like receptor (TLR) signals triggered by GM. When GM becomes depleted, the oral administration of lipopolysaccharide can help restore the immune function of AMs (38, 39). Interestingly, another study indicated that it was not the TLR, but Nod-like receptor (NLR) ligands, could rescue early bacterial clearance activity in lung (40). The reason of above contradict results is unknown, it may be because compared to NLR, TLR activation is more tightly restrained, so the effect of triggering the immune response to infection in the early stage is not as good as NLR (41). However, TLR and NLR pathways in the gut is complex, more experiments are needed to elucidate their role, understanding their mechanism facilitates more precise treatment.

2.1.2 Viral pneumonia

Previous studies have provided evidence of the immunomodulatory potential of AMs in viral infections (42). AM origin is a determinant in shaping extended function against pulmonary viral infections and affecting infection severity (43). In addition to necessary antiviral responses, tissue damage during infection (such as influenza and COVID-19) caused by exaggerated immune inflammation greatly contributes to the high mortality rates (44). Surprisingly, by inducing changes in bone marrow hematopoietic function, a high-fiber diet (HFD) prevents severe influenza infection by reducing tissue damage and enhancing adaptive antiviral immunity while significantly decreasing AM counts (45). The intermediate role of the bone marrow means that the gut–lung axis pathway is highly collaborative, so it is probably feasible to retain the necessary antiinfection ability and achieve the anti-inflammatory ability of AMs. Moreover, although with no research on the role of GM in viral pneumonia, considering its previous description in modulating bacterial clearance, it stands to reason that the GM also has a function here, the altered hematopoietic function is likely not the sole source to prevent tissue damage. The supplementation of Clostridium butyricum can prevent excessive inflammation during RSV infection, probably because of the M2 polarization of AMs. Oral administration of Lactobacillus rhamnosus CRL 1505 activates AMs and enhances their production of type I interferon and interferon-γ as well as their ability to combat RSV. The enhanced antiviral response of AMs may be attributed to the increase of circulating SCFAs (4648). These findings suggested that probiotics may be promising for preventing and treating viral infections; however, additional clinical studies are needed to further delineate their effects.

2.2 Inflammatory injury

AMs are the most abundant innate immune cell during acute lung injury (ALI) and the critical drivers of the inflammatory response (49). In elderly patients with ALI, changes in GM contribute to lung inflammation, which aggravates symptoms; this senescence-related susceptibility maybe due to AM aging (50). Intestinal ischemia/reperfusion (I/R) increases an individual’s susceptibility to ALI-related complications (51). Succinate, a metabolic byproduct of GM, accumulates during intestinal I/R and promotes lung damage by regulating AM polarization. Additionally, C5a produced during ALI promotes AM autophagy, leading to AM apoptosis and disruption in pulmonary homeostasis (52, 53). However, some treatment approaches play a protective role against ALI. The oral administration of propionic acid, another GM-derived metabolite, contributes to the suppression of AM-mediated inflammation during ALI in metal fume fever (54). Pretreatment with astragalus polysaccharides, a well-known immunoregulatory bioactive component, regulates AMs by changing the GM composition and increasing the SCFA level, which constitutes anti-inflammatory activity in ALI and plays a prophylactic role (55). Surprisingly, although MV is considered a routine and effective strategy to treat ALI, it probably increases gut permeability and enhances AM activation, which can exacerbate ALI and even cause systemic sepsis (56).

Chronic obstructive pulmonary disease (COPD) is a progressive disease with a significant incidence and mortality. Drug therapy can temporarily improve but not normalize lung function (57). Even after smoking cessation, other nondrug measures are necessary. AMs always remain activated in patients with COPD, aiding the characterization of the alveolar destruction that occurs during emphysema (58). The optimal depletion of potentially harmful bacteria and/or enrichment of beneficial bacteria in the gut may be alternative strategies for improving COPD. HFD can reduce the development of emphysema by inhibiting local and systemic inflammation. In previous studies, a significant decrease in the number of macrophages in bronchoalveolar lavage fluid was observed, as was a significant decrease in tissue destruction and the expression of healing-related factors that are mainly secreted by macrophages (59, 60). AMs play an important role in the response to pathogen infection in COPD lungs; however, additional studies should elucidate how to avoid damage to alveolar structures by strictly balancing AM activation.

Patients with chronic Inflammatory bowel disease (IBD) are more susceptible to developing chronic inflammatory lung diseases. In the absence of obvious lung disease, the lung function of IBD patients still worsens (61). Previous studies have reported that the lung tissue injury was accompanied by a significant increase of AMs in bronchoalveolar lavage fluid, while the increased expression of nicotinamide adenine dinucleotide phosphate oxidase 2 and C-C motif chemokine ligand 2 showed that AMs actively participated in the lung injury of patients with colitis (62). Thus, one may infer that GM disorder is an important driver for IBD complicated with chronic lung inflammation, as it is also a widely accepted important factor affecting AM functions during lung injury. In addition, colitis can affect bone marrow hematopoiesis, thereby enhancing monocyte release into the blood and infiltration into the lungs (63). Thus, monocyte-derived alveolar macrophages (Mo-AMs) may be an important driving factor of pulmonary inflammation in the context of colitis that is accomplished through a mechanism of the gut–bone marrow–lung axis, similar to our previous description of influenza infection and further extending that “double-edged benefit” concept.

Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal lung disease with limited treatment options and a huge demand for new therapies (64). Through inhibiting histone deacetylase, a promising therapeutic target for IPF, enterogenous butyric acid promotes the expression of antifibrosis factors to ameliorate IPF, probably because butyric acid regulates the imbalance of macrophage subpopulations by affecting the differentiation of macrophages to AMs (65, 66). As determined earlier, Mo-AMs, rather than TRAMs, are the main driving factor for the development of pulmonary fibrosis, and the selective targeting and regulation of AM differentiation can improve fibrosis without adverse consequences related to the depletion of whole monocytes or tissue-intrinsic AMs. It is worth noting that, after the fibrosis subsides, the Mo-AMs continue to exist in the lung for 1 year, indicating the need for continuous targeted therapy (67).

Pulmonary arterial hypertension (PAH) is characterized by occlusive pulmonary vascular remodeling, and the pulmonary inflammation mediated by macrophages around the pulmonary vessels is a key driver leading to pathological changes occurring within it (68). No effective treatment is available, but the gut–lung axis was recently highlighted in these patients. Patients with PAH tend to have proinflammatory gut dysbiosis, and HFD may help slow the disease progression, probably because metabolite butyric acid reduces hypoxia-induced AM accumulation. During hypoxia-induced PAH, AM activation is complex and partially dependent on the microenvironment; thus, in addition to directly affecting the number of AMs, butyrate has a protective effect on hypoxia-induced angiogenesis and endothelial barrier dysfunction (69, 70). Besides, structural changes in the lungs are rarely reversible because PAH regression takes more time than PAH development. Therefore, butyrate may be more effective at preventing than reversing PAH symptoms.

2.3 Allergic diseases

Macrophages were once called “forgotten cells in asthma.” Asthma-related alveolar dysfunction can cause serious respiratory diseases, including AM death and surfactant dysfunction (71). The replacement of TRAMs by Mo-AMs in the alveolar ecological niche promotes the transition to hyperreactivity (5).

The increased fungal burden in the gut is associated with the risk of severe asthma (72). Through Abx-induced GM imbalance and fungal overgrowth, AMs are polarized into M2 macrophages and airway allergic inflammation is promoted. Correspondingly, the adoptive transfer of AMs to naïve objects can induce similar phenomena (73). Conversely, the presence of normal microbiota is conducive to inducing innate immune tolerance in the lung (74).

Epidemiological studies suggest that, as the fiber content in the diet increases, the incidence of asthma decreases (75). This is likely because HFD produces higher levels of SCFAs compared to a low-fiber diet, thereby reducing M2 polarization of AMs and better control of allergic inflammation. In a prospective cohort study, an acute soluble fiber challenge had an acute antiinflammatory effect on the asthmatic airway. It was observed that the sputum sample macrophage counts decreased after 4 hours. In summary, these studies suggest that AMs, as target cells for the gut–lung axis, may play an important role in regulating allergic pulmonary inflammation. In terms of mechanism, SCFAs regulate AMs by activating GPR43 and/or GPR41; interestingly, their effects on both receptors are contradictory to each other (7678). However, the relationship between GPR41/GPR43 activation and AM differentiation remains unclear, and there are many possible explanations. Both receptors can be expressed in different cell subpopulations or tissues, thereby affecting different aspects of the immune response. The binding effect of GPR41/GPR43 receptors may be closely related to coreceptor or specific inflammatory cell differentiation, and further research must analyze the pleiotropic effects of GPR41/GPR43 (79, 80). It is worth noting that allergic inflammation usually exists long term and the TRAMs induced by the asthma response are replaced by Mo–AMs for at least 2 months after the allergy subsides (71). Most studies to date have focused only on the short-term benefits of HFD; thus, we must test whether a long-term HFD can promote the maintenance of the antiinflammatory phenotype of AMs. Moreover, AMs are associated with airway remodeling and airway hyperresponsiveness in chronic asthma (81, 82). Larger samples and longer clinical trials are necessary to determine the clinical effects of long-term supplementation.

2.4 Malignancy

Research is lacking on the effect of regulating AMs through the gut–lung axis on the occurrence and development of lung malignancies. AMs are widely involved in the formation of the tumor microenvironment, affecting tumor immunity and the immunotherapeutic response, which are important targets for cancer treatment (83, 84).

It has been reported that Influenza A virus infection allowing more effectively preventive antitumor immunity of mucosal resident AMs. This immune training strategy is particularly attractive to patients with extrapulmonary malignancies (such as melanoma and breast cancer) because they tend to develop lung metastases as well as for high-risk people with primary lung cancer. However, this kind of training immunity of AMs lasts for a limited time only (85). Maintaining sufficient long-term immune monitoring would be more beneficial for them, which illustrates the importance of continuous triggering factors. Nevertheless, it remains a promising approach to strengthening lung antitumor immunity.

Through long-term healthy dietary management, the risk of a new cancer diagnosis or cancer recurrence can be minimized, but most cancer patients struggle to maintain a beneficial and evidence-based diet after receiving the diagnosis, which may be due to differences in patient treatment and disease trajectory (86, 87). Providing appropriate dietary education is necessary; more importantly, understanding the relationship between diet and AM-mediated immunity is conducive to the development of personalized and practical nutritional plans. However, evidence of dietary interventions during tumor treatment is currently very limited (88). Via the gut–lung axis, it is unclear whether the anti-tumor function of AMs can be sustained. Additional research is urgently needed in this field. This treatment pattern, which focuses on the gut–distal organ axis–based regulation of tissue macrophages in the treatment of distal solid malignancy, not only provides new treatment targets and strategies to prevent and treat lung cancer, but it also provides new insights into the biological mechanism of systemic malignancy.

3 Conclusion

To our knowledge, the gut–lung axis is active throughout most of human life that has a continuous impact on AM function, origin, and renewal. A more thorough understanding of this multifaceted modulation has far-reaching significance. Dietary intervention, as we previously stated, has been widely studied for regulating AMs through the gut–lung axis (89). Dietary management is economical and universal, making it a feasible way to continuously trigger AMs to fight lung diseases. From above studies, dietary fiber can be transformed to prebiotics by probiotics, high-diet fiber diet tends to be beneficial in lung health by modulating proper AM immune response, but the effect of oral prebiotics is inconsistent, understanding their mechanism of action is conducive to the development of personalized and precision therapy. Moreover, although the concept that the gut–lung axis is a highly collaborative pathway based on AM plasticity is still in its infancy, its realization will make it feasible to develop personalized treatment with a double-edged benefit. However, the gut-lung axis is the mechanism with cross-organ communication, its further research on broader mechanisms are tough, but the application of induced pluripotent stem cell and multiorgan-on-a-chip technology have made it easier in vitro.

Author contributions

ZC: Conceptualization, Data curation, Formal Analysis, Methodology, Writing – original draft, Writing – review & editing. YL: Data curation, Formal Analysis, Writing – review & editing. WH: Conceptualization, Project administration, Supervision, Writing – review & editing.

Funding

The author(s) declare that no financial support was received for the research, authorship, and/or publication of this article.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Ashique S, De Rubis G, Sirohi E, Mishra N, Rihan M, Garg A, et al. Short Chain Fatty Acids: Fundamental mediators of the gut-lung axis and their involvement in pulmonary diseases. Chem Biol Interact (2022) 368:110231. doi: 10.1016/j.cbi.2022.110231

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Marín-Hinojosa C, Eraso CC, Sanchez-Lopez V, Hernández LC, Otero-Candelera R, Lopez-Campos JL. Nutriepigenomics and chronic obstructive pulmonary disease: potential role of dietary and epigenetics factors in disease development and management. Am J Clin Nutr (2021) 114:1894–906. doi: 10.1093/ajcn/nqab267

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Ma PJ, Wang MM, Wang Y. Gut microbiota: A new insight into lung diseases. BioMed Pharmacother (2022) 155:113810. doi: 10.1016/j.biopha.2022.113810

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Aegerter H, Lambrecht BN, Jakubzick CV. Biology of lung macrophages in health and disease. Immunity (2022) 55:1564–80. doi: 10.1016/j.immuni.2022.08.010

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Kulikauskaite J, Wack A. Teaching old dogs new tricks? The plasticity of lung alveolar macrophage subsets. Trends Immunol (2020) 41:864–77. doi: 10.1016/j.it.2020.08.008

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Torres A, Cilloniz C, Niederman MS, Menéndez R, Chalmers JD, Wunderink RG, et al. Pneumonia. Nat Rev Dis Primers (2021) 7:25. doi: 10.1038/s41572-021-00259-0

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Li L, Ma J, Yu Z, Li M, Zhang W, Sun H. Epidemiological characteristics and antibiotic resistance mechanisms of Streptococcus pneumoniae: An updated review. Microbiol Res (2023) 266:127221. doi: 10.1016/j.micres.2022.127221

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Schuijt TJ, Lankelma JM, Scicluna BP, de Sousa e Melo F, Roelofs JJ, de Boer JD, et al. The gut microbiota plays a protective role in the host defence against pneumococcal pneumonia. Gut (2016) 65:575–83. doi: 10.1136/gutjnl-2015-309728

