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










Publication year range
1.
J Infect Dis ; 2023 Dec 30.
Article in English | MEDLINE | ID: mdl-38159068

ABSTRACT

Staphylococcus aureus is a prevalent pathogen in pneumonia and harbors glycolipids which may serve as molecular patterns in Mincle (Macrophage inducible C-type lectin) dependent pathogen recognition. We examined the role of Mincle in lung defense against S. aureus in WT, Mincle KO and Mincle transgenic (tg) mice. Two glycolipids, glucosyl-diacylglycerol (Glc-DAG) and diglucosyl-diacylglycerol (Glc2-DAG) were purified, of which only Glc-DAG triggered Mincle reporter cell activation and professional phagocyte responses. Proteomic profiling revealed that Glc2-DAG blocked Glc-DAG-induced cytokine responses, thereby acting as inhibitor of Glc-DAG/Mincle-signaling. WT mice responded to S. aureus with a similar lung pathology as Mincle KO mice, most likely due to Glc2-DAG-dependent inhibition of Glc-DAG/Mincle-signaling. In contrast, ectopic Mincle expression caused severe lung pathology in S. aureus-infected mice characterized by bacterial outgrowth and fatal pneumonia. Collectively, Glc2-DAG inhibits Glc-DAG/Mincle-dependent responses in WT mice, whereas sustained Mincle expression overrides Glc2-DAG-mediated inhibitory effects, conferring increased host susceptibility to S. aureus.

2.
PLoS Pathog ; 19(7): e1011493, 2023 07.
Article in English | MEDLINE | ID: mdl-37467233

ABSTRACT

S100A8/A9 has important immunomodulatory roles in antibacterial defense, but its relevance in focal pneumonia caused by Streptococcus pneumoniae (S. pneumoniae) is understudied. We show that S100A9 was significantly increased in BAL fluids of patients with bacterial but not viral pneumonia and correlated with procalcitonin and sequential organ failure assessment scores. Mice deficient in S100A9 exhibited drastically elevated Zn2+ levels in lungs, which led to bacterial outgrowth and significantly reduced survival. In addition, reduced survival of S100A9 KO mice was characterized by excessive release of neutrophil elastase, which resulted in degradation of opsonophagocytically important collectins surfactant proteins A and D. All of these features were attenuated in S. pneumoniae-challenged chimeric WT→S100A9 KO mice. Similarly, therapy of S. pneumoniae-infected S100A9 KO mice with a mutant S100A8/A9 protein showing increased half-life significantly decreased lung bacterial loads and lung injury. Collectively, S100A9 controls central antibacterial immune mechanisms of the lung with essential relevance to survival of pneumococcal pneumonia. Moreover, S100A9 appears to be a promising biomarker to distinguish patients with bacterial from those with viral pneumonia. Trial registration: Clinical Trials register (DRKS00000620).


Subject(s)
Pneumonia, Pneumococcal , Mice , Animals , Calgranulin B/genetics , Calgranulin B/metabolism , Lung , Streptococcus pneumoniae/metabolism , Calgranulin A/genetics , Calgranulin A/metabolism , Bacteria/metabolism , Mice, Knockout
3.
J Immunol ; 209(11): 2172-2180, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36426980

ABSTRACT

The pathomechanisms underlying the frequently observed fatal outcome of Klebsiella pneumoniae pneumonia in elderly patients are understudied. In this study, we examined the early antibacterial immune response in young mice (age 2-3 mo) as compared with old mice (age 18-19 mo) postinfection with K. pneumoniae. Old mice exhibited significantly higher bacterial loads in lungs and bacteremia as early as 24 h postinfection compared with young mice, with neutrophilic pleuritis nearly exclusively developing in old but not young mice. Moreover, we observed heavily increased cytokine responses in lungs and pleural spaces along with increased mortality in old mice. Mechanistically, Nlrp3 inflammasome activation and caspase-1-dependent IL-1ß secretion contributed to the observed hyperinflammation, which decreased upon caspase-1 inhibitor treatment of K. pneumoniae-infected old mice. Irradiated old mice transplanted with the bone marrow of young mice did not show hyperinflammation or early bacteremia in response to K. pneumoniae. Collectively, the accentuated lung pathology observed in K. pneumoniae-infected old mice appears to be due to regulatory defects of the bone marrow but not the lung, while involving dysregulated activation of the Nlrp3/caspase-1/IL-1ß axis.


