1887

Abstract

The control of bacterial contaminants on meat is a key area of interest in the food industry. Bacteria are exposed to a variety of stresses during broiler processing which challenge bacterial structures and metabolic pathways causing death or sublethal injury. To counter these stresses, bacteria possess robust response systems that can induce shifts in the transcriptome and proteome to enable survival. Effective adaptive responses, such as biofilm formation, shock protein production and metabolic flexibility, require rapid induction and implementation at a cellular and community level to facilitate bacterial survival in adverse conditions. This review aims to provide an overview of the scientific literature pertaining to the regulation of complex adaptive processes used by bacteria to survive the processing environment, with particular focus on species that impact the quality and safety of poultry products like spp., and spp.

Funding
This study was supported by the:
  • Department of Agriculture, Food and the Marine (Award 17/F/275)
    • Principle Award Recipient: AmaliaG. M. Scannell
  • 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.0.001184
2022-04-22
2024-04-26
Loading full text...

Full text loading...

/deliver/fulltext/micro/168/4/mic001184.html?itemId=/content/journal/micro/10.1099/mic.0.001184&mimeType=html&fmt=ahah

References

  1. Schmid B, Klumpp J, Raimann E, Loessner MJ, Stephan R et al. Role of cold shock proteins in growth of Listeria monocytogenes under cold and osmotic stress conditions. Appl Environ Microbiol 2009; 75:1621–1627 [View Article] [PubMed]
    [Google Scholar]
  2. Lamas A, Regal P, Vázquez B, Miranda JM, Cepeda A et al. Salmonella and Campylobacter biofilm formation: a comparative assessment from farm to fork. J Sci Food Agric 2018; 98:4014–4032 [View Article] [PubMed]
    [Google Scholar]
  3. Meunier M, Guyard-Nicodème M, Dory D, Chemaly M. Control strategies against Campylobacter at the poultry production level: biosecurity measures, feed additives and vaccination. J Appl Microbiol 2016; 120:1139–1173 [View Article] [PubMed]
    [Google Scholar]
  4. Loretz M, Stephan R, Zweifel C. Antibacterial activity of decontamination treatments for pig carcasses. Food Control 2011; 22:1121–1125 [View Article]
    [Google Scholar]
  5. del Río E, Panizo-Morán M, Prieto M, Alonso-Calleja C, Capita R. Effect of various chemical decontamination treatments on natural microflora and sensory characteristics of poultry. Int J Food Microbiol 2007; 115:268–280 [View Article] [PubMed]
    [Google Scholar]
  6. Loretz M, Stephan R, Zweifel C. Antimicrobial activity of decontamination treatments for poultry carcasses: A literature survey. Food Control 2010; 21:791–804 [View Article]
    [Google Scholar]
  7. Lu T, Marmion M, Ferone M, Wall P, Scannell AGM. Processing and retail strategies to minimize Campylobacter contamination in retail chicken. J Food Process Preserv 2019; 43: [View Article]
    [Google Scholar]
  8. Buess S, Zurfluh K, Stephan R, Guldimann C. Quantitative microbiological slaughter process analysis in a large-scale Swiss poultry abattoir. Food Control 2019; 105:86–93 [View Article]
    [Google Scholar]
  9. Merino L, Procura F, Trejo FM, Bueno DJ, Golowczyc MA. Biofilm formation by Salmonella sp. in the poultry industry: Detection, control and eradication strategies. Food Res Int 2019; 119:530–540 [View Article] [PubMed]
    [Google Scholar]
  10. European Chemicals Agency. Directive EC 98/8/EC of the European Parliament and of the Council of 16 February 1998 concerning the placing of biocidal products on the market. [Internet]. Vol. 123. European Union; 1998 [cited 2022 Jan 28]. n.d https://echa.europa.eu/guidance-documents/guidance-on-biocides-legislation/biocidal-products-directive
  11. West AM, Teska PJ, Lineback CB, Oliver HF. Strain, disinfectant, concentration, and contact time quantitatively impact disinfectant efficacy. Antimicrob Resist Infect Control 2018; 7:49 [View Article] [PubMed]
    [Google Scholar]
  12. Møretrø T, Vestby LK, Nesse LL, Storheim SE, Kotlarz K et al. Evaluation of efficacy of disinfectants against Salmonella from the feed industry. J Appl Microbiol 2009; 106:1005–1012 [View Article] [PubMed]
    [Google Scholar]
  13. Steenackers H, Hermans K, Vanderleyden J, De Keersmaecker SCJ. Salmonella biofilms: an overview on occurrence, structure, regulation and eradication. Food Research International 2012; 45:502–531 [View Article]
    [Google Scholar]
  14. Lapointe C, Deschênes L, Ells TC, Bisaillon Y, Savard T. Interactions between spoilage bacteria in tri-species biofilms developed under simulated meat processing conditions. Food Microbiol 2019; 82:515–522 [View Article]
