Microbiota and Immunity during Respiratory Infections: Lung and Gut Affair
Abstract
:1. Introduction
2. Lung Microbiota
2.1. Lung Microbiota Modifications during Respiratory Infections
2.2. Lung Immune Responses and Host–Microbe Interactions during Respiratory Infections
3. The Gut–Lung Axis
3.1. Impact of Respiratory Infections on Gut Microbiota
3.2. Impact of Gut Microbiota on Respiratory Infection Outcome
4. Targeting Microbiota to Counteract Respiratory Infections
4.1. Microbiota Modulation in the Context of Viral Infections
4.2. Microbiota Modulation in the Context of Bacterial Infections
5. Concluding Remarks
Author Contributions
Funding
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
Abbreviations
ACE2 | Angiotensin-converting enzyme 2 |
BCG | Bacillo di Calmette-Guérin |
BALF | broncho alveolar lavage |
CAP | community acquired pneumonia |
CXCL | C-X-C motif chemokine ligand |
CCL | C-C chemokine ligand |
DAT | desaminotyrosine |
DCs | dendritic cells |
FOXP3 | Forkhead box P3 |
HSCT | hematopoietic stem cell transplantation |
ILCs | innate lymphoid cells |
IL | interleukin |
iNKT | invariant Natural Killer T cells |
IV | influenza virus |
NLR | NOD like receptors |
L. | Lactobacillus |
LRT | low respiratory tract |
MAIT | mucosal-associated invariant T |
MBL | mannan-binding lectin |
Mtb | Mycobacterium tuberculosis |
PI3K | phosphatidylinositol 3-kinase |
PRRs | pattern recognition receptors |
RSV | Respiratory Syncytial Virus |
S. | Streptococcus |
SARS-CoV-2 | Severe Acute Respiratory Syndrome Coronavirus 2 |
SFB | segmented filamentous bacteria |
TB | tuberculosis |
TLR | toll-like receptor |
TNF | Tumor Necrosis Factor |
TRM | resident memory T |
TH | T helper |
K. | Klebsiella |
URT | upper respiratory tract |
References
- Alimi, Y.; Lim, W.S.; Lansbury, L.; Leonardi-Bee, J.; Nguyen-Van-Tam, J.S. Systematic review of respiratory viral pathogens identified in adults with community-acquired pneumonia in Europe. J. Clin. Virol. 2017, 95, 26–35. [Google Scholar] [CrossRef] [PubMed]
- Ferkol, T.; Schraufnagel, D. The global burden of respiratory disease. Ann. Am. Thorac. Soc. 2014, 11, 404–406. [Google Scholar] [CrossRef] [PubMed]
- Wypych, T.P.; Wickramasinghe, L.C.; Marsland, B.J. The influence of the microbiome on respiratory health. Nat. Immunol. 2019, 20, 1279–1290. [Google Scholar] [CrossRef] [PubMed]
- Mathieu, E.; Escribano-Vazquez, U.; Descamps, D.; Cherbuy, C.; Langella, P.; Riffault, S.; Remot, A.; Thomas, M. Paradigms of Lung Microbiota Functions in Health and Disease, Particularly, in Asthma. Front. Physiol. 2018, 9, 1168. [Google Scholar] [CrossRef] [PubMed]
- O’Dwyer, D.N.; Dickson, R.P.; Moore, B.B. The Lung Microbiome, Immunity, and the Pathogenesis of Chronic Lung Disease. J. Immunol. 2016, 196, 4839–4847. [Google Scholar] [CrossRef] [PubMed]
- Sommariva, M.; Le Noci, V.; Bianchi, F.; Camelliti, S.; Balsari, A.; Tagliabue, E.; Sfondrini, L. The lung microbiota: Role in maintaining pulmonary immune homeostasis and its implications in cancer development and therapy. Cell. Mol. Life Sci. 2020, 77, 2739–2749. [Google Scholar] [CrossRef] [PubMed]
- Natalini, J.G.; Singh, S.; Segal, L.N. The dynamic lung microbiome in health and disease. Nat. Rev. Microbiol. 2023, 21, 222–235. [Google Scholar] [CrossRef] [PubMed]
- Man, W.H.; de Steenhuijsen Piters, W.A.; Bogaert, D. The microbiota of the respiratory tract: Gatekeeper to respiratory health. Nat. Rev. Microbiol. 2017, 15, 259–270. [Google Scholar] [CrossRef] [PubMed]
- Pattaroni, C.; Watzenboeck, M.L.; Schneidegger, S.; Kieser, S.; Wong, N.C.; Bernasconi, E.; Pernot, J.; Mercier, L.; Knapp, S.; Nicod, L.P.; et al. Early-Life Formation of the Microbial and Immunological Environment of the Human Airways. Cell Host Microbe 2018, 24, 857–865.e4. [Google Scholar] [CrossRef]
- Dickson, R.P.; Erb-Downward, J.R.; Martinez, F.J.; Huffnagle, G.B. The Microbiome and the Respiratory Tract. Annu. Rev. Physiol. 2016, 78, 481–504. [Google Scholar] [CrossRef]
- Hilty, M.; Burke, C.; Pedro, H.; Cardenas, P.; Bush, A.; Bossley, C.; Davies, J.; Ervine, A.; Poulter, L.; Pachter, L.; et al. Disordered microbial communities in asthmatic airways. PLoS ONE 2010, 5, e8578. [Google Scholar] [CrossRef] [PubMed]
- Bassis, C.M.; Erb-Downward, J.R.; Dickson, R.P.; Freeman, C.M.; Schmidt, T.M.; Young, V.B.; Beck, J.M.; Curtis, J.L.; Huffnagle, G.B. Analysis of the upper respiratory tract microbiotas as the source of the lung and gastric microbiotas in healthy individuals. mBio 2015, 6, e00037-15. [Google Scholar] [CrossRef] [PubMed]
- Dickson, R.P.; Erb-Downward, J.R.; Huffnagle, G.B. Homeostasis and its disruption in the lung microbiome. Am. J. Physiol. Lung Cell. Mol. Physiol. 2015, 309, L1047–L1055. [Google Scholar] [CrossRef] [PubMed]
- Wu, B.G.; Sulaiman, I.; Tsay, J.J.; Perez, L.; Franca, B.; Li, Y.; Wang, J.; Gonzalez, A.N.; El-Ashmawy, M.; Carpenito, J.; et al. Episodic Aspiration with Oral Commensals Induces a MyD88-dependent, Pulmonary T-Helper Cell Type 17 Response that Mitigates Susceptibility to Streptococcus pneumoniae. Am. J. Respir. Crit. Care Med. 2021, 203, 1099–1111. [Google Scholar] [CrossRef] [PubMed]
- Wu, B.G.; Segal, L.N. The Lung Microbiome and Its Role in Pneumonia. Clin. Chest Med. 2018, 39, 677–689. [Google Scholar] [CrossRef] [PubMed]
- Dickson, R.P.; Erb-Downward, J.R.; Freeman, C.M.; McCloskey, L.; Falkowski, N.R.; Huffnagle, G.B.; Curtis, J.L. Bacterial Topography of the Healthy Human Lower Respiratory Tract. mBio 2017, 8, e02287-16. [Google Scholar] [CrossRef]
- Lohmann, P.; Luna, R.A.; Hollister, E.B.; Devaraj, S.; Mistretta, T.A.; Welty, S.E.; Versalovic, J. The airway microbiome of intubated premature infants: Characteristics and changes that predict the development of bronchopulmonary dysplasia. Pediatr. Res. 2014, 76, 294–301. [Google Scholar] [CrossRef] [PubMed]
- Bosch, A.A.T.M.; de Steenhuijsen Piters, W.A.A.; van Houten, M.A.; Chu, M.L.J.N.; Biesbroek, G.; Kool, J.; Pernet, P.; de Groot, P.C.M.; Eijkemans, M.J.C.; Keijser, B.J.F.; et al. Maturation of the Infant Respiratory Microbiota, Environmental Drivers, and Health Consequences. A Prospective Cohort Study. Am. J. Respir. Crit. Care Med. 2017, 196, 1582–1590. [Google Scholar] [CrossRef]
- Gollwitzer, E.S.; Saglani, S.; Trompette, A.; Yadava, K.; Sherburn, R.; McCoy, K.D.; Nicod, L.P.; Lloyd, C.M.; Marsland, B.J. Lung microbiota promotes tolerance to allergens in neonates via PD-L1. Nat. Med. 2014, 20, 642–647. [Google Scholar] [CrossRef]
- Dickson, R.P.; Martinez, F.J.; Huffnagle, G.B. The role of the microbiome in exacerbations of chronic lung diseases. Lancet 2014, 384, 691–702. [Google Scholar] [CrossRef]
- Budden, K.F.; Shukla, S.D.; Rehman, S.F.; Bowerman, K.L.; Keely, S.; Hugenholtz, P.; Armstrong-James, D.P.H.; Adcock, I.M.; Chotirmall, S.H.; Chung, K.F.; et al. Functional effects of the microbiota in chronic respiratory disease. Lancet Respir. Med. 2019, 7, 907–920. [Google Scholar] [CrossRef] [PubMed]
- Yagi, K.; Huffnagle, G.B.; Lukacs, N.W.; Asai, N. The Lung Microbiome during Health and Disease. Int. J. Mol. Sci. 2021, 22, 10872. [Google Scholar] [CrossRef] [PubMed]
- Dickson, R.P.; Erb-Downward, J.R.; Freeman, C.M.; McCloskey, L.; Beck, J.M.; Huffnagle, G.B.; Curtis, J.L. Spatial Variation in the Healthy Human Lung Microbiome and the Adapted Island Model of Lung Biogeography. Ann. Am. Thorac. Soc. 2015, 12, 821–830. [Google Scholar] [CrossRef] [PubMed]
- Marimón, J.M.; Sorarrain, A.; Ercibengoa, M.; Azcue, N.; Alonso, M.; Vidaur, L. Lung microbiome on admission in critically ill patients with acute bacterial and viral pneumonia. Sci. Rep. 2023, 14, 2737. [Google Scholar] [CrossRef] [PubMed]
- Huang, X.; Luo, Y.; Wang, J.; Zhang, X.; Chen, L.; Wu, R.; Xue, Z.; Gu, H.; Li, D.; Tang, H.; et al. Integrative study of pulmonary microbiome, transcriptome and clinical outcomes in Mycoplasma pneumoniae pneumonia. Respir. Res. 2024, 25, 35. [Google Scholar] [CrossRef] [PubMed]
- Zhan, D.; Li, D.; Yuan, K.; Sun, Y.; He, L.; Zhong, J.; Wang, L. Characteristics of the pulmonary microbiota in patients with mild and severe pulmonary infection. Front. Cell. Infect. Microbiol. 2023, 13, 1227581. [Google Scholar] [CrossRef] [PubMed]
- Belizário, J.; Garay-Malpartida, M.; Faintuch, J. Lung microbiome and origins of the respiratory diseases. Curr. Res. Immunol. 2023, 4, 100065. [Google Scholar] [CrossRef] [PubMed]
- Whiteside, S.A.; McGinniss, J.E.; Collman, R.G. The lung microbiome: Progress and promise. J. Clin. Investig. 2021, 131, e150473. [Google Scholar] [CrossRef] [PubMed]
- Barcik, W.; Boutin, R.C.T.; Sokolowska, M.; Finlay, B.B. The Role of Lung and Gut Microbiota in the Pathology of Asthma. Immunity 2020, 52, 241–255. [Google Scholar] [CrossRef]
