1887

Abstract

Although infections caused by have historically been attributed to hospital acquisition, growing evidence supports the role of community acquisition in infection (CDI). Symptoms of CDI can range from mild, self-resolving diarrhoea to toxic megacolon, pseudomembranous colitis, and death. In this study, we sampled from clinical, environmental, and canine reservoirs in Flagstaff, Arizona, USA, to understand the distribution and transmission of the pathogen in a One Health framework; Flagstaff is a medium-sized, geographically isolated city with a single hospital system, making it an ideal site to characterize genomic overlap between sequenced isolates across reservoirs. An analysis of 562 genomes from Flagstaff isolates identified 65 sequence types (STs), with eight STs being found across all three reservoirs and another nine found across two reservoirs. A screen of toxin genes in the pathogenicity locus identified nine STs where all isolates lost the toxin genes needed for CDI manifestation (, ), demonstrating the widespread distribution of non-toxigenic (NTCD) isolates in all three reservoirs; 15 NTCD genomes were sequenced from symptomatic, clinical samples, including two from mixed infections that contained both and - isolates. A comparative single nucleotide polymorphism (SNP) analysis of clinically derived isolates identified 78 genomes falling within clusters separated by ≤2 SNPs, indicating that ~19 % of clinical isolates are associated with potential healthcare-associated transmission clusters; only symptomatic cases were sampled in this study, and we did not sample asymptomatic transmission. Using this same SNP threshold, we identified genomic overlap between canine and soil isolates, as well as putative transmission between environmental and human reservoirs. The core genome of isolates sequenced in this study plus a representative set of public genomes (=136), was 2690 coding region sequences, which constitutes ~70 % of an individual genome; this number is significantly higher than has been published in some other studies, suggesting that genome data quality is important in understanding the minimal number of genes needed by . This study demonstrates the close genomic overlap among isolates sampled across reservoirs, which was facilitated by maximizing the genomic search space used for comprehensive identification of potential transmission events. Understanding the distribution of toxigenic and non-toxigenic across reservoirs has implications for surveillance sampling strategies, characterizing routes of infections, and implementing mitigation measures to limit human infection.

Funding
This study was supported by the:
  • Flinn Foundation (Award 2234)
    • Principle Award Recipient: JasonW Sahl
  • Flinn Foundation (Award 2062)
    • Principle Award Recipient: PaulKeim
  • This is an open-access article distributed under the terms of the Creative Commons Attribution License.
Loading

Article metrics loading...

/content/journal/mgen/10.1099/mgen.0.001046
2023-06-22
2024-05-03
Loading full text...

Full text loading...

/deliver/fulltext/mgen/9/6/mgen001046.html?itemId=/content/journal/mgen/10.1099/mgen.0.001046&mimeType=html&fmt=ahah