    [Google Scholar]
  15. Barria C, Malecki M, Arraiano CM. Bacterial adaptation to cold. Microbiology (Reading) 2013; 159:2437–2443 [View Article] [PubMed]
    [Google Scholar]
  16. Nedwell D. Effect of low temperature on microbial growth: lowered affinity for substrates limits growth at low temperature. FEMS Microbiol Ecol 1999; 30:101–111 [View Article] [PubMed]
    [Google Scholar]
  17. King T, Kocharunchitt C, Gobius K, Bowman JP, Ross T. Physiological response of Escherichia coli O157:H7 Sakai to dynamic changes in temperature and water activity as experienced during carcass chilling. Mol Cell Proteomics 2016; 15:3331–3347 [View Article] [PubMed]
    [Google Scholar]
  18. Ito F, Tamiya T, Ohtsu I, Fujimura M, Fukumori F. Genetic and phenotypic characterization of the heat shock response in Pseudomonas putida. Microbiologyopen 2014; 3:922–936 [View Article] [PubMed]
    [Google Scholar]
  19. Lim B, Miyazaki R, Neher S, Siegele DA, Ito K et al. Heat shock transcription factor σ32 co-opts the signal recognition particle to regulate protein homeostasis in E. coli. PLoS Biol 2013; 11:e1001735 [View Article] [PubMed]
    [Google Scholar]
  20. Irshad A, Asrun TS. Scalding and its significance in livestock slaughter and wholesome meat production. Int J Livest Res 2013; 3:45–53
    [Google Scholar]
  21. Berrang ME, Meinersmann RJ, Adams ES. Water rinse and flowing steam to kill Campylobacter on broiler transport coop flooring. Food Control 2020; 114:107214 [View Article]
    [Google Scholar]
  22. Berrang ME, Hofacre CL, Frank JF. Controlling attachment and growth of Listeria monocytogenes in polyvinyl chloride model floor drains using a peroxide chemical, chitosan-arginine, or heat. J Food Prot 2014; 77:2129–2132 [View Article] [PubMed]
    [Google Scholar]
  23. Schuster CF, Wiedemann DM, Kirsebom FCM, Santiago M, Walker S et al. High-throughput transposon sequencing highlights the cell wall as an important barrier for osmotic stress in methicillin resistant Staphylococcus aureus and underlines a tailored response to different osmotic stressors. Microbiology 2019 [View Article]
    [Google Scholar]
  24. Liao X, Li J, Suo Y, Ahn J, Liu D et al. Effect of preliminary stresses on the resistance of Escherichia coli and Staphylococcus aureus toward non-thermal plasma (NTP) challenge. Food Res Int 2018; 105:178–183 [View Article] [PubMed]
    [Google Scholar]
  25. James C, Vincent C, de Andrade Lima TI, James SJ. The primary chilling of poultry carcasses—a review. International Journal of Refrigeration 2006; 29:847–862 [View Article]
    [Google Scholar]
  26. Marmion M, Macori G, Ferone M, Whyte P, Scannell AGM. Survive and thrive: Control mechanisms that facilitate bacterial adaptation to survive manufacturing-related stress. Int J Food Microbiol 2022; 368:109612 [View Article] [PubMed]
    [Google Scholar]
  27. Carrascosa C, Raheem D, Ramos F, Saraiva A, Raposo A. Microbial biofilms in the food industry-a comprehensive review. Int J Environ Res Public Health 2021; 18:2014 [View Article] [PubMed]
    [Google Scholar]
  28. Yin W, Wang Y, Liu L, He J. Biofilms: the microbial “Protective Clothing” in extreme environments. Int J Mol Sci 2019; 20:E3423 [View Article] [PubMed]
    [Google Scholar]
  29. Ciofu O, Moser C, Jensen , Høiby N. Tolerance and resistance of microbial biofilms. Nat Rev Microbiol 2022 [View Article] [PubMed]
    [Google Scholar]
  30. Wang R. Biofilms and meat safety: a mini-review. J Food Prot 2018; 82:120–127 [View Article]
    [Google Scholar]
  31. Giaouris E, Heir E, Desvaux M, Hébraud M, Møretrø T et al. Intra- and inter-species interactions within biofilms of important foodborne bacterial pathogens. Front Microbiol 2015; 6:841 [View Article] [PubMed]
    [Google Scholar]
  32. Slipski CJ, Zhanel GG, Bay DC. Characterization of proteobacterial plasmid integron-encoded qac efflux pump sequence diversity and quaternary ammonium compound antiseptic selection in Escherichia coli grown planktonically and as biofilms. Antimicrob Agents Chemother 2021; 65:e0106921 [View Article] [PubMed]
    [Google Scholar]
  33. Stalder T, Top E. Plasmid transfer in biofilms: a perspective on limitations and opportunities. NPJ Biofilms Microbiomes 2016; 2:16022 [View Article] [PubMed]
    [Google Scholar]
  34. Tsviklist V, Guest RL, Raivio TL. The Cpx stress response regulates turnover of respiratory chain proteins at the inner membrane of Escherichia coli. Front Microbiol 2021; 12:732288 [View Article] [PubMed]
    [Google Scholar]
