Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Biochem Biophys ; 67(2): 399-414, 2013 Nov.
Article in English | MEDLINE | ID: mdl-22183615

ABSTRACT

Adiponectin (Ad), an adipokine exclusively secreted by the adipose tissue, has emerged as a paracrine metabolic regulator as well as a protectant against oxidative stress. Pharmacological approaches of protecting against clinical hyperoxic lung injury during oxygen therapy/treatment are limited. We have previously reported that Ad inhibits the NADPH oxidase-catalyzed formation of superoxide from molecular oxygen in human neutrophils. Based on this premise, we conducted studies to determine whether (i) exogenous Ad would protect against the hyperoxia-induced barrier dysfunction in the lung endothelial cells (ECs) in vitro, and (ii) endogenously synthesized Ad would protect against hyperoxic lung injury in wild-type (WT) and Ad-overexpressing transgenic (AdTg) mice in vivo. The results demonstrated that exogenous Ad protected against the hyperoxia-induced oxidative stress, loss of glutathione (GSH), cytoskeletal reorganization, barrier dysfunction, and leak in the lung ECs in vitro. Furthermore, the hyperoxia-induced lung injury, vascular leak, and lipid peroxidation were significantly attenuated in AdTg mice in vivo. Also, AdTg mice exhibited elevated levels of total thiols and GSH in the lungs as compared with WT mice. For the first time, our studies demonstrated that Ad protected against the hyperoxia-induced lung damage apparently through attenuation of oxidative stress and modulation of thiol-redox status.


Subject(s)
Adiponectin/metabolism , Adiponectin/pharmacology , Blood Vessels/drug effects , Blood Vessels/pathology , Lung Injury/metabolism , Lung Injury/pathology , Adiponectin/genetics , Animals , Cattle , Cell Hypoxia/drug effects , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Glutathione/metabolism , Humans , Lipid Peroxidation/drug effects , Lung/pathology , Male , Mice , Mice, Transgenic , Oxidative Stress/drug effects , Permeability/drug effects , Reactive Oxygen Species/metabolism , Tight Junctions/drug effects , Tight Junctions/metabolism
2.
Autophagy ; 7(11): 1359-70, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21997369

ABSTRACT

Cystic fibrosis (CF) is the most common inherited lethal disease of Caucasians which results in multi organ dysfunction. However, 85% of the deaths are due to pulmonary infections. Infection by Burkholderia cenocepacia (B. cepacia) is a particularly lethal threat to CF patients because it causes severe and persistent lung inflammation and is resistant to nearly all available antibiotics. In CFTR ΔF508 mouse macrophages, B. cepacia persists in vacuoles that do not fuse with the lysosomes and mediates increased production of IL-1ß. It is believed that intracellular bacterial survival contributes to the persistence of the bacterium. Here we show for the first time that in wild-type macrophages but not in ΔF508 macrophages, many B. cepacia reside in autophagosomes that fuse with lysosomes at later stages of infection. Accordingly, association and intracellular survival of B. cepacia are higher in CFTR-ΔF508 (ΔF508) macrophages than in WT macrophages. An autophagosome is a compartment that engulfs non-functional organelles and parts of the cytoplasm then delivers them to the lysosome for degradation to produce nutrients during periods of starvation or stress. Furthermore, we show that B. cepacia downregulates autophagy genes in WT and ΔF508 macrophages. However, autophagy dysfunction is more pronounced in ΔF508 macrophages since they already have compromised autophagy activity. We demonstrate that the autophagy-stimulating agent, rapamycin markedly decreases B. cepacia infection in vitro by enhancing the clearance of B. cepacia via induced autophagy. In vivo, Rapamycin decreases bacterial burden in the lungs of CF mice and drastically reduces signs of lung inflammation. Together, our studies reveal that if efficiently activated, autophagy can control B. cepacia infection and ameliorate the associated inflammation. Therefore, autophagy is a novel target for new drug development for CF patients to control B. cepacia infection and accompanying inflammation.


Subject(s)
Autophagy/drug effects , Burkholderia Infections/drug therapy , Burkholderia cenocepacia/physiology , Cystic Fibrosis/drug therapy , Pneumonia/drug therapy , Sirolimus/pharmacology , Sirolimus/therapeutic use , Animals , Autophagy/genetics , Burkholderia Infections/complications , Burkholderia Infections/microbiology , Burkholderia Infections/pathology , Burkholderia cenocepacia/drug effects , Burkholderia cenocepacia/growth & development , Burkholderia cenocepacia/ultrastructure , Cystic Fibrosis/complications , Cystic Fibrosis/microbiology , Cystic Fibrosis/pathology , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Disease Models, Animal , Down-Regulation/genetics , Interleukin-1beta/biosynthesis , Intracellular Space/drug effects , Intracellular Space/microbiology , Lysosomes/drug effects , Lysosomes/microbiology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/microbiology , Macrophages/ultrastructure , Mice , Mice, Inbred C57BL , Microbial Viability/drug effects , Microtubule-Associated Proteins/metabolism , Mutation/genetics , Phagosomes/drug effects , Phagosomes/microbiology , Phagosomes/ultrastructure , Pneumonia/complications , Pneumonia/microbiology , RNA, Small Interfering/metabolism , Vacuoles/drug effects , Vacuoles/microbiology
3.
Am J Respir Cell Mol Biol ; 45(5): 999-1006, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21562315

ABSTRACT

Ets-2 is a ubiquitous transcription factor activated after phosphorylation at threonine-72. Previous studies highlighted the importance of phosphorylated ets-2 in lung inflammation and extracellular matrix remodeling, two pathways involved in pulmonary fibrosis. We hypothesized that phosphorylated ets-2 played an important role in pulmonary fibrosis, and we sought to determine the role of ets-2 in its pathogenesis. We challenged ets-2 (A72/A72) transgenic mice (harboring a mutated form of ets-2 at phosphorylation site threonine-72) and ets-2 (wild-type/wild-type [WT/WT]) control mice with sequential intraperitoneal injections of bleomycin, followed by quantitative measurements of lung fibrosis and inflammation and primary cell in vitro assays. Concentrations of phosphorylated ets-2 were detected via the single and dual immunohistochemical staining of murine lungs and lung sections from patients with idiopathic pulmonary fibrosis. Ets-2 (A72/A72) mice were protected from bleomycin-induced pulmonary fibrosis, compared with ets-2 (WT/WT) mice. This protection was characterized by decreased lung pathological abnormalities and the fibrotic gene expression of Type I collagen, Type III collagen, α-smooth muscle actin, and connective tissue growth factor. Immunohistochemical staining of lung sections from bleomycin-treated ets-2 (WT/WT) mice and from patients with idiopathic pulmonary fibrosis demonstrated increased staining of phosphorylated ets-2 that colocalized with Type I collagen expression and to fibroblastic foci. Lastly, primary lung fibroblasts from ets-2 (A72/A72) mice exhibited decreased expression of Type I collagen in response to stimulation with TGF-ß, compared with fibroblasts from ets-2 (WT/WT) mice. These data indicate the importance of phosphorylated ets-2 in the pathogenesis of pulmonary fibrosis through the expression of Type I collagen and (myo)fibroblast activation.


Subject(s)
Proto-Oncogene Protein c-ets-2/metabolism , Pulmonary Fibrosis/metabolism , Actins/biosynthesis , Actins/genetics , Animals , Bleomycin/adverse effects , Cells, Cultured , Collagen Type I/biosynthesis , Collagen Type I/genetics , Collagen Type III/biosynthesis , Collagen Type III/genetics , Connective Tissue Growth Factor/biosynthesis , Connective Tissue Growth Factor/genetics , Fibroblasts/metabolism , Gene Expression , Humans , Lung/chemistry , Lung/pathology , Male , Mice , Mice, Transgenic , Phosphorylation , Pneumonia/chemically induced , Pneumonia/pathology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology
4.
PLoS One ; 6(5): e19654, 2011 May 09.
Article in English | MEDLINE | ID: mdl-21573017

ABSTRACT

BACKGROUND: Thrombospondin-1 (TSP-1) is involved in many biological processes, including immune and tissue injury response, but its role in sepsis is unknown. Cell surface expression of TSP-1 on platelets is increased in sepsis and could activate the anti-inflammatory cytokine transforming growth factor beta (TGFß1) affecting outcome. Because of these observations we sought to determine the importance of TSP-1 in sepsis. METHODOLOGY/PRINCIPAL FINDINGS: We performed studies on TSP-1 null and wild type (WT) C57BL/6J mice to determine the importance of TSP-1 in sepsis. We utilized the cecal ligation puncture (CLP) and intraperitoneal E. coli injection (i.p. E. coli) models of peritoneal sepsis. Additionally, bone-marrow-derived macrophages (BMMs) were used to determine phagocytic activity. TSP-1-/- animals experienced lower mortality than WT mice after CLP. Tissue and peritoneal lavage TGFß1 levels were unchanged between animals of each genotype. In addition, there is no difference between the levels of major innate cytokines between the two groups of animals. PLF from WT mice contained a greater bacterial load than TSP-1-/- mice after CLP. The survival advantage for TSP-1-/- animals persisted when i.p. E. coli injections were performed. TSP-1-/- BMMs had increased phagocytic capacity compared to WT. CONCLUSIONS: TSP-1 deficiency was protective in two murine models of peritoneal sepsis, independent of TGFß1 activation. Our studies suggest TSP-1 expression is associated with decreased phagocytosis and possibly bacterial clearance, leading to increased peritoneal inflammation and mortality in WT mice. These data support the contention that TSP-1 should be more fully explored in the human condition.


