1887

Abstract

Systemic release of norepinephrine (NE) is a component of the acute host response to infection, and studies in the field of microbial endocrinology indicate generally that NE increases the bacterial growth rate and promotes invasive disease. However, NE attenuates experimental invasive pneumococcal disease. We determined that NE promoted pneumococcal growth but paradoxically decreased pneumococcal adhesion to host cells. This effect was independent of the classical adhesin CbpA. Microarray analysis indicated that the effect of NE involved two two-component regulatory systems that both regulate expression of the Piu iron uptake ABC transport operon. We propose that NE, a known siderophore, enhances iron availability to the bacteria, resulting in greater bacterial replication and decreased expression of Piu operon products. Downregulation of the operon includes decreased expression of the Piu-associated adhesin PiuD. Our results suggested that the iron-dependent inhibitory effect of NE on pneumococcal adherence is a mechanism underlying the amelioration of pneumococcal disease by NE.

Loading

Article metrics loading...

/content/journal/micro/10.1099/mic.0.065607-0
2013-11-01
2024-03-29
Loading full text...

Full text loading...

/deliver/fulltext/micro/159/11/2333.html?itemId=/content/journal/micro/10.1099/mic.0.065607-0&mimeType=html&fmt=ahah

References

  1. Bansal T., Englert D., Lee J., Hegde M., Wood T. K., Jayaraman A.( 2007). Differential effects of epinephrine, norepinephrine, and indole on Escherichia coli O157 : H7 chemotaxis, colonization, and gene expression. Infect Immun 75:4597–4607 [View Article][PubMed]
    [Google Scholar]
  2. Barnes P. J.( 1990). Neural control of airway function: new perspectives. Mol Aspects Med 11:351–423 [View Article][PubMed]
    [Google Scholar]
  3. Bearson B. L., Bearson S. M. D., Uthe J. J., Dowd S. E., Houghton J. O., Lee I., Toscano M. J., Lay D. C. Jr( 2008). Iron regulated genes of Salmonella enterica serovar Typhimurium in response to norepinephrine and the requirement of FepDGC for norepinephrine-enhanced growth. Microbes Infect 10:807–816 [View Article][PubMed]
    [Google Scholar]
  4. Burton C. L., Chhabra S. R., Swift S., Baldwin T. J., Withers H., Hill S. J., Williams P.( 2002). The growth response of Escherichia coli to neurotransmitters and related catecholamine drugs requires a functional enterobactin biosynthesis and uptake system. Infect Immun 70:5913–5923 [View Article][PubMed]
    [Google Scholar]
  5. Cakan G., Turkoz M., Turan T., Ahmed K., Nagatake T.( 2003). S-carboxymethylcysteine inhibits the attachment of Streptococcus pneumoniae to human pharyngeal epithelial cells. Microb Pathog 34:261–265 [View Article][PubMed]
    [Google Scholar]
  6. Chen C., Brown D. R., Xie Y., Green B. T., Lyte M.( 2003). Catecholamines modulate Escherichia coli O157 : H7 adherence to murine cecal mucosa. Shock 20:183–188 [View Article][PubMed]
    [Google Scholar]
  7. Clarke M. B., Hughes D. T., Zhu C., Boedeker E. C., Sperandio V.( 2006). The QseC sensor kinase: a bacterial adrenergic receptor. Proc Natl Acad Sci U S A 103:10420–10425 [View Article][PubMed]
    [Google Scholar]
  8. Cogan T. A., Thomas A. O., Rees L. E., Taylor A. H., Jepson M. A., Williams P. H., Ketley J., Humphrey T. J.( 2007). Norepinephrine increases the pathogenic potential of Campylobacter jejuni.. Gut 56:1060–1065 [View Article][PubMed]
    [Google Scholar]
  9. De Backer D., Biston P., Devriendt J., Madl C., Chochrad D., Aldecoa C., Brasseur A., Defrance P., Gottignies P., Vincent J.-L.SOAP II Investigators( 2010). Comparison of dopamine and norepinephrine in the treatment of shock. N Engl J Med 362:779–789 [View Article][PubMed]
    [Google Scholar]
  10. Esler M., Jennings G., Korner P., Blombery P., Sacharias N., Leonard P.( 1984). Measurement of total and organ-specific norepinephrine kinetics in humans. Am J Physiol 247:E21–E28[PubMed]
    [Google Scholar]
  11. Freestone P. P., Lyte M.( 2008). Microbial endocrinology: experimental design issues in the study of interkingdom signalling in infectious disease. Adv Appl Microbiol 64:75–105 [View Article][PubMed]
    [Google Scholar]
  12. Freestone P. P., Lyte M., Neal C. P., Maggs A. F., Haigh R. D., Williams P. H.( 2000). The mammalian neuroendocrine hormone norepinephrine supplies iron for bacterial growth in the presence of transferrin or lactoferrin. J Bacteriol 182:6091–6098 [View Article][PubMed]
    [Google Scholar]
  13. Freestone P. P. E., Haigh R. D., Williams P. H., Lyte M.( 2003). Involvement of enterobactin in norepinephrine-mediated iron supply from transferrin to enterohaemorrhagic Escherichia coli.. FEMS Microbiol Lett 222:39–43 [View Article][PubMed]
    [Google Scholar]
  14. Freestone P. P., Haigh R. D., Lyte M.( 2007). Specificity of catecholamine-induced growth in Escherichia coli O157 : H7, Salmonella enterica and Yersinia enterocolitica.. FEMS Microbiol Lett 269:221–228 [View Article][PubMed]
    [Google Scholar]
  15. Freestone P. P., Sandrini S. M., Haigh R. D., Lyte M.( 2008). Microbial endocrinology: how stress influences susceptibility to infection. Trends Microbiol 16:55–64 [View Article][PubMed]
    [Google Scholar]
  16. Gonzales X. F., Deshmukh A., Pulse M., Johnson K., Jones H. P.( 2008). Stress-induced differences in primary and secondary resistance against bacterial sepsis correspond with diverse corticotropin releasing hormone receptor expression by pulmonary CD11c+ MHC II+ and CD11c MHC II+ APCs. Brain Behav Immun 22:552–564 [View Article][PubMed]
    [Google Scholar]
  17. Hegde M., Wood T. K., Jayaraman A.( 2009). The neuroendocrine hormone norepinephrine increases Pseudomonas aeruginosa PA14 virulence through the las quorum-sensing pathway. Appl Microbiol Biotechnol 84:763–776 [View Article][PubMed]
    [Google Scholar]
  18. Hughes D. T., Clarke M. B., Yamamoto K., Rasko D. A., Sperandio V.( 2009). The QseC adrenergic signaling cascade in enterohemorrhagic E. coli (EHEC). PLoS Pathog 5:e1000553 [View Article][PubMed]
    [Google Scholar]
  19. Jomaa M., Yuste J., Paton J. C., Jones C., Dougan G., Brown J. S.( 2005). Antibodies to the iron uptake ABC transporter lipoproteins PiaA and PiuA promote opsonophagocytosis of Streptococcus pneumoniae.. Infect Immun 73:6852–6859 [View Article][PubMed]
    [Google Scholar]
  20. Jordan D.( 2001). Central nervous pathways and control of the airways. Respir Physiol 125:67–81 [View Article][PubMed]
    [Google Scholar]
  21. Kadioglu A., Weiser J. N., Paton J. C., Andrew P. W.( 2008). The role of Streptococcus pneumoniae virulence factors in host respiratory colonization and disease. Nat Rev Microbiol 6:288–301 [View Article][PubMed]
    [Google Scholar]
