Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

RNA landscape of the emerging cancer-associated microbe Fusobacterium nucleatum

Abstract

Fusobacterium nucleatum, long known as a constituent of the oral microflora, has recently garnered renewed attention for its association with several different human cancers. The growing interest in this emerging cancer-associated bacterium contrasts with a paucity of knowledge about its basic gene expression features and physiological responses. As fusobacteria lack all established small RNA-associated proteins, post-transcriptional networks in these bacteria are also unknown. In the present study, using differential RNA-sequencing, we generate high-resolution global RNA maps for five clinically relevant fusobacterial strains—F. nucleatum subspecies nucleatum, animalis, polymorphum and vincentii, as well as F. periodonticum—for early, mid-exponential growth and early stationary phase. These data are made available in an online browser, and we use these to uncover fundamental aspects of fusobacterial gene expression architecture and a suite of non-coding RNAs. Developing a vector for functional analysis of fusobacterial genes, we discover a conserved fusobacterial oxygen-induced small RNA, FoxI, which serves as a post-transcriptional repressor of the major outer membrane porin FomA. Our findings provide a crucial step towards delineating the regulatory networks enabling F. nucleatum adaptation to different environments, which may elucidate how these bacteria colonize different compartments of the human body.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Differential RNA-seq for F. nucleatum subsp. nucleatum.
Fig. 2: Transcriptome features of known virulence factors and prediction of small proteins in F. nucleatum subsp. nucleatum.
Fig. 3: Identification of core ncRNAs and an active CRISPR–Cas system in F. nucleatum subsp. nucleatum.
Fig. 4: The sRNA landscape in Fusobacterium sp.
Fig. 5: FoxI is an oxygen-induced conserved sRNA.
Fig. 6: Overexpression of FoxI identifies the OM protein FomA as a potential target of the sRNA.

Similar content being viewed by others

Data availability

RNA-seq data can be accessed at NCBI’s GEO (https://www.ncbi.nlm.nih.gov/geo) under the accession no. GSE161360. MS data can be accessed at the Proteomics Identification Database PRIDE (https://www.ebi.ac.uk/pride) under the accession no. PXD022474. The Rfam database can be accessed at http://rfam.xfam.org. The Uniprot database can be accessed at https://www.uniprot.org/uniprot/?query=taxonomy:190304. Source data are provided with this paper.

References

  1. Dzutsev, A. et al. Microbes and cancer. Annu. Rev. Immunol. 35, 199–228 (2017).

    Article  CAS  PubMed  Google Scholar 

  2. Parhi, L. et al. Breast cancer colonization by Fusobacterium nucleatum accelerates tumor growth and metastatic progression. Nat. Commun. 11, 3259 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Kostic, A. D. et al. Genomic analysis identifies association of Fusobacterium with colorectal carcinoma. Genome Res. 22, 292–298 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Castellarin, M. et al. Fusobacterium nucleatum infection is prevalent in human colorectal carcinoma. Genome Res. 22, 299–306 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Hussan, H., Clinton, S. K., Roberts, K. & Bailey, M. T. Fusobacterium’s link to colorectal neoplasia sequenced: a systematic review and future insights. World J. Gastroenterol. 23, 8626–8650 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Brennan, C. A. & Garrett, W. S. Fusobacterium nucleatum—symbiont, opportunist and oncobacterium. Nat. Rev. Microbiol. 17, 156–166 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Bullman, S. et al. Analysis of Fusobacterium persistence and antibiotic response in colorectal cancer. Science 358, 1443–1448 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Amitay, E. L. et al. Fusobacterium and colorectal cancer: causal factor or passenger? Results from a large colorectal cancer screening study. Carcinogenesis 38, 781–788 (2017).

    Article  PubMed  Google Scholar 

  9. Yu, T. et al. Fusobacterium nucleatum promotes chemoresistance to colorectal cancer by modulating autophagy. Cell 170, 548–563.e516 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Gaiser, R. A. et al. Enrichment of oral microbiota in early cystic precursors to invasive pancreatic cancer. Gut 68, 2186–2194 (2019).

    Article  CAS  PubMed  Google Scholar 

  11. Yamamura, K. et al. Fusobacterium nucleatum in gastroenterological cancer: evaluation of measurement methods using quantitative polymerase chain reaction and a literature review. Oncol. Lett. 14, 6373–6378 (2017).

    PubMed  PubMed Central  Google Scholar 

  12. Yamamura, K. et al. Human microbiome Fusobacterium nucleatum in esophageal cancer tissue is associated with prognosis. Clin. Cancer Res. 22, 5574–5581 (2016).

    Article  CAS  PubMed  Google Scholar 

  13. Rubinstein, M. R. et al. Fusobacterium nucleatum promotes colorectal carcinogenesis by modulating E-cadherin/beta-catenin signaling via its FadA adhesin. Cell Host Microbe 14, 195–206 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Abed, J. et al. Fap2 mediates Fusobacterium nucleatum colorectal adenocarcinoma enrichment by binding to tumor-expressed Gal-GalNAc. Cell Host Microbe 20, 215–225 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Han, Y. W. et al. Identification and characterization of a novel adhesin unique to oral fusobacteria. J. Bacteriol. 187, 5330–5340 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Holt, R. A. & Cochrane, K. Tumor potentiating mechanisms of Fusobacterium nucleatum, a multifaceted microbe. Gastroenterology 152, 694–696 (2017).

    Article  PubMed  Google Scholar 

  17. Kaplan, C. W. et al. Fusobacterium nucleatum outer membrane proteins Fap2 and RadD induce cell death in human lymphocytes. Infect. Immun. 78, 4773–4778 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Gur, C. et al. Binding of the Fap2 protein of Fusobacterium nucleatum to human inhibitory receptor TIGIT protects tumors from immune cell attack. Immunity 42, 344–355 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Cochrane, K., Robinson, A. V., Holt, R. A. & Allen-Vercoe, E. A survey of Fusobacterium nucleatum genes modulated by host cell infection. Microb. Genom. https://doi.org/10.1099/mgen.0.000300 (2020).

  20. Abed, J. et al. Colon cancer-associated Fusobacterium nucleatum may originate from the oral cavity and reach colon tumors via the circulatory system. Front. Cell Infect. Microbiol. 10, 400 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Han, Y. W. Fusobacterium nucleatum: a commensal-turned pathogen. Curr. Opin. Microbiol. 23, 141–147 (2015).

    Article  CAS  PubMed  Google Scholar 

  22. Manson McGuire, A. et al. Evolution of invasion in a diverse set of Fusobacterium species. mBio 5, e01864 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Sanders, B. E., Umana, A., Lemkul, J. A. & Slade, D. J. FusoPortal: an interactive repository of hybrid MinION-sequenced Fusobacterium genomes improves gene identification and characterization. mSphere https://doi.org/10.1128/mSphere.00228-18 (2018).

  24. Kook, J. K. et al. Genome-based reclassification of Fusobacterium nucleatum subspecies at the species level. Curr. Microbiol. 74, 1137–1147 (2017).

    Article  CAS  PubMed  Google Scholar 

  25. Merritt, J., Niu, G., Okinaga, T. & Qi, F. Autoaggregation response of Fusobacterium nucleatum. Appl. Environ. Microbiol. 75, 7725–7733 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Mutha, N. V. R. et al. Transcriptional responses of Streptococcus gordonii and Fusobacterium nucleatum to coaggregation. Mol. Oral Microbiol. 33, 450–464 (2018).

    Article  CAS  PubMed  Google Scholar 

  27. Sasaki-Imamura, T., Yano, A. & Yoshida, Y. Production of indole from l-tryptophan and effects of these compounds on biofilm formation by Fusobacterium nucleatum ATCC 25586. Appl. Environ. Microbiol. 76, 4260–4268 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Wagner, E. G. & Romby, P. Small RNAs in bacteria and archaea: who they are, what they do, and how they do it. Adv. Genet. 90, 133–208 (2015).

    Article  CAS  PubMed  Google Scholar 

  29. Hör, J., Gorski, S. A. & Vogel, J. Bacterial RNA biology on a genome scale. Mol. Cell 70, 785–799 (2018).

    Article  PubMed  CAS  Google Scholar 

  30. Hör, J., Matera, G., Vogel, J., Gottesman, S. & Storz, G. Trans-acting small RNAs and their effects on gene expression in Escherichia coli and Salmonella enterica. EcoSal Plus https://doi.org/10.1128/ecosalplus.ESP-0030-2019 (2020)

  31. Babitzke, P., Lai, Y. J., Renda, A. J. & Romeo, T. Posttranscription initiation control of gene expression mediated by bacterial RNA-binding proteins. Annu. Rev. Microbiol. 73, 43–67 (2019).

    Article  CAS  PubMed  Google Scholar 

  32. Barquist, L. & Vogel, J. Accelerating discovery and functional analysis of small RNAs with new technologies. Annu. Rev. Genet. 49, 367–394 (2015).

