1887

Abstract

One third of people with CF in the UK are co-infected by both and . Chronic bacterial infection in CF contributes to the gradual destruction of lung tissue, and eventually respiratory failure in this group.

The contribution of to cystic fibrosis (CF) lung decline in the presence or absence of is unclear. Defining the molecular and phenotypic characteristics of a range of clinical isolates will help further understand its pathogenic capabilities.

Our objective was to use molecular and phenotypic tools to characterise twenty-five clinical isolates collected from mono- and coinfection with from people with CF at the Royal Victoria Infirmary, Newcastle upon Tyne.

Genomic DNA was extracted and sequenced. Multilocus sequence typing was used to construct phylogeny from the seven housekeeping genes. A pangenome was calculated using Roary, and cluster of Orthologous groups were assigned using eggNOG-mapper which were used to determine differences within core, accessory, and unique genomes. Characterisation of sequence type, clonal complex, and types was carried out using PubMLST, eBURST, AgrVATE and spaTyper, respectively. Antibiotic resistance was determined using Kirby-Bauer disc diffusion tests. Phenotypic testing of haemolysis was carried out using ovine red blood cell agar plates and mucoid phenotypes visualised using Congo red agar.

Clinical strains clustered closely based on type, sequence type and clonal complex. COG analysis revealed statistically significant enrichment of COG families between core, accessory and unique pangenome groups. The unique genome was significantly enriched for replication, recombination and repair, and defence mechanisms. The presence of known virulence genes and toxins were high within this group, and unique genes were identified in 11 strains. Strains which were isolated from the same patient all surpassed average nucleotide identity thresholds, however, differed in phenotypic traits. Antimicrobial resistance to macrolides was significantly higher in the coinfection group.

There is huge variation in genetic and phenotypic capabilities of strains. Further studies on how these may differ in relation to other species in the CF lung may give insight into inter-species interactions.

Funding
This study was supported by the:
  • National Health and Medical Research Council (Award APP2002921)
    • Principle Award Recipient: Jhih-HangJiang
  • Wellcome Trust (Award 224151/Z/21/Z)
    • Principle Award Recipient: TracyPalmer
  • Wellcome Trust (Award 10183/Z/15/Z)
    • Principle Award Recipient: TracyPalmer
  • This is an open-access article distributed under the terms of the Creative Commons Attribution License. This article was made open access via a Publish and Read agreement between the Microbiology Society and the corresponding author’s institution.
Loading

Article metrics loading...

/content/journal/jmm/10.1099/jmm.0.001703
2023-06-08
2024-04-29
Loading full text...

Full text loading...

/deliver/fulltext/jmm/72/6/jmm001703.html?itemId=/content/journal/jmm/10.1099/jmm.0.001703&mimeType=html&fmt=ahah

References

  1. Riordan JR, Rommens JM, Kerem B, Alon N, Rozmahel R et al. Identification of the cystic fibrosis gene: cloning and characterization of complementary DNA. Science 1989; 245:1066–1073 [View Article] [PubMed]
    [Google Scholar]
  2. CF Trust UK Uk Cystic Fibrosis Registry Annual Data Report; 2019
  3. Maliniak ML, Stecenko AA, McCarty NA. A longitudinal analysis of chronic MRSA and Pseudomonas aeruginosa co-infection in cystic fibrosis: a single-center study. J Cyst Fibros 2016; 15:350–356 [View Article] [PubMed]
    [Google Scholar]
  4. Limoli DH, Yang J, Khansaheb MK, Helfman B, Peng L et al. Staphylococcus aureus and Pseudomonas aeruginosa co-infection is associated with cystic fibrosis-related diabetes and poor clinical outcomes. Eur J Clin Microbiol Infect Dis 2016; 35:947–953 [View Article] [PubMed]
    [Google Scholar]
  5. Rosenbluth DB, Wilson K, Ferkol T, Schuster DP. Lung function decline in cystic fibrosis patients and timing for lung transplantation referral. Chest 2004; 126:412–419 [View Article] [PubMed]
    [Google Scholar]
  6. Briaud P, Bastien S, Camus L, Boyadjian M, Reix P et al. Impact of coexistence phenotype between Staphylococcus aureus and Pseudomonas aeruginosa isolates on clinical outcomes among cystic fibrosis patients. Front Cell Infect Microbiol 2020; 10:266 [View Article] [PubMed]
    [Google Scholar]
  7. Ahlgren HG, Benedetti A, Landry JS, Bernier J, Matouk E et al. Clinical outcomes associated with Staphylococcus aureus and Pseudomonas aeruginosa airway infections in adult cystic fibrosis patients. BMC Pulm Med 2015; 15:67 [View Article] [PubMed]
    [Google Scholar]
  8. Junge S, Görlich D, den Reijer M, Wiedemann B, Tümmler B et al. Factors associated with worse Lung function in Cystic Fibrosis patients with persistent Staphylococcus aureus. PLoS One 2016; 11:e0166220 [View Article] [PubMed]
    [Google Scholar]
  9. Limoli DH, Hoffman LR. Help, hinder, hide and harm: what can we learn from the interactions between Pseudomonas aeruginosa and Staphylococcus aureus during respiratory infections?. Thorax 2019; 74:684–692 [View Article] [PubMed]
    [Google Scholar]
  10. Camus L, Briaud P, Vandenesch F, Doléans-Jordheim A, Moreau K. Mixed populations and Co-Infection: Pseudomonas aeruginosa and Staphylococcus aureus. Adv Exp Med Biol 2022; 1386:397–424 [View Article] [PubMed]
    [Google Scholar]
  11. Valentini M, Gonzalez D, Mavridou DA, Filloux A. Lifestyle transitions and adaptive pathogenesis of Pseudomonas aeruginosa. Curr Opin Microbiol 2018; 41:15–20 [View Article] [PubMed]
    [Google Scholar]
  12. Goerke C, Wolz C. Adaptation of Staphylococcus aureus to the cystic fibrosis lung. Int J Med Microbiol 2010; 300:520–525 [View Article] [PubMed]
    [Google Scholar]
  13. Tomlinson KL, Lung TWF, Dach F, Annavajhala MK, Gabryszewski SJ et al. Staphylococcus aureus induces an itaconate-dominated immunometabolic response that drives biofilm formation. Nat Commun 2021; 12:1399 [View Article] [PubMed]
    [Google Scholar]
  14. Hirschhausen N, Block D, Bianconi I, Bragonzi A, Birtel J et al. Extended Staphylococcus aureus persistence in cystic fibrosis is associated with bacterial adaptation. Int J Med Microbiol 2013; 303:685–692 [View Article] [PubMed]
    [Google Scholar]
  15. Schwartbeck B, Birtel J, Treffon J, Langhanki L, Mellmann A et al. Dynamic in vivo mutations within the ica operon during persistence of Staphylococcus aureus in the airways of cystic fibrosis patients. PLoS Pathog 2016; 12:e1006024 [View Article] [PubMed]
    [Google Scholar]
  16. Biswas L, Gotz F. Molecular mechanisms of Staphylococcus and Pseudomonas interactions in cystic fibrosis. Front Cell Infect Microbiol 2021; 11:824042
    [Google Scholar]
  17. Graham BL, Steenbruggen I, Miller MR, Barjaktarevic IZ, Cooper BG et al. Standardization of spirometry 2019 update. An official American thoracic society and European respiratory society technical statement. Am J Respir Crit Care Med 2019; 200:e70–e88 [View Article] [PubMed]
    [Google Scholar]
  18. Quanjer PH, Stanojevic S, Cole TJ, Baur X, Hall GL et al. Multi-ethnic reference values for spirometry for the 3–95-yr age range: the global lung function 2012 equations. Eur Respir J 2012; 40:1324–1343 [View Article]
    [Google Scholar]
  19. Wick RR, Judd LM, Gorrie CL, Holt KE. Unicycler: resolving bacterial genome assemblies from short and long sequencing reads. PLoS Comput Biol 2017; 13:e1005595 [View Article] [PubMed]
    [Google Scholar]
  20. Garrett SR, Mariano G, Palmer T. Genomic analysis of the progenitor strains of Staphylococcus aureus RN6390. Access Microbiol 2022; 4:11 [View Article]
    [Google Scholar]
  21. Darling ACE, Mau B, Blattner FR, Perna NT. Mauve: multiple alignment of conserved genomic sequence with rearrangements. Genome Res 2004; 14:1394–1403 [View Article] [PubMed]
    [Google Scholar]
  22. Seemann T. Prokka: rapid prokaryotic genome annotation. Bioinformatics 2014; 30:2068–2069 [View Article]
    [Google Scholar]
  23. Edgar RC. MUSCLE: a multiple sequence alignment method with reduced time and space complexity. BMC Bioinformatics 2004; 5:113 [View Article] [PubMed]
    [Google Scholar]
  24. Edgar RC. MUSCLE: multiple sequence alignment with high accuracy and high throughput. Nucleic Acid Res 2004; 32:1792–1797 [View Article]
    [Google Scholar]
  25. Kumar S, Stecher G, Li M, Knyaz C, Tamura K. MEGA X: molecular evolutionary genetics analysis across computing platforms. Mol Biol Evol 2018; 35:1547–1549 [View Article] [PubMed]
    [Google Scholar]
  26. Letunic I, Bork P. Interactive Tree Of Life (iTOL) v5: an online tool for phylogenetic tree display and annotation. Nucleic Acids Res 2021; 49:W293–W296 [View Article] [PubMed]
    [Google Scholar]
  27. Page AJ, Cummins CA, Hunt M, Wong VK, Reuter S et al. Roary: rapid large-scale prokaryote pan genome analysis. Bioinformatics 2015; 31:3691–3693 [View Article] [PubMed]
    [Google Scholar]
  28. Cantalapiedra CP, Hernández-Plaza A, Letunic I, Bork P, Huerta-Cepas J. eggNOG-mapper v2: functional annotation, orthology assignments, and domain prediction at the metagenomic scale. Mol Biol Evol 2021; 38:5825–5829 [View Article] [PubMed]
    [Google Scholar]
  29. Tettelin H, Riley D, Cattuto C, Medini D. Comparative genomics: the bacterial pan-genome. Curr Opin Microbiol 2008; 11:472–477 [View Article] [PubMed]
    [Google Scholar]
  30. Jolley KA, Bray JE, Maiden MCJ. Open-access bacterial population genomics: BIGSdb software, the PubMLST.org website and their applications. Wellcome Open Res 2018; 3:124 [View Article] [PubMed]
    [Google Scholar]
  31. Francisco AP, Bugalho M, Ramirez M, Carriço JA. Global optimal eBURST analysis of multilocus typing data using a graphic matroid approach. BMC Bioinformatics 2009; 10:152 [View Article] [PubMed]
    [Google Scholar]
  32. Raghuram V, Alexander AM, Loo HQ, Petit RA, Goldberg JB et al. Species-wide phylogenomics of the Staphylococcus aureus Agr Operon ervealed convergent evolution of frameshift mutations. Microbiol Spectr 2022; 10:e0133421 [View Article] [PubMed]
    [Google Scholar]
  33. Sanchez-Herrero JF. SpaTyper: staphylococcal protein A (spa) characterization pipeline. Zenodo 2020
    [Google Scholar]
  34. Jain C, Rodriguez-R LM, Phillippy AM, Konstantinidis KT, Aluru S. High throughput ANI analysis of 90K prokaryotic genomes reveals clear species boundaries. Nat Commun 2018; 9:5114 [View Article] [PubMed]
    [Google Scholar]
  35. Hunt M, Bradley P, Lapierre SG, Heys S, Thomsit M et al. Antibiotic resistance prediction for Mycobacterium tuberculosis from genome sequence data with Mykrobe. Wellcome Open Res 2019; 4:191 [View Article] [PubMed]
    [Google Scholar]
  36. Zankari E, Hasman H, Cosentino S, Vestergaard M, Rasmussen S et al. Identification of acquired antimicrobial resistance genes. J Antimicrob Chemother 2012; 67:2640–2644 [View Article] [PubMed]
    [Google Scholar]
  37. Carver T, Harris SR, Berriman M, Parkhill J, McQuillan JA. Artemis: an integrated platform for visualization and analysis of high-throughput sequence-based experimental data. Bioinformatics 2012; 28:464–469 [View Article] [PubMed]
    [Google Scholar]
  38. Larkin MA, Blackshields G, Brown NP, Chenna R, McGettigan PA et al. Clustal W and Clustal X version 2.0. Bioinformatics 2007; 23:2947–2948 [View Article] [PubMed]
    [Google Scholar]
  39. Waterhouse AM, Procter JB, Martin DMA, Clamp M, Barton GJ. Jalview Version 2--a multiple sequence alignment editor and analysis workbench. Bioinformatics 2009; 25:1189–1191 [View Article] [PubMed]
    [Google Scholar]
  40. Burnside K, Lembo A, de Los Reyes M, Iliuk A, Binhtran N-T et al. Regulation of hemolysin expression and virulence of Staphylococcus aureus by a serine/threonine kinase and phosphatase. PLoS One 2010; 5:e11071 [View Article] [PubMed]
    [Google Scholar]
  41. Feil EJ, Li BC, Aanensen DM, Hanage WP, Spratt BG. eBURST: inferring patterns of evolutionary descent among clusters of related bacterial genotypes from multilocus sequence typing data. J Bacteriol 2004; 186:1518–1530 [View Article] [PubMed]
    [Google Scholar]
  42. Cao Z, Casabona MG, Kneuper H, Chalmers JD, Palmer T. The type VII secretion system of Staphylococcus aureus secretes a nuclease toxin that targets competitor bacteria. Nat Microbiol 2016; 2:16183 [View Article] [PubMed]
    [Google Scholar]
  43. Ulhuq FR, Gomes MC, Duggan GM, Guo M, Mendonca C et al. A membrane-depolarizing toxin substrate of the Staphylococcus aureus type VII secretion system mediates intraspecies competition. Proc Natl Acad Sci U S A 2020; 117:20836–20847 [View Article] [PubMed]
    [Google Scholar]
  44. Bowman L, Palmer T. The type VII secretion system of Staphylococcus. Annu Rev Microbiol 2021; 75:471–494
    [Google Scholar]
  45. Warne B, Harkins CP, Harris SR, Vatsiou A, Stanley-Wall N et al. The Ess/Type VII secretion system of Staphylococcus aureus shows unexpected genetic diversity. BMC Genomics 2016; 17:222 [View Article] [PubMed]
    [Google Scholar]
  46. Ibberson CB, Stacy A, Fleming D, Dees JL, Rumbaugh K et al. Co-infecting microorganisms dramatically alter pathogen gene essentiality during polymicrobial infection. Nat Microbiol 2017; 2:17079 [View Article] [PubMed]
    [Google Scholar]
  47. Haaber J, Penadés JR, Ingmer H. Transfer of antibiotic resistance in Staphylococcus aureus. Trends Microbiol 2017; 25:893–905 [View Article] [PubMed]
    [Google Scholar]
  48. McCarthy AJ, Witney AA, Lindsay JA. Staphylococcus aureus temperate bacteriophage: carriage and horizontal gene transfer is lineage associated. Front Cell Infect Microbiol 2012; 2:6 [View Article] [PubMed]
    [Google Scholar]
  49. Oliveira PH, Touchon M, Rocha EPC. The interplay of restriction-modification systems with mobile genetic elements and their prokaryotic hosts. Nucleic Acids Res 2014; 42:10618–10631 [View Article] [PubMed]
    [Google Scholar]
  50. Abdulgader SM, van Rijswijk A, Whitelaw A, Newton-Foot M. The association between pathogen factors and clinical outcomes in patients with Staphylococcus aureus bacteraemia in a tertiary hospital, Cape Town. Int J Infect Dis 2020; 91:111–118 [View Article] [PubMed]
    [Google Scholar]
  51. Bernardy EE, Petit RA, Raghuram V, Alexander AM, Read TD et al. Genotypic and phenotypic diversity of Staphylococcus aureus Isolates from Cystic fibrosis patient Lung infections and their interactions with Pseudomonas aeruginosa. mBio 2020; 11:e00735-20 [View Article] [PubMed]
    [Google Scholar]
  52. Bibalan MH, Shakeri F, Javid N, Ghaemi A, Ghaemi EA. Accessory gene regulator types of Staphylococcus aureus isolated in Gorgan, North of Iran. J Clin Diagn Res 2014; 8:DC07–9 [View Article] [PubMed]
    [Google Scholar]
  53. Mazi W, Sangal V, Sandstrom G, Saeed A, Yu J. Evaluation of spa-typing of methicillin-resistant Staphylococcus aureus using high-resolution melting analysis. Int J Infect Dis 2015; 38:125–128 [View Article] [PubMed]
    [Google Scholar]
  54. Keitsch S, Riethmüller J, Soddemann M, Sehl C, Wilker B et al. Pulmonary infection of cystic fibrosis mice with Staphylococcus aureus requires expression of α-toxin. Biol Chem 2018; 399:1203–1213 [View Article] [PubMed]
    [Google Scholar]
  55. Camus L, Briaud P, Vandenesch F, Moreau K. How Bacterial Adaptation to Cystic Fibrosis Environment Shapes Interactions Between Pseudomonas aeruginosa and Staphylococcus aureus. Front Microbiol 2021; 12:617784 [View Article] [PubMed]
    [Google Scholar]
  56. Nguyen HTT, Nguyen TH, Otto M. The Staphylococcal exopolysaccharide PIA - Biosynthesis and role in biofilm formation, colonization, and infection. Comput Struct Biotechnol J 2020; 18:3324–3334 [View Article] [PubMed]
    [Google Scholar]
  57. Heilmann C, Schweitzer O, Gerke C, Vanittanakom N, Mack D et al. Molecular basis of intercellular adhesion in the biofilm-forming Staphylococcus epidermidis. Mol Microbiol 1996; 20:1083–1091 [View Article] [PubMed]
    [Google Scholar]
  58. Jefferson KK, Cramton SE, Götz F, Pier GB. Identification of a 5-nucleotide sequence that controls expression of the ica locus in Staphylococcus aureus and characterization of the DNA-binding properties of IcaR. Mol Microbiol 2003; 48:889–899 [View Article] [PubMed]
    [Google Scholar]
  59. Lennartz FE, Schwartbeck B, Dübbers A, Große-Onnebrink J, Kessler C et al. The prevalence of Staphylococcus aureus with mucoid phenotype in the airways of patients with cystic fibrosis-A prospective study. Int J Med Microbiol 2019; 309:283–287 [View Article] [PubMed]
    [Google Scholar]
  60. Nabb DL, Song S, Kluthe KE, Daubert TA, Luedtke BE et al. Polymicrobial interactions induce multidrug tolerance in Staphylococcus aureus through energy depletion. Front Microbiol 2019; 10:2803 [View Article] [PubMed]
    [Google Scholar]
  61. Beaudoin T, Yau YCW, Stapleton PJ, Gong Y, Wang PW et al. Staphylococcus aureus interaction with Pseudomonas aeruginosa biofilm enhances tobramycin resistance. NPJ Biofilms Microbiomes 2017; 3:25 [View Article]
    [Google Scholar]
  62. Briaud P, Camus L, Bastien S, Doléans-Jordheim A, Vandenesch F et al. Coexistence with Pseudomonas aeruginosa alters Staphylococcus aureus transcriptome, antibiotic resistance and internalization into epithelial cells. Sci Rep 2019; 9:16564 [View Article] [PubMed]
    [Google Scholar]
  63. Westh H, Hougaard DM, Vuust J, Rosdahl VT. Prevalence of erm gene classes in erythromycin-resistant Staphylococcus aureus strains isolated between 1959 and 1988. Antimicrob Agents Chemother 1995; 39:369–373 [View Article] [PubMed]
    [Google Scholar]
  64. Long DR, Wolter DJ, Lee M, Precit M, McLean K et al. Polyclonality, Shared strains, and convergent evolution in chronic Cystic Fibrosis Staphylococcus aureus airway infection. Am J Respir Crit Care Med 2021; 203:1127–1137 [View Article] [PubMed]
    [Google Scholar]
  65. Bosi E, Monk JM, Aziz RK, Fondi M, Nizet V et al. Comparative genome-scale modelling of Staphylococcus aureus strains identifies strain-specific metabolic capabilities linked to pathogenicity. Proc Natl Acad Sci U S A 2016; 113:E3801–9 [View Article] [PubMed]
    [Google Scholar]
  66. Donlan RM, Costerton JW. Biofilms: survival mechanisms of clinically relevant microorganisms. Clin Microbiol Rev 2002; 15:167–193 [View Article] [PubMed]
    [Google Scholar]
  67. Vuong C, Voyich JM, Fischer ER, Braughton KR, Whitney AR et al. Polysaccharide intercellular adhesin (PIA) protects Staphylococcus epidermidis against major components of the human innate immune system. Cell Microbiol 2004; 6:269–275 [View Article] [PubMed]
    [Google Scholar]
  68. Orazi G, Jean-Pierre F, O’Toole GA. Pseudomonas aeruginosa PA14 enhances the efficacy of Norfloxacin against Staphylococcus aureus newman biofilms. J Bacteriol 2020; 202:e00159-20 [View Article] [PubMed]
    [Google Scholar]
  69. Radlinski L, Rowe SE, Kartchner LB, Maile R, Cairns BA et al. Pseudomonas aeruginosa exoproducts determine antibiotic efficacy against Staphylococcus aureus. PLoS Biol 2017; 15:e2003981 [View Article] [PubMed]
    [Google Scholar]
  70. Goerke C, Gressinger M, Endler K, Breitkopf C, Wardecki K et al. High phenotypic diversity in infecting but not in colonizing Staphylococcus aureus populations. Environ Microbiol 2007; 9:3134–3142 [View Article] [PubMed]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/jmm/10.1099/jmm.0.001703
Loading
/content/journal/jmm/10.1099/jmm.0.001703
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