PubMed Abstract | CrossRef Full Text | Google Scholar

9. MaChado MG, Patente TA, Rouillé Y, Heumel S, Melo EM, Deruyter L, et al. Acetate Improves the Killing of Streptococcus pneumoniae by Alveolar Macrophages via NLRP3 Inflammasome and Glycolysis-HIF-1α Axis. Front Immunol (2022) 13:773261. doi: 10.3389/fimmu.2022.773261

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Wu T, Li H, Su C, Xu F, Yang G, Sun K, et al. Microbiota-derived short-chain fatty acids promote LAMTOR2-mediated immune responses in macrophages. mSystems (2020) 5:e00587–20. doi: 10.1128/mSystems.00587-20

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Wu T, Xu F, Su C, Li H, Lv N, Liu Y, et al. Alterations in the gut microbiome and cecal metabolome during klebsiella pneumoniae-induced pneumosepsis. Front Immunol (2020) 11:1331. doi: 10.3389/fimmu.2020.01331

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Lankelma JM, Birnie E, Weehuizen TAF, Scicluna BP, Belzer C, Houtkooper RH, et al. The gut microbiota as a modulator of innate immunity during melioidosis. PloS Negl Trop Dis (2017) 11:e0005548. doi: 10.1371/journal.pntd.0005548

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Yang W, Ansari AR, Niu X, Zou W, Lu M, Dong L, et al. Interaction between gut microbiota dysbiosis and lung infection as gut-lung axis caused by Streptococcus suis in mouse model. Microbiol Res (2022) 261:127047. doi: 10.1016/j.micres.2022.127047

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Mitsi E, Carniel B, Reiné J, Rylance J, Zaidi S, Soares-Schanoski A, et al. Nasal pneumococcal density is associated with microaspiration and heightened human alveolar macrophage responsiveness to bacterial pathogens. Am J Respir Crit Care Med (2020) 20:335–47. doi: 10.1164/rccm.201903-0607OC

CrossRef Full Text | Google Scholar

15. Fukunaga R, Glaziou P, Harris JB, Date A, Floyd K, Kasaeva T. Epidemiology of tuberculosis and progress toward meeting global targets - worldwide, 2019. MMWR Morb Mortal Wkly Rep (2021) 70:427–30. doi: 10.15585/mmwr.mm7012a4

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Khan N, Mendonca L, Dhariwal A, Fontes G, Menzies D, Xia J, et al. Intestinal dysbiosis compromises alveolar macrophage immunity to Mycobacterium tuberculosis. Mucosal Immunol (2019) 12:772–83. doi: 10.1038/s41385-019-0147-3

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Lee WJ, Hase K. Gut microbiota-generated metabolites in animal health and disease. Nat Chem Biol (2014) 10:416–24. doi: 10.1038/nchembio.1535

PubMed Abstract | CrossRef Full Text | Google Scholar

18. D’Agostino MR, Lai R, Afkhami S, Khera A, Yao Y, Vaseghi-Shanjani M, et al. Airway Macrophages Mediate Mucosal Vaccine-Induced Trained Innate Immunity against Mycobacterium tuberculosis in Early Stages of Infection. J Immunol (2020) 205:2750–62. doi: 10.4049/jimmunol.2000532

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Jeyanathan M, Vaseghi-Shanjani M, Afkhami S, Grondin JA, Kang A, D’Agostino MR, et al. Parenteral BCG vaccine induces lung-resident memory macrophages and trained immunity via the gut-lung axis [published correction appears in Nat Immunol. 2023 Jun;24(6):1049]. Nat Immunol (2022) 23:1687–702. doi: 10.1038/s41590-022-01354-4

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Netea MG, Domínguez-Andrés J, Barreiro LB, Chavakis T, Divangahi M, Fuchs E, et al. Defining trained immunity and its role in health and disease. Nat Rev Immunol (2020) 20:375–88. doi: 10.1038/s41577-020-0285-6

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Baker JR Jr, Farazuddin M, Wong PT, O’Konek JJ. The unfulfilled potential of mucosal immunization. J Allergy Clin Immunol (2022) 150:1–11. doi: 10.1016/j.jaci.2022.05.002

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Sadikot RT, Blackwell TS, Christman JW, Prince AS. Pathogen-host interactions in Pseudomonas aeruginosa pneumonia. Am J Respir Crit Care Med (2005) 171:1209–23. doi: 10.1164/rccm.200408-1044SO

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Ranzani OT, Niederman MS, Torres A. Ventilator-associated pneumonia. Intensive Care Med (2022) 48:1222–6. doi: 10.1007/s00134-022-06773-3

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Albin OR, Kaye KS, McCreary EK, Pogue JM. Less is more? Antibiotic treatment duration in pseudomonas aeruginosa ventilator-associated pneumonia. Clin Infect Dis (2023) 76:745–9. doi: 10.1093/cid/ciac784

