Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Immun Ageing ; 17: 12, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32467712

RESUMO

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 Immun ; 85(7)2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28438975

RESUMO

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.


Assuntos
Proteínas de Bactérias/metabolismo , Galectina 3/metabolismo , Interações Hospedeiro-Patógeno , Serina Endopeptidases/metabolismo , Staphylococcus aureus/enzimologia , Staphylococcus aureus/patogenicidade , Animais , Carga Bacteriana , Proteínas Sanguíneas , Modelos Animais de Doenças , Galectinas , Humanos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteólise , Infecções Cutâneas Estafilocócicas/microbiologia , Infecções Cutâneas Estafilocócicas/patologia , Virulência
3.
Genome Res ; 24(5): 839-49, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24717264

RESUMO

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.


Assuntos
Genoma Bacteriano , Staphylococcus aureus Resistente à Meticilina/genética , Modelos Genéticos , Virulência/genética , Estudo de Associação Genômica Ampla , Mutação INDEL , Staphylococcus aureus Resistente à Meticilina/patogenicidade , Polimorfismo de Nucleotídeo Único
4.
J Infect Dis ; 213(1): 139-48, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26136471

RESUMO

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.


Assuntos
Biofilmes/efeitos dos fármacos , Metaloendopeptidases/metabolismo , Staphylococcus aureus/metabolismo , Animais , Antibacterianos/farmacologia , Farmacorresistência Bacteriana/efeitos dos fármacos , Feminino , Fibrina/metabolismo , Masculino , Metaloendopeptidases/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Plasminogênio/metabolismo , Staphylococcus aureus/efeitos dos fármacos
5.
J Infect Dis ; 212(8): 1308-16, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-25838268

RESUMO

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.


Assuntos
Antirreumáticos/uso terapêutico , Artrite Infecciosa/imunologia , Artrite Reumatoide/imunologia , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/imunologia , Abatacepte/uso terapêutico , Animais , Artrite Infecciosa/microbiologia , Artrite Reumatoide/microbiologia , Etanercepte/uso terapêutico , Feminino , Articulações/imunologia , Articulações/patologia , Camundongos , Infecções Estafilocócicas/microbiologia , Fator de Necrose Tumoral alfa/imunologia
6.
J Infect Dis ; 208(6): 990-9, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23801604

RESUMO

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.


Assuntos
Regulação Bacteriana da Expressão Gênica , Metaloendopeptidases/metabolismo , Dermatopatias/microbiologia , Infecções Estafilocócicas/patologia , Abscesso/microbiologia , Animais , Biomarcadores/sangue , Feminino , Humanos , Masculino , Camundongos , Plasminogênio/metabolismo , Inibidor 1 de Ativador de Plasminogênio/sangue , Ativadores de Plasminogênio/farmacologia , Pele/microbiologia , Pele/patologia , Dermatopatias/patologia , Staphylococcus aureus/enzimologia
7.
PLoS Pathog ; 6(6): e1000964, 2010 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-20585570

RESUMO

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.


Assuntos
Adesinas Bacterianas/genética , Adesinas Bacterianas/metabolismo , Sequências Repetitivas de Ácido Nucleico/genética , Sepse/microbiologia , Infecções Estafilocócicas/patologia , Staphylococcus aureus/fisiologia , Virulência , Animais , Aderência Bacteriana , Western Blotting , Células Cultivadas , Endotélio Vascular/citologia , Endotélio Vascular/microbiologia , Ensaio de Imunoadsorção Enzimática , Humanos , Masculino , Camundongos , Sepse/metabolismo , Sepse/patologia , Infecções Estafilocócicas/metabolismo , Infecções Estafilocócicas/microbiologia , Veias Umbilicais/citologia , Veias Umbilicais/microbiologia
8.
J Infect Dis ; 204(3): 348-57, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21742832

RESUMO

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.


Assuntos
Antibacterianos/administração & dosagem , Artrite Infecciosa/tratamento farmacológico , Cloxacilina/administração & dosagem , Imunoglobulina G/administração & dosagem , Receptores do Fator de Necrose Tumoral/administração & dosagem , Infecções Estafilocócicas/tratamento farmacológico , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Animais , Quimioterapia Combinada , Etanercepte , Feminino , Proteína HMGB1/análise , Camundongos , Camundongos Endogâmicos BALB C
9.
Inflamm Res ; 60(11): 1021-9, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21786185

RESUMO

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.


Assuntos
Artrite Reumatoide/tratamento farmacológico , Colágeno/efeitos adversos , Fibrinólise , Animais , Artrite Reumatoide/induzido quimicamente , Modelos Animais de Doenças , Progressão da Doença , Fibrina/metabolismo , Produtos de Degradação da Fibrina e do Fibrinogênio/farmacologia , Fibrinogênio/química , Fibrinolisina/metabolismo , Imuno-Histoquímica/métodos , Interleucina-6/sangue , Masculino , Camundongos , Camundongos Endogâmicos DBA , Inibidor 1 de Ativador de Plasminogênio/sangue , Ativador de Plasminogênio Tecidual/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
10.
J Infect Dis ; 202(7): 1041-9, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20726765

RESUMO

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.


Assuntos
Proteínas de Bactérias/metabolismo , Metaloendopeptidases/metabolismo , Plasminogênio/metabolismo , Infecções Estafilocócicas/patologia , Staphylococcus aureus/patogenicidade , Animais , Proteínas de Bactérias/genética , Contagem de Colônia Microbiana , Modelos Animais de Doenças , Humanos , Rim/microbiologia , Metaloendopeptidases/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutagênese Insercional , Plasminogênio/genética , Recombinação Genética , Análise de Sobrevida
12.
Infect Dis (Lond) ; 51(6): 446-451, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30985241

RESUMO

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.


Assuntos
Biofilmes/crescimento & desenvolvimento , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/fisiologia , Staphylococcus aureus/patogenicidade , Antibacterianos/farmacologia , Proteínas de Bactérias/genética , Genótipo , Humanos , Staphylococcus aureus Resistente à Meticilina , Dermatopatias/microbiologia , Staphylococcus aureus/classificação , Transativadores/genética
13.
J Leukoc Biol ; 79(3): 482-8, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16387838

RESUMO

Uric acid, the naturally occurring degradation product of purine metabolism, is a danger signal, driving maturation of dendritic cells. It is well known that uric acid crystals display potent proinflammatory properties--the cause of gout--whereas the biological properties of soluble uric acid are less well documented. We have demonstrated previously that nucleic acids of endogenous and exogenous origin display proinflammatory properties. The aim of the present study was to assess the impact of soluble uric acid on in vivo inflammatory responses. Mice were administered with uric acid suspension in saline or saline alone prior to induction of neutrophil-mediated inflammation, delayed-type hypersensitivity, histamin-induced edema (measure of vasodilation capacity), as well as double-stranded (ds)RNA-triggered arthritis. Frequency and severity of arthritis were decreased significantly in mice exposed to dsRNA and simultaneously treated with uric acid as compared with saline-treated controls. Also, granulocyte-mediated inflammatory response and vasodilation capacity were reduced significantly in mice treated with uric acid as compared with their control group. The data suggest that down-regulation of inflammation was mediated by skewing the inflammatory response from the peripheral sites to the peritoneal cavity and down-regulating vasodilatatory capacity and thereby affecting leukocyte migration. In contrast, the T cell-mediated delayed-type hypersensitivity reaction was not affected significantly in mice exposed to uric acid. These findings demonstrate that uric acid displays a potent, distant anti-inflammatory effect in vivo. This property seems to be mediated by down-regulation of neutrophil influx to the site of inflammatory insult.


Assuntos
Artrite Experimental/imunologia , Regulação para Baixo/imunologia , Imunossupressores/imunologia , Ácidos Nucleicos/imunologia , RNA de Cadeia Dupla/imunologia , Ácido Úrico/imunologia , Animais , Artrite Experimental/induzido quimicamente , Artrite Experimental/tratamento farmacológico , Quimiotaxia/efeitos dos fármacos , Quimiotaxia/imunologia , Quimiotaxia de Leucócito/efeitos dos fármacos , Quimiotaxia de Leucócito/imunologia , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Edema/induzido quimicamente , Edema/imunologia , Edema/fisiopatologia , Feminino , Hipersensibilidade Tardia/induzido quimicamente , Hipersensibilidade Tardia/imunologia , Hipersensibilidade Tardia/fisiopatologia , Imunossupressores/metabolismo , Imunossupressores/farmacologia , Mediadores da Inflamação/efeitos adversos , Mediadores da Inflamação/imunologia , Articulações/efeitos dos fármacos , Articulações/imunologia , Articulações/fisiopatologia , Camundongos , Neutrófilos/efeitos dos fármacos , Neutrófilos/imunologia , Ácidos Nucleicos/metabolismo , RNA de Cadeia Dupla/efeitos adversos , Ácido Úrico/metabolismo , Ácido Úrico/farmacologia , Vasodilatação/efeitos dos fármacos , Vasodilatação/imunologia
14.
PLoS One ; 12(3): e0173492, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28264025

RESUMO

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.


Assuntos
Abatacepte/farmacologia , Antígeno CTLA-4/antagonistas & inibidores , Dermatite/microbiologia , Etanercepte/farmacologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/efeitos dos fármacos , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Animais , Apoptose , Carga Bacteriana , Citocinas/sangue , Citocinas/metabolismo , Dermatite/tratamento farmacológico , Dermatite/imunologia , Dermatite/metabolismo , Modelos Animais de Doenças , Feminino , Hiperpigmentação , Imunossupressores/farmacologia , Camundongos , Monócitos/imunologia , Monócitos/metabolismo , Monócitos/patologia , Necrose , Fenótipo , Índice de Gravidade de Doença , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/metabolismo , Staphylococcus aureus/imunologia
15.
Microbes Infect ; 7(15): 1501-11, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16039897

RESUMO

We have previously shown that staphylococcal protein A (SpA) anchored to the cell wall of Staphylococcus aureus acts as a virulence factor in septic arthritis. Apart from the ability of SpA to interact with Fcgamma, it also binds to Fab-regions with immunoglobulin heavy chains encoded by the V(H) clan III gene family. The objective of the present study was to investigate whether in vivo expression of SpA by staphylococci induces V(H)III-dependent supraclonal B-cell responses, and whether such responses may affect the ability of the host to produce anti-staphylococcal antibodies. Upon primary infection of mice, a SpA-expressing staphylococcal strain gave rise to significantly higher serum levels of V(H)III-encoded antibodies specific for SpA devoid of Fcgamma-binding ability (MSpA) than an isogeneic spa deletion mutant strain. The V(H)III-dependence of MSpA-specific antibody responses was affected by the size of the staphylococcal inoculum, and differed for IgM and IgG isotypes. Mice that had recovered from a prior mild infection from a SpA-expressing strain were protected against infection-induced weight loss upon reinfection. Although no lasting MSpA-specific IgG was induced by previous mild infection, these protected mice possessed IgG specific for clumping factor A, a conventional staphylococcal protein antigen. Our findings demonstrate that the expression of a B-cell superantigen during staphylococcal infection causes supraclonal changes to the immune system. Notably, while superantigen-triggered B-cell responses do not favor the development of SpA-specific memory B-cells, such responses do not interfere with the development of antibodies specific for a staphylococcal protein antigen associated with protective immunity.


Assuntos
Linfócitos B/imunologia , Parede Celular/imunologia , Infecções Estafilocócicas/imunologia , Proteína Estafilocócica A/imunologia , Staphylococcus aureus/imunologia , Superantígenos/imunologia , Animais , Anticorpos Antibacterianos/sangue , Peso Corporal , Parede Celular/química , Coagulase/imunologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Feminino , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Camundongos
16.
PLoS One ; 10(7): e0131645, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26135738

RESUMO

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.


Assuntos
Artrite Infecciosa/tratamento farmacológico , Proteína Antagonista do Receptor de Interleucina 1/uso terapêutico , Sepse/tratamento farmacológico , Infecções Estafilocócicas/tratamento farmacológico , Animais , Artrite Experimental/tratamento farmacológico , Artrite Experimental/microbiologia , Artrite Infecciosa/microbiologia , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Articulações/patologia , Rim/patologia , Macrófagos Peritoneais/metabolismo , Camundongos , Sepse/microbiologia , Infecções Estafilocócicas/microbiologia , Staphylococcus , Staphylococcus aureus , Microtomografia por Raio-X
17.
Arthritis Rheumatol ; 67(1): 107-16, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25302691

RESUMO

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.


Assuntos
Antibacterianos/farmacologia , Artrite Experimental/metabolismo , Artrite Experimental/microbiologia , Artrite Infecciosa/metabolismo , Artrite Infecciosa/microbiologia , Cloxacilina/farmacologia , Staphylococcus aureus/efeitos dos fármacos , Animais , Artrite Experimental/patologia , Artrite Infecciosa/patologia , Comunicação Celular/fisiologia , Modelos Animais de Doenças , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/patologia , Neutrófilos/patologia , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/deficiência , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/deficiência , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Índice de Gravidade de Doença , Receptor 2 Toll-Like/deficiência , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/metabolismo
18.
Microbes Infect ; 6(2): 196-201, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14998518

RESUMO

Clumping factor A (ClfA), a fibrinogen-binding protein expressed on the Staphylococcus aureus cell surface, has previously been shown to act as a virulence factor in experimental septic arthritis. Although the interaction between ClfA and fibrinogen is assumed to be of importance for the virulence of S. aureus, this has not been demonstrated in any in vivo model of infection. Therefore, the objective of this study was to investigate the contribution of this interaction to ClfA-mediated virulence in murine S. aureus-induced arthritis. Ancrod, a serine protease with thrombin-like activity, was used to induce in vivo depletion of fibrinogen in mice. Ancrod treatment significantly aggravated septic arthritis following inoculation with a ClfA-expressing strain (Newman) compared to control treatment. Also, ancrod treatment tended to enhance the arthritis induced by a clfA mutant strain (DU5876), indicating that fibrinogen depletion exacerbates septic arthritis in a ClfA-independent manner. Most importantly, the ClfA-expressing strain was much more arthritogenic than the isogenic clfA mutant, following inoculation of fibrinogen-depleted mice. This finding indicates that the interaction between ClfA and free fibrinogen is not required for ClfA-mediated functions contributing to S. aureus virulence. It is conceivable that ClfA contributes to the virulence of S. aureus through interactions with other host ligands than fibrinogen.


Assuntos
Coagulase/fisiologia , Fibrinogênio/metabolismo , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/patogenicidade , Virulência/fisiologia , Animais , Camundongos , Camundongos Endogâmicos , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/imunologia , Staphylococcus aureus/fisiologia , Virulência/efeitos dos fármacos
19.
Microbes Infect ; 6(2): 188-95, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14998517

RESUMO

Clumping factor A (ClfA), a fibrinogen-binding protein linked to the Staphylococcus aureus cell wall, is an important virulence factor in infection models, e.g., of septic arthritis. However, the mechanism(s) by which ClfA contributes to the virulence of the bacterium is unknown. In the present study, the impact of ClfA expression on the phagocytosis of S. aureus by macrophages was investigated using clfA-positive and clfA-negative isogenic strains. Furthermore, the possible contribution of ClfA to the proinflammatory and immunostimulatory activity of S. aureus was studied. Our results indicate that ClfA expression significantly protects S. aureus against macrophage phagocytosis. This protection does not require the presence of intact fibrinogen, a ligand for ClfA. ClfA expression by S. aureus enhanced the proliferative response of spleen cells. On the other hand, a clfA mutant strain caused more release of proinflammatory mediators by macrophages than its clfA-positive parental strain. Both the protection against phagocytosis and the enhanced immunostimulatory activity provided by ClfA expression are likely to contribute to the in vivo virulence of S. aureus.


Assuntos
Coagulase/toxicidade , Macrófagos/efeitos dos fármacos , Fagocitose/efeitos dos fármacos , Staphylococcus aureus/patogenicidade , Virulência/fisiologia , Animais , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/imunologia , Staphylococcus aureus/metabolismo , Virulência/efeitos dos fármacos
20.
FEMS Microbiol Lett ; 217(2): 125-32, 2002 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-12480095

RESUMO

Interactions between staphylococci and the joint tissues of the host lead typically to rapidly progressing and highly destructive processes. Staphylococci possess a vast arsenal of components and products that contribute to the pathogenesis of joint infection. Occasionally these compounds have overlapping activities and act either in concert or alone. Host responsiveness to staphylococcal infection displays an even more complex pattern. Most of the cells and molecules that participate in the innate immune system protect the host against bacteria. However, the staphylococci have developed systems that counteract endogenous protective mechanisms. Interestingly, certain cells and molecules of the acquired immune system potentiate the severity of infection by triggering exaggerated responses to the staphylococcal danger signals. This review deals with the intricate host-bacterium interactions that occur during experimental septic arthritis, and outlines potential preventive and treatment modalities.


Assuntos
Artrite Infecciosa/microbiologia , Infecções Estafilocócicas/microbiologia , Staphylococcus/patogenicidade , Animais , Artrite Infecciosa/imunologia , Artrite Infecciosa/terapia , Quimiocinas/metabolismo , Citocinas/metabolismo , Imunidade Ativa , Articulações/microbiologia , Camundongos , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/terapia , Staphylococcus/classificação , Staphylococcus/metabolismo , Resultado do Tratamento , Fatores de Virulência/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa