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
1.
Cells ; 11(3)2022 02 03.
Article in English | MEDLINE | ID: mdl-35159341

ABSTRACT

Pneumonia is a leading cause of morbidity and mortality. While inflammation is a host protective response that ensures bacterial clearance, a finely regulated response is necessary to prevent bystander tissue damage. Glucocorticoid (GC)-induced leucine zipper (GILZ) is a GC-induced protein with anti-inflammatory and proresolving bioactions, yet the therapeutical role of GILZ in infectious diseases remains unexplored. Herein, we investigate the role and effects of GILZ during acute lung injury (ALI) induced by LPS and Streptococcus pneumoniae infection. GILZ deficient mice (GILZ-/-) presented more severe ALI, characterized by increased inflammation, decreased macrophage efferocytosis and pronounced lung damage. In contrast, pulmonary inflammation, and damage were attenuated in WT mice treated with TAT-GILZ fusion protein. During pneumococcal pneumonia, TAT-GILZ reduced neutrophilic inflammation and prevented the associated lung damage. There was also enhanced macrophage efferocytosis and bacterial clearance in TAT-GILZ-treated mice. Mechanistically, TAT-GILZ enhanced macrophage phagocytosis of pneumococcus, which was lower in GILZ-/- macrophages. Noteworthy, early treatment with TAT-GILZ rescued 30% of S. pneumoniae-infected mice from lethal pneumonia. Altogether, we present evidence that TAT-GILZ enhances host resilience and resistance to pneumococcal pneumonia by controlling pulmonary inflammation and bacterial loads leading to decreased lethality. Exploiting GILZ pathways holds promise for the treatment of severe respiratory infections.


Subject(s)
Pneumonia, Pneumococcal , Animals , Glucocorticoids/pharmacology , Inflammation/metabolism , Leucine Zippers , Mice , Pneumonia, Pneumococcal/complications , Pneumonia, Pneumococcal/drug therapy , Streptococcus pneumoniae/metabolism , Transcription Factors/metabolism
2.
Pharmacol Res ; 163: 105292, 2021 01.
Article in English | MEDLINE | ID: mdl-33171305

ABSTRACT

Resolution failure of exacerbated inflammation triggered by Influenza A virus (IAV) prevents return of pulmonary homeostasis and survival, especially when associated with secondary pneumococcal infection. Therapeutic strategies based on pro-resolving molecules have great potential against acute inflammatory diseases. Angiotensin-(1-7) [Ang-(1-7)] is a pro-resolving mediator that acts on its Mas receptor (MasR) to promote resolution of inflammation. We investigated the effects of Ang-(1-7) and the role of MasR in the context of primary IAV infection and secondary pneumococcal infection and evaluated pulmonary inflammation, virus titers and bacteria counts, and pulmonary damage. Therapeutic treatment with Ang-(1-7) decreased neutrophil recruitment, lung injury, viral load and morbidity after a primary IAV infection. Ang-(1-7) induced apoptosis of neutrophils and efferocytosis of these cells by alveolar macrophages, but had no direct effect on IAV replication in vitro. MasR-deficient (MasR-/-) mice were highly susceptible to IAV infection, displaying uncontrolled inflammation, increased viral load and greater lethality rate, as compared to WT animals. Ang-(1-7) was not protective in MasR-/- mice. Interestingly, Ang-(1-7) given during a sublethal dose of IAV infection greatly reduced morbidity associated with a subsequent S. pneumoniae infection, as seen by decrease in the magnitude of neutrophil influx, number of bacteria in the blood leading to a lower lethality. Altogether, these results show that Ang-(1-7) is highly protective against severe primary IAV infection and protects against secondary bacterial infection of the lung. These effects are MasR-dependent. Mediators of resolution of inflammation, such as Ang-(1-7), should be considered for the treatment of pulmonary viral infections.


Subject(s)
Angiotensin I/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Peptide Fragments/therapeutic use , Pneumococcal Infections/drug therapy , Pneumonia, Viral/drug therapy , Proto-Oncogene Proteins/immunology , Receptors, G-Protein-Coupled/immunology , A549 Cells , Angiotensin I/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cytokines/immunology , Dogs , Humans , Influenza A virus , Lung/drug effects , Lung/immunology , Lung/pathology , Madin Darby Canine Kidney Cells , Male , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/drug effects , Neutrophils/immunology , Peptide Fragments/pharmacology , Peroxidase/immunology , Phagocytosis/drug effects , Pneumococcal Infections/immunology , Pneumococcal Infections/pathology , Pneumonia, Viral/immunology , Pneumonia, Viral/pathology , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Receptors, G-Protein-Coupled/genetics , Streptococcus pneumoniae
3.
Cell Rep ; 30(9): 2934-2947.e6, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32130898

ABSTRACT

Secondary bacterial infections often complicate viral respiratory infections. We hypothesize that perturbation of the gut microbiota during influenza A virus (IAV) infection might favor respiratory bacterial superinfection. Sublethal infection with influenza transiently alters the composition and fermentative activity of the gut microbiota in mice. These changes are attributed in part to reduced food consumption. Fecal transfer experiments demonstrate that the IAV-conditioned microbiota compromises lung defenses against pneumococcal infection. In mechanistic terms, reduced production of the predominant short-chain fatty acid (SCFA) acetate affects the bactericidal activity of alveolar macrophages. Following treatment with acetate, mice colonized with the IAV-conditioned microbiota display reduced bacterial loads. In the context of influenza infection, acetate supplementation reduces, in a free fatty acid receptor 2 (FFAR2)-dependent manner, local and systemic bacterial loads. This translates into reduced lung pathology and improved survival rates of double-infected mice. Lastly, pharmacological activation of the SCFA receptor FFAR2 during influenza reduces bacterial superinfection.


Subject(s)
Dysbiosis/microbiology , Fatty Acids, Volatile/biosynthesis , Gastrointestinal Tract/microbiology , Influenza, Human/microbiology , Lung/microbiology , Pneumococcal Infections/complications , Superinfection/complications , Superinfection/microbiology , Acetates/pharmacology , Animals , Dysbiosis/complications , Dysbiosis/virology , Feeding Behavior , Gastrointestinal Microbiome/drug effects , Gastrointestinal Tract/drug effects , Humans , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/microbiology , Macrophages, Alveolar/pathology , Mice, Inbred C57BL , Pneumococcal Infections/microbiology , Pneumococcal Infections/virology , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Respiratory Tract Infections/microbiology
4.
Front Immunol ; 8: 1799, 2017.
Article in English | MEDLINE | ID: mdl-29326698

ABSTRACT

RATIONALE: Influenza A infections are a leading cause of morbidity and mortality worldwide especially when associated with secondary pneumococcal infections. Inflammation is important to control pathogen proliferation but may also cause tissue injury and death. CXCR1/2 are chemokine receptors relevant for the recruitment of neutrophils. We investigated the role of CXCR1/2 during influenza, pneumococcal, and post-influenza pneumococcal infections. METHODS: Mice were infected with influenza A virus (IAV) or Streptococcus pneumoniae and then treated daily with the CXCR1/2 antagonist DF2162. To study secondary pneumococcal infection, mice were infected with a sublethal inoculum of IAV then infected with S. pneumoniae 14 days later. DF2162 was given in a therapeutic schedule from days 3 to 6 after influenza infection. Lethality, weight loss, inflammation, virus/bacteria counts, and lung injury were assessed. RESULTS: CXCL1 and CXCL2 were produced at high levels during IAV infection. DF2162 treatment decreased morbidity and this was associated with decreased infiltration of neutrophils in the lungs and reduced pulmonary damage and viral titers. During S. pneumoniae infection, DF2162 treatment decreased neutrophil recruitment, pulmonary damage, and lethality rates, without affecting bacteria burden. Therapeutic treatment with DF2162 during sublethal IAV infection reduced the morbidity associated with virus infection and also decreased the magnitude of inflammation, lung damage, and number of bacteria in the blood of mice subsequently infected with S. pneumoniae. CONCLUSION: Modulation of the inflammatory response by blocking CXCR1/2 improves disease outcome during respiratory influenza and pneumococcal infections, without compromising the ability of the murine host to deal with infection. Altogether, inhibition of CXCR1/2 may be a valid therapeutic strategy for treating lung infections caused by these pathogens, especially controlling secondary bacterial infection after influenza.

SELECTION OF CITATIONS
SEARCH DETAIL
...