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Tsay TB, Yang MC, Chang WH, Chen PH, Chen LW. Lactobacillus salivarius reverse antibiotic-induced lung defense impairment in a ventilator model. J Transl Med (2018) 16:225. doi: 10.1186/s12967-018-1597-1

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Kallet RH, Matthay MA. Hyperoxic acute lung injury. Respir Care (2013) 58:123–41. doi: 10.4187/respcare.01963

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Chen LW, Chen PH, Fung CP, Hsu CM. Dead bacteria reverse antibiotic-induced host defense impairment in burns. J Am Coll Surg (2014) 219:606–19. doi: 10.1016/j.jamcollsurg.2014.04.016

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Wang G, Wu X, Zhu G, Han S, Zhang J. Dexmedetomidine alleviates sleep-restriction-mediated exaggeration of postoperative immunosuppression via splenic TFF2 in aged mice. Aging (Albany NY) (2020) 12:5318–35. doi: 10.18632/aging.102952

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Sencio V, Barthelemy A, Tavares LP, MaChado MG, Soulard D, Cuinat C, et al. Gut Dysbiosis during Influenza Contributes to Pulmonary Pneumococcal Superinfection through Altered Short-Chain Fatty Acid Production. Cell Rep (2020) 30:2934–2947.e6. doi: 10.1016/j.celrep.2020.02.013

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Monaco CL, Gootenberg DB, Zhao G, Handley SA, Ghebremichael MS, Lim ES, et al. Altered virome and bacterial microbiome in human immunodeficiency virus-associated acquired immunodeficiency syndrome. Cell Host Microbe (2016) 19:311–22. doi: 10.1016/j.chom.2016.02.011

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Alexandrova Y, Costiniuk CT, Jenabian MA. Pulmonary immune dysregulation and viral persistence during HIV infection. Front Immunol (2022) 12:808722. doi: 10.3389/fimmu.2021.808722

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Perin J, Mulick A, Yeung D, Villavicencio F, Lopez G, Strong KL, et al. Global, regional, and national causes of under-5 mortality in 2000-19: an updated systematic analysis with implications for the Sustainable Development Goals [published correction appears in Lancet Child Adolesc Health. 2022 Jan;6(1):e4]. Lancet Child Adolesc Health (2022) 6:106–15. doi: 10.1016/S2352-4642(21)00311-4

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Arboleya S, Sánchez B, Milani C, Duranti S, Solís G, Fernández N, et al. Intestinal microbiota development in preterm neonates and effect of perinatal antibiotics. J Pediatr (2015) 166:538–44. doi: 10.1016/j.jpeds.2014.09.041

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Stevens J, Steinmeyer S, Bonfield M, Peterson L, Wang T, Gray J, et al. The balance between protective and pathogenic immune responses to pneumonia in the neonatal lung is enforced by gut microbiota. Sci Transl Med (2022) 14:eabl3981. doi: 10.1126/scitranslmed.abl3981

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Evren E, Ringqvist E, Doisne JM, Thaller A, Sleiers N, Flavell RA, et al. CD116+ fetal precursors migrate to the perinatal lung and give rise to human alveolar macrophages. J Exp Med (2022) 219:e20210987. doi: 10.1084/jem.20210987

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Ardeshir A, Narayan NR, Méndez-Lagares G, Lu D, Rauch M, Huang Y, et al. Breast-fed and bottle-fed infant rhesus macaques develop distinct gut microbiotas and immune systems. Sci Transl Med (2014) 6:252ra120. doi: 10.1126/scitranslmed.3008791

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Mitchell CM, Mazzoni C, Hogstrom L, Bryant A, Bergerat A, Cher A, et al. Delivery mode affects stability of early infant gut microbiota. Cell Rep Med (2020) 1:100156. doi: 10.1016/j.xcrm.2020.100156

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Tsay TB, Yang MC, Chen PH, Hsu CM, Chen LW. Gut flora enhance bacterial clearance in lung through toll-like receptors 4. J BioMed Sci (2011) 18:68. doi: 10.1186/1423-0127-18-68

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Chen LW, Chen PH, Hsu CM. Commensal microflora contribute to host defense against Escherichia coli pneumonia through Toll-like receptors. Shock (2011) 36:67–75. doi: 10.1097/SHK.0b013e3182184ee7

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Clarke TB. Early innate immunity to bacterial infection in the lung is regulated systemically by the commensal microbiota via nod-like receptor ligands. Infect Immun (2014) 82:4596–606. doi: 10.1128/IAI.02212-14

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Cario E. Bacterial interactions with cells of the intestinal mucosa: Toll-like receptors and NOD2. Gut (2005) 5:1182–93. doi: 10.1136/gut.2004.062794

