1887

Abstract

is a cariogenic bacterium that causes dental caries as well as being implicated in other dental pathologies and infective endocarditis. Bacitracin is a bactericidal antibiotic that induces cell wall stress in Gram-positive bacteria.

is among the most characterized Gram-positive bacteria. However, the transcriptome and proteome of have received less attention, and they are actually key in understanding the pathogenesis of any bacteria. In this study, we extracted the whole proteome of grown under bacitracin stress. Such a proteome is anticipated to offer deep insights related to physiological dynamic fluctuations and, consequently, it may provide ‘proteomic signatures’ to be identified as potential targets.

The aim of the study is to explore the general stress response that exhibits at the proteome level when cell wall stress is imposed on it.

A sub-MIC concentration of bacitracin was added to the growth media of followed by whole-cell protein extraction. The proteome was then subjected to high-throughput proteomics analysis, i.e. liquid chromatography tandem mass spectrometry (LC-MS/MS). Differentially expressed proteins obtained through LC-MS/MS underwent analyses such as gene ontology, KEGG (Kyoto Encyclopaedia of Genes and Genomes) and DAVID (Database for Annotation, Visualization and Integrated Discovery) analysis, and STRING for functional annotation, pathway enrichment and protein–protein interaction (PPI) networks, respectively. These proteins were also categorized into functional classes using the PANTHER (Protein Annotation Through Evolutionary Relationship) classification system.

LC-MS/MS produced data from 321 identified proteins. From these, 41 and 30 were found to be significantly over- (≥2 fold change) and underexpressed (≤0.4 fold change), respectively. In the upregulated proteins we mostly observed sortases and proteins involved in the EPS biosynthesis pathway, whereas among the downregulated proteins the majority related to glycolysis.

The sortase family of proteins appear to be potential targets because they regulate various virulence factors and therefore can be targeted to inhibit multiple virulence pathways simultaneously. This study offers an understanding of proteomic fluctuations in response to cell wall stress and can thus help in identifying key players mediating virulence.

Funding
This study was supported by the:
  • Department of Biotechnology, Ministry of Science and Technology, India (Award BT/PR40148/BTIS/137/20/2021)
    • Principle Award Recipient: AsadU Khan
Loading

Article metrics loading...

/content/journal/jmm/10.1099/jmm.0.001572
2022-08-30
2024-05-03
Loading full text...

Full text loading...

References

  1. Lemos JA, Burne RA. A model of efficiency: stress tolerance by Streptococcus mutans . Microbiology 2008; 154:3247–3255 [View Article] [PubMed]
    [Google Scholar]
  2. Lemos JA, Palmer SR, Zeng L, Wen ZT, Kajfasz JK et al. The biology of Streptococcus mutans . Microbiol Spectr 2019; 18:435–448
    [Google Scholar]
  3. Ajdić D, McShan WM, McLaughlin RE, Savić G, Chang J et al. Genome sequence of Streptococcus mutans UA159, a cariogenic dental pathogen. Proc Natl Acad Sci U S A 2002; 99:14434–14439 [View Article] [PubMed]
    [Google Scholar]
  4. Nakano K, Ooshima T. Serotype classification of Streptococcus mutans and its detection outside the oral cavity. Future Microbiol 2009; 4:891–902 [View Article] [PubMed]
    [Google Scholar]
  5. Dong L, Tong Z, Linghu D, Lin Y, Tao R et al. Effects of sub-minimum inhibitory concentrations of antimicrobial agents on Streptococcus mutans biofilm formation. Int J Antimicrob Agents 2012; 39:390–395 [View Article]
    [Google Scholar]
  6. Pollock TJ, Thorne L, Yamazaki M, Mikolajczak MJ, Armentrout RW. Mechanism of bacitracin resistance in gram-negative bacteria that synthesize exopolysaccharides. J Bacteriol 1994; 176:6229–6237 [View Article] [PubMed]
    [Google Scholar]
  7. Gottschalk S, Gottlieb CT, Vestergaard M, Hansen PR, Gram L et al. Amphibian antimicrobial peptide fallaxin analogue FL9 affects virulence gene expression and DNA replication in Staphylococcus aureus. J Med Microbiol 2015; 64:1504–1513 [View Article]
    [Google Scholar]
  8. Aswal M, Garg A, Singhal N, Kumar M. Comparative in-silico proteomic analysis discerns potential granuloma proteins of Yersinia pseudotuberculosis. Sci Rep 2020; 10:1–13 [View Article] [PubMed]
    [Google Scholar]
  9. Polikanov YS, Aleksashin NA, Beckert B, Wilson DN. The mechanisms of action of ribosome-targeting peptide antibiotics. Front Mol Biosci 2018; 5:48 [View Article] [PubMed]
    [Google Scholar]
  10. Ming LJ, Epperson JD. Metal binding and structure-activity relationship of the metalloantibiotic peptide bacitracin. J Inorg Biochem 2002; 91:46–58 [View Article] [PubMed]
    [Google Scholar]
  11. Mi H, Muruganujan A, Casagrande JT, Thomas PD. Large-scale gene function analysis with the PANTHER classification system. Nat Protoc 2013; 8:1551–1566 [View Article] [PubMed]
    [Google Scholar]
  12. Zhu M, Ajdić D, Liu Y, Lynch D, Merritt J et al. Role of the Streptococcus mutans irvA gene in GbpC-independent, dextran-dependent aggregation and biofilm formation. Appl Environ Microbiol 2009; 75:7037–7043 [View Article] [PubMed]
    [Google Scholar]
  13. Sharma D, Garg A, Kumar M, Khan AU. Proteome profiling of carbapenem-resistant K. pneumoniae clinical isolate (NDM-4): Exploring the mechanism of resistance and potential drug targets. J Proteomics 2019; 200:102–110 [View Article]
    [Google Scholar]
  14. Dey AK, Kumar B, Singh AK, Ranjan P, Thiruvengadam R et al. Salivary proteome signatures in the early and middle stages of human pregnancy with term birth outcome. Sci Rep 2020; 10:1–15 [View Article] [PubMed]
    [Google Scholar]
  15. UniProt Consortium UniProt: a worldwide hub of protein knowledge. Nucleic Acids Res 2019; 47:D506–D515 [View Article] [PubMed]
    [Google Scholar]
  16. Carbon S. Expansion of the gene ontology knowledgebase and resources. Nucleic Acids Res 2017; 45:D331–D338 [View Article] [PubMed]
    [Google Scholar]
  17. Mi H, Muruganujan A, Huang X, Ebert D, Mills C et al. Protocol Update for large-scale genome and gene function analysis with the PANTHER classification system (v.14.0). Nat Protoc 2019; 14:703–721 [View Article] [PubMed]
    [Google Scholar]
  18. Altermann E, Klaenhammer TR. Pathway voyager: pathway mapping using the Kyoto Encyclopedia of Genes and Genomes (KEGG) database. BMC Genomics 2005; 6:1–7 [View Article]
    [Google Scholar]
  19. Huang DW, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc 2009; 4:44–57 [View Article] [PubMed]
    [Google Scholar]
  20. Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S et al. STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res 2019; 47:D607–D613 [View Article] [PubMed]
    [Google Scholar]
  21. Ahn S-J, Desai S, Blanco L, Lin M, Rice KC. Acetate and potassium modulate the stationary-phase activation of lrgAB in Streptococcus mutans . Front Microbiol 2020; 11:401 [View Article]
    [Google Scholar]
  22. Mihara H, Esaki N. Bacterial cysteine desulfurases: their function and mechanisms. Appl Microbiol Biotechnol 2002; 60:12–23
    [Google Scholar]
  23. Shen CJ, Kuo TY, Lin CC, Chow LP, Chen WJ. Proteomic identification of membrane proteins regulating antimicrobial peptide resistance in Vibrio parahaemolyticus. J Appl Microbiol 2010; 108:1398–1407 [View Article] [PubMed]
    [Google Scholar]
  24. Maaß S, Bartel J, Mücke P-A, Schlüter R, Sura T et al. Proteomic adaptation of Clostridioides difficile to treatment with the antimicrobial peptide nisin. Cells 2021; 10:372 [View Article] [PubMed]
    [Google Scholar]
  25. Fränzel B, Frese C, Penkova M, Metzler-Nolte N, Bandow JE et al. Corynebacterium glutamicum exhibits a membrane-related response to a small ferrocene-conjugated antimicrobial peptide. J Biol Inorg Chem 2010; 15:1293–1303 [View Article] [PubMed]
    [Google Scholar]
  26. Gümüş D, Kalaycı-Yüksek F, Yörük E, Uz G, Çelik E et al. Alterations of growth rate and gene expression levels of UPEC by antibiotics at sub-MIC. Folia Microbiol 2018; 63:451–457 [View Article] [PubMed]
    [Google Scholar]
  27. Westhoff S, van Leeuwe TM, Qachach O, Zhang Z, van Wezel GP et al. The evolution of no-cost resistance at sub-MIC concentrations of streptomycin in Streptomyces coelicolor . ISME J 2017; 11:1168–1178 [View Article] [PubMed]
    [Google Scholar]
  28. Braga PC, Piatti G. Favourable effects of sub-MIC rufloxacin concentrations in decreasing the pathogen-host cell adhesion. Pharmacol Res 1993; 28:11–19 [View Article] [PubMed]
    [Google Scholar]
  29. Pandit S, Chang KW, Jeon JG. Effects of Withania somnifera on the growth and virulence properties of Streptococcus mutans and Streptococcus sobrinus at sub-MIC levels. Anaerobe 2013; 19:1–8 [View Article]
    [Google Scholar]
  30. Kato C, Nakano Y, Lis M, Kuramitsu HK. Molecular genetic analysis of the catalytic site of Streptococcus mutans glucosyltransferases. Biochem Biophys Res Commun 1992; 189:1184–1188 [View Article] [PubMed]
    [Google Scholar]
  31. Shan Y, Brown Gandt A, Rowe SE, Deisinger JP, Conlon BP et al. ATP-dependent persister formation in Escherichia coli . mBio 2017; 8:e02267–e02316 [View Article] [PubMed]
    [Google Scholar]
  32. Tiwari S, Jamal SB, Hassan SS, Carvalho PVSD, Almeida S et al. Two-component signal transduction systems of pathogenic bacteria as targets for antimicrobial therapy: an overview. Front Microbiol 2017; 8:1878 [View Article] [PubMed]
    [Google Scholar]
  33. Moye ZD, Zeng L, Burne RA. Fueling the caries process: carbohydrate metabolism and gene regulation by Streptococcus mutans . J Oral Microbiol 2014; 6:24878 [View Article] [PubMed]
    [Google Scholar]
  34. Simmons LA, Foti JJ, Cohen SE, Walker GC. The SOS regulatory network. EcoSal Plus 2008; 2008: [View Article] [PubMed]
    [Google Scholar]
  35. Bar-Nun S, Glickman MH. Proteasomal AAA-ATPases: structure and function. Biochim Biophys Acta 2012; 1823:67–82 [View Article]
    [Google Scholar]
  36. Allison SD, Gartner TB, Mack MC, McGuire K, Treseder K. Nitrogen alters carbon dynamics during early succession in boreal forest. Soil Biol Biochem 2010; 42:1157–1164 [View Article]
    [Google Scholar]
  37. Chróst R. Environmental control of the synthesis and activity of aquatic microbial ectoenzymes. In Chróst RJ. eds Microbial Enzymes in Aquatic Environments NY: Springer; 1991 pp 29–59
    [Google Scholar]
  38. Landi L, Renella G, Giagnoni L, Nannipieri P. Activities of Proteolytic Enzymes. In Methods of Soil Enzymology vol 247 John Wiley & Sons Ltd; 2011 p 60
    [Google Scholar]
  39. Vermassen A, Leroy S, Talon R, Provot C, Popowska M et al. Cell wall hydrolases in bacteria: insight on the diversity of cell wall amidases, glycosidases and peptidases toward peptidoglycan. Front Microbiol 2019; 10:331 [View Article] [PubMed]
    [Google Scholar]
  40. Chaffin DO, Taylor D, Skerrett SJ, Rubens CE. Changes in the Staphylococcus aureus transcriptome during early adaptation to the lung. PLoS One 2012; 7:e41329 [View Article] [PubMed]
    [Google Scholar]
  41. Pearson JP, Van Delden C, Iglewski BH. Active efflux and diffusion are involved in transport of Pseudomonas aeruginosa cell-to-cell signals. J Bacteriol 1999; 181:1203–1210 [View Article] [PubMed]
    [Google Scholar]
  42. Conlon BP, Rowe SE, Gandt AB, Nuxoll AS, Donegan NP et al. Persister formation in Staphylococcus aureus is associated with ATP depletion. Nat Microbiol 2016; 1:1–7 [View Article] [PubMed]
    [Google Scholar]
  43. Wishart DS, Knox C, Guo AC, Cheng D, Shrivastava S et al. DrugBank: a knowledgebase for drugs, drug actions and drug targets. Nucleic Acids Res 2008; 36:D901–6 [View Article] [PubMed]
    [Google Scholar]
  44. Mishra NM, Briers Y, Lamberigts C, Steenackers H, Robijns S et al. Evaluation of the antibacterial and antibiofilm activities of novel CRAMP-vancomycin conjugates with diverse linkers. Org Biomol Chem 2015; 13:7477–7486 [View Article] [PubMed]
    [Google Scholar]
  45. Ribeiro SM, de la Fuente-Núñez C, Baquir B, Faria-Junior C, Franco OL et al. Antibiofilm peptides increase the susceptibility of carbapenemase-producing Klebsiella pneumoniae clinical isolates to β-lactam antibiotics. Antimicrob Agents Chemother 2015; 59:3906–3912 [View Article]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/jmm/10.1099/jmm.0.001572
Loading
/content/journal/jmm/10.1099/jmm.0.001572
Loading

Data & Media loading...

Supplements

Supplementary material 1

PDF
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