References

  1. George WL, Sutter VL, Goldstein EJ, Ludwig SL, Finegold SM. Aetiology of antimicrobial-agent-associated colitis. Lancet 1978; 1:802–803 [View Article] [PubMed]
    [Google Scholar]
  2. George RH, Symonds JM, Dimock F, Brown JD, Arabi Y et al. Identification of Clostridium difficile as a cause of pseudomembranous colitis. Br Med J 1978; 1:695 [View Article] [PubMed]
    [Google Scholar]
  3. Larson HE, Price AB, Honour P, Borriello SP. Clostridium difficile and the aetiology of pseudomembranous colitis. Lancet 1978; 1:1063–1066 [View Article] [PubMed]
    [Google Scholar]
  4. Bartlett JG, Moon N, Chang TW, Taylor N, Onderdonk AB. Role of Clostridium difficile in antibiotic-associated pseudomembranous colitis. Gastroenterology 1978; 75:778–782 [View Article] [PubMed]
    [Google Scholar]
  5. Hammond GA, Johnson JL. The toxigenic element of Clostridium difficile strain VPI 10463. Microb Pathog 1995; 19:203–213 [View Article] [PubMed]
    [Google Scholar]
  6. Dingle KE, Elliott B, Robinson E, Griffiths D, Eyre DW et al. Evolutionary history of the Clostridium difficile pathogenicity locus. Genome Biol Evol 2014; 6:36–52 [View Article] [PubMed]
    [Google Scholar]
  7. Mani N, Dupuy B. Regulation of toxin synthesis in Clostridium difficile by an alternative RNA polymerase sigma factor. Proc Natl Acad Sci 2001; 98:5844–5849 [View Article] [PubMed]
    [Google Scholar]
  8. Matamouros S, England P, Dupuy B. Clostridium difficile toxin expression is inhibited by the novel regulator TcdC. Mol Microbiol 2007; 64:1274–1288 [View Article] [PubMed]
    [Google Scholar]
  9. Tan KS, Wee BY, Song KP. Evidence for holin function of tcdE gene in the pathogenicity of Clostridium difficile. J Med Microbiol 2001; 50:613–619 [View Article] [PubMed]
    [Google Scholar]
  10. Gerding DN, Johnson S, Rupnik M, Aktories K. Clostridium difficile binary toxin CDT: mechanism, epidemiology, and potential clinical importance. Gut Microbes 2014; 5:15–27 [View Article] [PubMed]
    [Google Scholar]
  11. Brazier JS. The epidemiology and typing of Clostridium difficile. J Antimicrob Chemother 1998; 41 Suppl 3:47–57 [View Article] [PubMed]
    [Google Scholar]
  12. Kuijper EJ, Coignard B, Tüll P et al. Emergence of Clostridium difficile-associated disease in North America and Europe. Clin Microbiol Infect 2006; 12 Suppl 6:2–18 [View Article] [PubMed]
    [Google Scholar]
  13. Eyre DW, Cule ML, Wilson DJ, Griffiths D, Vaughan A et al. Diverse sources of C. difficile infection identified on whole-genome sequencing. N Engl J Med 2013; 369:1195–1205 [View Article] [PubMed]
    [Google Scholar]
  14. Dingle KE, Didelot X, Quan TP, Eyre DW, Stoesser N et al. Effects of control interventions on Clostridium difficile infection in England: an observational study. Lancet Infect Dis 2017; 17:411–421 [View Article] [PubMed]
    [Google Scholar]
  15. Feazel LM, Malhotra A, Perencevich EN, Kaboli P, Diekema DJ et al. Effect of antibiotic stewardship programmes on Clostridium difficile incidence: a systematic review and meta-analysis. J Antimicrob Chemother 2014; 69:1748–1754 [View Article] [PubMed]
    [Google Scholar]
  16. Smits WK, Lyras D, Lacy DB, Wilcox MH, Kuijper EJ. Clostridium difficile infection. Nat Rev Dis Primers 2016; 2:16020 [View Article] [PubMed]
    [Google Scholar]
  17. Lim SC, Knight DR, Riley TV. Clostridium difficile and one health. Clin Microbiol Infect 2020; 26:857–863 [View Article] [PubMed]
    [Google Scholar]
  18. Ofori E, Ramai D, Dhawan M, Mustafa F, Gasperino J et al. Community-acquired Clostridium difficile: epidemiology, ribotype, risk factors, hospital and intensive care unit outcomes, and current and emerging therapies. J Hosp Infect 2018; 99:436–442 [View Article] [PubMed]
    [Google Scholar]
