Skip to main content
Erschienen in: Current Allergy and Asthma Reports 12/2017

01.12.2017 | Basic and Applied Science (I Lewkowich, Section Editor)

Staphylococcal Biofilms in Atopic Dermatitis

verfasst von: Tammy Gonzalez, Jocelyn M. Biagini Myers, Andrew B. Herr, Gurjit K. Khurana Hershey

Erschienen in: Current Allergy and Asthma Reports | Ausgabe 12/2017

Einloggen, um Zugang zu erhalten

Abstract

Purpose of Review

Atopic dermatitis (AD) is a chronic, relapsing inflammatory skin disorder that is a major public health burden worldwide. AD lesions are often colonized by Staphylococcus aureus and Staphylococcus epidermidis. An important aspect of Staphylococcus spp. is their propensity to form biofilms, adhesive surface-attached colonies that become highly resistant to antibiotics and immune responses, and recent studies have found that clinical isolates colonizing AD skin are often biofilm-positive. Biofilm formation results in complex bacterial communities that have unique effects on keratinocytes and host immunity. This review will summarize recent studies exploring the role of staphyloccocal biofilms in atopic dermatitis and the implications for treatment.

Recent Findings

Recent studies suggest an important role for biofilms in the pathogenesis of numerous dermatologic diseases including AD. S. aureus biofilms have been found to colonize the eccrine ducts of AD skin, and these biofilms influence secretion of keratinocyte cytokines and trigger differentiation and apoptosis of keratinocytes. These activities may act to disrupt barrier function and promote disease pathogenesis as well as allergen sensitization.

Summary

Formation of biofilm is a successful strategy that protects the bacteria from environmental danger, antibiotics, and phagocytosis, enabling chronic persistence in the host. An increasing number of S. aureus skin isolates are resistant to conventional antibiotics, and staphylococcal biofilm communities are prevalent on the skin of individuals with AD. Staphylococcal colonization of the skin impacts skin barrier function and plays multiple important roles in AD pathogenesis.
Literatur
2.
Zurück zum Zitat Drucker AM, Wang AR, Li WQ, Sevetson E, Block JK, Qureshi AA. The burden of atopic dermatitis: summary of a report for the National Eczema Association. J Invest Dermatol. 2017;137(1):26–30.PubMedCrossRef Drucker AM, Wang AR, Li WQ, Sevetson E, Block JK, Qureshi AA. The burden of atopic dermatitis: summary of a report for the National Eczema Association. J Invest Dermatol. 2017;137(1):26–30.PubMedCrossRef
3.
Zurück zum Zitat Biagini Myers JM, Khurana Hershey GK. Eczema in early life: genetics, the skin barrier, and lessons learned from birth cohort studies. J Pediatr. 2010;157(5):704–14.PubMedPubMedCentralCrossRef Biagini Myers JM, Khurana Hershey GK. Eczema in early life: genetics, the skin barrier, and lessons learned from birth cohort studies. J Pediatr. 2010;157(5):704–14.PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Zheng T, Yu J, Oh MH, Zhu Z. The atopic march: progression from atopic dermatitis to allergic rhinitis and asthma. Allergy Asthma Immunol Res. 2011;3(2):67–73.PubMedPubMedCentralCrossRef Zheng T, Yu J, Oh MH, Zhu Z. The atopic march: progression from atopic dermatitis to allergic rhinitis and asthma. Allergy Asthma Immunol Res. 2011;3(2):67–73.PubMedPubMedCentralCrossRef
6.
7.
Zurück zum Zitat Kobayashi T, Glatz M, Horiuchi K, Kawasaki H, Akiyama H, Kaplan DH, et al. Dysbiosis and Staphylococcus aureus colonization drives inflammation in atopic dermatitis. Immunity. 2015;42(4):756–66.PubMedPubMedCentralCrossRef Kobayashi T, Glatz M, Horiuchi K, Kawasaki H, Akiyama H, Kaplan DH, et al. Dysbiosis and Staphylococcus aureus colonization drives inflammation in atopic dermatitis. Immunity. 2015;42(4):756–66.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Williams MR, Gallo RL. The role of the skin microbiome in atopic dermatitis. Curr Allergy Asthma Rep. 2015;15(11):65.PubMedCrossRef Williams MR, Gallo RL. The role of the skin microbiome in atopic dermatitis. Curr Allergy Asthma Rep. 2015;15(11):65.PubMedCrossRef
10.
Zurück zum Zitat Ovaere P, Lippens S, Vandenabeele P, Declercq W. The emerging roles of serine protease cascades in the epidermis. Trends Biochem Sci. 2009;34(9):453–63.PubMedCrossRef Ovaere P, Lippens S, Vandenabeele P, Declercq W. The emerging roles of serine protease cascades in the epidermis. Trends Biochem Sci. 2009;34(9):453–63.PubMedCrossRef
12.
Zurück zum Zitat Archer NK, Mazaitis MJ, Costerton JW, Leid JG, Powers ME, Shirtliff ME. Staphylococcus aureus biofilms: Properties, regulation and roles in human disease. Virulence. 2011;2(5):445-59. Archer NK, Mazaitis MJ, Costerton JW, Leid JG, Powers ME, Shirtliff ME. Staphylococcus aureus biofilms: Properties, regulation and roles in human disease. Virulence. 2011;2(5):445-59.
13.
14.
Zurück zum Zitat •• Allen HB, Vaze ND, Choi C, Hailu T, Tulbert BH, Cusack CA, et al. The presence and impact of biofilm-producing staphylococci in atopic dermatitis. JAMA Dermatol. 2014;150(3):260–5. The initial study reporting near-ubiquitous S. aureus biofilms in AD lesional skin and showing the activation of TLR2 adjacent to the sweat ducts. PubMedCrossRef •• Allen HB, Vaze ND, Choi C, Hailu T, Tulbert BH, Cusack CA, et al. The presence and impact of biofilm-producing staphylococci in atopic dermatitis. JAMA Dermatol. 2014;150(3):260–5. The initial study reporting near-ubiquitous S. aureus biofilms in AD lesional skin and showing the activation of TLR2 adjacent to the sweat ducts. PubMedCrossRef
15.
Zurück zum Zitat Nutten S. Atopic dermatitis: global epidemiology and risk factors. Ann Nutr Metab. 2015;66(Suppl 1):8–16.PubMedCrossRef Nutten S. Atopic dermatitis: global epidemiology and risk factors. Ann Nutr Metab. 2015;66(Suppl 1):8–16.PubMedCrossRef
16.
Zurück zum Zitat Williams H, Flohr C. How epidemiology has challenged 3 prevailing concepts about atopic dermatitis. J Allergy Clin Immunol. 2006;118(1):209–13.PubMedCrossRef Williams H, Flohr C. How epidemiology has challenged 3 prevailing concepts about atopic dermatitis. J Allergy Clin Immunol. 2006;118(1):209–13.PubMedCrossRef
18.
Zurück zum Zitat Williams H, Stewart A, von Mutius E, Cookson W, Anderson HR. Is eczema really on the increase worldwide? J Allergy Clin Immunol. 2008;121(4):947–54. e15PubMedCrossRef Williams H, Stewart A, von Mutius E, Cookson W, Anderson HR. Is eczema really on the increase worldwide? J Allergy Clin Immunol. 2008;121(4):947–54. e15PubMedCrossRef
19.
Zurück zum Zitat Strachan D, Sibbald B, Weiland S, Ait-Khaled N, Anabwani G, Anderson HR, et al. Worldwide variations in prevalence of symptoms of allergic rhinoconjunctivitis in children: the International Study of Asthma and Allergies in Childhood (ISAAC). Pediatr Allergy Immunol. 1997;8(4):161–76.PubMedCrossRef Strachan D, Sibbald B, Weiland S, Ait-Khaled N, Anabwani G, Anderson HR, et al. Worldwide variations in prevalence of symptoms of allergic rhinoconjunctivitis in children: the International Study of Asthma and Allergies in Childhood (ISAAC). Pediatr Allergy Immunol. 1997;8(4):161–76.PubMedCrossRef
21.
Zurück zum Zitat Wang M, Karlsson C, Olsson C, Adlerberth I, Wold AE, Strachan DP, et al. Reduced diversity in the early fecal microbiota of infants with atopic eczema. J Allergy Clin Immunol. 2008;121(1):129–34.PubMedCrossRef Wang M, Karlsson C, Olsson C, Adlerberth I, Wold AE, Strachan DP, et al. Reduced diversity in the early fecal microbiota of infants with atopic eczema. J Allergy Clin Immunol. 2008;121(1):129–34.PubMedCrossRef
22.
Zurück zum Zitat Okada H, Kuhn C, Feillet H, Bach JF. The ‘hygiene hypothesis’ for autoimmune and allergic diseases: an update. Clin Exp Immunol. 2010;160(1):1–9.PubMedPubMedCentralCrossRef Okada H, Kuhn C, Feillet H, Bach JF. The ‘hygiene hypothesis’ for autoimmune and allergic diseases: an update. Clin Exp Immunol. 2010;160(1):1–9.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat United Nations. World Urbanization Prospects; the 2007 revision. United Nations Department of Economic and Social Affairs, Population Division.. New York; 2008. United Nations. World Urbanization Prospects; the 2007 revision. United Nations Department of Economic and Social Affairs, Population Division.. New York; 2008.
24.
Zurück zum Zitat Bendiks M, Kopp MV. The relationship between advances in understanding the microbiome and the maturing hygiene hypothesis. Curr Allergy Asthma Rep. 2013;13(5):487–94.PubMedCrossRef Bendiks M, Kopp MV. The relationship between advances in understanding the microbiome and the maturing hygiene hypothesis. Curr Allergy Asthma Rep. 2013;13(5):487–94.PubMedCrossRef
26.
Zurück zum Zitat Ong PY, Leung DY. Bacterial and viral infections in atopic dermatitis: a comprehensive review. Clin Rev Allergy Immunol. 2016;51(3):329–37.PubMedCrossRef Ong PY, Leung DY. Bacterial and viral infections in atopic dermatitis: a comprehensive review. Clin Rev Allergy Immunol. 2016;51(3):329–37.PubMedCrossRef
27.
Zurück zum Zitat Lakatos G, Soproni K, Doka A, Miklosi A. A comparative approach to dogs’ (Canis familiaris) and human infants’ comprehension of various forms of pointing gestures. Anim Cogn. 2009;12(4):621–31.PubMedCrossRef Lakatos G, Soproni K, Doka A, Miklosi A. A comparative approach to dogs’ (Canis familiaris) and human infants’ comprehension of various forms of pointing gestures. Anim Cogn. 2009;12(4):621–31.PubMedCrossRef
28.
Zurück zum Zitat Kawasaki H, Nagao K, Kubo A, Hata T, Shimizu A, Mizuno H, et al. Altered stratum corneum barrier and enhanced percutaneous immune responses in filaggrin-null mice. J Allergy Clin Immunol. 2012;129(6):1538–46. e6PubMedCrossRef Kawasaki H, Nagao K, Kubo A, Hata T, Shimizu A, Mizuno H, et al. Altered stratum corneum barrier and enhanced percutaneous immune responses in filaggrin-null mice. J Allergy Clin Immunol. 2012;129(6):1538–46. e6PubMedCrossRef
29.
Zurück zum Zitat Oyoshi MK, Murphy GF, Geha RS. Filaggrin-deficient mice exhibit TH17-dominated skin inflammation and permissiveness to epicutaneous sensitization with protein antigen. J Allergy Clin Immunol. 2009;124(3):485–93. 93 e1PubMedPubMedCentralCrossRef Oyoshi MK, Murphy GF, Geha RS. Filaggrin-deficient mice exhibit TH17-dominated skin inflammation and permissiveness to epicutaneous sensitization with protein antigen. J Allergy Clin Immunol. 2009;124(3):485–93. 93 e1PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Gupta J, Grube E, Ericksen MB, Stevenson MD, Lucky AW, Sheth AP, et al. Intrinsically defective skin barrier function in children with atopic dermatitis correlates with disease severity. J Allergy Clin Immunol. 2008;121(3):725–30. e2PubMedCrossRef Gupta J, Grube E, Ericksen MB, Stevenson MD, Lucky AW, Sheth AP, et al. Intrinsically defective skin barrier function in children with atopic dermatitis correlates with disease severity. J Allergy Clin Immunol. 2008;121(3):725–30. e2PubMedCrossRef
31.
Zurück zum Zitat de Veer SJ, Furio L, Harris JM, Hovnanian A. Proteases: common culprits in human skin disorders. Trends Mol Med. 2014;20(3):166–78.PubMedCrossRef de Veer SJ, Furio L, Harris JM, Hovnanian A. Proteases: common culprits in human skin disorders. Trends Mol Med. 2014;20(3):166–78.PubMedCrossRef
32.
Zurück zum Zitat Fischer J, Meyer-Hoffert U. Regulation of kallikrein-related peptidases in the skin—from physiology to diseases to therapeutic options. Thromb Haemost. 2013;110(3):442–9.PubMedCrossRef Fischer J, Meyer-Hoffert U. Regulation of kallikrein-related peptidases in the skin—from physiology to diseases to therapeutic options. Thromb Haemost. 2013;110(3):442–9.PubMedCrossRef
33.
Zurück zum Zitat Deraison C, Bonnart C, Lopez F, Besson C, Robinson R, Jayakumar A, et al. LEKTI fragments specifically inhibit KLK5, KLK7, and KLK14 and control desquamation through a pH-dependent interaction. Mol Biol Cell. 2007;18(9):3607–19.PubMedPubMedCentralCrossRef Deraison C, Bonnart C, Lopez F, Besson C, Robinson R, Jayakumar A, et al. LEKTI fragments specifically inhibit KLK5, KLK7, and KLK14 and control desquamation through a pH-dependent interaction. Mol Biol Cell. 2007;18(9):3607–19.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Weidinger S, Baurecht H, Wagenpfeil S, Henderson J, Novak N, Sandilands A, et al. Analysis of the individual and aggregate genetic contributions of previously identified serine peptidase inhibitor Kazal type 5 (SPINK5), kallikrein-related peptidase 7 (KLK7), and filaggrin (FLG) polymorphisms to eczema risk. J Allergy Clin Immunol. 2008;122(3):560–8. e4PubMedCrossRef Weidinger S, Baurecht H, Wagenpfeil S, Henderson J, Novak N, Sandilands A, et al. Analysis of the individual and aggregate genetic contributions of previously identified serine peptidase inhibitor Kazal type 5 (SPINK5), kallikrein-related peptidase 7 (KLK7), and filaggrin (FLG) polymorphisms to eczema risk. J Allergy Clin Immunol. 2008;122(3):560–8. e4PubMedCrossRef
35.
Zurück zum Zitat Walley AJ, Chavanas S, Moffatt MF, Esnouf RM, Ubhi B, Lawrence R, et al. Gene polymorphism in Netherton and common atopic disease. Nat Genet. 2001;29(2):175–8.PubMedCrossRef Walley AJ, Chavanas S, Moffatt MF, Esnouf RM, Ubhi B, Lawrence R, et al. Gene polymorphism in Netherton and common atopic disease. Nat Genet. 2001;29(2):175–8.PubMedCrossRef
36.
Zurück zum Zitat Wang XW, Wang JJ, Gutowska-Owsiak D, Salimi M, Selvakumar TA, Gwela A, et al. Deficiency of filaggrin regulates endogenous cysteine protease activity, leading to impaired skin barrier function. Clin Exp Dermatol. 2017;42(6):622–31.PubMedCrossRef Wang XW, Wang JJ, Gutowska-Owsiak D, Salimi M, Selvakumar TA, Gwela A, et al. Deficiency of filaggrin regulates endogenous cysteine protease activity, leading to impaired skin barrier function. Clin Exp Dermatol. 2017;42(6):622–31.PubMedCrossRef
37.
Zurück zum Zitat Williams MR, Nakatsuji T, Sanford JA, Vrbanac AF, Gallo RL. Staphylococcus aureus induces increased serine protease activity in keratinocytes. J Invest Dermatol. 2017;137(2):377–84.PubMedCrossRef Williams MR, Nakatsuji T, Sanford JA, Vrbanac AF, Gallo RL. Staphylococcus aureus induces increased serine protease activity in keratinocytes. J Invest Dermatol. 2017;137(2):377–84.PubMedCrossRef
38.
Zurück zum Zitat Gallo RL. Human skin is the largest epithelial surface for interaction with microbes. J Invest Dermatol. 2017;137(6):1213–4.PubMedCrossRef Gallo RL. Human skin is the largest epithelial surface for interaction with microbes. J Invest Dermatol. 2017;137(6):1213–4.PubMedCrossRef
40.
Zurück zum Zitat Belkaid Y, Segre JA. Dialogue between skin microbiota and immunity. Science (New York, NY). 2014;346(6212):954–9.CrossRef Belkaid Y, Segre JA. Dialogue between skin microbiota and immunity. Science (New York, NY). 2014;346(6212):954–9.CrossRef
41.
Zurück zum Zitat SanMiguel A, Grice EA. Interactions between host factors and the skin microbiome. Cell Mol Life Sci. 2015;72(8):1499–515.PubMedCrossRef SanMiguel A, Grice EA. Interactions between host factors and the skin microbiome. Cell Mol Life Sci. 2015;72(8):1499–515.PubMedCrossRef
43.
