Characterization of sialidase and disruption of its role in host–pathogen interactions Free

Abstract

Key to onset and progression of periodontitis is a complex relationship between oral bacteria and the host. The organisms most associated with severe periodontitis are the periodontal pathogens of the red complex: , and . These organisms express sialidases, which cleave sialic acid from host glycoproteins, and contribute to disease through various mechanisms. Here, we expressed and purified recombinant sialidase SiaPG (PG_0352) and characterized its activity on a number of substrates, including host sialoglycoproteins and highlighting the inability to cleave diacetylated sialic acids – a phenomenon overcome by the NanS sialate-esterase from . Indeed SiaPG required NanS to maximize sialic acid harvesting from heavily O-acetylated substrates such as bovine salivary mucin, hinting at the possibility of interspecies cooperation in sialic acid release from host sources by these members of the oral microbiota. Activity of SiaPG and was inhibited using the commercially available chemotherapeutic zanamivir, indicating its potential as a virulence inhibitor, which also inhibited sialic acid release from mucin, and was capable of inhibiting biofilm formation of on oral glycoprotein sources. Zanamivir also inhibited attachment and invasion of oral epithelial cells by and other periodontal pathogens, both in monospecies but also in multispecies infection experiments, indicating potential to suppress host–pathogen interactions of a mixed microbial community. This study broadens our understanding of the multifarious roles of bacterial sialidases in virulence, and indicates that their inhibition with chemotherapeutics could be a promising strategy for periodontitis therapy.

Loading

Article metrics loading...

/content/journal/micro/10.1099/mic.0.000851
2019-11-01
2024-03-28
Loading full text...

Full text loading...

/deliver/fulltext/micro/165/11/1181.html?itemId=/content/journal/micro/10.1099/mic.0.000851&mimeType=html&fmt=ahah