Subject(s)
Immunity, Innate/immunology , Sepsis/immunology , Sepsis/pathology , Thrombospondin 1/metabolism , Animals , Bacterial Load/immunology , Cecum/microbiology , Cecum/pathology , Cell Count , Cytokines/blood , Cytoprotection , Disease Models, Animal , Humans , Inflammation Mediators/metabolism , Ligation , Macrophages/cytology , Mice , Peritoneal Lavage , Peritoneum/microbiology , Peritoneum/pathology , Phagocytosis , Punctures , Sepsis/blood , Sepsis/microbiology , Survival Analysis , Thrombospondin 1/deficiency , Wound Healing
5.
Biomaterials ; 32(2): 538-46, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20875916

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is an interstitial lung disease characterized by irreversible scarring. Collagen deposition, myofibroblast expansion, and the development of fibroblastic foci are the hallmark pathological events. The origin and mechanism of recruitment of myofibroblasts, the key cell contributing to these events, is unknown. We hypothesize that the fibrotic lung microenvironment causes differentiation of arriving bone marrow-derived cells into myofibroblasts. Therefore, a method of isolating the effects of fibrotic microenvironment components on various cell types was developed. Electrospun nanofibers were coated with lung extracts from fibrotic or non-fibrotic mice and used to determine effects on bone marrow cells from naïve mice. Varying moduli nanofibers were also employed to determine matrix stiffness effects on these cells. At structured time points, bone marrow cell morphology was recorded and changes in fibrotic gene expression determined by real-time PCR. Cells plated on extracts isolated from fibrotic murine lungs secreted larger amounts of extracellular matrix, adopted a fibroblastic morphology, and exhibited increased myofibroblast gene expression after 8 and 14 days; cells plated on extracts from non-fibrotic lungs did not. Similar results were observed when the nanofiber modulus was increased. This ex vivo system appears to recapitulate the three-dimensional fibrotic lung microenvironment.


Subject(s)
Nanofibers , Animals , Bone Marrow Cells/cytology , Cell Culture Techniques/methods , Fibroblasts/cytology , Male , Mice , Polyesters/chemistry , Pulmonary Fibrosis/metabolism
6.
Am J Respir Cell Mol Biol ; 44(4): 556-61, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20581099

ABSTRACT

Thrombospondin-1 (TSP-1) is an extracellular protein critical to normal lung homeostasis, and is reported to activate latent transforming growth factor-ß (TGF-ß). Because active TGF-ß is causally involved in lung fibrosis after bleomycin challenge, alterations in TSP-1 may be relevant to pulmonary fibrosis. We sought to determine the effects of TSP-1 deficiency on the susceptibility to bleomycin-induced pulmonary fibrosis in a murine model. Age-matched and sex-matched C57BL/6 wild-type (WT) and TSP-1-deficient mice were treated twice weekly for 4 weeks with intraperitoneal bleomycin (0.035 U/g) or PBS, and were allowed to rest 1 week before being killed. Their lungs were inflated with PBS, fixed in formalin, paraffin-embedded, and sectioned. A certified veterinary pathologist blindly scored each slide for inflammation and fibrosis. Lungs were homogenized to obtain RNA and protein for the real-time RT-PCR analysis of connective tissue growth factor (CTGF) and collagen I, and for Western blotting to detect phospho-Smad2, or total Smad2/3, respectively. In response to bleomycin treatment, measures of fibrosis and inflammation, along with CTGF and collagen I mRNA concentrations, were increased in TSP-1-deficient mice compared with WT mice. Notably, Smad 2/3 signaling was of equal strength in WT and TSP-1 knockout mice treated with bleomycin, suggesting that TSP-1 is not required for the activation of TGF-ß. These results demonstrate that TSP-1 deficiency does not protect mice from systemic bleomycin challenge, and that TSP-1 deficiency is associated with increased expression of lung collagen and CTGF.