  19. Khanna S, Pardi DS, Aronson SL, Kammer PP, Orenstein R et al. The epidemiology of community-acquired Clostridium difficile infection: a population-based study. Am J Gastroenterol 2012; 107:89–95 [View Article] [PubMed]
    [Google Scholar]
  20. Hensgens MPM, Keessen EC, Squire MM, Riley TV, Koene MGJ et al. Clostridium difficile infection in the community: a zoonotic disease?. Clin Microbiol Infect 2012; 18:635–645 [View Article] [PubMed]
    [Google Scholar]
  21. Rodriguez-Palacios A, Stämpfli HR, Duffield T, Peregrine AS, Trotz-Williams LA et al. Clostridium difficile PCR ribotypes in calves, Canada. Emerg Infect Dis 2006; 12:1730–1736 [View Article] [PubMed]
    [Google Scholar]
  22. Knight DR, Thean S, Putsathit P, Fenwick S, Riley TV. Cross-sectional study reveals high prevalence of Clostridium difficile non-PCR ribotype 078 strains in Australian veal calves at slaughter. Appl Environ Microbiol 2013; 79:2630–2635 [View Article] [PubMed]
    [Google Scholar]
  23. Rodriguez C, Hakimi D-E, Vanleyssem R, Taminiau B, Van Broeck J et al. Clostridium difficile in beef cattle farms, farmers and their environment: assessing the spread of the bacterium. Vet Microbiol 2017; 210:183–187 [View Article] [PubMed]
    [Google Scholar]
  24. Hain-Saunders NMR, Knight DR, Bruce M, Riley TV. Clostridioides difficile infection and one health: an equine perspective. Environ Microbiol 2022; 24:985–997 [View Article] [PubMed]
    [Google Scholar]
  25. Knight DR, Squire MM, Riley TV. Nationwide surveillance study of Clostridium difficile in Australian neonatal pigs shows high prevalence and heterogeneity of PCR ribotypes. Appl Environ Microbiol 2015; 81:119–123 [View Article] [PubMed]
    [Google Scholar]
  26. Keel K, Brazier JS, Post KW, Weese S, Songer JG. Prevalence of PCR ribotypes among Clostridium difficile isolates from pigs, calves, and other species. J Clin Microbiol 2007; 45:1963–1964 [View Article] [PubMed]
    [Google Scholar]
  27. Zidaric V, Zemljic M, Janezic S, Kocuvan A, Rupnik M. High diversity of Clostridium difficile genotypes isolated from a single poultry farm producing replacement laying hens. Anaerobe 2008; 14:325–327 [View Article] [PubMed]
    [Google Scholar]
  28. Stone NE, Sidak-Loftis LC, Sahl JW, Vazquez AJ, Wiggins KB et al. More than 50% of Clostridium difficile isolates from pet dogs in Flagstaff, USA, carry toxigenic genotypes. PLoS One 2016; 11:e0164504 [View Article] [PubMed]
    [Google Scholar]
  29. Rabold D, Espelage W, Abu Sin M, Eckmanns T, Schneeberg A et al. The zoonotic potential of Clostridium difficile from small companion animals and their owners. PLoS One 2018; 13:e0193411 [View Article] [PubMed]
    [Google Scholar]
  30. Rodriguez C, Taminiau B, Bouchafa L, Romijn S, Van Broeck J et al. Clostridium difficile beyond stools: dog nasal discharge as a possible new vector of bacterial transmission. Heliyon 2019; 5:e01629 [View Article] [PubMed]
    [Google Scholar]
  31. Orden C, Neila C, Blanco JL, Álvarez-Pérez S, Harmanus C et al. Recreational sandboxes for children and dogs can be a source of epidemic ribotypes of clostridium difficile. Zoonoses Public Health 2018; 65:88–95 [View Article] [PubMed]
    [Google Scholar]
  32. Shivaperumal N, Chang BJ, Riley TV. High prevalence of clostridium difficile in home gardens in western Australia. Appl Environ Microbiol 2020; 87:e01572-20 [View Article] [PubMed]
    [Google Scholar]
  33. Zidaric V, Beigot S, Lapajne S, Rupnik M. The occurrence and high diversity of clostridium difficile genotypes in rivers. Anaerobe 2010; 16:371–375 [View Article] [PubMed]
    [Google Scholar]
  34. Janezic S, Potocnik M, Zidaric V, Rupnik M. Highly divergent clostridium difficile strains isolated from the environment. PLoS One 2016; 11:e0167101 [View Article] [PubMed]
    [Google Scholar]
  35. Janezic S, Smrke J, Rupnik M. Isolation of Clostridioides difficile from different outdoor sites in the domestic environment. Anaerobe 2020; 62:102183 [View Article] [PubMed]
    [Google Scholar]
  36. Lim S-C, Hain-Saunders NMR, Imwattana K, Putsathit P, Collins DA et al. Genetically related Clostridium difficile from water sources and human CDI cases revealed by whole-genome sequencing. Environ Microbiol 2022; 24:1221–1230 [View Article] [PubMed]
    [Google Scholar]
  37. Rodriguez-Palacios A, Lejeune JT. Moist-heat resistance, spore aging, and superdormancy in Clostridium difficile. Appl Environ Microbiol 2011; 77:3085–3091 [View Article] [PubMed]
    [Google Scholar]
  38. Knight DR, Kullin B, Androga GO, Barbut F, Eckert C et al. Evolutionary and genomic insights into Clostridioides difficile sequence type 11: a diverse zoonotic and antimicrobial-resistant lineage of global one health importance. mBio 2019; 10:e00446-19 [View Article] [PubMed]
    [Google Scholar]
  39. Knight DR, Riley TV. Genomic delineation of zoonotic origins of. Front Public Health 2019; 7:164
    [Google Scholar]
  40. Knetsch CW, Kumar N, Forster SC, Connor TR, Browne HP et al. Zoonotic transfer of clostridium difficile harboring antimicrobial resistance between farm animals and humans. J Clin Microbiol 2018; 56:e01384-17 [View Article] [PubMed]
    [Google Scholar]
  41. Turner NA, Smith BA, Lewis SS. Novel and emerging sources of Clostridioides difficile infection. PLoS Pathog 2019; 15:e1008125 [View Article] [PubMed]
    [Google Scholar]
  42. Zhou Y, Zhou W, Xiao T, Chen Y, Lv T et al. Comparative genomic and transmission analysis of Clostridioides difficile between environmental, animal, and clinical sources in China. Emerg Microbes Infect 2021; 10:2244–2255 [View Article] [PubMed]
    [Google Scholar]
  43. Knetsch CW, Connor TR, Mutreja A, van Dorp SM, Sanders IM et al. Whole genome sequencing reveals potential spread of clostridium difficile between humans and farm animals in the Netherlands, 2002 to 2011. Euro Surveill 2014; 19:20954 [View Article] [PubMed]
    [Google Scholar]
  44. Rodríguez-Pallares S, Fernández-Palacios P, Jurado-Tarifa E, Arroyo F, Rodríguez-Iglesias MA et al. Transmission of toxigenic Clostridiodes difficile between a pet dog with diarrhea and a 10-month-old infant. Anaerobe 2022; 74:102519 [View Article] [PubMed]
    [Google Scholar]
  45. Bidet P, Barbut F, Lalande V, Burghoffer B, Petit JC. Development of a new PCR-ribotyping method for clostridium difficile based on ribosomal RNA gene sequencing. FEMS Microbiol Lett 1999; 175:261–266 [View Article] [PubMed]
    [Google Scholar]
  46. Knetsch CW, Lawley TD, Hensgens MP, Corver J, Wilcox MW et al. Current application and future perspectives of molecular typing methods to study clostridium difficile infections. Euro Surveill 2013; 18:20381 [View Article] [PubMed]
    [Google Scholar]
  47. Stabler RA, He M, Dawson L, Martin M, Valiente E et al. Comparative genome and phenotypic analysis of clostridium difficile 027 strains provides insight into the evolution of a hypervirulent bacterium. Genome Biol 2009; 10:R102 [View Article] [PubMed]
    [Google Scholar]
  48. Dingle KE, Griffiths D, Didelot X, Evans J, Vaughan A et al. Clinical clostridium difficile: clonality and pathogenicity locus diversity. PLoS One 2011; 6:e19993 [View Article] [PubMed]
    [Google Scholar]
  49. Griffiths D, Fawley W, Kachrimanidou M, Bowden R, Crook DW et al. Multilocus sequence typing of Clostridium difficile. J Clin Microbiol 2010; 48:770–778 [View Article] [PubMed]
    [Google Scholar]
  50. Janezic S, Rupnik M. Genomic diversity of Clostridium difficile strains. Res Microbiol 2015; 166:353–360 [View Article] [PubMed]
    [Google Scholar]
