DOI QR코드

DOI QR Code

A Moonlighting Protein Secreted by a Nasal Microbiome Fortifies the Innate Host Defense Against Bacterial and Viral Infections

  • Gwanghee Kim (Department of Microbiology and Immunology, Yonsei University College of Medicine) ;
  • Yoojin Lee (Department of Microbiology and Immunology, Yonsei University College of Medicine) ;
  • Jin Sun You (Department of Microbiology and Immunology, Yonsei University College of Medicine) ;
  • Wontae Hwang (Department of Microbiology and Immunology, Yonsei University College of Medicine) ;
  • Jeewon Hwang (Department of Microbiology and Immunology, Yonsei University College of Medicine) ;
  • Hwa Young Kim (Department of Microbiology and Immunology, Yonsei University College of Medicine) ;
  • Jieun Kim (Department of Microbiology and Immunology, Yonsei University College of Medicine) ;
  • Ara Jo (Department of Otorhinolaryngology, Seoul National University College of Medicine) ;
  • In ho Park (Department of Microbiology and Immunology, Yonsei University College of Medicine) ;
  • Mohammed Ali (Department of Microbiology and Immunology, Yonsei University College of Medicine) ;
  • Jongsun Kim (Department of Microbiology and Immunology, Yonsei University College of Medicine) ;
  • Jeon-Soo Shin (Department of Microbiology and Immunology, Yonsei University College of Medicine) ;
  • Ho-Keun Kwon (Department of Microbiology and Immunology, Yonsei University College of Medicine) ;
  • Hyun Jik Kim (Department of Otorhinolaryngology, Seoul National University College of Medicine) ;
  • Sang Sun Yoon (Department of Microbiology and Immunology, Yonsei University College of Medicine)
  • Received : 2023.05.02
  • Accepted : 2023.06.19
  • Published : 2023.08.31

Abstract

Evidence suggests that the human respiratory tract, as with the gastrointestinal tract, has evolved to its current state in association with commensal microbes. However, little is known about how the airway microbiome affects the development of airway immune system. Here, we uncover a previously unidentified mode of interaction between host airway immunity and a unique strain (AIT01) of Staphylococcus epidermidis, a predominant species of the nasal microbiome. Intranasal administration of AIT01 increased the population of neutrophils and monocytes in mouse lungs. The recruitment of these immune cells resulted in the protection of the murine host against infection by Pseudomonas aeruginosa, a pathogenic bacterium. Interestingly, an AIT01-secreted protein identified as GAPDH, a well-known bacterial moonlighting protein, mediated this protective effect. Intranasal delivery of the purified GAPDH conferred significant resistance against other Gram-negative pathogens (Klebsiella pneumoniae and Acinetobacter baumannii) and influenza A virus. Our findings demonstrate the potential of a native nasal microbe and its secretory protein to enhance innate immune defense against airway infections. These results offer a promising preventive measure, particularly relevant in the context of global pandemics.

Keywords

Acknowledgement

This work was supported by grants from the National Research Foundation of Korea (NRF) grant funded by the Korean Government (2022R1A2C1091845 and 2022M3A9F3017506). This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT) (2022R1A2C2011867 and awarded to HJK). This research was also supported by a grant from the Korean Health Technology Research and Development Project through the Korean Health Industry Development Institute, funded by the Ministry of Health and Welfare of the Republic of Korea (HI23C0795 awarded to HJK). This research was supported by the National Research Foundation of Korea (NRF-2022M3A9I2017587).

References

  1. Ruff WE, Greiling TM, Kriegel MA. Host-microbiota interactions in immune-mediated diseases. Nat Rev Microbiol 2020;18:521-538. https://doi.org/10.1038/s41579-020-0367-2
  2. Zheng D, Liwinski T, Elinav E. Interaction between microbiota and immunity in health and disease. Cell Res 2020;30:492-506. https://doi.org/10.1038/s41422-020-0332-7
  3. Belkaid Y, Hand TW. Role of the microbiota in immunity and inflammation. Cell 2014;157:121-141. https://doi.org/10.1016/j.cell.2014.03.011
  4. Littman DR, Pamer EG. Role of the commensal microbiota in normal and pathogenic host immune responses. Cell Host Microbe 2011;10:311-323.  https://doi.org/10.1016/j.chom.2011.10.004
  5. Hill DA, Artis D. Intestinal bacteria and the regulation of immune cell homeostasis. Annu Rev Immunol 2010;28:623-667. https://doi.org/10.1146/annurev-immunol-030409-101330
  6. Becattini S, Taur Y, Pamer EG. Antibiotic-induced changes in the intestinal microbiota and disease. Trends Mol Med 2016;22:458-478. https://doi.org/10.1016/j.molmed.2016.04.003
  7. Dethlefsen L, Relman DA. Incomplete recovery and individualized responses of the human distal gut microbiota to repeated antibiotic perturbation. Proc Natl Acad Sci U S A 2011;108 Suppl 1:4554-4561. https://doi.org/10.1073/pnas.1000087107
  8. You JS, Yong JH, Kim GH, Moon S, Nam KT, Ryu JH, Yoon MY, Yoon SS. Commensal-derived metabolites govern Vibrio cholerae pathogenesis in host intestine. Microbiome 2019;7:132.
  9. Khosravi A, Yanez A, Price JG, Chow A, Merad M, Goodridge HS, Mazmanian SK. Gut microbiota promote hematopoiesis to control bacterial infection. Cell Host Microbe 2014;15:374-381. https://doi.org/10.1016/j.chom.2014.02.006
  10. McKenney PT, Pamer EG. From hype to hope: the gut microbiota in enteric infectious disease. Cell 2015;163:1326-1332. https://doi.org/10.1016/j.cell.2015.11.032
  11. Parlet CP, Brown MM, Horswill AR. Commensal staphylococci influence Staphylococcus aureus skin colonization and disease. Trends Microbiol 2019;27:497-507. https://doi.org/10.1016/j.tim.2019.01.008
  12. Nakatsuji T, Chen TH, Narala S, Chun KA, Two AM, Yun T, Shafiq F, Kotol PF, Bouslimani A, Melnik AV, et al. Antimicrobials from human skin commensal bacteria protect against Staphylococcus aureus and are deficient in atopic dermatitis. Sci Transl Med 2017;9:eaah4680.
  13. Khan R, Petersen FC, Shekhar S. Commensal bacteria: an emerging player in defense against respiratory pathogens. Front Immunol 2019;10:1203.
  14. Brown RL, Sequeira RP, Clarke TB. The microbiota protects against respiratory infection via GM-CSF signaling. Nat Commun 2017;8:1512.
  15. Ichinohe T, Pang IK, Kumamoto Y, Peaper DR, Ho JH, Murray TS, Iwasaki A. Microbiota regulates immune defense against respiratory tract influenza A virus infection. Proc Natl Acad Sci U S A 2011;108:5354-5359. https://doi.org/10.1073/pnas.1019378108
  16. Zanin M, Baviskar P, Webster R, Webby R. The interaction between respiratory pathogens and mucus. Cell Host Microbe 2016;19:159-168. https://doi.org/10.1016/j.chom.2016.01.001
  17. Kim HJ, Jo A, Jeon YJ, An S, Lee KM, Yoon SS, Choi JY. Nasal commensal Staphylococcus epidermidis enhances interferon-λ-dependent immunity against influenza virus. Microbiome 2019;7:80.
  18. Liu Q, Liu Q, Meng H, Lv H, Liu Y, Liu J, Wang H, He L, Qin J, Wang Y, et al. Staphylococcus epidermidis contributes to healthy maturation of the nasal microbiome by stimulating antimicrobial peptide production. Cell Host Microbe 2020;27:68-78.e5. https://doi.org/10.1016/j.chom.2019.11.003
  19. Iwase T, Uehara Y, Shinji H, Tajima A, Seo H, Takada K, Agata T, Mizunoe Y. Staphylococcus epidermidis Esp inhibits Staphylococcus aureus biofilm formation and nasal colonization. Nature 2010;465:346-349. https://doi.org/10.1038/nature09074
  20. Cogen AL, Yamasaki K, Sanchez KM, Dorschner RA, Lai Y, MacLeod DT, Torpey JW, Otto M, Nizet V, Kim JE, et al. Selective antimicrobial action is provided by phenol-soluble modulins derived from Staphylococcus epidermidis, a normal resident of the skin. J Invest Dermatol 2010;130:192-200. https://doi.org/10.1038/jid.2009.243
  21. Zipperer A, Konnerth MC, Laux C, Berscheid A, Janek D, Weidenmaier C, Burian M, Schilling NA, Slavetinsky C, Marschal M, et al. Human commensals producing a novel antibiotic impair pathogen colonization. Nature 2016;535:511-516. https://doi.org/10.1038/nature18634
  22. Laborel-Preneron E, Bianchi P, Boralevi F, Lehours P, Fraysse F, Morice-Picard F, Sugai M, Sato'o Y, Badiou C, Lina G, et al. Effects of the Staphylococcus aureus and Staphylococcus epidermidis secretomes isolated from the skin microbiota of atopic children on CD4+ T cell activation. PLoS One 2015;10:e0141067.
  23. Henderson B, Martin A. Bacterial virulence in the moonlight: multitasking bacterial moonlighting proteins are virulence determinants in infectious disease. Infect Immun 2011;79:3476-3491. https://doi.org/10.1128/IAI.00179-11
  24. Sun X, Wang J, Zhou J, Wang H, Wang X, Wu J, He Y, Yin Y, Zhang X, Xu W. Subcutaneous immunization with Streptococcus pneumoniae GAPDH confers effective protection in mice via TLR2 and TLR4. Mol Immunol 2017;83:1-12. https://doi.org/10.1016/j.molimm.2017.01.002
  25. Elhaik Goldman S, Dotan S, Talias A, Lilo A, Azriel S, Malka I, Portnoi M, Ohayon A, Kafka D, Ellis R, et al. Streptococcus pneumoniae fructose-1,6-bisphosphate aldolase, a protein vaccine candidate, elicits Th1/Th2/Th17-type cytokine responses in mice. Int J Mol Med 2016;37:1127-1138. https://doi.org/10.3892/ijmm.2016.2512
  26. Feng Y, Pan X, Sun W, Wang C, Zhang H, Li X, Ma Y, Shao Z, Ge J, Zheng F, et al. Streptococcus suis enolase functions as a protective antigen displayed on the bacterial cell surface. J Infect Dis 2009;200:1583-1592. https://doi.org/10.1086/644602
  27. Nielsen KL, Olsen MH, Palleja A, Ebdrup SR, Sorensen N, Lukjancenko O, Marvig RL, Moller K, Frimodt-Moller N, Hertz FB. Microbiome compositions and resistome levels after antibiotic treatment of critically ill patients: an observational cohort study. Microorganisms 2021;9:2542.
  28. Ayeni FA, Andersen C, Norskov-Lauritsen N. Comparison of growth on mannitol salt agar, matrix-assisted laser desorption/ionization time-of-flight mass spectrometry, VITEK® 2 with partial sequencing of 16S rRNA gene for identification of coagulase-negative staphylococci. Microb Pathog 2017;105:255-259. https://doi.org/10.1016/j.micpath.2017.02.034
  29. An S, Jeon YJ, Jo A, Lim HJ, Han YE, Cho SW, Kim HY, Kim HJ. Initial influenza virus replication can be limited in allergic asthma through rapid induction of type III interferons in respiratory epithelium. Front Immunol 2018;9:986.
  30. Olson ME, Todd DA, Schaeffer CR, Paharik AE, Van Dyke MJ, Buttner H, Dunman PM, Rohde H, Cech NB, Fey PD, et al. Staphylococcus epidermidis agr quorum-sensing system: signal identification, cross talk, and importance in colonization. J Bacteriol 2014;196:3482-3493. https://doi.org/10.1128/JB.01882-14
  31. Camp JV, Jonsson CB. A role for neutrophils in viral respiratory disease. Front Immunol 2017;8:550.
  32. Seo SH, Webster RG. Tumor necrosis factor alpha exerts powerful anti-influenza virus effects in lung epithelial cells. J Virol 2002;76:1071-1076. https://doi.org/10.1128/JVI.76.3.1071-1076.2002
  33. Zhou W, Spoto M, Hardy R, Guan C, Fleming E, Larson PJ, Brown JS, Oh J. Host-specific evolutionary and transmission dynamics shape the functional diversification of Staphylococcus epidermidis in human skin. Cell 2020;180:454-470.e18. https://doi.org/10.1016/j.cell.2020.01.006
  34. Tan L, Li SR, Jiang B, Hu XM, Li S. Therapeutic targeting of the Staphylococcus aureus accessory gene regulator (agr) system. Front Microbiol 2018;9:55.
  35. Henderson B, Martin AC. Protein moonlighting: a new factor in biology and medicine. Biochem Soc Trans 2014;42:1671-1678. https://doi.org/10.1042/BST20140273
  36. Netea M. Trained immunity: a memory for innate host defense. Eur J Clin Invest 2019;49:26. 
  37. Ciarlo E, Heinonen T, Theroude C, Asgari F, Le Roy D, Netea MG, Roger T. Trained immunity confers broad-spectrum protection against bacterial infections. J Infect Dis 2020;222:1869-1881. https://doi.org/10.1093/infdis/jiz692
  38. Shi C, Hohl TM, Leiner I, Equinda MJ, Fan X, Pamer EG. Ly6G+ neutrophils are dispensable for defense against systemic Listeria monocytogenes infection. J Immunol 2011;187:5293-5298. https://doi.org/10.4049/jimmunol.1101721
  39. Moorlag SJ, Rodriguez-Rosales YA, Gillard J, Fanucchi S, Theunissen K, Novakovic B, de Bont CM, Negishi Y, Fok ET, Kalafati L, et al. BCG vaccination induces long-term functional reprogramming of human neutrophils. Cell Reports 2020;33:108387.
  40. Kalafati L, Kourtzelis I, Schulte-Schrepping J, Li X, Hatzioannou A, Grinenko T, Hagag E, Sinha A, Has C, Dietz S, et al. Innate immune training of granulopoiesis promotes anti-tumor activity. Cell 2020;183:771-785.e12. https://doi.org/10.1016/j.cell.2020.09.058