  35. Alvarez-Ordóñez A, Broussolle V, Colin P, Nguyen-The C, Prieto M. The adaptive response of bacterial food-borne pathogens in the environment, host and food: Implications for food safety. Int J Food Microbiol 2015; 213:99–109 [View Article] [PubMed]
    [Google Scholar]
  36. Shah J, Desai PT, Chen D, Stevens JR, Weimer BC. Preadaptation to cold stress in Salmonella enterica serovar Typhimurium increases survival during subsequent acid stress exposure. Appl Environ Microbiol 2013; 79:7281–7289 [View Article] [PubMed]
    [Google Scholar]
  37. Jennings MC, Minbiole KPC, Wuest WM. Quaternary ammonium compounds: an antimicrobial mainstay and platform for innovation to address bacterial resistance. ACS Infect Dis 2015; 1:288–303 [View Article] [PubMed]
    [Google Scholar]
  38. Takeuchi K, Imai M, Shimada I. Conformational equilibrium defines the variable induction of the multidrug-binding transcriptional repressor QacR. Proc Natl Acad Sci U S A 2019; 116:19963–19972 [View Article] [PubMed]
    [Google Scholar]
  39. Jennings MC, Forman ME, Duggan SM, Minbiole KPC, Wuest WM. Efflux pumps might not be the major drivers of QAC resistance in methicillin-resistant Staphylococcus aureus. Chembiochem 2017; 18:1573–1577 [View Article] [PubMed]
    [Google Scholar]
  40. Bremer E, Krämer R. Responses of microorganisms to osmotic stress. Annu Rev Microbiol 2019; 73:313–334 [View Article] [PubMed]
    [Google Scholar]
  41. Wall E, Majdalani N, Gottesman S. The complex Rcs regulatory cascade. Annu Rev Microbiol 2018; 72:111–139 [View Article] [PubMed]
    [Google Scholar]
  42. Zhang Y, Burkhardt DH, Rouskin S, Li G-W, Weissman JS et al. A stress response that monitors and regulates mRNA structure is central to cold shock adaptation. Mol Cell 2018; 70:274–286 [View Article] [PubMed]
    [Google Scholar]
  43. Begley M, Hill C. Stress adaptation in foodborne pathogens. Annu Rev Food Sci Technol 2015; 6:191–210 [View Article] [PubMed]
    [Google Scholar]
  44. Delhaye A, Collet JF, Laloux G. A fly on the wall: how stress response systems can sense and respond to damage to peptidoglycan. Front Cell Infect Microbiol 2019; 9:380 [View Article] [PubMed]
    [Google Scholar]
  45. Laubacher ME, Ades SE. The Rcs phosphorelay is a cell envelope stress response activated by peptidoglycan stress and contributes to intrinsic antibiotic resistance. J Bacteriol 2008; 190:2065–2074 [View Article] [PubMed]
    [Google Scholar]
  46. Ranjit DK, Young KD. The Rcs stress response and accessory envelope proteins are required for de novo generation of cell shape in Escherichia coli. J Bacteriol 2013; 195:2452–2462 [View Article] [PubMed]
    [Google Scholar]
  47. Howery KE, Clemmer KM, Rather PN. The Rcs regulon in Proteus mirabilis: implications for motility, biofilm formation, and virulence. Curr Genet 2016; 62:775–789 [View Article] [PubMed]
    [Google Scholar]
  48. Majdalani N, Hernandez D, Gottesman S. Regulation and mode of action of the second small RNA activator of RpoS translation, RprA. Mol Microbiol 2002; 46:813–826 [View Article] [PubMed]
    [Google Scholar]
  49. Davis MC, Kesthely CA, Franklin EA, MacLellan SR. The essential activities of the bacterial sigma factor. Can J Microbiol 2017; 63:89–99 [View Article] [PubMed]
    [Google Scholar]
  50. Wang H, Dong Y, Wang G, Xu X, Zhou G. Effect of growth media on gene expression levels in Salmonella Typhimurium biofilm formed on stainless steel surface. Food Control 2016; 59:546–552 [View Article]
    [Google Scholar]
  51. Franchini AG, Ihssen J, Egli T. Effect of Global Regulators RpoS and Cyclic-AMP/CRP on the Catabolome and Transcriptome of Escherichia coli K12 during Carbon- and Energy-Limited Growth. PLoS One 2015; 10:e0133793 [View Article] [PubMed]
    [Google Scholar]
  52. Horn N, Bhunia AK. Food-associated stress primes foodborne pathogens for the gastrointestinal phase of infection. Front Microbiol 2018; 9:1962 [View Article] [PubMed]
    [Google Scholar]
  53. Donlan RM. Biofilms: microbial life on surfaces. Emerg Infect Dis 2002; 8:881–890 [View Article] [PubMed]
    [Google Scholar]
  54. Spöring I, Felgner S, Preuße M, Eckweiler D, Rohde M et al. Regulation of flagellum biosynthesis in response to cell envelope stress in Salmonella enterica Serovar Typhimurium. mBio 2018; 9:e00736-17 [View Article] [PubMed]
    [Google Scholar]
  55. Ferenci T, Galbiati HF, Betteridge T, Phan K, Spira B. The constancy of global regulation across a species: the concentrations of ppGpp and RpoS are strain-specific in Escherichia coli. BMC Microbiol 2011; 11:62 [View Article] [PubMed]
    [Google Scholar]