  51. Bletz S, Janezic S, Harmsen D, Rupnik M, Mellmann A. Defining and evaluating a core genome multilocus sequence typing scheme for genome-wide typing of Clostridium difficile. J Clin Microbiol 2018; 56:e01987-17 [View Article] [PubMed]
    [Google Scholar]
  52. Scaria J, Ponnala L, Janvilisri T, Yan W, Mueller LA et al. Analysis of ultra low genome conservation in Clostridium difficile. PLoS One 2010; 5:e15147 [View Article] [PubMed]
    [Google Scholar]
  53. O’Grady K, Knight DR, Riley TV. Antimicrobial resistance in Clostridioides difficile. Eur J Clin Microbiol Infect Dis 2021; 40:2459–2478 [View Article] [PubMed]
    [Google Scholar]
  54. Lewis BB, Carter RA, Ling L, Leiner I, Taur Y et al. Pathogenicity locus, core genome, and accessory Gene contributions to virulence. MBio 2017; 8:
    [Google Scholar]
  55. Williamson CHD, Stone NE, Nunnally AE, Roe CC, Vazquez AJ et al. Identification of novel, cryptic Clostridioides species isolates from environmental samples collected from diverse geographical locations. Microb Genom 2022; 8:000742 [View Article] [PubMed]
    [Google Scholar]
  56. Pritchard L, Glover RH, Humphris S, Elphinstone JG, Toth IK. Genomics and taxonomy in diagnostics for food security: soft-rotting enterobacterial plant pathogens. Anal Methods 2016; 8:12–24 [View Article]
    [Google Scholar]
  57. 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]
  58. Williamson CHD, Stone NE, Nunnally AE, Hornstra HM, Wagner DM et al. A global to local genomics analysis of Clostridioides difficile ST1/RT027 identifies cryptic transmission events in a northern Arizona healthcare network. Microb Genom 2019; 5:e000271 [View Article] [PubMed]
    [Google Scholar]
  59. Bankevich A, Nurk S, Antipov D, Gurevich AA, Dvorkin M et al. SPAdes: a new genome assembly algorithm and its applications to single-cell sequencing. J Comput Biol 2012; 19:455–477 [View Article] [PubMed]
    [Google Scholar]
  60. Walker BJ, Abeel T, Shea T, Priest M, Abouelliel A et al. Pilon: an integrated tool for comprehensive microbial variant detection and genome assembly improvement. PLoS One 2014; 9:e112963 [View Article] [PubMed]
    [Google Scholar]
  61. Li H. Minimap2: pairwise alignment for nucleotide sequences. Bioinformatics 2009; 25:3094–3100 [View Article] [PubMed]
    [Google Scholar]
  62. Li H, Handsaker B, Wysoker A, Fennell T, Ruan J et al. The sequence alignment/Map format and SAMtools. Bioinformatics 2009; 25:2078–2079 [View Article] [PubMed]
    [Google Scholar]
  63. Sayers EW, Agarwala R, Bolton EE, Brister JR, Canese K et al. Database resources of the national center for biotechnology information. Nucleic Acids Res 2019; 47:D23–D28 [View Article] [PubMed]
    [Google Scholar]
  64. Camacho C, Coulouris G, Avagyan V, Ma N, Papadopoulos J et al. BLAST+: architecture and applications. BMC Bioinformatics 2009; 10:421 [View Article] [PubMed]
    [Google Scholar]
  65. Stone NE, Nunnally AE, Jimenez V, Cope EK, Sahl JW et al. Domestic canines do not display evidence of gut microbial dysbiosis in the presence of Clostridioides (Clostridium) difficile, despite cellular susceptibility to its toxins. Anaerobe 2019; 58:53–72
    [Google Scholar]
  66. Souvorov A, Agarwala R, Lipman DJ. SKESA: strategic k-mer extension for scrupulous assemblies. Genome Biol 2018; 19:153 [View Article] [PubMed]
    [Google Scholar]
  67. Kurtz S, Phillippy A, Delcher AL, Smoot M, Shumway M et al. Versatile and open software for comparing large genomes. Genome Biol 2004; 5:R12 [View Article] [PubMed]
    [Google Scholar]
  68. Knight DR, Imwattana K, Kullin B, Guerrero-Araya E, Paredes-Sabja D et al. Major genetic discontinuity and novel toxigenic species in Clostridioides difficile taxonomy. Elife 2021; 10: [View Article]
    [Google Scholar]
  69. Guerrero-Araya E, Muñoz M, Rodríguez C, Paredes-Sabja D. FastMLST: a multi-core tool for multilocus sequence typing of draft genome assemblies. Bioinform Biol Insights 2021; 15:11779322211059238 [View Article] [PubMed]
