Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Immun Ageing ; 17: 12, 2020.
Article in English | MEDLINE | ID: mdl-32467712

ABSTRACT

OBJECTIVE: Exercise can improve immune health and is beneficial for physical function in patients with rheumatoid arthritis (RA), but the immunological mechanisms are largely unknown. We evaluated the effect of moderate- to high intensity exercise with person-centred guidance on cells of the immune system, with focus on regulatory cell populations, in older adults with RA. METHODS: Older adults (≥65 years) with RA were randomized to either 20-weeks of moderate - to high intensity aerobic and resistance exercise (n = 24) or to an active control group performing home-based exercise of light intensity (n = 25). Aerobic capacity, muscle strength, DAS28 and CRP were evaluated. Blood samples were collected at baseline and after 20 weeks. The frequency of immune cells defined as adaptive regulatory populations, CD4 + Foxp3 + CD25 + CD127- T regulatory cells (Tregs) and CD19 + CD24hiCD38hi B regulatory cells (Bregs) as well as HLA-DR-/lowCD33 + CD11b + myeloid derived suppressor cells (MDSCs), were assessed using flow cytometry. RESULTS: After 20 weeks of moderate- to high intensity exercise, aerobic capacity and muscle strength were significantly improved but there were no significant changes in Disease Activity Score 28 (DAS28) or CRP. The frequency of Tregs and Bregs decreased significantly in the intervention group, but not in the active control group. The exercise intervention had no effect on MDSCs. The reduction in regulatory T cells in the intervention group was most pronounced in the female patients. CONCLUSION: Moderate- to high intensity exercise in older adults with RA led to a decreased proportion of Tregs and Bregs, but that was not associated with increased disease activity or increased inflammation. TRIAL REGISTRATION: Improved Ability to Cope With Everyday Life Through a Person-centered Training Program in Elderly Patients With Rheumatoid Arthritis - PEP-walk Study, NCT02397798. Registered at ClinicalTrials.gov March 19, 2015.

2.
Infect Dis (Lond) ; 51(6): 446-451, 2019 06.
Article in English | MEDLINE | ID: mdl-30985241

ABSTRACT

BACKGROUND: Biofilms are involved in many Staphylococcus aureus infections, but relation of biofilm formation and the infection types or the clinical outcomes remain unclear. METHODS: We measured biofilm formation, with a microtiter plate assay, of a collection of methicillin-sensitive clinical isolates from 159 invasive S. aureus infections, encompassing all cases occurring within a hospital catchment area during two years, and from additional 49 non-invasive skin infections from the same region. Results were related to available clinical and microbiological documentation. RESULTS: Isolates from medical device infections (intravenous line-associated and prosthetic joint infections), as well as isolates from superficial skin infections, were particularly proficient in forming biofilms. No increased biofilm-forming capacity was seen in isolates from endocarditis, osteomyelitis, or other infections. There was also a correlation of biofilm formation with the agr type of the isolates. Thicker biofilms were more resistant to antibiotic treatment in vitro. No correlation between biofilm formation and clinical outcomes was noted. CONCLUSIONS: S. aureus isolates from 'classical' biofilm-related infections, but also from superficial skin infections, are especially proficient in forming biofilms. There is, however, no obvious relation of biofilm-forming capacity of isolates and the clinical outcome of the infection, and more studies on this issue are needed.


Subject(s)
Biofilms/growth & development , Staphylococcal Infections/microbiology , Staphylococcus aureus/physiology , Staphylococcus aureus/pathogenicity , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Genotype , Humans , Methicillin-Resistant Staphylococcus aureus , Skin Diseases/microbiology , Staphylococcus aureus/classification , Trans-Activators/genetics
3.
Infect Immun ; 85(7)2017 07.
Article in English | MEDLINE | ID: mdl-28438975

ABSTRACT

Staphylococcus aureus is a major cause of skin and soft tissue infection. The bacterium expresses four major proteases that are emerging as virulence factors: aureolysin (Aur), V8 protease (SspA), staphopain A (ScpA), and staphopain B (SspB). We hypothesized that human galectin-3, a ß-galactoside-binding lectin involved in immune regulation and antimicrobial defense, is a target for these proteases and that proteolysis of galectin-3 is a novel immune evasion mechanism. Indeed, supernatants from laboratory strains and clinical isolates of S. aureus caused galectin-3 degradation. Similar proteolytic capacities were found in Staphylococcus epidermidis isolates but not in Staphylococcus saprophyticus Galectin-3-induced activation of the neutrophil NADPH oxidase was abrogated by bacterium-derived proteolysis of galectin-3, and SspB was identified as the major protease responsible. The impact of galectin-3 and protease expression on S. aureus virulence was studied in a murine skin infection model. In galectin-3+/+ mice, SspB-expressing S. aureus caused larger lesions and resulted in higher bacterial loads than protease-lacking bacteria. No such difference in bacterial load or lesion size was detected in galectin-3-/- mice, which overall showed smaller lesion sizes than the galectin-3+/+ animals. In conclusion, the staphylococcal protease SspB inactivates galectin-3, abrogating its stimulation of oxygen radical production in human neutrophils and increasing tissue damage during skin infection.


