Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Innate Immun ; : 1-15, 2022 May 10.
Article in English | MEDLINE | ID: mdl-35537415

ABSTRACT

Humans with dysfunctional Bruton's tyrosine kinase (Btk) are highly susceptible to bacterial infections. Compelling evidence indicates that Btk is essential for B cell-mediated immunity, whereas its role in myeloid cell-mediated immunity against infections is controversial. In this study, we determined the contribution of Btk in B cells and neutrophils to host defense against the extracellular bacterial pathogen Klebsiella pneumoniae, a common cause of pulmonary infections and sepsis. Btk-/- mice were highly susceptible to Klebsiella infection, which was not reversed by Btk re-expression in B cells and restoration of natural antibody levels. Neutrophil-specific Btk deficiency impaired host defense against Klebsiella to a similar extent as complete Btk deficiency. Neutrophil-specific Btk deficiency abolished extracellular reactive oxygen species production in response to Klebsiella. These data indicate that expression of Btk in neutrophils is crucial, while in B cells, it is dispensable for in vivo host defense against K. pneumoniae.

2.
Front Immunol ; 12: 723967, 2021.
Article in English | MEDLINE | ID: mdl-34552589

ABSTRACT

Bruton's tyrosine kinase (Btk) is a cytoplasmic kinase expressed in B cells and myeloid cells. It is essential for B cell development and natural antibody-mediated host defense against bacteria in humans and mice, but little is known about the role of Btk in innate host defense in vivo. Previous studies have indicated that lack of (natural) antibodies is paramount for impaired host defense against Streptococcus (S.) pneumoniae in patients and mice with a deficiency in functional Btk. In the present study, we re-examined the role of Btk in B cells and myeloid cells during pneumococcal pneumonia and sepsis in mice. The antibacterial defense of Btk-/- mice was severely impaired during pneumococcal pneumosepsis and restoration of natural antibody production in Btk-/- mice by transgenic expression of Btk specifically in B cells did not suffice to protect against infection. Btk-/- mice with reinforced Btk expression in MhcII+ cells, including B cells, dendritic cells and macrophages, showed improved antibacterial defense as compared to Btk-/- mice. Bacterial outgrowth in Lysmcre-Btkfl/Y mice was unaltered despite a reduced capacity of Btk-deficient alveolar macrophages to respond to pneumococci. Mrp8cre-Btkfl/Y mice with a neutrophil specific paucity in Btk expression, however, demonstrated impaired antibacterial defense. Neutrophils of Mrp8cre-Btkfl/Y mice displayed reduced release of granule content after pulmonary installation of lipoteichoic acid, a gram-positive bacterial cell wall component relevant for pneumococci. Moreover, Btk deficient neutrophils showed impaired degranulation and phagocytosis upon incubation with pneumococci ex vivo. Taken together, the results of our study indicate that besides regulating B cell-mediated immunity, Btk is critical for regulation of myeloid cell-mediated, and particularly neutrophil-mediated, innate host defense against S. pneumoniae in vivo.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/metabolism , Immunity, Innate , Myeloid Cells/immunology , Pneumonia, Pneumococcal/immunology , Pneumonia, Pneumococcal/metabolism , Sepsis/metabolism , Agammaglobulinaemia Tyrosine Kinase/genetics , Animals , B-Lymphocytes/metabolism , Disease Models, Animal , Female , Humans , Lipopolysaccharides , Lung/pathology , Macrophages, Alveolar/metabolism , Male , Mice , Mice, Inbred C57BL , Phagocytosis , Pneumonia, Pneumococcal/genetics , Signal Transduction , Streptococcus pneumoniae/physiology , Teichoic Acids
3.
Am J Physiol Lung Cell Mol Physiol ; 319(1): L105-L114, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32401674

ABSTRACT

Klebsiella (K.) pneumoniae is a common cause of gram-negative pneumonia and sepsis. Caspase-11 is an intracellular receptor for lipopolysaccharide and regulates pyroptosis, a specific form of inflammatory cell death, which aids in host defense against intracellular gram-negative bacteria. Recently, caspase-11 has also been implicated in blood coagulation. Previously, we found that local fibrin formation contributes to protective immunity against Klebsiella infection of the lung. The aim of the present study was to determine the role of caspase-11 in host defense during K. pneumoniae-evoked pneumonia and sepsis. Therefore, we infected wild-type and caspase-11-deficient (Casp11-/-) mice with a low-dose K. pneumoniae via the airways to induce a gradually evolving pneumosepsis. Casp11-/- mice displayed increased bacterial numbers in the lung 12 h and 48 h after inoculation. Analysis of pulmonary IL-1α, IL-1ß, and TNF levels showed reduced IL-1α levels in bronchoalveolar lavage fluid and increased TNF levels in the lung of Casp11-/- mice at 48 h after inoculation. Lung γH2AX staining (marker for cell death), lung pathology and neutrophil influx in the lung, as well as bacterial dissemination and organ damage, however, were not altered in Casp11-/- mice after Klebsiella infection. Strikingly, analysis of cross-linked fibrin and D-dimer (markers for coagulation) revealed significantly less fibrin formation in the lungs of Casp11-/- mice at either time point after Klebsiella infection. These data reveal that caspase-11 contributes to protective immunity against K. pneumoniae possibly by activation of blood coagulation in the lung.


Subject(s)
Blood Coagulation/physiology , Caspases, Initiator/metabolism , Host-Pathogen Interactions , Klebsiella Infections/immunology , Klebsiella Infections/microbiology , Klebsiella pneumoniae/physiology , Lung/immunology , Lung/microbiology , Animals , Caspases, Initiator/deficiency , Cell Death , Cytokines/metabolism , Fibrin/metabolism , Inflammation Mediators/metabolism , Klebsiella Infections/blood , Lung/pathology , Mice, Inbred C57BL , Neutrophils/metabolism , Pneumonia/blood , Pneumonia/complications
4.
Respir Res ; 20(1): 165, 2019 Jul 24.
Article in English | MEDLINE | ID: mdl-31340811

ABSTRACT

BACKGROUND: Complement factor C5 can either aggravate or attenuate the T-helper type 2 (TH2) immune response and airway hyperresponsiveness (AHR) in murine models of allergic asthma. The effect of C5 during the effector phase of allergen-induced asthma is ill-defined. OBJECTIVES: We aimed to determine the effect of C5 blockade during the effector phase on the pulmonary TH2 response and AHR in a house dust mite (HDM) driven murine asthma model. METHODS: BALB/c mice were sensitized and challenged repeatedly with HDM via the airways to induce allergic lung inflammation. Sensitized mice received twice weekly injections with a blocking anti-C5 or control antibody 24 h before the first challenge. RESULTS: HDM challenge in sensitized mice resulted in elevated C5a levels in bronchoalveolar lavage fluid. Anti-C5 administered to sensitized mice prior to the first HDM challenge prevented this rise in C5a, but did not influence the influx of eosinophils or neutrophils. While anti-C5 did not impact the recruitment of CD4 T cells upon HDM challenge, it reduced the proportion of TH2 cells recruited to the airways, attenuated IL-4 release by regional lymph nodes restimulated with HDM ex vivo and mitigated the plasma IgE response. Anti-C5 did not affect innate lymphoid cell (ILC) proliferation or group 2 ILC (ILC2) differentiation. Anti-C5 attenuated HDM induced AHR in the absence of an effect on lung histopathology, mucus production or vascular leak. CONCLUSIONS: Generation of C5a during the effector phase of HDM induced allergic lung inflammation contributes to TH2 cell differentiation and AHR without impacting ILC2 cells.


Subject(s)
Asthma/immunology , Complement C5a/antagonists & inhibitors , Complement C5a/immunology , Immunity, Innate/immunology , Pyroglyphidae/immunology , Th2 Cells/immunology , Animals , Asthma/chemically induced , Asthma/prevention & control , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Immunity, Innate/drug effects , Lymphocytes/drug effects , Lymphocytes/immunology , Methacholine Chloride/pharmacology , Mice , Mice, Inbred BALB C , Th2 Cells/drug effects
5.
Thromb Haemost ; 119(6): 930-940, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30873567

ABSTRACT

Platelet Bruton's tyrosine kinase (Btk) is an essential signalling protein for the collagen receptor glycoprotein VI (GPVI) and podoplanin receptor C-type-lectin-like receptor-2, which are platelet receptors implicated in the maintenance of vascular integrity during inflammation. Moreover, platelets, platelet GPVI and Btk are important for host defence during murine bacterial pneumosepsis. The aim of this study was to determine the role of platelet Btk in vascular integrity and host defence during murine pneumosepsis caused by the common human pathogens Streptococcus pneumoniae and Klebsiella pneumoniae. Using the Cre-loxP system, male platelet-specific Btk-deficient mice (PF4creBtkfl/Y) were created. Similar to platelets from total Btk-deficient mice, platelets from PF4creBtkfl/Y mice showed abrogated aggregation and P-selectin expression when stimulated with the GPVI ligand cross-linked collagen-related peptide. Upon infection with S. pneumoniae, PF4creBtkfl/Y mice showed increased lung bleeding, but unimpaired anti-bacterial defence. During pneumosepsis evoked by K. pneumoniae, platelet Btk deficiency was not associated with lung bleeding and did not impact on host defence, even when platelet function was further compromised by blocking secondary platelet activation by the P2Y12 receptor antagonist clopidogrel. Together, these data indicate that, while platelet Btk is not important for anti-bacterial defence in pneumosepsis, its role in maintaining vascular integrity in the lung depends on the causative pathogen.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/metabolism , Blood Platelets/physiology , Klebsiella pneumoniae/physiology , Lung/pathology , Pneumonia, Pneumococcal/metabolism , Sepsis/metabolism , Streptococcus pneumoniae/physiology , Agammaglobulinaemia Tyrosine Kinase/genetics , Animals , Disease Models, Animal , Hemorrhage , Humans , Immunity , Lung/blood supply , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Platelet Aggregation/genetics , Platelet Membrane Glycoproteins/metabolism , Pneumonia, Pneumococcal/genetics , Regional Blood Flow , Signal Transduction
6.
Mol Med ; 25(1): 3, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30646846

ABSTRACT

BACKGROUND: Streptococcus pneumoniae is a major causative agent in community-acquired pneumonia and sepsis. Overwhelming lung inflammation during pneumococcal pneumonia may hamper lung function. Ibrutinib is an irreversible inhibitor of Bruton's tyrosine kinase (Btk), a key signaling protein controlling the activation of various immune cells, including macrophages and neutrophils. The aim of this study was to determine whether ibrutinib treatment ameliorates acute lung inflammation during pneumococcal pneumonia. METHODS: Mice were treated orally with ibrutinib and the effect on acute pulmonary inflammation elicited by the gram-positive bacterial cell wall component lipoteichoic acid (LTA) and during ceftriaxone-treated pneumococcal pneumonia was assessed. RESULTS: Treatment with ibrutinib prior to and after intranasal LTA instillation reduced alveolar macrophage activation, neutrophil influx, cytokine release and plasma leakage into the lung. Postponed treatment with ibrutinib supplementing antibiotic therapy during ongoing pneumococcal pneumonia did not impair bacterial killing in lung, blood and spleen. In this setting, ibrutinib reduced alveolar macrophage and systemic neutrophil activation and substantially diminished further monocyte and neutrophil influx in the lung. In vitro, ibrutinib inhibited macrophage TNF secretion and neutrophil activation upon LTA and pneumococcal stimulation. CONCLUSIONS: Taken together, these data indicate that the Btk inhibitor ibrutinib reduces inflammatory myeloid cell responses during acute pulmonary inflammation evoked by LTA and antibiotic-treated pneumococcal pneumonia and suggest that ibrutinib has the potential to inhibit ongoing lung inflammation in an acute infectious setting.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Anti-Inflammatory Agents/therapeutic use , Pneumonia, Pneumococcal/drug therapy , Protein Kinase Inhibitors/therapeutic use , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Adenine/analogs & derivatives , Animals , Anti-Bacterial Agents/therapeutic use , Bronchoalveolar Lavage Fluid/cytology , Ceftriaxone/therapeutic use , Lipopolysaccharides , Lung/drug effects , Lung/immunology , Male , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/drug effects , Myeloid Cells/immunology , Piperidines , Pneumonia, Pneumococcal/immunology , Teichoic Acids
7.
Eur J Immunol ; 48(1): 66-79, 2018 01.
Article in English | MEDLINE | ID: mdl-28971472

ABSTRACT

Streptococcus (S.) pneumoniae is the most common cause of community-acquired pneumonia. The Nod-like receptor family pyrin domain containing 3 (NLRP3) inflammasome, consisting of NLRP3, ASC (the adaptor apoptosis-associated speck-like protein containing a CARD) and caspase-1, has been implicated in protective immunity during pneumonia induced by high doses of S. pneumoniae serotype 2. Here we investigated the role of the NLRP3 inflammasome in the host response during lethal airway infection with a low dose of serotype 3 S. pneumoniae. Mice were euthanized at predefined endpoints for analysis or observed in survival studies. In additional studies, Tlr2-/- /Tlr4-/- mice and Myd88-/- mice incapable of Toll-like receptor signaling were studied. In stark contrast with existing literature, both Nlrp3-/- and Asc-/- mice showed a strongly improved host defense, as reflected by a markedly reduced mortality rate accompanied by diminished bacterial growth and dissemination. Host defense was unaltered in Tlr2-/- /Tlr4-/- mice and Myd88-/- mice. These results show that the NLRP3 inflammasome impairs host defense during lethal pneumonia caused by serotype 3 S. pneumoniae. Our findings challenge the current paradigm that proximal innate detection systems are indispensable for an adequate host immune response against bacteria.


Subject(s)
CARD Signaling Adaptor Proteins/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Pneumonia, Pneumococcal/immunology , Streptococcus pneumoniae/immunology , Animals , CARD Signaling Adaptor Proteins/genetics , Caspase 1/immunology , Community-Acquired Infections/immunology , Community-Acquired Infections/microbiology , Immunity, Innate/genetics , Immunity, Innate/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Pneumonia, Pneumococcal/pathology , Signal Transduction/immunology , Streptococcus pneumoniae/classification , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics
8.
Gut ; 65(4): 575-83, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26511795

ABSTRACT

OBJECTIVE: Pneumonia accounts for more deaths than any other infectious disease worldwide. The intestinal microbiota supports local mucosal immunity and is increasingly recognised as an important modulator of the systemic immune system. The precise role of the gut microbiota in bacterial pneumonia, however, is unknown. Here, we investigate the function of the gut microbiota in the host defence against Streptococcus pneumoniae infections. DESIGN: We depleted the gut microbiota in C57BL/6 mice and subsequently infected them intranasally with S. pneumoniae. We then performed survival and faecal microbiota transplantation (FMT) experiments and measured parameters of inflammation and alveolar macrophage whole-genome responses. RESULTS: We found that the gut microbiota protects the host during pneumococcal pneumonia, as reflected by increased bacterial dissemination, inflammation, organ damage and mortality in microbiota-depleted mice compared with controls. FMT in gut microbiota-depleted mice led to a normalisation of pulmonary bacterial counts and tumour necrosis factor-α and interleukin-10 levels 6 h after pneumococcal infection. Whole-genome mapping of alveolar macrophages showed upregulation of metabolic pathways in the absence of a healthy gut microbiota. This upregulation correlated with an altered cellular responsiveness, reflected by a reduced responsiveness to lipopolysaccharide and lipoteichoic acid. Compared with controls, alveolar macrophages derived from gut microbiota-depleted mice showed a diminished capacity to phagocytose S. pneumoniae. CONCLUSIONS: This study identifies the intestinal microbiota as a protective mediator during pneumococcal pneumonia. The gut microbiota enhances primary alveolar macrophage function. Novel therapeutic strategies could exploit the gut-lung axis in bacterial infections.


Subject(s)
Gastrointestinal Microbiome/immunology , Immunity, Mucosal , Pneumonia, Pneumococcal/immunology , Animals , Anti-Bacterial Agents/pharmacology , Bacterial Load , Feces/microbiology , Interleukin-10/metabolism , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Mice , Mice, Inbred C57BL , Pneumonia, Pneumococcal/metabolism , Pneumonia, Pneumococcal/microbiology , Sepsis/immunology , Sepsis/metabolism , Sepsis/microbiology , Streptococcus pneumoniae/immunology , Transcriptome , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
9.
PLoS One ; 10(2): e0118181, 2015.
Article in English | MEDLINE | ID: mdl-25700108

ABSTRACT

Toll-like receptors (TLR) and the downstream adaptor protein MyD88 are considered crucial for protective immunity during bacterial infections. Streptococcus (S.) pneumoniae is a human respiratory pathogen and a large majority of clinical pneumococcal isolates expresses an external polysaccharide capsule. We here sought to determine the role of pneumococcal capsule in MyD88-mediated antibacterial defense during S. pneumonia pneumonia. Wild type (WT) and Myd88(-/-) mice were inoculated intranasally with serotype 2 S. pneumoniae D39 or with an isogenic capsule locus deletion mutant (D39∆cps), and analysed for bacterial outgrowth and inflammatory responses in the lung. As compared to WT mice, Myd88(-/-) mice infected with D39 demonstrated a modestly impaired bacterial clearance accompanied by decreased inflammatory responses in the lung. Strikingly, while WT mice rapidly cleared D39∆cps, Myd88(-/-) mice showed 105-fold higher bacterial burdens in their lungs and dissemination to blood 24 hours after infection. These data suggest that the pneumococcal capsule impairs recognition of TLR ligands expressed by S. pneumoniae and thereby partially impedes MyD88-mediated antibacterial defense.


Subject(s)
Bacterial Capsules/immunology , Myeloid Differentiation Factor 88/genetics , Pneumonia, Bacterial/immunology , Polysaccharides, Bacterial/immunology , Streptococcus pneumoniae/immunology , Animals , Bacterial Capsules/genetics , Gene Deletion , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/immunology
10.
J Infect Dis ; 210(9): 1376-84, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-24823624

ABSTRACT

BACKGROUND: Streptococcus pneumoniae is the most common causative pathogen in community-acquired pneumonia. Mast cells (MCs) are located mainly at the host-environment interface where they function as sentinels. OBJECTIVE: Our goal was to study the role of MCs during pneumonia caused by S. pneumoniae. METHODS: Lung tissue of patients who had died from pneumococcal pneumonia or a nonpulmonary cause was stained for MCs and tryptase. Wild-type (WT) and MC-deficient (Kit(W-sh/W-sh)) mice were observed or sacrificed after induction of pneumonia by intranasal inoculation of S. pneumoniae. In separate experiments, WT mice were treated with doxantrazole or cromoglycate, which are MC stabilizing agents. RESULTS: The constitutive presence of tryptase-positive MCs was reduced in affected lungs from pneumonia patients. Kit(W-sh/W-sh) mice showed a prolonged survival during the first few days after median lethal dose (LD)100 and LD50 infection, while overall mortality did not differ from that in WT mice. Relative to WT mice, Kit(W-sh/W-sh) mice showed reduced bacterial counts with less bacterial dissemination to distant organs and less inflammation. Neither doxantrazole nor cromoglycate influenced antibacterial defense or inflammatory responses after airway infection with S. pneumoniae. CONCLUSIONS: MCs exhibit an unfavorable role in host defense during pneumococcal pneumonia by a mechanism independent of degranulation.


Subject(s)
Mast Cells/physiology , Pneumonia, Pneumococcal/immunology , Streptococcus pneumoniae/immunology , Animals , Bacterial Load , Female , Host-Pathogen Interactions/immunology , Humans , Lung/microbiology , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Pneumonia, Pneumococcal/microbiology , Pneumonia, Pneumococcal/pathology
11.
J Innate Immun ; 6(2): 219-26, 2014.
Article in English | MEDLINE | ID: mdl-24157568

ABSTRACT

BACKGROUND: Mast cells are implicated in allergic and innate immune responses in asthma, although their role in models using an allergen relevant for human disease is incompletely understood. House dust mite (HDM) allergy is common in asthma patients. Our aim was to investigate the role of mast cells in HDM-induced allergic lung inflammation. METHODS: Wild-type (Wt) and mast cell-deficient Kit(w-sh) mice on a C57BL/6 background were repetitively exposed to HDM via the airways. RESULTS: HDM challenge resulted in a rise in tryptase activity in bronchoalveolar lavage fluid (BALF) of Wt mice, indicative of mast cell activation. Kit(w-sh) mice showed a strongly attenuated HDM- induced recruitment of eosinophils in BALF and lung tissue, accompanied by reduced pulmonary levels of the eosinophil chemoattractant eotaxin. Remarkably, Kit(w-sh) mice demonstrated an unaltered capacity to develop lung pathology and increased mucus production in response to HDM. The increased plasma IgE in response to HDM in Wt mice was absent in Kit(w-sh) mice. CONCLUSION: These data contrast with previous reports on the role of mast cells in models using ovalbumin as allergen in that C57BL/6 Kit(w-sh) mice display a selective impairment of eosinophil recruitment without differences in other features of allergic inflammation.


Subject(s)
Eosinophils/immunology , Mast Cells/immunology , Pneumonia/immunology , Proto-Oncogene Proteins c-kit/immunology , Pyroglyphidae/immunology , Allergens/immunology , Animals , Asthma/genetics , Asthma/immunology , Asthma/metabolism , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Chemokine CCL11/immunology , Chemokine CCL11/metabolism , Disease Models, Animal , Eosinophils/metabolism , Humans , Immunoglobulin E/blood , Immunoglobulin E/immunology , Immunohistochemistry , Lung/immunology , Lung/metabolism , Lung/pathology , Mast Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucus/immunology , Mucus/metabolism , Ovalbumin/immunology , Pneumonia/genetics , Pneumonia/metabolism , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Tryptases/immunology , Tryptases/metabolism
12.
J Innate Immun ; 5(6): 603-12, 2013.
Article in English | MEDLINE | ID: mdl-23774862

ABSTRACT

Streptococcus pneumoniae is one of the most common causes of sepsis. Sepsis is associated with the release of 'damage-associated molecular patterns' (DAMPs). The receptor for advanced glycation end products (RAGE) is a multiligand receptor, abundantly expressed in the lungs, that recognizes several of these DAMPs. Triggering of RAGE leads to activation of the NF-κB pathway and perpetuation of inflammation. Earlier investigations have shown that the absence of RAGE reduces inflammation and bacterial dissemination and increases survival in sepsis caused by S. pneumoniae pneumonia. We hypothesized that the detrimental role of RAGE depends on the level of RAGE expression in the primary organ of infection. By directly injecting S. pneumoniae intravenously, thereby circumventing the extensive RAGE-expressing lung, we here determined whether RAGE contributes to an adverse outcome of bacteremia or whether its role is restricted to primary lung infection. During late-stage infection (48 h), rage(-/-) mice had an attenuated systemic inflammatory response, as reflected by lower plasma levels of proinflammatory cytokines, reduced endothelial cell activation (as measured by E-selectin levels) and less neutrophil accumulation in lung tissue. However, RAGE deficiency did not influence bacterial loads or survival in this model. In accordance, plasma markers for cell injury were similar in both mouse strains. These results demonstrate that while RAGE plays a harmful part in S. pneumoniae sepsis originating from the respiratory tract, this receptor has a limited role in the outcome of primary bloodstream infection by this pathogen.


Subject(s)
Bacteremia/immunology , Pneumococcal Infections/immunology , Receptors, Immunologic/immunology , Streptococcus pneumoniae/immunology , Animals , Bacteremia/genetics , Bacteremia/microbiology , Cytokines/blood , Cytokines/immunology , E-Selectin/immunology , E-Selectin/metabolism , Endothelial Cells/immunology , Endothelial Cells/metabolism , Host-Pathogen Interactions/immunology , Immunohistochemistry , Lung/immunology , Lung/metabolism , Lung/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration/genetics , Neutrophil Infiltration/immunology , Pneumococcal Infections/genetics , Pneumococcal Infections/microbiology , Receptor for Advanced Glycation End Products , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Sepsis/genetics , Sepsis/immunology , Sepsis/microbiology , Streptococcus pneumoniae/physiology , Survival Analysis , Time Factors
13.
Am J Respir Cell Mol Biol ; 48(3): 382-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23239494

ABSTRACT

The complex biology of asthma compels the use of more relevant human allergens, such as house dust mite (HDM), to improve the translation of animal models into human asthma. LPS exposure is associated with aggravations of asthma, but the mechanisms remain unclear. Here, we studied the effects of increasing LPS doses on HDM-evoked allergic lung inflammation. To this end, mice were intranasally sensitized and challenged with HDM with or without increasing doses of LPS (0.001-10 µg). LPS dose-dependently inhibited HDM-induced eosinophil recruitment into the lungs and mucus production in the airways. LPS attenuated the production of Th2 cytokines (IL-4, IL-5, IL-10, and IL-13) in HDM-challenged lungs, while enhancing the HDM-induced release of IL-17, IL-33, IFN-γ, and TNF-α. The shift toward a Th1 inflammatory response was further illustrated by predominant neutrophilic lung inflammation after LPS administration at higher doses. LPS did not influence HDM-induced plasma IgE concentrations. Although LPS did not significantly affect the activation of coagulation or complement in HDM-challenged lungs, it reduced HDM-initiated endothelial cell activation. This study is the first to provide insights into the effects of LPS in an allergic lung inflammation model making use of a clinically relevant allergen without a systemic adjuvant, revealing that LPS dose-dependently inhibits HDM-induced pulmonary Th2 responses.


Subject(s)
Antigens, Dermatophagoides/immunology , Lipopolysaccharides/pharmacology , Lung/immunology , Pneumonia/immunology , Pyroglyphidae/immunology , Th2 Cells/drug effects , Th2 Cells/immunology , Animals , Asthma/immunology , Complement Activation/immunology , Cytokines/immunology , Disease Models, Animal , Endothelial Cells/immunology , Eosinophils/immunology , Immunoglobulin E/immunology , Lipopolysaccharides/immunology , Mice , Mice, Inbred C57BL , Mucus/immunology , Respiratory Mucosa/immunology , Th1 Cells/immunology
14.
Mol Immunol ; 48(12-13): 1468-76, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21529952

ABSTRACT

Monocyte chemoattractant protein 1 (MCP-1) plays an important role in leukocyte recruitment to sites of infection and inflammation. In addition, MCP-1 may attenuate inflammation by virtue of its capacity to inhibit the production of proinflammatory cytokines. We here investigated the role of MCP-1 in lung inflammation induced by lipopolysaccharide (LPS) or lipoteichoic acid (LTA), constituents of the gram-negative and gram-positive bacterial cell wall, respectively. Healthy humans demonstrated elevated MCP-1 concentrations in their bronchoalveolar lavage fluid (BALF) 6h after inhalation of LPS. Similarly, intranasal administration of LPS or LTA to mice resulted in a rise in BALF MCP-1 levels. Murine alveolar macrophage-like cells released significant amounts of MCP-1 upon stimulation with LPS or LTA in vitro. Compared to Wt mice, MCP-1(-/-) mice demonstrated lower TNF-α levels and a diminished neutrophil influx into their bronchoalveolar space after either LPS or LTA instillation. After intrapulmonary delivery of LPS MCP-1(-/-) mice had decreased interleukin-6 and KC concentrations and less severe lung inflammation upon histopathological examination. Remarkably, MCP-1 deficiency was associated with an early enhancement of interleukin-10 release in BALF after both LPS and LTA instillation. These data suggest that MCP-1 is a proinflammatory mediator during pulmonary inflammation induced by either LPS or LTA.


Subject(s)
Chemokine CCL2/physiology , Lipopolysaccharides/immunology , Lung/immunology , Pneumonia/immunology , Teichoic Acids/immunology , Animals , Bronchoalveolar Lavage Fluid/chemistry , Cell Count , Chemokine CCL2/deficiency , Chemotaxis, Leukocyte , Cytokines/biosynthesis , Cytokines/immunology , Cytokines/metabolism , Female , Humans , Interleukin-10/blood , Interleukin-6/blood , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neutrophils/immunology , Tumor Necrosis Factor-alpha/blood , Young Adult
15.
J Infect Dis ; 200(5): 765-73, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19627249

ABSTRACT

BACKGROUND: The receptor for advanced glycation end products (RAGE) mediates a variety of inflammatory responses. METHODS: To determine the role of RAGE in the innate immune response to abdominal sepsis caused by Escherichia coli, RAGE-deficient (RAGE(-/-)) and normal wild-type mice were intraperitoneally injected with E. coli. In a separate experiment, wild-type mice received either anti-RAGE immunoglobulin (Ig) G or control IgG. RESULTS: E. coli sepsis resulted in an up-regulation of RAGE in the liver but not in the lungs. RAGE-deficient mice demonstrated an enhanced bacterial outgrowth in their peritoneal cavity and increased dissemination of the infection, accompanied by increased hepatocellular injury and exaggerated systemic cytokine release and coagulation activation, 20 h after intraperitoneal administration of E. coli. Wild-type mice treated with anti-RAGE IgG also displayed a diminished defense against the growth and/or dissemination of E. coli. RAGE was important for the initiation of the host response, as reflected by a reduced immune and procoagulant response early after intraperitoneal injection of E. coli lipopolysaccharide. CONCLUSION: These data are the first to suggest that intact RAGE signaling contributes to an effective antibacterial defense during E. coli sepsis, thereby limiting the accompanying inflammatory and procoagulant response.


Subject(s)
Escherichia coli Infections/immunology , Escherichia coli/immunology , Receptors, Immunologic/immunology , Sepsis/immunology , Animals , Blood Coagulation , Colony Count, Microbial , Cytokines/blood , Escherichia coli Infections/microbiology , Female , Liver/immunology , Liver/pathology , Lung/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Peritoneal Cavity/microbiology , Receptor for Advanced Glycation End Products , Receptors, Immunologic/deficiency , Sepsis/microbiology
16.
PLoS Pathog ; 5(5): e1000447, 2009 May.
Article in English | MEDLINE | ID: mdl-19461880

ABSTRACT

The causative agent of Lyme borreliosis, the spirochete Borrelia burgdorferi, has been shown to induce expression of the urokinase receptor (uPAR); however, the role of uPAR in the immune response against Borrelia has never been investigated. uPAR not only acts as a proteinase receptor, but can also, dependently or independently of ligation to uPA, directly affect leukocyte function. We here demonstrate that uPAR is upregulated on murine and human leukocytes upon exposure to B. burgdorferi both in vitro as well as in vivo. Notably, B. burgdorferi-inoculated C57BL/6 uPAR knock-out mice harbored significantly higher Borrelia numbers compared to WT controls. This was associated with impaired phagocytotic capacity of B. burgdorferi by uPAR knock-out leukocytes in vitro. B. burgdorferi numbers in vivo, and phagocytotic capacity in vitro, were unaltered in uPA, tPA (low fibrinolytic activity) and PAI-1 (high fibrinolytic activity) knock-out mice compared to WT controls. Strikingly, in uPAR knock-out mice partially backcrossed to a B. burgdorferi susceptible C3H/HeN background, higher B. burgdorferi numbers were associated with more severe carditis and increased local TLR2 and IL-1beta mRNA expression. In conclusion, in B. burgdorferi infection, uPAR is required for phagocytosis and adequate eradication of the spirochete from the heart by a mechanism that is independent of binding of uPAR to uPA or its role in the fibrinolytic system.


Subject(s)
Borrelia burgdorferi/immunology , Lyme Disease/immunology , Receptors, Urokinase Plasminogen Activator/metabolism , Animals , Arthritis, Infectious/microbiology , Cell Movement , Heart/microbiology , Histocytochemistry , Humans , Leukocytes/metabolism , Lyme Disease/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocarditis/microbiology , Phagocytosis , Receptors, Urokinase Plasminogen Activator/genetics , Skin/metabolism , Skin/microbiology , Statistics, Nonparametric , Up-Regulation , Urinary Bladder/metabolism , Urinary Bladder/microbiology , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism
17.
Am J Pathol ; 174(6): 2182-9, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19435793

ABSTRACT

Patients with respiratory failure often require supplemental oxygen therapy and mechanical ventilation. Although both supportive measures are necessary to guarantee adequate oxygen uptake, they can also cause or worsen lung inflammation and injury. Hyperoxia-induced lung injury is characterized by neutrophil infiltration into the lungs. The urokinase plasminogen activator receptor (uPAR) has been deemed important for leukocyte trafficking. To determine the expression and function of neutrophil uPAR during hyperoxia-induced lung injury, uPAR expression was determined on pulmonary neutrophils of mice exposed to hyperoxia. Hyperoxia exposure (O2>80%) for 4 days elicited a pulmonary inflammatory response as reflected by a profound rise in the number of neutrophils that were recovered from bronchoalveolar lavage fluid and lung cell suspensions, as well as increased bronchoalveolar keratinocyte-derived chemokine, interleukin-6, total protein, and alkaline phosphatase levels. In addition, hyperoxia induced the migration of uPAR-positive granulocytes into lungs from wild-type mice compared with healthy control mice (exposed to room air). uPAR deficiency was associated with diminished neutrophil influx into both lung tissues and bronchoalveolar spaces, which was accompanied by a strong reduction in lung injury. Furthermore, in uPAR(-/-) mice, activation of coagulation was diminished. These data suggest that uPAR plays a detrimental role in hyperoxia-induced lung injury and that uPAR deficiency is associated with diminished neutrophil influx into both lung tissues and bronchoalveolar spaces, accompanied by decreased pulmonary injury.


Subject(s)
Hyperoxia/immunology , Lung Injury/immunology , Neutrophils/metabolism , Receptors, Urokinase Plasminogen Activator/biosynthesis , Animals , Blotting, Western , Bronchoalveolar Lavage Fluid/immunology , Chemokines/biosynthesis , Enzyme-Linked Immunosorbent Assay , Female , Interleukin-6/biosynthesis , Lung Injury/pathology , Mice , Mice, Inbred C57BL , Neutrophil Infiltration/immunology , Receptors, Urokinase Plasminogen Activator/deficiency
18.
J Immunol ; 180(5): 3357-65, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18292561

ABSTRACT

Pseudomonas aeruginosa is a major cause of nosocomial pneumonia, which is associated with high morbidity and mortality. Because of its ubiquitous nature and its ability to develop resistance to antibiotics, it is a problematic pathogen from a treatment perspective. Platelet-activating factor receptor (PAFR) is involved in phagocytosis of several pathogens. To determine the role of PAFR in the innate immune response to P. aeruginosa pneumonia, pafr gene-deficient (PAFR-/-) mice and normal wild-type (Wt) mice were intranasally inoculated with P. aeruginosa. PAFR deficiency impaired host defense as reflected by increased bacterial outgrowth and dissemination in mice with a targeted deletion of the PAFR gene. PAFR-/- neutrophils showed a diminished phagocytosing capacity of P. aeruginosa in vitro. Relative to Wt mice, PAFR-/- mice demonstrated increased lung inflammation and injury as reflected by histopathology, relative lung weights and total protein concentrations in bronchoalveolar lavage fluid, which was accompanied by higher levels of proinflammatory cytokines in lung homogenates and plasma. In addition, PAFR deficiency was associated with exaggerated local and systemic activation of coagulation as determined by fibrin staining of lung tissue and pulmonary and plasma concentrations of thrombin-antithrombin complexes and D-dimer. These data suggest that PAFR is an essential component of an effective host response to P. aeruginosa pneumonia, at least partly via its contribution to the phagocytic properties of professional granulocytes. Additionally, our results indicate that PAFR signaling is not essential for the induction of a local and systemic inflammatory and procoagulant response to Pseudomonas pneumonia.


Subject(s)
Inflammation Mediators , Platelet Activating Factor/metabolism , Platelet Membrane Glycoproteins/physiology , Pneumonia, Bacterial/metabolism , Pneumonia, Bacterial/prevention & control , Pseudomonas Infections/metabolism , Pseudomonas Infections/prevention & control , Receptors, G-Protein-Coupled/physiology , Animals , Inflammation Mediators/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet Membrane Glycoproteins/deficiency , Platelet Membrane Glycoproteins/genetics , Pneumonia, Bacterial/pathology , Pseudomonas Infections/pathology , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics
19.
Cell Microbiol ; 10(1): 81-7, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17645551

ABSTRACT

Burkholderia pseudomallei, which causes melioidosis, a severe, mainly pulmonary disease endemic in South-East Asia, is considered to be the most pathogenic of the Burkholderia genus. B. thailandensis, however, is considered avirulent. We determined differences in patterns of inflammation of B. pseudomallei 1026b (clinical virulent isolate), B. pseudomallei AJ1D8 (an in vitro invasion-deficient mutant generated from strain 1026b by Tn5-OT182 mutagenesis) and B. thailandensis by intranasally inoculating C57BL/6 mice with each strain. Mice infected with B. thailandensis showed a markedly decreased bacterial outgrowth from lungs, spleen and blood 24 h after inoculation, compared with infection with B. pseudomallei and the invasion mutant AJ1D8. Forty-eight hours after inoculation, B. thailandensis was no longer detectable. This was consistent with elevated pulmonary cytokine and chemokine concentrations after infection with B. pseudomallei 1026b and AJ1D8, and the absence of these mediators 48 h, but not 24 h, after inoculation with B. thailandensis. Histological examination, however, did show marked pulmonary inflammation in the mice infected with B. thailandensis, corresponding with substantial granulocyte influx and raised myeloperoxidase levels. Survival experiments showed that infection with 1 x 10(3) cfu B. thailandensis was not lethal, whereas inoculation with 1 x 10(6) cfu B. thailandensis was equally lethal as 1 x 10(3) cfu B. pseudomallei 1026b or AJ1D8. These data show that B. pseudomallei AJ1D8 is just as lethal as wild-type B. pseudomallei in an in vivo mouse model, and B. thailandensis is perhaps more virulent than is often recognized.


Subject(s)
Burkholderia/immunology , Burkholderia/pathogenicity , Inflammation/pathology , Melioidosis/microbiology , Melioidosis/pathology , Animals , Blood/microbiology , Burkholderia/genetics , Colony Count, Microbial , Cytokines/analysis , DNA Transposable Elements/genetics , Granulocytes/immunology , Lung/chemistry , Lung/microbiology , Lung/pathology , Melioidosis/immunology , Mice , Mice, Inbred C57BL , Mutagenesis, Insertional , Peroxidase/analysis , Spleen/microbiology , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...