Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Front Immunol ; 12: 695373, 2021.
Article in English | MEDLINE | ID: mdl-34512625

ABSTRACT

Langerhans cells (LCs) reside in the epidermis where they are poised to mount an antimicrobial response against microbial pathogens invading from the outside environment. To elucidate potential pathways by which LCs contribute to host defense, we mined published LC transcriptomes deposited in GEO and the scientific literature for genes that participate in antimicrobial responses. Overall, we identified 31 genes in LCs that encode proteins that contribute to antimicrobial activity, ten of which were cross-validated in at least two separate experiments. Seven of these ten antimicrobial genes encode chemokines, CCL1, CCL17, CCL19, CCL2, CCL22, CXCL14 and CXCL2, which mediate both antimicrobial and inflammatory responses. Of these, CCL22 was detected in seven of nine transcriptomes and by PCR in cultured LCs. Overall, the antimicrobial genes identified in LCs encode proteins with broad antibacterial activity, including against Staphylococcus aureus, which is the leading cause of skin infections. Thus, this study illustrates that LCs, consistent with their anatomical location, are programmed to mount an antimicrobial response against invading pathogens in skin.


Subject(s)
Antimicrobial Peptides/genetics , Epidermis/metabolism , Langerhans Cells/metabolism , Staphylococcal Skin Infections/genetics , Staphylococcus aureus/pathogenicity , Transcriptome , Cells, Cultured , Databases, Genetic , Epidermis/immunology , Epidermis/microbiology , Gene Expression Profiling , Host-Pathogen Interactions , Humans , Langerhans Cells/immunology , Langerhans Cells/microbiology , Staphylococcal Skin Infections/immunology , Staphylococcal Skin Infections/metabolism , Staphylococcal Skin Infections/microbiology , Staphylococcus aureus/immunology
2.
Infect Immun ; 88(8)2020 07 21.
Article in English | MEDLINE | ID: mdl-32513856

ABSTRACT

Staphylococcus aureus fatty acid kinase FakA is necessary for the incorporation of exogenous fatty acids into the lipid membrane. We previously demonstrated that the inactivation of fakA leads to decreased α-hemolysin (Hla) production but increased expression of the proteases SspAB and aureolysin in vitro, and that the ΔfakA mutant causes larger lesions than the wild type (WT) during murine skin infection. As expected, necrosis is Hla dependent in the presence or absence of FakA, as both hla and hla ΔfakA mutants are unable to cause necrosis of the skin. At day 4 postinfection, while the ΔfakA mutant maintains larger and more necrotic abscesses, bacterial numbers are similar to those of the WT, indicating the enhanced tissue damage of mice infected with the ΔfakA mutant is not due to an increase in bacterial burden. At this early stage of infection, skin infected with the ΔfakA mutant has decreased levels of proinflammatory cytokines, such as interleukin-17A (IL-17A) and IL-1α, compared to those of WT-infected skin. At a later stage of infection (day 7), abscess resolution and bacterial clearance are hindered in ΔfakA mutant-infected mice. The paradoxical findings of decreased Hla in vitro but increased necrosis in vivo led us to investigate the role of the proteases regulated by FakA. Utilizing Δaur and ΔsspAB mutants in both the WT and fakA mutant backgrounds, we found that the absence of these proteases in a fakA mutant reduced dermonecrosis to levels similar to those of the WT strain. These studies suggest that the overproduction of proteases is one factor contributing to the enhanced pathogenesis of the ΔfakA mutant during skin infection.


Subject(s)
Bacterial Proteins/immunology , Metalloendopeptidases/immunology , Phosphotransferases (Carboxyl Group Acceptor)/immunology , Serine Endopeptidases/immunology , Skin Ulcer/immunology , Staphylococcal Skin Infections/immunology , Staphylococcus aureus/pathogenicity , Animals , Bacterial Load , Bacterial Proteins/genetics , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Chemokine CCL4/genetics , Chemokine CCL4/immunology , Female , Gene Expression Regulation , Hemolysin Proteins/genetics , Hemolysin Proteins/immunology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-1alpha/genetics , Interleukin-1alpha/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Metalloendopeptidases/deficiency , Metalloendopeptidases/genetics , Mice , Phosphotransferases (Carboxyl Group Acceptor)/deficiency , Phosphotransferases (Carboxyl Group Acceptor)/genetics , Serine Endopeptidases/deficiency , Serine Endopeptidases/genetics , Signal Transduction , Skin/immunology , Skin/microbiology , Skin/pathology , Skin Ulcer/genetics , Skin Ulcer/microbiology , Skin Ulcer/pathology , Staphylococcal Skin Infections/genetics , Staphylococcal Skin Infections/microbiology , Staphylococcal Skin Infections/pathology , Staphylococcus aureus/enzymology , Staphylococcus aureus/genetics , Staphylococcus aureus/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Virulence Factors/genetics , Virulence Factors/immunology
4.
J Clin Invest ; 130(3): 1122-1127, 2020 03 02.
Article in English | MEDLINE | ID: mdl-31873074

