Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Am J Physiol Lung Cell Mol Physiol ; 295(2): L314-25, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18556802

ABSTRACT

Activation of the Fas/Fas ligand (FasL) system in the lungs results in a form of injury characterized by alveolar epithelial apoptosis and neutrophilic inflammation. Studies in vitro show that Fas activation induces apoptosis in alveolar epithelial cells and cytokine production in alveolar macrophages. The main goal of this study was to determine the contribution of alveolar macrophages to Fas-induced lung inflammation in mice, by depleting alveolar macrophages using clodronate-containing liposomes. Liposomes containing clodronate or PBS were instilled by intratracheal instillation. After 24 h, the mice received intratracheal instillations of the Fas-activating monoclonal antibody Jo2 or an isotype control antibody and were studied 18 h later. The Jo2 MAb induced increases in bronchoalveolar lavage fluid (BALF) total neutrophils, lung caspase-3 activity, and BALF total protein and worsened histological lung injury in the macrophage-depleted mice. Studies in vitro showed that Fas activation induced the release of the cytokine KC in a mouse lung epithelial cell line, MLE-12. These results suggest that the lung inflammatory response to Fas activation is not primarily dependent on resident alveolar macrophages and may instead depend on cytokine release by alveolar epithelial cells.


Subject(s)
Apoptosis , Fas Ligand Protein/metabolism , Macrophages, Alveolar/metabolism , Respiratory Distress Syndrome/metabolism , Respiratory Mucosa/metabolism , fas Receptor/metabolism , Animals , Antibodies, Monoclonal/toxicity , Apoptosis/drug effects , Apoptosis/immunology , Bone Density Conservation Agents/pharmacology , Bronchoalveolar Lavage Fluid/immunology , Cell Line , Chemokine CXCL1/biosynthesis , Chemokine CXCL1/immunology , Clodronic Acid/pharmacology , Fas Ligand Protein/immunology , Inflammation/chemically induced , Inflammation/immunology , Inflammation/metabolism , Macrophages, Alveolar/immunology , Macrophages, Alveolar/pathology , Male , Mice , Respiratory Distress Syndrome/chemically induced , Respiratory Distress Syndrome/immunology , Respiratory Mucosa/immunology , Respiratory Mucosa/pathology , Time Factors , fas Receptor/immunology
2.
Infect Immun ; 69(9): 5768-76, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11500454

ABSTRACT

The Fas/Fas ligand (FasL) system has been implicated in alveolar epithelial cell apoptosis during pulmonary fibrosis and acute respiratory distress syndrome. However, Fas ligation can also lead to cell activation and cytokine production. The goal of this study was to determine the role of the Fas/FasL system in host defenses against Escherichia coli, Staphylococcus aureus, and Streptococcus pneumoniae. We administered bacteria by aerosolization into the lungs of Fas-deficient (lpr) mice and wild-type (C57BL/6) mice and measured bacterial clearance at 6 and 12 h. One hour prior to euthanasia, the mice received an intraperitoneal injection of human serum albumin (HSA) for alveolar permeability determinations. At all times after bacterial challenges, the lungs of the lpr mice contained similar or lower numbers of bacteria than those of the C57BL/6 mice. Alveolar permeability changes, as determined by bronchoalveolar lavage fluid HSA concentrations, were less severe in the lpr mice 6 h after the challenges. In response to E. coli, the lpr mice had significantly more polymorphonuclear leukocytes (PMN) and macrophage inflammatory protein 2 in the lungs, whereas histopathologic changes were less severe. In contrast, in response to the gram-positive cocci, the lpr animals had similar or lower numbers of PMN. We conclude that the Fas/FasL system contributes to the development of permeability changes and tissue injury during-gram negative bacterial pneumonia. The Fas/FasL system did not have a major role in the clearance of aerosolized bacteria from the lungs at the bacterial doses tested.