References

  1. Hajishengallis G, Darveau RP, Curtis MA. The keystone-pathogen hypothesis. Nat Rev Microbiol 2012; 10:717–725 [View Article]
    [Google Scholar]
  2. Meuric V, Le Gall-David S, Boyer E, Acuña-Amador L, Martin B et al. Signature of microbial dysbiosis in periodontitis. Appl Environ Microbiol 2017; 83:1–13 [View Article]
    [Google Scholar]
  3. Socransky SS, Cugini MA, Cugini MA, Smith C, Kent RL. Microbial complexes in subgingival plaque. J Clin Periodontol 1998; 25:134–144 [View Article]
    [Google Scholar]
  4. Ximénez-Fyvie La, Haffajee AD, Socransky SS. Comparison of the microbiota of supra- and subgingival plaque in health and periodontitis. J Clin Periodontol 2000; 27:648–657 [View Article]
    [Google Scholar]
  5. Muniz FWMG, de Oliveira CC, de Sousa Carvalho R, Moreira MMSM, de Moraes MEA et al. Azithromycin: a new concept in adjuvant treatment of periodontitis. Eur J Pharmacol 2013; 705:135–139 [View Article]
    [Google Scholar]
  6. Colombo APV, Boches SK, Cotton SL, Goodson JM, Kent R et al. Comparisons of subgingival microbial profiles of refractory periodontitis, severe periodontitis, and periodontal health using the human oral microbe identification microarray. J Periodontol 2009; 80:1421–1432 [View Article]
    [Google Scholar]
  7. Löhr G, Beikler T, Hensel A. Inhibition of in vitro adhesion and virulence of Porphyromonas gingivalis by aqueous extract and polysaccharides from Rhododendron ferrugineum L. a new way for prophylaxis of periodontitis?. Fitoterapia 2015; 107:105–113 [View Article]
    [Google Scholar]
  8. Kariu T, Nakao R, Ikeda T, Nakashima K, Potempa J et al. Inhibition of gingipains and Porphyromonas gingivalis growth and biofilm formation by prenyl flavonoids. J Periodontal Res 2017; 52:89–96 [View Article]
    [Google Scholar]
  9. Huq NL, Seers CA, Toh ECY, Dashper SG, Slakeski N et al. Propeptide-mediated inhibition of cognate gingipain proteinases. PLoS One 2013; 8:e65447 [View Article]
    [Google Scholar]
  10. Dominy SS, Lynch C, Ermini F, Benedyk M, Marczyk A et al. Porphyromonas gingivalis in Alzheimer’s disease brains: Evidence for disease causation and treatment with small-molecule inhibitors. Sci. Adv. 2019; 5:eaau3333 [View Article]
    [Google Scholar]
  11. Parker D, Soong G, Planet P, Brower J, Ratner AJ et al. The nana neuraminidase of Streptococcus pneumoniae is involved in biofilm formation. Infect Immun 2009; 77:3722–3730 [View Article]
    [Google Scholar]
  12. Galen JE, Ketley JM, Fasano A, Richardson SH, Wasserman SS et al. Role of Vibrio cholerae neuraminidase in the function of cholera toxin. Infect Immun 1992; 60:406–415
    [Google Scholar]
  13. Stafford G, Roy S, Honma K, Sharma A. Sialic acid, periodontal pathogens and Tannerella forsythia: stick around and enjoy the feast!. Mol Oral Microbiol 2012; 27:11–22 [View Article]
    [Google Scholar]
  14. Palmer RJ. Composition and development of oral bacterial communities. Periodontol 2000 2014; 64:20–39 [View Article]
    [Google Scholar]
  15. Frey AM, Ansbro K, Kamble NS, Pham TK, Stafford GP. Characterisation and pure culture of putative health-associated oral bacterium BU063 (Tannerella sp. HOT-286) reveals presence of a potentially novel glycosylated S-layer. FEMS Microbiol Lett 2018; 365:1–8 [View Article]
    [Google Scholar]
  16. Roy S, Honma K, Douglas CWI, Sharma A, Stafford GP et al. Role of sialidase in glycoprotein utilization by Tannerella forsythia . Microbiology 2011; 157:3195–3202 [View Article]
    [Google Scholar]
  17. Li C, Kurniyati HB, Hu B, Bian J, Sun J et al. Abrogation of neuraminidase reduces biofilm formation, capsule biosynthesis, and virulence of Porphyromonas gingivalis . Infect Immun 2012; 80:3–13 [View Article]
    [Google Scholar]
  18. Aruni W, Vanterpool E, Osbourne D, Roy F, Muthiah A et al. Sialidase and sialoglycoproteases can modulate virulence in Porphyromonas gingivalis. Infect Immun 2011; 79:2779–2791 [View Article]
    [Google Scholar]
  19. Kurniyati K, Zhang W, Zhang K, Li C. A surface-exposed neuraminidase affects complement resistance and virulence of the oral spirochaete Treponema denticola . Mol Microbiol 2013; 89:842–856 [View Article]
