1887

Abstract

Bacteriophages represent the most extensive group of viruses within the human virome and have a significant impact on general health and well-being by regulating bacterial population dynamics. , found in the anterior nostrils, throat and skin, is an opportunistic pathobiont that can cause a wide range of diseases, from chronic inflammation to severe and acute infections. In this study, we developed a human cell-based homeostasis model between a clinically isolated strain of 141 and active phages for this strain (PYO) isolated from the commercial Pyophage cocktail (PYO). The cocktail is produced by Eliava BioPreparations Ltd. (Tbilisi, Georgia) and is used as an add-on therapy for bacterial infections, mainly in Georgia. The triptych interaction model was evaluated by time-dependent analysis of cell death and inflammatory response of the nasal and bronchial epithelial cells. Inflammatory mediators (IL-8, CCL5/RANTES, IL-6 and IL-1β) in the culture supernatants were measured by enzyme-linked immunosorbent assay and cell viability was determined by crystal violet staining. By measuring trans-epithelial electrical resistance, we assessed the epithelial integrity of nasal cells that had differentiated under air-liquid interface conditions. PYO was found to have a prophylactic effect on airway epithelial cells exposed to 141 by effectively down-regulating bacterial-induced inflammation, cell death and epithelial barrier disruption in a time-dependent manner. Overall, the proposed model represents an advance in the way multi-component biological systems can be simulated .

Funding
This study was supported by the:
  • HORIZON EUROPE Framework Programme (Award grant agreement No. 767015)
    • Principle Award Recipient: NotApplicable
Loading

Article metrics loading...

/content/journal/jgv/10.1099/jgv.0.001819
2022-12-19
2024-05-05
Loading full text...

Full text loading...

