Chronic Pseudomonas aeruginosa biofilm infection impairs murine S100A8/A9 and neutrophil effector cytokines-implications for delayed wound closure?

The impact of Pseudomonas aeruginosa biofilm infections in chronic wounds and clinical implication for healing is receiving increased attention. However, the pathophysiology of host/pathogen interplay is not fully understood. By further revealing the mechanisms, necessary new treatment strategies may be identified. Since the background for chronic wounds is diverse, representative animal models are important. We assessed host response and spontaneous wound closure in the relatively resistant C3H/HeN and the susceptible BALB/c mouse strain. Full-thickness burn wounds were inflicted in 108 mice. Pseudomonas aeruginosa biofilm (106 colony forming units) was injected subcutaneously in 72 mice, euthanised day 4, 7 or 10 days post-infection. Wounds were analysed for neutrophil host response markers: S100A8/A9, keratinocyte-derived chemokine and granulocyte-colony stimulating factor. Total peripheral blood leucocyte and polymorphonuclear count were assessed in parallel. Histopathology evaluated wound inflammatory burden. Photoplanimetry described macroscopical wound closure. Stable chronic wound infection was established in all challenged mice. Pseudomonas aeruginosa biofilm suppressed neutrophil host response in wounds. C3H/HeN mice achieved earlier systemic inflammatory control and healed faster than BALB/c mice. Pseudomonas aeruginosa biofilms perturb host defence thereby inducing a steady state of chronic infection which may impair wound healing. These results indicate therapeutic options for immune modulation of biofilm-infected wounds.

[1]  N. Høiby,et al.  Biofilms and host response – helpful or harmful , 2017, APMIS : acta pathologica, microbiologica, et immunologica Scandinavica.

[2]  A. Schmidtchen,et al.  Pseudomonas aeruginosa elastase cleaves a C-terminal peptide from human thrombin that inhibits host inflammatory responses , 2016, Nature Communications.

[3]  N. Høiby,et al.  S100A8/A9 is an important host defence mediator in neuropathic foot ulcers in patients with type 2 diabetes mellitus , 2016, Archives of Dermatological Research.

[4]  Hans C. Bernstein,et al.  Microsensor and transcriptomic signatures of oxygen depletion in biofilms associated with chronic wounds , 2016, Wound repair and regeneration : official publication of the Wound Healing Society [and] the European Tissue Repair Society.

[5]  Benjamin A Lipsky,et al.  Biofilms and Wounds: An Overview of the Evidence. , 2015, Advances in wound care.

[6]  N. Høiby,et al.  Bactericidal effect of colistin on planktonic Pseudomonas aeruginosa is independent of hydroxyl radical formation. , 2014, International journal of antimicrobial agents.

[7]  Raquel Navarro,et al.  S100A8-S100A9 protein complex mediates psoriasis by regulating the expression of complement factor C3. , 2013, Immunity.

[8]  P. Stewart,et al.  Biofilms and Inflammation in Chronic Wounds. , 2013, Advances in wound care.

[9]  N. Høiby,et al.  Pseudomonas aeruginosa biofilm aggravates skin inflammatory response in BALB/c mice in a novel chronic wound model , 2013, Wound repair and regeneration : official publication of the Wound Healing Society [and] the European Tissue Repair Society.

[10]  T. Standiford,et al.  Genome-wide transcriptional analysis of differentially expressed genes in flagellin-pretreated mouse corneal epithelial cells in response to Pseudomonas aeruginosa: involvement of S100A8/A9 , 2013, Mucosal Immunology.

[11]  N. Høiby,et al.  Bead‐size directed distribution of Pseudomonas aeruginosa results in distinct inflammatory response in a mouse model of chronic lung infection , 2012, Clinical and experimental immunology.

[12]  David W Williams,et al.  A review of the scientific evidence for biofilms in wounds , 2012, Wound repair and regeneration : official publication of the Wound Healing Society [and] the European Tissue Repair Society.