ABSTRACT

Staphylococcus aureus remains a leading cause of human infection. These infections frequently recur when the skin is a primary site of infection, especially in infants and children. In contrast, invasive staphylococcal disease is less commonly associated with reinfection, suggesting that tissue-specific mechanisms govern the development of immunity. Knowledge of how S. aureus manipulates protective immunity has been hampered by a lack of antigen-specific models to interrogate the T cell response. Using a chicken egg OVA-expressing S. aureus strain to analyze OVA-specific T cell responses, we demonstrated that primary skin infection was associated with impaired development of T cell memory. Conversely, invasive infection induced antigen-specific memory and protected against reinfection. This defect in adaptive immunity following skin infection was associated with a loss of DCs, attributable to S. aureus α-toxin (Hla) expression. Gene- and immunization-based approaches to protect against Hla during skin infection restored the T cell response. Within the human population, exposure to α-toxin through skin infection may modulate the establishment of T cell-mediated immunity, adversely affecting long-term protection. These studies prompt consideration that vaccination targeting S. aureus may be most effective if delivered prior to initial contact with the organism.


Subject(s)
Bacterial Toxins/immunology , Hemolysin Proteins/immunology , Immunity, Cellular , Immunologic Memory , Staphylococcal Skin Infections/immunology , Staphylococcus aureus/immunology , T-Lymphocytes/immunology , Animals , Bacterial Toxins/genetics , Hemolysin Proteins/genetics , Humans , Mice , Staphylococcal Skin Infections/genetics , Staphylococcal Skin Infections/pathology , Staphylococcal Vaccines/genetics , Staphylococcal Vaccines/immunology , Staphylococcal Vaccines/pharmacology , Staphylococcus aureus/genetics , T-Lymphocytes/pathology
5.
Sci Rep ; 9(1): 1271, 2019 02 04.
Article in English | MEDLINE | ID: mdl-30718644

ABSTRACT

Staphylococcus pseudintermedius is an opportunistic and emerging zoonotic pathogen that primarily colonises the skin of dogs. Many common variants are methicillin resistant (MRSP) or multidrug resistant (MDR), and drug resistance is increasingly reported across the globe. In New Zealand, MRSP isolation remains rare in clinics. To pre-emptively inform diagnostic and antimicrobial stewardship practices, we examine isolates of S. pseudintermedius, MRSP and MDR-MRSP from New Zealand dogs using a combination of methodologies. Genetic and genomic data combined with antimicrobial susceptibility screening identify common drug-resistance profiles and their genetic determinants. We demonstrate that sensitive and specific species-level identification of S. pseudintermedius can be achieved using Bruker MALDI-TOF MS and, further, that this technique can be used to identify some common subtype variants, providing a level of categorical precision that falls somewhere between single-locus and multi-locus sequence typing. Comparative genomics analysis of global S. pseudintermedius data shows that MRSP moves frequently across the globe, but that horizontal gene transfer events resulting in the acquisition of the SCCmec cassette (responsible for beta-lactam antibiotic resistance) are infrequent. This suggests that biosecurity and surveillance in addition to antibiotic stewardship should play important roles in mitigating the risk of MRSP, especially in countries such as New Zealand where MRSP is still rare.


