Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 149
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Crit Care ; 27(1): 374, 2023 09 29.
Article in English | MEDLINE | ID: mdl-37773186

ABSTRACT

BACKGROUND AND AIMS: The triggering factors of sepsis-induced myocardial dysfunction (SIMD) are poorly understood and are not addressed by current treatments. S100A8/A9 is a pro-inflammatory alarmin abundantly secreted by activated neutrophils during infection and inflammation. We investigated the efficacy of S100A8/A9 blockade as a potential new treatment in SIMD. METHODS: The relationship between plasma S100A8/A9 and cardiac dysfunction was assessed in a cohort of 62 patients with severe sepsis admitted to the intensive care unit of Linköping University Hospital, Sweden. We used S100A8/A9 blockade with the small-molecule inhibitor ABR-238901 and S100A9-/- mice for therapeutic and mechanistic studies on endotoxemia-induced cardiac dysfunction in mice. RESULTS: In sepsis patients, elevated plasma S100A8/A9 was associated with left-ventricular (LV) systolic dysfunction and increased SOFA score. In wild-type mice, 5 mg/kg of bacterial lipopolysaccharide (LPS) induced rapid plasma S100A8/A9 increase and acute LV dysfunction. Two ABR-238901 doses (30 mg/kg) administered intraperitoneally with a 6 h interval, starting directly after LPS or at a later time-point when LV dysfunction is fully established, efficiently prevented and reversed the phenotype, respectively. In contrast, dexamethasone did not improve cardiac function compared to PBS-treated endotoxemic controls. S100A8/A9 inhibition potently reduced systemic levels of inflammatory mediators, prevented upregulation of inflammatory genes and restored mitochondrial function in the myocardium. The S100A9-/- mice were protected against LPS-induced LV dysfunction to an extent comparable with pharmacologic S100A8/A9 blockade. The ABR-238901 treatment did not induce an additional improvement of LV function in the S100A9-/- mice, confirming target specificity. CONCLUSION: Elevated S100A8/A9 is associated with the development of LV dysfunction in severe sepsis patients and in a mouse model of endotoxemia. Pharmacological blockade of S100A8/A9 with ABR-238901 has potent anti-inflammatory effects, mitigates myocardial dysfunction and might represent a novel therapeutic strategy for patients with severe sepsis.


Subject(s)
Endotoxemia , Heart Diseases , Ventricular Dysfunction, Left , Humans , Mice , Animals , Endotoxemia/complications , Endotoxemia/drug therapy , Lipopolysaccharides , Calgranulin A/physiology , Calgranulin B/genetics , Myocardium , Inflammation/drug therapy
2.
BMC Gastroenterol ; 21(1): 337, 2021 Aug 28.
Article in English | MEDLINE | ID: mdl-34454419

ABSTRACT

BACKGROUND: Most patients with acute pancreatitis (AP) experience mild, self-limiting disease with little or no need for hospital care. However, 20-25% of patients develop a more severe and potentially life-threatening condition with progressive systemic inflammatory response syndrome (SIRS) and multiorgan failure, resulting in high morbidity and mortality rates. Predicting disease severity at an early stage is important, as immediate supportive care has been demonstrated to reduce the incidence of SIRS and organ failure, improving patient outcome. Several studies have demonstrated elevated levels of heparin-binding protein (HBP) in patients with sepsis and septic shock, and HBP is believed to play a part in endothelial dysfunction leading to vascular leakage. As HBP levels increase prior to other known biomarkers, HBP has emerged as a promising early predictor of severe sepsis with organ dysfunction. METHODS: Patients admitted to Skåne University Hospital in Malmö between 2010 and 2013 fulfilling the criteria for AP were identified in the emergency department and prospectively enrolled in this study. The primary outcome was measured levels of HBP upon hospital admission in patients with confirmed AP. Correlations among HBP concentrations, disease severity and fluid balance were considered secondary endpoints. The correlation between HBP levels and fluid balance were analysed using Pearson correlation, and the ability of HBP to predict moderately severe/severe AP was assessed using a receiver operating characteristic (ROC) curve. RESULTS: The overall median HBP level in this study was 529 (307-898) ng/ml. There were no significant group differences in HBP levels based on AP severity. Fluid balance differed significantly between patients with mild versus moderately severe and severe pancreatitis, but we found no correlation between HBP concentration and fluid balance. CONCLUSIONS: HBP levels are dramatically increased in patients with AP, and these levels far exceed those previously reported in other conditions. In this study, we did not observe any significant correlation between HBP levels and disease severity or the need for intravenous fluid. Additional studies on HBP are needed to further explore the role of HBP in the pathogenesis of AP and its possible clinical implications.


Subject(s)
Pancreatitis , Acute Disease , Antimicrobial Cationic Peptides , Blood Proteins , Carrier Proteins , Humans
3.
Crit Rev Microbiol ; 46(2): 121-135, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32141788

ABSTRACT

Hemostasis is a defence mechanism that protects the integrity of the vascular system and is comprised of the coagulation cascade, fibrinolysis, platelet aggregation, and vascular endothelium. Besides the primary function in preserving the vascular integrity, the haemostatic system cooperates with immune and inflammatory processes to eliminate invading pathogens during microbial infections. Under pathological manifestations, hemostasis must therefore interact in a coordinated manner with inflammatory responses and immune reactions. Several pathogens can modulate these host-derived countermeasures by specifically targeting certain haemostatic components for their own benefit. Thus, the ability to modulate host defence systems has to be considered as an essential bacterial virulence mechanism. Complications that bacterial pathogens can induce are therefore often the consequence of evoked host responses. A comprehensive understanding of the molecular mechanisms triggered in infectious processes may help to develop prophylactic methods and novel therapies for the patients suffering from a particular infectious disease. This review aims to provide a critical updated compiling of recent studies on how the pathogenic Leptospira can interact with and manipulate the host haemostatic systems and the consequences for leptospirosis pathogenesis.


Subject(s)
Hemostasis , Leptospira/physiology , Leptospirosis/blood , Animals , Fibrinolysis , Host-Pathogen Interactions , Humans , Leptospira/genetics , Leptospirosis/microbiology
4.
FASEB J ; 33(2): 2599-2609, 2019 02.
Article in English | MEDLINE | ID: mdl-30281335

ABSTRACT

Neutrophil recruitment and plasma exudation are key elements in the immune response to injury or infection. Activated neutrophils stimulate opening of the endothelial barrier; however, the underlying mechanisms have remained largely unknown. In this study, we identified a pivotal role of the proinflammatory kallikrein-kinin system and consequent formation of bradykinin in neutrophil-evoked vascular leak. In mouse and hamster models of acute inflammation, inhibitors of bradykinin generation, and signaling markedly reduced plasma exudation in response to chemoattractant activation of neutrophils. The neutrophil-driven leak was likewise suppressed in mice deficient in either the bradykinin B2 receptor or factor XII (initiator of the kallikrein-kinin system). In human endothelial cell monolayers, material secreted from activated neutrophils induced cytoskeletal rearrangement, leading to paracellular gap formation in a bradykinin-dependent manner. As a mechanistic basis, we found that a neutrophil-derived heparin-binding protein (HBP/azurocidin) displaced the bradykinin precursor high-molecular-weight kininogen from endothelial cells, thereby enabling proteolytic processing of kininogen into bradykinin by neutrophil and plasma proteases. These data provide novel insight into the signaling pathway by which neutrophils open up the endothelial barrier and identify the kallikrein-kinin system as a target for therapeutic interventions in acute inflammatory reactions.-Kenne, E., Rasmuson, J., Renné, T., Vieira, M. L., Müller-Esterl, W., Herwald, H., Lindbom, L. Neutrophils engage the kallikrein-kinin system to open up the endothelial barrier in acute inflammation.


Subject(s)
Cell Membrane Permeability , Endothelium, Vascular/physiology , Inflammation/pathology , Kallikrein-Kinin System/physiology , Neutrophils/metabolism , Pulmonary Edema/pathology , Animals , Bradykinin/metabolism , Endothelium, Vascular/cytology , Factor XII/metabolism , Female , Humans , Inflammation/metabolism , Kininogen, High-Molecular-Weight/metabolism , Male , Mesocricetus , Mice , Mice, Inbred C57BL , Neutrophil Infiltration , Pulmonary Edema/etiology , Pulmonary Edema/metabolism
5.
J Infect Dis ; 219(6): 996-1006, 2019 02 23.
Article in English | MEDLINE | ID: mdl-30299510

ABSTRACT

BACKGROUND: Leptospirosis, caused by spirochetes of the genus Leptospira, is one of the most widespread zoonoses worldwide. Efficient diagnostic methods for early diagnosis of leptospirosis are still lacking, and acute disease presents with nonspecific symptomatology and is often misdiagnosed. The leptospires pathogenic processes and virulence mechanisms remain virtually unknown. In severe infections, hemostatic impairment is frequently observed, and pathophysiological complications often develop when the host response is modulated by the pathogen. The neutrophil heparin-binding protein (HBP) is an inflammatory mediator and potent inducer of vascular leakage. RESULTS: In this study, we found that leptospires and their secreted products induce the release of HBP from stimulated neutrophils through a controlled degranulation mechanism. We acknowledged 2 leptospiral proteins as able to induce HBP degranulation. These findings have clinical implications, as high levels of HBP were detected in serum from patients with leptospirosis, especially at the early phase of the disease. CONCLUSION: In conclusion, we describe a new mechanism by which the leptospirosis pathophysiological complications may arise, such as vascular leakage and edema formation. We also propose HBP as a new early screening biomarker for human leptospirosis.


Subject(s)
Antimicrobial Cationic Peptides/blood , Bacterial Proteins/blood , Leptospira/pathogenicity , Leptospirosis/blood , Animals , Antimicrobial Cationic Peptides/pharmacology , Bacterial Proteins/pharmacology , Biomarkers/blood , Blood Proteins/pharmacology , Host-Pathogen Interactions , Humans , Leptospira/metabolism , Leptospirosis/diagnosis , Leptospirosis/physiopathology , Mice, Inbred BALB C , Muscle, Skeletal/blood supply , Muscle, Skeletal/drug effects , Neutrophils/drug effects , Neutrophils/metabolism , Recombinant Proteins/pharmacology
6.
Allergy ; 74(4): 663-674, 2019 04.
Article in English | MEDLINE | ID: mdl-30362569

ABSTRACT

BACKGROUND: In atopic asthma, chronic Th2-biased inflammation is associated with an increased risk of pneumococcal infection. The anionic phosphoglycoprotein osteopontin (OPN) is highly expressed in asthma and has been ascribed several roles during inflammation. This study aimed to investigate whether OPN affects inflammation and vulnerability to pneumococcal infection in atopic asthma. METHODS: House dust mite (HDM) extract was used to induce allergic airway inflammation in both wild-type (Spp1+/+ ) and OPN knockout (Spp1-/- ) C57BL/6J mice, and the airway was then infected with Streptococcus pneumoniae. Parameters reflecting inflammation, tissue injury, and bacterial burden were measured. In addition, samples from humans with allergic asthma were analyzed. RESULTS: Both allergen challenge in individuals with allergic asthma and the intranasal instillation of HDM in mice resulted in increased OPN levels in bronchoalveolar lavage fluid (BALF). More immune cells (including alveolar macrophages, neutrophils, eosinophils, and lymphocytes) and higher levels of proinflammatory cytokines were found in Spp1-/- mice than in Spp1+/+ mice. Moreover, OPN-deficient mice exhibited increased levels of markers reflecting tissue injury. Upon infection with S. pneumoniae, Spp1+/+ mice with allergic airway inflammation had a significantly lower bacterial burden in both BALF and lung tissue than did Spp1-/- mice. Furthermore, Spp1-/- mice had higher levels of cytokines and immune cells in BALF than did Spp1+/+ mice. CONCLUSION: OPN reduces inflammation, decreases tissue injury, and reduces bacterial loads during concurrent pneumococcal infection and allergic airway inflammation in a murine model. These findings suggest that OPN significantly affects vulnerability to pneumococcal infection in atopic asthma.


Subject(s)
Asthma/complications , Osteopontin/pharmacology , Pneumococcal Infections/prevention & control , Animals , Asthma/chemically induced , Asthma/microbiology , Bacterial Load/drug effects , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/immunology , Disease Models, Animal , Humans , Inflammation/chemically induced , Lung Injury/drug therapy , Lung Injury/prevention & control , Mice , Mice, Knockout , Osteopontin/genetics , Pneumococcal Infections/drug therapy , Protective Agents/pharmacology , Pyroglyphidae/immunology
7.
Cell Microbiol ; 20(1)2018 01.
Article in English | MEDLINE | ID: mdl-28973822

ABSTRACT

Streptococcus pyogenes of the M1 serotype is commonly associated with invasive streptococcal infections and development of streptococcal toxic shock syndrome. The M1 protein is a powerful inducer of inflammatory responses for several human cell types, but the reason why M1 protein-related strains is over-represented in invasive streptococcal diseases is still not understood. This study was undertaken to investigate if soluble M1 protein can aggravate the severity of streptococcal skin infections in respect to inflammation, leucocyte recruitment, and tissue remodelling as seen in patients with cellulitis and necrotizing fasciitis. We found that HaCaT cells are able to recruit activated leucocytes when encountering M1 protein. Neither the bacterial protein nor activated leucocytes caused cell damage on HaCaT cells, instead HaCaT cells responded to the bacterial virulence factor by releasing several proteins protective against bacterial infection and leucocyte responses. However, although not cytotoxic, M1 protein completely abolished wound healing abilities of HaCaT cells. Taken together, our results demonstrate that M1 protein is a critical virulence factor that can augment streptococcal skin infection suggesting that the protein is an interesting target for drug development.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/immunology , Carrier Proteins/immunology , Cellulitis/pathology , Fasciitis, Necrotizing/pathology , Leukocytes/immunology , Streptococcal Infections/pathology , Streptococcus pyogenes/pathogenicity , Cell Line , Cell Movement/physiology , Cell Proliferation , Cellulitis/microbiology , Fasciitis, Necrotizing/microbiology , Humans , Keratinocytes , Streptococcal Infections/microbiology , Streptococcus pyogenes/metabolism , Virulence Factors
8.
Biochem J ; 475(4): 775-786, 2018 02 23.
Article in English | MEDLINE | ID: mdl-29358188

ABSTRACT

The protein gC1qR (globular C1q receptor), also named p33, was originally identified as a binding partner of the globular heads of C1q in the complement system. gC1qR/p33 is abundantly expressed in many cell types, but the functional importance of this protein is not completely understood. Here, we investigate the impact of gC1qR/p33 on the production and function of the pathophysiologically important chemokine monocyte chemoattractant protein-1 (MCP-1) and the underlying molecular mechanisms. Knockdown of gC1qR/p33 negatively regulated the production of MCP-1, but had no effect on the expression of transcript for MCP-1 in human periodontal ligament cells, suggesting a translational/post-translational mechanism of action. Laser scanning confocal microscopy showed considerable cytosolic co-localization of gC1qR/p33 and MCP-1, and co-immunoprecipitation disclosed direct physical interaction between gC1qR/p33 and MCP-1. Surface plasmon resonance analysis revealed a high-affinity binding (KD = 10.9 nM) between gC1qR/p33 and MCP-1. Using a transwell migration assay, we found that recombinant gC1qR/p33 enhances MCP-1-induced migration of human THP-1 monocytes, pointing to a functional importance of the interaction between gC1qR/p33 and MCP-1. An in vitro assay revealed a rapid turnover of the MCP-1 protein and that gC1qR/p33 stabilizes MCP-1, hence preventing its degradation. We propose that endogenous gC1qR/p33 physically interacts with MCP-1 causing stabilization of the MCP-1 protein and stimulation of its activity in human periodontal ligament cells, suggesting a novel gC1qR/p33-mediated pro-inflammatory mechanism of action.


Subject(s)
Carrier Proteins/genetics , Chemokine CCL2/genetics , Inflammation/genetics , Mitochondrial Proteins/genetics , Periodontal Ligament/metabolism , Carrier Proteins/metabolism , Cell Movement/genetics , Chemokine CCL2/biosynthesis , Chemokine CCL2/chemistry , Cytosol/chemistry , Cytosol/metabolism , Gene Expression Regulation , Humans , Inflammation/metabolism , Inflammation/pathology , Microscopy, Confocal , Mitochondrial Proteins/metabolism , Monocytes/metabolism , Monocytes/pathology , Periodontal Ligament/growth & development , Periodontal Ligament/pathology , Protein Binding , Protein Processing, Post-Translational/genetics , Surface Plasmon Resonance
9.
J Proteome Res ; 17(1): 600-617, 2018 01 05.
Article in English | MEDLINE | ID: mdl-29160079

ABSTRACT

Streptococcus pyogenes is a major global health burden causing a wide variety of diseases. Because a vaccine against this bacterium is still lacking, vaccine candidates or antimicrobial therapies are urgently needed. Here we use an invasive and clinically relevant streptococcal M1 serotype to characterize the bacterial proteome in-depth. An elaborate fractionation technique is employed to separate the different cell fractions, followed by shotgun mass-spectrometry analysis, allowing us to confirm the expression of nearly two-thirds (1022) of the 1690 open reading frames predicted for the streptococcal M1 reference proteome. In contrast with other studies, we present the entire isolated membrane proteome, which opens up a whole new source for drug targets. We show both the unique and most prevalent proteins for each cellular fraction and analyze the presence of predicted cell-wall-anchored proteins and lipoproteins. With our approach, we also identify a variety of novel proteins whose presence has not been reported in previous proteome studies. Proteins of interest, potential virulence factors, and drug or vaccine targets are discussed for each cellular fraction. Overall, the results of this work represent the so-far widest proteomic approach to characterize the protein composition and localization in S. pyogenes.


Subject(s)
Proteome/analysis , Streptococcus pyogenes/chemistry , Bacterial Proteins/analysis , Mass Spectrometry/methods , Membrane Proteins/analysis , Subcellular Fractions/chemistry
10.
Biochem J ; 474(3): 411-425, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27784762

ABSTRACT

Pseudomonas aeruginosa airway infection is common in cystic fibrosis (CF), a disease also characterized by abundant extracellular DNA (eDNA) in the airways. The eDNA is mainly derived from neutrophils accumulating in the airways and contributes to a high sputum viscosity. The altered environment in the lower airways also paves the way for chronic P. aeruginosa infection. Here, we show that mice with P. aeruginosa airway infection have increased survival and decreased bacterial load after topical treatment with DNase. Furthermore, DNA from the sputum of CF patients showed increased bactericidal activity after treatment with DNase ex vivo. Both degraded DNA of neutrophil extracellular traps (NETs) and genomic DNA degraded by serum, acquired bactericidal activity against P. aeruginosa In vitro, small synthetic DNA-fragments (<100 base pairs) but not large fragments nor genomic DNA, were bactericidal against Gram-negative but not Gram-positive bacteria. The addition of divalent cations reduced bacterial killing, suggesting that chelation of divalent cations by DNA results in destabilization of the lipopolysaccharide (LPS) envelope. This is a novel antibacterial strategy where fragmentation of eDNA and DNA-fragments can be used to treat P. aeruginosa airway infection.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bronchoalveolar Lavage Fluid/chemistry , Chelating Agents/pharmacology , DNA/pharmacology , Neutrophils/chemistry , Pseudomonas Infections/drug therapy , Pseudomonas aeruginosa/drug effects , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/isolation & purification , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cations, Divalent , Chelating Agents/chemistry , Chelating Agents/isolation & purification , Cystic Fibrosis/drug therapy , Cystic Fibrosis/immunology , Cystic Fibrosis/microbiology , Cystic Fibrosis/pathology , DNA/chemistry , DNA/isolation & purification , DNA Fragmentation , Deoxyribonuclease I/chemistry , Extracellular Traps/chemistry , Extracellular Traps/immunology , Humans , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/chemistry , Male , Mice , Mice, Inbred C57BL , Microbial Sensitivity Tests , Neutrophil Activation , Neutrophils/immunology , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology , Pseudomonas Infections/mortality , Pseudomonas aeruginosa/growth & development , Sputum/chemistry , Sputum/cytology , Sputum/immunology
11.
Mediators Inflamm ; 2018: 7028267, 2018.
Article in English | MEDLINE | ID: mdl-29769838

ABSTRACT

The microbial etiology and source of sepsis influence the inflammatory response. Therefore, the plasma levels of cytokines (IL-6, IL-8, and IL-10), chemokines (CCL2/MCP-1, MIP-1ß), heparin-binding protein (HBP), soluble CD14 (sCD14), and cortisol were analyzed in blood from septic patients obtained during the first 96 hours of intensive care unit hospitalization. The etiology was established in 56 out of a total of 62 patients enrolled in the study. Plasma concentrations of MCP-1, sCD14, IL-6, and IL-10 were significantly higher in patients with community-acquired pneumonia (CAP; n = 10) and infective endocarditis (IE; n = 11) compared to those with bacterial meningitis (BM; n = 18). Next, cortisol levels were higher in IE patients than in those with BM and CAP, and at one time point, cortisol was also higher in patients with gram-negative sepsis when compared to those with gram-positive infections. Furthermore, cortisol and MCP-1 levels correlated positively with the daily measured SOFA score. In addition, HBP levels were significantly higher in patients with IE than in those with BM. Our findings suggest that MCP-1, sCD14, IL-6, IL-10, cortisol, and HBP are modulated by the source of sepsis and that elevated MCP-1 and cortisol plasma levels are associated with sepsis-induced organ dysfunction.


Subject(s)
Biomarkers/metabolism , Sepsis/metabolism , Aged , Antimicrobial Cationic Peptides/metabolism , Blood Proteins/metabolism , Carrier Proteins/metabolism , Chemokine CCL2/metabolism , Chemokine CCL4/metabolism , Critical Care , Female , Humans , Hydrocortisone/metabolism , Interleukin-10/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Lipopolysaccharide Receptors/metabolism , Male , Middle Aged
12.
Article in English | MEDLINE | ID: mdl-28652240

ABSTRACT

Macrolide antibiotics are used as anti-inflammatory agents, e.g., for prevention of exacerbations in chronic obstructive pulmonary disease and cystic fibrosis. Several studies have shown improved outcomes after the addition of macrolides to ß-lactam antibiotics for treatment of severe community-acquired pneumonia. However, a beneficial effect of macrolides in treating Gram-negative bacterial airway infections, e.g., those caused by Pseudomonas aeruginosa, remains to be shown. Macrolide antibiotics have significant side effects, in particular, motility-stimulating activity in the gastrointestinal tract and promotion of bacterial resistance. In this study, EM703, a modified macrolide lacking antibiotic and motility-stimulating activities but with retained anti-inflammatory properties, was used as an adjunct treatment for experimental P. aeruginosa lung infection, in combination with a conventional antibiotic. Airway infections in BALB/cJRj mice were induced by nasal instillation of P. aeruginosa; this was followed by treatment with the quinolone levofloxacin in the absence or presence of EM703. Survival, inflammatory responses, and cellular influx to the airways were monitored. Both pretreatment and simultaneous administration of EM703 dramatically improved survival in levofloxacin-treated mice with P. aeruginosa airway infections. In addition, EM703 reduced the levels of proinflammatory cytokines, increased the numbers of leukocytes in bronchoalveolar lavage fluid, and reduced the numbers of neutrophils present in lung tissue. In summary, the findings of this study show that the immunomodulatory properties of the modified macrolide EM703 can be important when treating Gram-negative pneumonia, as exemplified by P. aeruginosa infection in this study.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Erythromycin/analogs & derivatives , Levofloxacin/therapeutic use , Lung Diseases/drug therapy , Pseudomonas Infections/drug therapy , Pseudomonas aeruginosa/drug effects , Animals , Bronchoalveolar Lavage Fluid/cytology , Cytokines/blood , Drug Therapy, Combination , Erythromycin/therapeutic use , Female , Gastrointestinal Tract/drug effects , Leukocyte Count , Leukocytes/cytology , Lung/cytology , Lung Diseases/microbiology , Mice , Mice, Inbred BALB C , Neutrophils/cytology
13.
PLoS Pathog ; 11(12): e1005319, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26646682

ABSTRACT

The innate immune system relies to a great deal on the interaction of pattern recognition receptors with pathogen- or damage-associated molecular pattern molecules. Extracellular histones belong to the latter group and their release has been described to contribute to the induction of systemic inflammatory reactions. However, little is known about their functions in the early immune response to an invading pathogen. Here we show that extracellular histones specifically target monocytes in human blood and this evokes the mobilization of the chemotactic chemokines CXCL9 and CXCL10 from these cells. The chemokine induction involves the toll-like receptor 4/myeloid differentiation factor 2 complex on monocytes, and is under the control of interferon-γ. Consequently, subcutaneous challenge with extracellular histones results in elevated levels of CXCL10 in a murine air pouch model and an influx of leukocytes to the site of injection in a TLR4 dependent manner. When analyzing tissue biopsies from patients with necrotizing fasciitis caused by Streptococcus pyogenes, extracellular histone H4 and CXCL10 are immunostained in necrotic, but not healthy tissue. Collectively, these results show for the first time that extracellular histones have an important function as chemoattractants as their local release triggers the recruitment of immune cells to the site of infection.


Subject(s)
Chemokine CXCL10/biosynthesis , Chemokine CXCL9/biosynthesis , Chemotaxis, Leukocyte/immunology , Histones/immunology , Leukocytes/immunology , Animals , Chemokine CXCL10/immunology , Chemokine CXCL9/immunology , Chemokines/biosynthesis , Enzyme-Linked Immunosorbent Assay , Female , Humans , Leukocytes/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Immunoelectron , Monocytes/immunology , Surface Plasmon Resonance
14.
Biochem J ; 473(1): 87-98, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26508735

ABSTRACT

The human host-defence peptide (HDP) LL-37 not only displays anti-microbial activity but also immune-modulating properties that trigger intracellular signalling events in host cells. Since the cytolytic activity of high LL-37 concentrations affects cell viability, the function of LL-37 requires tight regulation. Eukaryotic cells therefore benefit from protective measures to prevent harmful effects of LL-37. p33, also known as globular C1q receptor (gC1qR), is reported to act as an LL-37 antagonist by binding the peptide, thereby reducing its cytotoxic activity. In the present report, we show that high levels of endogenous p33 correlate with an increased viability in human cells treated with LL-37. Sub-cellular localization analysis showed p33 distribution at the mitochondria, the plasma membrane and in the cytosol. Strikingly, cytosolic overexpression of p33 significantly antagonized detrimental effects of LL-37 on cell fitness, whereas the reverse effect was observed by siRNA-induced down-regulation of p33. However, modulation of p33 expression had no effect on LL-37-induced plasma membrane pore forming capacity pointing to an intracellular mechanism. A scavenging function of intracellular p33 is further supported by co-immunoprecipitation experiments, showing a direct interaction between intracellular p33 and LL-37. Thus, our findings support an important role of intracellular p33 in maintaining cell viability by counteracting LL-37-induced cytotoxicity.


Subject(s)
Antimicrobial Cationic Peptides/toxicity , Cell Membrane/drug effects , Cell Membrane/metabolism , Cytotoxins/toxicity , Membrane Glycoproteins/biosynthesis , Receptors, Complement/biosynthesis , Adolescent , Cells, Cultured , Dose-Response Relationship, Drug , Female , Gene Expression Regulation , HeLa Cells , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , Male , Membrane Glycoproteins/genetics , Receptors, Complement/genetics , Cathelicidins
15.
Infect Immun ; 83(12): 4673-81, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26416902

ABSTRACT

The human skin exerts many functions in order to maintain its barrier integrity and protect the host from invading microorganisms. One such pathogen is Streptococcus pyogenes, which can cause a variety of superficial skin wounds that may eventually progress into invasive deep soft tissue infections. Here we show that keratinocytes recognize soluble M1 protein, a streptococcal virulence factor, as a pathogen-associated molecular pattern to release alarming inflammatory responses. We found that this interaction initiates an inflammatory intracellular signaling cascade involving the activation of the mitogen-activated protein kinases extracellular signal-regulated kinase (ERK), p38, and Jun N-terminal protein kinase and the subsequent induction and mobilization of the transcription factors NF-κB and AP-1. We also determined the imprint of the inflammatory mediators released, such as interleukin-8 (IL-8), growth-related oncogene alpha, migration inhibitory factor, extracellular matrix metalloproteinase inducer, IL-1α, IL-1 receptor a, and ST2, in response to streptococcal M1 protein. The expression of IL-8 is dependent on Toll-like receptor 2 activity and subsequent activation of the mitogen-activated protein kinases ERK and p38. Notably, this signaling seems to be distinct for IL-8 release, and it is not shared with the other inflammatory mediators. We conclude that keratinocytes participate in a proinflammatory manner in streptococcal pattern recognition and that expression of the chemoattractant IL-8 by keratinocytes constitutes an important protective mechanism against streptococcal M1 protein.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/immunology , Carrier Proteins/immunology , Interleukin-8/immunology , Keratinocytes/immunology , Signal Transduction/immunology , Streptococcus pyogenes/immunology , Antigens, Bacterial/genetics , Bacterial Outer Membrane Proteins/genetics , Basigin/genetics , Basigin/immunology , Carrier Proteins/genetics , Cell Line, Transformed , Chemokine CXCL1/genetics , Chemokine CXCL1/immunology , Gene Expression Regulation , Host-Pathogen Interactions , Humans , Interleukin 1 Receptor Antagonist Protein/genetics , Interleukin 1 Receptor Antagonist Protein/immunology , Interleukin-1 Receptor-Like 1 Protein , Interleukin-1alpha/genetics , Interleukin-1alpha/immunology , Interleukin-8/genetics , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/immunology , Keratinocytes/microbiology , Macrophage Migration-Inhibitory Factors/genetics , Macrophage Migration-Inhibitory Factors/immunology , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/immunology , NF-kappa B/genetics , NF-kappa B/immunology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/immunology , Transcription Factor AP-1/genetics , Transcription Factor AP-1/immunology , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/immunology
16.
Am J Physiol Lung Cell Mol Physiol ; 308(11): L1159-67, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25840996

ABSTRACT

Streptococcus pyogenes cause infections ranging from mild pharyngitis to severe streptococcal toxic shock syndrome (STSS). The M1 serotype of Streptococcus pyogenes is most frequently associated with STSS. Herein, it was hypothesized that STAT3 signaling might be involved in M1 protein-evoked lung inflammation. The STAT3 inhibitor, S3I-201, was administered to male C57Bl/6 mice before iv challenge with M1 protein. Bronchoalveolar fluid and lung tissue were harvested for quantification of STAT3 activity, neutrophil recruitment, edema, and CXC chemokine formation. Neutrophil expression of Mac-1 was quantified by use of flow cytometry. Levels of IL-6 and HMGB1 were determined in plasma. CXCL2-induced neutrophil chemotaxis was studied in vitro. Administration of S3I-201 markedly reduced M1 protein-provoked STAT3 activity, neutrophil recruitment, edema formation, and inflammatory changes in the lung. In addition, M1 protein significantly increased Mac-1 expression on neutrophils and CXC chemokine levels in the lung. Treatment with S3I-201 had no effect on M1 protein-induced expression of Mac-1 on neutrophils. In contrast, inhibition of STAT3 activity greatly reduced M1 protein-induced formation of CXC chemokines in the lung. Interestingly, STAT3 inhibition markedly decreased plasma levels of IL-6 and HMGB1 in animals exposed to M1 protein. Moreover, we found that S3I-201 abolished CXCL2-induced neutrophil migration in vitro. In conclusion, these novel findings indicate that STAT3 signaling plays a key role in mediating CXC chemokine production and neutrophil infiltration in M1 protein-induced acute lung inflammation.


Subject(s)
Acute Lung Injury/immunology , Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/immunology , Carrier Proteins/immunology , Neutrophils/physiology , STAT3 Transcription Factor/physiology , Animals , Cell Movement , HMGB1 Protein/blood , Interleukin-6/blood , Lung/immunology , Lung/metabolism , Lung/pathology , Macrophage-1 Antigen/metabolism , Male , Mice, Inbred C57BL , Neutrophil Infiltration
17.
Microbiology (Reading) ; 161(Pt 3): 621-627, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25533444

ABSTRACT

Recent work has shown that coagulation and innate immunity are tightly interwoven host responses that help eradicate an invading pathogen. Some bacterial species, including Staphylococcus aureus, secrete pro-coagulant factors that, in turn, can modulate these immune reactions. Such mechanisms may not only protect the micro-organism from a lethal attack, but also promote bacterial proliferation and the establishment of infection. Our data showed that coagulase-positive S. aureus bacteria promoted clotting of plasma which was not seen when a coagulase-deficient mutant strain was used. Furthermore, in vitro studies showed that this ability constituted a mechanism that supported the aggregation, survival and persistence of the micro-organism within the fibrin network. These findings were also confirmed when agglutination and persistence of coagulase-positive S. aureus bacteria at the local focus of infection were studied in a subcutaneous murine infection model. In contrast, the coagulase-deficient S. aureus strain which was not able to induce clotting failed to aggregate and to persist in vivo. In conclusion, our data suggested that coagulase-positive S. aureus have evolved mechanisms that prevent their elimination within a fibrin clot.


Subject(s)
Blood Coagulation , Fibrin/metabolism , Immune Evasion , Staphylococcal Infections/blood , Staphylococcus aureus/immunology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Coagulase/genetics , Coagulase/metabolism , Fibrin/genetics , Humans , Mice , Mice, Inbred CBA , Microbial Viability , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcus aureus/enzymology , Staphylococcus aureus/genetics , Staphylococcus aureus/growth & development
18.
PLoS Pathog ; 9(8): e1003529, 2013.
Article in English | MEDLINE | ID: mdl-23935504

ABSTRACT

Previous studies have shown that stimulation of whole blood or peripheral blood mononuclear cells with bacterial virulence factors results in the sequestration of pro-coagulant microvesicles (MVs). These particles explore their clotting activity via the extrinsic and intrinsic pathway of coagulation; however, their pathophysiological role in infectious diseases remains enigmatic. Here we describe that the interaction of pro-coagulant MVs with bacteria of the species Streptococcus pyogenes is part of the early immune response to the invading pathogen. As shown by negative staining electron microscopy and clotting assays, pro-coagulant MVs bind in the presence of plasma to the bacterial surface. Fibrinogen was identified as a linker that, through binding to the M1 protein of S. pyogenes, allows the opsonization of the bacteria by MVs. Surface plasmon resonance analysis revealed a strong interaction between pro-coagulant MVs and fibrinogen with a KD value in the nanomolar range. When performing a mass-spectrometry-based strategy to determine the protein quantity, a significant up-regulation of the fibrinogen-binding integrins CD18 and CD11b on pro-coagulant MVs was recorded. Finally we show that plasma clots induced by pro-coagulant MVs are able to prevent bacterial dissemination and possess antimicrobial activity. These findings were confirmed by in vivo experiments, as local treatment with pro-coagulant MVs dampens bacterial spreading to other organs and improved survival in an invasive streptococcal mouse model of infection. Taken together, our data implicate that pro-coagulant MVs play an important role in the early response of the innate immune system in infectious diseases.


Subject(s)
Blood Coagulation/immunology , CD11b Antigen/immunology , CD18 Antigens/immunology , Cell-Derived Microparticles/immunology , Streptococcal Infections/metabolism , Streptococcus pyogenes/immunology , Animals , CD11b Antigen/metabolism , CD18 Antigens/metabolism , Cell-Derived Microparticles/metabolism , Cell-Derived Microparticles/microbiology , Cell-Derived Microparticles/ultrastructure , Humans , Mice , Mice, Inbred BALB C , Microscopy, Electron, Transmission , Streptococcal Infections/immunology , Streptococcal Infections/pathology , Streptococcus pyogenes/metabolism , Streptococcus pyogenes/ultrastructure
19.
J Immunol ; 191(11): 5714-21, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24174616

ABSTRACT

The innate immune system is the first line of defense against invading microbes. Its specificity relies a great deal on host pattern recognition molecules that sense pathogen-associated molecular patterns of the invading pathogen. However, full protection is not always guaranteed, and some early defense mechanisms involved in bacterial killing, such as the complement system, can also exert cytolytic activity against host cells. Although these cascades are tightly regulated, the host has to take additional precautions to prevent its cell destruction. In this study, we describe that p33, a negatively charged surface protein found on endothelial cells also known as gC1q receptor, protects host cells from a cytolytic attack by antimicrobial peptides (AMPs), such as LL37 and ß-defensin 3. To this end, we characterized the interaction of p33 with AMPs by biochemical and functional means. Our data show that p33 forms a doughnut-shaped trimer that can bind up to three AMPs, and we identified a segment in p33 forming a ß-sheet that mediates the binding to all AMPs. Moreover, our results show that p33 abolishes the lytic activity of AMPs at an equimolar ratio, and it protects endothelial cells and erythrocytes from AMP-induced lysis. Taken together, our data suggest a novel protective mechanism of p33 in modulating innate immune response by neutralizing cytotoxic AMPs at the host cell surface.


Subject(s)
Carrier Proteins/metabolism , Endothelial Cells/immunology , Erythrocytes/immunology , Mitochondrial Proteins/metabolism , Amino Acid Sequence , Antimicrobial Cationic Peptides , Binding Sites , Carrier Proteins/immunology , Cathelicidins/pharmacology , Cells, Cultured , Cytoprotection/drug effects , Cytotoxicity, Immunologic/drug effects , Endothelial Cells/drug effects , Erythrocytes/drug effects , Host-Pathogen Interactions , Humans , Mitochondrial Proteins/immunology , Molecular Sequence Data , Protein Binding , Protein Multimerization , beta-Defensins/pharmacology
20.
Curr Top Microbiol Immunol ; 368: 189-205, 2013.
Article in English | MEDLINE | ID: mdl-23224709

ABSTRACT

Haemostasis is maintained by a tightly regulated coagulation system that comprises platelets, procoagulant proteins, and anticoagulant proteins. During the local and systemic response to bacterial infection, the coagulation system becomes activated, and contributes to the pathophysiological response to infection. The significant human pathogen, Streptococcus pyogenes has multiple strategies to modulate coagulation. This can range from systemic activation of the intrinsic and extrinsic pathway of coagulation to local stimulation of fibrinolysis. Such diverse effects on this host system imply a finely tuned host-bacteria interaction. The molecular mechanisms that underlie this modulation of the coagulation system are discussed in this review.


Subject(s)
Blood Coagulation , Streptococcal Infections/blood , Streptococcus pyogenes/pathogenicity , Animals , Humans , Kinins/metabolism , Plasminogen/metabolism , Platelet Activation , Streptococcal Infections/immunology
SELECTION OF CITATIONS
SEARCH DETAIL