[13]  K. Tracey,et al.  Alarmins: awaiting a clinical response. , 2012, The Journal of clinical investigation.

[14]  R. Gallo,et al.  Antimicrobial peptides: Old Molecules with New Ideas , 2011, The Journal of investigative dermatology.

[15]  L. Jørgensen,et al.  S100A8/A9 deficiency in nonhealing venous leg ulcers uncovered by multiplexed antibody microarray profiling , 2011, The British journal of dermatology.

[16]  T. Bjarnsholt,et al.  Success Rate of Split-Thickness Skin Grafting of Chronic Venous Leg Ulcers Depends on the Presence of Pseudomonas aeruginosa: A Retrospective Study , 2011, PloS one.

[17]  A. Chang,et al.  A Comparison of Wound Area Measurement Techniques: Visitrak Versus Photography , 2011, Eplasty.

[18]  M. Mildner,et al.  The antimicrobial heterodimer S100A8/S100A9 (calprotectin) is upregulated by bacterial flagellin in human epidermal keratinocytes. , 2010, The Journal of investigative dermatology.

[19]  K. Harding,et al.  Non-healing is associated with persistent stimulation of the innate immune response in chronic venous leg ulcers. , 2010, Journal of dermatological science.

[20]  F. Gottrup,et al.  Cost of wound treatment to increase significantly in Denmark over the next decade. , 2010, Journal of wound care.

[21]  T. Tolker-Nielsen,et al.  Nonrandom Distribution of Pseudomonas aeruginosa and Staphylococcus aureus in Chronic Wounds , 2009, Journal of Clinical Microbiology.

[22]  G. Jacobsen,et al.  Thermal injury induces impaired function in polymorphonuclear neutrophil granulocytes and reduced control of burn wound infection , 2009, Clinical and experimental immunology.

[23]  M. Herzberg,et al.  Calprotectin S100A9 Calcium-binding Loops I and II Are Essential for Keratinocyte Resistance to Bacterial Invasion* , 2009, Journal of Biological Chemistry.

[24]  R. Gallo,et al.  Antimicrobial peptides, skin infections, and atopic dermatitis. , 2008, Seminars in cutaneous medicine and surgery.

[25]  T. Tolker-Nielsen,et al.  Distribution, Organization, and Ecology of Bacteria in Chronic Wounds , 2008, Journal of Clinical Microbiology.

[26]  Eric P. Skaar,et al.  Metal Chelation and Inhibition of Bacterial Growth in Tissue Abscesses , 2008, Science.

[27]  P. Williams,et al.  Rapid necrotic killing of polymorphonuclear leukocytes is caused by quorum-sensing-controlled production of rhamnolipid by Pseudomonas aeruginosa. , 2007, Microbiology.

[28]  T. Krieg,et al.  Inflammation in wound repair: molecular and cellular mechanisms. , 2007, The Journal of investigative dermatology.

[29]  D. Hassett,et al.  The Exopolysaccharide Alginate Protects Pseudomonas aeruginosa Biofilm Bacteria from IFN-γ-Mediated Macrophage Killing1 , 2005, The Journal of Immunology.

[30]  A. Kharazmi,et al.  Faster activation of polymorphonuclear neutrophils in resistant mice during early innate response to Pseudomonas aeruginosa lung infection , 2004, Clinical and experimental immunology.

[31]  J. Davidson,et al.  Migration inhibitory factor‐related protein (MRP)8 and MRP14 are differentially expressed in free‐electron laser and scalpel incisions , 2004, Wound repair and regeneration : official publication of the Wound Healing Society [and] the European Tissue Repair Society.

[32]  A. Jesaitis,et al.  Compromised Host Defense on Pseudomonas aeruginosa Biofilms: Characterization of Neutrophil and Biofilm Interactions 1 , 2003, The Journal of Immunology.

[33]  C. Moser,et al.  Cytokine and surface receptor diversity of NK cells in resistant C3H/HeN and susceptible BALB/c mice with chronic Pseudomonas aeruginosa lung infection , 2003, APMIS : acta pathologica, microbiologica, et immunologica Scandinavica.

[34]  S. Werner,et al.  Regulation of wound healing by growth factors and cytokines. , 2003, Physiological reviews.

[35]  I. Thorey,et al.  The Ca2+-binding Proteins S100A8 and S100A9 Are Encoded by Novel Injury-regulated Genes* , 2001, The Journal of Biological Chemistry.

[36]  M. Herzberg,et al.  Calprotectin Expression In Vitro by Oral Epithelial Cells Confers Resistance to Infection by Porphyromonas gingivalis , 2001, Infection and Immunity.

[37]  Shizhong Xu,et al.  Genetic control of the rate of wound healing in mice , 2001, Heredity.

[38]  M. Herzberg,et al.  Calprotectin Expression Inhibits Bacterial Binding to Mucosal Epithelial Cells , 2001, Infection and Immunity.

[39]  W. Payne,et al.  Maintenance of wound bacterial balance. , 1999, American journal of surgery.

[40]  A. Kharazmi,et al.  Early immune response in susceptible and resistant mice strains with chronic Pseudomonas aeruginosa lung infection determines the type of T‐helper cell response , 1999, APMIS : acta pathologica, microbiologica, et immunologica Scandinavica.

[41]  J. Rygaard,et al.  Chronic Pseudomonas aeruginosa lung infection is more severe in Th2 responding BALB/c mice compared to Th1 responding C3H/HeN mice , 1997, APMIS : acta pathologica, microbiologica, et immunologica Scandinavica.

[42]  M. Robson,et al.  Wound Infection: A Failure of Wound Healing Caused by an Imbalance of Bacteria , 1997 .

[43]  S. Cryz,et al.  Vaccination promotes TH1-like inflammation and survival in chronic Pseudomonas aeruginosa pneumonia. A new prophylactic principle. , 1997, Behring Institute Mitteilungen.

[44]  J. Rygaard,et al.  Interferon‐gamma (IFN‐γ) treatment decreases the inflammatory response in chronic Pseudomonas aeruginosa pneumonia in rats , 1996, Clinical and experimental immunology.

[45]  J. Rygaard,et al.  Immunization with Pseudomonas aeruginosa vaccines and adjuvant can modulate the type of inflammatory response subsequent to infection , 1994, Infection and immunity.

[46]  T. Theander,et al.  Dichotomy of the human T cell response to Leishmania antigens. I. Th1‐like response to Leishmania major promastigote antigens in individuals recovered from cutaneous leishmaniasis , 1994, Clinical and experimental immunology.

[47]  G. Pier,et al.  Suppression of lymphocyte and neutrophil functions by Pseudomonas aeruginosa mucoid exopolysaccharide (alginate): reversal by physicochemical, alginase, and specific monoclonal antibody treatments , 1993, Infection and immunity.

[48]  A. Kharazmi,et al.  Mechanisms involved in the evasion of the host defence by Pseudomonas aeruginosa. , 1991, Immunology letters.

[49]  E. Lingaas,et al.  Antimicrobial actions of calcium binding leucocyte L1 protein, calprotectin , 1990, The Lancet.

[50]  M. Röllinghoff,et al.  IL-1 beta is secreted by activated murine macrophages as biologically inactive precursor. , 1990, Journal of immunology.

[51]  P. Brandtzaeg,et al.  Epidermal and dermal distribution of a myelomonocytic antigen (L1) shared by epithelial cells in various inflammatory skin diseases. , 1986, Journal of the American Academy of Dermatology.

[52]  J. Albina,et al.  Wound macrophages as key regulators of repair: origin, phenotype, and function. , 2011, The American journal of pathology.

[53]  K. Krogfelt,et al.  Why chronic wounds will not heal: a novel hypothesis , 2008, Wound repair and regeneration : official publication of the Wound Healing Society [and] the European Tissue Repair Society.