Zurück zum Zitat Kennedy EA, Connolly J, Hourihane JO, Fallon PG, McLean WH, Murray D, et al. Skin microbiome before development of atopic dermatitis: early colonization with commensal staphylococci at 2 months is associated with a lower risk of atopic dermatitis at 1 year. J Allergy Clin Immunol. 2017;139(1):166–72.PubMedPubMedCentralCrossRef Kennedy EA, Connolly J, Hourihane JO, Fallon PG, McLean WH, Murray D, et al. Skin microbiome before development of atopic dermatitis: early colonization with commensal staphylococci at 2 months is associated with a lower risk of atopic dermatitis at 1 year. J Allergy Clin Immunol. 2017;139(1):166–72.PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat •• Kong HH, Oh J, Deming C, Conlan S, Grice EA, Beatson MA, et al. Temporal shifts in the skin microbiome associated with disease flares and treatment in children with atopic dermatitis. Genome Res. 2012;22(5):850–9. A study describing dysbiosis in active AD lesions using 16S rRNA sequencing, showing increased prevalence of both S. aureus and S. epidermidis . PubMedPubMedCentralCrossRef •• Kong HH, Oh J, Deming C, Conlan S, Grice EA, Beatson MA, et al. Temporal shifts in the skin microbiome associated with disease flares and treatment in children with atopic dermatitis. Genome Res. 2012;22(5):850–9. A study describing dysbiosis in active AD lesions using 16S rRNA sequencing, showing increased prevalence of both S. aureus and S. epidermidis . PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Poretsky R, Rodriguez RL, Luo C, Tsementzi D, Konstantinidis KT. Strengths and limitations of 16S rRNA gene amplicon sequencing in revealing temporal microbial community dynamics. PLoS One. 2014;9(4):e93827.PubMedPubMedCentralCrossRef Poretsky R, Rodriguez RL, Luo C, Tsementzi D, Konstantinidis KT. Strengths and limitations of 16S rRNA gene amplicon sequencing in revealing temporal microbial community dynamics. PLoS One. 2014;9(4):e93827.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat •• Byrd AL, Deming C, Cassidy SKB, Harrison OJ, Ng WI, Conlan S, et al. Staphylococcus aureus and Staphylococcus epidermidis strain diversity underlying pediatric atopic dermatitis. Sci Transl Med. 2017;9(397):eaal4651. A study using metagenomic shotgun sequencing to identify strain-level differences in S. aureus and S. epidermidis colonization in pediatric AD patients. PubMedCrossRef •• Byrd AL, Deming C, Cassidy SKB, Harrison OJ, Ng WI, Conlan S, et al. Staphylococcus aureus and Staphylococcus epidermidis strain diversity underlying pediatric atopic dermatitis. Sci Transl Med. 2017;9(397):eaal4651. A study using metagenomic shotgun sequencing to identify strain-level differences in S. aureus and S. epidermidis colonization in pediatric AD patients. PubMedCrossRef
47.
Zurück zum Zitat •• Oh J, Byrd AL, Deming C, Conlan S, Program NCS, Kong HH, et al. Biogeography and individuality shape function in the human skin metagenome. Nature. 2014;514(7520):59–64. The first metagenomic survey of different healthy human skin sites, which lays the foundation for studies to assess changes of the skin microbiome in disease. PubMedPubMedCentralCrossRef •• Oh J, Byrd AL, Deming C, Conlan S, Program NCS, Kong HH, et al. Biogeography and individuality shape function in the human skin metagenome. Nature. 2014;514(7520):59–64. The first metagenomic survey of different healthy human skin sites, which lays the foundation for studies to assess changes of the skin microbiome in disease. PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Brauweiler AM, Goleva E, Leung DY. Th2 cytokines increase Staphylococcus aureus alpha toxin-induced keratinocyte death through the signal transducer and activator of transcription 6 (STAT6). J Invest Dermatol. 2014;134(8):2114–21.PubMedPubMedCentralCrossRef Brauweiler AM, Goleva E, Leung DY. Th2 cytokines increase Staphylococcus aureus alpha toxin-induced keratinocyte death through the signal transducer and activator of transcription 6 (STAT6). J Invest Dermatol. 2014;134(8):2114–21.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Brauweiler AM, Bin L, Kim BE, Oyoshi MK, Geha RS, Goleva E, et al. Filaggrin-dependent secretion of sphingomyelinase protects against staphylococcal alpha-toxin-induced keratinocyte death. J Allergy Clin Immunol. 2013;131(2):421–7. e1-2PubMedCrossRef Brauweiler AM, Bin L, Kim BE, Oyoshi MK, Geha RS, Goleva E, et al. Filaggrin-dependent secretion of sphingomyelinase protects against staphylococcal alpha-toxin-induced keratinocyte death. J Allergy Clin Immunol. 2013;131(2):421–7. e1-2PubMedCrossRef
50.
Zurück zum Zitat Amagai M, Matsuyoshi N, Wang ZH, Andl C, Stanley JR. Toxin in bullous impetigo and staphylococcal scalded-skin syndrome targets desmoglein 1. Nat Med. 2000;6(11):1275–7.PubMedCrossRef Amagai M, Matsuyoshi N, Wang ZH, Andl C, Stanley JR. Toxin in bullous impetigo and staphylococcal scalded-skin syndrome targets desmoglein 1. Nat Med. 2000;6(11):1275–7.PubMedCrossRef
51.
Zurück zum Zitat Hanakawa Y, Schechter NM, Lin C, Nishifuji K, Amagai M, Stanley JR. Enzymatic and molecular characteristics of the efficiency and specificity of exfoliative toxin cleavage of desmoglein 1. J Biol Chem. 2004;279(7):5268–77.PubMedCrossRef Hanakawa Y, Schechter NM, Lin C, Nishifuji K, Amagai M, Stanley JR. Enzymatic and molecular characteristics of the efficiency and specificity of exfoliative toxin cleavage of desmoglein 1. J Biol Chem. 2004;279(7):5268–77.PubMedCrossRef
52.
Zurück zum Zitat Amagai M, Yamaguchi T, Hanakawa Y, Nishifuji K, Sugai M, Stanley JR. Staphylococcal exfoliative toxin B specifically cleaves desmoglein 1. J Invest Dermat. 2002;118(5):845–50.CrossRef Amagai M, Yamaguchi T, Hanakawa Y, Nishifuji K, Sugai M, Stanley JR. Staphylococcal exfoliative toxin B specifically cleaves desmoglein 1. J Invest Dermat. 2002;118(5):845–50.CrossRef
53.
Zurück zum Zitat Hauk PJ, Hamid QA, Chrousos GP, Leung DY. Induction of corticosteroid insensitivity in human PBMCs by microbial superantigens. J Allergy Clin Immunol. 2000;105(4):782–7.PubMedCrossRef Hauk PJ, Hamid QA, Chrousos GP, Leung DY. Induction of corticosteroid insensitivity in human PBMCs by microbial superantigens. J Allergy Clin Immunol. 2000;105(4):782–7.PubMedCrossRef
54.
Zurück zum Zitat Schlievert PM, Case LC, Strandberg KL, Abrams BB, Leung DY. Superantigen profile of Staphylococcus aureus isolates from patients with steroid-resistant atopic dermatitis. Clin Infect Dis. 2008;46(10):1562–7.PubMedPubMedCentralCrossRef Schlievert PM, Case LC, Strandberg KL, Abrams BB, Leung DY. Superantigen profile of Staphylococcus aureus isolates from patients with steroid-resistant atopic dermatitis. Clin Infect Dis. 2008;46(10):1562–7.PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Costerton W, Veeh R, Shirtliff M, Pasmore M, Post C, Ehrlich G. The application of biofilm science to the study and control of chronic bacterial infections. J Clin Invest. 2003;112(10):1466–77.PubMedPubMedCentralCrossRef Costerton W, Veeh R, Shirtliff M, Pasmore M, Post C, Ehrlich G. The application of biofilm science to the study and control of chronic bacterial infections. J Clin Invest. 2003;112(10):1466–77.PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Watters C, Fleming D, Bishop D, Rumbaugh KP. Host responses to biofilm. Prog Mol Biol Transl Sci. 2016;142:193–239.PubMedCrossRef Watters C, Fleming D, Bishop D, Rumbaugh KP. Host responses to biofilm. Prog Mol Biol Transl Sci. 2016;142:193–239.PubMedCrossRef