    Article  CAS  PubMed  Google Scholar 

  33. Ang, M. Y. et al. Comparative genome analysis of Fusobacterium nucleatum. Genome Biol. Evol. 8, 2928–2938 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Pyne, M. E., Bruder, M. R., Moo-Young, M., Chung, D. A. & Chou, C. P. Harnessing heterologous and endogenous CRISPR–Cas machineries for efficient markerless genome editing in Clostridium. Sci. Rep. 6, 25666 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Li, Y. et al. Harnessing Type I and Type III CRISPR–Cas systems for genome editing. Nucleic Acids Res. 44, e34 (2016).

    Article  PubMed  Google Scholar 

  36. Sharma, C. M. et al. The primary transcriptome of the major human pathogen Helicobacter pylori. Nature 464, 250–255 (2010).

    Article  CAS  PubMed  Google Scholar 

  37. Minarovits, J. Anaerobic bacterial communities associated with oral carcinoma: Intratumoral, surface-biofilm and salivary microbiota. Anaerobe 68, 102300 (2021).

    Article  CAS  PubMed  Google Scholar 

  38. Sharma, C. M. & Vogel, J. Differential RNA-seq: the approach behind and the biological insight gained. Curr. Opin. Microbiol. 19, 97–105 (2014).

    Article  CAS  PubMed  Google Scholar 

  39. Yu, S.-H., Vogel, J. & Förstner, K. U. ANNOgesic: a Swiss army knife for the RNA-seq based annotation of bacterial/archaeal genomes. GigaScience 7, giy096 (2018).

    Article  PubMed Central  CAS  Google Scholar 

  40. Georg, J. & Hess, W. R. Widespread antisense transcription in prokaryotes. Microbiol. Spectrum https://doi.org/10.1128/microbiolspec.RWR-0029-2018 (2018).

  41. Feklistov, A., Sharon, B. D., Darst, S. A. & Gross, C. A. Bacterial sigma factors: a historical, structural, and genomic perspective. Annu. Rev. Microbiol. 68, 357–376 (2014).

    Article  CAS  PubMed  Google Scholar 

  42. Browning, D. F. & Busby, S. J. W. The regulation of bacterial transcription initiation. Nat. Rev. Microbiol. 2, 57 (2004).

    Article  CAS  PubMed  Google Scholar 

  43. Karpathy, S. E. et al. Genome sequence of Fusobacterium nucleatum subspecies polymorphum—a genetically tractable fusobacterium. PLoS ONE 2, e659 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Kapatral, V. et al. Genome sequence and analysis of the oral bacterium Fusobacterium nucleatum strain ATCC 25586. J. Bacteriol. 184, 2005–2018 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Bailey, T. L., Johnson, J., Grant, C. E. & Noble, W. S. The MEME suite. Nucleic Acids Res. 43, W39–W49 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Dugar, G. et al. High-resolution transcriptome maps reveal strain-specific regulatory features of multiple Campylobacter jejuni isolates. PLoS Genet. 9, e1003495 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Zanzoni, A., Spinelli, L., Braham, S. & Brun, C. Perturbed human sub-networks by Fusobacterium nucleatum candidate virulence proteins. Microbiome 5, 89 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Kapatral, V. et al. Genome analysis of F. nucleatum sub spp vincentii and its comparison with the genome of F. nucleatum ATCC 25586. Genome Res. 13, 1180–1189 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Desvaux, M., Khan, A., Beatson, S. A., Scott-Tucker, A. & Henderson, I. R. Protein secretion systems in Fusobacterium nucleatum: genomic identification of Type 4 piliation and complete Type V pathways brings new insight into mechanisms of pathogenesis. Biochim. Biophys. Acta 1713, 92–112 (2005).

    Article  CAS  PubMed  Google Scholar 

  50. Umana, A. et al. Utilizing whole Fusobacterium genomes to identify, correct, and characterize potential virulence protein families. J. Bacteriol. https://doi.org/10.1128/JB.00273-19 (2019).

  51. Xu, M. et al. FadA from Fusobacterium nucleatum utilizes both secreted and nonsecreted forms for functional oligomerization for attachment and invasion of host cells. J. Biol. Chem. 282, 25000–25009 (2007).

    Article  CAS  PubMed  Google Scholar 

  52. Kaplan, C. W., Lux, R., Haake, S. K. & Shi, W. The Fusobacterium nucleatum outer membrane protein RadD is an arginine-inhibitable adhesin required for inter-species adherence and the structured architecture of multispecies biofilm. Mol. Microbiol. 71, 35–47 (2009).

    Article  CAS  PubMed  Google Scholar 

  53. Engevik, M. et al. Fusobacterium nucleatum adheres to Clostridioides difficile via the RadD adhesin to enhance biofilm formation in intestinal mucus. Gastroenterology https://doi.org/10.1053/j.gastro.2020.11.034 (2020).