  56. Hecker M, Pané-Farré J, Völker U. SigB-dependent general stress response in Bacillus subtilis and related gram-positive bacteria. Annu Rev Microbiol 2007; 61:215–236 [View Article] [PubMed]
    [Google Scholar]
  57. Zetzmann M, Bucur FI, Crauwels P, Borda D, Nicolau AI et al. Characterization of the biofilm phenotype of a Listeria monocytogenes mutant deficient in agr peptide sensing. Microbiologyopen 2019; 8:e00826 [View Article] [PubMed]
    [Google Scholar]
  58. Reder A, Höper D, Gerth U, Hecker M. Contributions of individual σB-dependent general stress genes to oxidative stress resistance of Bacillus subtilis. J Bacteriol 2012; 194:3601–3610 [View Article] [PubMed]
    [Google Scholar]
  59. Tran V, Geraci K, Midili G, Satterwhite W, Wright R et al. Resilience to oxidative and nitrosative stress is mediated by the stressosome, RsbP and SigB in Bacillus subtilis. J Basic Microbiol 2019; 59:834–845 [View Article] [PubMed]
    [Google Scholar]
  60. Bartolini M, Cogliati S, Vileta D, Bauman C, Ramirez W et al. Stress-responsive alternative sigma factor SigB plays a positive role in the antifungal proficiency of Bacillus subtilis. Appl Environ Microbiol 2019; 85:e00178-19 [View Article] [PubMed]
    [Google Scholar]
  61. Rodriguez Ayala F, Bartolini M, Grau R. The stress-responsive alternative sigma factor SigB of Bacillus subtilis and its relatives: an old friend with new functions. Front Microbiol 2020; 11:1761 [View Article] [PubMed]
    [Google Scholar]
  62. Xue Y, Osborn J, Panchal A, Mellies JL. The RpoE stress response pathway mediates reduction of the virulence of enteropathogenic Escherichia coli by zinc. Appl Environ Microbiol 2015; 81:3766–3774 [View Article] [PubMed]
    [Google Scholar]
  63. Missiakas D, Mayer MP, Lemaire M, Georgopoulos C, Raina S. Modulation of the Escherichia coli sigmaE (RpoE) heat-shock transcription-factor activity by the RseA, RseB and RseC proteins. Mol Microbiol 1997; 24:355–371 [View Article]
    [Google Scholar]
  64. Li Q, Peng T, Klug G. The PhyR homolog RSP_1274 of Rhodobacter sphaeroides is involved in defense of membrane stress and has a moderate effect on RpoE (RSP_1092) activity. BMC Microbiol 2018; 18: [View Article] [PubMed]
    [Google Scholar]
  65. Miladi H, Elabed H, Ben Slama R, Rhim A, Bakhrouf A. Molecular analysis of the role of osmolyte transporters opuCA and betL in Listeria monocytogenes after cold and freezing stress. Arch Microbiol 2017; 199:259–265 [View Article] [PubMed]
    [Google Scholar]
  66. Gu D, Zhang J, Hao Y, Xu R, Zhang Y et al. Alternative sigma factor RpoX is a part of the RpoE regulon and plays distinct roles in stress responses, motility, biofilm formation, and hemolytic activities in the marine pathogen Vibrio alginolyticus. Appl Environ Microbiol 2019; 85: [View Article] [PubMed]
    [Google Scholar]
  67. Ghazaei C. Role and mechanism of the Hsp70 molecular chaperone machines in bacterial pathogens. J Med Microbiol 2017; 66:259–265 [View Article] [PubMed]
    [Google Scholar]
  68. Usaga J, Worobo RW, Padilla-Zakour OI. Effect of acid adaptation and acid shock on thermal tolerance and survival of Escherichia coli O157:H7 and O111 in apple juice. J Food Prot 2014; 77:1656–1663 [View Article] [PubMed]
    [Google Scholar]
  69. Fahmi T, Port GC, Cho KH. c-di-AMP: an essential molecule in the signaling pathways that regulate the viability and virulence of gram-positive bacteria. Genes (Basel) 2017; 8:197 [View Article] [PubMed]
    [Google Scholar]
  70. Wirebrand L, Österberg S, López-Sánchez A, Govantes F, Shingler V. PP4397/FlgZ provides the link between PP2258 c-di-GMP signalling and altered motility in Pseudomonas putida. Sci Rep 2018; 8:12205 [View Article] [PubMed]
    [Google Scholar]
  71. Pultz IS, Christen M, Kulasekara HD, Kennard A, Kulasekara B et al. The response threshold of Salmonella PilZ domain proteins is determined by their binding affinities for c-di-GMP. Mol Microbiol 2012; 86:1424–1440 [View Article] [PubMed]
    [Google Scholar]
  72. Kuan N-L, Yeh K-S. Arginine within a specific motif near the N-terminal of FimY is critical for the maximal production of type 1 fimbriae in Salmonella enterica serovar Typhimurium. Microbiologyopen 2019; 8:e00846 [View Article] [PubMed]
    [Google Scholar]
  73. Dieltjens L, Appermans K, Lissens M, Lories B, Kim W et al. Inhibiting bacterial cooperation is an evolutionarily robust anti-biofilm strategy. Nat Commun 2020; 11:107 [View Article] [PubMed]
    [Google Scholar]
  74. He J, Yin W, Galperin MY, Chou S-H. Cyclic di-AMP, a second messenger of primary importance: tertiary structures and binding mechanisms. Nucleic Acids Res 2020; 48:2807–2829 [View Article] [PubMed]
    [Google Scholar]