- Segal, L.N.; Clemente, J.C.; Tsay, J.C.; Koralov, S.B.; Keller, B.C.; Wu, B.G.; Li, Y.; Shen, N.; Ghedin, E.; Morris, A.; et al. Enrichment of the lung microbiome with oral taxa is associated with lung inflammation of a Th17 phenotype. Nat. Microbiol. 2016, 1, 16031. [Google Scholar] [CrossRef]
- Segal, L.N.; Clemente, J.C.; Li, Y.; Ruan, C.; Cao, J.; Danckers, M.; Morris, A.; Tapyrik, S.; Wu, B.G.; Diaz, P.; et al. Anaerobic Bacterial Fermentation Products Increase Tuberculosis Risk in Antiretroviral-Drug-Treated HIV Patients. Cell Host Microbe 2017, 21, 530–537.e4. [Google Scholar] [CrossRef] [PubMed]
- Gustafson, C.E.; Kim, C.; Weyand, C.M.; Goronzy, J.J. Influence of immune aging on vaccine responses. J. Allergy Clin. Immunol. 2020, 145, 1309–1321. [Google Scholar] [CrossRef] [PubMed]
- Drigot, Z.G.; Clark, S.E. Insights into the role of the respiratory tract microbiome in defense against bacterial pneumonia. Curr. Opin. Microbiol. 2024, 77, 102428. [Google Scholar] [CrossRef] [PubMed]
- Yadava, K.; Pattaroni, C.; Sichelstiel, A.K.; Trompette, A.; Gollwitzer, E.S.; Salami, O.; von Garnier, C.; Nicod, L.P.; Marsland, B.J. Microbiota Promotes Chronic Pulmonary Inflammation by Enhancing IL-17A and Autoantibodies. Am. J. Respir. Crit. Care Med. 2016, 193, 975–987. [Google Scholar] [CrossRef] [PubMed]
- Larsen, J.M.; Musavian, H.S.; Butt, T.M.; Ingvorsen, C.; Thysen, A.H.; Brix, S. Chronic obstructive pulmonary disease and asthma-associated Proteobacteria, but not commensal Prevotella spp., promote Toll-like receptor 2-independent lung inflammation and pathology. Immunology 2015, 144, 333–342. [Google Scholar] [CrossRef] [PubMed]
- Jin, C.; Lagoudas, G.K.; Zhao, C.; Bullman, S.; Bhutkar, A.; Hu, B.; Ameh, S.; Sandel, D.; Liang, X.S.; Mazzilli, S.; et al. Commensal Microbiota Promote Lung Cancer Development via γδ T Cells. Cell 2019, 176, 998–1013.e16. [Google Scholar] [CrossRef]
- Kim, Y.G.; Udayanga, K.G.; Totsuka, N.; Weinberg, J.B.; 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. [Google Scholar] [CrossRef]
- Hufnagl, K.; Pali-Schöll, I.; Roth-Walter, F.; Jensen-Jarolim, E. Dysbiosis of the gut and lung microbiome has a role in asthma. Semin. Immunopathol. 2020, 42, 75–93. [Google Scholar] [CrossRef]
- Wiertsema, S.P.; van Bergenhenegouwen, J.; Garssen, J.; Knippels, L.M.J. The Interplay between the Gut Microbiome and the Immune System in the Context of Infectious Diseases throughout Life and the Role of Nutrition in Optimizing Treatment Strategies. Nutrients 2021, 13, 886. [Google Scholar] [CrossRef]
- Haro, C.; Rangel-Zúñiga, O.A.; Alcalá-Díaz, J.F.; Gómez-Delgado, F.; Pérez-Martínez, P.; Delgado-Lista, J.; Quintana-Navarro, G.M.; Landa, B.B.; Navas-Cortés, J.A.; Tena-Sempere, M.; et al. Intestinal Microbiota Is Influenced by Gender and Body Mass Index. PLoS ONE 2016, 11, e0154090. [Google Scholar] [CrossRef]
- Fauci, A.S. Infectious diseases: Considerations for the 21st century. Clin. Infect. Dis. 2001, 32, 675–685. [Google Scholar] [CrossRef] [PubMed]
- Gopallawa, I.; Dehinwal, R.; Bhatia, V.; Gujar, V.; Chirmule, N. A four-part guide to lung immunology: Invasion, inflammation, immunity, and intervention. Front. Immunol. 2023, 14, 1119564. [Google Scholar] [CrossRef] [PubMed]
- Dickson, R.P.; Erb-Downward, J.R.; Prescott, H.C.; Martinez, F.J.; Curtis, J.L.; Lama, V.N.; Huffnagle, G.B. Analysis of culture-dependent versus culture-independent techniques for identification of bacteria in clinically obtained bronchoalveolar lavage fluid. J. Clin. Microbiol. 2014, 52, 3605–3613. [Google Scholar] [CrossRef] [PubMed]
- Vissing, N.H.; Chawes, B.L.; Bisgaard, H. Increased risk of pneumonia and bronchiolitis after bacterial colonization of the airways as neonates. Am. J. Respir. Crit. Care Med. 2013, 188, 1246–1252. [Google Scholar] [CrossRef] [PubMed]
- Lee, K.H.; Gordon, A.; Shedden, K.; Kuan, G.; Ng, S.; Balmaseda, A.; Foxman, B. The respiratory microbiome and susceptibility to influenza virus infection. PLoS ONE 2019, 14, e0207898. [Google Scholar] [CrossRef] [PubMed]
- Kaul, D.; Rathnasinghe, R.; Ferres, M.; Tan, G.S.; Barrera, A.; Pickett, B.E.; Methe, B.A.; Das, S.R.; Budnik, I.; Halpin, R.A.; et al. Microbiome disturbance and resilience dynamics of the upper respiratory tract during influenza A virus infection. Nat. Commun. 2020, 11, 2537. [Google Scholar] [CrossRef] [PubMed]
- Teo, S.M.; Mok, D.; Pham, K.; Kusel, M.; Serralha, M.; Troy, N.; Holt, B.J.; Hales, B.J.; Walker, M.L.; Hollams, E.; et al. The infant nasopharyngeal microbiome impacts severity of lower respiratory infection and risk of asthma development. Cell Host Microbe 2015, 17, 704–715. [Google Scholar] [CrossRef] [PubMed]
- Toivonen, L.; Hasegawa, K.; Waris, M.; Ajami, N.J.; Petrosino, J.F.; Camargo, C.A.; Peltola, V. Early nasal microbiota and acute respiratory infections during the first years of life. Thorax 2019, 74, 592–599. [Google Scholar] [CrossRef] [PubMed]
- Gu, L.; Deng, H.; Ren, Z.; Zhao, Y.; Yu, S.; Guo, Y.; Dai, J.; Chen, X.; Li, K.; Li, R.; et al. Dynamic Changes in the Microbiome and Mucosal Immune Microenvironment of the Lower Respiratory Tract by Influenza Virus Infection. Front. Microbiol. 2019, 10, 2491. [Google Scholar] [CrossRef]
- Barbosa-Amezcua, M.; Galeana-Cadena, D.; Alvarado-Peña, N.; Silva-Herzog, E. The Microbiome as Part of the Contemporary View of Tuberculosis Disease. Pathogens 2022, 11, 584. [Google Scholar] [CrossRef]
- Pant, A.; Das, B.; Arimbasseri, G.A. Host microbiome in tuberculosis: Disease, treatment, and immunity perspectives. Front. Microbiol. 2023, 14, 1236348. [Google Scholar] [CrossRef] [PubMed]
- Mori, G.; Morrison, M.; Blumenthal, A. Microbiome-immune interactions in tuberculosis. PLoS Pathog. 2021, 17, e1009377. [Google Scholar] [CrossRef]
- Hu, Y.; Kang, Y.; Liu, X.; Cheng, M.; Dong, J.; Sun, L.; Zhu, Y.; Ren, X.; Yang, Q.; Chen, X.; et al. Distinct lung microbial community states in patients with pulmonary tuberculosis. Sci. China Life Sci. 2020, 63, 1522–1533. [Google Scholar] [CrossRef]
- Dickson, R.P.; Erb-Downward, J.R.; Huffnagle, G.B. The role of the bacterial microbiome in lung disease. Expert Rev. Respir. Med. 2013, 7, 245–257. [Google Scholar] [CrossRef] [PubMed]
- Namasivayam, S.; Sher, A.; Glickman, M.S.; Wipperman, M.F. The Microbiome and Tuberculosis: Early Evidence for Cross Talk. mBio 2018, 9, e01420-18. [Google Scholar] [CrossRef] [PubMed]
- Wu, J.; Liu, W.; He, L.; Huang, F.; Chen, J.; Cui, P.; Shen, Y.; Zhao, J.; Wang, W.; Zhang, Y.; et al. Sputum microbiota associated with new, recurrent and treatment failure tuberculosis. PLoS ONE 2013, 8, e83445. [Google Scholar] [CrossRef]
- Krishna, P.; Jain, A.; Bisen, P.S. Microbiome diversity in the sputum of patients with pulmonary tuberculosis. Eur. J. Clin. Microbiol. Infect. Dis. 2016, 35, 1205–1210. [Google Scholar] [CrossRef]
- Ding, L.; Liu, Y.; Wu, X.; Wu, M.; Luo, X.; Ouyang, H.; Xia, J.; Liu, X.; Ding, T. Pathogen Metagenomics Reveals Distinct Lung Microbiota Signatures Between Bacteriologically Confirmed and Negative Tuberculosis Patients. Front. Cell. Infect. Microbiol. 2021, 11, 708827. [Google Scholar] [CrossRef]
- Vázquez-Pérez, J.A.; Carrillo, C.O.; Iñiguez-García, M.A.; Romero-Espinoza, I.; Márquez-García, J.E.; Falcón, L.I.; Torres, M.; Herrera, M.T. Alveolar microbiota profile in patients with human pulmonary tuberculosis and interstitial pneumonia. Microb. Pathog. 2020, 139, 103851. [Google Scholar] [CrossRef]
- Nakhaee, M.; Rezaee, A.; Basiri, R.; Soleimanpour, S.; Ghazvini, K. Relation between lower respiratory tract microbiota and type of immune response against tuberculosis. Microb. Pathog. 2018, 120, 161–165. [Google Scholar] [CrossRef]
- Huang, Y.; Tang, J.H.; Cai, Z.; Qi, Y.; Jiang, S.; Ma, T.T.; Yue, Y.; Huang, F.; Yang, H.; Ma, Y.Y. Alterations in the nasopharyngeal microbiota associated with active and latent tuberculosis. Tuberculosis 2022, 136, 102231. [Google Scholar] [CrossRef]
- Ancona, G.; Alagna, L.; Alteri, C.; Palomba, E.; Tonizzo, A.; Pastena, A.; Muscatello, A.; Gori, A.; Bandera, A. Gut and airway microbiota dysbiosis and their role in COVID-19 and long-COVID. Front. Immunol. 2023, 14, 1080043. [Google Scholar] [CrossRef] [PubMed]
- Merenstein, C.; Bushman, F.D.; Collman, R.G. Alterations in the respiratory tract microbiome in COVID-19: Current observations and potential significance. Microbiome 2022, 10, 165. [Google Scholar] [CrossRef] [PubMed]
- Sulaiman, I.; Wu, B.G.; Li, Y.; Tsay, J.C.; Sauthoff, M.; Scott, A.S.; Ji, K.; Koralov, S.B.; Weiden, M.; Clemente, J.C.; et al. Functional lower airways genomic profiling of the microbiome to capture active microbial metabolism. Eur. Respir. J. 2021, 58, 2003434. [Google Scholar] [CrossRef] [PubMed]
