1887

Abstract

DNA damage often causes an arrest of the cell cycle that provides time for genome integrity to be restored. In bacteria, the classical SOS DNA damage response leads to an inhibition of cell division resulting in temporarily filamentous growth. This raises the question as to whether such a response mechanism might similarly function in naturally filamentous bacteria such as exhibit two functionally distinct forms of cell division: cross-wall formation in vegetative hyphae and sporulation septation in aerial hyphae. Here, we show that the genotoxic agent mitomycin C confers a block in sporulation septation in in a mechanism that involves, at least in part, the downregulation of . Notably, this DNA damage response does not appear to block cross-wall formation and may be independent of canonical SOS and developmental regulators. We also show that the mitomycin C-induced block in sporulation can be partially bypassed by the constitutive expression of , though this appears to be largely limited to mitomycin C treatment and the resultant spore-like cells have reduced viability.

Funding
This study was supported by the:
  • Biotechnology and Biological Sciences Research Council (Award BB/J004561/1)
    • Principle Award Recipient: NotApplicable
  • Royal Society (Award RGF\EA\181026)
    • Principle Award Recipient: SusanSchlimpert
  • Royal Society (Award URF\R1\180075)
    • Principle Award Recipient: SusanSchlimpert
  • Institute of Infection and Immunity (Award MID-406688)
    • Principle Award Recipient: JustinR. Nodwell
  • Mitacs (Award IT14924)
    • Principle Award Recipient: JanVT. Falguera
  • 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/micro/10.1099/mic.0.001198
2022-06-15
2024-04-19
Loading full text...

Full text loading...

/deliver/fulltext/micro/168/6/mic001198.html?itemId=/content/journal/micro/10.1099/mic.0.001198&mimeType=html&fmt=ahah