  22. Kontiokari T., Uhari M., Koskela M.( 1998). Antiadhesive effects of xylitol on otopathogenic bacteria. J Antimicrob Chemother 41:563–565 [View Article][PubMed]
    [Google Scholar]
  23. Kurola P., Tapiainen T., Sevander J., Kaijalainen T., Leinonen M., Uhari M., Saukkoriipi A.( 2011). Effect of xylitol and other carbon sources on Streptococcus pneumoniae biofilm formation and gene expression in vitro.. APMIS 119:135–142 [View Article][PubMed]
    [Google Scholar]
  24. Kvetnansky R.( 2004). Stressor specificity and effect of prior experience on catecholamine biosynthetic enzyme phenylethanolamine N-methyltransferase. Ann N Y Acad Sci 1032:117–129 [View Article][PubMed]
    [Google Scholar]
  25. Lyte M., Erickson A. K., Arulanandam B. P., Frank C. D., Crawford M. A., Francis D. H.( 1997). Norepinephrine-induced expression of the K99 pilus adhesin of enterotoxigenic Escherichia coli.. Biochem Biophys Res Commun 232:682–686 [View Article][PubMed]
    [Google Scholar]
  26. Lyte M., Freestone P. P., Neal C. P., Olson B. A., Haigh R. D., Bayston R., Williams P. H.( 2003). Stimulation of Staphylococcus epidermidis growth and biofilm formation by catecholamine inotropes. Lancet 361:130–135 [View Article][PubMed]
    [Google Scholar]
  27. Mann B., Orihuela C., Antikainen J., Gao G., Sublett J., Korhonen T. K., Tuomanen E.( 2006). Multifunctional role of choline binding protein G in pneumococcal pathogenesis. Infect Immun 74:821–829 [View Article][PubMed]
    [Google Scholar]
  28. Methner U., Rabsch W., Reissbrodt R., Williams P. H.( 2008). Effect of norepinephrine on colonisation and systemic spread of Salmonella enterica in infected animals: role of catecholate siderophore precursors and degradation products. Int J Med Microbiol 298:429–439 [View Article][PubMed]
    [Google Scholar]
  29. Miethke M., Skerra A.( 2010). Neutrophil gelatinase-associated lipocalin expresses antimicrobial activity by interfering with L-norepinephrine-mediated bacterial iron acquisition. Antimicrob Agents Chemother 54:1580–1589 [View Article][PubMed]
    [Google Scholar]
  30. Moreira C. G., Weinshenker D., Sperandio V.( 2010). QseC mediates Salmonella enterica serovar typhimurium virulence in vitro and in vivo.. Infect Immun 78:914–926 [View Article][PubMed]
    [Google Scholar]
  31. Moscoso M., García E., López R.( 2006). Biofilm formation by Streptococcus pneumoniae: role of choline, extracellular DNA, and capsular polysaccharide in microbial accretion. J Bacteriol 188:7785–7795 [View Article][PubMed]
    [Google Scholar]
  32. O’Brien K. L., Wolfson L. J., Watt J. P., Henkle E., Deloria-Knoll M., McCall N., Lee E., Mulholland K., Levine O. S., Cherian T.Hib and Pneumococcal Global Burden of Disease Study Team( 2009). Burden of disease caused by Streptococcus pneumoniae in children younger than 5 years: global estimates. Lancet 374:893–902 [View Article][PubMed]
    [Google Scholar]
  33. O’Donnell P. M., Aviles H., Lyte M., Sonnenfeld G.( 2006). Enhancement of in vitro growth of pathogenic bacteria by norepinephrine: importance of inoculum density and role of transferrin. Appl Environ Microbiol 72:5097–5099 [View Article][PubMed]
    [Google Scholar]
  34. Orihuela C. J., Gao G., Francis K. P., Yu J., Tuomanen E. I.( 2004). Tissue-specific contributions of pneumococcal virulence factors to pathogenesis. J Infect Dis 190:1661–1669 [View Article][PubMed]
    [Google Scholar]