CrossRef Full Text | Google Scholar

42. Wang Z, Li S, Huang B. Alveolar macrophages: Achilles’ heel of SARS-CoV-2 infection. Signal Transduct Target Ther (2022) 7:242. doi: 10.1038/s41392-022-01106-8

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Li F, Piattini F, Pohlmeier L, Feng Q, Rehrauer H, Kopf M. Monocyte-derived alveolar macrophages autonomously determine severe outcome of respiratory viral infection. Sci Immunol (2022) 7:eabj5761. doi: 10.1126/sciimmunol.abj5761

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Cao X. COVID-19: immunopathology and its implications for therapy. Nat Rev Immunol (2020) 20:269–70. doi: 10.1038/s41577-020-0308-3

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Trompette A, Gollwitzer ES, Pattaroni C, Lopez-Mejia IC, Riva E, Pernot J, et al. Dietary fiber confers protection against flu by shaping ly6c- patrolling monocyte hematopoiesis and CD8+ T cell metabolism. Immunity (2018) 48:992–1005.e8. doi: 10.1016/j.immuni.2018.04.022

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Zhu W, Wang J, Zhao N, Zheng R, Wang D, Liu W, et al. Oral administration of Clostridium butyricum rescues streptomycin-exacerbated respiratory syncytial virus-induced lung inflammation in mice. Virulence (2021) 12:2133–48. doi: 10.1080/21505594.2021.1962137

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Garcia-Castillo V, Tomokiyo M, Raya Tonetti F, Islam MA, Takahashi H, Kitazawa H, et al. Alveolar macrophages are key players in the modulation of the respiratory antiviral immunity induced by orally administered lacticaseibacillus rhamnosus CRL1505. Front Immunol (2020) 11:568636. doi: 10.3389/fimmu.2020.568636

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Ji JJ, Sun QM, Nie DY, Wang Q, Zhang H, Qin FF, et al. Probiotics protect against RSV infection by modulating the microbiota-alveolar-macrophage axis. Acta Pharmacol Sin (2021) 42:1630–41. doi: 10.1038/s41401-020-00573-5

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Huang X, Xiu H, Zhang S, Zhang G. The role of macrophages in the pathogenesis of ALI/ARDS. Mediators Inflammation (2018) 2018:1264913. doi: 10.1155/2018/1264913

CrossRef Full Text | Google Scholar

50. Brandenberger C, Kling KM, Vital M, Christian M. The role of pulmonary and systemic immunosenescence in acute lung injury. Aging Dis (2018) 9:553–65. doi: 10.14336/AD.2017.0902

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Meyer NJ, Gattinoni L, Calfee CS. Acute respiratory distress syndrome. Lancet (2021) 398:622–37. doi: 10.1016/S0140-6736(21)00439-6

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Wang YH, Yan ZZ, Luo SD, Hu JJ, Wu M, Zhao J, et al. Gut microbiota-derived succinate aggravates acute lung injury after intestinal ischaemia/reperfusion in mice. Eur Respir J (2023) 61:2200840. doi: 10.1183/13993003.00840-2022

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Hu R, Chen ZF, Yan J, Li QF, Huang Y, Xu H, et al. Complement C5a exacerbates acute lung injury induced through autophagy-mediated alveolar macrophage apoptosis. Cell Death Dis (2014) 5:e1330. doi: 10.1038/cddis.2014.274

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Zhang Y, Zhang L, Mao L, Fan J, Jiang X, Li N, et al. Intestinal microbiota-derived propionic acid protects against zinc oxide nanoparticle-induced lung injury. Am J Respir Cell Mol Biol (2022) 67:680–94. doi: 10.1165/rcmb.2021-0515OC

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Ming K, Zhuang S, Ma N, Nan S, Li Q, Ding M, et al. Astragalus polysaccharides alleviates lipopolysaccharides-induced inflammatory lung injury by altering intestinal microbiota in mice. Front Microbiol (2022) 13:1033875. doi: 10.3389/fmicb.2022.1033875

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Jin S, Ding X, Yang C, Li W, Deng M, Liao H, et al. Mechanical ventilation exacerbates poly (I:C) induced acute lung injury: central role for caspase-11 and gut-lung axis. Front Immunol (2021) 12:693874. doi: 10.3389/fimmu.2021.693874

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Agustí A, Celli BR, Criner GJ, Halpin D, Anzueto A, Barnes P, et al. Global initiative for chronic obstructive lung disease 2023 report: GOLD executive summary. Am J Respir Crit Care Med (2023) 207:819–37. doi: 10.1164/rccm.202301-0106PP

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Barnes PJ. Alveolar macrophages as orchestrators of COPD. COPD (2004) 1:59–70. doi: 10.1081/COPD-120028701

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Lai HC, Lin TL, Chen TW, Kuo YL, Chang CJ, Wu TR, et al. Gut microbiota modulates COPD pathogenesis: role of anti-inflammatory Parabacteroides goldsteinii lipopolysaccharide. Gut (2022) 71:309–21. doi: 10.1136/gutjnl-2020-322599