  54. Doron, L. et al. Identification and characterization of fusolisin, the Fusobacterium nucleatum autotransporter serine protease. PLoS ONE 9, e111329 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Kumar, A., Thotakura, P. L., Tiwary, B. K. & Krishna, R. Target identification in Fusobacterium nucleatum by subtractive genomics approach and enrichment analysis of host–pathogen protein–protein interactions. BMC Microbiol. 16, 84 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Storz, G., Wolf, Y. I. & Ramamurthi, K. S. Small proteins can no longer be ignored. Annu. Rev. Biochem. 83, 753–777 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Cao, H., Ma, Q., Chen, X. & Xu, Y. DOOR: a prokaryotic operon database for genome analyses and functional inference. Brief. Bioinform. 20, 1568–1577 (2019).

    Article  CAS  PubMed  Google Scholar 

  58. Casasanta, M. A. et al. A chemical and biological toolbox for Type Vd secretion: characterization of the phospholipase A1 autotransporter FplA from Fusobacterium nucleatum. J. Biol. Chem. 292, 20240–20254 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Loh, E., Righetti, F., Eichner, H., Twittenhoff, C. & Narberhaus, F. RNA thermometers in bacterial pathogens. Microbiol. Spectrum https://doi.org/10.1128/microbiolspec.RWR-0012-2017 (2018).

  60. McCown, P. J., Corbino, K. A., Stav, S., Sherlock, M. E. & Breaker, R. R. Riboswitch diversity and distribution. RNA 23, 995–1011 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Kalvari, I. et al. Rfam 13.0: shifting to a genome-centric resource for non-coding RNA families. Nucleic Acids Res. 46, D335–D342 (2018).

    Article  CAS  PubMed  Google Scholar 

  62. Winkler, W. C., Nahvi, A., Roth, A., Collins, J. A. & Breaker, R. R. Control of gene expression by a natural metabolite-responsive ribozyme. Nature 428, 281–286 (2004).

    Article  CAS  PubMed  Google Scholar 

  63. Johnsen, M., Christensen, T., Dennis, P. P. & Fiil, N. P. Autogenous control: ribosomal protein L10–L12 complex binds to the leader sequence of its mRNA. EMBO J. 1, 999–1004 (1982).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Yakhnin, H., Yakhnin, A. V. & Babitzke, P. Ribosomal protein L10(L12)4 autoregulates expression of the Bacillus subtilis rplJL operon by a transcription attenuation mechanism. Nucleic Acids Res. 43, 7032–7043 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Bonner, E. R., D’Elia, J. N., Billips, B. K. & Switzer, R. L. Molecular recognition of pyr mRNA by the Bacillus subtilis attenuation regulatory protein PyrR. Nucleic Acids Res. 29, 4851–4865 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Akopian, D., Shen, K., Zhang, X. & Shan, S. O. Signal recognition particle: an essential protein-targeting machine. Annu. Rev. Biochem. 82, 693–721 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Hartmann, E. & Hartmann, R. K. The enigma of ribonuclease P evolution. Trends Genet. 19, 561–569 (2003).

    Article  CAS  PubMed  Google Scholar 

  68. Wassarman, K. M. 6S RNA, a global regulator of transcription. Microbiol. Spectrum https://doi.org/10.1128/microbiolspec.RWR-0019-2018 (2018).

  69. Wehner, S., Damm, K., Hartmann, R. K. & Marz, M. Dissemination of 6S RNA among bacteria. RNA Biol. 11, 1467–1478 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  70. Cochrane, K. et al. Complete genome sequences and analysis of the Fusobacterium nucleatum subspecies animalis 7-1 bacteriophage ɸFunu1 and ɸFunu2. Anaerobe 38, 125–129 (2016).

    Article  CAS  PubMed  Google Scholar 

  71. Richter, H. et al. Characterization of CRISPR RNA processing in Clostridium thermocellum and Methanococcus maripaludis. Nucleic Acids Res. 40, 9887–9896 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Millman, A. et al. Bacterial retrons function in anti-phage defense. Cell https://doi.org/10.1016/j.cell.2020.09.065 (2020).

  73. Melamed, S., Adams, P. P., Zhang, A., Zhang, H. & Storz, G. RNA–RNA interactomes of ProQ and Hfq reveal overlapping and competing roles. Mol. Cell 77, 411–425 e417 (2020).

