Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
2.
PLoS Pathog ; 9(2): e1003188, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23468627

ABSTRACT

Influenza viruses (IV) cause pneumonia in humans with progression to lung failure and fatal outcome. Dysregulated release of cytokines including type I interferons (IFNs) has been attributed a crucial role in immune-mediated pulmonary injury during severe IV infection. Using ex vivo and in vivo IV infection models, we demonstrate that alveolar macrophage (AM)-expressed IFN-ß significantly contributes to IV-induced alveolar epithelial cell (AEC) injury by autocrine induction of the pro-apoptotic factor TNF-related apoptosis-inducing ligand (TRAIL). Of note, TRAIL was highly upregulated in and released from AM of patients with pandemic H1N1 IV-induced acute lung injury. Elucidating the cell-specific underlying signalling pathways revealed that IV infection induced IFN-ß release in AM in a protein kinase R- (PKR-) and NF-κB-dependent way. Bone marrow chimeric mice lacking these signalling mediators in resident and lung-recruited AM and mice subjected to alveolar neutralization of IFN-ß and TRAIL displayed reduced alveolar epithelial cell apoptosis and attenuated lung injury during severe IV pneumonia. Together, we demonstrate that macrophage-released type I IFNs, apart from their well-known anti-viral properties, contribute to IV-induced AEC damage and lung injury by autocrine induction of the pro-apoptotic factor TRAIL. Our data suggest that therapeutic targeting of the macrophage IFN-ß-TRAIL axis might represent a promising strategy to attenuate IV-induced acute lung injury.


Subject(s)
Acute Lung Injury/metabolism , Influenza, Human/metabolism , Interferon-beta/metabolism , Macrophages, Alveolar/metabolism , Pneumonia, Viral/metabolism , Respiratory Mucosa/metabolism , Acute Lung Injury/immunology , Acute Lung Injury/pathology , Adult , Animals , Apoptosis , Disease Models, Animal , Humans , Influenza, Human/immunology , Influenza, Human/pathology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mosaicism , Pneumonia, Viral/immunology , Pneumonia, Viral/pathology , Respiratory Mucosa/immunology , Respiratory Mucosa/pathology , TNF-Related Apoptosis-Inducing Ligand/metabolism
3.
J Clin Invest ; 122(10): 3652-64, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22996662

ABSTRACT

Influenza viruses (IVs) cause pneumonia in humans with progression to lung failure. Pulmonary DCs are key players in the antiviral immune response, which is crucial to restore alveolar barrier function. The mechanisms of expansion and activation of pulmonary DC populations in lung infection remain widely elusive. Using mouse BM chimeric and cell-specific depletion approaches, we demonstrated that alveolar epithelial cell (AEC) GM-CSF mediates recovery from IV-induced injury by affecting lung DC function. Epithelial GM-CSF induced the recruitment of CD11b+ and monocyte-derived DCs. GM-CSF was also required for the presence of CD103+ DCs in the lung parenchyma at baseline and for their sufficient activation and migration to the draining mediastinal lymph nodes (MLNs) during IV infection. These activated CD103+ DCs were indispensable for sufficient clearance of IVs by CD8+ T cells and for recovery from IV-induced lung injury. Moreover, GM-CSF applied intratracheally activated CD103+ DCs, inducing increased migration to MLNs, enhanced viral clearance, and attenuated lung injury. Together, our data reveal that GM-CSF-dependent cross-talk between IV-infected AECs and CD103+ DCs is crucial for effective viral clearance and recovery from injury, which has potential implications for GM-CSF treatment in severe IV pneumonia.


Subject(s)
Dendritic Cells/physiology , Epithelial Cells/physiology , Influenza, Human/immunology , Orthomyxoviridae Infections/immunology , Pneumonia, Viral/immunology , Pulmonary Alveoli/pathology , Animals , Antigens, CD/analysis , Antigens, Surface/genetics , Bone Marrow Transplantation , Cells, Cultured/immunology , Cells, Cultured/virology , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza, Human/pathology , Instillation, Drug , Integrin alpha Chains/analysis , Lectins, C-Type/genetics , Lung/immunology , Lung/virology , Mannose-Binding Lectins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Orthomyxoviridae Infections/pathology , Pneumonia, Viral/pathology , Pulmonary Alveoli/immunology , Radiation Chimera , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Specific Pathogen-Free Organisms , Trachea
4.
Am J Respir Crit Care Med ; 183(10): 1380-90, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21278303

ABSTRACT

RATIONALE: Exudate macrophages are key players in host defense toward invading pathogens. Their antiinflammatory and epithelial-protective potential in gram-negative pneumonia, however, remains elusive. OBJECTIVES: We investigated whether exudate macrophages contributed to preservation of alveolar epithelial barrier integrity and analyzed the molecular pathways involved. METHODS: We evaluated the antiinflammatory and epithelial-protective effects of exudate macrophages in a model of LPS- and Klebsiella pneumoniae-induced lung injury comparing wild-type and CC-chemokine receptor 2 (CCR2)-deficient mice with defective lung macrophage recruitment and in in vitro studies using primary alveolar epithelial cells. MEASUREMENTS AND MAIN RESULTS: CCR2(-/-) mice exhibited enhanced alveolar epithelial cell apoptosis and lung leakage on intratracheal LPS treatment, which could be attributed to lack of exudate macrophage recruitment from the circulating pool as demonstrated in a model of wild-type/CCR2(-/-) bone-marrow chimeric mice. Among various antiinflammatory and proliferative mediators analyzed, the endogenous counterpart of resident macrophage-expressed IL-1ß, IL-1 receptor antagonist (IL-1ra), was highly up-regulated in flow-sorted exudate macrophages in LPS-treated wild-type mice. LPS/IL-1ß-induced impairment of alveolar epithelial cell integrity was antagonized by IL-1ra in vitro. Finally, intratracheal substitution of IL-1ra or intravenous adoptive transfer of IL-1ra(+/+) but not IL-1ra(-/-) blood mononuclear cells attenuated alveolar inflammation, epithelial apoptosis, and loss of barrier function in LPS-challenged or K. pneumoniae-infected CCR2(-/-) mice and enhanced survival after K. pneumoniae infection. CONCLUSIONS: We conclude that recruited lung macrophages attenuate IL-1ß-mediated acute lung injury in gram-negative pneumonia by release of IL-1ra.


Subject(s)
Acute Lung Injury/immunology , Exudates and Transudates/immunology , Interleukin 1 Receptor Antagonist Protein/immunology , Klebsiella Infections/immunology , Macrophages, Alveolar/immunology , Pneumonia, Bacterial/immunology , Animals , Apoptosis/immunology , Cell Culture Techniques , Disease Models, Animal , Humans , Klebsiella pneumoniae/immunology , Lung/immunology , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...