- Gaibani, P.; Viciani, E.; Bartoletti, M.; Lewis, R.E.; Tonetti, T.; Lombardo, D.; Castagnetti, A.; Bovo, F.; Horna, C.S.; Ranieri, M.; et al. The lower respiratory tract microbiome of critically ill patients with COVID-19. Sci. Rep. 2021, 11, 10103. [Google Scholar] [CrossRef]
- Cyprian, F.; Sohail, M.U.; Abdelhafez, I.; Salman, S.; Attique, Z.; Kamareddine, L.; Al-Asmakh, M. SARS-CoV-2 and immune-microbiome interactions: Lessons from respiratory viral infections. Int. J. Infect. Dis. 2021, 105, 540–550. [Google Scholar] [CrossRef] [PubMed]
- Fan, J.; Li, X.; Gao, Y.; Zhou, J.; Wang, S.; Huang, B.; Wu, J.; Cao, Q.; Chen, Y.; Wang, Z.; et al. The lung tissue microbiota features of 20 deceased patients with COVID-19. J. Infect. 2020, 81, e64–e67. [Google Scholar] [CrossRef]
- De Maio, F.; Posteraro, B.; Ponziani, F.R.; Cattani, P.; Gasbarrini, A.; Sanguinetti, M. Nasopharyngeal Microbiota Profiling of SARS-CoV-2 Infected Patients. Biol. Proced. Online 2020, 22, 18. [Google Scholar] [CrossRef] [PubMed]
- Minich, J.J.; Ali, F.; Marotz, C.; Belda-Ferre, P.; Chiang, L.; Shaffer, J.P.; Carpenter, C.S.; McDonald, D.; Gilbert, J.; Allard, S.M.; et al. Feasibility of using alternative swabs and storage solutions for paired SARS-CoV-2 detection and microbiome analysis in the hospital environment. Microbiome 2021, 9, 25. [Google Scholar] [CrossRef]
- Ren, L.; Wang, Y.; Zhong, J.; Li, X.; Xiao, Y.; Li, J.; Yang, J.; Fan, G.; Guo, L.; Shen, Z.; et al. Dynamics of the Upper Respiratory Tract Microbiota and Its Association with Mortality in COVID-19. Am. J. Respir. Crit. Care Med. 2021, 204, 1379–1390. [Google Scholar] [CrossRef]
- Zhu, T.; Jin, J.; Chen, M.; Chen, Y. The impact of infection with COVID-19 on the respiratory microbiome: A narrative review. Virulence 2022, 13, 1076–1087. [Google Scholar] [CrossRef] [PubMed]
- Shaykhiev, R.; Bals, R. Interactions between epithelial cells and leukocytes in immunity and tissue homeostasis. J. Leukoc. Biol. 2007, 82, 1–15. [Google Scholar] [CrossRef] [PubMed]
- Invernizzi, R.; Lloyd, C.M.; Molyneaux, P.L. Respiratory microbiome and epithelial interactions shape immunity in the lungs. Immunology 2020, 160, 171–182. [Google Scholar] [CrossRef] [PubMed]
- Knowles, M.R.; Boucher, R.C. Mucus clearance as a primary innate defense mechanism for mammalian airways. J. Clin. Investig. 2002, 109, 571–577. [Google Scholar] [CrossRef] [PubMed]
- Ehre, C.; Worthington, E.N.; Liesman, R.M.; Grubb, B.R.; Barbier, D.; O’Neal, W.K.; Sallenave, J.M.; Pickles, R.J.; Boucher, R.C. Overexpressing mouse model demonstrates the protective role of Muc5ac in the lungs. Proc. Natl. Acad. Sci. USA 2012, 109, 16528–16533. [Google Scholar] [CrossRef] [PubMed]
- Roy, M.G.; Livraghi-Butrico, A.; Fletcher, A.A.; McElwee, M.M.; Evans, S.E.; Boerner, R.M.; Alexander, S.N.; Bellinghausen, L.K.; Song, A.S.; Petrova, Y.M.; et al. Muc5b is required for airway defence. Nature 2014, 505, 412–416. [Google Scholar] [CrossRef]
- Pilette, C.; Ouadrhiri, Y.; Godding, V.; Vaerman, J.P.; Sibille, Y. Lung mucosal immunity: Immunoglobulin-A revisited. Eur. Respir. J. 2001, 18, 571–588. [Google Scholar] [CrossRef] [PubMed]
- Janoff, E.N.; Fasching, C.; Orenstein, J.M.; Rubins, J.B.; Opstad, N.L.; Dalmasso, A.P. Killing of Streptococcus pneumoniae by capsular polysaccharide-specific polymeric IgA, complement, and phagocytes. J. Clin. Investig. 1999, 104, 1139–1147. [Google Scholar] [CrossRef] [PubMed]
- Rodríguez, A.; Tjärnlund, A.; Ivanji, J.; Singh, M.; García, I.; Williams, A.; Marsh, P.D.; Troye-Blomberg, M.; Fernández, C. Role of IgA in the defense against respiratory infections IgA deficient mice exhibited increased susceptibility to intranasal infection with Mycobacterium bovis BCG. Vaccine 2005, 23, 2565–2572. [Google Scholar] [CrossRef]
- Joseph, J. Harnessing Nasal Immunity with IgA to Prevent Respiratory Infections. Immuno 2022, 2, 571–583. [Google Scholar] [CrossRef]
- Hiemstra, P.S.; Amatngalim, G.D.; van der Does, A.M.; Taube, C. Antimicrobial Peptides and Innate Lung Defenses: Role in Infectious and Noninfectious Lung Diseases and Therapeutic Applications. Chest 2016, 149, 545–551. [Google Scholar] [CrossRef]
- Malainou, C.; Abdin, S.M.; Lachmann, N.; Matt, U.; Herold, S. Alveolar macrophages in tissue homeostasis, inflammation, and infection: Evolving concepts of therapeutic targeting. J. Clin. Investig. 2023, 133, e170501. [Google Scholar] [CrossRef] [PubMed]
- Neupane, A.S.; Willson, M.; Chojnacki, A.K.; Vargas E Silva Castanheira, F.; Morehouse, C.; Carestia, A.; Keller, A.E.; Peiseler, M.; DiGiandomenico, A.; Kelly, M.M.; et al. Patrolling Alveolar Macrophages Conceal Bacteria from the Immune System to Maintain Homeostasis. Cell 2020, 183, 110–125.e11. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef]
- Santos, L.D.; Antunes, K.H.; Muraro, S.P.; de Souza, G.F.; da Silva, A.G.; Felipe, J.S.; Zanetti, L.C.; Czepielewski, R.S.; Magnus, K.; Scotta, M.; et al. TNF-mediated alveolar macrophage necroptosis drives disease pathogenesis during respiratory syncytial virus infection. Eur. Respir. J. 2021, 57, 2003764. [Google Scholar] [CrossRef] [PubMed]
- Bain, C.C.; Lucas, C.D.; Rossi, A.G. Pulmonary macrophages and SARS-Cov2 infection. Int. Rev. Cell Mol. Biol. 2022, 367, 1–28. [Google Scholar] [CrossRef] [PubMed]
- Lin, K.L.; Suzuki, Y.; Nakano, H.; Ramsburg, E.; Gunn, M.D. CCR2+ monocyte-derived dendritic cells and exudate macrophages produce influenza-induced pulmonary immune pathology and mortality. J. Immunol. 2008, 180, 2562–2572. [Google Scholar] [CrossRef]
- Antoine, B.M.K. Contribution of Dendritic Cell Responses to Sepsis-Induced Immunosuppression and to Susceptibility to Secondary Pneumonia. Front. Immunol. 2018, 9, 2590. [Google Scholar] [CrossRef]
- Kim, H.; Shin, S.J. Pathological and protective roles of dendritic cells in. Front. Cell. Infect. Microbiol. 2022, 12, 891878. [Google Scholar] [CrossRef]
- Campana, P.; Parisi, V.; Leosco, D.; Bencivenga, D.; Della Ragione, F.; Borriello, A. Dendritic Cells and SARS-CoV-2 Infection: Still an Unclarified Connection. Cells 2020, 9, 2046. [Google Scholar] [CrossRef]
- Stehle, C.; Hernández, D.C.; Romagnani, C. Innate lymphoid cells in lung infection and immunity. Immunol. Rev. 2018, 286, 102–119. [Google Scholar] [CrossRef]
- Monticelli, L.A.; Sonnenberg, G.F.; Abt, M.C.; Alenghat, T.; Ziegler, C.G.; Doering, T.A.; Angelosanto, J.M.; Laidlaw, B.J.; Yang, C.Y.; Sathaliyawala, T.; et al. Innate lymphoid cells promote lung-tissue homeostasis after infection with influenza virus. Nat. Immunol. 2011, 12, 1045–1054. [Google Scholar] [CrossRef] [PubMed]
- Das, S.; Chauhan, K.S.; Ahmed, M.; Akter, S.; Lu, L.; Colonna, M.; Khader, S.A. Lung type 3 innate lymphoid cells respond early following. mBio 2024, e03299-23. [Google Scholar] [CrossRef]
- Silverstein, N.J.; Wang, Y.; Manickas-Hill, Z.; Carbone, C.; Dauphin, A.; Boribong, B.P.; Loiselle, M.; Davis, J.; Leonard, M.M.; Kuri-Cervantes, L.; et al. Innate lymphoid cells and COVID-19 severity in SARS-CoV-2 infection. eLife 2022, 11, e74681. [Google Scholar] [CrossRef] [PubMed]
- Gold, M.C.; Cerri, S.; Smyk-Pearson, S.; Cansler, M.E.; Vogt, T.M.; Delepine, J.; Winata, E.; Swarbrick, G.M.; Chua, W.J.; Yu, Y.Y.; et al. Human mucosal associated invariant T cells detect bacterially infected cells. PLoS Biol. 2010, 8, e1000407. [Google Scholar] [CrossRef]
- Le Bourhis, L.; Martin, E.; Péguillet, I.; Guihot, A.; Froux, N.; Coré, M.; Lévy, E.; Dusseaux, M.; Meyssonnier, V.; Premel, V.; et al. Antimicrobial activity of mucosal-associated invariant T cells. Nat. Immunol. 2010, 11, 701–708. [Google Scholar] [CrossRef]
- Wen, X.; Zhang, X.; Nian, S.; Wei, G.; Guo, X.; Yu, H.; Xie, X.; Ye, Y.; Yuan, Q. Title of article: Mucosal-associated invariant T cells in lung diseases. Int. Immunopharmacol. 2021, 94, 107485. [Google Scholar] [CrossRef] [PubMed]
- Malka-Ruimy, C.; Ben Youssef, G.; Lambert, M.; Tourret, M.; Ghazarian, L.; Faye, A.; Caillat-Zucman, S.; Houdouin, V. Mucosal-Associated Invariant T Cell Levels Are Reduced in the Peripheral Blood and Lungs of Children with Active Pulmonary Tuberculosis. Front. Immunol. 2019, 10, 206. [Google Scholar] [CrossRef] [PubMed]
- Huang, X.; Kantonen, J.; Nowlan, K.; Nguyen, N.A.; Jokiranta, S.T.; Kuivanen, S.; Heikkilä, N.; Mahzabin, S.; Kantele, A.; Vapalahti, O.; et al. Mucosal-Associated Invariant T Cells are not susceptible in vitro to SARS-CoV-2 infection but accumulate into the lungs of COVID-19 patients. Virus Res. 2024, 341, 199315. [Google Scholar] [CrossRef]
- Jeong, D.; Woo, Y.D.; Chung, D.H. Invariant natural killer T cells in lung diseases. Exp. Mol. Med. 2023, 55, 1885–1894. [Google Scholar] [CrossRef]
- Im, J.S.; Kang, T.J.; Lee, S.B.; Kim, C.H.; Lee, S.H.; Venkataswamy, M.M.; Serfass, E.R.; Chen, B.; Illarionov, P.A.; Besra, G.S.; et al. Alteration of the relative levels of iNKT cell subsets is associated with chronic mycobacterial infections. Clin. Immunol. 2008, 127, 214–224. [Google Scholar] [CrossRef]
- Zingaropoli, M.A.; Perri, V.; Pasculli, P.; Cogliati Dezza, F.; Nijhawan, P.; Savelloni, G.; La Torre, G.; D’Agostino, C.; Mengoni, F.; Lichtner, M.; et al. Major reduction of NKT cells in patients with severe COVID-19 pneumonia. Clin. Immunol. 2021, 222, 108630. [Google Scholar] [CrossRef] [PubMed]
- Tang, J.; Sun, J. Lung tissue-resident memory T cells: The gatekeeper to respiratory viral (re)-infection. Curr. Opin. Immunol. 2023, 80, 102278. [Google Scholar] [CrossRef] [PubMed]
- Cheon, I.S.; Son, Y.M.; Sun, J. Tissue-resident memory T cells and lung immunopathology. Immunol. Rev. 2023, 316, 63–83. [Google Scholar] [CrossRef]
- Pizzolla, A.; Nguyen, T.H.; Sant, S.; Jaffar, J.; Loudovaris, T.; Mannering, S.I.; Thomas, P.G.; Westall, G.P.; Kedzierska, K.; Wakim, L.M. Influenza-specific lung-resident memory T cells are proliferative and polyfunctional and maintain diverse TCR profiles. J. Clin. Investig. 2018, 128, 721–733. [Google Scholar] [CrossRef]
- Jozwik, A.; Habibi, M.S.; Paras, A.; Zhu, J.; Guvenel, A.; Dhariwal, J.; Almond, M.; Wong, E.H.C.; Sykes, A.; Maybeno, M.; et al. RSV-specific airway resident memory CD8+ T cells and differential disease severity after experimental human infection. Nat. Commun. 2015, 6, 10224. [Google Scholar] [CrossRef] [PubMed]
- Grau-Expósito, J.; Sánchez-Gaona, N.; Massana, N.; Suppi, M.; Astorga-Gamaza, A.; Perea, D.; Rosado, J.; Falcó, A.; Kirkegaard, C.; Torrella, A.; et al. Peripheral and lung resident memory T cell responses against SARS-CoV-2. Nat. Commun. 2021, 12, 3010. [Google Scholar] [CrossRef] [PubMed]
- Kirby, A.C.; Newton, D.J.; Carding, S.R.; Kaye, P.M. Evidence for the involvement of lung-specific gammadelta T cell subsets in local responses to Streptococcus pneumoniae infection. Eur. J. Immunol. 2007, 37, 3404–3413. [Google Scholar] [CrossRef] [PubMed]
- Sanz, M.; Mann, B.T.; Chitrakar, A.; Soriano-Sarabia, N. Defying convention in the time of COVID-19: Insights into the role of γδ T cells. Front. Immunol. 2022, 13, 819574. [Google Scholar] [CrossRef]
- Chen, K.; McAleer, J.P.; Lin, Y.; Paterson, D.L.; Zheng, M.; Alcorn, J.F.; Weaver, C.T.; Kolls, J.K. Th17 cells mediate clade-specific, serotype-independent mucosal immunity. Immunity 2011, 35, 997–1009. [Google Scholar] [CrossRef]
- Malley, R.; Anderson, P.W. Serotype-independent pneumococcal experimental vaccines that induce cellular as well as humoral immunity. Proc. Natl. Acad. Sci. USA 2012, 109, 3623–3627. [Google Scholar] [CrossRef] [PubMed]
- Gopal, R.; Monin, L.; Slight, S.; Uche, U.; Blanchard, E.; Fallert Junecko, B.A.; Ramos-Payan, R.; Stallings, C.L.; Reinhart, T.A.; Kolls, J.K.; et al. Unexpected role for IL-17 in protective immunity against hypervirulent Mycobacterium tuberculosis HN878 infection. PLoS Pathog. 2014, 10, e1004099. [Google Scholar] [CrossRef] [PubMed]
- Martonik, D.; Parfieniuk-Kowerda, A.; Rogalska, M.; Flisiak, R. The Role of Th17 Response in COVID-19. Cells 2021, 10, 1550. [Google Scholar] [CrossRef] [PubMed]
- Jovisic, M.; Mambetsariev, N.; Singer, B.D.; Morales-Nebreda, L. Differential roles of regulatory T cells in acute respiratory infections. J. Clin. Investig. 2023, 133, e170505. [Google Scholar] [CrossRef] [PubMed]
- Crother, T.R.; Schröder, N.W.; Karlin, J.; Chen, S.; Shimada, K.; Slepenkin, A.; Alsabeh, R.; Peterson, E.; Arditi, M. Chlamydia pneumoniae infection induced allergic airway sensitization is controlled by regulatory T-cells and plasmacytoid dendritic cells. PLoS ONE 2011, 6, e20784. [Google Scholar] [CrossRef] [PubMed]
- Neill, D.R.; Fernandes, V.E.; Wisby, L.; Haynes, A.R.; Ferreira, D.M.; Laher, A.; Strickland, N.; Gordon, S.B.; Denny, P.; Kadioglu, A.; et al. T regulatory cells control susceptibility to invasive pneumococcal pneumonia in mice. PLoS Pathog. 2012, 8, e1002660. [Google Scholar] [CrossRef]
- Betts, R.J.; Prabhu, N.; Ho, A.W.; Lew, F.C.; Hutchinson, P.E.; Rotzschke, O.; Macary, P.A.; Kemeny, D.M. Influenza A virus infection results in a robust, antigen-responsive, and widely disseminated Foxp3+ regulatory T cell response. J. Virol. 2012, 86, 2817–2825. [Google Scholar] [CrossRef] [PubMed]
- Chang, Y.J.; Kim, H.Y.; Albacker, L.A.; Baumgarth, N.; McKenzie, A.N.; Smith, D.E.; Dekruyff, R.H.; Umetsu, D.T. Innate lymphoid cells mediate influenza-induced airway hyper-reactivity independently of adaptive immunity. Nat. Immunol. 2011, 12, 631–638. [Google Scholar] [CrossRef] [PubMed]
- Wang, H.; Wang, Z.; Cao, W.; Wu, Q.; Yuan, Y.; Zhang, X. Regulatory T cells in COVID-19. Aging Dis. 2021, 12, 1545–1553. [Google Scholar] [CrossRef]
- Swiatczak, B.; Cohen, I.R. Gut feelings of safety: Tolerance to the microbiota mediated by innate immune receptors. Microbiol. Immunol. 2015, 59, 573–585. [Google Scholar] [CrossRef]
- Uehara, A.; Fujimoto, Y.; Fukase, K.; Takada, H. Various human epithelial cells express functional Toll-like receptors, NOD1 and NOD2 to produce anti-microbial peptides, but not proinflammatory cytokines. Mol. Immunol. 2007, 44, 3100–3111. [Google Scholar] [CrossRef] [PubMed]
- Brown, R.L.; Sequeira, R.P.; Clarke, T.B. The microbiota protects against respiratory infection via GM-CSF signaling. Nat. Commun. 2017, 8, 1512. [Google Scholar] [CrossRef] [PubMed]
- Butcher, S.K.; O’Carroll, C.E.; Wells, C.A.; Carmody, R.J. Toll-Like Receptors Drive Specific Patterns of Tolerance and Training on Restimulation of Macrophages. Front. Immunol. 2018, 9, 933. [Google Scholar] [CrossRef]
- Zeng, Q.; Jewell, C.M. Directing toll-like receptor signaling in macrophages to enhance tumor immunotherapy. Curr. Opin. Biotechnol. 2019, 60, 138–145. [Google Scholar] [CrossRef] [PubMed]
- Belkaid, Y.; Harrison, O.J. Homeostatic Immunity and the Microbiota. Immunity 2017, 46, 562–576. [Google Scholar] [CrossRef]
- Belkaid, Y.; Hand, T.W. Role of the microbiota in immunity and inflammation. Cell 2014, 157, 121–141. [Google Scholar] [CrossRef] [PubMed]
- Kau, A.L.; Ahern, P.P.; Griffin, N.W.; Goodman, A.L.; Gordon, J.I. Human nutrition, the gut microbiome and the immune system. Nature 2011, 474, 327–336. [Google Scholar] [CrossRef] [PubMed]
- Zheng, D.; Liwinski, T.; Elinav, E. Interaction between microbiota and immunity in health and disease. Cell Res. 2020, 30, 492–506. [Google Scholar] [CrossRef] [PubMed]
- Müller, M.M.; Baldauf, C.; Hornischer, S.; Klassert, T.E.; Schneegans, A.; Behnert, A.; Pletz, M.W.; Hagel, S.; Slevogt, H. Staphylococcus aureus induces tolerance in human monocytes accompanied with expression changes of cell surface markers. Front. Immunol. 2023, 14, 1046374. [Google Scholar] [CrossRef]
- Ratner, A.J.; Lysenko, E.S.; Paul, M.N.; Weiser, J.N. Synergistic proinflammatory responses induced by polymicrobial colonization of epithelial surfaces. Proc. Natl. Acad. Sci. USA 2005, 102, 3429–3434. [Google Scholar] [CrossRef]
- Yao, Y.; Jeyanathan, M.; Haddadi, S.; Barra, N.G.; Vaseghi-Shanjani, M.; Damjanovic, D.; Lai, R.; Afkhami, S.; Chen, Y.; Dvorkin-Gheva, A.; et al. Induction of Autonomous Memory Alveolar Macrophages Requires T Cell Help and Is Critical to Trained Immunity. Cell 2018, 175, 1634–1650.e17. [Google Scholar] [CrossRef] [PubMed]
- Fagundes, C.T.; Amaral, F.A.; Vieira, A.T.; Soares, A.C.; Pinho, V.; Nicoli, J.R.; Vieira, L.Q.; Teixeira, M.M.; Souza, D.G. Transient TLR activation restores inflammatory response and ability to control pulmonary bacterial infection in germfree mice. J. Immunol. 2012, 188, 1411–1420. [Google Scholar] [CrossRef] [PubMed]
- Fox, A.C.; McConnell, K.W.; Yoseph, B.P.; Breed, E.; Liang, Z.; Clark, A.T.; O’Donnell, D.; Zee-Cheng, B.; Jung, E.; Dominguez, J.A.; et al. The endogenous bacteria alter gut epithelial apoptosis and decrease mortality following Pseudomonas aeruginosa pneumonia. Shock 2012, 38, 508–514. [Google Scholar] [CrossRef] [PubMed]
- Herbst, T.; Sichelstiel, A.; Schär, C.; Yadava, K.; Bürki, K.; Cahenzli, J.; McCoy, K.; Marsland, B.J.; Harris, N.L. Dysregulation of allergic airway inflammation in the absence of microbial colonization. Am. J. Respir. Crit. Care Med. 2011, 184, 198–205. [Google Scholar] [CrossRef] [PubMed]
- Naik, S.; Bouladoux, N.; Linehan, J.L.; Han, S.J.; Harrison, O.J.; Wilhelm, C.; Conlan, S.; Himmelfarb, S.; Byrd, A.L.; Deming, C.; et al. Commensal-dendritic-cell interaction specifies a unique protective skin immune signature. Nature 2015, 520, 104–108. [Google Scholar] [CrossRef] [PubMed]
- Barfod, K.K.; Vrankx, K.; Mirsepasi-Lauridsen, H.C.; Hansen, J.S.; Hougaard, K.S.; Larsen, S.T.; Ouwenhand, A.C.; Krogfelt, K.A. The Murine Lung Microbiome Changes during Lung Inflammation and Intranasal Vancomycin Treatment. Open Microbiol. J. 2015, 9, 167–179. [Google Scholar] [CrossRef] [PubMed]