Subject(s)
Bacterial Proteins/metabolism , Galectin 3/metabolism , Host-Pathogen Interactions , Serine Endopeptidases/metabolism , Staphylococcus aureus/enzymology , Staphylococcus aureus/pathogenicity , Animals , Bacterial Load , Blood Proteins , Disease Models, Animal , Galectins , Humans , Mice, Inbred C57BL , Mice, Knockout , Proteolysis , Staphylococcal Skin Infections/microbiology , Staphylococcal Skin Infections/pathology , Virulence
4.
PLoS One ; 12(3): e0173492, 2017.
Article in English | MEDLINE | ID: mdl-28264025

ABSTRACT

BACKGROUND: RA patients being treated with biologics are known to have an increased risk of infections. We recently demonstrated that both CTLA4 Ig and anti-TNF treatment aggravate systemic Staphylococcus aureus (S. aureus) infection in mice, but with distinct clinical manifestations. However, the effects of CTLA4 Ig and anti-TNF treatments on a local S. aureus infection (e.g., skin infection) might differ from their effects on a systemic infection. AIMS: The aim of this study was to examine the differential effects of anti-TNF versus CTLA4 Ig treatment on S. aureus skin infections in mice. METHOD: Abatacept (CTLA4 Ig), etanercept (anti-TNF treatment) or PBS was given to NMRI mice subcutaneously inoculated with S. aureus strain SH1000. The clinical signs of dermatitis, along with histopathological changes due to skin infection, were compared between the groups. RESULTS: Both CTLA4 Ig and anti-TNF treatment resulted in less severe skin infections and smaller post-infectious hyperpigmentation compared with controls. Consistent with the clinical signs of dermatitis, smaller lesion size, more epithelial hyperplasia and more granulation were found in skin biopsies from mice receiving anti-TNF compared with PBS controls. However, both CTLA4 Ig and anti-TNF therapy tended to prolong the healing time, although this finding was not statistically significant. Serum MCP-1 levels were elevated in the anti-TNF group relative to the CTLA4 Ig and PBS groups, whereas IL-6 levels were higher in PBS controls than in the other two groups. Both anti-TNF and CTLA4 Ig treatments tended to down-regulate the necrosis/apoptosis ratio in the locally infected skin tissue. Importantly, no tangible difference was found in the bacterial burden among groups. CONCLUSION: Both CTLA4 Ig and anti-TNF therapies attenuate disease severity but may prolong the healing time required for S. aureus skin infections. Neither treatment has an impact on bacterial clearance in skin tissues.


Subject(s)
Abatacept/pharmacology , CTLA-4 Antigen/antagonists & inhibitors , Dermatitis/microbiology , Etanercept/pharmacology , Staphylococcal Infections/microbiology , Staphylococcus aureus/drug effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Apoptosis , Bacterial Load , Cytokines/blood , Cytokines/metabolism , Dermatitis/drug therapy , Dermatitis/immunology , Dermatitis/metabolism , Disease Models, Animal , Female , Hyperpigmentation , Immunosuppressive Agents/pharmacology , Mice , Monocytes/immunology , Monocytes/metabolism , Monocytes/pathology , Necrosis , Phenotype , Severity of Illness Index , Staphylococcal Infections/drug therapy , Staphylococcal Infections/immunology , Staphylococcal Infections/metabolism , Staphylococcus aureus/immunology
5.
J Infect Dis ; 213(1): 139-48, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26136471

ABSTRACT

Staphylococcus aureus biofilms, a leading cause of persistent infections, are highly resistant to immune defenses and antimicrobial therapies. In the present study, we investigated the contribution of fibrin and staphylokinase (Sak) to biofilm formation. In both clinical S. aureus isolates and laboratory strains, high Sak-producing strains formed less biofilm than strains that lacked Sak, suggesting that Sak prevents biofilm formation. In addition, Sak induced detachment of mature biofilms. This effect depended on plasminogen activation by Sak. Host-derived fibrin, the main substrate cleaved by Sak-activated plasminogen, was a major component of biofilm matrix, and dissolution of this fibrin scaffold greatly increased susceptibility of biofilms to antibiotics and neutrophil phagocytosis. Sak also attenuated biofilm-associated catheter infections in mouse models. In conclusion, our results reveal a novel role for Sak-induced plasminogen activation that prevents S. aureus biofilm formation and induces detachment of existing biofilms through proteolytic cleavage of biofilm matrix components.


