#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Genome Analysis of a Transmissible Lineage of Reveals Pathoadaptive Mutations and Distinct Evolutionary Paths of Hypermutators


Genome sequencing of bacterial pathogens has advanced our understanding of their evolution, epidemiology, and response to antibiotic therapy. However, we still have only a limited knowledge of the molecular changes in in vivo evolving bacterial populations in relation to long-term, chronic infections. For example, it remains unclear what genes are mutated to facilitate the establishment of long-term existence in the human host environment, and in which way acquisition of a hypermutator phenotype with enhanced rates of spontaneous mutations influences the evolutionary trajectory of the pathogen. Here we perform a retrospective study of the DK2 clone type of P. aeruginosa isolated from Danish patients suffering from cystic fibrosis (CF), and analyze the genomes of 55 bacterial isolates collected from 21 infected individuals over 38 years. Our phylogenetic analysis of 8,530 mutations in the DK2 genomes shows that the ancestral DK2 clone type spread among CF patients through several independent transmission events. Subsequent to transmission, sub-lineages evolved independently for years in separate hosts, creating a unique possibility to study parallel evolution and identification of genes targeted by mutations to optimize pathogen fitness (pathoadaptive mutations). These genes were related to antibiotic resistance, the cell envelope, or regulatory functions, and we find that the prevalence of pathoadaptive mutations correlates with evolutionary success of co-evolving sub-lineages.

The long-term co-existence of both normal and hypermutator populations enabled comparative investigations of the mutation dynamics in homopolymeric sequences in which hypermutators are particularly prone to mutations. We find a positive exponential correlation between the length of the homopolymer and its likelihood to acquire mutations and identify two homopolymer-containing genes preferentially mutated in hypermutators. This homopolymer facilitated differential mutagenesis provides a novel genome-wide perspective on the different evolutionary trajectories of hypermutators, which may help explain their emergence in CF infections.


Vyšlo v časopise: Genome Analysis of a Transmissible Lineage of Reveals Pathoadaptive Mutations and Distinct Evolutionary Paths of Hypermutators. PLoS Genet 9(9): e32767. doi:10.1371/journal.pgen.1003741
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003741

Souhrn

Genome sequencing of bacterial pathogens has advanced our understanding of their evolution, epidemiology, and response to antibiotic therapy. However, we still have only a limited knowledge of the molecular changes in in vivo evolving bacterial populations in relation to long-term, chronic infections. For example, it remains unclear what genes are mutated to facilitate the establishment of long-term existence in the human host environment, and in which way acquisition of a hypermutator phenotype with enhanced rates of spontaneous mutations influences the evolutionary trajectory of the pathogen. Here we perform a retrospective study of the DK2 clone type of P. aeruginosa isolated from Danish patients suffering from cystic fibrosis (CF), and analyze the genomes of 55 bacterial isolates collected from 21 infected individuals over 38 years. Our phylogenetic analysis of 8,530 mutations in the DK2 genomes shows that the ancestral DK2 clone type spread among CF patients through several independent transmission events. Subsequent to transmission, sub-lineages evolved independently for years in separate hosts, creating a unique possibility to study parallel evolution and identification of genes targeted by mutations to optimize pathogen fitness (pathoadaptive mutations). These genes were related to antibiotic resistance, the cell envelope, or regulatory functions, and we find that the prevalence of pathoadaptive mutations correlates with evolutionary success of co-evolving sub-lineages.

The long-term co-existence of both normal and hypermutator populations enabled comparative investigations of the mutation dynamics in homopolymeric sequences in which hypermutators are particularly prone to mutations. We find a positive exponential correlation between the length of the homopolymer and its likelihood to acquire mutations and identify two homopolymer-containing genes preferentially mutated in hypermutators. This homopolymer facilitated differential mutagenesis provides a novel genome-wide perspective on the different evolutionary trajectories of hypermutators, which may help explain their emergence in CF infections.


Zdroje

1. LiebermanTD, MichelJB, AingaranM, Potter-BynoeG, RouxD, et al. (2011) Parallel bacterial evolution within multiple patients identifies candidate pathogenicity genes. Nat Genet 43: 1275–1280.

2. MutrejaA, KimDW, ThomsonNR, ConnorTR, LeeJH, et al. (2011) Evidence for several waves of global transmission in the seventh cholera pandemic. Nature 477: 462–465.

3. SmithEE, BuckleyDG, WuZ, SaenphimmachakC, HoffmanLR, et al. (2006) Genetic adaptation by Pseudomonas aeruginosa to the airways of cystic fibrosis patients. Proc Natl Acad Sci U S A 103: 8487–8492.

4. HarrisSR, ClarkeIN, Seth-SmithHM, SolomonAW, CutcliffeLT, et al. (2012) Whole-genome analysis of diverse Chlamydia trachomatis strains identifies phylogenetic relationships masked by current clinical typing. Nat Genet 44: 413–419, S411.

5. CroucherNJ, HarrisSR, FraserC, QuailMA, BurtonJ, et al. (2011) Rapid pneumococcal evolution in response to clinical interventions. Science 331: 430–434.

6. MorelliG, SongY, MazzoniCJ, EppingerM, RoumagnacP, et al. (2010) Yersinia pestis genome sequencing identifies patterns of global phylogenetic diversity. Nat Genet 42: 1140–1143.

7. HoltKE, BakerS, WeillFX, HolmesEC, KitchenA, et al. (2012) Shigella sonnei genome sequencing and phylogenetic analysis indicate recent global dissemination from Europe. Nat Genet 44: 1056–1059.

8. SokurenkoEV, HastyDL, DykhuizenDE (1999) Pathoadaptive mutations: gene loss and variation in bacterial pathogens. Trends Microbiol 7: 191–195.

9. RauMH, MarvigRL, EhrlichGD, MolinS, JelsbakL (2012) Deletion and acquisition of genomic content during early stage adaptation of Pseudomonas aeruginosa to a human host environment. Environ Microbiol 14: 2200–2211.

10. YangL, JelsbakL, MarvigRL, DamkiaerS, WorkmanCT, et al. (2011) Evolutionary dynamics of bacteria in a human host environment. Proc Natl Acad Sci U S A 108: 7481–7486.

11. OliverA, CantonR, CampoP, BaqueroF, BlazquezJ (2000) High frequency of hypermutable Pseudomonas aeruginosa in cystic fibrosis lung infection. Science 288: 1251–1254.

12. MaticI, RadmanM, TaddeiF, PicardB, DoitC, et al. (1997) Highly variable mutation rates in commensal and pathogenic Escherichia coli. Science 277: 1833–1834.

13. MaoEF, LaneL, LeeJ, MillerJH (1997) Proliferation of mutators in A cell population. J Bacteriol 179: 417–422.

14. LeClercJE, LiB, PayneWL, CebulaTA (1996) High mutation frequencies among Escherichia coli and Salmonella pathogens. Science 274: 1208–1211.

15. RichardsonAR, YuZ, PopovicT, StojiljkovicI (2002) Mutator clones of Neisseria meningitidis in epidemic serogroup A disease. Proc Natl Acad Sci U S A 99: 6103–6107.

16. SniegowskiPD, GerrishPJ, LenskiRE (1997) Evolution of high mutation rates in experimental populations of E. coli. Nature 387: 703–705.

17. ChungJC, BecqJ, FraserL, Schulz-TrieglaffO, BondNJ, et al. (2012) Genomic Variation among Contemporary Pseudomonas aeruginosa Isolates from Chronically Infected Cystic Fibrosis Patients. J Bacteriol 194: 4857–4866.

18. OliverA, BaqueroF, BlazquezJ (2002) The mismatch repair system (mutS, mutL and uvrD genes) in Pseudomonas aeruginosa: molecular characterization of naturally occurring mutants. Mol Microbiol 43: 1641–1650.

19. SchaaperRM, DunnRL (1987) Spectra of spontaneous mutations in Escherichia coli strains defective in mismatch correction: the nature of in vivo DNA replication errors. Proc Natl Acad Sci U S A 84: 6220–6224.

20. BicharaM, WagnerJ, LambertIB (2006) Mechanisms of tandem repeat instability in bacteria. Mutat Res 598: 144–163.

21. StrandM, ProllaTA, LiskayRM, PetesTD (1993) Destabilization of tracts of simple repetitive DNA in yeast by mutations affecting DNA mismatch repair. Nature 365: 274–276.

22. LevinsonG, GutmanGA (1987) High frequencies of short frameshifts in poly-CA/TG tandem repeats borne by bacteriophage M13 in Escherichia coli K-12. Nucleic Acids Res 15: 5323–5338.

23. MoxonER, RaineyPB, NowakMA, LenskiRE (1994) Adaptive evolution of highly mutable loci in pathogenic bacteria. Curr Biol 4: 24–33.

24. MoyanoAJ, SmaniaAM (2009) Simple sequence repeats and mucoid conversion: biased mucA mutagenesis in mismatch repair-deficient Pseudomonas aeruginosa. PLoS One 4: e8203.

25. LiberatiNT, UrbachJM, MiyataS, LeeDG, DrenkardE, et al. (2006) An ordered, nonredundant library of Pseudomonas aeruginosa strain PA14 transposon insertion mutants. Proc Natl Acad Sci U S A 103: 2833–2838.

26. YangL, HaagensenJA, JelsbakL, JohansenHK, SternbergC, et al. (2008) In situ growth rates and biofilm development of Pseudomonas aeruginosa populations in chronic lung infections. J Bacteriol 190: 2767–2776.

27. HarrisSR, FeilEJ, HoldenMT, QuailMA, NickersonEK, et al. (2010) Evolution of MRSA during hospital transmission and intercontinental spread. Science 327: 469–474.

28. MarvigRL, SondergaardMS, DamkiaerS, HoibyN, JohansenHK, et al. (2012) Mutations in 23S rRNA confer resistance against azithromycin in Pseudomonas aeruginosa. Antimicrob Agents Chemother 56: 4519.

29. SeverinovK, MarkovD, SeverinovaE, NikiforovV, LandickR, et al. (1995) Streptolydigin-resistant mutants in an evolutionarily conserved region of the beta' subunit of Escherichia coli RNA polymerase. J Biol Chem 270: 23926–23929.

30. YeeYC, KisslingerB, YuVL, JinDJ (1996) A mechanism of rifamycin inhibition and resistance in Pseudomonas aeruginosa. J Antimicrob Chemother 38: 133–137.

31. LiaoX, HancockRE (1995) Cloning and characterization of the Pseudomonas aeruginosa pbpB gene encoding penicillin-binding protein 3. Antimicrob Agents Chemother 39: 1871–1874.

32. LomovskayaO, LewisK (1992) Emr, an Escherichia coli locus for multidrug resistance. Proc Natl Acad Sci U S A 89: 8938–8942.

33. BieleckiP, LukatP, HuseckenK, DotschA, SteinmetzH, et al. (2012) Mutation in elongation factor G confers resistance to the antibiotic argyrin in the opportunistic pathogen Pseudomonas aeruginosa. Chembiochem 13: 2339–2345.

34. HancockRE (1998) Resistance mechanisms in Pseudomonas aeruginosa and other nonfermentative gram-negative bacteria. Clin Infect Dis 27 Suppl 1: S93–99.

35. StratevaT, YordanovD (2009) Pseudomonas aeruginosa - a phenomenon of bacterial resistance. J Med Microbiol 58: 1133–1148.

36. MoskowitzSM, ErnstRK, MillerSI (2004) PmrAB, a two-component regulatory system of Pseudomonas aeruginosa that modulates resistance to cationic antimicrobial peptides and addition of aminoarabinose to lipid A. J Bacteriol 186: 575–579.

37. WangY, HaU, ZengL, JinS (2003) Regulation of membrane permeability by a two-component regulatory system in Pseudomonas aeruginosa. Antimicrob Agents Chemother 47: 95–101.

38. NguyenD, SinghPK (2006) Evolving stealth: genetic adaptation of Pseudomonas aeruginosa during cystic fibrosis infections. Proc Natl Acad Sci U S A 103: 8305–8306.

39. MoskowitzSM, BrannonMK, DasguptaN, PierM, SgambatiN, et al. (2012) PmrB mutations promote polymyxin resistance of Pseudomonas aeruginosa isolated from colistin-treated cystic fibrosis patients. Antimicrob Agents Chemother 56: 1019–1030.

40. PhilippeN, CrozatE, LenskiRE, SchneiderD (2007) Evolution of global regulatory networks during a long-term experiment with Escherichia coli. Bioessays 29: 846–860.

41. WaineDJ, HoneybourneD, SmithEG, WhitehouseJL, DowsonCG (2008) Association between hypermutator phenotype, clinical variables, mucoid phenotype, and antimicrobial resistance in Pseudomonas aeruginosa. J Clin Microbiol 46: 3491–3493.

42. FerroniA, GuillemotD, MoumileK, BernedeC, Le BourgeoisM, et al. (2009) Effect of mutator P. aeruginosa on antibiotic resistance acquisition and respiratory function in cystic fibrosis. Pediatr Pulmonol 44: 820–825.

43. CiofuO, RiisB, PresslerT, PoulsenHE, HoibyN (2005) Occurrence of hypermutable Pseudomonas aeruginosa in cystic fibrosis patients is associated with the oxidative stress caused by chronic lung inflammation. Antimicrob Agents Chemother 49: 2276–2282.

44. Hoiby N, Frederiksen B (2000) Microbiology of cystic fibrosis. In: Hodson M, Geddes D, editors. Cystic fibrosis. 2nd ed. London, United Kingdom: Arnold. pp. 83–107.

45. KrawitzP, RodelspergerC, JagerM, JostinsL, BauerS, et al. (2010) Microindel detection in short-read sequence data. Bioinformatics 26: 722–729.

46. LiH, HandsakerB, WysokerA, FennellT, RuanJ, et al. (2009) The Sequence Alignment/Map format and SAMtools. Bioinformatics 25: 2078–2079.

47. DrummondAJ, SuchardMA, XieD, RambautA (2012) Bayesian phylogenetics with BEAUti and the BEAST 1.7. Mol Biol Evol 29: 1969–1973.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2013 Číslo 9
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#