58.
Zurück zum Zitat Otto M. Staphylococcal infections: mechanisms of biofilm maturation and detachment as critical determinants of pathogenicity. Annu Rev Med. 2013;64:175–88.PubMedCrossRef Otto M. Staphylococcal infections: mechanisms of biofilm maturation and detachment as critical determinants of pathogenicity. Annu Rev Med. 2013;64:175–88.PubMedCrossRef
60.
Zurück zum Zitat Vlassova N, Han A, Zenilman JM, James G, Lazarus GS. New horizons for cutaneous microbiology: the role of biofilms in dermatological disease. Br J Dermatol. 2011;165(4):751–9.PubMedCrossRef Vlassova N, Han A, Zenilman JM, James G, Lazarus GS. New horizons for cutaneous microbiology: the role of biofilms in dermatological disease. Br J Dermatol. 2011;165(4):751–9.PubMedCrossRef
62.
Zurück zum Zitat Moormeier DE, Bose JL, Horswill AR, Bayles KW. Temporal and stochastic control of Staphylococcus aureus biofilm development. MBio. 2014;5(5):e01341–14.PubMedPubMedCentralCrossRef Moormeier DE, Bose JL, Horswill AR, Bayles KW. Temporal and stochastic control of Staphylococcus aureus biofilm development. MBio. 2014;5(5):e01341–14.PubMedPubMedCentralCrossRef
63.
Zurück zum Zitat Sharp JA, Echague CG, Hair PS, Ward MD, Nyalwidhe JO, Geoghegan JA, et al. Staphylococcus aureus surface protein SdrE binds complement regulator factor H as an immune evasion tactic. PLoS One. 2012;7(5):e38407.PubMedPubMedCentralCrossRef Sharp JA, Echague CG, Hair PS, Ward MD, Nyalwidhe JO, Geoghegan JA, et al. Staphylococcus aureus surface protein SdrE binds complement regulator factor H as an immune evasion tactic. PLoS One. 2012;7(5):e38407.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Foster TJ, Hook M. Surface protein adhesins of Staphylococcus aureus. Trends Microbiol. 1998;6(12):484–8.PubMedCrossRef Foster TJ, Hook M. Surface protein adhesins of Staphylococcus aureus. Trends Microbiol. 1998;6(12):484–8.PubMedCrossRef
65.
Zurück zum Zitat Ponnuraj K, Bowden MG, Davis S, Gurusiddappa S, Moore D, Choe D, et al. A “dock, lock, and latch” structural model for a staphylococcal adhesin binding to fibrinogen. Cell. 2003;115(2):217–28.PubMedCrossRef Ponnuraj K, Bowden MG, Davis S, Gurusiddappa S, Moore D, Choe D, et al. A “dock, lock, and latch” structural model for a staphylococcal adhesin binding to fibrinogen. Cell. 2003;115(2):217–28.PubMedCrossRef
66.
Zurück zum Zitat Zhang X, Wu M, Zhuo W, Gu J, Zhang S, Ge J, et al. Crystal structures of Bbp from Staphylococcus aureus reveal the ligand binding mechanism with Fibrinogen alpha. Protein Cell. 2015;6(10):757–66.PubMedPubMedCentralCrossRef Zhang X, Wu M, Zhuo W, Gu J, Zhang S, Ge J, et al. Crystal structures of Bbp from Staphylococcus aureus reveal the ligand binding mechanism with Fibrinogen alpha. Protein Cell. 2015;6(10):757–66.PubMedPubMedCentralCrossRef
67.
Zurück zum Zitat Ganesh VK, Rivera JJ, Smeds E, Ko YP, Bowden MG, Wann ER, et al. A structural model of the Staphylococcus aureus ClfA-fibrinogen interaction opens new avenues for the design of anti-staphylococcal therapeutics. PLoS Pathog. 2008;4(11):e1000226.PubMedPubMedCentralCrossRef Ganesh VK, Rivera JJ, Smeds E, Ko YP, Bowden MG, Wann ER, et al. A structural model of the Staphylococcus aureus ClfA-fibrinogen interaction opens new avenues for the design of anti-staphylococcal therapeutics. PLoS Pathog. 2008;4(11):e1000226.PubMedPubMedCentralCrossRef
68.
Zurück zum Zitat Xiang H, Feng Y, Wang J, Liu B, Chen Y, Liu L, et al. Crystal structures reveal the multi-ligand binding mechanism of Staphylococcus aureus ClfB. PLoS Pathog. 2012;8(6):e1002751.PubMedPubMedCentralCrossRef Xiang H, Feng Y, Wang J, Liu B, Chen Y, Liu L, et al. Crystal structures reveal the multi-ligand binding mechanism of Staphylococcus aureus ClfB. PLoS Pathog. 2012;8(6):e1002751.PubMedPubMedCentralCrossRef
69.
Zurück zum Zitat Askarian F, Ajayi C, Hanssen AM, van Sorge NM, Pettersen I, Diep DB, et al. The interaction between Staphylococcus aureus SdrD and desmoglein 1 is important for adhesion to host cells. Sci Rep. 2016;6:22134.PubMedPubMedCentralCrossRef Askarian F, Ajayi C, Hanssen AM, van Sorge NM, Pettersen I, Diep DB, et al. The interaction between Staphylococcus aureus SdrD and desmoglein 1 is important for adhesion to host cells. Sci Rep. 2016;6:22134.PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat Barbu EM, Ganesh VK, Gurusiddappa S, Mackenzie RC, Foster TJ, Sudhof TC, et al. Beta-neurexin is a ligand for the Staphylococcus aureus MSCRAMM SdrC. PLoS Pathog. 2010;6(1):e1000726.PubMedPubMedCentralCrossRef Barbu EM, Ganesh VK, Gurusiddappa S, Mackenzie RC, Foster TJ, Sudhof TC, et al. Beta-neurexin is a ligand for the Staphylococcus aureus MSCRAMM SdrC. PLoS Pathog. 2010;6(1):e1000726.PubMedPubMedCentralCrossRef
71.
Zurück zum Zitat Cho SH, Strickland I, Boguniewicz M, Leung DY. Fibronectin and fibrinogen contribute to the enhanced binding of Staphylococcus aureus to atopic skin. J Allergy Clin Immunol. 2001;108(2):269–74.PubMedCrossRef Cho SH, Strickland I, Boguniewicz M, Leung DY. Fibronectin and fibrinogen contribute to the enhanced binding of Staphylococcus aureus to atopic skin. J Allergy Clin Immunol. 2001;108(2):269–74.PubMedCrossRef
72.
Zurück zum Zitat Abraham NM, Jefferson KK. Staphylococcus aureus clumping factor B mediates biofilm formation in the absence of calcium. Microbiology. 2012;158(Pt 6):1504–12.PubMedPubMedCentralCrossRef Abraham NM, Jefferson KK. Staphylococcus aureus clumping factor B mediates biofilm formation in the absence of calcium. Microbiology. 2012;158(Pt 6):1504–12.PubMedPubMedCentralCrossRef
73.
Zurück zum Zitat Fleury OM, McAleer MA, Feuillie C, Formosa-Dague C, Sansevere E, Bennett DE, et al. Clumping Factor B promotes adherence of Staphylococcus aureus to corneocytes in atopic dermatitis. Infect Immun. 2017;85(6):e00994.PubMedCrossRefPubMedCentral Fleury OM, McAleer MA, Feuillie C, Formosa-Dague C, Sansevere E, Bennett DE, et al. Clumping Factor B promotes adherence of Staphylococcus aureus to corneocytes in atopic dermatitis. Infect Immun. 2017;85(6):e00994.PubMedCrossRefPubMedCentral
74.
Zurück zum Zitat Vuong C, Voyich JM, Fischer ER, Braughton KR, Whitney AR, DeLeo FR, et al. Polysaccharide intercellular adhesin (PIA) protects Staphylococcus epidermidis against major components of the human innate immune system. Cell Microbiol. 2004;6(3):269–75.PubMedCrossRef Vuong C, Voyich JM, Fischer ER, Braughton KR, Whitney AR, DeLeo FR, et al. Polysaccharide intercellular adhesin (PIA) protects Staphylococcus epidermidis against major components of the human innate immune system. Cell Microbiol. 2004;6(3):269–75.PubMedCrossRef
75.