Subject(s)
Biofilms/drug effects , Metalloendopeptidases/metabolism , Staphylococcus aureus/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/drug effects , Female , Fibrin/metabolism , Male , Metalloendopeptidases/pharmacology , Mice , Mice, Inbred C57BL , Plasminogen/metabolism , Staphylococcus aureus/drug effects
6.
PLoS One ; 10(7): e0131645, 2015.
Article in English | MEDLINE | ID: mdl-26135738

ABSTRACT

BACKGROUND: Interleukin-1 receptor antagonist (IL-1Ra) is the primary therapy against autoinflammatory syndromes with robust efficacy in reducing systemic inflammation and associated organ injury. However, patients receiving IL-1Ra might be at increased risk of acquiring serious infections. AIMS: To study whether IL-1Ra treatment deteriorates Staphylococcus aureus (S. aureus) septic arthritis and sepsis in mice. METHOD: NMRI mice were treated with anakinra (IL-1Ra) daily for 7 days before intravenous inoculation with S. aureus strain Newman in both arthritogenic and lethal doses. The clinical course of septic arthritis, histopathological and radiological changes of the joints, as well as the mortality were compared between IL-1Ra treated and control groups. RESULTS: IL-1Ra treated mice developed more frequent and severe clinical septic arthritis. Also, the frequency of polyarthritis was significantly higher in the mice receiving IL-1Ra therapy. In line with the data from clinical arthritis, both histological and radiological signs of septic arthritis were more pronounced in IL-1Ra treated group compared to controls. Importantly, the mortality of IL-1Ra treated mice was significantly higher than PBS treated controls. CONCLUSION: IL-1Ra treatment significantly aggravated S. aureus induced septic arthritis and increased the mortality in these mice.


Subject(s)
Arthritis, Infectious/drug therapy , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Sepsis/drug therapy , Staphylococcal Infections/drug therapy , Animals , Arthritis, Experimental/drug therapy , Arthritis, Experimental/microbiology , Arthritis, Infectious/microbiology , Cytokines/metabolism , Disease Models, Animal , Female , Inflammation/drug therapy , Inflammation/metabolism , Joints/pathology , Kidney/pathology , Macrophages, Peritoneal/metabolism , Mice , Sepsis/microbiology , Staphylococcal Infections/microbiology , Staphylococcus , Staphylococcus aureus , X-Ray Microtomography
7.
J Infect Dis ; 212(8): 1308-16, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-25838268

ABSTRACT

BACKGROUND: The development of biologics has greatly increased the quality of life and the life expectancy of many patients with rheumatoid arthritis. However, a large number of these patients have an increased risk of developing serious infections. The aim of this study was to examine differential effects of anti-tumor necrosis factor (TNF) treatment and CTLA4 immunoglobulin (Ig) treatment on both immunological response and host defense in a murine model of septic arthritis. METHODS: Abatacept (CTLA4-Ig), etanercept (anti-TNF), or phosphate-buffered saline were given to NMRI mice intravenously inoculated with Staphylococcus aureus. The clinical course of septic arthritis and histopathological and radiological changes of joints were compared among the groups. RESULTS: Mice receiving CTLA4-Ig treatment had more-severe septic arthritis, compared with controls and mice receiving anti-TNF treatment. Anti-TNF treatment led to more-severe weight loss and kidney abscesses, as well as a higher bacterial burden in the kidneys. Mice receiving CTLA4-Ig therapy had lower serum levels of interleukin 4, whereas mice receiving anti-TNF therapy had higher levels of TNF-α. Both iNOS and arginase-1 expression were reduced in peritoneal macrophages from mice receiving CTLA4-Ig, compared with expression in the anti-TNF group. CONCLUSIONS: CTLA4-Ig therapy significantly increased the susceptibility to S. aureus septic arthritis in mice, whereas anti-TNF therapy deteriorated host bacterial clearance, resulting in more-severe weight loss and kidney abscesses.