- Ubags, N.D.J.; Marsland, B.J. Mechanistic insight into the function of the microbiome in lung diseases. Eur. Respir. J. 2017, 50, 1602467. [Google Scholar] [CrossRef]
- Schuijt, T.J.; Lankelma, J.M.; Scicluna, B.P.; de Sousa e Melo, F.; Roelofs, J.J.; de Boer, J.D.; Hoogendijk, A.J.; de Beer, R.; de Vos, A.; Belzer, C.; et al. The gut microbiota plays a protective role in the host defence against pneumococcal pneumonia. Gut 2016, 65, 575–583. [Google Scholar] [CrossRef]
- Schroeder, B.O.; Bäckhed, F. Signals from the gut microbiota to distant organs in physiology and disease. Nat. Med. 2016, 22, 1079–1089. [Google Scholar] [CrossRef]
- Feng, Q.; Chen, W.D.; Wang, Y.D. Gut Microbiota: An Integral Moderator in Health and Disease. Front. Microbiol. 2018, 9, 151. [Google Scholar] [CrossRef]
- Chakradhar, S. A curious connection: Teasing apart the link between gut microbes and lung disease. Nat. Med. 2017, 23, 402–404. [Google Scholar] [CrossRef] [PubMed]
- Li, Z.; Li, Y.; Sun, Q.; Wei, J.; Li, B.; Qiu, Y.; Liu, K.; Shao, D.; Ma, Z. Targeting the Pulmonary Microbiota to Fight against Respiratory Diseases. Cells 2022, 11, 916. [Google Scholar] [CrossRef] [PubMed]
- Li, R.; Li, J.; Zhou, X. Lung microbiome: New insights into the pathogenesis of respiratory diseases. Signal Transduct. Target. Ther. 2024, 9, 19. [Google Scholar] [CrossRef] [PubMed]
- Hu, Y.; Feng, Y.; Wu, J.; Liu, F.; Zhang, Z.; Hao, Y.; Liang, S.; Li, B.; Li, J.; Lv, N.; et al. The Gut Microbiome Signatures Discriminate Healthy From Pulmonary Tuberculosis Patients. Front. Cell. Infect. Microbiol. 2019, 9, 90. [Google Scholar] [CrossRef] [PubMed]
- Luo, M.; Liu, Y.; Wu, P.; Luo, D.X.; Sun, Q.; Zheng, H.; Hu, R.; Pandol, S.J.; Li, Q.F.; Han, Y.P.; et al. Alternation of Gut Microbiota in Patients with Pulmonary Tuberculosis. Front. Physiol. 2017, 8, 822. [Google Scholar] [CrossRef]
- Li, W.; Zhu, Y.; Liao, Q.; Wang, Z.; Wan, C. Characterization of gut microbiota in children with pulmonary tuberculosis. BMC Pediatr. 2019, 19, 445. [Google Scholar] [CrossRef] [PubMed]
- Osei Sekyere, J.; Maningi, N.E.; Fourie, P.B. Mycobacterium tuberculosis, antimicrobials, immunity, and lung-gut microbiota crosstalk: Current updates and emerging advances. Ann. N. Y. Acad. Sci. 2020, 1467, 21–47. [Google Scholar] [CrossRef] [PubMed]
- Naidoo, C.C.; Nyawo, G.R.; Wu, B.G.; Walzl, G.; Warren, R.M.; Segal, L.N.; Theron, G. The microbiome and tuberculosis: State of the art, potential applications, and defining the clinical research agenda. Lancet Respir. Med. 2019, 7, 892–906. [Google Scholar] [CrossRef] [PubMed]
- Leylabadlo, H.E.; Ghotaslou, R.; Feizabadi, M.M.; Farajnia, S.; Moaddab, S.Y.; Ganbarov, K.; Khodadadi, E.; Tanomand, A.; Sheykhsaran, E.; Yousefi, B.; et al. The critical role of Faecalibacterium prausnitzii in human health: An overview. Microb. Pathog. 2020, 149, 104344. [Google Scholar] [CrossRef]
- Qin, N.; Zheng, B.; Yao, J.; Guo, L.; Zuo, J.; Wu, L.; Zhou, J.; Liu, L.; Guo, J.; Ni, S.; et al. Influence of H7N9 virus infection and associated treatment on human gut microbiota. Sci. Rep. 2015, 5, 14771. [Google Scholar] [CrossRef]
- Gu, S.; Chen, Y.; Wu, Z.; Gao, H.; Lv, L.; Guo, F.; Zhang, X.; Luo, R.; Huang, C.; Lu, H.; et al. Alterations of the Gut Microbiota in Patients with Coronavirus Disease 2019 or H1N1 Influenza. Clin. Infect. Dis. 2020, 71, 2669–2678. [Google Scholar] [CrossRef]
- Woodall, C.A.; McGeoch, L.J.; Hay, A.D.; Hammond, A. Respiratory tract infections and gut microbiome modifications: A systematic review. PLoS ONE 2022, 17, e0262057. [Google Scholar] [CrossRef] [PubMed]
- Xiao, F.; Tang, M.; Zheng, X.; Liu, Y.; Li, X.; Shan, H. Evidence for Gastrointestinal Infection of SARS-CoV-2. Gastroenterology 2020, 158, 1831–1833.e3. [Google Scholar] [CrossRef]
- Lamers, M.M.; Haagmans, B.L. SARS-CoV-2 pathogenesis. Nat. Rev. Microbiol. 2022, 20, 270–284. [Google Scholar] [CrossRef]
- Zuo, T.; Zhang, F.; Lui, G.C.Y.; Yeoh, Y.K.; Li, A.Y.L.; Zhan, H.; Wan, Y.; Chung, A.C.K.; Cheung, C.P.; Chen, N.; et al. Alterations in Gut Microbiota of Patients with COVID-19 during Time of Hospitalization. Gastroenterology 2020, 159, 944–955.e8. [Google Scholar] [CrossRef]
- Albrich, W.C.; Ghosh, T.S.; Ahearn-Ford, S.; Mikaeloff, F.; Lunjani, N.; Forde, B.; Suh, N.; Kleger, G.R.; Pietsch, U.; Frischknecht, M.; et al. A high-risk gut microbiota configuration associates with fatal hyperinflammatory immune and metabolic responses to SARS-CoV-2. Gut Microbes 2022, 14, 2073131. [Google Scholar] [CrossRef] [PubMed]
- Zhou, B.; Pang, X.; Wu, J.; Liu, T.; Wang, B.; Cao, H. Gut microbiota in COVID-19: New insights from inside. Gut Microbes 2023, 15, 2201157. [Google Scholar] [CrossRef] [PubMed]
- Bernard-Raichon, L.; Venzon, M.; Klein, J.; Axelrad, J.E.; Zhang, C.; Sullivan, A.P.; Hussey, G.A.; Casanovas-Massana, A.; Noval, M.G.; Valero-Jimenez, A.M.; et al. Gut microbiome dysbiosis in antibiotic-treated COVID-19 patients is associated with microbial translocation and bacteremia. Nat. Commun. 2022, 13, 5926. [Google Scholar] [CrossRef]
- Samuelson, D.R.; Charles, T.P.; de la Rua, N.M.; Taylor, C.M.; Blanchard, E.E.; Luo, M.; Shellito, J.E.; Welsh, D.A. Analysis of the intestinal microbial community and inferred functional capacities during the host response to Pneumocystis pneumonia. Exp. Lung Res. 2016, 42, 425–439. [Google Scholar] [CrossRef]
- Groves, H.T.; Cuthbertson, L.; James, P.; Moffatt, M.F.; Cox, M.J.; Tregoning, J.S. Respiratory Disease following Viral Lung Infection Alters the Murine Gut Microbiota. Front. Immunol. 2018, 9, 182. [Google Scholar] [CrossRef]
- Deriu, E.; Boxx, G.M.; He, X.; Pan, C.; Benavidez, S.D.; Cen, L.; Rozengurt, N.; Shi, W.; Cheng, G. Influenza Virus Affects Intestinal Microbiota and Secondary Salmonella Infection in the Gut through Type I Interferons. PLoS Pathog. 2016, 12, e1005572. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Li, F.; Wei, H.; Lian, Z.X.; Sun, R.; Tian, Z. Respiratory influenza virus infection induces intestinal immune injury via microbiota-mediated Th17 cell-dependent inflammation. J. Exp. Med. 2014, 211, 2397–2410. [Google Scholar] [CrossRef] [PubMed]
- Sze, M.A.; Tsuruta, M.; Yang, S.W.; Oh, Y.; Man, S.F.; Hogg, J.C.; Sin, D.D. Changes in the bacterial microbiota in gut, blood, and lungs following acute LPS instillation into mice lungs. PLoS ONE 2014, 9, e111228. [Google Scholar] [CrossRef] [PubMed]
- Jiang, Q.; Xu, Q.; Kenéz, Á.; Chen, S.; Yang, G. Klebsiella pneumoniae infection is associated with alterations in the gut microbiome and lung metabolome. Microbiol. Res. 2022, 263, 127139. [Google Scholar] [CrossRef] [PubMed]
- Wolff, N.S.; Jacobs, M.C.; Wiersinga, W.J.; Hugenholtz, F. Pulmonary and intestinal microbiota dynamics during Gram-negative pneumonia-derived sepsis. Intensive Care Med. Exp. 2021, 9, 35. [Google Scholar] [CrossRef] [PubMed]
- Groves, H.T.; Higham, S.L.; Moffatt, M.F.; Cox, M.J.; Tregoning, J.S. Respiratory Viral Infection Alters the Gut Microbiota by Inducing Inappetence. mBio 2020, 11, e03236-19. [Google Scholar] [CrossRef] [PubMed]
- Sencio, V.; Machado, M.G.; Trottein, F. The lung-gut axis during viral respiratory infections: The impact of gut dysbiosis on secondary disease outcomes. Mucosal Immunol. 2021, 14, 296–304. [Google Scholar] [CrossRef]
- Tulic, M.K.; Piche, T.; Verhasselt, V. Lung-gut cross-talk: Evidence, mechanisms and implications for the mucosal inflammatory diseases. Clin. Exp. Allergy 2016, 46, 519–528. [Google Scholar] [CrossRef]
- Majewski, S.; Piotrowski, W. Pulmonary manifestations of inflammatory bowel disease. Arch. Med. Sci. 2015, 11, 1179–1188. [Google Scholar] [CrossRef]
- Black, H.; Mendoza, M.; Murin, S. Thoracic manifestations of inflammatory bowel disease. Chest 2007, 131, 524–532. [Google Scholar] [CrossRef]
- Kuzela, L.; Vavrecka, A.; Prikazska, M.; Drugda, B.; Hronec, J.; Senkova, A.; Drugdova, M.; Oltman, M.; Novotna, T.; Brezina, M.; et al. Pulmonary complications in patients with inflammatory bowel disease. Hepatogastroenterology 1999, 46, 1714–1719. [Google Scholar] [PubMed]
- Ogimi, C.; Krantz, E.M.; Golob, J.L.; Waghmare, A.; Liu, C.; Leisenring, W.M.; Woodard, C.R.; Marquis, S.; Kuypers, J.M.; Jerome, K.R.; et al. Antibiotic Exposure Prior to Respiratory Viral Infection Is Associated with Progression to Lower Respiratory Tract Disease in Allogeneic Hematopoietic Cell Transplant Recipients. Biol. Blood Marrow Transplant. 2018, 24, 2293–2301. [Google Scholar] [CrossRef] [PubMed]
- Lee, W.C.; Chou, H.S.; Wu, T.H.; Cheng, C.H.; Lee, C.F.; Wang, Y.C.; Wu, T.J.; Chan, K.M. Low-dose anti-hepatitis B immunoglobulin regimen as prophylaxis for hepatitis B recurrence after liver transplantation. Transpl. Infect. Dis. 2019, 21, e13190. [Google Scholar] [CrossRef] [PubMed]