Zurück zum Zitat Formosa-Dague C, Feuillie C, Beaussart A, Derclaye S, Kucharikova S, Lasa I, et al. Sticky matrix: adhesion mechanism of the Staphylococcal polysaccharide intercellular adhesin. ACS Nano. 2016;10(3):3443–52.PubMedCrossRef Formosa-Dague C, Feuillie C, Beaussart A, Derclaye S, Kucharikova S, Lasa I, et al. Sticky matrix: adhesion mechanism of the Staphylococcal polysaccharide intercellular adhesin. ACS Nano. 2016;10(3):3443–52.PubMedCrossRef
76.
Zurück zum Zitat Cramton SE, Gerke C, Schnell NF, Nichols WW, Gotz F. The intercellular adhesion (ica) locus is present in Staphylococcus aureus and is required for biofilm formation. Infect Immun. 1999;67(10):5427–33.PubMedPubMedCentral Cramton SE, Gerke C, Schnell NF, Nichols WW, Gotz F. The intercellular adhesion (ica) locus is present in Staphylococcus aureus and is required for biofilm formation. Infect Immun. 1999;67(10):5427–33.PubMedPubMedCentral
77.
78.
Zurück zum Zitat Schaeffer CR, Hoang TN, Sudbeck CM, Alawi M, Tolo IE, Robinson DA, et al. Versatility of biofilm matrix molecules in Staphylococcus epidermidis clinical isolates and importance of polysaccharide intercellular adhesin expression during high shear stress. mSphere. 2016;1(5):e00165.PubMedPubMedCentralCrossRef Schaeffer CR, Hoang TN, Sudbeck CM, Alawi M, Tolo IE, Robinson DA, et al. Versatility of biofilm matrix molecules in Staphylococcus epidermidis clinical isolates and importance of polysaccharide intercellular adhesin expression during high shear stress. mSphere. 2016;1(5):e00165.PubMedPubMedCentralCrossRef
79.
Zurück zum Zitat Yarawsky AE, English LR, Whitten ST, Herr AB. The proline/glycine-rich region of the biofilm adhesion protein Aap forms an extended stalk that resists compaction. J Mol Biol. 2017;429(2):261–79.PubMedCrossRef Yarawsky AE, English LR, Whitten ST, Herr AB. The proline/glycine-rich region of the biofilm adhesion protein Aap forms an extended stalk that resists compaction. J Mol Biol. 2017;429(2):261–79.PubMedCrossRef
80.
Zurück zum Zitat Hussain M, Herrmann M, von Eiff C, Perdreau-Remington F, Peters G. A 140-kilodalton extracellular protein is essential for the accumulation of Staphylococcus epidermidis strains on surfaces. Infect Immun. 1997;65(2):519–24.PubMedPubMedCentral Hussain M, Herrmann M, von Eiff C, Perdreau-Remington F, Peters G. A 140-kilodalton extracellular protein is essential for the accumulation of Staphylococcus epidermidis strains on surfaces. Infect Immun. 1997;65(2):519–24.PubMedPubMedCentral
81.
Zurück zum Zitat Paharik AE, Kotasinska M, Both A, Hoang TN, Buttner H, Roy P, et al. The metalloprotease SepA governs processing of accumulation-associated protein and shapes intercellular adhesive surface properties in Staphylococcus epidermidis. Mol Microbiol. 2017;103(5):860–74.PubMedCrossRef Paharik AE, Kotasinska M, Both A, Hoang TN, Buttner H, Roy P, et al. The metalloprotease SepA governs processing of accumulation-associated protein and shapes intercellular adhesive surface properties in Staphylococcus epidermidis. Mol Microbiol. 2017;103(5):860–74.PubMedCrossRef
82.
Zurück zum Zitat Rohde H, Burdelski C, Bartscht K, Hussain M, Buck F, Horstkotte MA, et al. Induction of Staphylococcus epidermidis biofilm formation via proteolytic processing of the accumulation-associated protein by staphylococcal and host proteases. Mol Microbiol. 2005;55(6):1883–95.PubMedCrossRef Rohde H, Burdelski C, Bartscht K, Hussain M, Buck F, Horstkotte MA, et al. Induction of Staphylococcus epidermidis biofilm formation via proteolytic processing of the accumulation-associated protein by staphylococcal and host proteases. Mol Microbiol. 2005;55(6):1883–95.PubMedCrossRef
83.
Zurück zum Zitat Corrigan RM, Rigby D, Handley P, Foster TJ. The role of Staphylococcus aureus surface protein SasG in adherence and biofilm formation. Microbiology. 2007;153(Pt 8):2435–46.PubMedCrossRef Corrigan RM, Rigby D, Handley P, Foster TJ. The role of Staphylococcus aureus surface protein SasG in adherence and biofilm formation. Microbiology. 2007;153(Pt 8):2435–46.PubMedCrossRef
84.
Zurück zum Zitat Conrady DG, Brescia CC, Horii K, Weiss AA, Hassett DJ, Herr AB. A zinc-dependent adhesion module is responsible for intercellular adhesion in staphylococcal biofilms. Proc Natl Acad Sci U S A. 2008;105(49):19456–61.PubMedPubMedCentralCrossRef Conrady DG, Brescia CC, Horii K, Weiss AA, Hassett DJ, Herr AB. A zinc-dependent adhesion module is responsible for intercellular adhesion in staphylococcal biofilms. Proc Natl Acad Sci U S A. 2008;105(49):19456–61.PubMedPubMedCentralCrossRef
85.
Zurück zum Zitat Herr AB, Conrady DG. Thermodynamic analysis of metal ion-induced protein assembly. Methods Enzymol. 2011;488:101–21.PubMedCrossRef Herr AB, Conrady DG. Thermodynamic analysis of metal ion-induced protein assembly. Methods Enzymol. 2011;488:101–21.PubMedCrossRef
86.
Zurück zum Zitat Conrady DG, Wilson JJ, Herr AB. Structural basis for Zn2+-dependent intercellular adhesion in staphylococcal biofilms. Proc Natl Acad Sci U S A. 2013;110(3):E202–11.PubMedCrossRef Conrady DG, Wilson JJ, Herr AB. Structural basis for Zn2+-dependent intercellular adhesion in staphylococcal biofilms. Proc Natl Acad Sci U S A. 2013;110(3):E202–11.PubMedCrossRef
87.
Zurück zum Zitat Shelton CL, Conrady DG, Herr AB. Functional consequences of B-repeat sequence variation in the staphylococcal biofilm protein Aap: deciphering the assembly code. Biochem J. 2017;474(3):427–43.PubMedCrossRef Shelton CL, Conrady DG, Herr AB. Functional consequences of B-repeat sequence variation in the staphylococcal biofilm protein Aap: deciphering the assembly code. Biochem J. 2017;474(3):427–43.PubMedCrossRef
88.
Zurück zum Zitat Chaton CT, Herr AB. Defining the metal specificity of a multifunctional biofilm adhesion protein. Protein Sci. 2017;26:1964–73.PubMedCrossRef Chaton CT, Herr AB. Defining the metal specificity of a multifunctional biofilm adhesion protein. Protein Sci. 2017;26:1964–73.PubMedCrossRef
89.
Zurück zum Zitat Geoghegan JA, Corrigan RM, Gruszka DT, Speziale P, O'Gara JP, Potts JR, et al. Role of surface protein SasG in biofilm formation by Staphylococcus aureus. J Bacteriol. 2010;192(21):5663–73.PubMedPubMedCentralCrossRef Geoghegan JA, Corrigan RM, Gruszka DT, Speziale P, O'Gara JP, Potts JR, et al. Role of surface protein SasG in biofilm formation by Staphylococcus aureus. J Bacteriol. 2010;192(21):5663–73.PubMedPubMedCentralCrossRef
90.
Zurück zum Zitat • Formosa-Dague C, Speziale P, Foster TJ, Geoghegan JA, Dufrene YF. Zinc-dependent mechanical properties of Staphylococcus aureus biofilm-forming surface protein SasG. Proc Natl Acad Sci U S A. 2016;113(2):410–5. A study demonstrating Zn2+-dependent intercellular adhesion between S. aureus cells mediated by SasG, and heterophilic adhesion between S. aureus and S. epidermidis mediated by SasG/Aap, using single-cell force microscopy. PubMedCrossRef • Formosa-Dague C, Speziale P, Foster TJ, Geoghegan JA, Dufrene YF. Zinc-dependent mechanical properties of Staphylococcus aureus biofilm-forming surface protein SasG. Proc Natl Acad Sci U S A. 2016;113(2):410–5. A study demonstrating Zn2+-dependent intercellular adhesion between S. aureus cells mediated by SasG, and heterophilic adhesion between S. aureus and S. epidermidis mediated by SasG/Aap, using single-cell force microscopy. PubMedCrossRef
91.
Zurück zum Zitat Peters BM, Jabra-Rizk MA, O'May GA, Costerton JW, Shirtliff ME. Polymicrobial interactions: impact on pathogenesis and human disease. Clin Microbiol Rev. 2012;25(1):193–213.PubMedPubMedCentralCrossRef Peters BM, Jabra-Rizk MA, O'May GA, Costerton JW, Shirtliff ME. Polymicrobial interactions: impact on pathogenesis and human disease. Clin Microbiol Rev. 2012;25(1):193–213.PubMedPubMedCentralCrossRef
92.
Zurück zum Zitat Stacy A, McNally L, Darch SE, Brown SP, Whiteley M. The biogeography of polymicrobial infection. Nat Rev Microbiol. 2016;14(2):93–105.PubMedCrossRef Stacy A, McNally L, Darch SE, Brown SP, Whiteley M. The biogeography of polymicrobial infection. Nat Rev Microbiol. 2016;14(2):93–105.PubMedCrossRef
93.
Zurück zum Zitat Wolcott R, Costerton JW, Raoult D, Cutler SJ. The polymicrobial nature of biofilm infection. Clin Microbiol Infect. 2013;19(2):107–12.PubMedCrossRef Wolcott R, Costerton JW, Raoult D, Cutler SJ. The polymicrobial nature of biofilm infection. Clin Microbiol Infect. 2013;19(2):107–12.PubMedCrossRef
95.
Zurück zum Zitat Silverman RJ, Nobbs AH, Vickerman MM, Barbour ME, Jenkinson HF. Interaction of Candida albicans cell wall Als3 protein with Streptococcus gordonii SspB adhesin promotes development of mixed-species communities. Infect Immun. 2010;78(11):4644–52.PubMedPubMedCentralCrossRef Silverman RJ, Nobbs AH, Vickerman MM, Barbour ME, Jenkinson HF. Interaction of Candida albicans cell wall Als3 protein with Streptococcus gordonii SspB adhesin promotes development of mixed-species communities. Infect Immun. 2010;78(11):4644–52.PubMedPubMedCentralCrossRef
96.
Zurück zum Zitat Stewart EJ, Payne DE, Ma TM, VanEpps JS, Boles BR, Younger JG, et al. Effect of antimicrobial and physical treatments on growth of multispecies Staphylococcal biofilms. Appl Environ Microbiol. 2017;83(12):e03483-16. Stewart EJ, Payne DE, Ma TM, VanEpps JS, Boles BR, Younger JG, et al. Effect of antimicrobial and physical treatments on growth of multispecies Staphylococcal biofilms. Appl Environ Microbiol. 2017;83(12):e03483-16.
97.
Zurück zum Zitat Stoodley P, Conti SF, DeMeo PJ, Nistico L, Melton-Kreft R, Johnson S, et al. Characterization of a mixed MRSA/MRSE biofilm in an explanted total ankle arthroplasty. FEMS Immunol Med Microbiol. 2011;62(1):66–74.PubMedCrossRef Stoodley P, Conti SF, DeMeo PJ, Nistico L, Melton-Kreft R, Johnson S, et al. Characterization of a mixed MRSA/MRSE biofilm in an explanted total ankle arthroplasty. FEMS Immunol Med Microbiol. 2011;62(1):66–74.PubMedCrossRef
99.
Zurück zum Zitat Ong PY, Ohtake T, Brandt C, Strickland I, Boguniewicz M, Ganz T, et al. Endogenous antimicrobial peptides and skin infections in atopic dermatitis. N Engl J Med. 2002;347(15):1151–60.PubMedCrossRef Ong PY, Ohtake T, Brandt C, Strickland I, Boguniewicz M, Ganz T, et al. Endogenous antimicrobial peptides and skin infections in atopic dermatitis. N Engl J Med. 2002;347(15):1151–60.PubMedCrossRef
100.
Zurück zum Zitat Powers CE, McShane DB, Gilligan PH, Burkhart CN, Morrell DS. Microbiome and pediatric atopic dermatitis. J Dermatol. 2015;42(12):1137–42.PubMedCrossRef Powers CE, McShane DB, Gilligan PH, Burkhart CN, Morrell DS. Microbiome and pediatric atopic dermatitis. J Dermatol. 2015;42(12):1137–42.PubMedCrossRef
101.
Zurück zum Zitat Nomura I, Goleva E, Howell MD, Hamid QA, Ong PY, Hall CF, et al. Cytokine milieu of atopic dermatitis, as compared to psoriasis, skin prevents induction of innate immune response genes. J Immunol. 2003;171(6):3262–9.PubMedCrossRef Nomura I, Goleva E, Howell MD, Hamid QA, Ong PY, Hall CF, et al. Cytokine milieu of atopic dermatitis, as compared to psoriasis, skin prevents induction of innate immune response genes. J Immunol. 2003;171(6):3262–9.PubMedCrossRef
103.
Zurück zum Zitat •• Nakatsuji T, Chen TH, Narala S, Chun KA, Two AM, Yun T, et al. Antimicrobials from human skin commensal bacteria protect against Staphylococcus aureus and are deficient in atopic dermatitis. Sci Transl Med. 2017;9(378):eaah4680. A study describing AMPs that inhibit S. aureus from strains of coagulase-negative staphylococci common on the skin of healthy individuals but rare in AD patients. PubMedPubMedCentralCrossRef •• Nakatsuji T, Chen TH, Narala S, Chun KA, Two AM, Yun T, et al. Antimicrobials from human skin commensal bacteria protect against Staphylococcus aureus and are deficient in atopic dermatitis. Sci Transl Med. 2017;9(378):eaah4680. A study describing AMPs that inhibit S. aureus from strains of coagulase-negative staphylococci common on the skin of healthy individuals but rare in AD patients. PubMedPubMedCentralCrossRef
104.
Zurück zum Zitat de Koning HD, Kamsteeg M, Rodijk-Olthuis D, van Vlijmen-Willems IM, van Erp PE, Schalkwijk J, et al. Epidermal expression of host response genes upon skin barrier disruption in normal skin and uninvolved skin of psoriasis and atopic dermatitis patients. J Invest Dermatol. 2011;131(1):263–6.PubMedCrossRef de Koning HD, Kamsteeg M, Rodijk-Olthuis D, van Vlijmen-Willems IM, van Erp PE, Schalkwijk J, et al. Epidermal expression of host response genes upon skin barrier disruption in normal skin and uninvolved skin of psoriasis and atopic dermatitis patients. J Invest Dermatol. 2011;131(1):263–6.PubMedCrossRef
105.
Zurück zum Zitat • Scherr TD, Hanke ML, Huang O, James DB, Horswill AR, Bayles KW, et al. Staphylococcus aureus biofilms induce macrophage dysfunction through leukocidin AB and alpha-toxin. MBio. 2015;6(4):e01021. A paper identifying protein factors secreted from S. aureus biofilms that inhibit macrophage phagocytosis, illustrating how S. aureus biofilms can evade host defense. PubMedPubMedCentralCrossRef • Scherr TD, Hanke ML, Huang O, James DB, Horswill AR, Bayles KW, et al. Staphylococcus aureus biofilms induce macrophage dysfunction through leukocidin AB and alpha-toxin. MBio. 2015;6(4):e01021. A paper identifying protein factors secreted from S. aureus biofilms that inhibit macrophage phagocytosis, illustrating how S. aureus biofilms can evade host defense. PubMedPubMedCentralCrossRef
106.
Zurück zum Zitat Scherr TD, Heim CE, Morrison JM, Kielian T. Hiding in plain sight: interplay between Staphylococcal biofilms and host immunity. Front Immunol. 2014;5:37.PubMedPubMedCentralCrossRef Scherr TD, Heim CE, Morrison JM, Kielian T. Hiding in plain sight: interplay between Staphylococcal biofilms and host immunity. Front Immunol. 2014;5:37.PubMedPubMedCentralCrossRef
107.
Zurück zum Zitat Paharik AE, Horswill AR. The staphylococcal biofilm: adhesins, regulation, and host response. Microbiol Spectr. 2016;4(2):VMBF-0022-2015. Paharik AE, Horswill AR. The staphylococcal biofilm: adhesins, regulation, and host response. Microbiol Spectr. 2016;4(2):VMBF-0022-2015.
108.
Zurück zum Zitat Thurlow LR, Hanke ML, Fritz T, Angle A, Aldrich A, Williams SH, et al. Staphylococcus aureus biofilms prevent macrophage phagocytosis and attenuate inflammation in vivo. J Immunol. 2011;186(11):6585–96.PubMedPubMedCentralCrossRef Thurlow LR, Hanke ML, Fritz T, Angle A, Aldrich A, Williams SH, et al. Staphylococcus aureus biofilms prevent macrophage phagocytosis and attenuate inflammation in vivo. J Immunol. 2011;186(11):6585–96.PubMedPubMedCentralCrossRef
109.
Zurück zum Zitat Cerca F, Andrade F, Franca A, Andrade EB, Ribeiro A, Almeida AA, et al. Staphylococcus epidermidis biofilms with higher proportions of dormant bacteria induce a lower activation of murine macrophages. J Med Microbiol. 2011;60(Pt 12):1717–24.PubMedCrossRef Cerca F, Andrade F, Franca A, Andrade EB, Ribeiro A, Almeida AA, et al. Staphylococcus epidermidis biofilms with higher proportions of dormant bacteria induce a lower activation of murine macrophages. J Med Microbiol. 2011;60(Pt 12):1717–24.PubMedCrossRef
110.
Zurück zum Zitat • Tankersley A, Frank MB, Bebak M, Brennan R. Early effects of Staphylococcus aureus biofilm secreted products on inflammatory responses of human epithelial keratinocytes. J Inflamm (Lond). 2014;11:17. A paper demonstrating that S. aureus biofilm conditioned media induces significantly stronger inflammatory responses in human keratinocytes compared to planktonic conditioned media. CrossRef • Tankersley A, Frank MB, Bebak M, Brennan R. Early effects of Staphylococcus aureus biofilm secreted products on inflammatory responses of human epithelial keratinocytes. J Inflamm (Lond). 2014;11:17. A paper demonstrating that S. aureus biofilm conditioned media induces significantly stronger inflammatory responses in human keratinocytes compared to planktonic conditioned media. CrossRef
111.
Zurück zum Zitat Takai T. TSLP expression: cellular sources, triggers, and regulatory mechanisms. Allergol Int. 2012;61(1):3–17.PubMedCrossRef Takai T. TSLP expression: cellular sources, triggers, and regulatory mechanisms. Allergol Int. 2012;61(1):3–17.PubMedCrossRef
112.
Zurück zum Zitat Wilson SR, The L, Batia LM, Beattie K, Katibah GE, McClain SP, et al. The epithelial cell-derived atopic dermatitis cytokine TSLP activates neurons to induce itch. Cell. 2013;155(2):285–95.PubMedPubMedCentralCrossRef Wilson SR, The L, Batia LM, Beattie K, Katibah GE, McClain SP, et al. The epithelial cell-derived atopic dermatitis cytokine TSLP activates neurons to induce itch. Cell. 2013;155(2):285–95.PubMedPubMedCentralCrossRef
113.
Zurück zum Zitat • Son ED, Kim HJ, Park T, Shin K, Bae IH, Lim KM, et al. Staphylococcus aureus inhibits terminal differentiation of normal human keratinocytes by stimulating interleukin-6 secretion. J Dermatol Sci. 2014;74(1):64–71. A study exploring the impact of S. aureus on keratinocyte differentiation, describing the increase in IL-6 and decrease in filaggrin and other differentiation markers upon exposure to S. aureus . PubMedCrossRef • Son ED, Kim HJ, Park T, Shin K, Bae IH, Lim KM, et al. Staphylococcus aureus inhibits terminal differentiation of normal human keratinocytes by stimulating interleukin-6 secretion. J Dermatol Sci. 2014;74(1):64–71. A study exploring the impact of S. aureus on keratinocyte differentiation, describing the increase in IL-6 and decrease in filaggrin and other differentiation markers upon exposure to S. aureus . PubMedCrossRef
114.
Zurück zum Zitat den Reijer PM, Haisma EM, Lemmens-den Toom NA, Willemse J, Koning RI, Demmers JA, et al. Detection of alpha-toxin and other virulence factors in biofilms of Staphylococcus aureus on polystyrene and a human epidermal model. PLoS One. 2016;11(1):e0145722.CrossRef den Reijer PM, Haisma EM, Lemmens-den Toom NA, Willemse J, Koning RI, Demmers JA, et al. Detection of alpha-toxin and other virulence factors in biofilms of Staphylococcus aureus on polystyrene and a human epidermal model. PLoS One. 2016;11(1):e0145722.CrossRef
115.
Zurück zum Zitat Eriksson S, van der Plas MJA, Morgelin M, Sonesson A. Antibacterial and antibiofilm effects of sodium hypochlorite against Staphylococcus aureus isolates derived from patients with atopic dermatitis. Br J Dermatol. 2017;177(2):513–21.PubMedCrossRef Eriksson S, van der Plas MJA, Morgelin M, Sonesson A. Antibacterial and antibiofilm effects of sodium hypochlorite against Staphylococcus aureus isolates derived from patients with atopic dermatitis. Br J Dermatol. 2017;177(2):513–21.PubMedCrossRef
116.
Zurück zum Zitat Batoni G, Maisetta G, Esin S. Antimicrobial peptides and their interaction with biofilms of medically relevant bacteria. Biochim Biophys Acta. 2016;1858(5):1044–60.PubMedCrossRef Batoni G, Maisetta G, Esin S. Antimicrobial peptides and their interaction with biofilms of medically relevant bacteria. Biochim Biophys Acta. 2016;1858(5):1044–60.PubMedCrossRef
117.
118.
Zurück zum Zitat Haisma EM, de Breij A, Chan H, van Dissel JT, Drijfhout JW, Hiemstra PS, et al. LL-37-derived peptides eradicate multidrug-resistant Staphylococcus aureus from thermally wounded human skin equivalents. Antimicrob Agents Chemother. 2014;58(8):4411–9.PubMedPubMedCentralCrossRef Haisma EM, de Breij A, Chan H, van Dissel JT, Drijfhout JW, Hiemstra PS, et al. LL-37-derived peptides eradicate multidrug-resistant Staphylococcus aureus from thermally wounded human skin equivalents. Antimicrob Agents Chemother. 2014;58(8):4411–9.PubMedPubMedCentralCrossRef
119.
Zurück zum Zitat Myles IA, Williams KW, Reckhow JD, Jammeh ML, Pincus NB, Sastalla I, et al. Transplantation of human skin microbiota in models of atopic dermatitis. JCI Insight. 2016;1(10):e86955.PubMedPubMedCentralCrossRef Myles IA, Williams KW, Reckhow JD, Jammeh ML, Pincus NB, Sastalla I, et al. Transplantation of human skin microbiota in models of atopic dermatitis. JCI Insight. 2016;1(10):e86955.PubMedPubMedCentralCrossRef
Metadaten
Titel
Staphylococcal Biofilms in Atopic Dermatitis
verfasst von
Tammy Gonzalez
Jocelyn M. Biagini Myers
Andrew B. Herr
Gurjit K. Khurana Hershey
Publikationsdatum
01.12.2017
Verlag
Springer US
Erschienen in
Current Allergy and Asthma Reports / Ausgabe 12/2017
Print ISSN: 1529-7322
Elektronische ISSN: 1534-6315
DOI
https://doi.org/10.1007/s11882-017-0750-x

Weitere Artikel der Ausgabe 12/2017

Current Allergy and Asthma Reports 12/2017 Zur Ausgabe

Food Allergy (T Green, Section Editor)

Non-IgE-mediated Adverse Food Reactions

Allergic Skin Diseases (L Fonacier, Section Editor)

Pathomechanisms of Contact Sensitization

Basic and Applied Science (I Lewkowich, Section Editor)

Endoplasmic Reticulum Stress and Allergic Diseases

Update HNO

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert – ganz bequem per eMail.