    [Google Scholar]
  70. Sahl JW, Lemmer D, Travis J, Schupp JM, Gillece JD et al. NASP: an accurate, rapid method for the identification of SNPs in WGS datasets that supports flexible input and output formats. Microb Genom 2016; 2:e000074 [View Article] [PubMed]
    [Google Scholar]
  71. Minh BQ, Schmidt HA, Chernomor O, Schrempf D, Woodhams MD et al. Corrigendum to: IQ-TREE 2: new models and efficient methods for phylogenetic inference in the genomic era. Mol Biol Evol 2020; 37:2461 [View Article] [PubMed]
    [Google Scholar]
  72. Kalyaanamoorthy S, Minh BQ, Wong TKF, von Haeseler A, Jermiin LS. ModelFinder: fast model selection for accurate phylogenetic estimates. Nat Methods 2017; 14:587–589 [View Article] [PubMed]
    [Google Scholar]
  73. Letunic I, Bork P. Interactive tree of life (iTOL) v3: an online tool for the display and annotation of phylogenetic and other trees. Nucleic Acids Res 2016; 44:W242–5 [View Article] [PubMed]
    [Google Scholar]
  74. McKenna A, Hanna M, Banks E, Sivachenko A, Cibulskis K et al. The genome analysis toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res 2010; 20:1297–1303 [View Article] [PubMed]
    [Google Scholar]
  75. Schwengers O, Jelonek L, Dieckmann MA, Beyvers S, Blom J et al. Bakta: rapid and standardized annotation of bacterial genomes via alignment-free sequence identification. Microb Genom 2021; 7:000685 [View Article] [PubMed]
    [Google Scholar]
  76. Tonkin-Hill G, MacAlasdair N, Ruis C, Weimann A, Horesh G et al. Producing polished prokaryotic pangenomes with the Panaroo pipeline. Genome Biol 2020; 21:180 [View Article] [PubMed]
    [Google Scholar]
  77. Sahl JW, Caporaso JG, Rasko DA, Keim P. The large-scale blast score ratio (LS-BSR) pipeline: a method to rapidly compare genetic content between bacterial genomes. PeerJ 2014; 2:e332 [View Article] [PubMed]
    [Google Scholar]
  78. Kent WJ. BLAT--the BLAST-like alignment tool. Genome Res 2002; 12:656–664 [View Article] [PubMed]
    [Google Scholar]
  79. Rasko DA, Myers GSA, Ravel J. Visualization of comparative genomic analyses by BLAST score ratio. BMC Bioinformatics 2005; 6:2 [View Article] [PubMed]
    [Google Scholar]
  80. Roberts MC. Tetracycline resistance determinants: mechanisms of action, regulation of expression, genetic mobility, and distribution. FEMS Microbiol Rev 1996; 19:1–24 [View Article] [PubMed]
    [Google Scholar]
  81. Banawas SS. Infections: a global overview of drug sensitivity and resistance mechanisms. Biomed Res Int 2018; 2018:8414257
    [Google Scholar]
  82. Imwattana K, Putsathit P, Knight DR, Kiratisin P, Riley TV. Molecular characterization of, and antimicrobial resistance in, Clostridioides difficile from Thailand, 2017-2018. Microb Drug Resist 2021; 27:1505–1512 [View Article] [PubMed]
    [Google Scholar]
  83. Cizek A, Masarikova M, Mares J, Brajerova M, Krutova M. Detection of plasmid-mediated resistance to metronidazole in Clostridioides difficile from river water. Microbiol Spectr 2022; 10:e0080622 [View Article] [PubMed]
    [Google Scholar]
  84. Roxas BAP, Roxas JL, Claus-Walker R, Harishankar A, Mansoor A et al. Phylogenomic analysis of Clostridioides difficile ribotype 106 strains reveals novel genetic islands and emergent phenotypes. Sci Rep 2020; 10:22135 [View Article] [PubMed]
    [Google Scholar]
  85. Lim S-C, Collins DA, Imwattana K, Knight DR, Perumalsamy S et al. Whole-genome sequencing links clostridium (Clostridioides) difficile in a single hospital to diverse environmental sources in the community. J Appl Microbiol 2022; 133:1156–1168 [View Article] [PubMed]
    [Google Scholar]
  86. Balaji A, Ozer EA, Kociolek LK. Clostridioides difficile whole-genome sequencing reveals limited within-host genetic diversity in a pediatric cohort. J Clin Microbiol 2019; 57:e00559-19 [View Article] [PubMed]
    [Google Scholar]
  87. Miles-Jay A, Young VB, Pamer EG, Savidge TC, Kamboj M et al. A multisite genomic epidemiology study of Clostridioides difficile infections in the USA supports differential roles of healthcare versus community spread for two common strains. Microb Genom 2021; 7:000590 [View Article] [PubMed]
    [Google Scholar]
  88. Cariss SJL, Constantinidou C, Patel MD, Takebayashi Y, Hobman JL et al. YieJ (CbrC) mediates CreBC-dependent colicin E2 tolerance in Escherichia coli. J Bacteriol 2010; 192:3329–3336 [View Article] [PubMed]
    [Google Scholar]
  89. Jobe BA, Grasley A, Deveney KE, Deveney CW, Sheppard BC. Clostridium difficile colitis: an increasing hospital-acquired illness. Am J Surg 1995; 169:480–483 [View Article] [PubMed]
    [Google Scholar]
  90. Gupta A, Khanna S. Community-acquired clostridium difficile infection: an increasing public health threat. Infect Drug Resist 2014; 7:63–72 [View Article] [PubMed]
    [Google Scholar]
  91. Candel-Pérez C, Ros-Berruezo G, Martínez-Graciá C. A review of Clostridioides [Clostridium] difficile occurrence through the food chain. Food Microbiol 2019; 77:118–129 [View Article] [PubMed]
    [Google Scholar]
  92. Janezic S, Mlakar S, Rupnik M. Dissemination of clostridium difficile spores between environment and households: dog paws and shoes. Zoonoses Public Health 2018; 65:669–674 [View Article] [PubMed]
    [Google Scholar]
  93. Clooten J, Kruth S, Arroyo L, Weese JS. Prevalence and risk factors for clostridium difficile colonization in dogs and cats hospitalized in an intensive care unit. Vet Microbiol 2008; 129:209–214 [View Article] [PubMed]
    [Google Scholar]
  94. Davis GS, Waits K, Nordstrom L, Weaver B, Aziz M et al. Intermingled Klebsiella pneumoniae populations between retail meats and human urinary tract infections. Clin Infect Dis 2015; 61:892–899 [View Article] [PubMed]
    [Google Scholar]
  95. Behroozian AA, Chludzinski JP, Lo ES, Ewing SA, Waslawski S et al. Detection of mixed populations of clostridium difficile from symptomatic patients using capillary-based polymerase chain reaction ribotyping. Infect Control Hosp Epidemiol 2013; 34:961–966 [View Article] [PubMed]
    [Google Scholar]
  96. Bjöersdorff OG, Lindberg S, Kiil K, Persson S, Guardabassi L et al. Dogs are carriers of Clostridioides difficile lineages associated with human community-acquired infections. Anaerobe 2021; 67:102317 [View Article] [PubMed]
    [Google Scholar]
  97. Alves F, Nunes A, Castro R, Sequeira A, Moreira O et al. Assessment of the transmission dynamics of Clostridioides difficile in a farm environment reveals the presence of a new toxigenic strain connected to swine production. Front Microbiol 2022; 13:858310 [View Article] [PubMed]
    [Google Scholar]
  98. Wilson KH, Sheagren JN. Antagonism of toxigenic clostridium difficile by nontoxigenic C. difficile. J Infect Dis 1983; 147:733–736 [View Article] [PubMed]
    [Google Scholar]
  99. Borriello SP, Barclay FE. Protection of hamsters against clostridium difficile ileocaecitis by prior colonisation with non-pathogenic strains. J Med Microbiol 1985; 19:339–350 [View Article] [PubMed]
    [Google Scholar]
  100. Natarajan M, Walk ST, Young VB, Aronoff DM. A clinical and epidemiological review of non-toxigenic Clostridium difficile. Anaerobe 2013; 22:1–5 [View Article] [PubMed]
    [Google Scholar]
  101. Villano SA, Seiberling M, Tatarowicz W, Monnot-Chase E, Gerding DN. Evaluation of an oral suspension of VP20621, spores of nontoxigenic Clostridium difficile strain M3, in healthy subjects. Antimicrob Agents Chemother 2012; 56:5224–5229 [View Article] [PubMed]
    [Google Scholar]
  102. Nagaro KJ, Phillips ST, Cheknis AK, Sambol SP, Zukowski WE et al. Nontoxigenic Clostridium difficile protects hamsters against challenge with historic and epidemic strains of toxigenic BI/NAP1/027 C. difficile. Antimicrob Agents Chemother 2013; 57:5266–5270 [View Article] [PubMed]
    [Google Scholar]
  103. Gerding DN, Sambol SP, Johnson S. Non-toxigenic Clostridioides (Formerly Clostridium) difficile for prevention of C. difficile infection: from bench to bedside back to bench and back to bedside. Front Microbiol 2018; 9:1700 [View Article]
    [Google Scholar]
  104. Seth-Smith HMB, Biggel M, Roloff T, Hinic V, Bodmer T et al. Transition from PCR-ribotyping to whole genome sequencing based typing of Clostridioides difficile. Front Cell Infect Microbiol 2021; 11:681518 [View Article]
    [Google Scholar]
  105. Ngamskulrungroj P, Sanmee S, Putsathit P, Piewngam P, Elliott B et al. Correction: molecular epidemiology of Clostridium difficile infection in a large teaching hospital in Thailand. PLoS One 2015; 10:e0134771 [View Article] [PubMed]
    [Google Scholar]
  106. Sahl JW, Matalka MN, Rasko DA. Phylomark, a tool to identify conserved phylogenetic markers from whole-genome alignments. Appl Environ Microbiol 2012; 78:4884–4892 [View Article] [PubMed]
    [Google Scholar]
  107. Ducarmon QR, van der Bruggen T, Harmanus C, Sanders IMJG, Daenen LGM et al. Clostridioides difficile PCR ribotype 151 is polyphyletic and includes pathogenic isolates from cryptic clade C-II with mono-toxin pathogenicity loci that can escape routine diagnostics. bioRxiv 2022 [View Article]
    [Google Scholar]
  108. Jolley KA, Bliss CM, Bennett JS, Bratcher HB, Brehony C et al. Ribosomal multilocus sequence typing: universal characterization of bacteria from domain to strain. Microbiology 2012; 158:1005–1015 [View Article] [PubMed]
    [Google Scholar]
  109. García-Fernández S, Frentrup M, Steglich M, Gonzaga A, Cobo M et al. Whole-genome sequencing reveals nosocomial Clostridioides difficile transmission and a previously unsuspected epidemic scenario. Sci Rep 2019; 9:6959 [View Article] [PubMed]
    [Google Scholar]
  110. Frentrup M, Zhou Z, Steglich M, Meier-Kolthoff JP, Göker M et al. A publicly accessible database for Clostridioides difficile genome sequences supports tracing of transmission chains and epidemics. Microb Genom 2020; 6:mgen000410 [View Article] [PubMed]
    [Google Scholar]
  111. Mawer DPC, Eyre DW, Griffiths D, Fawley WN, Martin JSH et al. Contribution to clostridium difficile transmission of symptomatic patients with toxigenic strains who are fecal toxin negative. Clin Infect Dis 2017; 64:1163–1170 [View Article] [PubMed]
    [Google Scholar]
  112. Weller C, Wu M. A generation-time effect on the rate of molecular evolution in bacteria. Evolution 2015; 69:643–652 [View Article] [PubMed]
    [Google Scholar]
  113. Callister SJ, McCue LA, Turse JE, Monroe ME, Auberry KJ et al. Comparative bacterial proteomics: analysis of the core genome concept. PLoS One 2008; 3:e1542 [View Article] [PubMed]
    [Google Scholar]
  114. Forgetta V, Oughton MT, Marquis P, Brukner I, Blanchette R et al. Fourteen-genome comparison identifies DNA markers for severe-disease-associated strains of clostridium difficile. J Clin Microbiol 2011; 49:2230–2238 [View Article] [PubMed]
    [Google Scholar]
  115. Bertels F, Silander OK, Pachkov M, Rainey PB, van Nimwegen E. Automated reconstruction of whole-genome phylogenies from short-sequence reads. Mol Biol Evol 2014; 31:1077–1088 [View Article] [PubMed]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/mgen/10.1099/mgen.0.001046
Loading
/content/journal/mgen/10.1099/mgen.0.001046
Loading

Data & Media loading...

Supplements

Supplementary material 1

PDF

Supplementary material 2

EXCEL
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