  75. Commichau FM, Heidemann JL, Ficner R, Stülke J. Making and breaking of an essential poison: the cyclases and phosphodiesterases that produce and degrade the essential second messenger cyclic di-AMP in bacteria. J Bacteriol 2019; 201:e00462-18 [View Article] [PubMed]
    [Google Scholar]
  76. Poole K. Bacterial stress responses as determinants of antimicrobial resistance. J Antimicrob Chemother 2012; 67:2069–2089 [View Article] [PubMed]
    [Google Scholar]
  77. Kanjee U, Ogata K, Houry WA. Direct binding targets of the stringent response alarmone (p)ppGpp. Mol Microbiol 2012; 85:1029–1043 [View Article] [PubMed]
    [Google Scholar]
  78. Hughes D, Andersson DI. Environmental and genetic modulation of the phenotypic expression of antibiotic resistance. FEMS Microbiol Rev 2017; 41:374–391 [View Article] [PubMed]
    [Google Scholar]
  79. Ritzert JT, Minasov G, Embry R, Schipma MJ, Satchell KJF. The cyclic AMP receptor protein regulates quorum sensing and global gene expression in Yersinia pestis during planktonic growth and growth in biofilms. mBio 2019; 10:e02613-19 [View Article] [PubMed]
    [Google Scholar]
  80. Ono K, Oka R, Toyofuku M, Sakaguchi A, Hamada M et al. CAMP signaling affects irreversible attachment during biofilm formation by Pseudomonas aeruginosa PAO1. Microbes Environ 2014; 29:104–106 [View Article] [PubMed]
    [Google Scholar]
  81. Semsey S, Jauffred L, Csiszovszki Z, Erdossy J, Stéger V et al. The effect of LacI autoregulation on the performance of the lactose utilization system in Escherichia coli. Nucleic Acids Res 2013; 41:6381–6390 [View Article] [PubMed]
    [Google Scholar]
  82. Zhao P, Wang W, Tian P. Development of cyclic AMP receptor protein-based artificial transcription factor for intensifying gene expression. Appl Microbiol Biotechnol 2018; 102:1673–1685 [View Article] [PubMed]
    [Google Scholar]
  83. Huynh TT, McDougald D, Klebensberger J, Al Qarni B, Barraud N et al. Glucose starvation-induced dispersal of Pseudomonas aeruginosa biofilms is cAMP and energy dependent. PLoS One 2012; 7:e42874 [View Article] [PubMed]
    [Google Scholar]
  84. Patiño-Ruiz M, Ganea C, Fendler K, Călinescu O. Competition is the basis of the transport mechanism of the NhaB Na+/H+ exchanger from Klebsiella pneumoniae. PLoS One 2017; 12:e0182293 [View Article]
    [Google Scholar]
  85. Padan E, Danieli T, Keren Y, Alkoby D, Masrati G et al. NhaA antiporter functions using 10 helices, and an additional 2 contribute to assembly/stability. Proc Natl Acad Sci U S A 2015; 112:E5575–82 [View Article] [PubMed]
    [Google Scholar]
  86. Toyofuku M, Inaba T, Kiyokawa T, Obana N, Yawata Y et al. Environmental factors that shape biofilm formation. Biosci Biotechnol Biochem 2016; 80:7–12 [View Article] [PubMed]
    [Google Scholar]
  87. Solano C, Echeverz M, Lasa I. Biofilm dispersion and quorum sensing. Curr Opin Microbiol 2014; 18:96–104 [View Article] [PubMed]
    [Google Scholar]
  88. Jesudhasan PR, Cepeda ML, Widmer K, Dowd SE, Soni KA et al. Transcriptome analysis of genes controlled by luxS/autoinducer-2 in Salmonella enterica serovar Typhimurium. Foodborne Pathog Dis 2010; 7:399–410 [View Article] [PubMed]
    [Google Scholar]
  89. Whiteley M, Diggle SP, Greenberg EP. Progress in and promise of bacterial quorum sensing research. Nature 2017; 551:313–320 [View Article] [PubMed]
    [Google Scholar]
  90. Melo RT, Mendonça EP, Monteiro GP, Siqueira MC, Pereira CB et al. Intrinsic and extrinsic aspects on Campylobacter jejuni biofilms. Front Microbiol 2017; 8:1332 [View Article] [PubMed]
    [Google Scholar]
  91. Liao L, Schaefer AL, Coutinho BG, Brown PJB, Greenberg EP. An aryl-homoserine lactone quorum-sensing signal produced by a dimorphic prosthecate bacterium. Proc Natl Acad Sci U S A 2018; 115:7587–7592 [View Article] [PubMed]
    [Google Scholar]
  92. Smith JN, Ahmer BMM. Detection of other microbial species by Salmonella: expression of the SdiA regulon. J Bacteriol 2003; 185:1357–1366 [View Article] [PubMed]
    [Google Scholar]
  93. Elvers KT, Park SF. Quorum sensing in Campylobacter jejuni: detection of a luxS encoded signalling molecule. Microbiology (Reading) 2002; 148:1475–1481 [View Article] [PubMed]
    [Google Scholar]
  94. Mukherjee S, Bassler BL. Bacterial quorum sensing in complex and dynamically changing environments. Nat Rev Microbiol 2019; 17:371–382 [View Article] [PubMed]
    [Google Scholar]
  95. Teh AHT, Lee SM, Dykes GA. Does Campylobacter jejuni form biofilms in food-related environments?. Appl Environ Microbiol 2014; 80:5154–5160 [View Article] [PubMed]
    [Google Scholar]
  96. Lu J, Jin M, Nguyen SH, Mao L, Li J et al. Non-antibiotic antimicrobial triclosan induces multiple antibiotic resistance through genetic mutation. Environ Int 2018; 118:257–265 [View Article] [PubMed]
    [Google Scholar]
  97. Wouters JA, Rombouts FM, Kuipers OP, de Vos WM, Abee T. The role of cold-shock proteins in low-temperature adaptation of food-related bacteria. Syst Appl Microbiol 2000; 23:165–173 [View Article] [PubMed]
    [Google Scholar]
  98. Alreshidi MM, Dunstan RH, Macdonald MM, Gottfries J et al. Metabolomic and proteomic responses of Staphylococcus aureus to prolonged cold stress. J Proteomics 2015; 121:44–55 [View Article] [PubMed]
    [Google Scholar]
  99. Qu A, Brulc JM, Wilson MK, Law BF, Theoret JR et al. Comparative metagenomics reveals host specific metavirulomes and horizontal gene transfer elements in the chicken cecum microbiome. PLoS One 2008; 3:e2945 [View Article] [PubMed]
    [Google Scholar]
  100. Apata DF. Antibiotic resistance in poultry. International J of Poultry Science 2009; 8:404–408 [View Article]
    [Google Scholar]
  101. Granstad S, Kristoffersen AB, Benestad SL, Sjurseth SK, David B et al. Effect of feed additives as alternatives to in-feed antimicrobials on production performance and intestinal Clostridium perfringens counts in broiler chickens. Animals (Basel) 2020; 10:240 [View Article] [PubMed]
    [Google Scholar]
  102. Madec JY, Haenni M. Antimicrobial resistance plasmid reservoir in food and food-producing animals. Plasmid 2018; 99:72–81 [View Article] [PubMed]
    [Google Scholar]
  103. Lewis K. Platforms for antibiotic discovery. Nat Rev Drug Discov 2013; 12:371–387 [View Article] [PubMed]
    [Google Scholar]
  104. Murphy CP, Carson C, Smith BA, Chapman B, Marrotte J et al. Factors potentially linked with the occurrence of antimicrobial resistance in selected bacteria from cattle, chickens and pigs: A scoping review of publications for use in modelling of antimicrobial resistance (IAM.AMR Project). Zoonoses Public Health 2018; 65:957–971 [View Article] [PubMed]
    [Google Scholar]
  105. García V, García P, Rodríguez I, Rodicio R, Rodicio MR. The role of IS26 in evolution of a derivative of the virulence plasmid of Salmonella enterica serovar Enteritidis which confers multiple drug resistance. Infect Genet Evol 2016; 45:246–249 [View Article] [PubMed]
    [Google Scholar]
  106. Jensen SO, Apisiridej S, Kwong SM, Yang YH, Skurray RA et al. Analysis of the prototypical Staphylococcus aureus multiresistance plasmid pSK1. Plasmid 2010; 64:135–142 [View Article] [PubMed]
    [Google Scholar]
  107. McMahon MAS, Xu J, Moore JE, Blair IS, McDowell DA. Environmental stress and antibiotic resistance in food-related pathogens. Appl Environ Microbiol 2007; 73:211–217 [View Article] [PubMed]
    [Google Scholar]
  108. Simpson AE, Skurray RA, Firth N. A single gene on the staphylococcal multiresistance plasmid pSK1 encodes A novel partitioning system. J Bacteriol 2003; 185:2143–2152 [View Article] [PubMed]
    [Google Scholar]
  109. Ranjana KC, Shrestha U, Kumar S, Ranaweera I, Kakarla P et al. Molecular biology of multidrug resistance efflux pumps of the major facilitator superfamily from bacterial food pathogens. Foodborne Pathog Antibiot Resist 2017
    [Google Scholar]
  110. Seasotiya L, Siwach P, Bharti P, Bai S, Malik A et al. A cross sectional study on prevalence of antibiotic resistance and role of efflux pumps in fluoroquinolone resistance by using efflux pump inhibitors in isolated cultures from poultry, dairy farms and MTCC strains from reservoirs. BMRJ 2015; 5:107–116 [View Article] [PubMed]
    [Google Scholar]
  111. Abraham A, Ifeanyi SS, Muinah F, Ibidunni Oreoluwa BS, Coulibaly KJ et al. Plasmid profile and role in virulence of Salmonella enterica serovars isolated from food animals and humans in Lagos Nigeria. Pathog Glob Health 2019; 113:282–287 [View Article] [PubMed]
    [Google Scholar]
  112. Hong SI, Lee Y-M, Park K-H, Ryu B-H, Hong K-W et al. Clinical and molecular characteristics of qacA- and qacB-positive methicillin-resistant Staphylococcus aureus causing bloodstream infections. Antimicrob Agents Chemother 2019; 63:e02157-18 [View Article]
    [Google Scholar]
  113. Seribelli AA, Cruz MF, Vilela FP, Frazão MR, Paziani MH et al. Phenotypic and genotypic characterization of Salmonella Typhimurium isolates from humans and foods in Brazil. PLoS One 2020; 15:e0237886 [View Article] [PubMed]
    [Google Scholar]
  114. Salvail H, Groisman EA. The phosphorelay BarA/SirA activates the non-cognate regulator RcsB in Salmonella enterica. PLoS Genet 2020; 16:e1008722 [View Article] [PubMed]
    [Google Scholar]