    Article  CAS  PubMed  Google Scholar 

  74. Smirnov, A. et al. Grad-seq guides the discovery of ProQ as a major small RNA-binding protein. Proc. Natl Acad. Sci. USA 113, 11591–11596 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Bak, G. et al. Identification of novel sRNAs involved in biofilm formation, motility, and fimbriae formation in Escherichia coli. Sci. Rep. 5, 15287 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Loh, E. et al. A trans-acting riboswitch controls expression of the virulence regulator PrfA in Listeria monocytogenes. Cell 139, 770–779 (2009).

    Article  CAS  PubMed  Google Scholar 

  77. Koonin, E. V., Makarova, K. S. & Wolf, Y. I. Evolutionary genomics of defense systems in Archaea and bacteria. Annu. Rev. Microbiol 71, 233–261 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Fagan, R. P. & Fairweather, N. F. Clostridium difficile has two parallel and essential Sec secretion systems. J. Biol. Chem. 286, 27483–27493 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Mann, M., Wright, P. R. & Backofen, R. IntaRNA 2.0: enhanced and customizable prediction of RNA–RNA interactions. Nucleic Acids Res. 45, W435–W439 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Wu, C. et al. Forward genetic dissection of biofilm development by Fusobacterium nucleatum: novel functions of cell division proteins FtsX and EnvC. mBio https://doi.org/10.1128/mBio.00360-18 (2018).

  81. Coppenhagen-Glazer, S. et al. Fap2 of Fusobacterium nucleatum is a galactose-inhibitable adhesin involved in coaggregation, cell adhesion, and preterm birth. Infect. Immun. 83, 1104–1113 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Brown, S. P., Cornforth, D. M. & Mideo, N. Evolution of virulence in opportunistic pathogens: generalism, plasticity, and control. Trends Microbiol. 20, 336–342 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Bayley, D. P., Rocha, E. R. & Smith, C. J. Analysis of cepA and other Bacteroides fragilis genes reveals a unique promoter structure. FEMS Microbiol. Lett. 193, 149–154 (2000).

    Article  CAS  PubMed  Google Scholar 

  84. Ryan, D., Jenniches, L., Reichardt, S., Barquist, L. & Westermann, A. J. A high-resolution transcriptome map identifies small RNA regulation of metabolism in the gut microbe Bacteroides thetaiotaomicron. Nat. Commun. 11, 3557 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Jackson, C. A. et al. A consensus Porphyromonas gingivalis promoter sequence. FEMS Microbiol. Lett. 186, 133–138 (2000).

    Article  CAS  PubMed  Google Scholar 

  86. Michaux, C. et al. Single-nucleotide RNA maps for the two major nosocomial pathogens Enterococcus faecalis and Enterococcus faecium. Front. Cell Infect. Microbiol. 10, 600325 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Olejniczak, M. & Storz, G. ProQ/FinO-domain proteins: another ubiquitous family of RNA matchmakers? Mol. Microbiol. 104, 905–915 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Holmqvist, E. & Vogel, J. RNA-binding proteins in bacteria. Nat. Rev. Microbiol. 16, 601–615 (2018).

    Article  CAS  PubMed  Google Scholar 

  89. Zheng, J. J., Perez, A. J., Tsui, H. T., Massidda, O. & Winkler, M. E. Absence of the KhpA and KhpB (JAG/EloR) RNA-binding proteins suppresses the requirement for PBP2b by overproduction of FtsA in Streptococcus pneumoniae D39. Mol. Microbiol. 106, 793–814 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Lamm-Schmidt, V. et al. Grad-seq identifies KhpB as a global RNA-binding protein in Clostridioides difficile that regulates toxin production. microLife https://doi.org/10.1093/femsml/uqab004 (2021).

  91. Hör, J. et al. Grad-seq in a Gram-positive bacterium reveals exonucleolytic sRNA activation in competence control. EMBO J. 39, e103852 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  92. Altuvia, S., Weinstein-Fischer, D., Zhang, A., Postow, L. & Storz, G. A small, stable RNA induced by oxidative stress: role as a pleiotropic regulator and antimutator. Cell 90, 43–53 (1997).

    Article  CAS  PubMed  Google Scholar 

  93. Pfeiffer, V. et al. A small non-coding RNA of the invasion gene island (SPI-1) represses outer membrane protein synthesis from the Salmonella core genome. Mol. Microbiol. 66, 1174–1191 (2007).

    Article  CAS  PubMed  Google Scholar 

  94. Guisbert, E., Rhodius, V. A., Ahuja, N., Witkin, E. & Gross, C. A. Hfq modulates the sigmaE-mediated envelope stress response and the sigma32-mediated cytoplasmic stress response in Escherichia coli. J. Bacteriol. 189, 1963–1973 (2007).

    Article  CAS  PubMed  Google Scholar 

  95. Vanderpool, C. K. & Gottesman, S. The novel transcription factor SgrR coordinates the response to glucose-phosphate stress. J. Bacteriol. 189, 2238–2248 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Nejman, D. et al. The human tumor microbiome is composed of tumor type-specific intracellular bacteria. Science 368, 973–980 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Swidsinski, A. et al. Acute appendicitis is characterised by local invasion with Fusobacterium nucleatum/necrophorum. Gut 60, 34–40 (2011).

    Article  PubMed  Google Scholar 

  98. Han, X. Y. et al. Fusobacterial brain abscess: a review of five cases and an analysis of possible pathogenesis. J. Neurosurg. 99, 693–700 (2003).

    Article  PubMed  Google Scholar 

  99. Gregory, S. W., Boyce, T. G., Larson, A. N., Patel, R. & Jackson, M. A. Fusobacterium nucleatum osteomyelitis in 3 previously healthy children: a case series and review of the literature. J. Pediatr. Infect. Dis. Soc. 4, e155–e159 (2015).

    Article  Google Scholar 

  100. Truant, A. L., Menge, S., Milliorn, K., Lairscey, R. & Kelly, M. T. Fusobacterium nucleatum pericarditis. J. Clin. Microbiol. 17, 349–351 (1983).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Altshuler, G. & Hyde, S. Clinicopathologic considerations of fusobacteria chorioamnionitis. Acta Obstet. Gynecol. Scand. 67, 513–517 (1988).

    Article  CAS  PubMed  Google Scholar 

  102. Toussi, D. N., Liu, X. & Massari, P. The FomA porin from Fusobacterium nucleatum is a Toll-like receptor 2 agonist with immune adjuvant activity. Clin. Vaccin. Immunol. 19, 1093–1101 (2012).

    Article  CAS  Google Scholar 

  103. Westermann, A. J. et al. Dual RNA-seq unveils noncoding RNA functions in host–pathogen interactions. Nature 529, 496–501 (2016).

    Article  CAS  PubMed  Google Scholar 

  104. Imdahl, F., Vafadarnejad, E., Homberger, C., Saliba, A. E. & Vogel, J. Single-cell RNA-sequencing reports growth-condition-specific global transcriptomes of individual bacteria. Nat. Microbiol. https://doi.org/10.1038/s41564-020-0774-1 (2020).

  105. Blattman, S. B., Jiang, W., Oikonomou, P. & Tavazoie, S. Prokaryotic single-cell RNA sequencing by in situ combinatorial indexing. Nat. Microbiol. 5, 1192–1201 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Ji, Y. et al. Identification of critical staphylococcal genes using conditional phenotypes generated by antisense RNA. Science 293, 2266–2269 (2001).

    Article  CAS  PubMed  Google Scholar 

  107. Blomberg, P., Wagner, E. G. & Nordstrom, K. Control of replication of plasmid R1: the duplex between the antisense RNA, CopA, and its target, CopT, is processed specifically in vivo and in vitro by RNase III. EMBO J. 9, 2331–2340 (1990).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Berezikov, E. et al. Diversity of microRNAs in human and chimpanzee brain. Nat. Genet. 38, 1375–1377 (2006).

    Article  CAS  PubMed  Google Scholar 

  109. Förstner, K. U., Vogel, J. & Sharma, C. M. READemption: a tool for the computational analysis of deep-sequencing-based transcriptome data. Bioinformatics 30, 3421–3423 (2014).

    Article  PubMed  CAS  Google Scholar 

  110. Otto, C., Stadler, P. F. & Hoffmann, S. Fast and sensitive mapping of bisulfite-treated sequencing data. Bioinformatics 28, 1698–1704 (2012).

    Article  CAS  PubMed  Google Scholar 

  111. Kingsford, C. L., Ayanbule, K. & Salzberg, S. L. Rapid, accurate, computational discovery of Rho-independent transcription terminators illuminates their relationship to DNA uptake. Genome Biol. 8, R22 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  112. Quinlan, A. R. BEDTools: the Swiss-army tool for genome feature analysis. Curr. Protoc. Bioinform. 47, 11.12.11–34 (2014).

    Article  Google Scholar 

  113. Haake, S. K., Yoder, S. C., Attarian, G. & Podkaminer, K. Native plasmids of Fusobacterium nucleatum: characterization and use in development of genetic systems. J. Bacteriol. 182, 1176–1180 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Knoke, L. R. et al. Agrobacterium tumefaciens small lipoprotein Atu8019 is involved in selective outer membrane vesicle (OMV) docking to bacterial cells. Front. Microbiol. 11, 1228 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  115. Cox, J. & Mann, M. MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nat. Biotechnol. 26, 1367–1372 (2008).

    Article  CAS  PubMed  Google Scholar 

  116. Drozdetskiy, A., Cole, C., Procter, J. & Barton, G. J. JPred4: a protein secondary structure prediction server. Nucleic Acids Res. 43, W389–W394 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Almagro Armenteros, J. J. et al. SignalP 5.0 improves signal peptide predictions using deep neural networks. Nat. Biotechnol. 37, 420–423 (2019).

    Article  CAS  PubMed  Google Scholar 

  118. Lorenz, R. et al. ViennaRNA package 2.0. Algorithms Mol. Biol. 6, 26 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank Y. el Mouali Benomar, J. Hör, D. Ryan and G. Bachrach for fruitful discussions, E. Venturini for support with the subcellular fractionation, G. Bachrach (Hebrew University of Jerusalem, Israel) for providing pORI92, E. Allen-Vercoe (University of Guelph, Canada) for providing Fusobacterium strains and L. Jenniches for help with launching the web browser. We thank S. Lamer and A. Schlosser for MS analysis. We thank the Vogel Stiftung Dr. Eckernkamp for supporting F.P. with a Dr. Eckernkamp Fellowship. This work was funded by a DFG Gottfried Wilhelm Leibniz award to J.V. (DFG Vo875‐18). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

F.P. performed most of the experiments. C.T. and Y.Z. conducted experiments for determining growth stages. F.P. performed data analysis. F.P., F.F. and J.V. designed research. L.B. set up the web browser. J.V. directed research. F.P., F.F. and J.V. wrote the manuscript.

Corresponding author

Correspondence to Jörg Vogel.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Microbiology thanks Cari Vanderpool and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Overview of the dRNA-seq analysis for F. nucleatum subsp. polymorphum.

a, Venn diagram showing the number and class-distribution of detected TSS for F. nucleatum subsp. polymorphum. b, Length distribution and corresponding occurrences of all 800 5′-UTR yielding from a pTSS (black) or sTSS (red) are shown. The consensus Shine Dalgarno sequence is displayed with the average distance from the start codon. c, Motif search using MEME in the 50-nt region upstream of pTSS identified an extended -10 box similar to the σ70 binding site in E. coli for ~92% of pTSS. While most of the sequences harbour an AT-rich stretch preceding the -10 box, a -35 box was only detected in ~28% of promoter regions.

Extended Data Fig. 2 Overview of the dRNA-seq analysis for F. nucleatum subsp. animalis.

a, Venn diagram showing the number and class-distribution of detected TSS for F. nucleatum subsp. animalis. b, Length distribution and corresponding occurrences of all 758 5′-UTR yielding from a pTSS (black) or sTSS (red) are shown. The consensus Shine Dalgarno sequence is displayed with the average distance from the start codon. c, Motif search using MEME in the 50-nt region upstream of pTSS identified an extended −10 box similar and −35 box to the σ70 binding site in E. coli for ~92% of pTSS separated by an AT-rich stretch.

Extended Data Fig. 3 Overview of the dRNA-seq analysis for F. nucleatum subsp. vincentii.

a, Venn diagram showing the number and class-distribution of detected TSS for F. nucleatum subsp. vincentii. b, Length distribution and corresponding occurrences of all 589 5′-UTR yielding from a pTSS (black) or sTSS (red) are shown. The consensus Shine Dalgarno sequence is displayed with the average distance from the start codon. c, Motif search using MEME in the 50-nt region upstream of pTSS identified an extended -10 box similar to the σ70 binding site in E. coli for ~78% of pTSS. While most of the sequences harbour an AT-rich stretch preceding the -10 box, a -35 box was only detected in ~31% of promoter regions.

Extended Data Fig. 4 Overview of the dRNA-seq analysis for F. periodonticum.

a, Venn diagram showing the number and class-distribution of detected TSS for F. periodonticum. b, Length distribution and corresponding occurrences of all 638 5′-UTR yielding from a pTSS (black) or sTSS (red) are shown. The consensus Shine Dalgarno sequence is displayed with the average distance from the start codon. c, Motif search using MEME in the 50-nt region upstream of pTSS identified an extended -10 box similar and -35 box to the σ70 binding site in E. coli for ~88% of pTSS separated by an AT-rich stretch.

Extended Data Fig. 5 Annotation of riboswitches and cis-acting elements in Fnn.

Overview of genomic locations for predicted riboswitches that were identified by RFAM analysis.

Extended Data Fig. 6 Sequence conservation and secondary structure of 4.5S RNA in Fnn.

a, Alignment of 4.5S RNA sequences from representative strains of different Fusobacterium species and the subspecies of Fusobacterium nucleatum (FNN = F. n. subsp. nucleatum; FNA = F. n. subsp. animalis; FNP = F. n. subsp. polymorphum; FNV = F. n. subsp. vincentii; FuH = F. hwasookii; FuP = F. periodonticum; FuG = F. gonidiaiformans; FuM = F. mortiferum; FuN = F. necrophorum; FuU = F. ulcerans; FuV = F. varium). The TSS and the conserved apical tetraloop are indicated. b, Shown is prediction for the secondary structure of the 4.5S RNA in Fnn.

Extended Data Fig. 7 Sequence conservation of tmRNA in the Fusobacterium genus.

a, Alignment of tmRNA sequences from representative strains of Fusobacterium species and subspecies of Fusobacterium nucleatum (FNN = F. n. subsp. nucleatum; FNA = F. n. subsp. animalis; FNP = F. n. subsp. polymorphum; FNV = F. n. subsp. vincentii; FuH = F. hwasookii; FuP = F. periodonticum; FuG = F. gonidiaiformans; FuM = F. mortiferum; FuN = F. necrophorum; FuU = F. ulcerans; FuV = F. varium). The conserved tag peptide is highlighted (yellow). b, Alignment of the ORF for the tag peptide reveals sequence dichotomy between oral isolates and fusobacteria found elsewhere.

Extended Data Fig. 8 Alignment of 6S RNA sequences identified in the Fusobacterium genus.

Shown is the alignment for the 6S RNA in representative strains of Fusobacterium species and subspecies of Fusobacterium nucleatum (FNN = F. n. subsp. nucleatum; FNA = F. n. subsp. animalis; FNP = F. n. subsp. polymorphum; FNV = F. n. subsp. vincentii; FuH = F. hwasookii; FuP = F. periodonticum; FuG = F. gonidiaiformans; FuM = F. mortiferum; FuN = F. necrophorum; FuU = F. ulcerans; FuV = F. varium). The region encoding for the pRNA is highlighted (blue).

Extended Data Fig. 9 Analysis of FoxI overexpression.

a, Comparison of Fnn (ATCC 23726) growth harbouring empty vector control (ctrl.), FoxI-expressing (FoxI) or FoxI-3C-expressing (FoxI-3C) plasmid, respectively. A 24 h pre-culture was diluted 1:50 into fresh Columbia Broth and measured every 20 min with 10 s shaking prior to each measurement. Growth data are represented as the mean (± standard deviation) from three biological replicates. b, Northern blot analysis of FoxI overexpression during mid-exponential growth phase. c-f,Comparison of the excised bands for the region of interest for two replicates for either the empty vector control (ctrl.), the overexpression of FoxI (FoxI) or the mutant FoxI-3C (FoxI-3C). Shown are the estimated protein abundances by iBAQ quantification for all proteins detected in both replicates of each group. c, Comparison of total protein samples for the empty vector control. d, Comparison of total protein samples for the FoxI overexpression. e, Comparison of total protein samples for the FoxI-3C overexpression. f, Comparison of the same region for the outer membrane fraction in the control (Fig. 6b). FomA is indicated (red) and was detected as the most abundant protein in all samples with a clear reduction in the FoxI-overexpression.

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–4 and references.

Reporting Summary

Supplementary Table 1

Overview of detected TSSs.

Supplementary Table 2

Results of differential gene expression analysis.

Supplementary Table 3

Overview of predicted 5′-UTRs.

Supplementary Table 4

Comparison of putative virulence factors to pTSSs in Fnn.

Supplementary Table 5

Reannotated CDSs in Fnn.

Supplementary Table 6

Predicted operons in Fnn.

Supplementary Table 7

Overview of predicted riboregulatory elements.

Supplementary Table 8

Overview of predicted sRNAs.

Supplementary Table 9

Used oligonucleotides, plasmids and strains in the present study.

Source data

Source Data Fig. 3

Unprocessed northern blots.

Source Data Fig. 4

Unprocessed northern blots.

Source Data Fig. 5

Unprocessed northern blots.

Source Data Fig. 6

Unprocessed northern blots, western blots and/or gels.

Source Data Extended Data Fig. 9

Unprocessed northern blots.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ponath, F., Tawk, C., Zhu, Y. et al. RNA landscape of the emerging cancer-associated microbe Fusobacterium nucleatum. Nat Microbiol 6, 1007–1020 (2021). https://doi.org/10.1038/s41564-021-00927-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-021-00927-7

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing