1887

Abstract

Finding a safe innate immune response stimulator is one of the greatest challenges facing immunologists and vaccine manufacturers.

The role of sterile bacterial secretions (SBSs) of in stimulating the innate immune response was not investigated previously.

The comparative effect of SBSs and bacterial cells of isolates isolated from freshwater (PAE) and infected wounds (PAC) on the respiratory tract innate immune response.

Four test mice groups were instilled intranasally (i.n.) with 10 c.f.u of PAC, 10 c.f.u of PAE, SBS of PAC, and SBS of PAE. Two control groups were given i.n. either LB broth or PBS. Time-course changes in IL-1 beta mRNA, TNF-alpha mRNA, IL-1β and TNF-α, leukocyte count, bacterial uptake, and intracellular bacterial killing by mouse alveolar macrophages (AMs) and histological changes were examined. Lung bacterial burdens were counted in first and second test groups.

The maximum level of IL-1β was seen as early as 2 h (1360±180 pg ml) post-instillation (i.n.) with SBS of PAC and 1 h (1910±244 pgml) post-instillation with SBS of PAE. The maximum level of TNF-α was seen as early as 4 h (953±192 pg ml) post-instillation with SBS of PAC and (1197±298 pg ml) post-instillation with SBS of PAE. These values were almost in line with IL-1β and TNF-α gene expression. Moderate infiltration of leukocytes in bronchoalveolar lavage (BAL) and lung sections and moderate activity of AMs (bacterial uptake and bacterial killing) were observed. The above innate immune response parameters in mice instilled i.n. with PAC and PAE were higher (<0.05) than in the mice groups instilled i.n. with SBSs. The PAC was persistent in the lungs of mice for up to 72 h (3.5±0.22 log of c.f.u. g) and up to 48 h (2.05±0.21 log of c.f.u. g) for PAE.

The administration of mice with SBS i.n. stimulates cellular and molecular arms of the innate immune response in the respiratory tract, opening the door to the possibility of using SBS of as an adjuvant.

Loading

Article metrics loading...

/content/journal/jmm/10.1099/jmm.0.001588
2022-10-26
2024-05-02
Loading full text...

Full text loading...

References

  1. Lyczak JB, Cannon CL, Pier GB. Lung infections associated with cystic fibrosis. Clin Microbiol Rev 2002; 15:194–222 [View Article]
    [Google Scholar]
  2. Govan JR, Deretic V. Microbial pathogenesis in cystic fibrosis: mucoid Pseudomonas aeruginosa and Burkholderia cepacia. Microbiol Rev 1996; 60:539–574 [View Article]
    [Google Scholar]
  3. Jones AM, Horsley A, Denning DW. What is the importance of classifying Aspergillus disease in cystic fibrosis patients?. Expert Rev Respir Med 2014; 8:389–392 [View Article]
    [Google Scholar]
  4. Vives-Flórez M, Garnica D. Comparison of virulence between clinical and environmental Pseudomonas aeruginosa isolates. Int Microbiol 2006; 4:247–252
    [Google Scholar]
  5. Winstanley C, O’Brien S, Brockhurst MA. Pseudomonas aeruginosa evolutionary adaptation and diversification in cystic fibrosis chronic lung infections. Trends Microbiol 2016; 24:327–337 [View Article]
    [Google Scholar]
  6. Wang Y, Cela E, Gagnon S, Sweezey NB. Estrogen aggravates inflammation in Pseudomonas aeruginosa pneumonia in cystic fibrosis mice. Respir Res 2010; 11:166 [View Article]
    [Google Scholar]
  7. Maurice NM, Bedi B, Sadikot RT. Pseudomonas aeruginosa Biofilms: host response and clinical implications in Lung infections. Am J Respir Cell Mol Biol 2018; 58:428–439 [View Article] [PubMed]
    [Google Scholar]
  8. Moser C, Pedersen HT, Lerche CJ, Kolpen M, Line L et al. Biofilms and host response - helpful or harmful. APMIS 2017; 125:320–338 [View Article] [PubMed]
    [Google Scholar]
  9. Shen Z, Fang L, Zhao L, Lei H. β-defensin 2 ameliorates lung injury caused by Pseudomonas infection and regulates proinflammatory and anti-inflammatory cytokines in rat. Int J Mol Sci 2014; 15:13372–13387 [View Article] [PubMed]
    [Google Scholar]
  10. Xia X, Lin Z, Shao K, Wang X, Xu J et al. Combination of white tea and peppermint demonstrated synergistic antibacterial and anti-inflammatory activities. J Sci Food Agric 2021; 101:2500–2510 [View Article] [PubMed]
    [Google Scholar]
  11. Esoda CN, Kuehn MJ. Pseudomonas aeruginosa leucine aminopeptidase influences early iofilm Composition and Structure via Vesicle-Associated Antibiofilm Activity. mBio 2019; 10:e02548-19 [View Article]
    [Google Scholar]
  12. Ghafil JA. Immobilization of chitinase improves ability of enzyme to hydrolyze chitin. World J Exp Biosci 2013; 1:33–36
    [Google Scholar]
  13. Al-Hinai AH, Al-Sadi AM, Al-Bahry SN, Mothershaw AS, Al-Said FA. Isolation and characterization of Pseudomonas aeruginosa with antagonistic activity against pythium aphanidermatum. J Plant Pathol 2010; 92:653–660
    [Google Scholar]
  14. Funke G, Monnet D, deBernardis C, von Graevenitz A, Freney J. Evaluation of the VITEK 2 system for rapid identification of medically relevant gram-negative rods. J Clin Microbiol 1998; 36:1948–1952 [View Article]
    [Google Scholar]
  15. Zgair AK, Chhibber S. Stenotrophomonas maltophilia flagellin induces a compartmentalized innate immune response in mouse lung. J Med Microbiol 2010; 59:913–919 [View Article]
    [Google Scholar]
  16. Moors MA, Li L, Mizel SB. Activation of interleukin-1 receptor-associated kinase by gram-negative flagellin. Infect Immun 2001; 69:4424–4429 [View Article] [PubMed]
    [Google Scholar]
  17. Subhi IM, Zgair AK. Estimation of levels of interleukin-1 beta and interleukin-10 in sera of some iraqi patients with chronic rheumatoid arthritis. Iraqi J Sci 2018; 59:1554–1559
    [Google Scholar]
  18. Kagari T, Doi H, Shimozato T. The importance of IL-1 beta and TNF-alpha, and the noninvolvement of IL-6, in the development of monoclonal antibody-induced arthritis. J Immunol 2002; 169:1459–1466 [View Article] [PubMed]
    [Google Scholar]
  19. Liu DF, Wei W, Song LH. Upregulation of TNF-alpha and IL-6 mRNA in mouse liver induced by bacille Calmette-Guerin plus lipopolysaccharide. Acta Pharmacol Sin 2006; 27:460–468 [View Article] [PubMed]
    [Google Scholar]
  20. Allen PM, Fisher D, Saunders JR, Hart CA. The role of capsular polysaccharide K21b of Klebsiella and of the structurally related colanic-acid polysaccharide of Escherichia coli in resistance to phagocytosis and serum killing. J Med Microbiol 1987; 24:363–370 [View Article]
    [Google Scholar]
  21. Zgair AK, Chhibber S. Stenotrophomonas maltophilia flagellin restricts bacterial colonization in BALB/c mouse lung in vivo. FEMS Immunol Med Microbiol 2012; 66:191–200 [View Article]
    [Google Scholar]
  22. Uderhardt S, Martins AJ, Tsang JS, Lämmermann T, Germain RN. Resident Macrophages Cloak Tissue Microlesions to Prevent Neutrophil-Driven Inflammatory Damage. Cell 2019; 177:541–555 [View Article]
    [Google Scholar]
  23. Hogardt M, Heesemann J. Adaptation of Pseudomonas aeruginosa during persistence in the cystic fibrosis lung. Int J Med Microbiol 2010; 300:557–562 [View Article]
    [Google Scholar]
  24. Schick A, Kassen R. Rapid diversification of Pseudomonas aeruginosa in cystic fibrosis lung-like conditions. Proc Natl Acad Sci USA 2018; 115:10714–10719 [View Article]
    [Google Scholar]
  25. Olszowiec-Chlebna M, Koniarek-Maniecka A, Brzozowska A, Błauż A, Rychlik B et al. Vitamin D inhibits pro-inflammatory cytokines in the airways of cystic fibrosis patients infected by Pseudomonas aeruginosa- pilot study. Ital J Pediatr 2019; 45:41 [View Article]
    [Google Scholar]
  26. Lou T-L, Ji T, Peng X, Ji W-W, Yuan L-X et al. Extract from Tetrastigma hemsleyanum Leaf Alleviates Pseudomonas aeruginosa Lung infection: network pharmacology analysis and experimental evidence. Front Pharmacol 2021; 12:12 [View Article]
    [Google Scholar]
  27. Isopi E, Mattoscio D, Codagnone M, Mari VC, Lamolinara A et al. Resolvin D1 reduces Lung infection and Inflammation Activating Resolution in Cystic Fibrosis. Front Immunol 2020; 11:581 [View Article]
    [Google Scholar]
  28. Jain-Vora S, LeVine AM, Chroneos Z, Ross GF, Hull WM et al. Interleukin-4 enhances pulmonary clearance of Pseudomonas aeruginosa. Infect Immun 1998; 66:4229–4236 [View Article]
    [Google Scholar]
  29. Glasser SW, Senft AP, Whitsett JA, Maxfield MD, Ross GF et al. Macrophage dysfunction and susceptibility to pulmonary Pseudomonas aeruginosa infection in surfactant protein C-deficient mice. J Immunol 2008; 181:621–628 [View Article] [PubMed]
    [Google Scholar]
  30. Hector A, Kröner C, Carevic M, Bakele M, Rieber N et al. The chemokine CCL18 characterises Pseudomonas infections in cystic fibrosis lung disease. Eur Respir J 2014; 44:1608–1615 [View Article] [PubMed]
    [Google Scholar]
  31. Zeng M, Sang W, Chen S, Chen R, Zhang H et al. 4-PBA inhibits LPS-induced inflammation through regulating ER stress and autophagy in acute lung injury models. Toxicol Lett 2017; 271:26–37 [View Article]
    [Google Scholar]
  32. Evers A, Atanasova S, Fuchs-Moll G, Petri K, Wilker S et al. Adaptive and innate immune responses in a rat orthotopic lung transplant model of chronic lung allograft dysfunction. Transpl Int 2015; 28:95–107 [View Article]
    [Google Scholar]
  33. Periselneris J, Ercoli G, Pollard T, Chimalapati S, Camberlein E et al. Relative contributions of extracellular and internalized bacteria to early macrophage roinflammatory responses to Streptococcus pneumoniae. mBio 2019; 10:e02144-19 [View Article]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/jmm/10.1099/jmm.0.001588
Loading
/content/journal/jmm/10.1099/jmm.0.001588
Loading

Data & Media loading...

This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error