  35. Paris I., Martinez-Alvarado P., Perez-Pastene C., Vieira M. N., Olea-Azar C., Raisman-Vozari R., Cardenas S., Graumann R., Caviedes P., Segura-Aguilar J.( 2005). Monoamine transporter inhibitors and norepinephrine reduce dopamine-dependent iron toxicity in cells derived from the substantia nigra. J Neurochem 92:1021–1032 [View Article][PubMed]
    [Google Scholar]
  36. Rasko D. A., Moreira C. G., Li R., Reading N. C., Ritchie J. M., Waldor M. K., Williams N., Taussig R., Wei S.& other authors ( 2008). Targeting QseC signaling and virulence for antibiotic development. Science 321:1078–1080 [View Article][PubMed]
    [Google Scholar]
  37. Rosch J. W., Mann B., Thornton J., Sublett J., Tuomanen E.( 2008). Convergence of regulatory networks on the pilus locus of Streptococcus pneumoniae.. Infect Immun 76:3187–3196 [View Article][PubMed]
    [Google Scholar]
  38. Sandrini S. M., Shergill R., Woodward J., Muralikuttan R., Haigh R. D., Lyte M., Freestone P. P.( 2010). Elucidation of the mechanism by which catecholamine stress hormones liberate iron from the innate immune defense proteins transferrin and lactoferrin. J Bacteriol 192:587–594 [View Article][PubMed]
    [Google Scholar]
  39. Scheckelhoff M. R., Telford S. R., Wesley M., Hu L. T.( 2007). Borrelia burgdorferi intercepts host hormonal signals to regulate expression of outer surface protein A. Proc Natl Acad Sci U S A 104:7247–7252 [View Article][PubMed]
    [Google Scholar]
  40. Schreurs A. J., Nijkamp F. P.( 1982). Haemophilus influenzae induced loss of lung beta-adrenoceptor binding sites and modulation by changes in peripheral catecholaminergic input. Eur J Pharmacol 77:95–102 [View Article][PubMed]
    [Google Scholar]
  41. Schreurs A. J., Versteeg D. H., Nijkamp F. P.( 1982). Involvement of catecholamines in Haemophilus influenzae induced decrease of beta-adrenoceptor function. Naunyn Schmiedebergs Arch Pharmacol 320:235–239 [View Article][PubMed]
    [Google Scholar]
  42. Standish A. J., Stroeher U. H., Paton J. C.( 2007). The pneumococcal two-component signal transduction system RR/HK06 regulates CbpA and PspA by two distinct mechanisms. J Bacteriol 189:5591–5600 [View Article][PubMed]
    [Google Scholar]
  43. Suer E., Sayrac S., Sarinay E., Ozturk H. E., Turkoz M., Ichinose A., Nagatake T., Ahmed K.( 2008). Variation in the attachment of Streptococcus pneumoniae to human pharyngeal epithelial cells after treatment with S-carboxymethylcysteine. J Infect Chemother 14:333–336 [View Article][PubMed]
    [Google Scholar]
  44. Talbot U. M., Paton A. W., Paton J. C.( 1996). Uptake of Streptococcus pneumoniae by respiratory epithelial cells. Infect Immun 64:3772–3777[PubMed]
    [Google Scholar]
  45. Tapiainen T., Sormunen R., Kaijalainen T., Kontiokari T., Ikäheimo I., Uhari M.( 2004). Ultrastructure of Streptococcus pneumoniae after exposure to xylitol. J Antimicrob Chemother 54:225–228 [View Article][PubMed]
    [Google Scholar]
  46. Vlisidou I., Lyte M., van Diemen P. M., Hawes P., Monaghan P., Wallis T. S., Stevens M. P.( 2004). The neuroendocrine stress hormone norepinephrine augments Escherichia coli O157 : H7-induced enteritis and adherence in a bovine ligated ileal loop model of infection. Infect Immun 72:5446–5451 [View Article][PubMed]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/micro/10.1099/mic.0.065607-0
Loading
/content/journal/micro/10.1099/mic.0.065607-0
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