Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Am J Respir Cell Mol Biol ; 59(6): 770-781, 2018 12.
Article in English | MEDLINE | ID: mdl-30110182

ABSTRACT

Influenza viruses can result in significant lung injury with significant morbidity and mortality. In this study, we evaluated the impact of cigarette smoke (CS) exposure on the pulmonary fibroblastic response after influenza infection. We used a murine model in which animals were exposed to CS or room air and subsequently infected with H1N1 influenza virus. Inflammatory and fibrotic responses were measured at different time points after influenza infection. Primary fibroblasts were isolated from the lungs of mice and their characteristics were evaluated. Exposure to CS significantly increased the amount of collagen in the lungs of mice infected with influenza virus compared with the nonsmoking group at 30 days after infection. Furthermore, the presence of fibroblast-specific protein-positive cells increased in the lungs of influenza-infected mice that were exposed to CS compared with the infection-alone group. The smoking group also showed delays in weight recovery and higher cell counts in BAL fluid after infection. Active transforming growth factor ß1 levels in BAL fluid increased in both groups; however, CS-exposed mice had a later surge in active transforming growth factor ß1 (Day 24). Ex vivo cultures of lung-derived fibroblasts from CS-exposed mice with influenza infection showed rapid proliferation, increased expression of α-smooth muscle actin-stained stress fibers, and higher expression of growth factors compared with fibroblasts from room air-exposed lungs after infection. These results suggest that CS exposure changes the fibroblastic potential, leading to increased fibrosis after influenza infection.


Subject(s)
Cigarette Smoking/adverse effects , Fibroblasts/immunology , Influenza A virus/pathogenicity , Orthomyxoviridae Infections/complications , Pneumonia, Viral/complications , Pulmonary Fibrosis/etiology , Animals , Cells, Cultured , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Male , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Pneumonia, Viral/pathology , Pneumonia, Viral/virology , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology
3.
Infect Immun ; 84(7): 2094-2104, 2016 07.
Article in English | MEDLINE | ID: mdl-27141083

ABSTRACT

Pseudomonas aeruginosa causes hospital-acquired pneumonia and is associated with high mortality. An effective response to such an infection includes efficient clearance of pathogenic organisms while limiting collateral damage from the host inflammatory response, known as host resistance and host tolerance, respectively. P. aeruginosa expresses a type III secretion system (T3SS) needle complex that induces NLRC4 (NOD-like receptor C4) activation, interleukin-1ß (IL-1ß) production, and host tissue damage. Chitinase 3-like-1 (Chil1) is expressed during infection and binds to its receptor, IL-13 receptor α2 (IL-13Rα2), to regulate the pathogen-host response during Streptococcus pneumoniae infection, but the role Chil1 plays in balancing the host resistance and host tolerance during P. aeruginosa pneumonia is not known. We conducted experiments using C57BL/6 mice with or without a genetic deficiency of Chil1 and demonstrated that Chil1-deficient mice succumb to P. aeruginosa infection more rapidly than the wild type (WT). The decreased survival time in infected Chil1-deficient mice is associated with more neutrophils recruited to the airways, more lung parenchymal damage, and increased pulmonary consolidation while maintaining equivalent bacterial killing compared to WT mice. Infected Chil1-deficient mice and bone marrow-derived macrophages (BMDMs) from Chil1-deficient mice have increased production of tumor necrosis factor alpha (TNF-α) and IL-1ß compared to infected WT mice and macrophages. Infection of Chil1-deficient BMDMs with non-NLRC4-triggering P. aeruginosa, which is deficient in the T3SS needle complex, did not alter the excessive IL-1ß production compared to BMDMs from WT mice. The addition of recombinant Chil1 decreases the excessive IL-1ß production but only partially rescues stimulated BMDMs from IL-13Rα2-deficient mice. Our data provide mechanistic insights into how Chil1 regulates P. aeruginosa-induced host responses.


Subject(s)
Chitinase-3-Like Protein 1/metabolism , Macrophages/metabolism , Pneumonia, Bacterial/metabolism , Pneumonia, Bacterial/microbiology , Pseudomonas Infections/metabolism , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa , Animals , Bacterial Load , Cell Death/genetics , Cell Death/immunology , Chemotaxis, Leukocyte/genetics , Chemotaxis, Leukocyte/immunology , Chitinase-3-Like Protein 1/genetics , Disease Models, Animal , Gene Expression , Interleukin-1beta/metabolism , Mice , Mice, Knockout , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/mortality , Prognosis , Pseudomonas Infections/immunology , Pseudomonas Infections/mortality , Tumor Necrosis Factor-alpha/metabolism
4.
Am J Respir Cell Mol Biol ; 53(6): 822-33, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25923039

ABSTRACT

Virus-induced exacerbations often lead to further impairment of lung function in chronic obstructive pulmonary disease. IL-15 is critical in antiviral immune responses. Retinoic acid (RA) signaling plays an important role in tissue maintenance and repair, particularly in the lung. We studied RA signaling and its relation to IL-15 in the lung during cigarette smoke (CS) exposure and influenza virus infection. In vivo studies show that RA signaling is diminished by long-term CS exposure or influenza virus infection alone, which is further attenuated during infection after CS exposure. RA receptor ß (RARß) is specifically decreased in the lung of IL-15 transgenic (overexpression; IL-15Tg) mice, and a greater reduction in RARß is found in these mice compared with wild-type (WT) mice after infection. RARß is increased in IL-15 knockout (IL-15KO) mice compared with WT mice after infection, and the additive effect of CS and virus on RARß down-regulation is diminished in IL-15KO mice. IL-15 receptor α (IL-15Rα) is increased and RARß is significantly decreased in lung interstitial macrophages from IL-15Tg mice compared with WT mice. In vitro studies show that IL-15 down-regulates RARß in macrophages via IL-15Rα signaling during influenza virus infection. These studies suggest that RA signaling is significantly diminished in the lung by CS exposure and influenza virus infection. IL-15 specifically down-regulates RARß expression, and RARß may play a protective role in lung injury caused by CS exposure and viral infections.


Subject(s)
Influenza, Human/metabolism , Interleukin-15/physiology , Lung/metabolism , Retinoid X Receptor beta/metabolism , Retinoid X Receptor gamma/metabolism , Smoking/metabolism , Animals , Cell Line , Down-Regulation , Gene Expression Regulation , Humans , Influenza, Human/immunology , Lung/immunology , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin-15/metabolism , Retinoid X Receptor beta/genetics , Retinoid X Receptor gamma/genetics , Smoking/immunology
5.
Am J Physiol Lung Cell Mol Physiol ; 306(6): L487-96, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24441872

ABSTRACT

The pathogenesis of chronic obstructive pulmonary disease (COPD) remains poorly understood. Cellular senescence and apoptosis contribute to the development of COPD; however, crucial regulators of these underlying mechanisms remain unknown. Macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine that antagonizes both apoptosis and premature senescence and may be important in the pathogenesis of COPD. This study examines the role of MIF in the pathogenesis of COPD. Mice deficient in MIF (Mif(-/-)) or the MIF receptor CD74 (Cd74(-/-)) and wild-type (WT) controls were aged for 6 mo. Both Mif(-/-) and Cd74(-/-) mice developed spontaneous emphysema by 6 mo of age compared with WT mice as measured by lung volume and chord length. This was associated with activation of the senescent pathway markers p53/21 and p16. Following exposure to cigarette smoke, Mif(-/-) mice were more susceptible to the development of COPD and apoptosis compared with WT mice. MIF plasma concentrations were measured in a cohort of 224 human participants. Within a subgroup of older current and former smokers (n = 72), MIF concentrations were significantly lower in those with COPD [8.8, 95%CI (6.7-11.0)] compared with those who did not exhibit COPD [12.7 ng/ml, 95%CI (10.6-14.8)]. Our results suggest that both MIF and the MIF receptor CD74 are required for maintenance of normal alveolar structure in mice and that decreases in MIF are associated with COPD in human subjects.


Subject(s)
Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/genetics , Pulmonary Disease, Chronic Obstructive/genetics , Receptors, Immunologic/blood , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Animals , Apoptosis/drug effects , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Emphysema/genetics , Female , Humans , Intramolecular Oxidoreductases/deficiency , Lung/metabolism , Macrophage Migration-Inhibitory Factors/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Smoke/adverse effects , Smoking/adverse effects , Nicotiana/adverse effects , Tumor Suppressor Protein p53/metabolism , Young Adult
6.
Sci Signal ; 11(528)2018 05 01.
Article in English | MEDLINE | ID: mdl-29717063

ABSTRACT

Sepsis-induced organ damage is caused by systemic inflammatory response syndrome (SIRS), which results in substantial comorbidities. Therefore, it is of medical importance to identify molecular brakes that can be exploited to dampen inflammation and prevent the development of SIRS. We investigated the role of phosphatase and tensin homolog (PTEN) in suppressing SIRS, increasing microbial clearance, and preventing lung damage. Septic patients and mice with sepsis exhibited increased PTEN expression in leukocytes. Myeloid-specific Pten deletion in an animal model of sepsis increased bacterial loads and cytokine production, which depended on enhanced myeloid differentiation primary response gene 88 (MyD88) abundance and resulted in mortality. PTEN-mediated induction of the microRNAs (miRNAs) miR125b and miR203b reduced the abundance of MyD88. Loss- and gain-of-function assays demonstrated that PTEN induced miRNA production by associating with and facilitating the nuclear localization of Drosha-Dgcr8, part of the miRNA-processing complex. Reconstitution of PTEN-deficient mouse embryonic fibroblasts with a mutant form of PTEN that does not localize to the nucleus resulted in retention of Drosha-Dgcr8 in the cytoplasm and impaired production of mature miRNAs. Thus, we identified a regulatory pathway involving nuclear PTEN-mediated miRNA generation that limits the production of MyD88 and thereby limits sepsis-associated mortality.


Subject(s)
MicroRNAs/genetics , Myeloid Differentiation Factor 88/genetics , PTEN Phosphohydrolase/genetics , Regulon/genetics , Sepsis/genetics , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , Female , Gene Expression Profiling , Humans , Inflammation/genetics , Inflammation/metabolism , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/chemistry , Myeloid Differentiation Factor 88/metabolism , PTEN Phosphohydrolase/metabolism , Peptides/pharmacology , RNA Interference , Sepsis/metabolism , Sepsis/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL