#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Strategies to Avoid Killing by Human Neutrophils


Staphylococcus epidermidis is a leading nosocomial pathogen. In contrast to its more aggressive relative S. aureus, it causes chronic rather than acute infections. In highly virulent S. aureus, phenol-soluble modulins (PSMs) contribute significantly to immune evasion and aggressive virulence by their strong ability to lyse human neutrophils. Members of the PSM family are also produced by S. epidermidis, but their role in immune evasion is not known. Notably, strong cytolytic capacity of S. epidermidis PSMs would be at odds with the notion that S. epidermidis is a less aggressive pathogen than S. aureus, prompting us to examine the biological activities of S. epidermidis PSMs. Surprisingly, we found that S. epidermidis has the capacity to produce PSMδ, a potent leukocyte toxin, representing the first potent cytolysin to be identified in that pathogen. However, production of strongly cytolytic PSMs was low in S. epidermidis, explaining its low cytolytic potency. Interestingly, the different approaches of S. epidermidis and S. aureus to causing human disease are thus reflected by the adaptation of biological activities within one family of virulence determinants, the PSMs. Nevertheless, S. epidermidis has the capacity to evade neutrophil killing, a phenomenon we found is partly mediated by resistance mechanisms to antimicrobial peptides (AMPs), including the protease SepA, which degrades AMPs, and the AMP sensor/resistance regulator, Aps (GraRS). These findings establish a significant function of SepA and Aps in S. epidermidis immune evasion and explain in part why S. epidermidis may evade elimination by innate host defense despite the lack of cytolytic toxin expression. Our study shows that the strategy of S. epidermidis to evade elimination by human neutrophils is characterized by a passive defense approach and provides molecular evidence to support the notion that S. epidermidis is a less aggressive pathogen than S. aureus.


Vyšlo v časopise: Strategies to Avoid Killing by Human Neutrophils. PLoS Pathog 6(10): e32767. doi:10.1371/journal.ppat.1001133
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1001133

Souhrn

Staphylococcus epidermidis is a leading nosocomial pathogen. In contrast to its more aggressive relative S. aureus, it causes chronic rather than acute infections. In highly virulent S. aureus, phenol-soluble modulins (PSMs) contribute significantly to immune evasion and aggressive virulence by their strong ability to lyse human neutrophils. Members of the PSM family are also produced by S. epidermidis, but their role in immune evasion is not known. Notably, strong cytolytic capacity of S. epidermidis PSMs would be at odds with the notion that S. epidermidis is a less aggressive pathogen than S. aureus, prompting us to examine the biological activities of S. epidermidis PSMs. Surprisingly, we found that S. epidermidis has the capacity to produce PSMδ, a potent leukocyte toxin, representing the first potent cytolysin to be identified in that pathogen. However, production of strongly cytolytic PSMs was low in S. epidermidis, explaining its low cytolytic potency. Interestingly, the different approaches of S. epidermidis and S. aureus to causing human disease are thus reflected by the adaptation of biological activities within one family of virulence determinants, the PSMs. Nevertheless, S. epidermidis has the capacity to evade neutrophil killing, a phenomenon we found is partly mediated by resistance mechanisms to antimicrobial peptides (AMPs), including the protease SepA, which degrades AMPs, and the AMP sensor/resistance regulator, Aps (GraRS). These findings establish a significant function of SepA and Aps in S. epidermidis immune evasion and explain in part why S. epidermidis may evade elimination by innate host defense despite the lack of cytolytic toxin expression. Our study shows that the strategy of S. epidermidis to evade elimination by human neutrophils is characterized by a passive defense approach and provides molecular evidence to support the notion that S. epidermidis is a less aggressive pathogen than S. aureus.


Zdroje

1. OttoM

2008 Staphylococcal biofilms. Curr Top Microbiol Immunol 322 207 228

2. OttoM

2009 Staphylococcus epidermidis - the ‘accidental’ pathogen. Nat Rev Microbiol 7 555 567

3. NauseefWM

2007 How human neutrophils kill and degrade microbes: an integrated view. Immunol Rev 219 88 102

4. CostertonJW

StewartPS

GreenbergEP

1999 Bacterial biofilms: a common cause of persistent infections. Science 284 1318 1322

5. KocianovaS

VuongC

YaoY

VoyichJM

FischerER

2005 Key role of poly-gamma-DL-glutamic acid in immune evasion and virulence of Staphylococcus epidermidis. J Clin Invest 115 688 694

6. VuongC

VoyichJM

FischerER

BraughtonKR

WhitneyAR

2004 Polysaccharide intercellular adhesin (PIA) protects Staphylococcus epidermidis against major components of the human innate immune system. Cell Microbiol 6 269 275

7. FosterTJ

2005 Immune evasion by staphylococci. Nat Rev Microbiol 3 948 958

8. RooijakkersSH

van KesselKP

van StrijpJA

2005 Staphylococcal innate immune evasion. Trends Microbiol 13 596 601

9. WoodinA

1970 Staphylococcal leukocidin.

MontjeT

KadisS

AjlS

Microbial toxins New York Academic Press, Inc 327 355

10. WangR

BraughtonKR

KretschmerD

BachTH

QueckSY

2007 Identification of novel cytolytic peptides as key virulence determinants for community-associated MRSA. Nat Med 13 1510 1514

11. McKevittAI

BjornsonGL

MauracherCA

ScheifeleDW

1990 Amino acid sequence of a deltalike toxin from Staphylococcus epidermidis. Infect Immun 58 1473 1475

12. MehlinC

HeadleyCM

KlebanoffSJ

1999 An inflammatory polypeptide complex from Staphylococcus epidermidis: isolation and characterization. J Exp Med 189 907 918

13. YaoY

SturdevantDE

OttoM

2005 Genomewide analysis of gene expression in Staphylococcus epidermidis biofilms: insights into the pathophysiology of S. epidermidis biofilms and the role of phenol-soluble modulins in formation of biofilms. J Infect Dis 191 289 298

14. GillSR

FoutsDE

ArcherGL

MongodinEF

DeboyRT

2005 Insights on evolution of virulence and resistance from the complete genome analysis of an early methicillin-resistant Staphylococcus aureus strain and a biofilm-producing methicillin-resistant Staphylococcus epidermidis strain. J Bacteriol 187 2426 2438

15. HajjarAM

O'MahonyDS

OzinskyA

UnderhillDM

AderemA

2001 Cutting edge: functional interactions between toll-like receptor (TLR) 2 and TLR1 or TLR6 in response to phenol-soluble modulin. J Immunol 166 15 19

16. KlebanoffSJ

KazaziF

Van VoorhisWC

SchlechteKG

1994 Activation of the human immunodeficiency virus long terminal repeat in THP-1 cells by a staphylococcal extracellular product. Proc Natl Acad Sci U S A 91 10615 10619

17. LilesWC

ThomsenAR

O'MahonyDS

KlebanoffSJ

2001 Stimulation of human neutrophils and monocytes by staphylococcal phenol-soluble modulin. J Leukoc Biol 70 96 102

18. HashimotoM

TawaratsumidaK

KariyaH

KiyoharaA

SudaY

2006 Not lipoteichoic acid but lipoproteins appear to be the dominant immunobiologically active compounds in Staphylococcus aureus. J Immunol 177 3162 3169

19. FaurschouM

BorregaardN

2003 Neutrophil granules and secretory vesicles in inflammation. Microbes Infect 5 1317 1327

20. HancockRE

DiamondG

2000 The role of cationic antimicrobial peptides in innate host defences. Trends Microbiol 8 402 410

21. LaiY

VillaruzAE

LiM

ChaDJ

SturdevantDE

2007 The human anionic antimicrobial peptide dermcidin induces proteolytic defence mechanisms in staphylococci. Mol Microbiol 63 497 506

