1887

Abstract

The increased accessibility of next generation sequencing has allowed enough genomes from a given bacterial species to be sequenced to describe the distribution of genes in the pangenome, without limiting analyses to genes present in reference strains. Although some taxa have thousands of whole genome sequences available on public databases, most genomes were sequenced with short read technology, resulting in incomplete assemblies. Studying pangenomes could lead to important insights into adaptation, pathogenicity, or molecular epidemiology, however given the known information loss inherent in analyzing contig-level assemblies, these inferences may be biased or inaccurate. In this study we describe the pangenome of a clonally evolving pathogen, , and examine the utility of gene content variation in outbreak investigation. We constructed the pangenome using 1463 assembled genomes. We tested the assumption of strict clonal evolution by studying evidence of recombination in core genes and analyzing the distribution of accessory genes among core monophyletic groups. To determine if gene content variation could be utilized in outbreak investigation, we carefully examined accessory genes detected in a well described outbreak in Minnesota. We found significant errors in accessory gene classification. After accounting for these errors, we show that has a much smaller accessory genome than previously described and provide evidence supporting ongoing clonal evolution and a closed pangenome, with little gene content variation generated over outbreaks. We also identified frameshift mutations in multiple genes, including a mutation in , which has recently been associated with antibiotic tolerance in . A pangenomic approach enables a more comprehensive analysis of genome dynamics than is possible with reference-based approaches; however, without critical evaluation of accessory gene content, inferences of transmission patterns employing these loci could be misguided.

  • 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.0.000839
2022-06-28
2024-04-26
Loading full text...

Full text loading...

/deliver/fulltext/mgen/8/6/mgen000839.html?itemId=/content/journal/mgen/10.1099/mgen.0.000839&mimeType=html&fmt=ahah

References

  1. Ceres KM, Stanhope MJ, Gröhn YT. A critical evaluation of Mycobacterium bovis pangenomics, with reference to its utility in outbreak investigation Figshare 2022 [View Article]
    [Google Scholar]
  2. Boritsch EC, Khanna V, Pawlik A, Honoré N, Navas VH et al. Key experimental evidence of chromosomal DNA transfer among selected tuberculosis-causing mycobacteria. Proc Natl Acad Sci U S A 2016; 113:9876–9881 [View Article] [PubMed]
    [Google Scholar]
  3. Chiner-Oms Á, Sánchez-Busó L, Corander J, Gagneux S, Harris SR et al. Genomic determinants of speciation and spread of the Mycobacterium tuberculosis complex. Sci Adv 2019; 5:eaaw3307 [View Article] [PubMed]
    [Google Scholar]
  4. Patané JSL, Martins J, Beatriz Castelão A, Nishibe C, Montera L et al. Patterns and processes of Mycobacterium bovis evolution revealed by phylogenomic analyses. Genome Biol Evol 2017 [View Article] [PubMed]
    [Google Scholar]
  5. Kavvas ES, Catoiu E, Mih N, Yurkovich JT, Seif Y et al. Machine learning and structural analysis of Mycobacterium tuberculosis pan-genome identifies genetic signatures of antibiotic resistance. Nat Commun 2018; 9:4306 [View Article] [PubMed]
    [Google Scholar]
  6. Reis AC, Cunha MV. The open pan-genome architecture and virulence landscape of Mycobacterium bovis. Microb Genom 2021; 7:10 [View Article] [PubMed]
    [Google Scholar]
  7. Reis AC, Cunha MV. Genome-wide estimation of recombination, mutation and positive selection enlightens diversification drivers of Mycobacterium bovis. Sci Rep 2021; 11:18789 [View Article] [PubMed]
    [Google Scholar]
  8. Namouchi A, Didelot X, Schöck U, Gicquel B, Rocha EPC. After the bottleneck: genome-wide diversification of the Mycobacterium tuberculosis complex by mutation, recombination, and natural selection. Genome Res 2012; 22:721–734 [View Article] [PubMed]
    [Google Scholar]
  9. Godfroid M, Dagan T, Kupczok A. Recombination signal in Mycobacterium tuberculosis stems from reference-guided assemblies and alignment artefacts. Genome Biol Evol 2018; 10:1920–1926 [View Article] [PubMed]
    [Google Scholar]
  10. Almaw G, Mekonnen GA, Mihret A, Aseffa A, Taye H et al. Population structure and transmission of Mycobacterium bovis in Ethiopia. Microb Genom 2021; 7: [View Article] [PubMed]
    [Google Scholar]
  11. Loiseau C, Menardo F, Aseffa A, Hailu E, Gumi B et al. An African origin for Mycobacterium bovis. Evol Med Public Health 2020; 2020:49–59 [View Article]
    [Google Scholar]
  12. Orloski K, Robbe-Austerman S, Stuber T, Hench B, Schoenbaum M. Whole genome sequencing of Mycobacterium bovis isolated from livestock in the United States, 1989-2018. Front Vet Sci 2018; 5:253 [View Article] [PubMed]
    [Google Scholar]
  13. Rodrigues R de A, Ribeiro Araújo F, Rivera Dávila AM, Etges RN, Parkhill J et al. Genomic and temporal analyses of Mycobacterium bovis in southern Brazil. Microb Genom 2021; 7: [View Article] [PubMed]
    [Google Scholar]
  14. Zwyer M, Çavusoglu C, Ghielmetti G, Pacciarini ML, Scaltriti E et al. A new nomenclature for the livestock-associated Mycobacterium tuberculosis complex based on phylogenomics. Open Res Europe 2021; 1:100 [View Article]
    [Google Scholar]
  15. Prjibelski A, Antipov D, Meleshko D, Lapidus A, Korobeynikov A. Using SPAdes de novo assembler. Curr Protoc Bioinformatics 2020; 70:e102 [View Article] [PubMed]
    [Google Scholar]
  16. 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]
  17. Gurevich A, Saveliev V, Vyahhi N, Tesler G. QUAST: quality assessment tool for genome assemblies. Bioinformatics 2013; 29:1072–1075 [View Article] [PubMed]
    [Google Scholar]
  18. Parks DH, Imelfort M, Skennerton CT, Hugenholtz P, Tyson GW. CheckM: assessing the quality of microbial genomes recovered from isolates, single cells, and metagenomes. Genome Res 2015; 25:1043–1055 [View Article] [PubMed]
    [Google Scholar]
  19. Seemann T. Prokka: rapid prokaryotic genome annotation. Bioinformatics 2014; 30:2068–2069 [View Article] [PubMed]
    [Google Scholar]
  20. Malone KM, Farrell D, Stuber TP, Schubert OT, Aebersold R et al. Updated reference genome sequence and annotation of. Genome Announc 2017; 5:14 [View Article]