  115. Martínez LC, Yakhnin H, Camacho MI, Georgellis D, Babitzke P et al. Integration of a complex regulatory cascade involving the SirA/BarA and Csr global regulatory systems that controls expression of the Salmonella SPI-1 and SPI-2 virulence regulons through HilD. Mol Microbiol 2011; 80:1637–1656 [View Article] [PubMed]
    [Google Scholar]
  116. Baxter MA, Jones BD. Two-component regulators control hilA expression by controlling fimZ and hilE expression within Salmonella enterica serovar Typhimurium. Infect Immun 2015; 83:978–985 [View Article] [PubMed]
    [Google Scholar]
  117. de Jong HK, Parry CM, van der Poll T, Wiersinga WJ. Host-pathogen interaction in invasive Salmonellosis. PLoS Pathog 2012; 8:e1002933 [View Article] [PubMed]
    [Google Scholar]
  118. Banda MM, Zavala-Alvarado C, Pérez-Morales D, Bustamante VH. SlyA and HilD counteract H-NS-mediated repression on the ssrAB virulence operon of Salmonella enterica Serovar Typhimurium and thus promote its activation by OmpR. J Bacteriol 2019; 201:e00530-18 [View Article] [PubMed]
    [Google Scholar]
  119. Hueffer K, Galán JE. Salmonella-induced macrophage death: multiple mechanisms, different outcomes. Cell Microbiol 2004; 6:1019–1025 [View Article] [PubMed]
    [Google Scholar]
  120. Mangat CS, Bekal S, Irwin RJ, Mulvey MR. A novel hybrid plasmid carrying multiple antimicrobial resistance and virulence genes in Salmonella enterica serovar dublin. Antimicrob Agents Chemother 2017; 61:e02601-16 [View Article] [PubMed]
    [Google Scholar]
  121. Kumar H, Franzetti L, Kaushal A, Kumar D. Pseudomonas fluorescens: a potential food spoiler and challenges and advances in its detection. Ann Microbiol 2019; 69:873–883 [View Article]
    [Google Scholar]
  122. Kalia VC. Quorum sensing inhibitors: an overview. Biotechnol Adv 2013; 31:224–245 [View Article] [PubMed]
    [Google Scholar]
  123. Norizan SNM, Yin W-F, Chan K-G. Caffeine as a potential quorum sensing inhibitor. Sensors (Basel) 2013; 13:5117–5129 [View Article] [PubMed]
    [Google Scholar]
  124. Balaban N, Cirioni O, Giacometti A, Ghiselli R, Braunstein JB et al. Treatment of Staphylococcus aureus biofilm infection by the quorum-sensing inhibitor RIP. Antimicrob Agents Chemother 2007; 51:2226–2229 [View Article] [PubMed]
    [Google Scholar]
  125. Kalia VC, Patel SKS, Kang YC, Lee JK. Quorum sensing inhibitors as antipathogens: biotechnological applications. Biotechnol Adv 2019; 37:68–90 [View Article] [PubMed]
    [Google Scholar]
  126. O’Loughlin CT, Miller LC, Siryaporn A, Drescher K, Semmelhack MF et al. A quorum-sensing inhibitor blocks Pseudomonas aeruginosa virulence and biofilm formation. Proc Natl Acad Sci U S A 2013; 110:17981–17986 [View Article] [PubMed]
    [Google Scholar]
  127. Nahar S, Jeong HL, Kim Y, Ha AJ-W, Roy PK et al. Inhibitory effects of Flavourzyme on biofilm formation, quorum sensing, and virulence genes of foodborne pathogens Salmonella Typhimurium and Escherichia coli. Food Res Int 2021; 147:110461 [View Article] [PubMed]
    [Google Scholar]
  128. Gunaratnam S, Millette M, McFarland LV, DuPont HL, Lacroix M. Potential role of probiotics in reducing Clostridioides difficile virulence: Interference with quorum sensing systems. Microb Pathog 2021; 153:104798 [View Article] [PubMed]
    [Google Scholar]
  129. Velikova N, Fulle S, Manso AS, Mechkarska M, Finn P et al. Putative histidine kinase inhibitors with antibacterial effect against multi-drug resistant clinical isolates identified by in vitro and in silico screens. Sci Rep 2016; 6:26085 [View Article] [PubMed]
    [Google Scholar]
  130. Koolman L, Whyte P, Burgess C, Bolton D. Virulence gene expression, adhesion and invasion of Campylobacter jejuni exposed to oxidative stress (H2O2). Int J Food Microbiol 2016; 220:33–38 [View Article] [PubMed]
    [Google Scholar]
  131. Koolman L, Whyte P, Meade J, Lyng J, Bolton D. Use of chemical treatments applied alone and in combination to reduce Campylobacter on raw poultry. Food Control 2014; 46:299–303 [View Article]
    [Google Scholar]
  132. Dann AB, Hontela A. Triclosan: environmental exposure, toxicity and mechanisms of action. J Appl Toxicol 2011; 31:285–311 [View Article] [PubMed]
    [Google Scholar]
  133. Pedrós-Garrido S, Clemente I, Calanche JB, Condón-Abanto S, Beltrán JA et al. Antimicrobial activity of natural compounds against listeria spp. and their effects on sensory attributes in salmon (Salmo salar) and cod (Gadus morhua). Food Control 2020; 107:106768 [View Article]
    [Google Scholar]
  134. Biswas D, Tiwari M, Tiwari V. Molecular mechanism of antimicrobial activity of chlorhexidine against carbapenem-resistant Acinetobacter baumannii. PLoS One 2019; 14:e0224107 [View Article] [PubMed]
