1887

Abstract

Trachoma, a neglected tropical disease caused by (Ct) serovars A–C, is the leading infectious cause of blindness worldwide. Africa bears the highest burden, accounting for over 86 % of global trachoma cases. We investigated Ct serovar A (SvA) and B (SvB) whole genome sequences prior to the induction of mass antibiotic drug administration in The Gambia. Here, we explore the factors contributing to Ct strain diversification and the implications for Ct evolution within the context of ocular infection. A cohort study in 2002–2003 collected ocular swabs across nine Gambian villages during a 6 month follow-up study. To explore the genetic diversity of Ct within and between individuals, we conducted whole-genome sequencing (WGS) on a limited number (=43) of Ct-positive samples with an B load ≥10 from four villages. WGS was performed using target enrichment with SureSelect and Illumina paired-end sequencing. Out of 43 WGS samples, 41 provided sufficient quality for further analysis. A analysis revealed that 11 samples had highest identity to A from strain A/HAR13 (NC_007429) and 30 had highest identity to A from strain B/Jali20 (NC_012686). While SvB genome sequences formed two distinct village-driven subclades, the heterogeneity of SvA sequences led to the formation of many individual branches within the Gambian SvA subclade. Comparing the Gambian SvA and SvB sequences with their reference strains, Ct A/HAR13 and Ct B/Jali20, indicated an single nucleotide polymorphism accumulation rate of 2.4×10 per site per year for the Gambian SvA and 1.3×10 per site per year for SvB variants (<0.0001). Variant calling resulted in a total of 1371 single nucleotide variants (SNVs) with a frequency >25 % in SvA sequences, and 438 SNVs in SvB sequences. Of note, in SvA variants, highest evolutionary pressure was recorded on genes responsible for host cell modulation and intracellular survival mechanisms, whereas in SvB variants this pressure was mainly on genes essential for DNA replication/repair mechanisms and protein synthesis. A comparison of the sequences between observed separate infection events (4–20 weeks between infections) suggested that the majority of the variations accumulated in genes responsible for host–pathogen interaction such as CTA_0166 (phospholipase D-like protein), CTA_0498 (TarP) and CTA_0948 (deubiquitinase). This comparison of Ct SvA and SvB variants within a trachoma endemic population focused on their local evolutionary adaptation. We found a different variation accumulation pattern in the Gambian SvA chromosomal genes compared with SvB, hinting at the potential of Ct serovar-specific variation in diversification and evolutionary fitness. These findings may have implications for optimizing trachoma control and prevention strategies.

Funding
This study was supported by the:
  • Horizon 2020 Framework Programme (Award 733373)
    • Principle Award Recipient: MartinJ Holland
  • Wellcome Trust (Award 079246/Z/06)
    • Principle Award Recipient: MartinJ Holland
  • Medical Research Council (Award G9826361/ ID48103)
    • Principle Award Recipient: MartinJ Holland
  • Austrian Science Fund (Award J-4608)
    • Principle Award Recipient: EhsanGhasemian
  • 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/mgen/10.1099/mgen.0.001210
2024-03-06
2024-04-27
Loading full text...

Full text loading...

/deliver/fulltext/mgen/10/3/mgen001210.html?itemId=/content/journal/mgen/10.1099/mgen.0.001210&mimeType=html&fmt=ahah

References

  1. Solomon AW, Burton MJ, Gower EW, Harding-Esch EM, Oldenburg CE et al. Trachoma. Nat Rev Dis Primers 2022; 8:32 [View Article] [PubMed]
    [Google Scholar]
  2. Taylor HR, Burton MJ, Haddad D, West S, Wright H. Trachoma. Lancet 2014; 384:2142–2152 [View Article] [PubMed]
    [Google Scholar]
  3. Pickering H, Chernet A, Sata E, Zerihun M, Williams CA et al. Genomics of ocular Chlamydia trachomatis after 5 years of SAFE interventions for Trachoma in Amhara, Ethiopia. J Infect Dis 2022; 225:994–1004 [View Article] [PubMed]
    [Google Scholar]
  4. Andreasen AA, Burton MJ, Holland MJ, Polley S, Faal N et al. Chlamydia trachomatis ompA variants in trachoma: what do they tell us?. PLoS Negl Trop Dis 2008; 2:e306 [View Article] [PubMed]
    [Google Scholar]
  5. Wolle MA, West SK. Ocular Chlamydia trachomatis infection: elimination with mass drug administration. Expert Rev Anti Infect Ther 2019; 17:189–200 [View Article] [PubMed]
    [Google Scholar]
  6. Trachoma WHO; 2022 https://www.Who.Int/News-Room/Fact-Sheets/Detail/Trachoma accessed 5 October 2022
  7. WHO alliance for the global elimination of Trachoma: progress report on elimination of Trachoma, 2022. Wkly Epidemiol Rec 2023; 28:297–314
    [Google Scholar]
  8. Abdelsamed H, Peters J, Byrne GI. Genetic variation in Chlamydia trachomatis and their hosts: impact on disease severity and tissue tropism. Future Microbiol 2013; 8:1129–1146 [View Article] [PubMed]
    [Google Scholar]
  9. Hadfield J, Bénard A, Domman D, Thomson N. The hidden genomics of Chlamydia trachomatis. Curr Top Microbiol Immunol 2018; 412:107–131 [View Article] [PubMed]
    [Google Scholar]
  10. Joseph SJ, Didelot X, Gandhi K, Dean D, Read TD. Interplay of recombination and selection in the genomes of Chlamydia trachomatis. Biol Direct 2011; 6:28 [View Article] [PubMed]
    [Google Scholar]
  11. Joseph SJ, Didelot X, Rothschild J, de Vries HJC, Morré SA et al. Population genomics of Chlamydia trachomatis: insights on drift, selection, recombination, and population structure. Mol Biol Evol 2012; 29:3933–3946 [View Article] [PubMed]
    [Google Scholar]
  12. Somboonna N, Mead S, Liu J, Dean D. Discovering and differentiating new and emerging clonal populations of Chlamydia trachomatis with a novel shotgun cell culture harvest assay. Emerg Infect Dis 2008; 14:445–453 [View Article] [PubMed]
    [Google Scholar]
  13. Last AR, Pickering H, Roberts CH, Coll F, Phelan J et al. Population-based analysis of ocular Chlamydia trachomatis in trachoma-endemic West African communities identifies genomic markers of disease severity. Genome Med 2018; 10:15 [View Article] [PubMed]
    [Google Scholar]
  14. Carlson JH, Hughes S, Hogan D, Cieplak G, Sturdevant DE et al. Polymorphisms in the Chlamydia trachomatis cytotoxin locus associated with ocular and genital isolates. Infect Immun 2004; 72:7063–7072 [View Article] [PubMed]
    [Google Scholar]
  15. Alkhidir AAI, Holland MJ, Elhag WI, Williams CA, Breuer J et al. Whole-genome sequencing of ocular Chlamydia trachomatis isolates from Gadarif State, Sudan. Parasit Vectors 2019; 12:518 [View Article] [PubMed]
    [Google Scholar]
  16. Eder T, Kobus S, Stallmann S, Stepanow S, Köhrer K et al. Genome sequencing of Chlamydia trachomatis serovars E and F reveals substantial genetic variation. Pathog Dis 2017; 75:ftx120 [View Article] [PubMed]
    [Google Scholar]
  17. Beder T, Saluz HP. Virulence-related comparative transcriptomics of infectious and non-infectious chlamydial particles. BMC Genomics 2018; 19:575 [View Article] [PubMed]
    [Google Scholar]