Subject(s)
Bacteremia , Pleurisy , Pneumonia , Mice , Animals , Klebsiella , Klebsiella pneumoniae , NLR Family, Pyrin Domain-Containing 3 Protein , Caspase 1
4.
Respir Res ; 23(1): 180, 2022 Jul 08.
Article in English | MEDLINE | ID: mdl-35804363

ABSTRACT

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a diffuse parenchymal lung disease characterized by exuberant deposition of extracellular matrix (ECM) proteins in the lung interstitium, which contributes to substantial morbidity and mortality in IPF patients. Matrix metalloproteinases (MMPs) are a large family of zinc-dependent endopeptidases, many of which have been implicated in the regulation of ECM degradation in lung fibrosis. However, the roles of MMP-2 and -9 (also termed gelatinases A and B) have not yet been explored in lung fibrosis in detail. METHODS: AdTGF-ß1 was applied via orotracheal routes to the lungs of WT, MMP-2 KO, MMP-9 KO and MMP-2/-9 dKO mice on day 0 to induce lung fibrosis. Using hydroxyproline assay, FlexiVent based lung function measurement, histopathology, western blot and ELISA techniques, we analyzed MMP-2 and MMP-9 levels in BAL fluid and lung, collagen contents in lung and lung function in mice on day 14 and 21 post-treatment. RESULT: IPF lung homogenates exhibited significantly increased levels of MMP-2 and MMP-9, relative to disease controls. Enzymatically active MMP-2 and MMP-9 was increased in lungs of mice exposed to adenoviral TGF-ß1, suggesting a role for these metalloproteinases in lung fibrogenesis. However, we found that neither MMP-2 or MMP-9 nor combined MMP-2/-9 deletion had any effect on experimental lung fibrosis in mice. CONCLUSION: Together, our data strongly suggest that both gelatinases MMP-2 and MMP-9 play only a subordinate role in experimental lung fibrosis in mice.


Subject(s)
Idiopathic Pulmonary Fibrosis , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Animals , Extracellular Matrix Proteins/metabolism , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/metabolism , Lung/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinases/metabolism , Mice
5.
JCI Insight ; 6(3)2021 02 08.
Article in English | MEDLINE | ID: mdl-33554955

ABSTRACT

Alpha-1 antitrypsin (AAT) is a major inhibitor of serine proteases in mammals. Therefore, its deficiency leads to protease-antiprotease imbalance and a risk for developing lung emphysema. Although therapy with human plasma-purified AAT attenuates AAT deficiency-related emphysema, its impact on lung antibacterial immunity is poorly defined. Here, we examined the effect of AAT therapy on lung protective immunity in AAT-deficient (KO) mice challenged with Streptococcus pneumoniae. AAT-KO mice were highly susceptible to S. pneumoniae, as determined by severe lobar pneumonia and early mortality. Mechanistically, we found that neutrophil-derived elastase (NE) degraded the opsonophagocytically important collectins, surfactant protein A (SP-A) and D (SP-D), which was accompanied by significantly impaired lung bacterial clearance in S. pneumoniae-infected AAT-KO mice. Treatment of S. pneumoniae-infected AAT-KO mice with human AAT protected SP-A and SP-D from NE-mediated degradation and corrected the pulmonary pathology observed in these mice. Likewise, treatment with Sivelestat, a specific inhibitor of NE, also protected collectins from degradation and significantly decreased bacterial loads in S. pneumoniae-infected AAT-KO mice. Our findings show that NE is responsible for the degradation of lung SP-A and SP-D in AAT-KO mice affecting lung protective immunity in AAT deficiency.


Subject(s)
Lung/immunology , Lung/microbiology , Streptococcus pneumoniae/immunology , Streptococcus pneumoniae/pathogenicity , alpha 1-Antitrypsin Deficiency/immunology , Animals , Female , Humans , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumonia, Pneumococcal/drug therapy , Pneumonia, Pneumococcal/etiology , Pneumonia, Pneumococcal/immunology , Pulmonary Emphysema/etiology , alpha 1-Antitrypsin/genetics , alpha 1-Antitrypsin/immunology , alpha 1-Antitrypsin/metabolism , alpha 1-Antitrypsin/pharmacokinetics , alpha 1-Antitrypsin/therapeutic use , alpha 1-Antitrypsin Deficiency/complications , alpha 1-Antitrypsin Deficiency/genetics
6.
Am J Physiol Lung Cell Mol Physiol ; 320(3): L377-L392, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33296268

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is an interstitial lung disease (ILD) associated with high morbidity and mortality. Patients with ILD frequently develop an acute exacerbation of their disease, which may be triggered by viral and/or bacterial infections. Prostaglandin E2 (PGE2) is an eicosanoid released in a cyclooxygenase-2 (COX2)-dependent manner and is considered to contribute to regulation of lung fibrosis. However, its role in infection-induced exacerbation of lung fibrosis is poorly defined. We found significantly increased levels of PGE2 in lung tissue of patients with ILD. Increased levels of PGE2 were also found in lung tissue of mice with AdTGF-ß1-induced lung fibrosis and even more so in Streptococcus pneumoniae exacerbated lung fibrosis. Type II alveolar epithelial cells (AT II cells) and alveolar macrophages (AM) contributed to PGE2 release during exacerbating fibrosis. Application of parecoxib to inhibit PGE2 synthesis ameliorated lung fibrosis, whereas intratracheal application of PGE2 worsened lung fibrosis in mice. Both interventions had no effect on S. pneumoniae-exacerbated lung fibrosis. Together, we found that the COX2-PGE2 axis has dual roles in fibrosis that may offset each other: PGE2 helps resolve infection/attenuate inflammation in fibrosis exacerbation but accentuates TGF-ß/AT II cell-mediated fibrosis. These data support the efficacy of COX/PGE2 interventions in the setting of non-exacerbating lung fibrosis.


Subject(s)
Alveolar Epithelial Cells/metabolism , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Pneumonia, Pneumococcal/metabolism , Pulmonary Fibrosis/metabolism , Signal Transduction , Streptococcus pneumoniae/metabolism , Alveolar Epithelial Cells/microbiology , Alveolar Epithelial Cells/pathology , Animals , Disease Models, Animal , Female , Isoxazoles/pharmacology , Mice , Pneumonia, Pneumococcal/pathology , Pulmonary Fibrosis/microbiology , Pulmonary Fibrosis/pathology , Transforming Growth Factor beta/metabolism
7.
J Immunol ; 205(12): 3390-3399, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33158955

ABSTRACT

Macrophage-inducible C-type lectin (Mincle)-dependent sensing of pathogens triggers proinflammatory immune responses in professional phagocytes that contribute to protecting the host against pathogen invasion. In this study, we examined whether overexpression of Mincle that is designed to improve early pathogen sensing by professional phagocytes would improve lung-protective immunity against Streptococcus pneumoniae in mice. Proteomic profiling of alveolar macrophages of Mincle transgenic (tg) mice stimulated with the Mincle-specific pneumococcal ligand glucosyl-diacylglycerol (Glc-DAG) revealed increased Nlrp3 inflammasome activation and downstream IL-1ß cytokine release that was not observed in Glc-DAG-stimulated Mincle knockout or Nlrp3 knockout macrophages. Along this line, Mincle tg mice also responded with a stronger Nlrp3 expression and early proinflammatory cytokine release after challenge with S. pneumoniae, ultimately leading to fatal pneumonia in the Mincle tg mice. Importantly, Nlrp3 inhibitor treatment of Mincle tg mice significantly mitigated the observed hyperinflammatory response to pneumococcal challenge. Together, we show that overexpression of the pattern recognition receptor Mincle triggers increased Glc-DAG-dependent Nlrp3 inflammasome activation in professional phagocytes leading to fatal pneumococcal pneumonia in mice that is amenable to Nlrp3 inhibitor treatment. These data show that ectopic expression of the Mincle receptor confers increased susceptibility rather than resistance to S. pneumoniae in mice, thus highlighting the importance of an inducible Mincle receptor expression in response to microbial challenge.


Subject(s)
Lectins, C-Type/immunology , Macrophages, Alveolar/immunology , Membrane Proteins/immunology , Pneumonia, Pneumococcal/immunology , Streptococcus pneumoniae/immunology , Animals , Inflammasomes/genetics , Inflammasomes/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Lectins, C-Type/genetics , Macrophages, Alveolar/pathology , Membrane Proteins/genetics , Mice , Mice, Transgenic , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Pneumonia, Pneumococcal/genetics , Pneumonia, Pneumococcal/pathology
8.
J Immunol ; 204(9): 2429-2438, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32213566

ABSTRACT

Patients with idiopathic pulmonary fibrosis (IPF) can experience life-threatening episodes of acute worsening of their disease, termed acute exacerbation of IPF, which may be caused by bacterial and/or viral infections. The potential for regulatory T cells (Tregs) to limit disease progression in bacterially triggered fibrosis exacerbation has not been explored so far. In the current study, we show that the number of Tregs was significantly increased in mice with established AdTGF-ß1-induced lung fibrosis and further increased in mice with pneumococcal infection-induced lung fibrosis exacerbation. Diphtheria toxin-induced depletion of Tregs significantly worsened infection-induced fibrosis exacerbation as determined by increased lung collagen deposition, lung histology, and elevated pulmonary Th1/Th2 cytokine levels. Conversely, IL-2 complex-induced Treg expansion in wild-type mice with established lung fibrosis completely inhibited pneumococcal infection-induced fibrosis exacerbation as efficaciously as antibiotic treatment while preserving lung antibacterial immunity in mice. Collectively, these findings demonstrate the efficacy of Tregs as "silencers," suppressing infection-induced exacerbation of lung fibrosis in mice, and their expansion may offer a novel adjunctive treatment to limit acute exacerbations in patients with IPF.


Subject(s)
Idiopathic Pulmonary Fibrosis/immunology , Pneumococcal Infections/immunology , Streptococcus pneumoniae/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cytokines/immunology , Disease Models, Animal , Disease Progression , Female , Idiopathic Pulmonary Fibrosis/microbiology , Interleukin-2/immunology , Lung/immunology , Lung/microbiology , Male , Mice , Mice, Inbred C57BL , Pneumococcal Infections/microbiology , Th1 Cells/immunology , Th17 Cells/immunology
9.
Thorax ; 74(10): 947-957, 2019 10.
Article in English | MEDLINE | ID: mdl-31076499

ABSTRACT

RATIONALE: Dendritic cells (DC) accumulate in the lungs of patients with idiopathic lung fibrosis, but their pathogenetic relevance is poorly defined. OBJECTIVES: To assess the role of the FMS-like tyrosine kinase-3 ligand (Flt3L)-lung dendritic cell axis in lung fibrosis. MEASUREMENTS AND MAIN RESULTS: We demonstrate in a model of adenoviral gene transfer of active TGF-ß1 that established lung fibrosis was accompanied by elevated serum Flt3L levels and subsequent accumulation of CD11bpos DC in the lungs of mice. Patients with idiopathic pulmonary fibrosis also demonstrated increased levels of Flt3L protein in serum and lung tissue and accumulation of lung DC in explant subpleural lung tissue specimen. Mice lacking Flt3L showed significantly reduced lung DC along with worsened lung fibrosis and reduced lung function relative to wild-type (WT) mice, which could be inhibited by administration of recombinant Flt3L. Moreover, therapeutic Flt3L increased numbers of CD11bpos DC and improved lung fibrosis in WT mice exposed to AdTGF-ß1. In this line, RNA-sequencing analysis of CD11bpos DC revealed significantly enriched differentially expressed genes within extracellular matrix degrading enzyme and matrix metalloprotease gene clusters. In contrast, the CD103pos DC subset did not appear to be involved in pulmonary fibrogenesis. CONCLUSIONS: We show that Flt3L protein and numbers of lung DC are upregulated in mice and humans during pulmonary fibrogenesis, and increased mobilisation of lung CD11bpos DC limits the severity of lung fibrosis in mice. The current study helps to inform the development of DC-based immunotherapy as a novel intervention against lung fibrosis in humans.


Subject(s)
Collagen/metabolism , Dendritic Cells/metabolism , Lung/metabolism , Pulmonary Fibrosis/metabolism , fms-Like Tyrosine Kinase 3/metabolism , Animals , Dendritic Cells/pathology , Disease Models, Animal , Ligands , Lung/pathology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
10.
Eur J Immunol ; 48(6): 965-974, 2018 06.
Article in English | MEDLINE | ID: mdl-29543979

ABSTRACT

Nasopharyngeal colonization with Streptococcus pneumoniae (the pneumococcus) is known to mount protective adaptive immune responses in rodents and humans. However, the cellular response of the nasopharyngeal compartment to pneumococcal colonization and its importance for the ensuing adaptive immune response is only partially defined. Here we show that nasopharyngeal colonization with S. pneumoniae triggered substantial expansion of both integrin αE (CD103) positive dendritic cells (DC) and T lymphocytes in nasopharynx, nasal-associated lymphoid tissue (NALT) and cervical lymph nodes (CLN) of WT mice. However, nasopharyngeal de-colonization and pneumococcus-specific antibody responses were similar between WT and CD103 KO mice or Batf3 KO mice. Also, naïve WT mice passively immunized with antiserum from previously colonized WT and CD103 KO mice were similarly protected against invasive pneumococcal disease (IPD). In summary, the data show that CD103 is dispensable for pneumococcal colonization-induced adaptive immune responses in mice.


Subject(s)
Antigens, CD/metabolism , Dendritic Cells/immunology , Integrin alpha Chains/metabolism , Lymphoid Tissue/immunology , Nasopharyngeal Diseases/immunology , Pneumococcal Infections/immunology , Streptococcus pneumoniae/physiology , T-Lymphocytes/immunology , Adaptive Immunity , Animals , Antibodies, Bacterial/metabolism , Antigens, CD/genetics , Basic-Leucine Zipper Transcription Factors/genetics , Cell Differentiation , Cells, Cultured , Disease Models, Animal , Female , Humans , Integrin alpha Chains/genetics , Lymphocyte Activation , Lymphoid Tissue/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Repressor Proteins/genetics
11.
Immunobiology ; 223(3): 331-341, 2018 03.
Article in English | MEDLINE | ID: mdl-29089144

ABSTRACT

Granulocyte macrophage-colony stimulating factor (GM-CSF) is a hematopoietic growth factor involved in differentiation, survival and activation of myeloid and non-myeloid cells with important implications for lung antibacterial immunity. Here we examined the effect of pulmonary adenoviral vector-mediated delivery of GM-CSF (AdGM-CSF) on anti-mycobacterial immunity in M. bovis BCG infected mice. Exposure of M. bovis BCG infected mice to AdGM-CSF either applied on 6h, or 6h and 7days post-infection substantially increased alveolar recruitment of iNOS and IL-12 expressing macrophages, and significantly increased accumulation of IFNγpos T cells and particularly regulatory T cells (Tregs). This was accompanied by significantly reduced mycobacterial loads in the lungs of mice. Importantly, diphtheria toxin-induced depletion of Tregs did not influence mycobacterial loads, but accentuated immunopathology in AdGM-CSF-exposed mice infected with M. bovis BCG. Together, the data demonstrate that AdGM-CSF therapy improves lung protective immunity against M. bovis BCG infection in mice independent of co-recruited Tregs, which however critically contribute to limit lung immunopathology in BCG-infected mice. These data may be relevant to the development of immunomodulatory strategies to limit immunopathology-based lung injury in tuberculosis in humans.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Lung/physiology , Macrophages/immunology , Mycobacterium bovis/physiology , T-Lymphocytes, Regulatory/immunology , Tuberculosis/immunology , Adenoviridae/genetics , Animals , Cell Movement , Cells, Cultured , Gene Transfer Techniques , Genetic Vectors , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Immunity, Cellular , Interferon-gamma/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Tuberculosis/veterinary
12.
Eur J Immunol ; 47(3): 540-551, 2017 03.
Article in English | MEDLINE | ID: mdl-28101913

ABSTRACT

Nasopharyngeal colonization with Streptococcus pneumoniae (Spn) is an important precondition for the development of pneumococcal pneumonia. At the same time, nasopharyngeal colonization with Spn has been shown to mount adaptive immune responses against Spn in mice and humans. Cellular responses of the nasopharyngeal compartment, including the nasal-associated lymphoid tissue, to pneumococcal colonization and their importance for developing adaptive immune responses are poorly defined. We show that nasopharyngeal colonization with S. pneumoniae led to substantial expansion of dendritic cells (DCs) both in nasopharyngeal tissue and nasal-associated lymphoid tissue of mice. Depletion of DCs achieved by either diphtheria toxin (DT) treatment of chimeric zDC+/DTR mice, or by use of FMS-like tyrosine kinase 3 ligand (Flt3L) KO mice exhibiting congenitally reduced DC pool sizes, significantly diminished antibody responses after colonization with Spn, along with impaired protective immunity against invasive pneumococcal disease. Collectively, the data show that classical DCs contribute to pneumococcal colonization induced adaptive immune responses against invasive pneumococcal disease in two different mouse models. These data may be useful for future nasopharyngeal vaccination strategies against pneumococcal diseases in humans.


Subject(s)
Dendritic Cells/physiology , Nasopharynx/immunology , Pneumonia, Pneumococcal/immunology , Streptococcus pneumoniae/immunology , Adaptive Immunity , Animals , Antibody Formation/genetics , Cell Proliferation/genetics , Cells, Cultured , Dendritic Cells/microbiology , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Nasopharynx/microbiology , Streptococcus pneumoniae/growth & development , fms-Like Tyrosine Kinase 3/genetics
13.
PLoS Pathog ; 12(12): e1006038, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27923071

ABSTRACT

Among various innate immune receptor families, the role of C-type lectin receptors (CLRs) in lung protective immunity against Streptococcus pneumoniae (S. pneumoniae) is not fully defined. We here show that Mincle gene expression was induced in alveolar macrophages and neutrophils in bronchoalveolar lavage fluids of mice and patients with pneumococcal pneumonia. Moreover, S. pneumoniae directly triggered Mincle reporter cell activation in vitro via its glycolipid glucosyl-diacylglycerol (Glc-DAG), which was identified as the ligand recognized by Mincle. Purified Glc-DAG triggered Mincle reporter cell activation and stimulated inflammatory cytokine release by human alveolar macrophages and alveolar macrophages from WT but not Mincle KO mice. Mincle deficiency led to increased bacterial loads and decreased survival together with strongly dysregulated cytokine responses in mice challenged with focal pneumonia inducing S. pneumoniae, all of which was normalized in Mincle KO mice reconstituted with a WT hematopoietic system. In conclusion, the Mincle-Glc-DAG axis is a hitherto unrecognized element of lung protective immunity against focal pneumonia induced by S. pneumoniae.


Subject(s)
Glycolipids/metabolism , Lectins, C-Type/immunology , Macrophages, Alveolar/immunology , Pneumonia, Pneumococcal/immunology , Receptors, Immunologic/immunology , Streptococcus pneumoniae/immunology , Animals , Chromatography, High Pressure Liquid , Flow Cytometry , Glycolipids/immunology , Humans , Immunophenotyping , Lectins, C-Type/metabolism , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Real-Time Polymerase Chain Reaction , Receptors, Immunologic/metabolism , Streptococcus pneumoniae/metabolism
14.
Thorax ; 70(7): 636-46, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25964315

ABSTRACT

RATIONALE: Respiratory tract infections are common in patients suffering from pulmonary fibrosis. The interplay between bacterial infection and fibrosis is characterised poorly. OBJECTIVES: To assess the effect of Gram-positive bacterial infection on fibrosis exacerbation in mice. METHODS: Fibrosis progression in response to Streptococcus pneumoniae was examined in two different mouse models of pulmonary fibrosis. MEASUREMENTS AND MAIN RESULTS: We demonstrate that wild-type mice exposed to adenoviral vector delivery of active transforming growth factor-ß1 (TGFß1) or diphteria toxin (DT) treatment of transgenic mice expressing the DT receptor (DTR) under control of the surfactant protein C (SPC) promoter (SPC-DTR) to induce pulmonary fibrosis developed progressive fibrosis following infection with Spn, without exhibiting impaired lung protective immunity against Spn. Antibiotic treatment abolished infection-induced fibrosis progression. The cytotoxin pneumolysin (Ply) of Spn caused this phenomenon in a TLR4-independent manner, as Spn lacking Ply (SpnΔply) failed to trigger progressive fibrogenesis, whereas purified recombinant Ply did. Progressive fibrogenesis was also observed in AdTGFß1-exposed Ply-challenged TLR4 KO mice. Increased apoptotic cell death of alveolar epithelial cells along with an attenuated intrapulmonary release of antifibrogenic prostaglandin E2 was found to underlie progressive fibrogenesis in Ply-challenged AdTGFß1-exposed mice. Importantly, vaccination of mice with the non-cytotoxic Ply derivative B (PdB) substantially attenuated Ply-induced progression of lung fibrosis in AdTGFß1-exposed mice. CONCLUSIONS: Our data unravel a novel mechanism by which infection with Spn through Ply release induces progression of established lung fibrosis, which can be attenuated by protein-based vaccination of mice.


Subject(s)
Pneumonia, Pneumococcal/complications , Pulmonary Fibrosis/microbiology , Streptolysins/physiology , Animals , Anti-Bacterial Agents/therapeutic use , Apoptosis/drug effects , Bacterial Proteins/pharmacology , Bacterial Proteins/physiology , Bronchoalveolar Lavage Fluid/immunology , Diphtheria Toxin , Disease Models, Animal , Disease Progression , Epithelial Cells/drug effects , Female , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Pneumococcal Vaccines , Pneumonia, Pneumococcal/drug therapy , Pneumonia, Pneumococcal/immunology , Pneumonia, Pneumococcal/metabolism , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/pathology , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/prevention & control , Streptolysins/deficiency , Streptolysins/pharmacology , Transforming Growth Factor beta1/metabolism
15.
Infect Immun ; 83(1): 184-96, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25332121

ABSTRACT

The macrophage-inducible C-type lectin Mincle has recently been identified to be a pattern recognition receptor sensing mycobacterial infection via recognition of the mycobacterial cell wall component trehalose-6',6-dimycolate (TDM). However, its role in systemic mycobacterial infections has not been examined so far. Mincle-knockout (KO) mice were infected intravenously with Mycobacterium bovis BCG to mimic the systemic spread of mycobacteria under defined experimental conditions. After intravenous infection with M. bovis BCG, Mincle-KO mice responded with significantly higher numbers of mycobacterial CFU in spleen and liver, while reduced granuloma formation was observed only in the spleen. At the same time, reduced Th1 cytokine production and decreased numbers of gamma interferon-producing T cells were observed in the spleens of Mincle-KO mice relative to the numbers in the spleens of wild-type (WT) mice. The effect of adoptive transfer of defined WT leukocyte subsets generated from bone marrow cells of zDC(+/DTR) mice (which bear the human diphtheria toxin receptor [DTR] under the control of the classical dendritic cell-specific zinc finger transcription factor zDC) to specifically deplete Mincle-expressing classical dendritic cells (cDCs) but not macrophages after diphtheria toxin application on the numbers of splenic and hepatic CFU and T cell subsets was then determined. Adoptive transfer experiments revealed that Mincle-expressing splenic cDCs rather than Mincle-expressing macrophages contributed to the reconstitution of attenuated splenic antimycobacterial immune responses in Mincle-KO mice after intravenous challenge with BCG. Collectively, we show that expression of Mincle, particularly by cDCs, contributes to the control of splenic M. bovis BCG infection in mice.


Subject(s)
Dendritic Cells/immunology , Lectins, C-Type/metabolism , Membrane Proteins/metabolism , Mycobacterium bovis/immunology , Spleen/microbiology , Tuberculosis/immunology , Animals , Bacterial Load , Cation Transport Proteins , Colony Count, Microbial , Cytokines/metabolism , Disease Models, Animal , Granuloma/microbiology , Granuloma/pathology , Lectins, C-Type/deficiency , Liver/microbiology , Membrane Proteins/deficiency , Mice, Inbred C57BL , Mice, Knockout , Spleen/immunology , T-Lymphocytes/immunology
16.
J Exp Med ; 209(11): 1937-52, 2012 Oct 22.
Article in English | MEDLINE | ID: mdl-23071253

ABSTRACT

Apoptotic death of alveolar macrophages observed during lung infection with Streptococcus pneumoniae is thought to limit overwhelming lung inflammation in response to bacterial challenge. However, the underlying apoptotic death mechanism has not been defined. Here, we examined the role of the TNF superfamily member TNF-related apoptosis-inducing ligand (TRAIL) in S. pneumoniae-induced macrophage apoptosis, and investigated the potential benefit of TRAIL-based therapy during pneumococcal pneumonia in mice. Compared with WT mice, Trail(-/-) mice demonstrated significantly decreased lung bacterial clearance and survival in response to S. pneumoniae, which was accompanied by significantly reduced apoptosis and caspase 3 cleavage but rather increased necrosis in alveolar macrophages. In WT mice, neutrophils were identified as a major source of intraalveolar released TRAIL, and their depletion led to a shift from apoptosis toward necrosis as the dominant mechanism of alveolar macrophage cell death in pneumococcal pneumonia. Therapeutic application of TRAIL or agonistic anti-DR5 mAb (MD5-1) dramatically improved survival of S. pneumoniae-infected WT mice. Most importantly, neutropenic mice lacking neutrophil-derived TRAIL were protected from lethal pneumonia by MD5-1 therapy. We have identified a previously unrecognized mechanism by which neutrophil-derived TRAIL induces apoptosis of DR5-expressing macrophages, thus promoting early bacterial killing in pneumococcal pneumonia. TRAIL-based therapy in neutropenic hosts may represent a novel antibacterial treatment option.


Subject(s)
Antibodies, Monoclonal/pharmacology , Pneumonia, Pneumococcal/drug therapy , Streptococcus pneumoniae/drug effects , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Animals , Antibodies, Monoclonal/immunology , Apoptosis/drug effects , Caspase 3/metabolism , Cytokines/metabolism , Female , Flow Cytometry , Host-Pathogen Interactions/drug effects , Lung/metabolism , Lung/microbiology , Lung/pathology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Necrosis , Neutrophils/drug effects , Neutrophils/metabolism , Neutrophils/pathology , Pneumonia, Pneumococcal/genetics , Pneumonia, Pneumococcal/microbiology , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Streptococcus pneumoniae/physiology , Survival Analysis , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/metabolism , Treatment Outcome
17.
Infect Immun ; 80(12): 4281-90, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23006850

ABSTRACT

FMS-like tyrosine kinase-3 ligand (Flt3L) is a dendritic cell (DC) growth and differentiation factor with potential in antitumor therapies and antibacterial immunization strategies. However, the effect of systemic Flt3L treatment on lung-protective immunity against bacterial infection is incompletely defined. Here, we examined the impact of deficient (in Flt3L knockout [KO] mice), normal (in wild-type [WT] mice), or increased Flt3L availability (in WT mice pretreated with Flt3L for 3, 5, or 7 days) on lung DC subset profiles and lung-protective immunity against the major lung-tropic pathogen, Streptococcus pneumoniae. Although in Flt3L-deficient mice the numbers of DCs positive for CD11b (CD11b(pos) DCs) and for CD103 (CD103(pos) DCs) were diminished, lung permeability, a marker of injury, was unaltered in response to S. pneumoniae. In contrast, WT mice pretreated with Flt3L particularly responded with increased numbers of CD11b(pos) DCs and with less pronounced numbers of CD103(pos) DCs and impaired bacterial clearance and with increased lung permeability following S. pneumoniae challenge. Notably, infection of Flt3L-pretreated mice with S. pneumoniae lacking the pore-forming toxin, pneumolysin (PLY), resulted in substantially less lung CD11b(pos) DCs activation and reduced lung permeability. Collectively, this study establishes that Flt3L treatment enhances the accumulation of proinflammatory activated lung CD11b(pos) DCs which contribute to acute lung injury in response to PLY released by S. pneumoniae.


Subject(s)
Acute Lung Injury/immunology , Dendritic Cells/immunology , Membrane Proteins/therapeutic use , Streptococcus pneumoniae/pathogenicity , Streptolysins/metabolism , Acute Lung Injury/therapy , Animals , Bacterial Proteins/metabolism , CD11b Antigen/metabolism , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells/pathology , Humans , Inflammation/immunology , Ligands , Lung/drug effects , Lung/immunology , Lung/pathology , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumonia, Pneumococcal/immunology , Pneumonia, Pneumococcal/microbiology , Pneumonia, Pneumococcal/pathology
18.
J Immunol ; 189(6): 3121-9, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22869905

ABSTRACT

The role of macrophage-inducible C-type lectin Mincle in lung innate immunity against mycobacterial infection is incompletely defined. In this study, we show that wild-type (WT) mice responded with a delayed Mincle induction on resident alveolar macrophages and newly immigrating exudate macrophages to infection with Mycobacterium bovis bacillus Calmette-Guérin (BCG), peaking by days 14-21 posttreatment. As compared with WT mice, Mincle knockout (KO) mice exhibited decreased proinflammatory mediator responses and leukocyte recruitment upon M. bovis BCG challenge, and they demonstrated increased mycobacterial loads in pulmonary and extrapulmonary organ systems. Secondary mycobacterial infection on day 14 after primary BCG challenge led to increased cytokine gene expression in sorted alveolar macrophages of WT mice, but not Mincle KO mice, resulting in substantially reduced alveolar neutrophil recruitment and increased mycobacterial loads in the lungs of Mincle KO mice. Collectively, these data show that WT mice respond with a relatively late Mincle expression on lung sentinel cells to M. bovis BCG infection. Moreover, M. bovis BCG-induced upregulation of C-type lectin Mincle on professional phagocytes critically shapes antimycobacterial responses in both pulmonary and extrapulmonary organ systems of mice, which may be important for elucidating the role of Mincle in the control of mycobacterial dissemination in mice.


Subject(s)
Immunity, Innate , Lectins, C-Type/physiology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/pathology , Membrane Proteins/physiology , Mycobacterium bovis/immunology , Tuberculosis/immunology , Tuberculosis/metabolism , Animals , Immunity, Innate/genetics , Inflammation/immunology , Inflammation/microbiology , Inflammation/pathology , Lectins, C-Type/biosynthesis , Lectins, C-Type/deficiency , Lung/immunology , Lung/microbiology , Lung/pathology , Macrophages, Alveolar/microbiology , Membrane Proteins/biosynthesis , Membrane Proteins/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Neutrophils/microbiology , Neutrophils/pathology , Signal Transduction/genetics , Signal Transduction/immunology , Tuberculosis/prevention & control
19.
J Trauma Acute Care Surg ; 73(4): 861-8, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22914078

ABSTRACT

BACKGROUND: Patients with multiple injuries surviving the initial insult are highly susceptible to secondary pneumonia, frequently progressing into sepsis and multiorgan failure. However, the underlying mechanisms of posttraumatic immunosuppression are poorly understood. We hypothesized that dysregulated p38 mitogen-activated protein kinase (MAPK) signaling accounts for impaired lung protective immunity in a model of trauma/hemorrhage (T/H) and subsequent pneumococcal pneumonia in mice. METHODS: C57BL6/N mice were subjected to trauma by midline laparotomy, and T/H was induced by midline laparotomy followed by cannulation of femoral arteries and veins to induce hemorrhage. Subsequently, mice were infected with Streptococcus pneumoniae. In selected experiments, mice were treated with a p38 MAPK inhibitor or vehicle control immediately after induction of T/H. RESULTS: Mice subjected to T/H showed significantly increased p38 MAPK activation in their lungs, which was accompanied by a reduced Escherichia coli phagocytosis by macrophages from T/H mice in vitro and an impaired pneumococcal killing activity of T/H mice in vivo, overall resulting in increased mortality of T/H mice after infection with S. pneumoniae. Application of p38 MAPK inhibitor BIRB796 immediately after T/H induction improved the bacterial phagocytosis activity of macrophages from T/H mice in vitro and lung pneumococcal killing in vivo but did not improve the survival of T/H mice challenged with S. pneumoniae. CONCLUSION: T/H triggers sustained p38 MAPK activation in the lungs of mice, which attenuates lung macrophage antibacterial activities and renders mice more susceptible to pneumococcal pneumonia. However, no major role for dysregulated p38 MAPK to affect survival of T/H mice after pneumococcal challenge was detected, suggesting that dysregulated p38 MAPK activity may possibly play only a limited role in posttraumatic immunosuppression in mice.


Subject(s)
Enzyme Activation/immunology , Immune Tolerance/immunology , Immunocompromised Host , Pneumonia, Pneumococcal/immunology , Wounds and Injuries/immunology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Disease Models, Animal , Female , Macrophages, Alveolar/enzymology , Macrophages, Alveolar/immunology , Mice , Mice, Inbred C57BL , Pneumonia, Pneumococcal/enzymology , Pneumonia, Pneumococcal/etiology , Signal Transduction/immunology , Streptococcus pneumoniae/immunology , Wounds and Injuries/complications , Wounds and Injuries/enzymology , p38 Mitogen-Activated Protein Kinases/immunology
20.
J Immunol ; 188(9): 4476-87, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22461690

ABSTRACT

The neutrophil serine proteases cathepsin G (CG) and neutrophil elastase (NE) are involved in immune-regulatory processes and exert antibacterial activity against various pathogens. To date, their role and their therapeutic potential in pulmonary host defense against mycobacterial infections are poorly defined. In this work, we studied the roles of CG and NE in the pulmonary resistance against Mycobacterium bovis bacillus Calmette-Guérin (BCG). CG-deficient mice and even more pronounced CG/NE-deficient mice showed significantly impaired pathogen elimination to infection with M. bovis BCG in comparison to wild-type mice. Moreover, granuloma formation was more pronounced in M. bovis BCG-infected CG/NE-deficient mice in comparison to CG-deficient and wild-type mice. A close examination of professional phagocyte subsets revealed that exclusively neutrophils shuttled CG and NE into the bronchoalveolar space of M. bovis BCG-infected mice. Accordingly, chimeric wild-type mice with a CG/NE-deficient hematopoietic system displayed significantly increased lung bacterial loads in response to M. bovis BCG infection. Therapeutically applied human CG/NE encapsulated in liposomes colocalized with mycobacteria in alveolar macrophages, as assessed by laser scanning and electron microscopy. Importantly, therapy with CG/NE-loaded liposomes significantly reduced mycobacterial loads in the lungs of mice. Together, neutrophil-derived CG and NE critically contribute to deceleration of pathogen replication during the early phase of antimycobacterial responses. In addition, to our knowledge, we show for the first time that liposomal encapsulated CG/NE exhibit therapeutic potential against pulmonary mycobacterial infections. These findings may be relevant for novel adjuvant approaches in the treatment of tuberculosis in humans.


Subject(s)
Cathepsin G/immunology , Leukocyte Elastase/immunology , Macrophages, Alveolar/immunology , Mycobacterium bovis/immunology , Tuberculosis, Pulmonary/immunology , Animals , Cathepsin G/genetics , Cathepsin G/metabolism , Female , Humans , Leukocyte Elastase/genetics , Leukocyte Elastase/metabolism , Macrophages, Alveolar/enzymology , Macrophages, Alveolar/microbiology , Mice , Mice, Mutant Strains , Tuberculosis, Pulmonary/drug therapy , Tuberculosis, Pulmonary/enzymology , Tuberculosis, Pulmonary/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...