22. LiM

LaiY

VillaruzAE

ChaDJ

SturdevantDE

2007 Gram-positive three-component antimicrobial peptide-sensing system. Proc Natl Acad Sci U S A 104 9469 9474

23. HerbertS

BeraA

NerzC

KrausD

PeschelA

2007 Molecular basis of resistance to muramidase and cationic antimicrobial peptide activity of lysozyme in staphylococci. PLoS Pathog 3 e102

24. LiM

ChaDJ

LaiY

VillaruzAE

SturdevantDE

2007 The antimicrobial peptide-sensing system aps of Staphylococcus aureus. Mol Microbiol 66 1136 1147

25. PeschelA

OttoM

JackRW

KalbacherH

JungG

1999 Inactivation of the dlt operon in Staphylococcus aureus confers sensitivity to defensins, protegrins, and other antimicrobial peptides. J Biol Chem 274 8405 8410

26. PeschelA

JackRW

OttoM

CollinsLV

StaubitzP

2001 Staphylococcus aureus resistance to human defensins and evasion of neutrophil killing via the novel virulence factor MprF is based on modification of membrane lipids with l-lysine. J Exp Med 193 1067 1076

27. VoyichJM

BraughtonKR

SturdevantDE

WhitneyAR

Said-SalimB

2005 Insights into mechanisms used by Staphylococcus aureus to avoid destruction by human neutrophils. J Immunol 175 3907 3919

28. ZhangYQ

RenSX

LiHL

WangYX

FuG

2003 Genome-based analysis of virulence genes in a non-biofilm-forming Staphylococcus epidermidis strain (ATCC 12228). Mol Microbiol 49 1577 1593

29. QueckSY

Jameson-LeeM

VillaruzAE

BachTH

KhanBA

2008 RNAIII-Independent Target Gene Control by the agr Quorum-Sensing System: Insight into the Evolution of Virulence Regulation in Staphylococcus aureus. Mol Cell 32 150 158

30. VuongC

DurrM

CarmodyAB

PeschelA

KlebanoffSJ

2004 Regulated expression of pathogen-associated molecular pattern molecules in Staphylococcus epidermidis: quorum-sensing determines pro-inflammatory capacity and production of phenol-soluble modulins. Cell Microbiol 6 753 759

31. MellorIR

ThomasDH

SansomMS

1988 Properties of ion channels formed by Staphylococcus aureus delta-toxin. Biochim Biophys Acta 942 280 294

32. TalbotJC

ThiaudiereE

VincentM

GallayJ

SiffertO

2001 Dynamics and orientation of amphipathic peptides in solution and bound to membranes: a steady-state and time-resolved fluorescence study of staphylococcal delta-toxin and its synthetic analogues. Eur Biophys J 30 147 161

33. CogenAL

YamasakiK

SanchezKM

DorschnerRA

LaiY

2010 Selective antimicrobial action is provided by phenol-soluble modulins derived from Staphylococcus epidermidis, a normal resident of the skin. J Invest Dermatol 130 192 200

34. OttoM

O'MahoneyDS

GuinaT

KlebanoffSJ

2004 Activity of Staphylococcus epidermidis phenol-soluble modulin peptides expressed in Staphylococcus carnosus. J Infect Dis 190 748 755

35. VuongC

KocianovaS

YaoY

CarmodyAB

OttoM

2004 Increased colonization of indwelling medical devices by quorum-sensing mutants of Staphylococcus epidermidis in vivo. J Infect Dis 190 1498 1505

36. VuongC

GerkeC

SomervilleGA

FischerER

OttoM

2003 Quorum-sensing control of biofilm factors in Staphylococcus epidermidis. J Infect Dis 188 706 718

37. LeY

MurphyPM

WangJM

2002 Formyl-peptide receptors revisited. Trends Immunol 23 541 548

38. SomervilleGA

CockayneA

DurrM

PeschelA

OttoM

2003 Synthesis and deformylation of Staphylococcus aureus delta-toxin are linked to tricarboxylic acid cycle activity. J Bacteriol 185 6686 6694

