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.
J Appl Physiol (1985) ; 111(1): 177-84, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21512145

ABSTRACT

Elevated soluble tumor necrosis factor-α receptor (sTNFR) levels in bronchoalveolar lavage fluid (BALF) are associated with poor patient outcome in acute lung injury (ALI). The mechanisms underlying these increases are unknown, but it is possible that pulmonary inflammation and increased alveolar epithelial permeability may individually contribute. We investigated mechanisms of elevated BALF sTNFRs in two in vivo mouse models of ALI. Anesthetized mice were challenged with intratracheal lipopolysaccharide or subjected to injurious mechanical ventilation. Lipopolysaccharide instillation produced acute intra-alveolar inflammation, but minimal alveolar epithelial permeability changes, with increased BALF sTNFR p75, but not p55. Increased p75 levels were markedly attenuated by alveolar macrophage depletion. In contrast, injurious ventilation induced substantial alveolar epithelial permeability, with increased BALF p75 and p55, which strongly correlated with total protein. BALF sTNFRs were not increased in isolated buffer-perfused lungs (devoid of circulating sTNFRs) subjected to injurious ventilation. These results suggest that lipopolysaccharide-induced intra-alveolar inflammation upregulates alveolar macrophage-mediated production of sTNFR p75, whereas enhanced alveolar epithelial permeability following mechanical ventilation leads to increased BALF p75 and p55 via plasma leakage. These data provide new insights into differential regulation of intra-alveolar sTNFR levels during ALI and may suggest sTNFRs as potential markers for evaluating the pathophysiology of ALI.


Subject(s)
Acute Lung Injury/immunology , Pneumonia/immunology , Pulmonary Alveoli/immunology , Receptors, Tumor Necrosis Factor, Type II/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Ventilator-Induced Lung Injury/immunology , Acute Lung Injury/chemically induced , Animals , Bronchoalveolar Lavage Fluid/immunology , Capillary Permeability , Disease Models, Animal , Epithelial Cells/immunology , Lipopolysaccharides , Macrophages, Alveolar/immunology , Male , Mice , Mice, Inbred C57BL , Pneumonia/etiology , Pulmonary Alveoli/blood supply , Respiration, Artificial/adverse effects , Time Factors , Up-Regulation , Ventilator-Induced Lung Injury/etiology
3.
Am J Physiol Lung Cell Mol Physiol ; 300(5): L781-9, 2011 May.
Article in English | MEDLINE | ID: mdl-21378027

ABSTRACT

TNF plays a crucial role in the pathogenesis of acute lung injury. However, the expression profile of its two receptors, p55 and p75, on pulmonary endothelium and their influence on TNF signaling during lung microvascular inflammation remain uncertain. Using flow cytometry, we characterized the expression profile of TNF receptors on the surface of freshly harvested pulmonary endothelial cells (PECs) from mice and found expression of both receptors with dominance of p55. To investigate the impact of stimulating individual TNF receptors, we treated wild-type and TNF receptor knockout mice with intravenous TNF and determined surface expression of adhesion molecules (E-selectin, VCAM-1, ICAM-1) on PECs by flow cytometry. TNF-induced upregulation of all adhesion molecules was substantially attenuated by absence of p55, whereas lack of p75 had a similar but smaller effect that varied between adhesion molecules. Selective blockade of individual TNF receptors by specific antibodies in wild-type primary PEC culture confirmed that the in vivo findings were due to direct effects of TNF receptor inhibition on endothelium and not other cells (e.g., circulating leukocytes). Finally, we found that PEC surface expression of p55 dramatically decreased in the early stages of endotoxemia following intravenous LPS, while no change in p75 expression was detected. These data demonstrate a crucial in vivo role of p55 and an auxiliary role of p75 in TNF-mediated adhesion molecule upregulation on PECs. It is possible that the importance of the individual receptors varies at different stages of pulmonary microvascular inflammation following changes in their relative expression.


Subject(s)
Endothelial Cells/metabolism , Lung/physiology , Receptors, Tumor Necrosis Factor, Type II/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction/physiology , Tumor Necrosis Factor Decoy Receptors/metabolism , Animals , E-Selectin/metabolism , Endotoxemia/physiopathology , Intercellular Adhesion Molecule-1/biosynthesis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumonia/physiopathology , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation , Vascular Cell Adhesion Molecule-1/biosynthesis
4.
PLoS One ; 5(7): e11565, 2010 Jul 13.
Article in English | MEDLINE | ID: mdl-20644637

ABSTRACT

BACKGROUND: Pulmonary inflammation is a major contributor to morbidity in a variety of respiratory disorders, but treatment options are limited. Here we investigate the efficacy, safety and mechanism of action of low dose inhaled carbon monoxide (CO) using a mouse model of lipopolysaccharide (LPS)-induced pulmonary inflammation. METHODOLOGY: Mice were exposed to 0-500 ppm inhaled CO for periods of up to 24 hours prior to and following intratracheal instillation of 10 ng LPS. Animals were sacrificed and assessed for intraalveolar neutrophil influx and cytokine levels, flow cytometric determination of neutrophil number and activation in blood, lung and lavage fluid samples, or neutrophil mobilisation from bone marrow. PRINCIPAL FINDINGS: When administered for 24 hours both before and after LPS, inhaled CO of 100 ppm or more reduced intraalveolar neutrophil infiltration by 40-50%, although doses above 100 ppm were associated with either high carboxyhemoglobin, weight loss or reduced physical activity. This anti-inflammatory effect of CO did not require pre-exposure before induction of injury. 100 ppm CO exposure attenuated neutrophil sequestration within the pulmonary vasculature as well as LPS-induced neutrophilia at 6 hours after LPS, likely due to abrogation of neutrophil mobilisation from bone marrow. In contrast to such apparently beneficial effects, 100 ppm inhaled CO induced an increase in pulmonary barrier permeability as determined by lavage fluid protein content and translocation of labelled albumin from blood to the alveolar space. CONCLUSIONS: Overall, these data confirm some protective role for inhaled CO during pulmonary inflammation, although this required a dose that produced carboxyhemoglobin values close to potentially toxic levels for humans, and increased lung permeability.


Subject(s)
Carbon Monoxide/adverse effects , Carbon Monoxide/therapeutic use , Lung/drug effects , Lung/metabolism , Pneumonia/drug therapy , Administration, Inhalation , Animals , Carbon Monoxide/administration & dosage , Flow Cytometry , Lipopolysaccharides/toxicity , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Neutrophil Infiltration/drug effects , Pneumonia/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL
...