- Haak, B.W.; Littmann, E.R.; Chaubard, J.L.; Pickard, A.J.; Fontana, E.; Adhi, F.; Gyaltshen, Y.; Ling, L.; Morjaria, S.M.; Peled, J.U.; et al. Impact of gut colonization with butyrate-producing microbiota on respiratory viral infection following allo-HCT. Blood 2018, 131, 2978–2986. [Google Scholar] [CrossRef] [PubMed]
- Tang, B.M.; Shojaei, M.; Teoh, S.; Meyers, A.; Ho, J.; Ball, T.B.; Keynan, Y.; Pisipati, A.; Kumar, A.; Eisen, D.P.; et al. Neutrophils-related host factors associated with severe disease and fatality in patients with influenza infection. Nat. Commun. 2019, 10, 3422. [Google Scholar] [CrossRef] [PubMed]
- Sokol, H.; Contreras, V.; Maisonnasse, P.; Desmons, A.; Delache, B.; Sencio, V.; Machelart, A.; Brisebarre, A.; Humbert, L.; Deryuter, L.; et al. SARS-CoV-2 infection in nonhuman primates alters the composition and functional activity of the gut microbiota. Gut Microbes 2021, 13, 1–19. [Google Scholar] [CrossRef] [PubMed]
- Nagata, N.; Takeuchi, T.; Masuoka, H.; Aoki, R.; Ishikane, M.; Iwamoto, N.; Sugiyama, M.; Suda, W.; Nakanishi, Y.; Terada-Hirashima, J.; et al. Human Gut Microbiota and Its Metabolites Impact Immune Responses in COVID-19 and Its Complications. Gastroenterology 2023, 164, 272–288. [Google Scholar] [CrossRef]
- Mendes de Almeida, V.; Engel, D.F.; Ricci, M.F.; Cruz, C.S.; Lopes, Í.; Alves, D.A.; d’Auriol, M.; Magalhães, J.; Machado, E.C.; Rocha, V.M.; et al. Gut microbiota from patients with COVID-19 cause alterations in mice that resemble post-COVID symptoms. Gut Microbes 2023, 15, 2249146. [Google Scholar] [CrossRef] [PubMed]
- Yang, T.; Chakraborty, S.; Saha, P.; Mell, B.; Cheng, X.; Yeo, J.Y.; Mei, X.; Zhou, G.; Mandal, J.; Golonka, R.; et al. Gnotobiotic Rats Reveal That Gut Microbiota Regulates Colonic mRNA of. Hypertension 2020, 76, e1–e3. [Google Scholar] [CrossRef]
- Dhar, D.; Mohanty, A. Gut microbiota and Covid-19- possible link and implications. Virus Res. 2020, 285, 198018. [Google Scholar] [CrossRef]
- Chen, M.; Shen, W.; Rowan, N.R.; Kulaga, H.; Hillel, A.; Ramanathan, M.; Lane, A.P. Elevated ACE-2 expression in the olfactory neuroepithelium: Implications for anosmia and upper respiratory SARS-CoV-2 entry and replication. Eur. Respir. J. 2020, 56, 2001948. [Google Scholar] [CrossRef] [PubMed]
- Villapol, S. Gastrointestinal symptoms associated with COVID-19: Impact on the gut microbiome. Transl. Res. 2020, 226, 57–69. [Google Scholar] [CrossRef] [PubMed]
- Koester, S.T.; Li, N.; Lachance, D.M.; Morella, N.M.; Dey, N. Variability in digestive and respiratory tract Ace2 expression is associated with the microbiome. PLoS ONE 2021, 16, e0248730. [Google Scholar] [CrossRef] [PubMed]
- Rosshart, S.P.; Vassallo, B.G.; Angeletti, D.; Hutchinson, D.S.; Morgan, A.P.; Takeda, K.; Hickman, H.D.; McCulloch, J.A.; Badger, J.H.; Ajami, N.J.; et al. Wild Mouse Gut Microbiota Promotes Host Fitness and Improves Disease Resistance. Cell 2017, 171, 1015–1028.e13. [Google Scholar] [CrossRef] [PubMed]
- Steed, A.L.; Christophi, G.P.; Kaiko, G.E.; Sun, L.; Goodwin, V.M.; Jain, U.; Esaulova, E.; Artyomov, M.N.; Morales, D.J.; Holtzman, M.J.; et al. The microbial metabolite desaminotyrosine protects from influenza through type I interferon. Science 2017, 357, 498–502. [Google Scholar] [CrossRef] [PubMed]
- Ichinohe, T.; Pang, I.K.; Kumamoto, Y.; Peaper, D.R.; Ho, J.H.; Murray, T.S.; Iwasaki, A. Microbiota regulates immune defense against respiratory tract influenza A virus infection. Proc. Natl. Acad. Sci. USA 2011, 108, 5354–5359. [Google Scholar] [CrossRef] [PubMed]
- Sudo, N.; Aiba, Y.; Oyama, N.; Yu, X.N.; Matsunaga, M.; Koga, Y.; Kubo, C. Dietary nucleic acid and intestinal microbiota synergistically promote a shift in the Th1/Th2 balance toward Th1-skewed immunity. Int. Arch. Allergy Immunol. 2004, 135, 132–135. [Google Scholar] [CrossRef]
- Atarashi, K.; Tanoue, T.; Oshima, K.; Suda, W.; Nagano, Y.; Nishikawa, H.; Fukuda, S.; Saito, T.; Narushima, S.; Hase, K.; et al. Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature 2013, 500, 232–236. [Google Scholar] [CrossRef]
- Huang, Y.; Mao, K.; Chen, X.; Sun, M.A.; Kawabe, T.; Li, W.; Usher, N.; Zhu, J.; Urban, J.F.; Paul, W.E.; et al. S1P-dependent interorgan trafficking of group 2 innate lymphoid cells supports host defense. Science 2018, 359, 114–119. [Google Scholar] [CrossRef]
- Gasteiger, G.; Fan, X.; Dikiy, S.; Lee, S.Y.; Rudensky, A.Y. Tissue residency of innate lymphoid cells in lymphoid and nonlymphoid organs. Science 2015, 350, 981–985. [Google Scholar] [CrossRef]
- Eksteen, B.; Grant, A.J.; Miles, A.; Curbishley, S.M.; Lalor, P.F.; Hübscher, S.G.; Briskin, M.; Salmon, M.; Adams, D.H. Hepatic endothelial CCL25 mediates the recruitment of CCR9+ gut-homing lymphocytes to the liver in primary sclerosing cholangitis. J. Exp. Med. 2004, 200, 1511–1517. [Google Scholar] [CrossRef] [PubMed]
- Biagi, E.; Nylund, L.; Candela, M.; Ostan, R.; Bucci, L.; Pini, E.; Nikkïla, J.; Monti, D.; Satokari, R.; Franceschi, C.; et al. Through ageing, and beyond: Gut microbiota and inflammatory status in seniors and centenarians. PLoS ONE 2010, 5, e10667. [Google Scholar] [CrossRef]
- Verdam, F.J.; Fuentes, S.; de Jonge, C.; Zoetendal, E.G.; Erbil, R.; Greve, J.W.; Buurman, W.A.; de Vos, W.M.; Rensen, S.S. Human intestinal microbiota composition is associated with local and systemic inflammation in obesity. Obesity 2013, 21, E607–E615. [Google Scholar] [CrossRef]
- Guarner, F.; Malagelada, J.R. Gut flora in health and disease. Lancet 2003, 361, 512–519. [Google Scholar] [CrossRef]
- Abt, M.C.; Osborne, L.C.; Monticelli, L.A.; Doering, T.A.; Alenghat, T.; Sonnenberg, G.F.; Paley, M.A.; Antenus, M.; Williams, K.L.; Erikson, J.; et al. Commensal bacteria calibrate the activation threshold of innate antiviral immunity. Immunity 2012, 37, 158–170. [Google Scholar] [CrossRef] [PubMed]
- Grayson, M.H.; Camarda, L.E.; Hussain, S.A.; Zemple, S.J.; Hayward, M.; Lam, V.; Hunter, D.A.; Santoro, J.L.; Rohlfing, M.; Cheung, D.S.; et al. Intestinal Microbiota Disruption Reduces Regulatory T Cells and Increases Respiratory Viral Infection Mortality Through Increased IFNγ Production. Front. Immunol. 2018, 9, 1587. [Google Scholar] [CrossRef]
- Khan, N.; Vidyarthi, A.; Nadeem, S.; Negi, S.; Nair, G.; Agrewala, J.N. Alteration in the Gut Microbiota Provokes Susceptibility to Tuberculosis. Front. Immunol. 2016, 7, 529. [Google Scholar] [CrossRef]
- Negi, S.; Pahari, S.; Bashir, H.; Agrewala, J.N. Gut Microbiota Regulates Mincle Mediated Activation of Lung Dendritic Cells to Protect Against. Front. Immunol. 2019, 10, 1142. [Google Scholar] [CrossRef] [PubMed]
- Dumas, A.; Corral, D.; Colom, A.; Levillain, F.; Peixoto, A.; Hudrisier, D.; Poquet, Y.; Neyrolles, O. The Host Microbiota Contributes to Early Protection against Lung Colonization by. Front. Immunol. 2018, 9, 2656. [Google Scholar] [CrossRef]
- Ngo, V.L.; Lieber, C.M.; Kang, H.J.; Sakamoto, K.; Kuczma, M.; Plemper, R.K.; Gewirtz, A.T. Intestinal microbiota programming of alveolar macrophages influences severity of respiratory viral infection. Cell Host Microbe 2024, 32, 335–348.e8. [Google Scholar] [CrossRef]
- Grijalva, C.G.; Griffin, M.R.; Edwards, K.M.; Williams, J.V.; Gil, A.I.; Verastegui, H.; Hartinger, S.M.; Vidal, J.E.; Klugman, K.P.; Lanata, C.F. The role of influenza and parainfluenza infections in nasopharyngeal pneumococcal acquisition among young children. Clin. Infect. Dis. 2014, 58, 1369–1376. [Google Scholar] [CrossRef] [PubMed]
- Shrestha, S.; Foxman, B.; Weinberger, D.M.; Steiner, C.; Viboud, C.; Rohani, P. Identifying the interaction between influenza and pneumococcal pneumonia using incidence data. Sci. Transl. Med. 2013, 5, 191ra184. [Google Scholar] [CrossRef] [PubMed]
- Sencio, V.; Barthelemy, A.; Tavares, L.P.; Machado, M.G.; Soulard, D.; Cuinat, C.; Queiroz-Junior, C.M.; Noordine, M.L.; Salomé-Desnoulez, S.; Deryuter, L.; 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. [Google Scholar] [CrossRef] [PubMed]
- Cucchiari, D.; Pericàs, J.M.; Riera, J.; Gumucio, R.; Md, E.C.; Nicolás, D.; Team, H.C.H. Pneumococcal superinfection in COVID-19 patients: A series of 5 cases. Med. Clin. (Engl. Ed.) 2020, 155, 502–505. [Google Scholar] [CrossRef] [PubMed]
- Peddu, V.; Shean, R.C.; Xie, H.; Shrestha, L.; Perchetti, G.A.; Minot, S.S.; Roychoudhury, P.; Huang, M.L.; Nalla, A.; Reddy, S.B.; et al. Metagenomic Analysis Reveals Clinical SARS-CoV-2 Infection and Bacterial or Viral Superinfection and Colonization. Clin. Chem. 2020, 66, 966–972. [Google Scholar] [CrossRef] [PubMed]
- Garcia-Vidal, C.; Sanjuan, G.; Moreno-García, E.; Puerta-Alcalde, P.; Garcia-Pouton, N.; Chumbita, M.; Fernandez-Pittol, M.; Pitart, C.; Inciarte, A.; Bodro, M.; et al. Incidence of co-infections and superinfections in hospitalized patients with COVID-19: A retrospective cohort study. Clin. Microbiol. Infect. 2021, 27, 83–88. [Google Scholar] [CrossRef] [PubMed]
- Alexandre, Y.; Le Berre, R.; Barbier, G.; Le Blay, G. Screening of Lactobacillus spp. for the prevention of Pseudomonas aeruginosa pulmonary infections. BMC Microbiol. 2014, 14, 107. [Google Scholar] [CrossRef] [PubMed]
- Trompette, A.; Gollwitzer, E.S.; Pattaroni, C.; Lopez-Mejia, I.C.; Riva, E.; Pernot, J.; Ubags, N.; Fajas, L.; Nicod, L.P.; Marsland, B.J. Dietary Fiber Confers Protection against Flu by Shaping Ly6c. Immunity 2018, 48, 992–1005.e8. [Google Scholar] [CrossRef] [PubMed]
- Teng, C.F.; Hsieh, W.C.; Wu, H.C.; Lin, Y.J.; Tsai, H.W.; Huang, W.; Su, I.J. Hepatitis B Virus Pre-S2 Mutant Induces Aerobic Glycolysis through Mammalian Target of Rapamycin Signal Cascade. PLoS ONE 2015, 10, e0122373. [Google Scholar] [CrossRef]
- Shi, Y.; Yamazaki, T.; Okubo, Y.; Uehara, Y.; Sugane, K.; Agematsu, K. Regulation of aged humoral immune defense against pneumococcal bacteria by IgM memory B cell. J. Immunol. 2005, 175, 3262–3267. [Google Scholar] [CrossRef]
- Waki, N.; Matsumoto, M.; Fukui, Y.; Suganuma, H. Effects of probiotic Lactobacillus brevis KB290 on incidence of influenza infection among schoolchildren: An open-label pilot study. Lett. Appl. Microbiol. 2014, 59, 565–571. [Google Scholar] [CrossRef]
- Ceccarelli, G.; Marazzato, M.; Celani, L.; Lombardi, F.; Piccirilli, A.; Mancone, M.; Trinchieri, V.; Pugliese, F.; Mastroianni, C.M.; d’Ettorre, G. Oxygen Sparing Effect of Bacteriotherapy in COVID-19. Nutrients 2021, 13, 2898. [Google Scholar] [CrossRef] [PubMed]
- Lehtoranta, L.; Latvala, S.; Lehtinen, M.J. Role of Probiotics in Stimulating the Immune System in Viral Respiratory Tract Infections: A Narrative Review. Nutrients 2020, 12, 3163. [Google Scholar] [CrossRef]
- Mahooti, M.; Abdolalipour, E.; Salehzadeh, A.; Mohebbi, S.R.; Gorji, A.; Ghaemi, A. Immunomodulatory and prophylactic effects of Bifidobacterium bifidum probiotic strain on influenza infection in mice. World J. Microbiol. Biotechnol. 2019, 35, 91. [Google Scholar] [CrossRef] [PubMed]
- Wang, Q.; Fang, Z.; Li, L.; Wang, H.; Zhu, J.; Zhang, P.; Lee, Y.K.; Zhao, J.; Zhang, H.; Lu, W.; et al. Lactobacillus mucosae exerted different antiviral effects on respiratory syncytial virus infection in mice. Front. Microbiol. 2022, 13, 1001313. [Google Scholar] [CrossRef]
- Jounai, K.; Sugimura, T.; Ohshio, K.; Fujiwara, D. Oral administration of Lactococcus lactis subsp. lactis JCM5805 enhances lung immune response resulting in protection from murine parainfluenza virus infection. PLoS ONE 2015, 10, e0119055. [Google Scholar] [CrossRef]
- Belkacem, N.; Serafini, N.; Wheeler, R.; Derrien, M.; Boucinha, L.; Couesnon, A.; Cerf-Bensussan, N.; Gomperts Boneca, I.; Di Santo, J.P.; Taha, M.K.; et al. Lactobacillus paracasei feeding improves immune control of influenza infection in mice. PLoS ONE 2017, 12, e0184976. [Google Scholar] [CrossRef]
- Harata, G.; He, F.; Hiruta, N.; Kawase, M.; Kubota, A.; Hiramatsu, M.; Yausi, H. Intranasal administration of Lactobacillus rhamnosus GG protects mice from H1N1 influenza virus infection by regulating respiratory immune responses. Lett. Appl. Microbiol. 2010, 50, 597–602. [Google Scholar] [CrossRef] [PubMed]
- Goto, H.; Sagitani, A.; Ashida, N.; Kato, S.; Hirota, T.; Shinoda, T.; Yamamoto, N. Anti-influenza virus effects of both live and non-live Lactobacillus acidophilus L-92 accompanied by the activation of innate immunity. Br. J. Nutr. 2013, 110, 1810–1818. [Google Scholar] [CrossRef]
- Suez, J.; Zmora, N.; Segal, E.; Elinav, E. The pros, cons, and many unknowns of probiotics. Nat. Med. 2019, 25, 716–729. [Google Scholar] [CrossRef]
- Piqué, N.; Berlanga, M.; Miñana-Galbis, D. Health Benefits of Heat-Killed (Tyndallized) Probiotics: An Overview. Int. J. Mol. Sci. 2019, 20, 2534. [Google Scholar] [CrossRef] [PubMed]
- Antunes, K.H.; Fachi, J.L.; de Paula, R.; da Silva, E.F.; Pral, L.P.; Dos Santos, A.; Dias, G.B.M.; Vargas, J.E.; Puga, R.; Mayer, F.Q.; et al. Microbiota-derived acetate protects against respiratory syncytial virus infection through a GPR43-type 1 interferon response. Nat. Commun. 2019, 10, 3273. [Google Scholar] [CrossRef]
- Antunes, K.H.; Singanayagam, A.; Williams, L.; Faiez, T.S.; Farias, A.; Jackson, M.M.; Faizi, F.K.; Aniscenko, J.; Kebadze, T.; Chander Veerati, P.; et al. Airway-delivered short-chain fatty acid acetate boosts antiviral immunity during rhinovirus infection. J. Allergy Clin. Immunol. 2023, 151, 447–457.e5. [Google Scholar] [CrossRef] [PubMed]
- Luoto, R.; Ruuskanen, O.; Waris, M.; Kalliomäki, M.; Salminen, S.; Isolauri, E. Prebiotic and probiotic supplementation prevents rhinovirus infections in preterm infants: A randomized, placebo-controlled trial. J. Allergy Clin. Immunol. 2014, 133, 405–413. [Google Scholar] [CrossRef] [PubMed]
- Cruz, C.S.; Ricci, M.F.; Vieira, A.T. Gut Microbiota Modulation as a Potential Target for the Treatment of Lung Infections. Front. Pharmacol. 2021, 12, 724033. [Google Scholar] [CrossRef] [PubMed]
- Forestier, C.; Guelon, D.; Cluytens, V.; Gillart, T.; Sirot, J.; De Champs, C. Oral probiotic and prevention of Pseudomonas aeruginosa infections: A randomized, double-blind, placebo-controlled pilot study in intensive care unit patients. Crit. Care 2008, 12, R69. [Google Scholar] [CrossRef] [PubMed]
- Morrow, J.B.; Arango, C.P.; Holbrook, R.D. Association of quantum dot nanoparticles with Pseudomonas aeruginosa biofilm. J. Environ. Qual. 2010, 39, 1934–1941. [Google Scholar] [CrossRef]
- Shimizu, K.; Yamada, T.; Ogura, H.; Mohri, T.; Kiguchi, T.; Fujimi, S.; Asahara, T.; Ojima, M.; Ikeda, M.; Shimazu, T. Synbiotics modulate gut microbiota and reduce enteritis and ventilator-associated pneumonia in patients with sepsis: A randomized controlled trial. Crit. Care 2018, 22, 239. [Google Scholar] [CrossRef]
- Villena, J.; Barbieri, N.; Salva, S.; Herrera, M.; Alvarez, S. Enhanced immune response to pneumococcal infection in malnourished mice nasally treated with heat-killed Lactobacillus casei. Microbiol. Immunol. 2009, 53, 636–646. [Google Scholar] [CrossRef]
- Villena, J.; Racedo, S.; Agüero, G.; Alvarez, S. Yoghurt accelerates the recovery of defence mechanisms against Streptococcus pneumoniae in protein-malnourished mice. Br. J. Nutr. 2006, 95, 591–602. [Google Scholar] [CrossRef]
- Villena, J.; Racedo, S.; Agüero, G.; Bru, E.; Medina, M.; Alvarez, S. Lactobacillus casei improves resistance to pneumococcal respiratory infection in malnourished mice. J. Nutr. 2005, 135, 1462–1469. [Google Scholar] [CrossRef] [PubMed]
- Alvarez, S.; Herrero, C.; Bru, E.; Perdigon, G. Effect of Lactobacillus casei and yogurt administration on prevention of Pseudomonas aeruginosa infection in young mice. J. Food Prot. 2001, 64, 1768–1774. [Google Scholar] [CrossRef] [PubMed]
- Khailova, L.; Baird, C.H.; Rush, A.A.; McNamee, E.N.; Wischmeyer, P.E. Lactobacillus rhamnosus GG improves outcome in experimental Pseudomonas aeruginosa pneumonia: Potential role of regulatory T cells. Shock 2013, 40, 496–503. [Google Scholar] [CrossRef] [PubMed]
- Fangous, M.S.; Alexandre, Y.; Hymery, N.; Gouriou, S.; Arzur, D.; Blay, G.L.; Berre, R.L. Lactobacilli intra-tracheal administration protects from. Benef. Microbes 2019, 10, 893–900. [Google Scholar] [CrossRef] [PubMed]
- Vieira, A.T.; Rocha, V.M.; Tavares, L.; Garcia, C.C.; Teixeira, M.M.; Oliveira, S.C.; Cassali, G.D.; Gamba, C.; Martins, F.S.; Nicoli, J.R. Control of Klebsiella pneumoniae pulmonary infection and immunomodulation by oral treatment with the commensal probiotic Bifidobacterium longum 51A. Microbes Infect. 2016, 18, 180–189. [Google Scholar] [CrossRef] [PubMed]
- Galvão, I.; Tavares, L.P.; Corrêa, R.O.; Fachi, J.L.; Rocha, V.M.; Rungue, M.; Garcia, C.C.; Cassali, G.; Ferreira, C.M.; Martins, F.S.; et al. The Metabolic Sensor GPR43 Receptor Plays a Role in the Control of. Front. Immunol. 2018, 9, 142. [Google Scholar] [CrossRef] [PubMed]
- Chen, L.; Zhang, G.; Li, G.; Wang, W.; Ge, Z.; Yang, Y.; He, X.; Liu, Z.; Zhang, Z.; Mai, Q.; et al. Ifnar gene variants influence gut microbial production of palmitoleic acid and host immune responses to tuberculosis. Nat. Metab. 2022, 4, 359–373. [Google Scholar] [CrossRef] [PubMed]
- Kanmani, P.; Clua, P.; Vizoso-Pinto, M.G.; Rodriguez, C.; Alvarez, S.; Melnikov, V.; Takahashi, H.; Kitazawa, H.; Villena, J. Respiratory Commensal Bacteria. Front. Microbiol. 2017, 8, 1613. [Google Scholar] [CrossRef]
- Chotirmall, S.H.; Bogaert, D.; Chalmers, J.D.; Cox, M.J.; Hansbro, P.M.; Huang, Y.J.; Molyneaux, P.L.; O’Dwyer, D.N.; Pragman, A.A.; Rogers, G.B.; et al. Therapeutic Targeting of the Respiratory Microbiome. Am. J. Respir. Crit. Care Med. 2022, 206, 535–544. [Google Scholar] [CrossRef]
- Dashiff, A.; Junka, R.A.; Libera, M.; Kadouri, D.E. Predation of human pathogens by the predatory bacteria Micavibrio aeruginosavorus and Bdellovibrio bacteriovorus. J. Appl. Microbiol. 2011, 110, 431–444. [Google Scholar] [CrossRef]
- Shatzkes, K.; Singleton, E.; Tang, C.; Zuena, M.; Shukla, S.; Gupta, S.; Dharani, S.; Rinaggio, J.; Kadouri, D.E.; Connell, N.D. Examining the efficacy of intravenous administration of predatory bacteria in rats. Sci. Rep. 2017, 7, 1864. [Google Scholar] [CrossRef] [PubMed]
- Shatzkes, K.; Singleton, E.; Tang, C.; Zuena, M.; Shukla, S.; Gupta, S.; Dharani, S.; Onyile, O.; Rinaggio, J.; Connell, N.D.; et al. Predatory Bacteria Attenuate Klebsiella pneumoniae Burden in Rat Lungs. mBio 2016, 7, e01847-16. [Google Scholar] [CrossRef] [PubMed]
- Esposito, S.; Amirthalingam, G.; Bassetti, M.; Blasi, F.; De Rosa, F.G.; Halasa, N.B.; Hung, I.; Osterhaus, A.; Tan, T.; Torres, J.P.; et al. Monoclonal antibodies for prophylaxis and therapy of respiratory syncytial virus, SARS-CoV-2, human immunodeficiency virus, rabies and bacterial infections: An update from the World Association of Infectious Diseases and Immunological Disorders and the Italian Society of Antinfective Therapy. Front. Immunol. 2023, 14, 1162342. [Google Scholar] [CrossRef] [PubMed]
- DiGiandomenico, A.; Keller, A.E.; Gao, C.; Rainey, G.J.; Warrener, P.; Camara, M.M.; Bonnell, J.; Fleming, R.; Bezabeh, B.; Dimasi, N.; et al. A multifunctional bispecific antibody protects against Pseudomonas aeruginosa. Sci. Transl. Med. 2014, 6, 262ra155. [Google Scholar] [CrossRef] [PubMed]
- Zurawski, D.V.; McLendon, M.K. Monoclonal Antibodies as an Antibacterial Approach Against Bacterial Pathogens. Antibiotics 2020, 9, 155. [Google Scholar] [CrossRef] [PubMed]
- Tabor, D.E.; Oganesyan, V.; Keller, A.E.; Yu, L.; McLaughlin, R.E.; Song, E.; Warrener, P.; Rosenthal, K.; Esser, M.; Qi, Y.; et al. Pseudomonas aeruginosa PcrV and Psl, the Molecular Targets of Bispecific Antibody MEDI3902, Are Conserved Among Diverse Global Clinical Isolates. J. Infect. Dis. 2018, 218, 1983–1994. [Google Scholar] [CrossRef] [PubMed]
- Thanabalasuriar, A.; Surewaard, B.G.; Willson, M.E.; Neupane, A.S.; Stover, C.K.; Warrener, P.; Wilson, G.; Keller, A.E.; Sellman, B.R.; DiGiandomenico, A.; et al. Bispecific antibody targets multiple Pseudomonas aeruginosa evasion mechanisms in the lung vasculature. J. Clin. Investig. 2017, 127, 2249–2261. [Google Scholar] [CrossRef]
- Aguiar-Alves, F.; Le, H.N.; Tran, V.G.; Gras, E.; Vu, T.T.T.; Dong, O.X.; Quetz, J.S.; Cheng, L.I.; Yu, L.; Sellman, B.R.; et al. Antivirulence Bispecific Monoclonal Antibody-Mediated Protection against Pseudomonas aeruginosa Ventilator-Associated Pneumonia in a Rabbit Model. Antimicrob. Agents Chemother. 2022, 66, e0202221. [Google Scholar] [CrossRef]
- Que, Y.A.; Lazar, H.; Wolff, M.; François, B.; Laterre, P.F.; Mercier, E.; Garbino, J.; Pagani, J.L.; Revelly, J.P.; Mus, E.; et al. Assessment of panobacumab as adjunctive immunotherapy for the treatment of nosocomial Pseudomonas aeruginosa pneumonia. Eur. J. Clin. Microbiol. Infect. Dis. 2014, 33, 1861–1867. [Google Scholar] [CrossRef]
- Secher, T.; Fauconnier, L.; Szade, A.; Rutschi, O.; Fas, S.C.; Ryffel, B.; Rudolf, M.P. Anti-Pseudomonas aeruginosa serotype O11 LPS immunoglobulin M monoclonal antibody panobacumab (KBPA101) confers protection in a murine model of acute lung infection. J. Antimicrob. Chemother. 2011, 66, 1100–1109. [Google Scholar] [CrossRef]
- Tabor, D.E.; Yu, L.; Mok, H.; Tkaczyk, C.; Sellman, B.R.; Wu, Y.; Oganesyan, V.; Slidel, T.; Jafri, H.; McCarthy, M.; et al. Staphylococcus aureus Alpha-Toxin Is Conserved among Diverse Hospital Respiratory Isolates Collected from a Global Surveillance Study and Is Neutralized by Monoclonal Antibody MEDI4893. Antimicrob. Agents Chemother. 2016, 60, 5312–5321. [Google Scholar] [CrossRef]
- Hua, L.; Hilliard, J.J.; Shi, Y.; Tkaczyk, C.; Cheng, L.I.; Yu, X.; Datta, V.; Ren, S.; Feng, H.; Zinsou, R.; et al. Assessment of an anti-alpha-toxin monoclonal antibody for prevention and treatment of Staphylococcus aureus-induced pneumonia. Antimicrob. Agents Chemother. 2014, 58, 1108–1117. [Google Scholar] [CrossRef] [PubMed]
- Kollef, M.H.; Betthauser, K.D. Monoclonal antibodies as antibacterial therapies: Thinking outside of the box. Lancet Infect. Dis. 2021, 21, 1201–1202. [Google Scholar] [CrossRef]
- Lin, D.M.; Koskella, B.; Lin, H.C. Phage therapy: An alternative to antibiotics in the age of multi-drug resistance. World J. Gastrointest. Pharmacol. Ther. 2017, 8, 162–173. [Google Scholar] [CrossRef]
- Forti, F.; Roach, D.R.; Cafora, M.; Pasini, M.E.; Horner, D.S.; Fiscarelli, E.V.; Rossitto, M.; Cariani, L.; Briani, F.; Debarbieux, L.; et al. Design of a Broad-Range Bacteriophage Cocktail That Reduces Pseudomonas aeruginosa Biofilms and Treats Acute Infections in Two Animal Models. Antimicrob. Agents Chemother. 2018, 62, e02573-17. [Google Scholar] [CrossRef] [PubMed]
- Singla, S.; Harjai, K.; Katare, O.P.; Chhibber, S. Bacteriophage-loaded nanostructured lipid carrier: Improved pharmacokinetics mediates effective resolution of Klebsiella pneumoniae-induced lobar pneumonia. J. Infect. Dis. 2015, 212, 325–334. [Google Scholar] [CrossRef]
- Maddocks, S.; Fabijan, A.P.; Ho, J.; Lin, R.C.Y.; Ben Zakour, N.L.; Dugan, C.; Kliman, I.; Branston, S.; Morales, S.; Iredell, J.R. Bacteriophage Therapy of Ventilator-associated Pneumonia and Empyema Caused by. Am. J. Respir. Crit. Care Med. 2019, 200, 1179–1181. [Google Scholar] [CrossRef] [PubMed]
- Witzenrath, M.; Schmeck, B.; Doehn, J.M.; Tschernig, T.; Zahlten, J.; Loeffler, J.M.; Zemlin, M.; Müller, H.; Gutbier, B.; Schütte, H.; et al. Systemic use of the endolysin Cpl-1 rescues mice with fatal pneumococcal pneumonia. Crit. Care Med. 2009, 37, 642–649. [Google Scholar] [CrossRef] [PubMed]
- Raz, A.; Serrano, A.; Hernandez, A.; Euler, C.W.; Fischetti, V.A. Isolation of Phage Lysins That Effectively Kill Pseudomonas aeruginosa in Mouse Models of Lung and Skin Infection. Antimicrob. Agents Chemother. 2019, 63, e00024-19. [Google Scholar] [CrossRef]
- Khan, N.; Patel, D.; Trivedi, C.; Pernes, T.; Kavani, H.; Xie, D.; Yang, Y.X. The impact of IBD medications on risk of pneumonia and pneumonia-related hospitalisation: A nationwide cohort study of 56,410 IBD patients. Aliment. Pharmacol. Ther. 2022, 55, 64–72. [Google Scholar] [CrossRef]
- Kusakabe, T.; Lin, W.Y.; Cheong, J.G.; Singh, G.; Ravishankar, A.; Yeung, S.T.; Mesko, M.; DeCelie, M.B.; Carriche, G.; Zhao, Z.; et al. Fungal microbiota sustains lasting immune activation of neutrophils and their progenitors in severe COVID-19. Nat. Immunol. 2023, 24, 1879–1889. [Google Scholar] [CrossRef] [PubMed]
- Mindt, B.C.; DiGiandomenico, A. Microbiome Modulation as a Novel Strategy to Treat and Prevent Respiratory Infections. Antibiotics 2022, 11, 474. [Google Scholar] [CrossRef] [PubMed]
- Ashley, S.L.; Sjoding, M.W.; Popova, A.P.; Cui, T.X.; Hoostal, M.J.; Schmidt, T.M.; Branton, W.R.; Dieterle, M.G.; Falkowski, N.R.; Baker, J.M.; et al. Lung and gut microbiota are altered by hyperoxia and contribute to oxygen-induced lung injury in mice. Sci. Transl. Med. 2020, 12, eaau9959. [Google Scholar] [CrossRef] [PubMed]
- Dickson, R.P.; Erb-Downward, J.R.; Falkowski, N.R.; Hunter, E.M.; Ashley, S.L.; Huffnagle, G.B. The Lung Microbiota of Healthy Mice Are Highly Variable, Cluster by Environment, and Reflect Variation in Baseline Lung Innate Immunity. Am. J. Respir. Crit. Care Med. 2018, 198, 497–508. [Google Scholar] [CrossRef]
- Urban-Chmiel, R.; Marek, A.; Stępień-Pyśniak, D.; Wieczorek, K.; Dec, M.; Nowaczek, A.; Osek, J. Antibiotic Resistance in Bacteria-A Review. Antibiotics 2022, 11, 1079. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Marrella, V.; Nicchiotti, F.; Cassani, B. Microbiota and Immunity during Respiratory Infections: Lung and Gut Affair. Int. J. Mol. Sci. 2024, 25, 4051. https://doi.org/10.3390/ijms25074051
Marrella V, Nicchiotti F, Cassani B. Microbiota and Immunity during Respiratory Infections: Lung and Gut Affair. International Journal of Molecular Sciences. 2024; 25(7):4051. https://doi.org/10.3390/ijms25074051
Chicago/Turabian StyleMarrella, Veronica, Federico Nicchiotti, and Barbara Cassani. 2024. "Microbiota and Immunity during Respiratory Infections: Lung and Gut Affair" International Journal of Molecular Sciences 25, no. 7: 4051. https://doi.org/10.3390/ijms25074051
APA StyleMarrella, V., Nicchiotti, F., & Cassani, B. (2024). Microbiota and Immunity during Respiratory Infections: Lung and Gut Affair. International Journal of Molecular Sciences, 25(7), 4051. https://doi.org/10.3390/ijms25074051