Subject(s)
Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/prevention & control , Thrombospondin 1/deficiency , Animals , Bleomycin , Collagen/metabolism , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Gene Expression Regulation/drug effects , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Phosphorylation/drug effects , Pneumonia/complications , Pneumonia/metabolism , Pneumonia/pathology , Pulmonary Fibrosis/complications , Pulmonary Fibrosis/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Smad2 Protein/metabolism , Thrombospondin 1/metabolism , Transforming Growth Factor beta/metabolism
7.
PLoS Pathog ; 5(4): e1000361, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19343209

ABSTRACT

Legionella pneumophila (L. pneumophila), the causative agent of a severe form of pneumonia called Legionnaires' disease, replicates in human monocytes and macrophages. Most inbred mouse strains are restrictive to L. pneumophila infection except for the A/J, Nlrc4(-/-) (Ipaf(-/-)), and caspase-1(-/-) derived macrophages. Particularly, caspase-1 activation is detected during L. pneumophila infection of murine macrophages while absent in human cells. Recent in vitro experiments demonstrate that caspase-7 is cleaved by caspase-1. However, the biological role for caspase-7 activation downstream of caspase-1 is not known. Furthermore, whether this reaction is pertinent to the apoptosis or to the inflammation pathway or whether it mediates a yet unidentified effect is unclear. Using the intracellular pathogen L. pneumophila, we show that, upon infection of murine macrophages, caspase-7 was activated downstream of the Nlrc4 inflammasome and required caspase-1 activation. Such activation of caspase-7 was mediated by flagellin and required a functional Naip5. Remarkably, mice lacking caspase-7 and its macrophages allowed substantial L. pneumophila replication. Permissiveness of caspase-7(-/-) macrophages to the intracellular pathogen was due to defective delivery of the organism to the lysosome and to delayed cell death during early stages of infection. These results reveal a new mechanism for caspase-7 activation downstream of the Nlrc4 inflammasome and present a novel biological role for caspase-7 in host defense against an intracellular bacterium.


Subject(s)
Apoptosis Regulatory Proteins/immunology , Calcium-Binding Proteins/immunology , Caspase 7/metabolism , Legionella pneumophila/growth & development , Legionella pneumophila/immunology , Legionnaires' Disease/immunology , Macrophages/immunology , Signal Transduction/immunology , Animals , Apoptosis , Caspase 1/metabolism , Cells, Cultured , Enzyme Activation , Flagellin/metabolism , Humans , Interleukin-18/immunology , Interleukin-1beta/immunology , Legionnaires' Disease/microbiology , Lysosomes/immunology , Macrophages/enzymology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Neuronal Apoptosis-Inhibitory Protein/immunology
8.
Am J Respir Crit Care Med ; 176(1): 78-89, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17431224

ABSTRACT

RATIONALE: An increase in the number of mononuclear phagocytes in lung biopsies from patients with idiopathic pulmonary fibrosis (IPF) worsens prognosis. Chemokines that recruit mononuclear phagocytes, such as CC chemokine ligand 2 (CCL2), are elevated in bronchoalveolar lavage (BAL) fluid (BALF) from patients with IPF. However, little attention is given to the role of the mononuclear phagocyte survival and recruitment factor, macrophage colony-stimulating factor (M-CSF), in pulmonary fibrosis. OBJECTIVES: To investigate the role of mononuclear phagocytes and M-CSF in pulmonary fibrosis. METHODS: Wild-type, M-CSF-/-, or CCL2-/- mice received intraperitoneal bleomycin. Lung inflammation and fibrosis were measured by immunohistochemistry, ELISA, collagen assay, BAL differentials, real-time polymerase chain reaction, and Western blot analysis. Human and mouse macrophages were stimulated with M-CSF for CCL2 expression. BALF from patients with IPF was examined for M-CSF and CCL2. MEASUREMENTS AND MAIN RESULTS: M-CSF-/- and CCL2-/- mice had less lung fibrosis, mononuclear phagocyte recruitment, collagen deposition, and connective tissue growth factor (CTGF) expression after bleomycin administration than wild-type littermates. Human and mouse macrophages stimulated with M-CSF had increased CCL2 production, and intratracheal administration of M-CSF in mice induced CCL2 production in BALF. Finally, BALF from patients with IPF contained significantly more M-CSF and CCL2 than BALF from normal volunteers. Elevated levels of M-CSF were associated with elevated CCL2 in BALF and the diagnosis of IPF. CONCLUSIONS: These data suggest that M-CSF contributes to the pathogenesis of pulmonary fibrosis in mice and in patients with IPF through the involvement of mononuclear phagocytes and CCL2 production.


Subject(s)
Chemokine CCL2/immunology , Macrophage Colony-Stimulating Factor/immunology , Macrophages, Alveolar/immunology , Pulmonary Fibrosis/immunology , Adult , Animals , Bleomycin , Bronchoalveolar Lavage Fluid/cytology , Case-Control Studies , Cells, Cultured , Chemokine CCL2/metabolism , Disease Models, Animal , Humans , Mice , Mice, Knockout , Phagocytes/metabolism , Pulmonary Fibrosis/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...