    [Google Scholar]
  20. Duran-Pinedo AE, Chen T, Teles R, Starr JR, Wang X et al. Community-Wide transcriptome of the oral microbiome in subjects with and without periodontitis. Isme J 2014; 8:1659–1672 [View Article]
    [Google Scholar]
  21. Gul SS, Griffiths GS, Stafford GP, Al-Zubidi MI, Rawlinson A et al. Investigation of a novel predictive biomarker profile for the outcome of periodontal treatment. J Periodontol 2017; 88:11351144 [View Article]
    [Google Scholar]
  22. Honma K, Mishima E, Sharma A. Role of Tannerella forsythia NanH sialidase in epithelial cell attachment. Infect Immun 2011; 79:393–401 [View Article]
    [Google Scholar]
  23. Dickson MA, Hahn WC, Ino Y, Ronfard V, Wu JY et al. Human keratinocytes that express hTERT and also bypass a p16(INK4a)-enforced mechanism that limits life span become immortal yet retain normal growth and differentiation characteristics. Mol Cell Biol 2000; 20:1436–1447 [View Article]
    [Google Scholar]
  24. Thomas GJ, Lewis MP, Whawell SA, Russell A, Sheppard D et al. Expression of the alphavbeta6 integrin promotes migration and invasion in squamous carcinoma cells. J Invest Dermatol 2001; 117:67–73
    [Google Scholar]
  25. Sugiyama M, Speight PM, Prime SS, Watt FM. Comparison of integrin expression and terminal differentiation capacity in cell lines derived from oral squamous cell carcinomas. Carcinogenesis 1993; 14:2171–2176 [View Article]
    [Google Scholar]
  26. Phansopa C, Kozak RP, Liew LP, Frey AM, Farmilo T et al. Characterization of a sialate-O-acetylesterase (NanS) from the oral pathogen Tannerella forsythia that enhances sialic acid release by NanH, its cognate sialidase. Biochem J 2015; 472:157–167 [View Article]
    [Google Scholar]
  27. Frey AM, Satur MJ, Phansopa C, Parker JL, Bradshaw D et al. Evidence for a novel carbohydrate binding module (CBM) of Tannerella forsythia NanH sialidase, key to interactions at the host-pathogen interface. Biochem J 2018; 20170592:BCJ20170592
    [Google Scholar]
  28. Honma K, Mishima E, Inagaki S, Sharma A. The oxyR homologue in Tannerella forsythia regulates expression of oxidative stress responses and biofilm formation. Microbiology 2009; 155:1912–1922 [View Article]
    [Google Scholar]
  29. Royle L, Mattu TS, Hart E, Langridge JI, Merry AH et al. An analytical and structural database provides a strategy for sequencing O-glycans from microgram quantities of glycoproteins. Anal Biochem 2002; 304:70–90 [View Article]
    [Google Scholar]
  30. 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]
    [Google Scholar]
  31. Komatsu T, Nagano K, Sugiura S, Hagiwara M, Tanigawa N et al. E-Selectin mediates Porphyromonas gingivalis adherence to human endothelial cells. Infect Immun 2012; 80:2570–2576 [View Article]
    [Google Scholar]
  32. Varki A, Diaz S. A neuraminidase from Streptococcus sanguis that can release O-acetylated sialic acids. J Biol Chem 1983; 258:12465–12471
    [Google Scholar]
  33. Suwannakul S, Stafford GP, Whawell SA, Douglas CWI. Identification of bistable populations of Porphyromonas gingivalis that differ in epithelial cell invasion. Microbiology 2010; 156:3052–3064 [View Article]
    [Google Scholar]
  34. Al-Taweel FB, Douglas CWI, Whawell SA. The periodontal pathogen Porphyromonas gingivalis preferentially interacts with oral epithelial cells in S phase of the cell cycle. Infect Immun 2016; 84:1966–1974 [View Article]
    [Google Scholar]
  35. Naylor KL, Widziolek M, Hunt S, Conolly M, Hicks M et al. Role of OmpA2 surface regions of Porphyromonas gingivalis in host – pathogen interactions with oral epithelial cells. Microbiol Open 20171–11
    [Google Scholar]
  36. Saito A, Kokubu E, Inagaki S, Imamura K, Kita D et al. Porphyromonas gingivalis entry into gingival epithelial cells modulated by Fusobacterium nucleatum is dependent on lipid rafts. Microb Pathog 2012; 53:234–242 [View Article]
    [Google Scholar]
  37. Inagaki S, Onishi S, Kuramitsu HK, Sharma A. Porphyromonas gingivalis vesicles enhance attachment, and the leucine-rich repeat BspA protein is required for invasion of epithelial cells by "Tannerella forsythia". Infect Immun 2006; 74:5023–5028 [View Article]
    [Google Scholar]
  38. Kirschbaum M, Schultze-Mosgau S, Pfister W, Eick S. Mixture of periodontopathogenic bacteria influences interaction with KB cells. Anaerobe 2010; 16:461–468 [View Article]
    [Google Scholar]
  39. Sharma A, Inagaki S, Sigurdson W, Kuramitsu HK. Synergy between Tannerella forsythia and Fusobacterium nucleatum in biofilm formation. Oral Microbiol Immunol 2005; 20:39–42 [View Article]
    [Google Scholar]
  40. Settem RP, El-Hassan AT, Honma K, Stafford GP, Sharma A. Fusobacterium nucleatum and Tannerella forsythia induce synergistic alveolar bone loss in a mouse periodontitis model. Infect Immun 2012; 80:2436–2443 [View Article]
    [Google Scholar]
  41. Tan KH, Seers CA, Dashper SG, Mitchell HL, Pyke JS et al. Porphyromonas gingivalis and Treponema denticola exhibit metabolic symbioses. PLoS Pathog 2014; 10:e1003955 [View Article]
    [Google Scholar]
  42. Govinden G, Parker JL, Naylor KL, Frey AM, Anumba DOC et al. Inhibition of sialidase activity and cellular invasion by the bacterial vaginosis pathogen Gardnerella vaginalis . Arch Microbiol 2018; 200:1129–1133 [View Article]
    [Google Scholar]
  43. Walther E, Richter M, Xu Z, Kramer C, von Grafenstein S et al. Antipneumococcal activity of neuraminidase inhibiting artocarpin. Int J Med Microbiol 2015; 305:289–297 [View Article]
    [Google Scholar]
  44. Roy S, Honma K, Douglas CWI, Sharma A, Stafford GP. Role of sialidase in glycoprotein utilization by Tannerella forsythia. Microbiology 2011; 157:3195–3202 [View Article]
    [Google Scholar]
  45. Lewis WG, Robinson LS, Perry J, Bick JL, Peipert JF et al. Hydrolysis of secreted sialoglycoprotein immunoglobulin A (IgA) in ex vivo and biochemical models of bacterial vaginosis. J Biol Chem 2012; 287:2079–2089 [View Article]
    [Google Scholar]
  46. Davey ME, Duncan MJ. Enhanced biofilm formation and loss of capsule synthesis: deletion of a putative glycosyltransferase in Porphyromonas gingivalis. J Bacteriol 2006; 188:5510–5523 [View Article]
    [Google Scholar]
  47. Bao K, Belibasakis GN, Thurnheer T, Aduse-Opoku J, Curtis MA et al. Role of Porphyromonas gingivalis gingipains in multi-species biofilm formation. BMC Microbiol 2014; 14:1–8 [View Article]
    [Google Scholar]
  48. Bloch S, Thurnheer T, Murakami Y, Belibasakis GN, Schäffer C et al. Behavior of two Tannerella forsythia strains and their cell surface mutants in multispecies oral biofilms. Mol Oral Microbiol 2017; 32:404–418 [View Article]
    [Google Scholar]
  49. Han YW, Ikegami A, Rajanna C, Kawsar HI, Zhou Y et al. Identification and characterization of a novel adhesin unique to oral fusobacteria. J Bacteriol 2005; 187:5330–5340 [View Article]
    [Google Scholar]
  50. Saito A, Inagaki S, Ishihara K. Differential ability of periodontopathic bacteria to modulate invasion of human gingival epithelial cells by Porphyromonas gingivalis . Microb Pathog 2009; 47:329–333 [View Article]
    [Google Scholar]
  51. Stamatos NM, Carubelli I, van de Vlekkert D, Bonten EJ, Papini N et al. Lps-Induced cytokine production in human dendritic cells is regulated by sialidase activity. J Leukoc Biol 2010; 88:1227–1239 [View Article]
    [Google Scholar]
  52. Hata K, Koseki K, Yamaguchi K, Moriya S, Suzuki Y et al. Limited inhibitory effects of oseltamivir and zanamivir on human sialidases. Antimicrob Agents Chemother 2008; 52:3484–3491 [View Article]
    [Google Scholar]
  53. Amith SR, Jayanth P, Franchuk S, Finlay T, Seyrantepe V et al. Neu1 desialylation of sialyl α-2,3-linked β-galactosyl residues of Toll-like receptor 4 is essential for receptor activation and cellular signaling. Cell Signal 2010; 22:314–324 [View Article]
    [Google Scholar]
  54. Amith SR, Jayanth P, Franchuk S, Siddiqui S, Seyrantepe V et al. Dependence of pathogen molecule-induced Toll-like receptor activation and cell function on NEU1 sialidase. Glycoconj J 2009; 26:1197–1212 [View Article]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/micro/10.1099/mic.0.000851
Loading
/content/journal/micro/10.1099/mic.0.000851
Loading

Data & Media loading...

Supplements

Supplementary material 1

PDF

Most cited Most Cited RSS feed