Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Nature ; 511(7508): 184-90, 2014 Jul 10.
Article in English | MEDLINE | ID: mdl-24930766

ABSTRACT

Disease tolerance is the ability of the host to reduce the effect of infection on host fitness. Analysis of disease tolerance pathways could provide new approaches for treating infections and other inflammatory diseases. Typically, an initial exposure to bacterial lipopolysaccharide (LPS) induces a state of refractoriness to further LPS challenge (endotoxin tolerance). We found that a first exposure of mice to LPS activated the ligand-operated transcription factor aryl hydrocarbon receptor (AhR) and the hepatic enzyme tryptophan 2,3-dioxygenase, which provided an activating ligand to the former, to downregulate early inflammatory gene expression. However, on LPS rechallenge, AhR engaged in long-term regulation of systemic inflammation only in the presence of indoleamine 2,3-dioxygenase 1 (IDO1). AhR-complex-associated Src kinase activity promoted IDO1 phosphorylation and signalling ability. The resulting endotoxin-tolerant state was found to protect mice against immunopathology in Gram-negative and Gram-positive infections, pointing to a role for AhR in contributing to host fitness.


Subject(s)
Disease Resistance/genetics , Disease Resistance/immunology , Receptors, Aryl Hydrocarbon/metabolism , Animals , Bacterial Infections/immunology , Bacterial Infections/metabolism , Disease Resistance/drug effects , Endotoxemia/genetics , Endotoxemia/immunology , Endotoxemia/metabolism , Enzyme Activation/drug effects , Gene Expression Regulation/drug effects , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Inflammation/enzymology , Inflammation/genetics , Inflammation/metabolism , Kynurenine/metabolism , Lipopolysaccharides/pharmacology , Mice , Phosphorylation , Receptors, Aryl Hydrocarbon/genetics , Signal Transduction , Tryptophan Oxygenase/metabolism , src-Family Kinases/metabolism
2.
Arthritis Rheum ; 64(4): 1089-97, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22042442

ABSTRACT

OBJECTIVE: To assess the role of matrix metalloproteinase 2 (MMP-2) in the evolution of septic arthritis induced by group B streptococci (GBS) in mice. METHODS: Mice deficient in MMP-2 (MMP-2(-/-) ) and wild-type controls were injected intravenously with 1 × 10(7) colony-forming units of type IV GBS (strain 1/82). Levels of MMP-2, mortality rates, evolution of arthritis, bacterial clearance, joint histopathologic features, and production of cytokines and chemokines were examined in both experimental groups of mice on days 3, 6, and 9 after infection. RESULTS: MMP-2 was produced during GBS infection. Disruption of the gene for MMP-2 resulted in a decrease in the incidence and severity of arthritis, as demonstrated by both clinical and histologic findings, without affecting mortality rates. Amelioration of arthritis was accompanied by a dramatic reduction in the local production of interleukin-1ß (IL-1ß), IL-6, macrophage inflammatory protein 1α (MIP-1α), and MIP-2 and a reduced bacterial burden. CONCLUSION: MMP-2, produced early during GBS infection in mice, is involved in the degradation of extracellular matrix components at the level of the joint. This degradation is the first step in a cascade of events (joint invasion by GBS, extravasation and accumulation of inflammatory cells, proinflammatory cytokine production), all of which contribute to the damage of articular tissue. Thus, MMP-2 should be regarded as a potential therapeutic target in GBS-induced arthritis.


Subject(s)
Arthritis, Experimental/metabolism , Arthritis, Infectious/metabolism , Matrix Metalloproteinase 2/metabolism , Streptococcal Infections/metabolism , Streptococcus agalactiae , Animals , Arthritis, Experimental/microbiology , Arthritis, Experimental/pathology , Arthritis, Infectious/microbiology , Arthritis, Infectious/pathology , Female , Interleukin-1beta/metabolism , Joints/metabolism , Joints/pathology , Matrix Metalloproteinase 2/genetics , Mice , Mice, Knockout , Streptococcal Infections/pathology
3.
Microbes Infect ; 12(4): 302-8, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20114085

ABSTRACT

Group B streptococci have long been known as a leading cause of life-threatening infection in neonates, young infants and pregnant women, and recently have been recognized as an ever-growing cause of serious invasive infections in nonpregnant adults. B7-1 and B7-2 are two molecules with immunoregulatory functions implicated in the differentiation of T cells. The present study examined the role of B7-1 and B7-2 during group B streptococci-induced sepsis and arthritis. B7-1- or B7-2-deficient mice were infected with 1x10(7) streptococci, and mortality, appearance of arthritis, growth of microorganisms in the organs and cytokine profile were assessed. Lack of B7-1 was associated with amelioration of arthritis, while worsening of articular lesions was found in B7-2 deficient mice, in comparison to controls. Amelioration of arthritis in B7-1 deficient mice was accompanied by a lower local production of IL-1 beta and IL-18, and increase in IL-4 and IL-10 secretion. On the contrary, B7-2 deficient mice showed an higher proinflammatory cytokine production and lower IL-10 secretion than controls. Taken together, our results provide evidence that signaling delivered by B7-1 and B7-2 plays a role in determining the outcome of group B streptococcal induced arthritis, likely due to the different local secretory pattern.


Subject(s)
Arthritis, Infectious/immunology , Arthritis, Infectious/pathology , B7-1 Antigen/immunology , B7-2 Antigen/immunology , Streptococcus agalactiae/pathogenicity , Animals , Arthritis, Infectious/microbiology , B7-1 Antigen/genetics , B7-2 Antigen/genetics , Colony Count, Microbial , Cytokines/metabolism , Female , Mice , Mice, Inbred BALB C , Mice, Knockout , Sepsis/immunology , Sepsis/microbiology , Sepsis/pathology , Survival Analysis
4.
Mediators Inflamm ; 2009: 394021, 2009.
Article in English | MEDLINE | ID: mdl-19606256

ABSTRACT

IL-4 is an anti-inflammatory cytokine that inhibits the onset and severity in different experimental arthritis models. Group B streptococci (GBS) have been recognized as an ever-growing cause of serious invasive infections in nonpregnant adults. Septic arthritis is a clinical manifestation of GBS infection. To investigate the role of IL-4 in experimental GBS infection, IL-4 deficient or competent mice were inoculated with 1 x 10(7) GBS/mouse. Mortality, appearance of arthritis, GBS growth in the organs, and local and systemic cytokine and chemokine production were examined. IL-4-/- mice showed lower mortality rates but increased severity of arthritis and exhibited a lower microbial load in blood, kidneys, and joints than wt mice. Increased local levels of IL-1 beta, IL-6, TNF-alpha, MIP-1alpha, and MIP-2 accompanied the more severe arthritis in IL-4-/- mice. Our results suggest a detrimental role of IL-4 in GBS sepsis, whereas it plays a beneficial effect on GBS-induced arthritis.


Subject(s)
Arthritis, Infectious/immunology , Arthritis, Infectious/mortality , Interleukin-4 , Streptococcal Infections/immunology , Streptococcal Infections/mortality , Streptococcus agalactiae , Animals , Arthritis, Infectious/microbiology , Arthritis, Infectious/pathology , Chemokines/immunology , Cytokines/immunology , Humans , Interleukin-4/deficiency , Interleukin-4/genetics , Interleukin-4/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Streptococcal Infections/pathology , Streptococcus agalactiae/immunology , Streptococcus agalactiae/pathogenicity , Survival Rate
5.
Infect Immun ; 77(4): 1524-31, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19179417

