1887

Abstract

has emerged as one of the most common multi-drug-resistant pathogens isolated from people with cystic fibrosis (CF). However, its adaptation over time to CF lungs has not been fully established.

Sequential isolates of from a Brazilian adult patient are clonally related and show a pattern of adaptation by loss of virulence factors.

To investigate antimicrobial susceptibility, clonal relatedness, mutation frequency, quorum sensing (QS) and selected virulence factors in sequential isolates from a Brazilian adult patient attending a CF referral centre in Buenos Aires, Argentina, between May 2014 and May 2018.

The antibiotic resistance of 11 S. isolates recovered from expectorations of an adult female with CF was determined. Clonal relatedness, mutation frequency, QS variants (RpfC–RpfF), QS autoinducer (DSF) and virulence factors were investigated in eight viable isolates.

Seven isolates were resistant to trimethoprim–sulfamethoxazole and five to levofloxacin. All isolates were susceptible to minocycline. Strong, weak and normomutators were detected, with a tendency to decreased mutation rate over time. PFGE revealed that seven isolates belong to two related clones. All isolates were RpfC–RpfF1 variants and DSF producers. Only two isolates produced weak biofilms, but none displayed swimming or twitching motility. Four isolates showed proteolytic activity and amplified and genes. Only the first three isolates were siderophore producers. Four isolates showed high resistance to oxidative stress, while the last four showed moderate resistance.

The present study shows the long-time persistence of two related clones in an adult female with CF. During the adaptation of the prevalent clones to the CF lungs over time, we identified a gradual loss of virulence factors that could be associated with the high amounts of DSF produced by the evolved isolates. Further, a decreased mutation rate was observed in the late isolates. The role of all these adaptations over time remains to be elucidated from a clinical perspective, probably focusing on the damage they can cause to CF lungs.

Funding
This study was supported by the:
  • Secretaria de Ciencia y Tecnica, Universidad de Buenos Aires, http://dx.doi.org/10.13039/501100007351 (Award UBACYT 20020170100473BA)
    • Principle Award Recipient: Not Applicable
Loading

Article metrics loading...

/content/journal/jmm/10.1099/jmm.0.001281
2020-12-01
2024-03-29
Loading full text...

Full text loading...

/deliver/fulltext/jmm/70/1/jmm001281.html?itemId=/content/journal/jmm/10.1099/jmm.0.001281&mimeType=html&fmt=ahah

References

  1. Brooke JS. New strategies against Stenotrophomonas maltophilia: a serious worldwide intrinsically drug-resistant opportunistic pathogen. Expert Rev Anti Infect Ther 2014; 12:1–4 [View Article][PubMed]
    [Google Scholar]
  2. Vidigal PG, Dittmer S, Steinmann E, Buer J, Rath P-M et al. Adaptation of Stenotrophomonas maltophilia in cystic fibrosis: molecular diversity, mutation frequency and antibiotic resistance. Int J Med Microbiol 2014; 304:613–619 [View Article][PubMed]
    [Google Scholar]
  3. Esposito A, Pompilio A, Bettua C, Crocetta V, Giacobazzi E et al. Evolution of Stenotrophomonas maltophilia in cystic fibrosis lung over chronic infection: a genomic and phenotypic population study. Front Microbiol 2017; 8:1590 [View Article][PubMed]
    [Google Scholar]
  4. Salsgiver EL, Fink AK, Knapp EA, LiPuma JJ, Olivier KN et al. Changing epidemiology of the respiratory bacteriology of patients with cystic fibrosis. Chest 2016; 149:390–400 [View Article][PubMed]
    [Google Scholar]
  5. Parkins MD, Floto RA. Emerging bacterial pathogens and changing concepts of bacterial pathogenesis in cystic fibrosis. J Cyst Fibros 2015; 14:293–304 [View Article][PubMed]
    [Google Scholar]
  6. Brooke JS. Stenotrophomonas maltophilia: an emerging global opportunistic pathogen. Clin Microbiol Rev 2012; 25:2–41 [View Article][PubMed]
    [Google Scholar]
  7. Waters V, Yau Y, Prasad S, Lu A, Atenafu E et al. Stenotrophomonas maltophilia in cystic fibrosis: serologic response and effect on lung disease. Am J Respir Crit Care Med 2011; 183:635–640 [View Article][PubMed]
    [Google Scholar]
  8. Waters V, Atenafu EG, Lu A, Yau Y, Tullis E et al. Chronic Stenotrophomonas maltophilia infection and mortality or lung transplantation in cystic fibrosis patients. J Cyst Fibros 2013; 12:482–486 [View Article][PubMed]
    [Google Scholar]
  9. Pompilio A, Crocetta V, Ghosh D, Chakrabarti M, Gherardi G et al. Stenotrophomonas maltophilia phenotypic and genotypic diversity during a 10-year colonization in the lungs of a cystic fibrosis patient. Front Microbiol 2016; 7:1551 [View Article][PubMed]
    [Google Scholar]
  10. Passerini de Rossi B, Calenda M, Vay C, Franco M. Biofilm formation by Stenotrophomonas maltophilia isolates from device-associated nosocomial infections. Rev Argent Microbiol 2007; 39:204–212[PubMed]
    [Google Scholar]
  11. Pompilio A, Piccolomini R, Picciani C, D'Antonio D, Savini V et al. Factors associated with adherence to and biofilm formation on polystyrene by Stenotrophomonas maltophilia: the role of cell surface hydrophobicity and motility. FEMS Microbiol Lett 2008; 287:41–47 [View Article][PubMed]
    [Google Scholar]
  12. Nicoletti M, Iacobino A, Prosseda G, Fiscarelli E, Zarrilli R et al. Stenotrophomonas maltophilia strains from cystic fibrosis patients: genomic variability and molecular characterization of some virulence determinants. Int J Med Microbiol 2011; 301:34–43 [View Article][PubMed]
    [Google Scholar]
  13. García CA, Passerini De Rossi B, Alcaraz E, Vay C, Franco M. Siderophores of Stenotrophomonas maltophilia: detection and determination of their chemical nature. Rev Argent Microbiol 2012; 44:150–154[PubMed]
    [Google Scholar]
  14. García CA, Alcaraz ES, Franco MA, Passerini de Rossi BN. Iron is a signal for Stenotrophomonas maltophilia biofilm formation, oxidative stress response, OMPs expression, and virulence. Front Microbiol 2015; 6:926 [View Article][PubMed]
    [Google Scholar]
  15. Huedo P, Coves X, Daura X, Gibert I, Yero D. Quorum sensing signaling and quenching in the multidrug-resistant pathogen Stenotrophomonas maltophilia . Front Cell Infect Microbiol 2018; 8:122 [View Article][PubMed]
    [Google Scholar]
  16. Sánchez MB. Antibiotic resistance in the opportunistic pathogen Stenotrophomonas maltophilia . Front Microbiol 2015; 6:658 [View Article][PubMed]
    [Google Scholar]
  17. Barbolla R, Catalano M, Orman BE, Famiglietti A, Vay C et al. Class 1 integrons increase trimethoprim-sulfamethoxazole MICs against epidemiologically unrelated Stenotrophomonas maltophilia isolates. Antimicrob Agents Chemother 2004; 48:666–669 [View Article][PubMed]
    [Google Scholar]
  18. Chang Y-T, Lin C-Y, Chen Y-H, Hsueh P-R. Update on infections caused by Stenotrophomonas maltophilia with particular attention to resistance mechanisms and therapeutic options. Front Microbiol 2015; 6:893 [View Article][PubMed]
    [Google Scholar]
  19. San Gabriel P, Zhou J, Tabibi S, Chen Y, Trauzzi M et al. Antimicrobial susceptibility and synergy studies of Stenotrophomonas maltophilia isolates from patients with cystic fibrosis. Antimicrob Agents Chemother 2004; 48:168–171 [View Article][PubMed]
    [Google Scholar]
  20. Valenza G, Tappe D, Turnwald D, Frosch M, König C et al. Prevalence and antimicrobial susceptibility of microorganisms isolated from sputa of patients with cystic fibrosis. J Cyst Fibros 2008; 7:123–127 [View Article][PubMed]
    [Google Scholar]
  21. Wei C, Ni W, Cai X, Zhao J, Cui J. Evaluation of trimethoprim/sulfamethoxazole (SXT), minocycline, tigecycline, moxifloxacin, and ceftazidime alone and in combinations for SXT-susceptible and SXT-resistant Stenotrophomonas maltophilia by in vitro time-kill experiments. PLoS One 2016; 11:e0152132 [View Article][PubMed]
    [Google Scholar]
  22. Folkesson A, Jelsbak L, Yang L, Johansen HK, Ciofu O et al. Adaptation of Pseudomonas aeruginosa to the cystic fibrosis airway: an evolutionary perspective. Nat Rev Microbiol 2012; 10:841–851 [View Article][PubMed]
    [Google Scholar]
  23. Høiby N. Recent advances in the treatment of Pseudomonas aeruginosa infections in cystic fibrosis. BMC Med 2011; 9:32 [View Article][PubMed]
    [Google Scholar]
  24. Cullen L, McClean S. Bacterial adaptation during chronic respiratory infections. Pathogens 2015; 4:66–89 [View Article][PubMed]
    [Google Scholar]
  25. Pompilio A, Pomponio S, Crocetta V, Gherardi G, Verginelli F et al. Phenotypic and genotypic characterization of Stenotrophomonas maltophilia isolates from patients with cystic fibrosis: genome diversity, biofilm formation, and virulence. BMC Microbiol 2011; 11:159 [View Article][PubMed]
    [Google Scholar]
  26. Chung H, Lieberman TD, Vargas SO, Flett KB, McAdam AJ et al. Global and local selection acting on the pathogen Stenotrophomonas maltophilia in the human lung. Nat Commun 2017; 8:14078 [View Article][PubMed]
    [Google Scholar]
  27. Crossman LC, Gould VC, Dow JM, Vernikos GS, Okazaki A et al. The complete genome, comparative and functional analysis of Stenotrophomonas maltophilia reveals an organism heavily shielded by drug resistance determinants. Genome Biol 2008; 9:R74 [View Article][PubMed]
    [Google Scholar]
  28. Huedo Moreno P. Fatty acid-mediated quorum sensing systems in Stenotrophomonas maltophilia . Doctoral thesis Thesis Universitat Autònoma de Barcelona; 2014
    [Google Scholar]
  29. Alcaraz E. Pacientes fibroquísticos colonizados crónicamente por Stenotrophomonas maltophilia: diversidad de expresión de factores de virulencia. Presentación de Casos Clínicos Stenotrophomonas maltophilia y Achromobacter spp: clínica, microbiología e investigación. 5° Congreso Argentino de Fibrosis Quística. Buenos Aires, Argentina: Asociación de Profesionales de la Fibrosis Quística 2019Personal comunication.
    [Google Scholar]
  30. Barber CE, Tang JL, Feng JX, Pan MQ, Wilson TJ et al. A novel regulatory system required for pathogenicity of Xanthomonas campestris is mediated by a small diffusible signal molecule. Mol Microbiol 1997; 24:555–566 [View Article][PubMed]
    [Google Scholar]
  31. CLSI Performance Standards for Antimicrobial Susceptibility Testing, CLSI supplement M100 Clinical and Laboratory Standards Institute, 27th ed. 2017
    [Google Scholar]
  32. Centrón D, Roy PH. Presence of a group II intron in a multiresistant Serratia marcescens strain that harbors three integrons and a novel gene fusion. Antimicrob Agents Chemother 2002; 46:1402–1409 [View Article][PubMed]
    [Google Scholar]
  33. Di Conza J, Ayala JA, Power P, Mollerach M, Gutkind G. Novel class 1 integron (InS21) carrying blaCTX-M-2 in Salmonella enterica serovar infantis. Antimicrob Agents Chemother 2002; 46:2257–2261 [View Article][PubMed]
    [Google Scholar]
  34. Márquez C, Labbate M, Ingold AJ, Roy Chowdhury P, Ramírez MS et al. Recovery of a functional class 2 integron from an Escherichia coli strain mediating a urinary tract infection. Antimicrob Agents Chemother 2008; 52:4153–4154 [View Article][PubMed]
    [Google Scholar]
  35. Turrientes MC, Baquero MR, Sánchez MB, Valdezate S, Escudero E et al. Polymorphic mutation frequencies of clinical and environmental Stenotrophomonas maltophilia populations. Appl Environ Microbiol 2010; 76:1746–1758 [View Article][PubMed]
    [Google Scholar]
  36. Alcaraz E, Garcia C, Papalia M, Vay C, Friedman L et al. Stenotrophomonas maltophilia isolated from patients exposed to invasive devices in a university hospital in Argentina: molecular typing, susceptibility and detection of potential virulence factors. J Med Microbiol 2018; 67:992–1002 [View Article][PubMed]
    [Google Scholar]
  37. Nazik H, Ongen B, Erturan Z, Salcioğlu M. Genotype and antibiotic susceptibility patterns of Pseudomonas aeruginosa and Stenotrophomonas maltophilia isolated from cystic fibrosis patients. Jpn J Infect Dis 2007; 60:82–86[PubMed]
    [Google Scholar]
  38. Ejrnaes K, Sandvang D, Lundgren B, Ferry S, Holm S et al. Pulsed-field gel electrophoresis typing of Escherichia coli strains from samples collected before and after pivmecillinam or placebo treatment of uncomplicated community-acquired urinary tract infection in women. J Clin Microbiol 2006; 44:1776–1781 [View Article][PubMed]
    [Google Scholar]
  39. Stepanović S, Cirković I, Ranin L, Svabić-Vlahović M. Biofilm formation by Salmonella spp. and Listeria monocytogenes on plastic surface. Lett Appl Microbiol 2004; 38:428–432 [View Article][PubMed]
    [Google Scholar]
  40. Rashid MH, Kornberg A. Inorganic polyphosphate is needed for swimming, swarming, and twitching motilities of Pseudomonas aeruginosa . Proc Natl Acad Sci U S A 2000; 97:4885–4890 [View Article][PubMed]
    [Google Scholar]
  41. Quiroga MP, Arduino SM, Merkier AK, Quiroga C, Petroni A et al. "Distribution and functional identification of complex class 1 integrons". Infect Genet Evol 2013; 19:88–96 [View Article][PubMed]
    [Google Scholar]
  42. Colque CA, Albarracín Orio AG, Feliziani S, Marvig RL, Tobares AR et al. Hypermutator Pseudomonas aeruginosa exploits multiple genetic pathways to develop multidrug resistance during long-term infections in the airways of cystic fibrosis patients. Antimicrob Agents Chemother 2020; 64:e02142–19 [View Article][PubMed]
    [Google Scholar]
  43. Carmody LA, Spilker T, LiPuma JJ. Reassessment of Stenotrophomonas maltophilia phenotype. J Clin Microbiol 2011; 49:1101–1103 [View Article][PubMed]
    [Google Scholar]
  44. Madi H, Lukić J, Vasiljević Z, Biočanin M, Kojić M et al. Genotypic and phenotypic characterization of Stenotrophomonas maltophilia strains from a pediatric tertiary care hospital in Serbia. PLoS One 2016; 11:e0165660 [View Article][PubMed]
    [Google Scholar]
  45. Pompilio A, Savini V, Fiscarelli E, Gherardi G, Di Bonaventura G. Clonal Diversity, Biofilm Formation, and Antimicrobial Resistance among Stenotrophomonas maltophilia strains from cystic fibrosis and Non-Cystic Fibrosis Patients. Antibiotics 2020; 9:15 [View Article][PubMed]
    [Google Scholar]
  46. Alcaraz E, Wendorff M, Garcia C, Hernández C, Isasmendi A et al. Stenotrophomonas maltophilia: patógeno emergente en pacientes fibroquísticos pediátricos de Argentina. 5° Congreso Argentino de Fibrosis Quística 2019. Oral comunication. Asociación de Profesionales de la Fibrosis Quística. Buenos Aires, Argentina. URL: https://www.apafiq.org/index.php/congresos/v-congreso-argentino-de-fibrosis-quistica-ano-2019 .
  47. Alcaraz E, García C, Friedman L, de Rossi BP, Passerini de Rossi B. The rpf/DSF signalling system of Stenotrophomonas maltophilia positively regulates biofilm formation, production of virulence-associated factors and β-lactamase induction. FEMS Microbiol Lett 2019; 366:1–10 [View Article]
    [Google Scholar]
  48. Torres PS, Malamud F, Rigano LA, Russo DM, Marano MR et al. Controlled synthesis of the DSF cell-cell signal is required for biofilm formation and virulence in Xanthomonas campestris . Environ Microbiol 2007; 9:2101–2109 [View Article][PubMed]
    [Google Scholar]
  49. Amin R, Waters V. Antibiotic treatment for Stenotrophomonas maltophilia in people with cystic fibrosis. Cochrane Database Syst Rev 2016; 7:CD009249 [View Article][PubMed]
    [Google Scholar]
  50. Barsky EE, Williams KA, Priebe GP, Sawicki GS. Incident Stenotrophomonas maltophilia infection and lung function decline in cystic fibrosis. Pediatr Pulmonol 2017; 52:1276–1282 [View Article][PubMed]
    [Google Scholar]
  51. Lieberman TD, Flett KB, Yelin I, Martin TR, McAdam AJ et al. Genetic variation of a bacterial pathogen within individuals with cystic fibrosis provides a record of selective pressures. Nat Genet 2014; 46:82–87 [View Article][PubMed]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/jmm/10.1099/jmm.0.001281
Loading
/content/journal/jmm/10.1099/jmm.0.001281
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