Subject(s)
Dog Diseases , Drug Resistance, Multiple, Bacterial , Genomics , Methicillin Resistance , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Staphylococcal Skin Infections , Staphylococcus , Animals , Dog Diseases/genetics , Dog Diseases/metabolism , Dog Diseases/microbiology , Dogs , New Zealand , Staphylococcal Skin Infections/genetics , Staphylococcal Skin Infections/metabolism , Staphylococcal Skin Infections/veterinary , Staphylococcus/genetics , Staphylococcus/metabolism
6.
Acta Derm Venereol ; 99(2): 181-187, 2019 Feb 01.
Article in English | MEDLINE | ID: mdl-30328471

ABSTRACT

Staphylococcus epidermidis is an abundant skin commensal capable of activating cutaneous defense responses, such as induction of cytokines and antimicrobial peptides. To permanently colonize human skin and prevent inflammation S. epidermidis needs to control the induction of host defense mediators. We report here that S. epidermidis induces expression of the host regulator protein A20 in human keratinocytes, thereby controlling expression and release of interleukin-1 beta. siRNA-mediated knockdown of A20 expression strongly enhanced the induction of interleukin-1 beta gene expression and protein release in keratinocytes stimulated with S. epidermidis. Furthermore, siRNA-mediated knockdown of A20 resulted in enhanced gene expression and secretion of the antimicrobial peptide human beta-defensin-2 in keratinocytes facing S. epidermidis. Mechanistically, A20 negatively controlled S. epidermidis-induced activation of the transcription factor NF-kappaB. Together, these data indicate that S. epidermidis exploits A20 to attenuate cutaneous defense responses, which may help S. epidermidis to persist on human skin.


Subject(s)
Interleukin-1beta/metabolism , Keratinocytes/microbiology , Skin/microbiology , Staphylococcal Skin Infections/microbiology , Staphylococcus epidermidis/pathogenicity , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism , beta-Defensins/metabolism , Cells, Cultured , Host-Pathogen Interactions , Humans , Interleukin-1beta/genetics , Keratinocytes/metabolism , NF-kappa B/metabolism , Signal Transduction , Skin/metabolism , Staphylococcal Skin Infections/genetics , Staphylococcal Skin Infections/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Up-Regulation , beta-Defensins/genetics
7.
PLoS Pathog ; 14(8): e1007244, 2018 08.
Article in English | MEDLINE | ID: mdl-30102746

ABSTRACT

The early events that shape the innate immune response to restrain pathogens during skin infections remain elusive. Methicillin-resistant Staphylococcus aureus (MRSA) infection engages phagocyte chemotaxis, abscess formation, and microbial clearance. Upon infection, neutrophils and monocytes find a gradient of chemoattractants that influence both phagocyte direction and microbial clearance. The bioactive lipid leukotriene B4 (LTB4) is quickly (seconds to minutes) produced by 5-lipoxygenase (5-LO) and signals through the G protein-coupled receptors LTB4R1 (BLT1) or BLT2 in phagocytes and structural cells. Although it is known that LTB4 enhances antimicrobial effector functions in vitro, whether prompt LTB4 production is required for bacterial clearance and development of an inflammatory milieu necessary for abscess formation to restrain pathogen dissemination is unknown. We found that LTB4 is produced in areas near the abscess and BLT1 deficient mice are unable to form an abscess, elicit neutrophil chemotaxis, generation of neutrophil and monocyte chemokines, as well as reactive oxygen species-dependent bacterial clearance. We also found that an ointment containing LTB4 synergizes with antibiotics to eliminate MRSA potently. Here, we uncovered a heretofore unknown role of macrophage-derived LTB4 in orchestrating the chemoattractant gradient required for abscess formation, while amplifying antimicrobial effector functions.


Subject(s)
Abscess/immunology , Bacterial Load/immunology , Leukotriene B4/physiology , Macrophages/metabolism , Methicillin-Resistant Staphylococcus aureus , Staphylococcal Skin Infections/immunology , Abscess/genetics , Abscess/microbiology , Abscess/pathology , Animals , Arachidonate 5-Lipoxygenase/genetics , Bacterial Load/genetics , Cells, Cultured , Female , Leukotriene B4/metabolism , Macrophages/immunology , Male , Methicillin-Resistant Staphylococcus aureus/growth & development , Methicillin-Resistant Staphylococcus aureus/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Leukotriene B4/genetics , Staphylococcal Skin Infections/genetics , Staphylococcal Skin Infections/pathology
8.
Dev Growth Differ ; 60(6): 306-315, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29873073

ABSTRACT

Inflammation at a wound site is essential for preventing infection. However, misregulated inflammation leads to pathologies of the healing process, including chronic non-healing wounds and scarring. MicroRNAs (miRNAs) are key regulators of the inflammatory response and tissue repair, acting by translational processing of target mRNAs. In the final step of miRNA processing, Argonaute 2 (Ago2)-bound mature miRNA complexes bind to target mRNAs and inhibit their translation. A variety of wound healing-related miRNAs have been identified and their misregulation likely contributes to wound pathologies, including scarring and chronic healing. Recently, we have developed an Ago2-bound mature miRNA purification system that uses Ago2 antibody to analyze the expression of miRNAs from wound tissues by microarray and next generation sequencing. We have identified several wound inflammation-related miRNAs via Ago2-target immunoprecipitation assays and next generation sequencing of wound tissues from wild-type and PU.1 knockout mice, which exhibit no inflammatory response because of a lack of immune cell lineages. We demonstrated that miR-142, an identified inflammation-related miRNA, is essential role for neutrophilic chemotaxis via inhibition of small GTPase translation; its misregulation leads to susceptibility to infection against Staphylococcus aureus at skin wound sites. In this review, we summarize recent advances of miRNA studies in skin wound healing, introduce our miRNA purification system using an immunoprecipitation assay method, and discuss the function of miR-142 in skin wound healing.


Subject(s)
MicroRNAs/metabolism , Skin/metabolism , Staphylococcal Skin Infections/metabolism , Staphylococcus aureus , Wound Healing , Wound Infection/metabolism , Wounds and Injuries/metabolism , Animals , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Humans , Mice , Mice, Knockout , MicroRNAs/genetics , Skin/injuries , Staphylococcal Skin Infections/genetics , Staphylococcal Skin Infections/pathology , Wound Infection/genetics , Wound Infection/microbiology , Wound Infection/pathology , Wounds and Injuries/genetics , Wounds and Injuries/pathology
9.
PLoS Pathog ; 14(3): e1006907, 2018 03.
Article in English | MEDLINE | ID: mdl-29554137

ABSTRACT

Staphylococcus aureus exhibits many defenses against host innate immunity, including the ability to replicate in the presence of nitric oxide (NO·). S. aureus NO· resistance is a complex trait and hinges on the ability of this pathogen to metabolically adapt to the presence of NO·. Here, we employed deep sequencing of transposon junctions (Tn-Seq) in a library generated in USA300 LAC to define the complete set of genes required for S. aureus NO· resistance. We compared the list of NO·-resistance genes to the set of genes required for LAC to persist within murine skin infections (SSTIs). In total, we identified 168 genes that were essential for full NO· resistance, of which 49 were also required for S. aureus to persist within SSTIs. Many of these NO·-resistance genes were previously demonstrated to be required for growth in the presence of this immune radical. However, newly defined genes, including those encoding SodA, MntABC, RpoZ, proteins involved with Fe-S-cluster repair/homeostasis, UvrABC, thioredoxin-like proteins and the F1F0 ATPase, have not been previously reported to contribute to S. aureus NO· resistance. The most striking finding was that loss of any genes encoding components of the F1F0 ATPase resulted in mutants unable to grow in the presence of NO· or any other condition that inhibits cellular respiration. In addition, these mutants were highly attenuated in murine SSTIs. We show that in S. aureus, the F1F0 ATPase operates in the ATP-hydrolysis mode to extrude protons and contribute to proton-motive force. Loss of efficient proton extrusion in the ΔatpG mutant results in an acidified cytosol. While this acidity is tolerated by respiring cells, enzymes required for fermentation cannot operate efficiently at pH ≤ 7.0 and the ΔatpG mutant cannot thrive. Thus, S. aureus NO· resistance requires a mildly alkaline cytosol, a condition that cannot be achieved without an active F1F0 ATPase enzyme complex.


Subject(s)
Bacterial Proteins/genetics , Immunity, Innate/immunology , Nitric Oxide/pharmacology , Staphylococcal Skin Infections/immunology , Staphylococcus aureus/drug effects , Virulence/immunology , Animals , Gene Expression Regulation, Bacterial , Gene Library , Immunity, Innate/drug effects , Immunity, Innate/genetics , Mice , Mice, Inbred C57BL , Staphylococcal Skin Infections/genetics , Staphylococcal Skin Infections/microbiology , Staphylococcus aureus/immunology , Virulence/drug effects , Virulence/genetics
10.
J Infect Dis ; 218(5): 791-800, 2018 07 24.
Article in English | MEDLINE | ID: mdl-29329449

ABSTRACT

Background: Lack of receptor for advanced glycation end products (RAGE) ameliorates several infections including Staphylococcus aureus pneumonia. We sought to investigate the role of RAGE in staphylococcal skin infection in mice. Methods: Wild-type (WT) and RAGE deficient (RAGE-/-) mice were subcutaneously inoculated with S. aureus SH1000 strain in abscess-forming dose or necrotic dose. Clinical signs of dermatitis, along with histopathological changes, were compared between the groups. Results: The skin lesion size was smaller in RAGE-/- mice. Infected RAGE-/- mice expressed lower proinflammatory cytokines in local skins compared to control mice. Low dose of bacteria caused more abscess formation in RAGE-/- mice compared to skin necrosis that was more often observed in WT mice. As a result of more abscess formation, the wound healing was prolonged in RAGE-/- mice. Importantly, RAGE-/- mice had lower bacterial loads in the skin than controls, which is correlated with higher local levels of myeloperoxidase before skin infection. In vitro, enhanced phagocytic capacity of neutrophils and macrophages obtained from RAGE-/- mice compared to control mice was observed. Conclusions: RAGE deficiency up-regulates phagocytic capacity of phagocytes, resulting in lower bacterial burden in local skin and milder skin lesions in mice with staphylococcal skin infection.


Subject(s)
Abscess/pathology , Receptor for Advanced Glycation End Products/deficiency , Skin/pathology , Staphylococcal Skin Infections/pathology , Wound Healing , Abscess/genetics , Animals , Bacterial Load , Cytokines/analysis , Disease Models, Animal , Female , Histocytochemistry , Macrophages/immunology , Male , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Phagocytosis , Receptor for Advanced Glycation End Products/metabolism , Staphylococcal Skin Infections/genetics
11.
J Immunol ; 200(2): 657-668, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29222165

ABSTRACT

Numerous studies have reported sex bias in infectious diseases, with bias direction dependent on pathogen and site of infection. Staphylococcus aureus is the most common cause of skin and soft tissue infections (SSTIs), yet sex bias in susceptibility to S. aureus SSTI has not been described. A search of electronic health records revealed an odds ratio of 2.4 for S. aureus SSTI in males versus females. To investigate the physiological basis of this bias, we compared outcomes between male and female mice in a model of S. aureus dermonecrosis. Consistent with the epidemiological data, female mice were better protected against SSTI, with reduced dermonecrosis followed later by increased bacterial clearance. Protection in females was disrupted by ovariectomy and restored by short-term estrogen administration. Importantly, this sex bias was mediated by a sex-specific response to the S. aureus-secreted virulence factor α-hemolysin (Hla). Infection with wild-type S. aureus suppressed inflammatory cytokine production in the skin of female, but not male, mice when compared with infection with an isogenic hla deletion mutant. This differential response was conserved following injection with Hla alone, demonstrating a direct response to Hla independent of bacterial burden. Additionally, neutrophils, essential for clearing S. aureus, demonstrated sex-specific S. aureus bactericidal capacity ex vivo. This work suggests that sex-specific skin innate responsiveness to Hla and neutrophil bactericidal capacity play important roles in limiting S. aureus SSTI in females. Understanding the molecular mechanisms controlling this sex bias may reveal novel targets to promote host innate defense against S. aureus skin infection.


Subject(s)
Bacterial Toxins/metabolism , Hemolysin Proteins/metabolism , Staphylococcal Skin Infections/microbiology , Staphylococcus aureus/pathogenicity , Animals , Cytokines/metabolism , Disease Models, Animal , Disease Resistance , Estrogens/metabolism , Female , Gene Expression , Immunity, Innate , Inflammasomes/metabolism , Inflammation Mediators , Male , Mice , Microbial Viability/immunology , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/microbiology , Sex Factors , Staphylococcal Skin Infections/genetics , Staphylococcal Skin Infections/immunology , Staphylococcal Skin Infections/metabolism , Virulence , Virulence Factors
12.
Comp Med ; 67(4): 344-349, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28830581

ABSTRACT

Staphylococcus xylosus is a commensal bacterium found on the skin and mucosal surfaces of SPF mice. S. xylosus is rarely pathogenic, most often causing skin lesions and dermatitis in immunocompromised mice, particularly those with impaired NADPH oxidase function. Here we report spontaneous infection with S. xylosus in Rag1-/-Tpl2-/- mice. Infection was characterized by the presence of alopecia, crusts, and scaly skin. S. xylosus was detected in the feces, skin, lymph nodes, and lungs of Rag1-/-Tpl2-/- mice and led to mortality or euthanasia due to humane endpoints. C57BL/6 mice were culture-positive for S. xylosus on the skin, and Rag1-/- and Tpl2-/- mice were culture-positive on the skin and occasionally in the feces. However, S. xylosus did not cause clinical symptoms in C57BL/6, Rag1-/-, or Tpl2-/- mice. Compared with those in Rag1-/- mice, relative concentrations of circulating monocytes, but not neutrophils or lymphocytes, were increased in Rag1-/-Tpl2-/- mice, consistent with their increased incidence of clinical symptoms. Overall, this case study suggests a novel role for Tpl2 in T-cell-independent host resistance to the otherwise commensal organism S. xylosus.


Subject(s)
Dermatitis/veterinary , Homeodomain Proteins/genetics , Immunocompromised Host , MAP Kinase Kinase Kinases/genetics , Opportunistic Infections/veterinary , Proto-Oncogene Proteins/genetics , Skin/microbiology , Staphylococcal Skin Infections/veterinary , Staphylococcus/pathogenicity , Animals , Bacterial Translocation , Dermatitis/genetics , Dermatitis/immunology , Dermatitis/microbiology , Feces/microbiology , Genetic Predisposition to Disease , Host-Pathogen Interactions , MAP Kinase Kinase Kinases/deficiency , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Monocytes/microbiology , Opportunistic Infections/genetics , Opportunistic Infections/immunology , Opportunistic Infections/microbiology , Phenotype , Proto-Oncogene Proteins/deficiency , Skin/immunology , Skin/pathology , Staphylococcal Skin Infections/genetics , Staphylococcal Skin Infections/immunology , Staphylococcal Skin Infections/microbiology , Staphylococcus/classification , Staphylococcus/immunology
13.
Br J Dermatol ; 177(5): 1394-1400, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28317091

ABSTRACT

BACKGROUND: Atopic dermatitis (AD) is a prevalent disease with significant impact on physical health and quality of life. Staphylococcus aureus has been directly correlated to disease severity, and may also be a contributing causal factor in the pathogenesis of AD. OBJECTIVES: The primary aim was to assess differences in S. aureus colonization in patients with AD with and without filaggrin gene mutations. The secondary aim was to assess disease severity in relation to S. aureus colonization. Exploratory analyses were performed to investigate S. aureus genetic lineages in relation to filaggrin gene (FLG) mutations and disease severity. METHODS: Adult patients with AD (n = 101) were included in the study. Bacterial swabs were taken from lesional skin, nonlesional skin and the nose. Swabs positive for S. aureus were characterized by spa and the respective clonal complex (CC) type assigned. Patients were characterized with respect to disease severity (Scoring Atopic Dermatitis) and FLG mutations (n = 88). Fisher's exact test was used to analyse differences in S. aureus colonization in relation to FLG mutations. RESULTS: Of the 101 patients included, 74 (73%) were colonized with S. aureus. Of the colonized patients, 70 (95%) carried only one CC type in all three different sampling sites. In lesional skin, S. aureus was found in 24 of 31 patients with FLG mutations vs. 24 of 54 wild-type patients (P = 0·0004). Staphylococcus aureusCC1 clonal lineage was more prevalent in patients with FLG mutations (n = 10) than in wild-type patients (n = 2) (P = 0·003). No specific bacterial lineage was linked to disease severity. CONCLUSIONS: Increased S. aureus colonization in patients with AD with FLG mutations, and increased prevalence of CC1 in patients with FLG mutations, suggest that host-microbe interactions and clonal differences in S. aureus are important for colonization of AD skin.


Subject(s)
Dermatitis, Atopic/microbiology , Host-Pathogen Interactions/genetics , Intermediate Filament Proteins/genetics , Mutation/genetics , Nose Diseases/microbiology , Staphylococcal Infections/genetics , Adult , Aged , Bacterial Proteins/metabolism , Dermatitis, Atopic/genetics , Female , Filaggrin Proteins , Humans , Male , Middle Aged , Nose Diseases/genetics , Staphylococcal Skin Infections/genetics , Staphylococcus aureus/classification , Young Adult
14.
Cell Rep ; 16(8): 2219-2230, 2016 08 23.
Article in English | MEDLINE | ID: mdl-27524612

ABSTRACT

Staphylococcus aureus triggers inflammation through inflammasome activation and recruitment of neutrophils, responses that are critical for pathogen clearance but are associated with substantial tissue damage. We postulated that necroptosis, cell death mediated by the RIPK1/RIPK3/MLKL pathway, would function to limit pathological inflammation. In models of skin infection or sepsis, Mlkl-/- mice had high bacterial loads, an inability to limit interleukin-1b (IL-1b) production, and excessive inflammation. Similarly, mice treated with RIPK1 or RIPK3 inhibitors had increased bacterial loads in a model of sepsis. Ripk3-/- mice exhibited increased staphylococcal clearance and decreased inflammation in skin and systemic infection, due to direct effects of RIPK3 on IL-1b activation and apoptosis. In contrast to Casp1/4-/- mice with defective S. aureus killing, the poor outcomes of Mlkl-/- mice could not be attributed to impaired phagocytic function. We conclude that necroptotic cell death limits the pathological inflammation induced by S. aureus.


Subject(s)
Apoptosis/immunology , Necrosis/immunology , Protein Kinases/immunology , Sepsis/immunology , Staphylococcal Infections/immunology , Staphylococcal Skin Infections/immunology , Staphylococcus aureus/immunology , Animals , Bacterial Load , Caspase 1/genetics , Caspase 1/immunology , Caspases/genetics , Caspases/immunology , Caspases, Initiator , Cell Line , Gene Expression Regulation , HEK293 Cells , Humans , Inflammasomes/genetics , Inflammasomes/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Keratinocytes , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Kinases/deficiency , Protein Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Sepsis/genetics , Sepsis/mortality , Sepsis/pathology , Signal Transduction , Staphylococcal Infections/genetics , Staphylococcal Infections/pathology , Staphylococcal Skin Infections/genetics , Staphylococcal Skin Infections/pathology , Staphylococcus aureus/growth & development , Staphylococcus aureus/pathogenicity , Survival Analysis
17.
J Chemother ; 28(5): 359-66, 2016 Oct.
Article in English | MEDLINE | ID: mdl-26027683

ABSTRACT

The objective was to analyse the genetic lineages of Staphylococcus aureus recovered from nasal and skin samples of atopic dermatitis (AD) paediatric patients, and to characterize the antimicrobial resistance phenotype-genotype and the immune-evasion-cluster (IEC) type of isolates. Forty S. aureus isolates from 35 patients (skin: 26; nasal samples: 14) were characterized. Isolates were submitted to spa-, agr- and multilocus sequence typing. All S. aureus strains analyzed were methicillin-susceptible (MSSA). High genetic diversity was detected among the 40 MSSA isolates (especially among skin isolates), with detection of 27 different spa-types, 20 sequence-types and 16 clonal complexes (CCs). Lineages CC30 and CC5 were predominant among nasal isolates (71% vs 23% skin). Thirteen different CCs were detected among skin isolates, with detection of clades CC1, CC9 and CC398. Antimicrobial resistance rates detected were higher in skin than in nasal isolates, especially for macrolides, aminoglycosides, lincosamides and mupirocin. MSSA strains were characterized into five IEC-types, being A, B and F the predominant ones. MSSA strains of lineages CC45 and CC5 were detected in almost all cases in AD patients with severe Scoring Atopic Dermatitis (SCORAD) and lineages CC8, and CC30 in those with mild or moderate one. As conclusion, high-clonal-diversity was detected among MSSA from AD patients, especially in skin-isolates. Colonization with S. aureus of some CCs seems more associated with AD severity than other lineages.


Subject(s)
Dermatitis, Atopic/microbiology , Drug Resistance, Bacterial/genetics , Staphylococcal Skin Infections/genetics , Staphylococcal Skin Infections/microbiology , Staphylococcus aureus/genetics , Child , Female , Genotype , Humans , Male , Staphylococcal Skin Infections/complications
18.
Infect Immun ; 83(11): 4427-37, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26351278

ABSTRACT

Staphylococcus aureus is the leading cause of skin and skin structure infections (SSSI) in humans. Moreover, the high frequency of recurring SSSI due to S. aureus, particularly methicillin-resistant S. aureus (MRSA) strains, suggests that infection induces suboptimal anamnestic defenses. The present study addresses the hypothesis that interleukin-17A (IL-17A) and IL-22 play distinct roles in immunity to cutaneous and invasive MRSA infection in a mouse model of SSSI. Mice were treated with specific neutralizing antibodies against IL-17A and/or IL-22 and infected with MRSA, after which the severity of infection and host immune response were determined. Neutralization of either IL-17A or IL-22 reduced T cell and neutrophil infiltration and host defense peptide elaboration in lesions. These events corresponded with increased abscess severity, MRSA viability, and CFU density in skin. Interestingly, combined inhibition of IL-17A and IL-22 did not worsen abscesses but did increase gamma interferon (IFN-γ) expression at these sites. The inhibition of IL-22 led to a reduction in IL-17A expression, but not vice versa. These results suggest that the expression of IL-17A is at least partially dependent on IL-22 in this model. Inhibition of IL-17A but not IL-22 led to hematogenous dissemination to kidneys, which correlated with decreased T cell infiltration in renal tissue. Collectively, these findings indicate that IL-17A and IL-22 have complementary but nonredundant roles in host defense against cutaneous versus hematogenous infection. These insights may support targeted immune enhancement or other novel approaches to address the challenge of MRSA infection.


Subject(s)
Hematologic Diseases/immunology , Interleukin-17/immunology , Interleukins/immunology , Methicillin-Resistant Staphylococcus aureus/immunology , Staphylococcal Skin Infections/immunology , Animals , Hematologic Diseases/genetics , Hematologic Diseases/microbiology , Humans , Interleukin-17/genetics , Interleukins/genetics , Male , Methicillin-Resistant Staphylococcus aureus/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Staphylococcal Skin Infections/genetics , Staphylococcal Skin Infections/microbiology , Interleukin-22
19.
Biochemistry ; 54(31): 4855-62, 2015 Aug 11.
Article in English | MEDLINE | ID: mdl-26177220

ABSTRACT

Staphylococcus aureus is a leading cause of surgical site infections that results in increased hospital stays due to the development of chronic wounds. Little is known about factors involved in S. aureus' ability to prevent wounds from healing. We discovered a novel secreted protein produced by a surgical site isolate of S. aureus that prevents keratinocyte proliferation. The protein has a molecular weight of 15.7 kDa and an isoelectric point of 8.9. The cloned and purified protein has cytotoxic and proinflammatory properties, as shown in vitro and in vivo. Potent biological effects on keratinocytes and rabbit skin suggest that this protein may play an important role in preventing re-epithelialization. Its lack of homology to known exotoxins suggests that this protein is novel, and this observation is likely to open a new field of research in S. aureus exotoxins. Due to its cytotoxic activities, we call this new protein ε-cytotoxin.


Subject(s)
Bacterial Proteins/metabolism , Cell Proliferation , Keratinocytes/metabolism , Staphylococcal Skin Infections/metabolism , Staphylococcus aureus/metabolism , Animals , Bacterial Proteins/genetics , Cell Line, Transformed , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/microbiology , Inflammation/pathology , Keratinocytes/pathology , Rabbits , Staphylococcal Skin Infections/genetics , Staphylococcal Skin Infections/pathology , Staphylococcus aureus/genetics , Staphylococcus aureus/pathogenicity
20.
Microbes Infect ; 17(9): 622-7, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26086798

ABSTRACT

The receptor for advanced glycation endproducts (RAGE) has been implicated in the regulation of skin inflammation. We here sought to study the role of RAGE in host defense during skin infection caused by Staphylococcus (S.) aureus, the most common pathogen in this condition. Wild-type (Wt) and RAGE deficient (rage(-/-)) mice were infected subcutaneously with S. aureus and bacterial loads and local inflammation were quantified at regular intervals up to 8 days after infection. While bacterial burdens were similar in both mouse strains at the primary site of infection, rage(-/-) mice had lower bacterial counts in lungs and liver. Skin cytokine and chemokine levels did not differ between groups. In accordance with the skin model, direct intravenous infection with S. aureus was associated with lower bacterial loads in lungs and liver of rage(-/-) mice. Together these data suggest that RAGE does not impact local host defense during S. aureus skin infection, but facilitates bacterial growth at distant body sites.


Subject(s)
Host-Pathogen Interactions/immunology , Receptor for Advanced Glycation End Products/immunology , Staphylococcal Skin Infections/immunology , Staphylococcal Skin Infections/microbiology , Animals , Bacteremia/microbiology , Cytokines , Host-Pathogen Interactions/genetics , Immunity, Innate , Liver/microbiology , Lung/microbiology , Mice , Mice, Knockout , Neutrophils , Receptor for Advanced Glycation End Products/genetics , Receptor for Advanced Glycation End Products/metabolism , Staphylococcal Skin Infections/genetics
SELECTION OF CITATIONS
SEARCH DETAIL