  18. Hadfield J, Harris SR, Seth-Smith HMB, Parmar S, Andersson P et al. Comprehensive global genome dynamics of Chlamydia trachomatis show ancient diversification followed by contemporary mixing and recent lineage expansion. Genome Res 2017; 27:1220–1229 [View Article] [PubMed]
    [Google Scholar]
  19. Somboonna N, Ziklo N, Ferrin TE, Hyuk Suh J, Dean D. Clinical persistence of Chlamydia trachomatis sexually transmitted strains involves novel mutations in the functional αββα tetramer of the tryptophan synthase operon. mBio 2019; 10:e01464-19 [View Article] [PubMed]
    [Google Scholar]
  20. Elwell C, Mirrashidi K, Engel J. Chlamydia cell biology and pathogenesis. Nat Rev Microbiol 2016; 14:385–400 [View Article] [PubMed]
    [Google Scholar]
  21. Nunes A, Gomes JP. Evolution, phylogeny, and molecular epidemiology of Chlamydia. Infect Genet Evol 2014; 23:49–64 [View Article] [PubMed]
    [Google Scholar]
  22. Almeida F, Borges V, Ferreira R, Borrego MJ, Gomes JP et al. Polymorphisms in inc proteins and differential expression of inc genes among Chlamydia trachomatis strains correlate with invasiveness and tropism of lymphogranuloma venereum isolates. J Bacteriol 2012; 194:6574–6585 [View Article] [PubMed]
    [Google Scholar]
  23. West ES, Munoz B, Mkocha H, Holland MJ, Aguirre A et al. Mass treatment and the effect on the load of Chlamydia trachomatis infection in a trachoma-hyperendemic community. Invest Ophthalmol Vis Sci 2005; 46:83–87 [View Article] [PubMed]
    [Google Scholar]
  24. Last A, Burr S, Alexander N, Harding-Esch E, Roberts CH et al. Spatial clustering of high load ocular Chlamydia trachomatis infection in trachoma: a cross-sectional population-based study. Pathog Dis 2017; 75:ftx050 [View Article] [PubMed]
    [Google Scholar]
  25. Solomon AW, Holland MJ, Alexander NDE, Massae PA, Aguirre A et al. Mass treatment with single-dose azithromycin for trachoma. N Engl J Med 2004; 351:1962–1971 [View Article] [PubMed]
    [Google Scholar]
  26. Nash SD, Chernet A, Moncada J, Stewart AEP, Astale T et al. Ocular Chlamydia trachomatis infection and infectious load among pre-school aged children within trachoma hyperendemic districts receiving the SAFE strategy, Amhara region, Ethiopia. PLoS Negl Trop Dis 2020; 14:e0008226 [View Article] [PubMed]
    [Google Scholar]
  27. Harding-Esch EM, Holland MJ, Schémann J-F, Sillah A, Sarr B et al. Impact of a single round of mass drug administration with azithromycin on active trachoma and ocular Chlamydia trachomatis prevalence and circulating strains in The Gambia and Senegal. Parasit Vectors 2019; 12:497 [View Article] [PubMed]
    [Google Scholar]
  28. Ghasemian E, Inic-Kanada A, Collingro A, Mejdoubi L, Alchalabi H et al. Comparison of genovars and Chlamydia trachomatis infection loads in ocular samples from children in two distinct cohorts in Sudan and Morocco. PLoS Negl Trop Dis 2021; 15:e0009655 [View Article] [PubMed]
    [Google Scholar]
  29. Barton A, Rosenkrands I, Pickering H, Faal N, Harte A et al. A systems serology approach to the investigation of infection-induced antibody responses and protection in trachoma. Front Immunol 2023; 14:1178741 [View Article] [PubMed]
    [Google Scholar]
  30. Lutter EI, Bonner C, Holland MJ, Suchland RJ, Stamm WE et al. Phylogenetic analysis of Chlamydia trachomatis tarp and correlation with clinical phenotype. Infect Immun 2010; 78:3678–3688 [View Article] [PubMed]
    [Google Scholar]
  31. Barton A, Faal N, Ramadhani A, Derrick T, Mafuru E et al. Longitudinal changes in tear cytokines and antimicrobial proteins in trachomatous disease. PLoS Negl Trop Dis 2023; 17:e0011689 [View Article] [PubMed]
    [Google Scholar]
  32. Faal N, Bailey RL, Jeffries D, Joof H, Sarr I et al. Conjunctival FOXP3 expression in trachoma: do regulatory T cells have a role in human ocular Chlamydia trachomatis infection?. PLoS Med 2006; 3:e266 [View Article] [PubMed]
    [Google Scholar]
  33. Holland MJ, Faal N, Sarr I, Joof H, Laye M et al. The frequency of Chlamydia trachomatis major outer membrane protein-specific CD8+ T lymphocytes in active trachoma is associated with current ocular infection. Infect Immun 2006; 74:1565–1572 [View Article] [PubMed]
    [Google Scholar]
  34. Thylefors B, Dawson CR, Jones BR, West SK, Taylor HR. A simple system for the assessment of trachoma and its complications. Bull World Health Organ 1987; 65:477–483 [PubMed]
    [Google Scholar]
  35. Solomon AW, Holland MJ, Burton MJ, West SK, Alexander NDE et al. Strategies for control of trachoma: observational study with quantitative PCR. Lancet 2003; 362:198–204 [View Article] [PubMed]
    [Google Scholar]
  36. Burton MJ, Holland MJ, Jeffries D, Mabey DCW, Bailey RL. Conjunctival chlamydial 16S ribosomal RNA expression in trachoma: is chlamydial metabolic activity required for disease to develop?. Clin Infect Dis 2006; 42:463–470 [View Article] [PubMed]
    [Google Scholar]
  37. Roberts CH, Last A, Molina-Gonzalez S, Cassama E, Butcher R et al. Development and evaluation of a next-generation digital PCR diagnostic assay for ocular Chlamydia trachomatis infections. J Clin Microbiol 2013; 51:2195–2203 [View Article] [PubMed]
    [Google Scholar]
  38. Last AR, Roberts C h, Cassama E, Nabicassa M, Molina-Gonzalez S et al. Plasmid copy number and disease severity in naturally occurring ocular Chlamydia trachomatis infection. J Clin Microbiol 2014; 52:324–327 [View Article] [PubMed]
    [Google Scholar]
  39. Bushnell B, Rood J, Singer E. BBMerge - accurate paired shotgun read merging via overlap. PLoS ONE 2017; 12:e0185056 [View Article] [PubMed]
    [Google Scholar]
  40. Andrews S. FastQC: A Quality Control Tool for High Throughput Sequence Data; 2010 http://www.Bioinformatics.Babraham.Ac.Uk/Projects/Fastqc
  41. Zerbino DR, Birney E. Velvet: algorithms for de novo short read assembly using de Bruijn graphs. Genome Res 2008; 18:821–829 [View Article] [PubMed]
    [Google Scholar]
  42. Li H. New strategies to improve minimap2 alignment accuracy. Bioinformatics 2021; 37:4572–4574 [View Article] [PubMed]
    [Google Scholar]
  43. Li H. Minimap2: pairwise alignment for nucleotide sequences. Bioinformatics 2018; 34:3094–3100 [View Article] [PubMed]
    [Google Scholar]
  44. Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nat Methods 2012; 9:357–359 [View Article] [PubMed]
    [Google Scholar]
  45. Seemann T. ABRicate: Mass Screening of Contigs for Antiobiotic Resistance Genes; 2016 https://Github.Com/Tseemann/Abricate
  46. 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]
  47. Bharat A, Petkau A, Avery BP, Chen JC, Folster JP et al. Correlation between phenotypic and in silico detection of antimicrobial resistance in Salmonella enterica in Canada using Staramr. Microorganisms 2022; 10:292 [View Article] [PubMed]
    [Google Scholar]
  48. Shabalina SA, Spiridonov NA, Kashina A. Sounds of silence: synonymous nucleotides as a key to biological regulation and complexity. Nucleic Acids Res 2013; 41:2073–2094 [View Article] [PubMed]
    [Google Scholar]
  49. Kryazhimskiy S, Plotkin JB. The population genetics of DN/DS. PLoS Genet 2008; 4:e1000304 [View Article] [PubMed]
    [Google Scholar]
  50. Jones CA, Hadfield J, Thomson NR, Cleary DW, Marsh P et al. The nature and extent of plasmid variation in Chlamydia trachomatis. Microorganisms 2020; 8:373 [View Article] [PubMed]
    [Google Scholar]