    [Google Scholar]
  135. Phongphakdee K, Nitisinprasert S. Combination inhibition activity of nisin and ethanol on the growth inhibition of pathogenic gram negative bacteria and their application as disinfectant solution. J Food Sci 2015; 80:M2241–6 [View Article] [PubMed]
    [Google Scholar]
  136. Flores MJ, Lescano MR, Brandi RJ, Cassano AE, Labas MD. A novel approach to explain the inactivation mechanism of Escherichia coli employing A commercially available peracetic acid. Water Sci Technol 2014; 69:358–363 [View Article] [PubMed]
    [Google Scholar]
  137. Flores MJ, Brandi RJ, Cassano AE, Labas MD. Kinetic model of water disinfection using peracetic acid including synergistic effects. Water Sci Technol 2016; 73:275–282 [View Article] [PubMed]
    [Google Scholar]
  138. Suzuki S, Horinouchi T, Furusawa C. Prediction of antibiotic resistance by gene expression profiles. Nat Commun 2014; 5:5792 [View Article] [PubMed]
    [Google Scholar]
  139. Barroso R, García-Mauriño SM, Tomás-Gallardo L, Andújar E, Pérez-Alegre M et al. The CbrB Regulon: Promoter dissection reveals novel insights into the CbrAB expression network in Pseudomonas putida. PLoS One 2018; 13:e0209191 [View Article] [PubMed]
    [Google Scholar]
  140. Teplitski M, Goodier RI, Ahmer BMM. Catabolite repression of the SirA regulatory cascade in Salmonella enterica. Int J Med Microbiol 2006; 296:449–466 [View Article] [PubMed]
    [Google Scholar]
  141. Teplitski M, Al-Agely A, Ahmer BMM. Contribution of the SirA regulon to biofilm formation in Salmonella enterica serovar Typhimurium. Microbiology (Reading) 2006; 152:3411–3424 [View Article] [PubMed]
    [Google Scholar]
  142. Leblanc SKD, Oates CW, Raivio TL. Characterization of the induction and cellular role of the BaeSR two-component envelope stress response of Escherichia coli. J Bacteriol 2011; 193:3367–3375 [View Article] [PubMed]
    [Google Scholar]
  143. Kurabayashi K, Hirakawa Y, Tanimoto K, Tomita H, Hirakawa H. Role of the CpxAR two-component signal transduction system in control of fosfomycin resistance and carbon substrate uptake. J Bacteriol 2014; 196:248–256 [View Article] [PubMed]
    [Google Scholar]
  144. Jerga A, Lu Y-J, Schujman GE, de Mendoza D, Rock CO. Identification of a soluble diacylglycerol kinase required for lipoteichoic acid production in Bacillus subtilis. J Biol Chem 2007; 282:21738–21745 [View Article] [PubMed]
    [Google Scholar]
  145. Newberry KJ, Huffman JL, Miller MC, Vazquez-Laslop N, Neyfakh AA et al. Structures of BmrR-drug complexes reveal a rigid multidrug binding pocket and transcription activation through tyrosine expulsion. J Biol Chem 2008; 283:26795–26804 [View Article] [PubMed]
    [Google Scholar]
  146. Miller JM, Chaudhary H, Marsee JD. Phylogenetic analysis predicts structural divergence for proteobacterial ClpC proteins. J Struct Biol 2018; 201:52–62 [View Article] [PubMed]
    [Google Scholar]
  147. Shen J, Liu Z, Yu H, Ye J, Long Y et al. Systematic stress adaptation of Bacillus subtilis to tetracycline exposure. Ecotoxicol Environ Saf 2020; 188:109910 [View Article] [PubMed]
    [Google Scholar]
  148. Shu J-C, Soo P-C, Chen J-C, Hsu S-H, Chen L-C et al. Differential regulation and activity against oxidative stress of Dps proteins in Bacillus cereus. Int J Med Microbiol 2013; 303:662–673 [View Article] [PubMed]
    [Google Scholar]
  149. Song Y, Nikoloff JM, Zhang D. Improving protein production on the level of regulation of both expression and secretion pathways in Bacillus subtilis. J Microbiol Biotechnol 2015; 25:963–977 [View Article] [PubMed]
    [Google Scholar]
  150. Prajapati B, Bernal-Cabas M, López-Álvarez M, Schaffer M, Bartel J et al. Double trouble: Bacillus depends on a functional Tat machinery to avoid severe oxidative stress and starvation upon entry into a NaCl-depleted environment. Biochim Biophys Acta Mol Cell Res 2021; 1868:118914 [View Article] [PubMed]
    [Google Scholar]
  151. Hecker M, Reder A, Fuchs S, Pagels M, Engelmann S. Physiological proteomics and stress/starvation responses in Bacillus subtilis and Staphylococcus aureus. Res Microbiol 2009; 160:245–258 [View Article] [PubMed]
    [Google Scholar]
  152. Paunkov A, Kupc M, Sóki J, Leitsch D. Characterization of the components of the thioredoxin system in Bacteroides fragilis and evaluation of its activity during oxidative stress. Anaerobe 2022; 73:102507 [View Article] [PubMed]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/micro/10.1099/mic.0.001184
Loading
/content/journal/micro/10.1099/mic.0.001184
Loading

Data & Media loading...

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