References

  1. Rohwer F, Segall AM. In retrospect: a century of phage lessons. Nature 2015; 528:46–48 [View Article]
    [Google Scholar]
  2. Fernández L, Rodríguez A, García P. Phage or foe: an insight into the impact of viral predation on microbial communities. ISME J 2018; 12:1171–1179 [View Article] [PubMed]
    [Google Scholar]
  3. Koskella B, Brockhurst MA. Bacteria-phage coevolution as a driver of ecological and evolutionary processes in microbial communities. FEMS Microbiol Rev 2014; 38:916–931 [View Article] [PubMed]
    [Google Scholar]
  4. Santiago-Rodriguez TM, Hollister EB. Human virome and disease: high-throughput sequencing for virus discovery, identification of phage-bacteria dysbiosis and development of therapeutic approaches with emphasis on the human gut. Viruses 2019; 11:656 [View Article]
    [Google Scholar]
  5. Lim YW, Schmieder R, Haynes M, Furlan M, Matthews TD et al. Mechanistic model of Rothia mucilaginosa adaptation toward persistence in the CF lung, based on a genome reconstructed from metagenomic data. PLoS One 2013; 8:e64285 [View Article]
    [Google Scholar]
  6. Moustafa A, Xie C, Kirkness E, Biggs W, Wong E et al. The blood DNA virome in 8,000 humans. PLoS Pathog 2017; 13:e1006292 [View Article]
    [Google Scholar]
  7. Gordillo Altamirano FL, Barr JJ. Phage therapy in the postantibiotic era. Clin Microbiol Rev 2019; 32:e00066-18 [View Article]
    [Google Scholar]
  8. Brown SP, Cornforth DM, Mideo N. Evolution of virulence in opportunistic pathogens: generalism, plasticity, and control. Trends Microbiol 2012; 20:336–342 [View Article] [PubMed]
    [Google Scholar]
  9. Wertheim HFL, Melles DC, Vos MC, van Leeuwen W, van Belkum A et al. The role of nasal carriage in Staphylococcus aureus infections. Lancet Infect Dis 2005; 5:751–762 [View Article] [PubMed]
    [Google Scholar]
  10. Verhoeven PO, Grattard F, Carricajo A, Lucht F, Cazorla C et al. An algorithm based on one or two nasal samples is accurate to identify persistent nasal carriers of Staphylococcus aureus. Clin Microbiol Infect 2012; 18:551–557 [View Article] [PubMed]
    [Google Scholar]
  11. Nouwen JL, Fieren MWJA, Snijders S, Verbrugh HA, van Belkum A. Persistent (not intermittent) nasal carriage of Staphylococcus aureus is the determinant of CPD-related infections. Kidney Int 2005; 67:1084–1092 [View Article] [PubMed]
    [Google Scholar]
  12. McCauley K, Durack J, Valladares R, Fadrosh DW, Lin DL et al. Distinct nasal airway bacterial microbiotas differentially relate to exacerbation in pediatric patients with asthma. J Allergy Clin Immunol 2019; 144:1187–1197 [View Article] [PubMed]
    [Google Scholar]
  13. Tzani-Tzanopoulou P, Skliros D, Megremis S, Xepapadaki P, Andreakos E et al. Interactions of bacteriophages and bacteria at the airway Mucosa: new insights into the pathophysiology of asthma. Front Allergy 2020; 1:617240 [View Article]
    [Google Scholar]
  14. Leitner L, Ujmajuridze A, Chanishvili N, Goderdzishvili M, Chkonia I et al. Intravesical bacteriophages for treating urinary tract infections in patients undergoing transurethral resection of the prostate: a randomised, placebo-controlled, double-blind clinical trial. Lancet Infect Dis 2021; 21:427–436 [View Article] [PubMed]
    [Google Scholar]
  15. Wang L, Tkhilaishvili T, Trampuz A, Gonzalez Moreno M. Evaluation of Staphylococcal bacteriophage Sb-1 as an adjunctive agent to antibiotics against rifampin-resistant Staphylococcus aureus biofilms. Front Microbiol 2020; 11:602057 [View Article]
    [Google Scholar]
  16. Abedon ST, Danis-Wlodarczyk KM, Alves DR. Phage therapy in the 21st century: is there modern, clinical evidence of phage-mediated efficacy?. Pharmaceuticals 2021; 14:1157 [View Article]
    [Google Scholar]
  17. Abatángelo V, Peressutti Bacci N, Boncompain CA, Amadio AF, Carrasco S et al. Broad-range lytic bacteriophages that kill Staphylococcus aureus local field strains. PLoS One 2017; 12:e0181671 [View Article]
    [Google Scholar]
  18. Ajuebor J, Buttimer C, Arroyo-Moreno S, Chanishvili N, Gabriel EM et al. Comparison of Staphylococcus phage K with close phage relatives commonly employed in phage therapeutics. Antibiotics 2018; 7:37 [View Article]
    [Google Scholar]
  19. Berryhill BA, Huseby DL, McCall IC, Hughes D, Levin BR. Evaluating the potential efficacy and limitations of a phage for joint antibiotic and phage therapy of Staphylococcus aureus infections. Proc Natl Acad Sci 2021; 118:10 [View Article]
    [Google Scholar]
  20. Xia G, Kohler T, Peschel A. The wall teichoic acid and lipoteichoic acid polymers of Staphylococcus aureus. Int J Med Microbiol 2010; 300:148–154 [View Article] [PubMed]
    [Google Scholar]
  21. Dąbrowska K, Abedon ST. Pharmacologically aware phage therapy: pharmacodynamic and pharmacokinetic obstacles to phage antibacterial action in animal and human bodies. Microbiol Mol Biol Rev 2019; 83:e00012-19 [View Article]
    [Google Scholar]
  22. Barr JJ, Auro R, Furlan M, Whiteson KL, Erb ML et al. Bacteriophage adhering to mucus provide a non-host-derived immunity. Proc Natl Acad Sci 2013; 110:10771–10776 [View Article]
    [Google Scholar]
  23. Nguyen S, Baker K, Padman BS, Patwa R, Dunstan RA et al. Bacteriophage transcytosis provides a mechanism to cross epithelial cell layers. mBio 2017; 8:e01874-17 [View Article]
    [Google Scholar]
  24. Tian Y, Wu M, Liu X, Liu Z, Zhou Q et al. Probing the endocytic pathways of the filamentous bacteriophage in live cells using ratiometric pH fluorescent indicator. Adv Healthc Mater 2015; 4:413–419 [View Article] [PubMed]
    [Google Scholar]
  25. Kamme C. Antibodies against staphylococcal bacteriophages in human sera. I. Assay of antibodies in healthy individuals and in patients with staphylococcal infections. Acta Pathol Microbiol Scand B Microbiol Immunol 1973; 81:741–748 [View Article] [PubMed]
    [Google Scholar]
  26. Kaźmierczak Z, Majewska J, Miernikiewicz P, Międzybrodzki R, Nowak S et al. Immune response to therapeutic Staphylococcal bacteriophages in mammals: kinetics of induction, immunogenic structural proteins, natural and induced antibodies. Front Immunol 2021; 12:639570 [View Article]
    [Google Scholar]
  27. McCallin S, Sarker SA, Sultana S, Oechslin F, Brüssow H. Metagenome analysis of Russian and Georgian Pyophage cocktails and a placebo-controlled safety trial of single phage versus phage cocktail in healthy Staphylococcus aureus carriers. Environ Microbiol 2018; 20:3278–3293 [View Article]
    [Google Scholar]
  28. Jahn MT, Arkhipova K, Markert SM, Stigloher C, Lachnit T et al. A phage protein aids bacterial symbionts in eukaryote immune evasion. Cell Host Microbe 2019; 26:542–550 [View Article]
    [Google Scholar]
  29. Davies EV, James CE, Kukavica-Ibrulj I, Levesque RC, Brockhurst MA et al. Temperate phages enhance pathogen fitness in chronic lung infection. ISME J 2016; 10:2553–2555 [View Article]
    [Google Scholar]
  30. Secor PR, Sweere JM, Michaels LA, Malkovskiy AV, Lazzareschi D et al. Filamentous bacteriophage promote biofilm assembly and function. Cell Host Microbe 2015; 18:549–559 [View Article]
    [Google Scholar]
  31. Carrolo M, Frias MJ, Pinto FR, Melo-Cristino J, Ramirez M. Prophage spontaneous activation promotes DNA release enhancing biofilm formation in Streptococcus pneumoniae. PLoS One 2010; 5:e15678 [View Article] [PubMed]
    [Google Scholar]
  32. Bille E, Meyer J, Jamet A, Euphrasie D, Barnier J-P et al. A virulence-associated filamentous bacteriophage of Neisseria meningitidis increases host-cell colonisation. PLoS Pathog 2017; 13:e1006495 [View Article]
    [Google Scholar]
  33. Górski A, Dąbrowska K, Hodyra-Stefaniak K, Borysowski J, Międzybrodzki R et al. Phages targeting infected tissues: novel approach to phage therapy. Future Microbiol 2015; 10:199–204 [View Article] [PubMed]
    [Google Scholar]
  34. Lan F, Zhang N, Holtappels G, De Ruyck N, Krysko O et al. Staphylococcus aureus induces a Mucosal type 2 immune response via epithelial cell-derived cytokines. Am J Respir Crit Care Med 2018; 198:452–463 [View Article]
    [Google Scholar]
  35. Wang J, Zhao F, Sun H, Wang Q, Zhang C et al. Isolation and characterization of the Staphylococcus aureus bacteriophage vB_SauS_SA2. AIMS Microbiol 2019; 5:285–307 [View Article]
    [Google Scholar]
  36. Sanders ER. Aseptic laboratory techniques: plating methods. J Vis Exp 2012; 2012:e3064 [View Article]
    [Google Scholar]
  37. Anderson B, Rashid MH, Carter C, Pasternack G, Rajanna C et al. Enumeration of bacteriophage particles: Comparative analysis of the traditional plaque assay and real-time QPCR- and nanosight-based assays. Bacteriophage 2011; 1:86–93 [View Article] [PubMed]
    [Google Scholar]
  38. Racenis K, Kroica J, Rezevska D, Avotins L, Skuditis E et al. S. aureus colonization, biofilm production, and phage susceptibility in peritoneal dialysis patients. Antibiotics 2020; 9:582 [View Article]
    [Google Scholar]
  39. Synnott AJ, Kuang Y, Kurimoto M, Yamamichi K, Iwano H et al. Isolation from sewage influent and characterization of novel Staphylococcus aureus bacteriophages with wide host ranges and potent lytic capabilities. Appl Environ Microbiol 2009; 75:4483–4490 [View Article] [PubMed]
    [Google Scholar]
  40. Hietala V, Horsma-Heikkinen J, Carron A, Skurnik M, Kiljunen S. The removal of endo- and enterotoxins from bacteriophage preparations. Front Microbiol 2019; 10:1674 [View Article]
    [Google Scholar]
  41. Bonilla N, Rojas MI, Netto Flores Cruz G, Hung S-H, Rohwer F et al. Phage on tap-a quick and efficient protocol for the preparation of bacteriophage laboratory stocks. PeerJ 2016; 4:e2261 [View Article]
    [Google Scholar]
  42. Müller L, Brighton LE, Carson JL, Fischer WA, Jaspers I. Culturing of human nasal epithelial cells at the air liquid interface. J Vis Exp 2013; 2013:80 [View Article]
    [Google Scholar]
  43. LaFayette SL, Houle D, Beaudoin T, Wojewodka G, Radzioch D et al. Cystic fibrosis-adapted Pseudomonas aeruginosa quorum sensing lasR mutants cause hyperinflammatory responses. Sci Adv 2015; 1:e1500199 [View Article]
    [Google Scholar]
  44. Zhao F, Klimecki WT. Culture conditions profoundly impact phenotype in BEAS-2B, a human pulmonary epithelial model. J Appl Toxicol 2015; 35:945–951 [View Article] [PubMed]
    [Google Scholar]
  45. Xing SF. Study on the criteria of fine needle aspiration cytology for the diagnosis and classification of malignant lymphomas. Zhonghua Bing Li Xue Za Zhi 1987; 16:174–176 [PubMed]
    [Google Scholar]
  46. Burrowes BH, Molineux IJ, Fralick JA. Directed in vitro evolution of therapeutic bacteriophages: the appelmans protocol. Viruses 2019; 11:241 [View Article]
    [Google Scholar]
  47. Vengarai Jagannathan B, Kitchens S, Vijayakumar PP, Price S, Morgan M. Potential for bacteriophage cocktail to complement commercial sanitizer use on produce against Escherichia coli O157:H7. Microorganisms 2020; 8:1316 [View Article]
    [Google Scholar]
  48. Almutary A, Sanderson BJS. The MTT and crystal violet assays: potential confounders in nanoparticle toxicity testing. Int J Toxicol 2016; 35:454–462 [View Article]
    [Google Scholar]
  49. Feoktistova M, Geserick P, Leverkus M. Crystal violet assay for determining viability of cultured cells. Cold Spring Harb Protoc 2016; 2016:pdb.prot087379 [View Article]
    [Google Scholar]
  50. Ramezanpour M, Moraitis S, Smith JLP, Wormald PJ, Vreugde S. Th17 cytokines disrupt the airway mucosal barrier in chronic Rhinosinusitis. Mediators Inflamm 2016; 2016:9798206 [View Article]
    [Google Scholar]
  51. Núñez-Sánchez MA, Colom J, Walsh L, Buttimer C, Bolocan AS et al. Characterizing phage-host interactions in a simplified human intestinal barrier model. Microorganisms 2020; 8:1374 [View Article]
    [Google Scholar]
  52. Bichet MC, Chin WH, Richards W, Lin Y-W, Avellaneda-Franco L et al. Bacteriophage uptake by mammalian cell layers represents a potential sink that may impact phage therapy. iScience 2021; 24:102287 [View Article]
    [Google Scholar]
  53. Shan J, Ramachandran A, Thanki AM, Vukusic FBI, Barylski J et al. Bacteriophages are more virulent to bacteria with human cells than they are in bacterial culture; insights from HT-29 cells. Sci Rep 2018; 8:5091 [View Article]
    [Google Scholar]
  54. Fey PD, Endres JL, Yajjala VK, Widhelm TJ, Boissy RJ et al. A genetic resource for rapid and comprehensive phenotype screening of nonessential Staphylococcus aureus genes. mBio 2013; 4:e00537–12 [View Article] [PubMed]
    [Google Scholar]
  55. Tkhilaishvili T, Wang L, Tavanti A, Trampuz A, Di Luca M. Antibacterial efficacy of two commercially available bacteriophage formulations, Staphylococcal bacteriophage and PYO bacteriophage, against methicillin-resistant Staphylococcus aureus: prevention and eradication of biofilm formation and control of a systemic infection of Galleria mellonella Larvae. Front Microbiol 2020; 11:110 [View Article]
    [Google Scholar]
  56. Kvachadze L, Balarjishvili N, Meskhi T, Tevdoradze E, Skhirtladze N et al. Evaluation of lytic activity of staphylococcal bacteriophage Sb-1 against freshly isolated clinical pathogens. Microb Biotechnol 2011; 4:643–650 [View Article] [PubMed]
    [Google Scholar]
  57. Sakr A, Brégeon F, Mège J-L, Rolain J-M, Blin O. Staphylococcus aureus nasal colonization: an update on mechanisms, epidemiology, risk factors, and subsequent infections. Front Microbiol 2018; 9:2419 [View Article]
    [Google Scholar]
  58. Nouwen JL, Ott A, Kluytmans-Vandenbergh MFQ, Boelens HAM, Hofman A et al. Predicting the Staphylococcus aureus nasal carrier state: derivation and validation of a “culture rule.”. Clin Infect Dis 2004; 39:806–811 [View Article] [PubMed]
    [Google Scholar]
  59. Eriksen NH, Espersen F, Rosdahl VT, Jensen K. Carriage of Staphylococcus aureus among 104 healthy persons during a 19-month period. Epidemiol Infect 1995; 115:51–60 [View Article] [PubMed]
    [Google Scholar]
  60. Sakwinska O, Blanc DS, Lazor-Blanchet C, Moreillon M, Giddey M et al. Ecological temporal stability of Staphylococcus aureus nasal carriage. J Clin Microbiol 2010; 48:2724–2728 [View Article] [PubMed]
    [Google Scholar]
  61. Baur S, Rautenberg M, Faulstich M, Grau T, Severin Y et al. A nasal epithelial receptor for Staphylococcus aureus WTA governs adhesion to epithelial cells and modulates nasal colonization. PLoS Pathog 2014; 10:e1004089 [View Article]
    [Google Scholar]
  62. Krut O, Utermöhlen O, Schlossherr X, Krönke M. Strain-specific association of cytotoxic activity and virulence of clinical Staphylococcus aureus isolates. Infect Immun 2003; 71:2716–2723 [View Article]
    [Google Scholar]
  63. Wigington CH, Sonderegger D, Brussaard CPD, Buchan A, Finke JF et al. Author correction: re-examination of the relationship between marine virus and microbial cell abundances. Nat Microbiol 2017; 2:1571 [View Article]
    [Google Scholar]
  64. Górski A, Weber-Dabrowska B. The potential role of endogenous bacteriophages in controlling invading pathogens. Cell Mol Life Sci 2005; 62:511–519 [View Article]
    [Google Scholar]
  65. Górski A, Borysowski J, Miȩdzybrodzki R. Bacteriophage interactions with epithelial cells: therapeutic implications. Front Microbiol 2020; 11:631161 [View Article]
    [Google Scholar]
  66. Trend S, Fonceca AM, Ditcham WG, Kicic A, Cf A. The potential of phage therapy in cystic fibrosis: essential human-bacterial-phage interactions and delivery considerations for use in Pseudomonas aeruginosa-infected airways. J Cyst Fibros 2017; 16:663–670 [View Article]
    [Google Scholar]
  67. Shelley JR, Davidson DJ, Dorin JR. The dichotomous responses driven by β-defensins. Front Immunol 2020; 11:1176 [View Article]
    [Google Scholar]
  68. Fujisawa T, Kato Y, Atsuta J, Terada A, Iguchi K et al. Chemokine production by the BEAS-2B human bronchial epithelial cells: differential regulation of eotaxin, IL-8, and RANTES by TH2- and TH1-derived cytokines. J Allergy Clin Immunol 2000; 105:126–133 [View Article] [PubMed]
    [Google Scholar]
  69. O’Brien GJ, Riddell G, Elborn JS, Ennis M, Skibinski G. Staphylococcus aureus enterotoxins induce IL-8 secretion by human nasal epithelial cells. Respir Res 2006; 7:115 [View Article]
    [Google Scholar]
  70. Wang ZM, Liu C, Dziarski R. Chemokines are the main proinflammatory mediators in human monocytes activated by Staphylococcus aureus, peptidoglycan, and endotoxin. J Biol Chem 2000; 275:20260–20267 [View Article] [PubMed]
    [Google Scholar]
  71. Ahmed EB, Wang T, Daniels M, Alegre ML, Chong AS. IL-6 induced by Staphylococcus aureus infection prevents the induction of skin allograft acceptance in mice. Am J Transplant 2011; 11:936–946 [View Article] [PubMed]
    [Google Scholar]
  72. Zhang L, Hou X, Sun L, He T, Wei R et al. Staphylococcus aureus bacteriophage suppresses LPS-induced inflammation in MAC-T bovine mammary epithelial cells. Front Microbiol 2018; 9:1614 [View Article]
    [Google Scholar]
  73. Dickson RP, Erb-Downward JR, Martinez FJ, Huffnagle GB. The microbiome and the respiratory tract. Annu Rev Physiol 2016; 78:481–504 [View Article]
    [Google Scholar]
  74. De Rudder C, Calatayud Arroyo M, Lebeer S, Van de Wiele T. Dual and triple epithelial coculture model systems with donor-derived microbiota and THP-1 macrophages to mimic host-microbe interactions in the human sinonasal cavities. mSphere 2020; 5:e00916-19 [View Article]
    [Google Scholar]
  75. Van Belleghem JD, Dąbrowska K, Vaneechoutte M, Barr JJ, Bollyky PL. Interactions between bacteriophage, bacteria, and the mammalian immune system. Viruses 2018; 11:10 [View Article]
    [Google Scholar]
  76. Barr JJ. Missing a phage: unraveling tripartite symbioses within the human gut. mSystems 2019; 4:e00105-19 [View Article]
    [Google Scholar]
  77. Styles KM, Brown AT, Sagona AP. A review of using mathematical modeling to improve our understanding of bacteriophage, bacteria, and eukaryotic interactions. Front Microbiol 2021; 12:724767 [View Article]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/jgv/10.1099/jgv.0.001819
Loading
/content/journal/jgv/10.1099/jgv.0.001819
Loading

Data & Media loading...

Supplements

Supplementary material 1

PDF
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