  51. Hurst LD. The Ka/Ks ratio: diagnosing the form of sequence evolution. Trends Genet 2002; 18:486 [View Article] [PubMed]
    [Google Scholar]
  52. Jukes TH. “Evolution of protein molecules.” in mAammalian protein Metabol, 3rd. edn New York, NY: Academic Press; 1969 pp 21–132
    [Google Scholar]
  53. Nei M, Gojobori T. Simple methods for estimating the numbers of synonymous and nonsynonymous nucleotide substitutions. Mol Biol Evol 1986; 3:418–426 [View Article] [PubMed]
    [Google Scholar]
  54. Tamura K, Stecher G, Kumar S. MEGA11: Molecular Evolutionary Genetics Analysis version 11. Mol Biol Evol 2021; 38:3022–3027 [View Article] [PubMed]
    [Google Scholar]
  55. 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]
  56. Stamatakis A. RAxML version 8: a tool for phylogenetic analysis and post-analysis of large phylogenies. Bioinformatics 2014; 30:1312–1313 [View Article] [PubMed]
    [Google Scholar]
  57. Katoh K, Misawa K, Kuma K, Miyata T. MAFFT: a novel method for rapid multiple sequence alignment based on fast fourier transform. Nucleic Acids Res 2002; 30:3059–3066 [View Article] [PubMed]
    [Google Scholar]
  58. Katoh K, Standley DM. MAFFT multiple sequence alignment software version 7: improvements in performance and usability. Mol Biol Evol 2013; 30:772–780 [View Article] [PubMed]
    [Google Scholar]
  59. Guindon S, Dufayard J-F, Lefort V, Anisimova M, Hordijk W et al. New algorithms and methods to estimate maximum-likelihood phylogenies: assessing the performance of PhyML 3.0. Syst Biol 2010; 59:307–321 [View Article] [PubMed]
    [Google Scholar]
  60. Carlson JH, Porcella SF, McClarty G, Caldwell HD. Comparative genomic analysis of Chlamydia trachomatis oculotropic and genitotropic strains. Infect Immun 2005; 73:6407–6418 [View Article] [PubMed]
    [Google Scholar]
  61. Andersson P, Harris SR, Smith HMBS, Hadfield J, O’Neill C et al. Chlamydia trachomatis from Australian aboriginal people with trachoma are polyphyletic composed of multiple distinctive lineages. Nat Commun 2016; 7:10688 [View Article] [PubMed]
    [Google Scholar]
  62. Seth-Smith HMB, Harris SR, Persson K, Marsh P, Barron A et al. Co-evolution of genomes and plasmids within Chlamydia trachomatis and the emergence in Sweden of a new variant strain. BMC Genomics 2009; 10:239 [View Article] [PubMed]
    [Google Scholar]
  63. Fisher DJ, Fernández RE, Maurelli AT. Chlamydia trachomatis transports NAD via the Npt1 ATP/ADP translocase. J Bacteriol 2013; 195:3381–3386 [View Article] [PubMed]
    [Google Scholar]
  64. Rucks EA. Type III secretion in chlamydia. Microbiol Mol Biol Rev 2023; 87:e0003423 [View Article]
    [Google Scholar]
  65. Tanaka KJ, Song S, Mason K, Pinkett HW. Selective substrate uptake: the role of ATP-binding cassette (ABC) importers in pathogenesis. Biochim Biophys Acta Biomembr 2018; 1860:868–877 [View Article] [PubMed]
    [Google Scholar]
  66. Kari L, Whitmire WM, Carlson JH, Crane DD, Reveneau N et al. Pathogenic diversity among Chlamydia trachomatis ocular strains in nonhuman primates is affected by subtle genomic variations. J Infect Dis 2008; 197:449–456 [View Article] [PubMed]
    [Google Scholar]
  67. Braun C, Hegemann JH, Mölleken K. Insights into a Chlamydia pneumoniae-specific gene cluster of membrane binding proteins. Front Cell Infect Microbiol 2020; 10:565808 [View Article] [PubMed]
    [Google Scholar]
  68. Weber MM, Bauler LD, Lam J, Hackstadt T. Expression and localization of predicted inclusion membrane proteins in Chlamydia trachomatis. Infect Immun 2015; 83:4710–4718 [View Article] [PubMed]
    [Google Scholar]
  69. Weber MM, Noriea NF, Bauler LD, Lam JL, Sager J et al. A functional core of IncA is required for Chlamydia trachomatis inclusion fusion. J Bacteriol 2016; 198:1347–1355 [View Article] [PubMed]
    [Google Scholar]
  70. Thomson NR, Holden MTG, Carder C, Lennard N, Lockey SJ et al. Chlamydia trachomatis: genome sequence analysis of lymphogranuloma venereum isolates. Genome Res 2008; 18:161–171 [View Article] [PubMed]
    [Google Scholar]
  71. Saini A, Mapolelo DT, Chahal HK, Johnson MK, Outten FW. SufD and SufC ATPase activity are required for iron acquisition during in vivo Fe-S cluster formation on SufB. Biochemistry 2010; 49:9402–9412 [View Article] [PubMed]
    [Google Scholar]
  72. Hamaoui D, Cossé MM, Mohan J, Lystad AH, Wollert T et al. The Chlamydia effector CT622/TaiP targets a nonautophagy related function of ATG16L1. Proc Natl Acad Sci U S A 2020; 117:26784–26794 [View Article] [PubMed]
    [Google Scholar]
  73. Auer D, Hügelschäffer SD, Fischer AB, Rudel T. The chlamydial deubiquitinase Cdu1 supports recruitment of Golgi vesicles to the inclusion. Cell Microbiol 2020; 22:e13136 [View Article] [PubMed]
    [Google Scholar]
  74. Hausman JM, Kenny S, Iyer S, Babar A, Qiu J et al. The two deubiquitinating enzymes from Chlamydia trachomatis have distinct ubiquitin recognition properties. Biochemistry 2020; 59:1604–1617 [View Article] [PubMed]
    [Google Scholar]
  75. Belland RJ, Zhong G, Crane DD, Hogan D, Sturdevant D et al. Genomic transcriptional profiling of the developmental cycle of Chlamydia trachomatis. Proc Natl Acad Sci U S A 2003; 100:8478–8483 [View Article] [PubMed]
    [Google Scholar]
  76. Cortina ME, Bishop RC, DeVasure BA, Coppens I, Derré I. The inclusion membrane protein IncS is critical for initiation of the Chlamydia intracellular developmental cycle. PLoS Pathog 2022; 18:e1010818 [View Article] [PubMed]
    [Google Scholar]
  77. Cossé MM, Barta ML, Fisher DJ, Oesterlin LK, Niragire B et al. The loss of expression of a single type 3 effector (CT622) strongly reduces Chlamydia trachomatis infectivity and growth. Front Cell Infect Microbiol 2018; 8:145 [View Article] [PubMed]
    [Google Scholar]
  78. Huang Y, Wurihan W, Lu B, Zou Y, Wang Y et al. Robust Heat Shock Response in Chlamydia Lacking a Typical Heat Shock Sigma Factor. Front Microbiol 2021; 12:812448 [View Article]
    [Google Scholar]
  79. Benamri I, Azzouzi M, Moussa A, Radouani F. An in silico analysis of rpoB mutations to affect Chlamydia trachomatis sensitivity to rifamycin. J Genet Eng Biotechnol 2022; 20:146 [View Article] [PubMed]
    [Google Scholar]
  80. Gorbalenya AE, Koonin EV, Donchenko AP, Blinov VM. Two related superfamilies of putative helicases involved in replication, recombination, repair and expression of DNA and RNA genomes. Nucleic Acids Res 1989; 17:4713–4730 [View Article] [PubMed]
    [Google Scholar]
  81. Soules KR, LaBrie SD, May BH, Hefty PS. Sigma 54-regulated transcription is associated with membrane reorganization and type III secretion effectors during conversion to infectious forms of Chlamydia trachomatis. mBio 2020; 11:e01725-20 [View Article] [PubMed]
    [Google Scholar]
  82. Ilic S, Cohen S, Singh M, Tam B, Dayan A et al. DnaG Primase-a target for the development of novel antibacterial agents. Antibiotics 2018; 7:72 [View Article] [PubMed]
    [Google Scholar]
  83. Wylie JL, Berry JD, McClarty G. Chlamydia trachomatis CTP synthetase: molecular characterization and developmental regulation of expression. Mol Microbiol 1996; 22:631–642 [View Article] [PubMed]
    [Google Scholar]
  84. Lorca GL, Barabote RD, Zlotopolski V, Tran C, Winnen B et al. Transport capabilities of eleven gram-positive bacteria: comparative genomic analyses. Biochim Biophys Acta 2007; 1768:1342–1366 [View Article] [PubMed]
    [Google Scholar]
  85. Griffin J, Roshick C, Iliffe-Lee E, McClarty G. Catalytic mechanism of Chlamydia trachomatis flavin-dependent thymidylate synthase. J Biol Chem 2005; 280:5456–5467 [View Article] [PubMed]
    [Google Scholar]
  86. Kari L, Southern TR, Downey CJ, Watkins HS, Randall LB et al. Chlamydia trachomatis polymorphic membrane protein D is a virulence factor involved in early host-cell interactions. Infect Immun 2014; 82:2756–2762 [View Article] [PubMed]
    [Google Scholar]
  87. Yang M, Rajeeve K, Rudel T, Dandekar T. Comprehensive flux modeling of Chlamydia trachomatis proteome and QRT-PCR data indicate biphasic metabolic differences between elementary bodies and reticulate bodies during infection. Front Microbiol 2019; 10:2350 [View Article] [PubMed]
    [Google Scholar]
  88. Wang L, Hou Y, Yuan H, Chen H. The role of tryptophan in Chlamydia trachomatis persistence. Front Cell Infect Microbiol 2022; 12:931653 [View Article] [PubMed]
    [Google Scholar]
  89. Binet R, Fernandez RE, Fisher DJ, Maurelli AT. Identification and characterization of the Chlamydia trachomatis L2 S-adenosylmethionine transporter. mBio 2011; 2:e00051-11 [View Article] [PubMed]
    [Google Scholar]
  90. Stephens RS, Kalman S, Lammel C, Fan J, Marathe R et al. Genome sequence of an obligate intracellular pathogen of humans: Chlamydia trachomatis. Science 1998; 282:754–759 [View Article] [PubMed]
    [Google Scholar]
  91. Taylor LD, Nelson DE, Dorward DW, Whitmire WM, Caldwell HD. Biological characterization of Chlamydia trachomatis plasticity zone MACPF domain family protein CT153. Infect Immun 2010; 78:2691–2699 [View Article] [PubMed]
    [Google Scholar]
  92. Macleod CK, Butcher R, Mudaliar U, Natutusau K, Pavluck AL et al. Low prevalence of ocular Chlamydia trachomatis infection and active trachoma in the western division of fiji. PLoS Negl Trop Dis 2016; 10:e0004798 [View Article] [PubMed]
    [Google Scholar]
  93. Harris SR, Clarke IN, Seth-Smith HMB, Solomon AW, Cutcliffe LT et al. Whole-genome analysis of diverse Chlamydia trachomatis strains identifies phylogenetic relationships masked by current clinical typing. Nat Genet 2012; 44:413–419 [View Article] [PubMed]
    [Google Scholar]
  94. Jeffrey BM, Suchland RJ, Quinn KL, Davidson JR, Stamm WE et al. Genome sequencing of recent clinical Chlamydia trachomatis strains identifies loci associated with tissue tropism and regions of apparent recombination. Infect Immun 2010; 78:2544–2553 [View Article] [PubMed]
    [Google Scholar]
  95. Versteeg B, Bruisten SM, Pannekoek Y, Jolley KA, Maiden MCJ et al. Genomic analyses of the Chlamydia trachomatis core genome show an association between chromosomal genome, plasmid type and disease. BMC Genomics 2018; 19:130 [View Article] [PubMed]
    [Google Scholar]
  96. Seth-Smith HMB, Bénard A, Bruisten SM, Versteeg B, Herrmann B et al. Ongoing evolution of Chlamydia trachomatis lymphogranuloma venereum: exploring the genomic diversity of circulating strains. Microb Genom 2021; 7:000599 [View Article] [PubMed]
    [Google Scholar]
  97. Read TD, Joseph SJ, Didelot X, Liang B, Patel L et al. Comparative analysis of Chlamydia psittaci genomes reveals the recent emergence of a pathogenic lineage with a broad host range. mBio 2013; 4:e00604-12 [View Article] [PubMed]
    [Google Scholar]
  98. Williams MJ, Zapata L, Werner B, Barnes CP, Sottoriva A et al. Measuring the distribution of fitness effects in somatic evolution by combining clonal dynamics with DN/DS ratios. Elife 2020; 9:e48714 [View Article] [PubMed]
    [Google Scholar]
  99. Wolf JBW, Künstner A, Nam K, Jakobsson M, Ellegren H. Nonlinear dynamics of nonsynonymous (DN) and synonymous (DS) substitution rates affects inference of selection. Genome Biol Evol 2009; 1:308–319 [View Article]
    [Google Scholar]
  100. Kari L, Whitmire WM, Carlson JH, Crane DD, Reveneau N et al. Pathogenic diversity among Chlamydia trachomatis ocular strains in nonhuman primates is affected by subtle genomic variations. J Infect Dis 2008; 197:449–456 [View Article] [PubMed]
    [Google Scholar]
  101. Sigalova OM, Chaplin AV, Bochkareva OO, Shelyakin PV, Filaretov VA et al. Chlamydia pan-genomic analysis reveals balance between host adaptation and selective pressure to genome reduction. BMC Genomics 2019; 20:710 [View Article] [PubMed]
    [Google Scholar]
  102. Merkl R. Modelling the evolution of the archeal tryptophan synthase. BMC Evol Biol 2007; 7:59 [View Article] [PubMed]
    [Google Scholar]
  103. Busch F, Rajendran C, Heyn K, Schlee S, Merkl R et al. Ancestral tryptophan synthase reveals functional sophistication of primordial enzyme complexes. Cell Chem Biol 2016; 23:709–715 [View Article] [PubMed]
    [Google Scholar]
  104. Bastidas RJ, Elwell CA, Engel JN, Valdivia RH. Chlamydial intracellular survival strategies. Cold Spring Harb Perspect Med 2013; 3:a010256 [View Article] [PubMed]
    [Google Scholar]
  105. Ziklo N, Huston WM, Hocking JS, Timms P. Chlamydia trachomatis genital tract infections: when host immune response and the microbiome collide. Trends Microbiol 2016; 24:750–765 [View Article] [PubMed]
    [Google Scholar]
  106. Caldwell HD, Wood H, Crane D, Bailey R, Jones RB et al. Polymorphisms in Chlamydia trachomatis tryptophan synthase genes differentiate between genital and ocular isolates. J Clin Invest 2003; 111:1757–1769 [View Article] [PubMed]
    [Google Scholar]
  107. Yuan Y, Zhang YX, Watkins NG, Caldwell HD. Nucleotide and deduced amino acid sequences for the four variable domains of the major outer membrane proteins of the 15 Chlamydia trachomatis serovars. Infect Immun 1989; 57:1040–1049 [View Article] [PubMed]
    [Google Scholar]
  108. Lerat E, Ochman H. Recognizing the pseudogenes in bacterial genomes. Nucleic Acids Res 2005; 33:3125–3132 [View Article] [PubMed]
    [Google Scholar]
  109. Moran NA. Microbial minimalism. Cell 2002; 108:583–586 [View Article]
    [Google Scholar]
  110. Robinson SM, Rajachandran V, Majumdar S, Saha S, Das S et al. Distinct potentially adaptive accumulation of truncation mutations in Salmonella enterica serovar typhi and Salmonella enterica serovar paratyphi A. Microbiol Spectr 2022; 10:e0196921 [View Article] [PubMed]
    [Google Scholar]
  111. Smelov V, Vrbanac A, van Ess EF, Noz MP, Wan R et al. Chlamydia trachomatis strain types have diversified regionally and globally with evidence for recombination across geographic divides. Front Microbiol 2017; 8:2195 [View Article] [PubMed]
    [Google Scholar]
  112. Joseph SJ, Bommana S, Ziklo N, Kama M, Dean D et al. Patterns of within-host spread of Chlamydia trachomatis between vagina, endocervix and rectum revealed by comparative genomic analysis. Front Microbiol 2023; 14:1154664 [View Article] [PubMed]
    [Google Scholar]
  113. Bom RJM, Christerson L, Schim van der Loeff MF, Coutinho RA, Herrmann B et al. Evaluation of high-resolution typing methods for Chlamydia trachomatis in samples from heterosexual couples. J Clin Microbiol 2011; 49:2844–2853 [View Article] [PubMed]
    [Google Scholar]
  114. Chidambaram JD, Lee DC, Porco TC, Lietman TM. Mass antibiotics for trachoma and the allee effect. Lancet Infect Dis 2005; 5:194–196 [View Article] [PubMed]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/mgen/10.1099/mgen.0.001210
Loading
/content/journal/mgen/10.1099/mgen.0.001210
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