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Jang YO, Lee SH, Choi JJ, Kim DH, Choi JM, Kang MJ, et al. Fecal microbial transplantation and a high fiber diet attenuates emphysema development by suppressing inflammation and apoptosis. Exp Mol Med (2020) 52:1128–39. doi: 10.1038/s12276-020-0469-y

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Rogler G, Singh A, Kavanaugh A, Rubin DT. Extraintestinal manifestations of inflammatory bowel disease: current concepts, treatment, and implications for disease management. Gastroenterology (2021) 161:1118–32. doi: 10.1053/j.gastro.2021.07.042

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Raftery AL, O’Brien CA, Harris NL, Tsantikos E, Hibbs ML. Development of severe colitis is associated with lung inflammation and pathology. Front Immunol (2023) 14:1125260. doi: 10.3389/fimmu.2023.1125260

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Mateer SW, Mathe A, Bruce J, Liu G, Maltby S, Fricker M, et al. IL-6 drives neutrophil-mediated pulmonary inflammation associated with bacteremia in murine models of colitis. Am J Pathol (2018) 188:1625–39. doi: 10.1016/j.ajpath.2018.03.016

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Lederer DJ, Martinez FJ. Idiopathic pulmonary fibrosis. N Engl J Med (2018) 378:1811–23. doi: 10.1056/NEJMra1705751

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Park HJ, Jeong OY, Chun SH, Cheon YH, Kim M, Kim S, et al. Butyrate improves skin/lung fibrosis and intestinal dysbiosis in bleomycin-induced mouse models. Int J Mol Sci (2021) 22:2765. doi: 10.3390/ijms22052765

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Korfei M, Mahavadi P, Guenther A. Targeting histone deacetylases in idiopathic pulmonary fibrosis: A future therapeutic option. Cells (2022) 11:1626. doi: 10.3390/cells11101626

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Misharin AV, Morales-Nebreda L, Reyfman PA, Cuda CM, Walter JM, McQuattie-Pimentel AC, et al. Monocyte-derived alveolar macrophages drive lung fibrosis and persist in the lung over the life span. J Exp Med (2017) 214:2387–404. doi: 10.1084/jem.20162152

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Florentin J, Dutta P. Origin and production of inflammatory perivascular macrophages in pulmonary hypertension. Cytokine (2017) 100:11–5. doi: 10.1016/j.cyto.2017.08.015

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Moutsoglou DM, Tatah J, Prisco SZ, Prins KW, Staley C, Lopez S, et al. Pulmonary arterial hypertension patients have a proinflammatory gut microbiome and altered circulating microbial metabolites. Am J Respir Crit Care Med (2023) 207:740–56. doi: 10.1164/rccm.202203-0490OC

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Callejo M, Barberá JA, Duarte J, Perez-Vizcaino F. Impact of nutrition on pulmonary arterial hypertension. Nutrients (2020) 12:169. doi: 10.3390/nu12010169

PubMed Abstract | CrossRef Full Text | Google Scholar

71. Feo-Lucas L, Godio C, Minguito de la Escalera M, Alvarez-Ladrón N, Villarrubia LH, Vega-Pérez A, et al. Airway allergy causes alveolar macrophage death, profound alveolar disorganization and surfactant dysfunction. Front Immunol (2023) 14:1125984. doi: 10.3389/fimmu.2023.1125984

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Kanj AN, Kottom TJ, Schaefbauer KJ, Choudhury M, Limper AH, Skalski JH. Dysbiosis of the intestinal fungal microbiota increases lung resident group 2 innate lymphoid cells and is associated with enhanced asthma severity in mice and humans. Respir Res (2023) 24:144. doi: 10.1186/s12931-023-02422-5

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Kim YG, Udayanga KG, Totsuka N, Weinberg JB, Núñez G, Shibuya A. Gut dysbiosis promotes M2 macrophage polarization and allergic airway inflammation via fungi-induced PGE2. Cell Host Microbe (2014) 15:95–102. doi: 10.1016/j.chom.2013.12.010

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Marquant Q, Laubreton D, Drajac C, Mathieu E, Bouguyon E, Noordine ML, et al. The microbiota plays a critical role in the reactivity of lung immune components to innate ligands. FASEB J (2021) 35:e21348. doi: 10.1096/fj.202002338R

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Saeed MA, Gribben KC, Alam M, Lyden ER, Hanson CK, LeVan TD. Association of dietary fiber on asthma, respiratory symptoms, and inflammation in the adult national health and nutrition examination survey population. Ann Am Thorac Soc (2020) 17:1062–8. doi: 10.1513/AnnalsATS.201910-776OC

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Trompette A, Gollwitzer ES, Yadava K, Sichelstiel AK, Sprenger N, Ngom-Bru C, et al. Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat Med (2014) 20:159–66. doi: 10.1038/nm.3444

PubMed Abstract | CrossRef Full Text | Google Scholar

77. Huang C, Du W, Ni Y, Lan G, Shi G. The effect of short-chain fatty acids on M2 macrophages polarization in vitro and in vivo. Clin Exp Immunol (2022) 207:53–64. doi: 10.1093/cei/uxab028

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Halnes I, Baines KJ, Berthon BS, MacDonald-Wicks LK, Gibson PG, Wood LG. Soluble fibre meal challenge reduces airway inflammation and expression of GPR43 and GPR41 in asthma. Nutrients (2017) 9:57. doi: 10.3390/nu9010057

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Li X, Kolling FW, Aridgides D, Mellinger D, Ashare A, Jakubzick CV. ScRNA-seq expression of IFI27 and APOC2 identifies four alveolar macrophage superclusters in healthy BALF. Life Sci Alliance (2022) 5:e202201458. doi: 10.26508/lsa.202201458

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Sivaprakasam S, Prasad PD, Singh N. Benefits of short-chain fatty acids and their receptors in inflammation and carcinogenesis. Pharmacol Ther (2016) 164:144–51. doi: 10.1016/j.pharmthera.2016.04.007

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Lee YG, Jeong JJ, Nyenhuis S, Berdyshev E, Chung S, Ranjan R, et al. Recruited alveolar macrophages, in response to airway epithelial-derived monocyte chemoattractant protein 1/CCl2, regulate airway inflammation and remodeling in allergic asthma. Am J Respir Cell Mol Biol (2015) 52:772–84. doi: 10.1165/rcmb.2014-0255OC

PubMed Abstract | CrossRef Full Text | Google Scholar

82. Careau E, Bissonnette EY. Adoptive transfer of alveolar macrophages abrogates bronchial hyperresponsiveness. Am J Respir Cell Mol Biol (2004) 31:22–7. doi: 10.1165/rcmb.2003-0229OC

PubMed Abstract | CrossRef Full Text | Google Scholar

83. Casanova-Acebes M, Dalla E, Leader AM, LeBerichel J, Nikolic J, Morales BM, et al. Tissue-resident macrophages provide a pro-tumorigenic niche to early NSCLC cells. Nature (2021) 595:578–84. doi: 10.1038/s41586-021-03651-8

PubMed Abstract | CrossRef Full Text | Google Scholar

84. Loyher PL, Hamon P, Laviron M, Meghraoui-Kheddar A, Goncalves E, Deng Z, et al. Macrophages of distinct origins contribute to tumor development in the lung. J Exp Med (2018) 215:2536–53. doi: 10.1084/jem.20180534

PubMed Abstract | CrossRef Full Text | Google Scholar

85. Wang T, Zhang J, Wang Y, Li Y, Wang L, Yu Y, et al. Influenza-trained mucosal-resident alveolar macrophages confer long-term antitumor immunity in the lungs. Nat Immunol (2023) 24:423–38. doi: 10.1038/s41590-023-01428-x

PubMed Abstract | CrossRef Full Text | Google Scholar

86. Wiseman M. The second World Cancer Research Fund/American Institute for Cancer Research expert report. Food, nutrition, physical activity, and the prevention of cancer: a global perspective. Proc Nutr Soc (2008) 67:253–6. doi: 10.1017/S002966510800712X

PubMed Abstract | CrossRef Full Text | Google Scholar

87. Tan SY, Wong HY, Vardy JL. Do cancer survivors change their diet after cancer diagnosis? Support Care Cancer (2021) 29:6921–7. doi: 10.1007/s00520-021-06276-9

PubMed Abstract | CrossRef Full Text | Google Scholar

88. Ligibel JA, Bohlke K, May AM, Clinton SK, Demark-Wahnefried W, Gilchrist SC, et al. Exercise, diet, and weight management during cancer treatment: ASCO guideline. J Clin Oncol (2022) 40:2491–507. doi: 10.1200/JCO.22.00687

PubMed Abstract | CrossRef Full Text | Google Scholar

89. Anand S, Mande SS. Diet, microbiota and gut-lung connection. Front Microbiol (2018) 9:2147. doi: 10.3389/fmicb.2018.02147

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: alveolar macrophages, gut-lung axis, dietary, microbiota, lung diseases, immunity

Citation: Chen Z, Liu Y and Huang W (2023) Alveolar macrophage modulation via the gut–lung axis in lung diseases. Front. Immunol. 14:1279677. doi: 10.3389/fimmu.2023.1279677

Received: 18 August 2023; Accepted: 06 November 2023;
Published: 21 November 2023.

Edited by:

Christine Freeman, University of Michigan, United States

Reviewed by:

Coen Govers, Wageningen University and Research, Netherlands

Copyright © 2023 Chen, Liu and Huang. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Weizhe Huang, huangweizhe1980@163.com

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.