ABSTRACT

Group B streptococcus (GBS) has been recognized as an ever-growing cause of serious invasive infections in nonpregnant adults, in particular, in association with severe underlying diseases. The most common manifestations include primary bacteremia, urinary tract infections, pneumonia, meningitis, peritonitis, and osteoarticular infections. Toll-like receptor-2 (TLR2) mediates host responses to gram-positive bacteria. TLR2 function was investigated in murine GBS-induced sepsis and arthritis in wild-type (wt) and TLR2-deficient (TLR2(-/-)) mice. Mice were infected with different doses of GBS (10(7), 5 x 10(6), or 10(6) CFU per mouse). Mortality, appearance of arthritis, GBS growth in the organs, and local and systemic cytokine and chemokine production were examined. TLR2(-/-) mice showed earlier and higher mortality rates and increased incidence and severity of arthritis than wt mice at all the infecting doses employed. Histopathological analysis of the joints confirmed clinical observations. TLR2(-/-) mice exhibited a higher microbial load in blood, kidneys, and joints than wt animals. In vitro experiments performed with peritoneal polymorphonuclear cells and macrophages showed a significantly lower bactericidal ability of cells from TLR2(-/-) mice. Increased systemic and local levels of interleukin-1beta (IL-1beta), IL-6, tumor necrosis factor alpha, macrophage inflammatory protein-1alpha (MIP-1alpha), and MIP-2 accompanied the more severe development of sepsis and arthritis in TLR2(-/-) mice. In conclusion, the lack of TLR2 was associated with an impaired host resistance to GBS infection, likely due to a diminished bacterial clearing and a consequent enhanced inflammatory response.


Subject(s)
Arthritis, Infectious , Sepsis , Severity of Illness Index , Streptococcus agalactiae/pathogenicity , Toll-Like Receptor 2/deficiency , Animals , Arthritis, Infectious/immunology , Arthritis, Infectious/microbiology , Arthritis, Infectious/mortality , Arthritis, Infectious/physiopathology , Cytokines/biosynthesis , Humans , Joints/microbiology , Kidney/microbiology , Mice , Mice, Inbred C57BL , Rabbits , Sepsis/immunology , Sepsis/microbiology , Sepsis/mortality , Sepsis/physiopathology , Streptococcal Infections/immunology , Streptococcal Infections/microbiology , Streptococcal Infections/physiopathology , Streptococcus agalactiae/growth & development , Streptococcus agalactiae/isolation & purification
6.
J Bacteriol ; 190(6): 1956-65, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18203834

ABSTRACT

CsrRS (or CovRS) is a two-component regulatory system that controls expression of multiple virulence factors in the important human pathogen group B Streptococcus (GBS). We now report global gene expression studies in GBS strains 2603V/R and 515 and their isogenic csrR and csrS mutants. Together with data reported previously for strain NEM316, the results reveal a conserved 39-gene CsrRS regulon. In vitro phosphorylation-dependent binding of recombinant CsrR to promoter regions of both positively and negatively regulated genes suggests that direct binding of CsrR can mediate activation as well as repression of target gene expression. Distinct patterns of gene regulation in csrR versus csrS mutants in strain 2603V/R compared to 515 were associated with different hierarchies of relative virulence of wild-type, csrR, and csrS mutants in murine models of systemic infection and septic arthritis. We conclude that CsrRS regulates a core group of genes including important virulence factors in diverse strains of GBS but also displays marked variability in the repertoire of regulated genes and in the relative effects of CsrS signaling on CsrR-mediated gene regulation. Such variation is likely to play an important role in strain-specific adaptation of GBS to particular host environments and pathogenic potential in susceptible hosts.


Subject(s)
Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Regulon/genetics , Streptococcus agalactiae/genetics , Animals , Bacterial Proteins/metabolism , Bacterial Proteins/physiology , Electrophoretic Mobility Shift Assay , Gene Expression Profiling , Male , Mice , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Streptococcal Infections/microbiology , Streptococcus agalactiae/pathogenicity , Virulence/genetics
7.
Microbes Infect ; 8(9-10): 2376-83, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16844399

ABSTRACT

Group B streptococci (GBS) have been recognised as an ever-growing cause of serious invasive infections in non-pregnant adults, in particular in association with severe underlying diseases such as diabetes mellitus. In the present study we used mice rendered diabetic to gain further insights into host-pathogen interaction during induced GBS sepsis and septic arthritis. Type I diabetes was induced in adult CD-1 mice by low-dose streptozotocin treatment. Mice were then infected with different doses of GBS, and mortality, appearance of arthritis, growth of microorganisms in the organs and cytokine and chemokine profile were assessed in diabetic and control animals. The LD50 was significantly lower in diabetics than in controls, while both incidence and severity of arthritis were higher. A significantly higher number of microorganisms were recovered from the organs of diabetic mice than in controls. The worsening of sepsis and arthritis was associated with a significant increase in systemic and local production of IL-6, IL-1 beta, TNF-alpha, IL-10, macrophage inflammatory protein 1 alpha (MIP-1alpha), and MIP-2 and with a decrease in IFN-gamma production. Taken together, our results indicate an impaired host resistance to GBS infection in diabetics, likely due to a dysregulation of the cytokine network and prolonged local inflammatory response.


Subject(s)
Arthritis/microbiology , Bacteremia/blood , Diabetes Mellitus, Experimental/microbiology , Streptococcal Infections/complications , Streptococcus agalactiae/isolation & purification , Animals , Arthritis/immunology , Arthritis/pathology , Bacteremia/immunology , Bacteremia/microbiology , Chemokine CCL3 , Chemokine CCL4 , Chemokine CXCL2 , Chemokines/immunology , Diabetes Mellitus, Experimental/immunology , Interferon-gamma/immunology , Interleukin-10/immunology , Interleukin-1beta/immunology , Interleukin-6/immunology , Macrophage Inflammatory Proteins/immunology , Male , Mice , Streptococcal Infections/immunology , Streptococcal Infections/microbiology , Tumor Necrosis Factor-alpha/immunology
8.
J Med Microbiol ; 55(Pt 2): 229-235, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16434717

ABSTRACT

Corynebacterium diphtheriae is a well-known cause of localized respiratory tract infections. However, this micro-organism can also be associated with invasive infections, such as endocarditis, septic arthritis and osteomyelitis. Invasive infections are often caused by non-toxigenic strains. To set up an in vivo experimental model of C. diphtheriae infection, mice were infected intravenously with different doses (ranging from 1 x 10(7) to 5 x 10(8) bacteria per mouse) of three non-toxigenic strains, namely ISS-4749, ISS-4746 and ISS-3319. Similar mortality rates were observed with the three strains, with an LD50 ranging from 9 x 10(7) to 1.2 x 10(8). All strains were arthritogenic, although to different extents. ISS-4749 and ISS-4746 infection resulted in a maximum of 60 and 50 %, respectively, of animals with articular lesions, while in the ISS-3319-infected group only 25 % were positive. There were differences in systemic and joint cytokine production in the three experimental groups. ISS-4749- and ISS-4746-infected mice exhibited higher local levels of interleukin (IL)-6 and IL-1beta than ISS-3319-infected animals. At systemic levels, ISS-3319 was able to induce early and sustained production of interferon-gamma (IFN-gamma), but not IL-6. Conversely, infection with the other strains resulted in high IL-6, but not IFN-gamma, production. In conclusion, an experimental model of C. diphtheriae infection was set up, with development of septic arthritis. This model could be useful in studies on the pathogenicity and characterization of virulence factors other than toxin production.


Subject(s)
Arthritis, Infectious/immunology , Corynebacterium diphtheriae , Animals , Arthritis, Infectious/pathology , Cytokines/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Interferon-gamma/analysis , Interferon-gamma/metabolism , Interleukin-1/analysis , Interleukin-1/metabolism , Interleukin-6/analysis , Interleukin-6/metabolism , Joints/immunology , Joints/pathology , Male , Mice
9.
Infect Immun ; 72(11): 6367-72, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15501766

ABSTRACT

Glucuronoxylomannan (GXM), the principal constituent of the Cryptococcus neoformans capsule, modulates the inflammatory response of human monocytes in vitro. Here we examine the efficacy of GXM as a novel anti-inflammatory compound for use against experimental septic arthritis. Arthritis was induced in mice by the intravenous injection of 8 x 10(6) CFU of type IV group B streptococcus (GBS). GXM was administered intravenously in different doses (50, 100, or 200 microg/mouse) 1 day before and 1 day after bacterial inoculation. GXM treatment markedly decreased the incidence and severity of articular lesions. Histological findings showed limited periarticular inflammation in the joints of GXM-treated mice, confirming the clinical observations. The amelioration of arthritis was associated with a significant reduction in the local production of interleukin-6 (IL-6), IL-1beta, macrophage inflammatory protein 1alpha (MIP-1alpha), and MIP-2 and an increase in systemic IL-10 levels. Moreover, peritoneal macrophages derived from GXM-treated mice and stimulated in vitro with heat-inactivated GBS showed a similar pattern of cytokine production. The present study provides evidence for the modulation of the inflammatory response by GXM in vivo and suggests a potential therapeutic use for this compound in pathologies involving inflammatory processes.


Subject(s)
Arthritis, Infectious/drug therapy , Cryptococcus neoformans/chemistry , Polysaccharides/therapeutic use , Streptococcal Infections/drug therapy , Streptococcus agalactiae/drug effects , Animals , Animals, Outbred Strains , Arthritis, Infectious/immunology , Arthritis, Infectious/mortality , Arthritis, Infectious/pathology , Cytokines/metabolism , Disease Models, Animal , Female , Mice , Polysaccharides/administration & dosage , Severity of Illness Index , Streptococcal Infections/immunology , Streptococcal Infections/mortality , Streptococcal Infections/pathology , Streptococcus agalactiae/pathogenicity , Treatment Outcome
10.
Proc Natl Acad Sci U S A ; 101(40): 14491-6, 2004 Oct 05.
Article in English | MEDLINE | ID: mdl-15381763

ABSTRACT

Group B Streptococcus (GBS) is a major cause of pneumonia, bacteremia, and meningitis in neonates and has been found to persist inside host phagocytic cells. The pore-forming GBS beta-hemolysin/cytolysin (betaH/C) encoded by cylE is an important virulence factor as demonstrated in several in vivo models. Interestingly, cylE deletion results not only in the loss of betaH/C activity, but also in the loss of a carotenoid pigment of unknown function. In this study, we sought to define the mechanism(s) by which cylE may contribute to GBS phagocyte resistance and increased virulence potential. We found that cylE-deficient GBS was more readily cleared from a mouse's bloodstream, human whole blood, and isolated macrophage and neutrophil cultures. Survival was linked to the ability of betaH/C to induce cytolysis and apoptosis of the phagocytes. At a lower bacterial inoculum, cylE also contributed to enhanced survival within phagocytes that was attributed to the ability of carotenoid to shield GBS from oxidative damage. In oxidant killing assays, cylE mutants were shown to be more susceptible to hydrogen peroxide, hypochlorite, superoxide, and singlet oxygen. Together, these data suggest a mechanism by which the linked cylE-encoded phenotypes, betaH/C (sword) and carotenoid (shield), act in partnership to thwart the immune phagocytic defenses.


Subject(s)
Carotenoids/toxicity , Cytotoxins/toxicity , Hemolysin Proteins/toxicity , Phagocytes/drug effects , Phagocytes/immunology , Streptococcus agalactiae/pathogenicity , Animals , Apoptosis , Bacterial Proteins , Base Sequence , Carotenoids/genetics , Cell Line , Cytotoxins/genetics , DNA, Bacterial/genetics , Genes, Bacterial , Hemolysin Proteins/genetics , Humans , Mice , Mutation , Phagocytes/pathology , Reactive Oxygen Species/metabolism , Sepsis/immunology , Sepsis/pathology , Streptococcal Infections/immunology , Streptococcal Infections/pathology , Streptococcus agalactiae/genetics , Virulence/genetics , Virulence/physiology
12.
Arthritis Rheum ; 50(6): 2005-13, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15188378

ABSTRACT

OBJECTIVE: To assess the role of interleukin-18 (IL-18) in the evolution of septic arthritis induced by group B streptococci (GBS) in mice. METHODS: CD1 mice were inoculated intravenously with 8 x 10(6) colony-forming units (CFU) of type IV GBS (strain 1/82), and administered intraperitoneally 1 hour before infection with anti-IL-18 monoclonal antibodies (0.25 mg/mouse). In a subsequent set of experiments, mice infected with a suboptimal arthritogenic dose of GBS (4 x 10(6) CFU/mouse) were administered different doses of recombinant IL-18 for 4 days, starting 1 hour after infection. Mortality, evolution of arthritis, bacterial clearance, joint histopathology, and cytokine production were examined in infected mice that did or did not receive treatment with anti-IL-18 antibodies or IL-18. RESULTS: IL-18 was produced during GBS infection. Neutralization of IL-18 resulted in a decrease in mortality rates, and in the incidence and severity of arthritis. Amelioration of arthritis was accompanied by a dramatic reduction in local IL-1 beta, IL-6, macrophage inflammatory protein 1 alpha (MIP-1 alpha) and MIP-2 production, and reduced bacterial burden. Administration of exogenous IL-18 resulted in increased mortality rates and increased incidence and severity of GBS arthritis, concomitant with a higher number of GBS and increased levels of IL-6, IL-1 beta, MIP-1 beta, and MIP-2 production in the joints. CONCLUSION: The present study indicated some involvement of IL-18 in the pathogenesis of GBS-induced arthritis. The role of IL-18 in joint pathology is shown by a regulatory effect on inflammatory mediator levels and local cell influx. Thus, IL-18 should be regarded as a potential therapeutic target in GBS infection and arthritis.


Subject(s)
Arthritis, Infectious/immunology , Interleukin-18/immunology , Streptococcal Infections/immunology , Streptococcus agalactiae/immunology , Animals , Antibodies/pharmacology , Arthritis, Infectious/microbiology , Arthritis, Infectious/mortality , Cytokines/metabolism , Female , Incidence , Male , Mice , Mice, Inbred Strains , Severity of Illness Index , Streptococcal Infections/complications , Streptococcal Infections/mortality
13.
Cell Microbiol ; 4(10): 691-700, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12366405

ABSTRACT

Septic arthritis is a clinical manifestation of group B Streptococcus (GBS) infection in both neonates and adults. Because macrophages are known to participate in tissue injury, the role of this cell population in GBS-induced arthritis was investigated. Mice were rendered monocytopenic by administration of etoposide, a drug that selectively depletes the monocyte/macrophage population and then injected with GBS (1 x 10(7) colony-forming units per mouse). Appearance of arthritis, mortality, GBS growth in the organs, and local and systemic cytokine production were examined. Etoposide-treated mice had a significantly less severe arthritis than control animals. Histopathological analysis of the joints confirmed clinical observations. Decreased joint levels of the proinflammatory cytokines interleukin 1 (IL-1) beta and IL-6 accompanied the less severe development of arthritis in monocytopenic mice. In contrast, mortality was increased in the etoposide-treated mice compared with controls. Monocytopenic mice exhibited elevated bacterial load in the blood and kidneys at all time points examined. These results indicate that lack of macrophages leads to less severe joint lesions, but also results in impaired clearance of bacteria, and consequent enhancement of mortality rates.


Subject(s)
Arthritis, Infectious/immunology , Macrophages/physiology , Streptococcal Infections/immunology , Streptococcus agalactiae , Animals , Etoposide/pharmacology , Female , Interleukin-1/metabolism , Interleukin-6/metabolism , Joints/microbiology , Kidney/microbiology , Macrophages/immunology , Male , Mice , Monocytes/drug effects , Monocytes/metabolism , Nucleic Acid Synthesis Inhibitors/pharmacology , Streptococcal Infections/microbiology , Streptococcal Infections/pathology , Streptococcus agalactiae/isolation & purification , Tumor Necrosis Factor-alpha/metabolism
14.
Infect Immun ; 70(6): 2862-8, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12010973

ABSTRACT

Intravenous inoculation of CD-1 mice with 10(7) CFU of type IV group B Streptococcus (GBS) results in a high incidence of diffuse septic arthritis, associated with high levels of systemic and local production of interleukin-1beta (IL-1beta) and IL-6. In this study, the role of the anti-inflammatory cytokine IL-10 in the evolution of GBS systemic infection and arthritis was evaluated. IL-10 production was evident in sera and joints of GBS-infected mice. Neutralization of endogenous IL-10 by administration of anti-IL-10 antibodies (1 mg/mouse) at the time of infection resulted in worsening of articular lesions and 60% mortality associated with early sustained production of IL-6, IL-1beta, and tumor necrosis factor alpha (TNF-alpha). The effect of IL-10 supplementation was assessed by administering IL-10 (100, 200, or 400 ng/mouse) once a day for 5 days, starting 1 h after infection. Treatment with IL-10 had a beneficial effect on GBS arthritis, and there was a clear-cut dose dependence. The decrease in pathology was associated with a significant reduction in IL-6, IL-1beta, and TNF-alpha production. Histological findings showed limited periarticular inflammation and a few-cell influx in the articular cavity of IL-10-treated mice, confirming clinical observations. In conclusion, this study provides further information concerning the role of IL-10 in regulating the immune response and inflammation and calls attention to the potential therapeutic use of IL-10 in GBS arthritis.


Subject(s)
Arthritis, Infectious/immunology , Interleukin-10/immunology , Streptococcal Infections/immunology , Streptococcus agalactiae/immunology , Animals , Arthritis, Infectious/blood , Arthritis, Infectious/mortality , Disease Models, Animal , Female , Interleukin-1/immunology , Interleukin-10/administration & dosage , Interleukin-10/blood , Interleukin-6/immunology , Male , Mice , Streptococcal Infections/blood , Streptococcal Infections/mortality , Tumor Necrosis Factor-alpha/immunology
15.
Arthritis Rheum ; 46(3): 806-17, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11920419

ABSTRACT

OBJECTIVE: To assess the effect of interleukin-12 (IL-12) administration on the evolution of systemic infection and septic arthritis induced by group B streptococci (GBS) in mice. METHODS: CD1 mice were inoculated intravenously with arthritogenic strain 1/82 of type IV GBS. Exogenous murine IL-12 was administered intraperitoneally 18 hours or 5 days after infection with 1 x 10(7) GBS, at doses ranging from 0.5 to 2.5 microg per mouse. Mice were monitored daily for survival and for signs of arthritis. In a subsequent set of experiments, mice were killed at selected times for examination of bacterial clearance, histopathologic changes in the joints, and cytokine production. RESULTS: IL-12 administration before the onset of clinical signs had a beneficial effect on GBS-induced arthritis and was clearly dose-dependent. The 2.5-microg dose per mouse totally prevented death from GBS-induced arthritis. The decrease in pathology was associated with a reduction of the bacterial burden and a change in the cytokine profile. In particular, systemic and joint levels of interferon-gamma (IFN gamma) and IL-10 significantly increased in mice treated with IL-12, whereas a decrease in IL-6 and IL-1 beta production was observed. The beneficial effects of IL-12, in terms of the incidence and severity of articular lesions, were reversed by coadministration of anti-IFN gamma or anti-IL-10-neutralizing antibodies. CONCLUSION: The findings of this study demonstrate that IL-12 is important in controlling the cytokine production that leads to the evolution of GBS-induced experimental arthritis. The amelioration of articular lesions is mostly attributable to IL-12-induced IFN gamma, but with a relevant participation of IL-12-induced IL-10.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Arthritis, Infectious/drug therapy , Arthritis, Infectious/microbiology , Interferon-gamma/biosynthesis , Interleukin-10/biosynthesis , Interleukin-12/therapeutic use , Streptococcal Infections , Streptococcus agalactiae , Animals , Animals, Outbred Strains , Arthritis, Infectious/mortality , Arthritis, Infectious/physiopathology , Blood/microbiology , Female , Interferon-gamma/physiology , Interleukin-10/physiology , Joints/drug effects , Joints/metabolism , Joints/microbiology , Leukocyte Count , Mice , Streptococcus agalactiae/isolation & purification
16.
J Med Microbiol ; 48(11): 983-990, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10535641

ABSTRACT

Strains of a new polysaccharide type of group B streptococci (GBS), type VII, have been isolated from human carriers and invasive infections. Some of these strains bear the protein antigen c or R, as do other GBS serotypes. The capsular type polysaccharide is sialylated and this residue is involved in the immunodeterminant structure. All type VII strains examined were virulent in CD-1 mice; the LD50 after intraperitoneal (i.p.) challenge was 4.57 (SD 0.12) x10(7) cfu for the reference strain and 5.49 (SD 1.5) x10(7) cfu for clinical isolates. A particular feature of this serotype was the ability to induce septic arthritis not only when injected intravenously (i.v.), but also when injected i.p. Rabbit antiserum against the capsular type VII polysaccharide exhibited opsonic activity in a phagocytosis assay and protective activity against infection.


Subject(s)
Arthritis, Infectious/microbiology , Bacterial Capsules/immunology , Polysaccharides, Bacterial/immunology , Streptococcal Infections/microbiology , Streptococcus agalactiae/pathogenicity , Animals , Antibody Specificity , Bacterial Capsules/chemistry , Cross Reactions , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immune Sera/immunology , Immunodiffusion , Immunoelectrophoresis , Mice , Microscopy, Immunoelectron , N-Acetylneuraminic Acid/analysis , Phagocytosis , Polysaccharides, Bacterial/chemistry , Rabbits , Streptococcus agalactiae/immunology , Streptococcus agalactiae/ultrastructure , Virulence
17.
Clin Microbiol Infect ; 3(3): 324-328, 1997 Jun.
Article in English | MEDLINE | ID: mdl-11864128

ABSTRACT

OBJECTIVE: To study the prevalence of group B Streptococcus (GBS) colonization in pregnant women and their newborns at Perugia General Hospital. METHODS: The number of mother---child pairs examined was 2300. Vaginal swabs were collected from the mothers at delivery, and auricular and pharyngeal swabs and gastric aspirate from the newborns at birth. Maternal risk factors for GBS disease, including premature delivery, intrapartum fever, prolonged rupture of membranes and multiple births, were evaluated. RESULTS: Maternal and neonatal colonization rates were 11.3% and 4.6%, respectively. GBS was isolated in 41.5% of the neonates born to colonized mothers and in 0.1% of those born to non-colonized mothers. No significant difference was observed in vertical transmission rates in the presence or absence of maternal risk factors. The external auditory canal was the most frequent (93.5%) and heavily colonized body site. Type Ib was the most common serotype among GBS isolates from mothers and babies. C surface protein was not detected in serotype V and VIII isolates, but was frequent in all other serotypes. Early-onset disease was observed in 0.4/1000 live births. CONCLUSIONS: The prevalence of maternal and neonatal colonization at Perugia General Hospital was similar to that obtained in other studies performed in Italy. The external auditory canal was confirmed as the most reliable body site to be sampled for the detection of neonates exposed to maternal GBS colonization.

SELECTION OF CITATIONS
SEARCH DETAIL
...