Subject(s)
Lung/immunology , Lung/microbiology , Membrane Glycoproteins/metabolism , Pneumonia, Bacterial/immunology , fas Receptor/metabolism , Animals , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Escherichia coli/isolation & purification , Escherichia coli/pathogenicity , Fas Ligand Protein , Male , Mice , Mice, Inbred C57BL , Pneumonia, Bacterial/microbiology , Pneumonia, Pneumococcal/immunology , Pneumonia, Pneumococcal/microbiology , Pneumonia, Staphylococcal/immunology , Pneumonia, Staphylococcal/microbiology , Serum Albumin/metabolism , Staphylococcus aureus/isolation & purification , Staphylococcus aureus/pathogenicity , Streptococcus pneumoniae/isolation & purification , Streptococcus pneumoniae/pathogenicity
3.
Am J Physiol Lung Cell Mol Physiol ; 281(2): L328-35, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11435207

ABSTRACT

This study investigated whether recombinant human soluble Fas ligand (rh-sFasL) induces apoptosis of primary type II pneumocytes in vitro and lung injury in vivo. Type II cells isolated from normal rabbit lung expressed Fas on their surface and became apoptotic after an 18-h incubation with rh-sFasL. Fas expression in normal rabbit lungs was localized by immunohistochemistry to alveolar and airway epithelia and alveolar macrophages. The administration of 10 microg of rh-sFasL into the right lungs of rabbits resulted 24 h later in both significantly more bronchoalveolar lavage fluid total protein and significantly more tissue changes compared with those in the left lungs, which received rh-sFasL plus Fas:Ig (a fusion protein that binds and blocks sFasL). Tissue changes included thickening of the alveolar walls, neutrophilic infiltrates, apoptotic (terminal deoxynucleotidyltransferase-mediated dUTP nick end labeling-positive) cells in the alveolar walls, and increased expression of interleukin-8 by alveolar macrophages (as determined by immunohistochemistry). We conclude that the alveolar epithelium of normal rabbits expresses Fas and that sFasL induces lung injury and inflammation in rabbits.


Subject(s)
Apoptosis , Lung Diseases/chemically induced , Membrane Glycoproteins/pharmacology , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/physiology , Animals , Cells, Cultured , Epithelial Cells/drug effects , Epithelial Cells/physiology , Fas Ligand Protein , Female , Humans , Lung/metabolism , Pulmonary Alveoli/cytology , Pulmonary Alveoli/metabolism , Rabbits , Recombinant Proteins , fas Receptor/metabolism
4.
Am J Respir Crit Care Med ; 163(1): 234-43, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11208651

ABSTRACT

The major goal of this study was to investigate the mechanisms that link the host response to a local infection in the peritoneal cavity with the development of sepsis and lung injury. Rabbits were infected by intraperitoneal inoculation of fibrin clots containing Escherichia coli at 10(8), 10(9), or 10(10) cfu/clot. Physiologic, bacteriologic, and inflammatory responses were monitored, and the lungs were examined postmortem. At a dose of 10(8) cfu/clot the animals had resolving infection, and a dose of 10(9) cfu/clot resulted in persistent infection at 24 h, with minimal systemic manifestations. In contrast, inoculation of 10(10) cfu/clot resulted in rapidly lethal local infection, with septic shock and lung injury. The onset of septic shock was associated with a paradoxical lack of identifiable polymorphonuclear leukocytes (PMN; neutrophils) in the peritoneal cavity. The absence of PMN in the peritoneum was due in part to lysis of intraperitoneal PMN, because the peritoneal fluids contained free myeloperoxidase and induced rapid death of normal rabbit PMN in vitro. Although most animals became bacteremic, only those with a severe systemic inflammation response developed lung injury. These data show that control of an infection in the first compartment in which bacteria enter the host is a critical determinant of the systemic response. Above a threshold dose of bacteria, failure of the local neutrophil response is a key mechanism associated with deleterious systemic responses. Bacteremia alone is not sufficient to cause lung injury. Lung injury occurs only in the setting of a severe systemic inflammatory response and an inadequate leukocyte response at the primary site of infection.


Subject(s)
Neutrophils/immunology , Peritonitis/immunology , Respiratory Distress Syndrome/immunology , Shock, Septic/immunology , Animals , Body Fluids/cytology , Female , Peritonitis/complications , Rabbits , Respiratory Distress Syndrome/complications , Shock, Septic/complications
5.
Am J Pathol ; 158(1): 153-61, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11141488

ABSTRACT

Activation of the Fas/FasL system induces apoptosis of susceptible cells, but may also lead to nuclear factor kappaB activation. Our goal was to determine whether local Fas activation produces acute lung injury by inducing alveolar epithelial cell apoptosis and by generating local inflammatory responses. Normal mice (C57BL/6) and mice deficient in Fas (lpr) were treated by intranasal instillation of the Fas-activating monoclonal antibody (mAb) Jo2 or an irrelevant control mAb, and studied 6 or 24 hours later using bronchoalveolar lavage (BAL), histopathology, DNA nick-end-labeling assays, and electron microscopy. Normal mice treated with mAb Jo2 had significant increases in BAL protein at 6 hours, and BAL neutrophils at 24 hours, as compared to lpr mice and to mice treated with the irrelevant mAb. Neutrophil recruitment was preceded by increased mRNA expression for tumor necrosis factor-alpha, macrophage inflammatory protein-1alpha, macrophage inflammatory protein-2, macrophage chemotactic protein-1, and interleukin-6, but not interferon-gamma, transforming growth factor-ss, RANTES, eotaxin, or IP-10. Lung sections from Jo2-treated normal mice showed neutrophilic infiltrates, alveolar septal thickening, hemorrhage, and terminal dUTP nick-end-labeling-positive cells in the alveolar septae and airspaces. Type II pneumocyte apoptosis was confirmed by electron microscopy. Fas activation in vivo results in acute alveolar epithelial injury and lung inflammation, and may be important in the pathogenesis of acute lung injury.


Subject(s)
Apoptosis , Epithelial Cells/metabolism , Pulmonary Alveoli/metabolism , fas Receptor/metabolism , Acute Disease , Animals , Antibodies, Monoclonal/pharmacology , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Cytokines/genetics , Cytokines/metabolism , Epithelial Cells/cytology , Epithelial Cells/drug effects , Fas Ligand Protein , Gene Expression/drug effects , Leukocyte Count , Lung/drug effects , Lung/pathology , Lung/ultrastructure , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Microscopy, Electron , Neutrophils/cytology , Pneumonia/metabolism , Pneumonia/pathology , Pneumonia/prevention & control , Pulmonary Alveoli/cytology , Pulmonary Alveoli/drug effects , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , fas Receptor/genetics
6.
Methods Mol Biol ; 138: 319-30, 2000.
Article in English | MEDLINE | ID: mdl-10840772

ABSTRACT

To study the mechanisms that link sepsis with ARDS, many animal models have been developed. In this chapter, a rabbit model of sepsis secondary to an intrapulmonary or intraabdominal infection has been described. One advantage of the rabbit model of sepsis is that this species produces the C-X-C chemokine, IL-8. In contrast, rodents, which are often used in studies of sepsis and ARDS, lack this important chemokine. A second advantage is the rabbit's size. This species is large enough so that the measurement of physiological parameters (e.g., mean arterial pressure, heart rate, etc.) is not difficult, but they are not so large that they require large quantities of precious reagents (e.g., recombinant proteins and MAbs). A disadvantage of the rabbit model is that there are fewer reagents (e.g., recombinant cytokines and MAbs) available for the study of inflammation in rabbits when compared to mice.


Subject(s)
Disease Models, Animal , Peritonitis , Pneumonia , Respiratory Distress Syndrome , Animals , Humans , Infant, Newborn , Mice , Rabbits , Sepsis
7.
J Immunol ; 164(10): 5439-45, 2000 May 15.
Article in English | MEDLINE | ID: mdl-10799910

ABSTRACT

CD14, a pattern recognition receptor found on myeloid cells, is a critical component of the innate immune system that mediates local and systemic host responses to Gram-negative and Gram-positive bacterial products. Previous studies in normal animals have tested the effect of CD14 blockade on the systemic response to i.v. LPS. The goals of the study were to determine whether CD14 blockade protected against the deleterious systemic response associated with Escherichia coli pneumonia and to determine whether this strategy affected the pulmonary response to tissue infection. Rabbits were pretreated with either anti-CD14 mAb or isotype control mAb at 2.5 mg/kg. E. coli (1 x 109 CFU) was inoculated into the lungs, and the animals were observed for either 4 or 24 h. The blockade of CD14 improved the mean arterial blood pressure (p = 0.001) and decreased the i.v. fluid requirements (p = 0.01). Although this therapy protected the vascular compartment, rabbits treated with anti-CD14 mAb had increased bacterial burdens in the bronchoalveolar lavage fluid recovered from the instilled lung (p = 0.005) and widened alveolar-arterial oxygen difference. Blockade of CD14 prevents the deleterious systemic responses that occur in sepsis; however, other measures are necessary to control bacterial proliferation at the primary site of infection.


Subject(s)
Escherichia coli Infections/immunology , Lipopolysaccharide Receptors/immunology , Pneumonia, Bacterial/immunology , Sepsis/immunology , Animals , Antibodies, Monoclonal/pharmacology , Blood Pressure/immunology , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/immunology , Bronchoalveolar Lavage Fluid/microbiology , Chemokine CCL2/analysis , Chemotactic Factors/analysis , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Escherichia coli Infections/physiopathology , Growth Substances/analysis , Inflammation/immunology , Interleukin-8/analysis , Lung/immunology , Lung/microbiology , Lung/pathology , Pneumonia, Bacterial/microbiology , Pneumonia, Bacterial/pathology , Pneumonia, Bacterial/physiopathology , Pulmonary Gas Exchange/immunology , Rabbits , Sepsis/microbiology , Sepsis/pathology , Sepsis/physiopathology , Tumor Necrosis Factor-alpha/analysis
8.
Crit Care Med ; 28(1): 1-7, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10667491

ABSTRACT

OBJECTIVE: To determine whether bronchoalveolar lavage fluid (BALF) from patients either at risk for the acute respiratory distress syndrome (ARDS) or with sustained ARDS modulates neutrophil apoptosis; to measure the BALF concentrations of the apoptosis inhibitors granulocyte colony-stimulating factor (G-CSF) and granulocyte/macrophage colony-stimulating factor (GM-CSF) before and after the onset of ARDS; and to determine whether the BALF concentrations of G-CSF and/or GM-CSF are associated with clinical outcome. DESIGN: Prospective cohort study. SETTING: Tertiary university hospital. PATIENTS: Twenty patients at risk for ARDS and 45 patients with established ARDS. INTERVENTIONS: Patients at risk for ARDS underwent bronchoalveolar lavage within 24 hrs of being identified, then again 72 hrs later. Patients with ARDS underwent bronchoalveolar lavage within 24 hrs of meeting ARDS criteria, then again on days 3, 7, and 14 of the disease. MEASUREMENTS AND MAIN RESULTS: Normal peripheral blood neutrophil were incubated overnight in BALF from normal volunteers, from patients at risk for ARDS, or from patients with ARDS. neutrophil apoptosis was determined by flow cytometric analysis of annexin V binding. G-CSF and GM-CSF were measured in BALF by immunoassays. Compared with normal BALF, BALF from patients on days 1 and 3 of ARDS inhibited neutrophil apoptosis, but BALF from patients at later stages of ARDS, or from patients at risk for ARDS, did not. The BALF concentrations of both G-CSF and GM-CSF were elevated early in ARDS and decreased toward later stages. Patients who lived had significantly higher concentrations of GM-CSF in the BALF than those who died. CONCLUSIONS: We conclude that the antiapoptotic effect of ARDS BALF on normal neutrophil is highest during early ARDS, and decreases during late ARDS. G-CSF and GM-CSF are present in BALF from patients with ARDS, and their concentrations parallel the antiapoptotic effect of ARDS BALF. These data support the concept that the life-span of neutrophil in the air spaces is modulated during acute inflammation. GM-CSF in the air spaces is associated with improved survival in patients with ARDS.


Subject(s)
Apoptosis , Bronchoalveolar Lavage Fluid/immunology , Granulocyte Colony-Stimulating Factor/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Neutrophils/cytology , Respiratory Distress Syndrome/pathology , Adolescent , Adult , Aged , Bronchoalveolar Lavage Fluid/cytology , Cohort Studies , Female , Flow Cytometry , Humans , Immunoassay , Inflammation , Male , Middle Aged , Prospective Studies , Respiratory Distress Syndrome/immunology
9.
J Immunol ; 163(4): 2217-25, 1999 Aug 15.
Article in English | MEDLINE | ID: mdl-10438964

ABSTRACT

The goals of this study were to determine whether the Fas-dependent apoptosis pathway is active in the lungs of patients with the acute respiratory distress syndrome (ARDS), and whether this pathway can contribute to lung epithelial injury. We found that soluble Fas ligand (sFasL) is present in bronchoalveolar lavage (BAL) fluid of patients before and after the onset of ARDS. The BAL concentration of sFasL at the onset of ARDS was significantly higher in patients who died. BAL from patients with ARDS induced apoptosis of distal lung epithelial cells, which express Fas, and this effect was inhibited by blocking the Fas/FasL system using three different strategies: anti-FasL mAb, anti-Fas mAb, and a Fas-Ig fusion protein. In contrast, BAL from patients at risk for ARDS had no effect on distal lung epithelial cell apoptosis. These data indicate that sFasL is released in the airspaces of patients with acute lung injury and suggest that activation of the Fas/FasL system contributes to the severe epithelial damage that occurs in ARDS. These data provide the first evidence that FasL can be released as a biologically active, death-inducing mediator capable of inducing apoptosis of cells of the distal pulmonary epithelium during acute lung injury.


Subject(s)
Apoptosis/immunology , Epithelial Cells/cytology , Membrane Glycoproteins/physiology , Respiratory Distress Syndrome/immunology , Respiratory Distress Syndrome/pathology , fas Receptor/physiology , Bronchoalveolar Lavage Fluid/immunology , Cell Death/immunology , Cell-Free System/immunology , Cells, Cultured , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fas Ligand Protein , Humans , Ligands , Lung/immunology , Lung/metabolism , Membrane Glycoproteins/metabolism , Pulmonary Alveoli/immunology , Pulmonary Alveoli/metabolism , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/mortality , Risk Factors , Solubility , Survival Analysis
10.
Bone Marrow Transplant ; 21(11): 1125-30, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9645575

ABSTRACT

We investigated an association between pulmonary function testing (PFT) before bone marrow transplantation and the development of severe veno-occlusive disease (VOD) of the liver. We previously noted that reductions in diffusing capacity of the lung for carbon monoxide (corrected for hemoglobin) (D(L)COc) were associated with mortality after transplantation, but this was not caused by respiratory failure. We performed a case-series review of prospectively collected data from 307 marrow recipients who underwent PFT within 2 weeks of transplantation. Of these, 170 (56%) developed VOD; 39 (13%) mild, 81 (26%) moderate, and 50 (16%) severe or fatal. Both total lung capacity (TLC) and D(L)COc were associated with severe VOD in univariate analysis (P = 0.006 for each). However, D(L)COc entered logistic regression models that contained variables for all known risk factors for severe VOD, while TLC did not contribute additional predictive information. The odds ratio (OR) associated with a D(L)COc below the lower limits of normal (70% of predicted) was 2.4 (95 % CI, 1.0 to 5.4; P = 0.04). We conclude that reduced diffusion capacity of the lung measured before marrow transplantation is an independent risk for severe hepatic VOD. We speculate that the decreased D(l)COc indicates pre-existing systemic endothelial cell damage and a susceptibility to severe hepatic injury from chemotherapy.


Subject(s)
Bone Marrow Transplantation/adverse effects , Hepatic Veno-Occlusive Disease/etiology , Respiratory Function Tests , Adolescent , Adult , Child , Child, Preschool , Endothelium, Vascular/injuries , Humans , Middle Aged , Prospective Studies , Pulmonary Diffusing Capacity , Risk Factors , Total Lung Capacity , Transplantation Conditioning/adverse effects
11.
Am J Respir Crit Care Med ; 156(6): 1969-77, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9412582

ABSTRACT

Little is known about neutrophil (PMN) apoptosis in the acute respiratory distress syndrome (ARDS). We uses morphologic criteria to count apoptotic PMN in bronchoalveolar lavage fluid (BAL) of 35 patients on Days 1, 3, 7, 14, and 21 of ARDS and 13 patients on Days 1 and 3 of risk for ARDS. We found that the proportion of apoptotic PMN in BAL was low throughout the course of ARDS. There was no significant difference between the percentage of apoptotic PMN in patients at risk and patients with established ARDS or between patients who lived (2.4%) and patients who died (1.8%). When normal human PMN were incubated in ARDS BAL, a significantly lower proportion became apoptotic (50 +/- 4%), as compared with PMN incubated in lavage fluid from normal volunteers (76 +/- 7%, p < 0.05). This antiapoptotic effect of ARDS BAL was blocked by immunodepleting BAL of G-CSF and GM-CSF. We conclude that the proportion of apoptotic PMN recovered from the lungs of patients with ARDS is low throughout the course of ARD S. Furthermore, BAL from patients with ARDS prolongs survival of normal human PMN in vitro, and this effect is partially mediated by G-CSF and GM-CSF.


Subject(s)
Apoptosis , Neutrophils/pathology , Respiratory Distress Syndrome/pathology , Adult , Aged , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Cell Survival , Female , Granulocyte Colony-Stimulating Factor/analysis , Granulocyte-Macrophage Colony-Stimulating Factor/analysis , Humans , In Vitro Techniques , Interferon-gamma/analysis , Interleukin-6/analysis , Leukocyte Count , Male , Middle Aged , Respiratory Distress Syndrome/metabolism , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...