Subject(s)
Antirheumatic Agents/therapeutic use , Arthritis, Infectious/immunology , Arthritis, Rheumatoid/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Abatacept/therapeutic use , Animals , Arthritis, Infectious/microbiology , Arthritis, Rheumatoid/microbiology , Etanercept/therapeutic use , Female , Joints/immunology , Joints/pathology , Mice , Staphylococcal Infections/microbiology , Tumor Necrosis Factor-alpha/immunology
8.
Arthritis Rheumatol ; 67(1): 107-16, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25302691

ABSTRACT

OBJECTIVE: Permanent reduction in joint function is a severe postinfectious complication in patients with Staphylococcus aureus septic arthritis. We undertook this study to determine whether this reduction in joint function might be caused by persistent joint inflammation after the adequate eradication of bacteria by antibiotics. METHODS: After intraarticular injection of cloxacillin-killed S aureus into mouse knee joints, we investigated whether antibiotic-killed S aureus induced joint inflammation and elucidated the molecular and cellular mechanisms of this type of arthritis. RESULTS: Intraarticular injection of antibiotic-killed S aureus induced mild-to-moderate synovitis and bone erosions that lasted for a minimum of 14 days. Compared with wild-type animals, mice deficient in tumor necrosis factor receptor type I (TNFRI), receptor for advanced glycation end products (RAGE), or Toll-like receptor 2 (TLR-2) had a significantly reduced frequency and severity of synovitis. Combined depletion of monocytes and neutrophils also resulted in a significantly lower frequency of synovitis. Among bacterial factors, insoluble cell debris played a more important role than bacterial DNA or soluble components in inducing joint inflammation. Importantly, anti-TNF therapy abrogated joint inflammation induced by antibiotic-killed S aureus. CONCLUSION: Antibiotic-killed S aureus induced and maintained joint inflammation mediated through TLR-2, TNFRI, and RAGE. The cross-talk between neutrophils and monocytes is responsible for this type of arthritis. Anti-TNF therapy might be used as a novel strategy, in combination with antibiotics, to treat staphylococcal septic arthritis.


Subject(s)
Anti-Bacterial Agents/pharmacology , Arthritis, Experimental/metabolism , Arthritis, Experimental/microbiology , Arthritis, Infectious/metabolism , Arthritis, Infectious/microbiology , Cloxacillin/pharmacology , Staphylococcus aureus/drug effects , Animals , Arthritis, Experimental/pathology , Arthritis, Infectious/pathology , Cell Communication/physiology , Disease Models, Animal , Female , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Monocytes/pathology , Neutrophils/pathology , Receptor for Advanced Glycation End Products , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Receptors, Tumor Necrosis Factor, Type I/deficiency , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Severity of Illness Index , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism
9.
APMIS ; 122(12): 1240-50, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25051890

ABSTRACT

Staphylococcus aureus is the most common cause of skin infections that range from mild diseases up to life-threatening conditions. Mechanisms of S. aureus virulence in those infections remain poorly studied. To investigate the impact of S. aureus surface proteins on skin infection, we used mouse models of skin abscess formation and skin necrosis, induced by a subcutaneous injection of bacteria. In the skin abscess model, a sortase-deficient S. aureus strain lacking all of its cell-wall anchored proteins was less virulent than its wild-type strain. Also, strains specifically lacking protein A, fibronecting binding proteins, clumping factor A or surface protein SasF were impaired in their virulence. When a model of dermonecrosis was studied, the S. aureus surface proteins could not be shown to be involved. In summary, surface proteins play an important role in virulence of S. aureus skin abscess infections, but not in formation of skin necrosis.


Subject(s)
Coagulase/metabolism , Membrane Proteins/metabolism , Skin Diseases/microbiology , Staphylococcal Infections/pathology , Staphylococcal Protein A/metabolism , Staphylococcus aureus/pathogenicity , Animals , Cell Wall/metabolism , Cell Wall/microbiology , Disease Models, Animal , Female , Mice , Staphylococcal Infections/microbiology , Staphylococcus aureus/isolation & purification , Virulence Factors/metabolism
11.
Genome Res ; 24(5): 839-49, 2014 May.
Article in English | MEDLINE | ID: mdl-24717264

ABSTRACT

Microbial virulence is a complex and often multifactorial phenotype, intricately linked to a pathogen's evolutionary trajectory. Toxicity, the ability to destroy host cell membranes, and adhesion, the ability to adhere to human tissues, are the major virulence factors of many bacterial pathogens, including Staphylococcus aureus. Here, we assayed the toxicity and adhesiveness of 90 MRSA (methicillin resistant S. aureus) isolates and found that while there was remarkably little variation in adhesion, toxicity varied by over an order of magnitude between isolates, suggesting different evolutionary selection pressures acting on these two traits. We performed a genome-wide association study (GWAS) and identified a large number of loci, as well as a putative network of epistatically interacting loci, that significantly associated with toxicity. Despite this apparent complexity in toxicity regulation, a predictive model based on a set of significant single nucleotide polymorphisms (SNPs) and insertion and deletions events (indels) showed a high degree of accuracy in predicting an isolate's toxicity solely from the genetic signature at these sites. Our results thus highlight the potential of using sequence data to determine clinically relevant parameters and have further implications for understanding the microbial virulence of this opportunistic pathogen.


Subject(s)
Genome, Bacterial , Methicillin-Resistant Staphylococcus aureus/genetics , Models, Genetic , Virulence/genetics , Genome-Wide Association Study , INDEL Mutation , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Polymorphism, Single Nucleotide
12.
PLoS One ; 8(7): e67698, 2013.
Article in English | MEDLINE | ID: mdl-23861785

ABSTRACT

Mannitol (Mtl) fermentation, with the subsequent production of acid, is a species signature of Staphylococcus aureus, and discriminates it from most other members of the genus. Inactivation of the gene mtlD, encoding Mtl-1-P dehydrogenase was found to markedly reduce survival in the presence of the antimicrobial fatty acid, linoleic acid. We demonstrate that the sugar alcohol has a potentiating action for this membrane-acting antimicrobial. Analysis of cellular metabolites revealed that, during exponential growth, the mtlD mutant accumulated high levels of Mtl and Mtl-P. The latter metabolite was not detected in its isogenic parent strain or a deletion mutant of the entire mtlABFD operon. In addition, the mtlD mutant strain exhibited a decreased MIC for H2O2, however virulence was unaffected in a model of septic arthritis.


Subject(s)
Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Mannitol/metabolism , Skin/microbiology , Staphylococcus aureus/metabolism , Sugar Alcohol Dehydrogenases/genetics , Animals , Bacterial Proteins/metabolism , Humans , Hydrogen Peroxide/pharmacology , Linoleic Acid/pharmacology , Mice , Microbial Sensitivity Tests , Microbial Viability/drug effects , Mutation , Operon , Staphylococcal Skin Infections/microbiology , Staphylococcus aureus/genetics , Staphylococcus aureus/growth & development , Sugar Alcohol Dehydrogenases/deficiency , Virulence
13.
J Infect Dis ; 208(6): 990-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23801604

ABSTRACT

Skin infections are frequently caused by Staphylococcus aureus and can lead to a fatal sepsis. The microbial mechanisms controlling the initiation and progression from mild skin infection to a severe disseminated infection remain poorly understood. Using a combination of clinical data and in vitro and ex vivo assays, we show that staphylokinase, secreted by S. aureus, promoted the establishment of skin infections in humans and increased bacterial penetration through skin barriers by activating plasminogen. However, when infection was established, the interaction between staphylokinase and plasminogen did not promote systemic dissemination but induced the opening and draining of abscesses and decreased disease severity in neutropenic mice. Also, increased staphylokinase production was associated with noninvasive S. aureus infections in patients. Our results point out the dual roles of staphylokinase in S. aureus skin infections as promoting the establishment of infections while decreasing disease severity.


Subject(s)
Gene Expression Regulation, Bacterial , Metalloendopeptidases/metabolism , Skin Diseases/microbiology , Staphylococcal Infections/pathology , Abscess/microbiology , Animals , Biomarkers/blood , Female , Humans , Male , Mice , Plasminogen/metabolism , Plasminogen Activator Inhibitor 1/blood , Plasminogen Activators/pharmacology , Skin/microbiology , Skin/pathology , Skin Diseases/pathology , Staphylococcus aureus/enzymology
14.
J Infect Dis ; 204(3): 348-57, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21742832

ABSTRACT

BACKGROUND: Despite advances in medical practices, in recent decades permanent reductions in joint function have not been achieved, and the high mortality rate of patients with staphylococcal septic arthritis has not substantially improved. METHODS: We evaluated the effects of a combined tumor necrosis factor (TNF) inhibitor and antibiotic therapy on the course of Staphylococcus aureus arthritis and sepsis in mice. RESULTS: Treatment with the combination of a TNF inhibitor and an antibiotic resulted in a quicker relief of clinical arthritis in mice with septic arthritis, compared with an antibiotic monotherapy. Both histopathologically verified synovitis and the extent of joint destruction were reduced by this combined treatment. Importantly, anti-TNF treatment significantly improved the survival rate of mice with S. aureus sepsis and staphylococcal enterotoxin shock syndrome; this effect might be the result of a partial restoration of the hemostatic balance between coagulation and fibrinolysis. Finally, we demonstrated that anti-TNF treatment downregulates high-mobility group protein B1 in staphylococcal enterotoxin shock syndrome. CONCLUSIONS: Thus, simultaneous systemic TNF inhibition and antibiotic therapy has beneficial effects on the outcome of S. aureus arthritis and sepsis in a mouse model, suggesting that the combination of a TNF inhibitor and antibiotics represents a novel therapeutic strategy for the treatment of staphylococcal infections.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Arthritis, Infectious/drug therapy , Cloxacillin/administration & dosage , Immunoglobulin G/administration & dosage , Receptors, Tumor Necrosis Factor/administration & dosage , Staphylococcal Infections/drug therapy , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Drug Therapy, Combination , Etanercept , Female , HMGB1 Protein/analysis , Mice , Mice, Inbred BALB C
15.
Inflamm Res ; 60(11): 1021-9, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21786185

ABSTRACT

OBJECTIVE: Down-regulation of fibrinolysis and increased fibrin deposition in joints are hallmarks of rheumatoid arthritis (RA), and are believed to be involved in disease progression. The mouse model of collagen-induced arthritis (CIA) closely resembles RA and has been used to explore mechanism and treatments of RA, but neither the fibrinolytic system nor pro-fibrinolytic therapies were investigated in CIA. MATERIALS AND METHODS: Plasmin activity, levels of plasminogen activator inhibitor (PAI-1), D-dimer, and IL-6 were measured in plasma of CIA mice. Fibrin deposition and PAI-1 levels were also measured in inflamed joints. Mice were treated with plasminogen activators uPA (urokinase-type plasminogen activator) or tPA (tissue-type plasminogen activator). Effects of treatment on disease severity and fibrinolytic system were assessed. RESULTS: CIA caused decrease in plasmin activity, accompanied by increase in PAI-1 levels, in both blood and inflamed joints. This resulted in massive fibrin deposition in synovium. PAI-1 levels correlated negatively with plasmin activity and positively with IL-6. Treatments with uPA and tPA improved plasmin activity and removed fibrin deposits in inflamed joints. However, disease severity remained unchanged. CONCLUSIONS: Fibrinolytic changes in CIA parallel changes in RA, making CIA a suitable model to study fibrinolysis in RA. Normalization of plasmin activity in CIA after treatment with plasminogen activators had no effect on disease severity.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Collagen/adverse effects , Fibrinolysis , Animals , Arthritis, Rheumatoid/chemically induced , Disease Models, Animal , Disease Progression , Fibrin/metabolism , Fibrin Fibrinogen Degradation Products/pharmacology , Fibrinogen/chemistry , Fibrinolysin/metabolism , Immunohistochemistry/methods , Interleukin-6/blood , Male , Mice , Mice, Inbred DBA , Plasminogen Activator Inhibitor 1/blood , Tissue Plasminogen Activator/metabolism , Urokinase-Type Plasminogen Activator/metabolism
16.
J Infect Dis ; 202(7): 1041-9, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20726765

ABSTRACT

BACKGROUND: Staphylokinase (SAK) is produced by the majority of Staphylococcus aureus strains. It is an extracellular protein that activates the conversion of human plasminogen (plg) to plasmin. The role played by SAK in staphylococcal infection is unclear. METHODS: Wild-type S. aureus strain LS-1, which lacks the ability to produce SAK, was modified by an insertion of the sak gene into its chromosome. The sak gene was integrated in 2 forms--(1) linked to its own promoter and (2) fused to the promoter of the protein A gene--which resulted in the overexpression of SAK. SAK is highly specific for human plg and exhibits almost no activity toward murine plg. To investigate the role played by SAK in a murine infection model, human plg transgenic mice and their wild-type counterparts were inoculated intravenously with congenic S. aureus strains differing in SAK production. RESULTS: Human plg transgenic mice inoculated with SAK-expressing strains displayed significantly reduced mortality, less weight loss, and lower bacterial loads in kidneys than did the wild-type mice. No difference in the severity of sepsis was observed between transgenic and wild-type mice infected with a SAK-deficient strain. CONCLUSIONS: The results suggest that expression of SAK followed by activation of plg alleviates the course of S. aureus sepsis.


Subject(s)
Bacterial Proteins/metabolism , Metalloendopeptidases/metabolism , Plasminogen/metabolism , Staphylococcal Infections/pathology , Staphylococcus aureus/pathogenicity , Animals , Bacterial Proteins/genetics , Colony Count, Microbial , Disease Models, Animal , Humans , Kidney/microbiology , Metalloendopeptidases/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutagenesis, Insertional , Plasminogen/genetics , Recombination, Genetic , Survival Analysis
17.
PLoS Pathog ; 6(6): e1000964, 2010 Jun 24.
Article in English | MEDLINE | ID: mdl-20585570

ABSTRACT

Entry of Staphylococcus aureus into the bloodstream can lead to metastatic abscess formation and infective endocarditis. Crucial to the development of both these conditions is the interaction of S. aureus with endothelial cells. In vivo and in vitro studies have shown that the staphylococcal invasin FnBPA triggers bacterial invasion of endothelial cells via a process that involves fibronectin (Fn) bridging to alpha(5)beta(1) integrins. The Fn-binding region of FnBPA usually contains 11 non-identical repeats (FnBRs) with differing affinities for Fn, which facilitate the binding of multiple Fn molecules and may promote integrin clustering. We thus hypothesized that multiple repeats are necessary to trigger the invasion of endothelial cells by S. aureus. To test this we constructed variants of fnbA containing various combinations of FnBRs. In vitro assays revealed that endothelial cell invasion can be facilitated by a single high-affinity, but not low-affinity FnBR. Studies using a nisin-inducible system that controlled surface expression of FnBPA revealed that variants encoding fewer FnBRs required higher levels of surface expression to mediate invasion. High expression levels of FnBPA bearing a single low affinity FnBR bound Fn but did not invade, suggesting that FnBPA affinity for Fn is crucial for triggering internalization. In addition, multiple FnBRs increased the speed of internalization, as did higher expression levels of FnBPA, without altering the uptake mechanism. The relevance of these findings to pathogenesis was demonstrated using a murine sepsis model, which showed that multiple FnBRs were required for virulence. In conclusion, multiple FnBRs within FnBPA facilitate efficient Fn adhesion, trigger rapid bacterial uptake and are required for pathogenesis.


Subject(s)
Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Repetitive Sequences, Nucleic Acid/genetics , Sepsis/microbiology , Staphylococcal Infections/pathology , Staphylococcus aureus/physiology , Virulence , Animals , Bacterial Adhesion , Blotting, Western , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/microbiology , Enzyme-Linked Immunosorbent Assay , Humans , Male , Mice , Sepsis/metabolism , Sepsis/pathology , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Umbilical Veins/cytology , Umbilical Veins/microbiology
18.
Arthritis Res Ther ; 11(5): R132, 2009.
Article in English | MEDLINE | ID: mdl-19723321

ABSTRACT

INTRODUCTION: Dichloroacetate (DCA) has been in clinical use for the treatment of lactacidosis and inherited mitochondrial disorders. It has potent anti-tumor effects both in vivo and in vitro, facilitating apoptosis and inhibiting proliferation. The pro-apoptotic and anti-proliferative properties of DCA prompted us to investigate the effects of this compound in arthritis. METHODS: In the present study, we used DCA to treat murine collagen type II (CII)-induced arthritis (CIA), an experimental model of rheumatoid arthritis. DBA/1 mice were treated with DCA given in drinking water. RESULTS: Mice treated with DCA displayed much slower onset of CIA and significantly lower severity (P < 0.0001) and much lower frequency (36% in DCA group vs. 86% in control group) of arthritis. Also, cartilage and joint destruction was significantly decreased following DCA treatment (P = 0.005). Moreover, DCA prevented arthritis-induced cortical bone mineral loss. This clinical picture was also reflected by lower levels of anti-CII antibodies in DCA-treated versus control mice, indicating that DCA affected the humoral response. In contrast, DCA had no effect on T cell- or granulocyte-mediated responses. The beneficial effect of DCA was present in female DBA/1 mice only. This was due in part to the effect of estrogen, since ovariectomized mice did not benefit from DCA treatment to the same extent as sham-operated controls (day 30, 38.7% of ovarectomized mice had arthritis vs. only 3.4% in sham-operated group). CONCLUSION: Our results indicate that DCA delays the onset and alleviates the progression of CIA in an estrogen-dependent manner.


Subject(s)
Arthritis, Experimental/prevention & control , Arthritis, Rheumatoid/drug therapy , Dichloroacetic Acid/therapeutic use , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Autoantibodies/blood , Autoantibodies/drug effects , Bone Density/drug effects , Collagen/immunology , Estrogens/metabolism , Female , Hypersensitivity, Delayed/drug therapy , Hypersensitivity, Delayed/immunology , Mice , Mice, Inbred DBA , Ovariectomy , T-Lymphocytes/drug effects
19.
PLoS One ; 4(2): e4344, 2009.
Article in English | MEDLINE | ID: mdl-19183815

ABSTRACT

Staphylococcus aureus is an important human commensal and opportunistic pathogen responsible for a wide range of infections. Long chain unsaturated free fatty acids represent a barrier to colonisation and infection by S. aureus and act as an antimicrobial component of the innate immune system where they are found on epithelial surfaces and in abscesses. Despite many contradictory reports, the precise anti-staphylococcal mode of action of free fatty acids remains undetermined. In this study, transcriptional (microarrays and qRT-PCR) and translational (proteomics) analyses were applied to ascertain the response of S. aureus to a range of free fatty acids. An increase in expression of the sigma(B) and CtsR stress response regulons was observed. This included increased expression of genes associated with staphyloxanthin synthesis, which has been linked to membrane stabilisation. Similarly, up-regulation of genes involved in capsule formation was recorded as were significant changes in the expression of genes associated with peptidoglycan synthesis and regulation. Overall, alterations were recorded predominantly in pathways involved in cellular energetics. In addition, sensitivity to linoleic acid of a range of defined (sigB, arcA, sasF, sarA, agr, crtM) and transposon-derived mutants (vraE, SAR2632) was determined. Taken together, these data indicate a common mode of action for long chain unsaturated fatty acids that involves disruption of the cell membrane, leading to interference with energy production within the bacterial cell. Contrary to data reported for other strains, the clinically important EMRSA-16 strain MRSA252 used in this study showed an increase in expression of the important virulence regulator RNAIII following all of the treatment conditions tested. An adaptive response by S. aureus of reducing cell surface hydrophobicity was also observed. Two fatty acid sensitive mutants created during this study were also shown to diplay altered pathogenesis as assessed by a murine arthritis model. Differences in the prevalence and clinical importance of S. aureus strains might partly be explained by their responses to antimicrobial fatty acids.


Subject(s)
Fatty Acids, Unsaturated/pharmacology , Staphylococcus aureus/metabolism , Staphylococcus aureus/pathogenicity , Animals , Female , Genes, Bacterial , Mice , Mice, Inbred Strains , Polymerase Chain Reaction , Proteomics , Staphylococcus aureus/genetics , Transcription, Genetic , Up-Regulation , Virulence/genetics
20.
Microbiology (Reading) ; 155(Pt 3): 667-679, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19246738

ABSTRACT

The ability of Staphylococcus aureus to avoid innate immune responses including neutrophil-mediated phagocytosis is crucial for the organism to cause infection. This multifactorial process involves several secreted and cell-surface-associated proteins. In this paper we report a novel mechanism of combating neutrophils that involves iron-regulated surface determinant protein H (IsdH). The IsdH protein is part of a complex that is only expressed under iron-restricted conditions in order to bind haemoglobin and extract and transport haem into the cytoplasm. A null mutant defective in expression of IsdH, and mutants expressing variants of IsdH with substitutions in residues predicted to be involved in ligand binding, were generated from S. aureus 8325-4. The IsdH-defective mutants were shown by several measures to have reduced virulence compared with the wild-type. The mutant was engulfed more rapidly by human neutrophils in the presence of serum opsonins, survived poorly in fresh whole human blood and was less virulent in a mouse model of sepsis. The protective mechanism seems to stem from an accelerated degradation of the serum opsonin C3b.


Subject(s)
Antigens, Bacterial/immunology , Iron/metabolism , Neutrophils/microbiology , Receptors, Cell Surface/immunology , Staphylococcus aureus/immunology , Amino Acid Substitution , Animals , Antigens, Bacterial/genetics , Antigens, Bacterial/metabolism , Cloning, Molecular , Complement C3b/immunology , Disease Models, Animal , Female , Humans , Iron/immunology , Mice , Mutation, Missense , Neutrophils/immunology , Phagocytosis , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Staphylococcus aureus/pathogenicity , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...