39. KretschmerD

GleskeA

RautenbergM

WangR

KoberleM

2010 Human formyl peptide receptor 2 (FPR2/ALX) senses highly pathogenic Staphylococcus aureus. Cell Host Microbe. In press

40. KobayashiY

2008 The role of chemokines in neutrophil biology. Front Biosci 13 2400 2407

41. NizetV

2007 Understanding how leading bacterial pathogens subvert innate immunity to reveal novel therapeutic targets. J Allergy Clin Immunol 120 13 22

42. LaiY

Di NardoA

NakatsujiT

LeichtleA

YangY

2009 Commensal bacteria regulate Toll-like receptor 3-dependent inflammation after skin injury. Nat Med 15 1377 1382

43. VuongC

OttoM

2002 Staphylococcus epidermidis infections. Microbes Infect 4 481 489

44. KongKF

VuongC

OttoM

2006 Staphylococcus quorum sensing in biofilm formation and infection. Int J Med Microbiol 296 133 139

45. RogersDE

TompsettR

1952 The survival of staphylococci within human leukocytes. J Exp Med 95 209 230

46. YaoY

VuongC

KocianovaS

VillaruzAE

LaiY

2006 Characterization of the Staphylococcus epidermidis Accessory-Gene Regulator Response: Quorum-Sensing Regulation of Resistance to Human Innate Host Defense. J Infect Dis 193 841 848

47. GreshamHD

LowranceJH

CaverTE

WilsonBS

CheungAL

2000 Survival of Staphylococcus aureus inside neutrophils contributes to infection. J Immunol 164 3713 3722

48. ShompoleS

HenonKT

LiouLE

DziewanowskaK

BohachGA

2003 Biphasic intracellular expression of Staphylococcus aureus virulence factors and evidence for Agr-mediated diffusion sensing. Mol Microbiol 49 919 927

49. MackD

NedelmannM

KrokotschA

SchwarzkopfA

HeesemannJ

1994 Characterization of transposon mutants of biofilm-producing Staphylococcus epidermidis impaired in the accumulative phase of biofilm production: genetic identification of a hexosamine-containing polysaccharide intercellular adhesin. Infect Immun 62 3244 3253

50. ChristensenGD

BisnoAL

ParisiJT

McLaughlinB

HesterMG

1982 Nosocomial septicemia due to multiply antibiotic-resistant Staphylococcus epidermidis. Ann Intern Med 96 1 10

51. HeilmannC

GerkeC

Perdreau-RemingtonF

GotzF

1996 Characterization of Tn917 insertion mutants of Staphylococcus epidermidis affected in biofilm formation. Infect Immun 64 277 282

52. VuongC

GotzF

OttoM

2000 Construction and characterization of an agr deletion mutant of Staphylococcus epidermidis. Infect Immun 68 1048 1053

53. CDC

2003 Outbreaks of community-associated methicillin-resistant Staphylococcus aureus skin infections—Los Angeles County, California, 2002–2003. MMWR Morb Mortal Wkly Rep 52 88

54. CDC

1999 From the Centers for Disease Control and Prevention. Four pediatric deaths from community-acquired methicillin-resistant Staphylococcus aureus—Minnesota and North Dakota, 1997-1999. Jama 282 1123 1125

55. PeschelA

OttenwalderB

GotzF

1996 Inducible production and cellular location of the epidermin biosynthetic enzyme EpiB using an improved staphylococcal expression system. FEMS Microbiol Lett 137 279 284

56. VoyichJM

OttoM

MathemaB

BraughtonKR

WhitneyAR

2006 Is Panton-Valentine Leukocidin the Major Virulence Determinant in Community-Associated Methicillin-Resistant Staphylococcus aureus Disease? J Infect Dis 194 1761 1770

57. SreeramaN

WoodyRW

2004 Computation and analysis of protein circular dichroism spectra. Methods Enzymol 383 318 351

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2010 Číslo 10
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#