References

  1. Falguera JVT, Stratton KJ, Bush MJ, Jani C, Findlay KC et al. DNA damage-induced block of sporulation in Streptomyces venezuelae involves downregulation of ssgB. Figshare 2022 [View Article]
    [Google Scholar]
  2. Butala M, Zgur-Bertok D, Busby SJW. The bacterial LexA transcriptional repressor. Cell Mol Life Sci 2009; 66:82–93 [View Article] [PubMed]
    [Google Scholar]
  3. Little JW, Mount DW. The SOS regulatory system of Escherichia coli. Cell 1982; 29:11–22 [View Article] [PubMed]
    [Google Scholar]
  4. Kreuzer KN. DNA damage responses in prokaryotes: regulating gene expression, modulating growth patterns, and manipulating replication forks. Cold Spring Harb Perspect Biol 2013; 5:a012674 [View Article] [PubMed]
    [Google Scholar]
  5. Mukherjee A, Cao C, Lutkenhaus J. Inhibition of FtsZ polymerization by SulA, an inhibitor of septation in Escherichia coli. Proc Natl Acad Sci U S A 1998; 95:2885–2890 [View Article] [PubMed]
    [Google Scholar]
  6. Kawai Y, Moriya S, Ogasawara N. Identification of a protein, YneA, responsible for cell division suppression during the SOS response in Bacillus subtilis. Mol Microbiol 2003; 47:1113–1122 [View Article] [PubMed]
    [Google Scholar]
  7. Mo AH, Burkholder WF. YneA, an SOS-induced inhibitor of cell division in Bacillus subtilis, is regulated posttranslationally and requires the transmembrane region for activity. J Bacteriol 2010; 192:3159–3173 [View Article] [PubMed]
    [Google Scholar]
  8. Masser EA, Burby PE, Hawkins WD, Gustafson BR, Lenhart JS et al. DNA damage checkpoint activation affects peptidoglycan synthesis and late divisome components in Bacillus subtilis. Mol Microbiol 2021; 116:707–722 [View Article] [PubMed]
    [Google Scholar]
  9. Ogino H, Teramoto H, Inui M, Yukawa H. DivS, a novel SOS-inducible cell-division suppressor in Corynebacterium glutamicum. Mol Microbiol 2008; 67:597–608 [View Article] [PubMed]
    [Google Scholar]
  10. Modell JW, Hopkins AC, Laub MT. A DNA damage checkpoint in Caulobacter crescentus inhibits cell division through A direct interaction with FtsW. Genes Dev 2011; 25:1328–1343 [View Article] [PubMed]
    [Google Scholar]
  11. Modell JW, Kambara TK, Perchuk BS, Laub MT. A DNA damage-induced, SOS-independent checkpoint regulates cell division in Caulobacter crescentus. PLOS Biol 2014; 12:e1001977 [View Article] [PubMed]
    [Google Scholar]
  12. Chauhan A, Lofton H, Maloney E, Moore J, Fol M et al. Interference of Mycobacterium tuberculosis cell division by Rv2719c, a cell wall hydrolase. Mol Microbiol 2006; 62:132–147 [View Article] [PubMed]
    [Google Scholar]
  13. Bojer MS, Wacnik K, Kjelgaard P, Gallay C, Bottomley AL et al. SosA inhibits cell division in Staphylococcus aureus in response to DNA damage. Mol Microbiol 2019; 112:1116–1130 [View Article] [PubMed]
    [Google Scholar]
  14. Bush MJ, Tschowri N, Schlimpert S, Flärdh K, Buttner MJ. c-di-GMP signalling and the regulation of developmental transitions in streptomycetes. Nat Rev Microbiol 2015; 13:749–760 [View Article] [PubMed]
    [Google Scholar]
  15. Flärdh K, Buttner MJ. Streptomyces morphogenetics: dissecting differentiation in a filamentous bacterium. Nat Rev Microbiol 2009; 7:36–49 [View Article] [PubMed]
    [Google Scholar]
  16. McCormick JR, Su EP, Driks A, Losick R. Growth and viability of Streptomyces coelicolor mutant for the cell division gene ftsZ. Mol Microbiol 1994; 14:243–254 [View Article]
    [Google Scholar]
  17. Bisson-Filho AW, Hsu Y-P, Squyres GR, Kuru E, Wu F et al. Treadmilling by FtsZ filaments drives peptidoglycan synthesis and bacterial cell division. Science 2017; 355:739–743 [View Article] [PubMed]
    [Google Scholar]
  18. Yang X, Lyu Z, Miguel A, McQuillen R, Huang KC et al. GTPase activity-coupled treadmilling of the bacterial tubulin FtsZ organizes septal cell wall synthesis. Science 2017; 355:744–747 [View Article] [PubMed]
    [Google Scholar]
  19. McCormick JR. Cell division is dispensable but not irrelevant in Streptomyces. Curr Opin Microbiol 2009; 12:689–698 [View Article] [PubMed]
    [Google Scholar]
  20. Keijser BJF, Noens EEE, Kraal B, Koerten HK, van Wezel GP. The Streptomyces coelicolor ssgB gene is required for early stages of sporulation. FEMS Microbiol Lett 2003; 225:59–67 [View Article] [PubMed]
    [Google Scholar]
  21. Sevcikova B, Kormanec J. The ssgB gene, encoding a member of the regulon of stress-response sigma factor sigmaH, is essential for aerial mycelium septation in Streptomyces coelicolor A3(2). Arch Microbiol 2003; 180:380–384 [View Article] [PubMed]
    [Google Scholar]
  22. Willemse J, Borst JW, de Waal E, Bisseling T, van Wezel GP. Positive control of cell division: FtsZ is recruited by SsgB during sporulation of Streptomyces. Genes Dev 2011; 25:89–99 [View Article] [PubMed]
    [Google Scholar]
  23. Santos-Beneit F, Roberts DM, Cantlay S, McCormick JR, Errington J. A mechanism for FtsZ-independent proliferation in Streptomyces. Nat Commun 2017; 8:1378 [View Article] [PubMed]
    [Google Scholar]
  24. Bush MJ, Chandra G, Bibb MJ, Findlay KC, Buttner MJ et al. Genome-wide chromatin immunoprecipitation sequencing analysis shows that WhiB is a transcription factor that cocontrols its regulon with whia to initiate developmental cell division in Streptomyces. mBio 2016; 7: [View Article] [PubMed]
    [Google Scholar]
  25. Chater KF. A morphological and genetic mapping study of white colony mutants of Streptomyces coelicolor. J Gen Microbiol 1972; 72:9–28 [View Article] [PubMed]
    [Google Scholar]
  26. McAuley S, Huynh A, Howells A, Walpole C, Maxwell A et al. Discovery of a novel DNA gyrase-targeting antibiotic through the chemical perturbation of Streptomyces venezuelae sporulation. Cell Chem Biol 2019; 26:1274–1282 [View Article] [PubMed]
    [Google Scholar]
  27. Jani C, Tocheva EI, McAuley S, Craney A, Jensen GJ et al. Streptomyces: a screening tool for bacterial cell division inhibitors. J Biomol Screen 2015; 20:275–284 [View Article] [PubMed]
    [Google Scholar]
  28. Kieser T, Bibb MJ, Buttner MJ, Chater KF, Hopwood DA. Practical Streptomyces genetics Norwich: John Innes Foundation; 2000
    [Google Scholar]
  29. Paget MSB, Chamberlin L, Atrih A, Foster SJ, Buttner MJ. Evidence that the extracytoplasmic function sigma factor sigmaE is required for normal cell wall structure in Streptomyces coelicolor A3(2). J Bacteriol 1999; 181:204–211 [View Article] [PubMed]
    [Google Scholar]
  30. Fernández-Martínez LT, Bibb MJ. Use of the Meganuclease I-SceI of Saccharomycescerevisiae to select for gene deletions in actinomycetes. Sci Rep 2014; 4: [View Article] [PubMed]
    [Google Scholar]
  31. Gibson DG, Young L, Chuang RY, Venter JC, Hutchison CA et al. Enzymatic assembly of DNA molecules up to several hundred kilobases. Nat Methods 2009; 6:343–345 [View Article] [PubMed]
    [Google Scholar]
  32. Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 2013; 29:15–21 [View Article] [PubMed]
    [Google Scholar]
  33. Jani C. PhD thesis. McMaster University; 2015 http://hdl.handle.net/11375/18466
  34. Trapnell C, Roberts A, Goff L, Pertea G, Kim D et al. Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat Protoc 2012; 7:562–578 [View Article] [PubMed]
    [Google Scholar]
  35. Aranda PS, LaJoie DM, Jorcyk CL. Bleach gel: A simple agarose gel for analyzing RNA quality. Electrophoresis 2012; 33:366–369 [View Article] [PubMed]
    [Google Scholar]
  36. Jones SE, Leong V, Ortega J, Elliot MA. Development, antibiotic production, and ribosome assembly in Streptomyces venezuelae are impacted by RNase J and RNase III deletion. J Bacteriol 2014; 196:4253–4267 [View Article] [PubMed]
    [Google Scholar]
  37. Bush MJ, Chandra G, Al-Bassam MM, Findlay KC, Buttner MJ. BldC delays entry into development to produce a sustained period of vegetative growth in Streptomyces venezuelae. mBio 2019; 10:e02812-18 [View Article] [PubMed]
    [Google Scholar]
  38. Bush MJ, Gallagher KA, Chandra G, Findlay KC, Schlimpert S. Hyphal compartmentalization and sporulation in Streptomyces require the conserved cell division protein SepX. Nat Commun 2022; 13:71 [View Article] [PubMed]
    [Google Scholar]
  39. Ramos-León F, Bush MJ, Sallmen JW, Chandra G, Richardson J et al. A conserved cell division protein directly regulates FtsZ dynamics in filamentous and unicellular actinobacteria. Elife 2021; 10:e63387 [View Article] [PubMed]
    [Google Scholar]
  40. Bush MJ, Bibb MJ, Chandra G, Findlay KC, Buttner MJ. Genes required for aerial growth, cell division, and chromosome segregation are targets of WhiA before sporulation in Streptomyces venezuelae. mBio 2013; 4:e00684-13 [View Article] [PubMed]
    [Google Scholar]
  41. Schindelin J, Arganda-Carreras I, Frise E, Kaynig V, Longair M et al. Fiji: an open-source platform for biological-image analysis. Nat Methods 2012; 9:676–682 [View Article] [PubMed]
    [Google Scholar]
  42. Schlimpert S, Flärdh K, Buttner MJ. Fluorescence time-lapse imaging of the complete S. venezuelae life cycle using a microfluidic device. J Vis Exp 201653863 [View Article] [PubMed]
    [Google Scholar]
  43. Šmídová K, Ziková A, Pospíšil J, Schwarz M, Bobek J et al. DNA mapping and kinetic modeling of the HrdB regulon in Streptomyces coelicolor. Nucleic Acids Res 2019; 47:621–633 [View Article] [PubMed]
    [Google Scholar]
  44. Drlica K, Malik M, Kerns RJ, Zhao X. Quinolone-mediated bacterial death. Antimicrob Agents Chemother 2008; 52:385–392 [View Article] [PubMed]
    [Google Scholar]
  45. Chankova SG, Dimova E, Dimitrova M, Bryant PE. Induction of DNA double-strand breaks by zeocin in Chlamydomonas reinhardtii and the role of increased DNA double-strand breaks rejoining in the formation of an adaptive response. Radiat Environ Biophys 2007; 46:409–416 [View Article] [PubMed]
    [Google Scholar]
  46. Schlimpert S, Wasserstrom S, Chandra G, Bibb MJ, Findlay KC et al. Two dynamin-like proteins stabilize FtsZ rings during Streptomyces sporulation. Proc Natl Acad Sci U S A 2017; 114:E6176–E6183 [View Article] [PubMed]
    [Google Scholar]
  47. Kormanec J, Sevcikova B. The stress-response sigma factor sigma(H) controls the expression of ssgB, a homologue of the sporulation-specific cell division gene ssgA, in Streptomyces coelicolor A3(2). Mol Genet Genomics 2002; 267:536–543 [View Article] [PubMed]
    [Google Scholar]
  48. Goranov AI, Katz L, Breier AM, Burge CB, Grossman AD. A transcriptional response to replication status mediated by the conserved bacterial replication protein DnaA. Proc Natl Acad Sci U S A 2005; 102:12932–12937 [View Article] [PubMed]
    [Google Scholar]
  49. Daniel RA, Harry EJ, Katis VL, Wake RG, Errington J. Characterization of the essential cell division gene ftsL(yIID) of Bacillus subtilis and its role in the assembly of the division apparatus. Mol Microbiol 1998; 29:593–604 [View Article] [PubMed]
    [Google Scholar]
  50. Kim SH, Traag BA, Hasan AH, McDowall KJ, Kim B-G et al. Transcriptional analysis of the cell division-related ssg genes in Streptomyces coelicolor reveals direct control of ssgR by AtrA. Antonie van Leeuwenhoek 2015; 108:201–213 [View Article] [PubMed]
    [Google Scholar]
  51. Tschowri N, Schumacher MA, Schlimpert S, Chinnam NB, Findlay KC et al. Tetrameric c-di-GMP mediates effective transcription factor dimerization to control Streptomyces development. Cell 2014; 158:1136–1147 [View Article] [PubMed]
    [Google Scholar]
  52. Al-Bassam MM, Bibb MJ, Bush MJ, Chandra G, Buttner MJ. Response regulator heterodimer formation controls a key stage in Streptomyces development. PLOS Genet 2014; 10:e1004554 [View Article] [PubMed]
    [Google Scholar]
  53. Kawamoto S, Watanabe H, Hesketh A, Ensign JC, Ochi K. Expression analysis of the ssgA gene product, associated with sporulation and cell division in Streptomyces griseus. Microbiology (Reading) 1997; 143 (Pt 4):1077–1086 [View Article] [PubMed]
    [Google Scholar]
  54. van Wezel GP, Krabben P, Traag BA, Keijser BJF, Kerste R et al. Unlocking Streptomyces spp. for use as sustainable industrial production platforms by morphological engineering. Appl Environ Microbiol 2006; 72:5283–5288 [View Article] [PubMed]
    [Google Scholar]
  55. Traag BA, van Wezel GP. The SsgA-like proteins in actinomycetes: small proteins up to a big task. Antonie van Leeuwenhoek 2008; 94:85–97 [View Article] [PubMed]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/micro/10.1099/mic.0.001198
Loading
/content/journal/micro/10.1099/mic.0.001198
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