    [Google Scholar]
  21. Garrison E, Kronenberg ZN, Dawson ET, Pedersen BS, Prins P. Vcflib and tools for processing the VCF variant call format. Bioinformatics 2021 [View Article]
    [Google Scholar]
  22. 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]
  23. Mi H, Ebert D, Muruganujan A, Mills C, Albou L-P et al. PANTHER version 16: a revised family classification, tree-based classification tool, enhancer regions and extensive API. Nucleic Acids Res 2021; 49:D394–D403 [View Article] [PubMed]
    [Google Scholar]
  24. DeJesus MA, Gerrick ER, Xu W, Park SW, Long JE et al. Comprehensive essentiality analysis of the Mycobacterium tuberculosis genome via saturating transposon mutagenesis. mBio 2017; 8:e02133-16 [View Article] [PubMed]
    [Google Scholar]
  25. Farrell D, Crispell J, Gordon SV. Updated functional annotation of the Mycobacterium bovis AF2122/97 reference genome. Access Microbiol 2020; 2:acmi000129 [View Article] [PubMed]
    [Google Scholar]
  26. Treangen TJ, Ondov BD, Koren S, Phillippy AM. The Harvest suite for rapid core-genome alignment and visualization of thousands of intraspecific microbial genomes. Genome Biol 2014; 15:11 [View Article] [PubMed]
    [Google Scholar]
  27. Edgar RC. MUSCLE: multiple sequence alignment with high accuracy and high throughput. Nucleic Acids Res 2004; 32:1792–1797 [View Article] [PubMed]
    [Google Scholar]
  28. Price MN, Dehal PS, Arkin AP. FastTree 2--approximately maximum-likelihood trees for large alignments. PLoS One 2010; 5:e9490 [View Article] [PubMed]
    [Google Scholar]
  29. Croucher NJ, Page AJ, Connor TR, Delaney AJ, Keane JA et al. Rapid phylogenetic analysis of large samples of recombinant bacterial whole genome sequences using Gubbins. Nucleic Acids Res 2015; 43:e15 [View Article] [PubMed]
    [Google Scholar]
  30. Darling AE, Mau B, Perna NT. progressiveMauve: multiple genome alignment with gene gain, loss and rearrangement. PLoS One 2010; 5:e11147 [View Article] [PubMed]
    [Google Scholar]
  31. 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]
  32. Mostowy R, Croucher NJ, Andam CP, Corander J, Hanage WP et al. Efficient inference of recent and ancestral recombination within bacterial populations. Mol Biol Evol 2017; 34:1167–1182 [View Article] [PubMed]
    [Google Scholar]
  33. 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]
  34. Robinson JT, Thorvaldsdóttir H, Winckler W, Guttman M, Lander ES et al. Integrative genomics viewer. Nat Biotechnol 2011; 29:24–26 [View Article] [PubMed]
    [Google Scholar]
  35. Cingolani P, Platts A, Wang LL, Coon M, Nguyen T et al. A program for annotating and predicting the effects of single nucleotide polymorphisms, SnpEff: SNPs in the genome of drosophila melanogaster strain w1118; iso-2; iso-3. Fly (Austin) 2012; 6:80–92 [View Article] [PubMed]
    [Google Scholar]
  36. Konishi T, Matsukuma S, Fuji H, Nakamura D, Satou N et al. Principal component analysis applied directly to sequence matrix. Sci Rep 2019; 9:19297 [View Article] [PubMed]
    [Google Scholar]
  37. Bittinger K. abdiv: alpha and beta diversity measures. R package version 0.2.0 2020 https://CRAN.R-project.org/package=abdiv
    [Google Scholar]
  38. Team R. RStudio: Integrated Development for R. Boston, MA: 2020 http://www.rstudio.com
  39. Team RC. R: A Language and Environment for Statistical Computing Vienna, Austria: R Foundation for Statistical Computing; 2021
    [Google Scholar]
  40. Glaser L, Carstensen M, Shaw S, Robbe-Austerman S, Wunschmann A et al. Descriptive epidemiology and whole genome sequencing analysis for an outbreak of bovine tuberculosis in beef cattle and white-tailed deer in Northwestern Minnesota. PLoS One 2016; 11:e0145735 [View Article] [PubMed]
    [Google Scholar]
  41. Minh BQ, Schmidt HA, Chernomor O, Schrempf D, Woodhams MD et al. IQ-TREE 2: new models and efficient methods for phylogenetic inference in the genomic era. Mol Biol Evol 2020; 37:1530–1534 [View Article] [PubMed]
    [Google Scholar]
  42. Gupta A, Alland D. Reversible gene silencing through frameshift indels and frameshift scars provide adaptive plasticity for Mycobacterium tuberculosis. Nat Commun 2021; 12:4702 [View Article] [PubMed]
    [Google Scholar]
  43. 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]
  44. Zimpel CK, Brandão PE, de Souza Filho AF, de Souza RF, Ikuta CY et al. Complete genome sequencing of Mycobacterium bovis SP38 and comparative genomics of Mycobacterium bovis and M. tuberculosis strains. Front Microbiol 2017; 8:2389 [View Article]
    [Google Scholar]
  45. Zakham F, Sironen T, Vapalahti O, Kant R. Pan and core genome analysis of 183 Mycobacterium tuberculosis strains revealed a high inter-species diversity among the human adapted strains. Antibiotics (Basel) 2021; 10:500 [View Article] [PubMed]
    [Google Scholar]
  46. Dar HA, Zaheer T, Ullah N, Bakhtiar SM, Zhang T et al. Pangenome analysis of Mycobacterium tuberculosis reveals core-drug targets and screening of promising lead compounds for drug discovery. Antibiotics (Basel) 2020; 9:E819 [View Article] [PubMed]
    [Google Scholar]
  47. Yang T, Zhong J, Zhang J, Li C, Yu X et al. Pan-genomic study of Mycobacterium tuberculosis reflecting the primary/secondary genes, generality/individuality, and the interconversion through copy number variations. Front Microbiol 2018; 9:1886 [View Article] [PubMed]
    [Google Scholar]
  48. Meehan CJ, Goig GA, Kohl TA, Verboven L, Dippenaar A et al. Whole genome sequencing of Mycobacterium tuberculosis: current standards and open issues. Nat Rev Microbiol 2019; 17:533–545 [View Article] [PubMed]
    [Google Scholar]
  49. Safi H, Gopal P, Lingaraju S, Ma S, Levine C et al. Phase variation in Mycobacterium tuberculosis glpK produces transiently heritable drug tolerance. Proc Natl Acad Sci U S A 2019; 116:19665–19674 [View Article] [PubMed]
    [Google Scholar]
  50. Fan X, Abd Alla AAE, Xie J. Distribution and function of prophage phiRv1 and phiRv2 among Mycobacterium tuberculosis complex. J Biomol Struct Dyn 2016; 34:233–238 [View Article] [PubMed]
    [Google Scholar]
  51. Contreras-Moreira B, Vinuesa P. GET_HOMOLOGUES, a versatile software package for scalable and robust microbial pangenome analysis. Appl Environ Microbiol 2013; 79:7696–7701 [View Article] [PubMed]
    [Google Scholar]
  52. Salzberg SL. Next-generation genome annotation: we still struggle to get it right. Genome Biol 2019; 20:92 [View Article] [PubMed]
    [Google Scholar]
  53. Brockhurst MA, Harrison E, Hall JPJ, Richards T, McNally A et al. The ecology and evolution of pangenomes. Curr Biol 2019; 29:R1094–R1103 [View Article] [PubMed]
    [Google Scholar]
  54. Menardo F, Duchêne S, Brites D, Gagneux S. The molecular clock of Mycobacterium tuberculosis. PLoS Pathog 2019; 15:e1008067 [View Article] [PubMed]
    [Google Scholar]
  55. Bellerose MM, Baek S-H, Huang C-C, Moss CE, Koh E-I et al. Common variants in the glycerol kinase gene reduce tuberculosis drug efficacy. mBio 2019; 10:e00663-19 [View Article] [PubMed]
    [Google Scholar]
  56. Keating LA, Wheeler PR, Mansoor H, Inwald JK, Dale J et al. The pyruvate requirement of some members of the Mycobacterium tuberculosis complex is due to an inactive pyruvate kinase: implications for in vivo growth. Mol Microbiol 2005; 56:163–174 [View Article] [PubMed]
    [Google Scholar]
  57. Vargas R, Farhat MR. Antibiotic treatment and selection for. Proc Natl Acad Sci U S A 2020; 117:3910–3912 [View Article]
    [Google Scholar]
  58. Godfroid M, Dagan T, Merker M, Kohl TA, Diel R et al. Insertion and deletion evolution reflects antibiotics selection pressure in a Mycobacterium tuberculosis outbreak. PLoS Pathog 2020; 16:e1008357 [View Article] [PubMed]
    [Google Scholar]
  59. Chattopadhyay S, Weissman SJ, Minin VN, Russo TA, Dykhuizen DE et al. High frequency of hotspot mutations in core genes of Escherichia coli due to short-term positive selection. Proc Natl Acad Sci U S A 2009; 106:12412–12417 [View Article] [PubMed]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/mgen/10.1099/mgen.0.000839
Loading
/content/journal/mgen/10.1099/mgen.0.000839
Loading

Data & Media loading...

Supplements

Loading data from figshare Loading data from figshare
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