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










Publication year range
1.
Eur Respir J ; 39(1): 133-40, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21737551

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is characterised by myofibroblast proliferation leading to architectural destruction. Neither the origin nor the continued proliferation of myofibroblasts is well understood. Explanted human IPF lungs were stained by immunohistochemistry for calretinin, a marker of pleural mesothelial cells (PMCs). Chronic obstructive pulmonary disease (COPD) and cystic fibrosis (CF) lungs acted as controls. The number of PMCs per 100 nucleated cells and per photomicrograph was estimated along with the Ashcroft score of fibrosis. Mouse PMCs expressing green fluorescent protein (GFP) or labelled with nanoparticles were injected into the pleural space of mice given intranasal transforming growth factor (TGF)-ß1. Mouse lungs were lavaged and examined for the presence of GFP, smooth muscle α-actin (α-SMA) and calretinin. Calretinin-positive PMCs were found throughout IPF lungs, but not in COPD or CF lungs. The number of PMCs correlated with the Ashcroft score. In mice, nanoparticle-laden PMCs were recoverable by bronchoalveolar lavage, depending on the TGF-ß1 dose. Fluorescent staining showed α-SMA expression in GFP-expressing PMCs, with co-localisation of GFP and α-SMA. PMCs can traffic through the lung and show myofibroblast phenotypic markers. PMCs are present in IPF lungs, and their number correlates with IPF severity. Since IPF presumably begins subpleurally, PMCs could play a pathogenetic role via mesothelial-mesenchymal transition.


Subject(s)
Epithelium/pathology , Idiopathic Pulmonary Fibrosis/physiopathology , Lung/metabolism , S100 Calcium Binding Protein G/blood , Adolescent , Adult , Aged , Animals , Calbindin 2 , Cell Nucleus/metabolism , Child , Cystic Fibrosis/metabolism , Epithelial-Mesenchymal Transition , Female , GPI-Linked Proteins/blood , Humans , Immunohistochemistry/methods , Male , Mesothelin , Mice , Mice, Inbred C57BL , Middle Aged , Myofibroblasts/cytology , Pleura/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism
2.
Eur Respir J ; 29(4): 761-9, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17251235

ABSTRACT

Talc remains the most effective sclerosing agent for pleurodesis. However, its mechanism of action in resolving pleural malignant disease remains unclear. The present study evaluated the angiogenic balance in the pleural space in patients with malignant pleural effusions (MPE) following talc insufflation. Patient pleural fluid samples were collected both before and after talc insufflation. The ability of pleural mesothelial cells (PMC) and malignant mesothelioma cells (MMC) to produce endostatin in vitro was compared. The biological effects of pleural fluids and conditioned media from talc-activated PMC on endothelial cells were evaluated by performing proliferation, invasion, tube formation and apoptosis assays. Pleural fluids from patients with MPE who received thoracoscopic talc insufflation contained significantly higher levels of endostatin (median 16.75 ng.mL(-1)) compared with pre-talc instillation (1.06 ng.mL(-1)). Talc-activated PMC released significantly greater amounts of endostatin (mean+/-SEM 1052.39+/-38.66 pg.mL(-1)) when compared with a MMC line (134.73+/-8.72 pg.mL(-1)). In conclusion, talc alters the angiogenic balance in the pleural space from a biologically active and angiogenic environment to an angiostatic milieu. Functional improvement following talc poudrage in patients with malignant pleural effusions may, in part, reflect these alterations in the pleural space.


Subject(s)
Endostatins/metabolism , Neovascularization, Physiologic/drug effects , Pleurodesis/adverse effects , Talc/adverse effects , Talc/therapeutic use , Adult , Aged , Aged, 80 and over , Cell Proliferation , Epithelium/pathology , Female , Humans , Male , Middle Aged , Particle Size , Pleural Effusion, Malignant/therapy , Pleurodesis/methods
3.
J Appl Physiol (1985) ; 99(2): 677-82, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15817724

ABSTRACT

Active, nonanesthetized, tracheotomized rabbits were subjected to continuous positive airway pressure (CPAP) for 4 days to determine the effects of chronic mechanical strain on lung and airway function. Rabbits were maintained for 4 days at a CPAP of 6 cmH(2)O (high CPAP), at a CPAP of 0 cmH(2)O (low CPAP), or without tracheostomy (no CPAP). After treatment with CPAP, changes in respiratory resistance in response to increasing concentrations of inhaled ACh were measured during mechanical ventilation to evaluate respiratory system responsiveness in vivo. Intraparenchymal bronchial segments were isolated from the lungs of all animals to evaluate airway smooth muscle responsiveness and bronchial compliance in vitro. Rabbits maintained for 4 days at high CPAP demonstrated significantly lower responsiveness to ACh compared with rabbits that were maintained at low CPAP or with no CPAP. Airways isolated from the lungs of animals subjected to the chronic application of high CPAP were also less responsive to ACh in vitro than the airways isolated from animals subjected to low CPAP or no CPAP. The persistence of the decreased responsiveness in the excised airway tissues suggests that the decreased respiratory system responsiveness observed in vivo results primarily from direct effects on the airways. The results demonstrate that the application of prolonged mechanical strain in vivo can reduce airway reactivity.


Subject(s)
Lung/physiology , Positive-Pressure Respiration/methods , Pulmonary Ventilation/physiology , Tidal Volume/physiology , Acetylcholine , Adaptation, Physiological/physiology , Animals , Dose-Response Relationship, Drug , Lung/drug effects , Pulmonary Ventilation/drug effects , Rabbits , Respiratory Function Tests , Tidal Volume/drug effects
4.
Lung ; 181(2): 57-66, 2003.
Article in English | MEDLINE | ID: mdl-12953144

ABSTRACT

Patients with pulmonary tuberculosis develop pleural effusions with a high protein content. Pleural mesothelial adherens junctions promote mesothelial cell-cell adhesion and contribute to pleural integrity. In the present study we have investigated the effect of mycobacterium (BCG) on mesothelial cell adherens junction proteins and pleural permeability. BCG enhanced pleural mesothelial cell (PMC) release of vascular endothelial growth factor (VEGF), and decreased electrical resistance across the PMC monolayer. Neutralizing antibodies to VEGF significantly restored the drop in PMC electrical resistance caused by BCG. BCG infection down regulated beta-catenin (adherens junction protein) expression and caused increased permeability across confluent mesothelial monolayer. Our results suggest that in TB pleurisy, mycobacteria cause VEGF release from mesothelial cells and leads to protein exudation by altering mesothelial adherens junction proteins.


Subject(s)
Cytoskeletal Proteins/biosynthesis , Mycobacterium bovis , Trans-Activators/biosynthesis , Tuberculosis, Pleural/metabolism , Blotting, Western , Cadherins/biosynthesis , Down-Regulation , Electric Impedance , Endothelial Growth Factors/metabolism , Enzyme-Linked Immunosorbent Assay , Epithelium/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Lymphokines/metabolism , Permeability , Protein Isoforms , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , beta Catenin
5.
Eur Respir J ; 21(3): 539-44, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12662014

ABSTRACT

The pleural membrane consisting of pleural mesothelial cells and its underlying connective tissue layers play a critical role in immunological responses in both local and systemic diseases. The pleura, because of its intimate proximity to the lung, is positioned to respond to inflammatory changes in the lung parenchyma. Importantly, several systemic diseases have a predilection for expression on the pleural surface. Immunological responses in the pleura include the development of pleural permeability and pleural effusion formation as well as the development of pleural fibrosis and scarring. Under either circumstance, the normal functioning of the pleura is impaired and has multiple consequences leading to increased morbidity and even mortality for the patient. During infections in the pleural space, the pleural mesothelium responds by actively recruiting inflammatory phagocytic cells and allowing the movement of proteins from the vascular compartment into the pleural space. The release of chemokines by the pleural mesothelium allows for directed migration of phagocytic cells from the basilar surface of the pleura towards the apical surface. In malignant disease, the pleura may be the site of primary tumours such as mesothelioma and also the site for malignant metastatic deposits. Certain cancers such as cancers of the breast, ovary, lung, and stomach have a predilection for the pleural mesothelium. The process whereby malignant cells attach to the pleural mesothelium and develop autocrine mechanisms for survival in the pleural space are elucidated in this review. The pleura functions not only as a mechanical barrier, but also as an immunologically and metabolically responsive membrane that is involved in maintaining a dynamic homeostasis in the pleural space.


Subject(s)
Chemokines/metabolism , Cytokines/metabolism , Inflammation Mediators/analysis , Pleural Diseases/immunology , Pleural Diseases/pathology , Biomarkers/analysis , Chemokines/analysis , Cytokines/analysis , Epithelium/pathology , Humans , Pleura/anatomy & histology , Pleura/physiology , Pleural Effusion/immunology , Pleural Effusion/pathology , Prognosis , Sensitivity and Specificity , Severity of Illness Index
7.
Am J Physiol Lung Cell Mol Physiol ; 281(1): L119-25, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11404254

ABSTRACT

Pneumonia remains one of the most common infectious causes of mortality. Patients with pneumonia develop parapneumonic effusions with a high neutrophil count as well as high protein concentrations. We hypothesized that pulmonary parenchymal bacterial infection causes a permeability change in the pleural mesothelium by inducing the production of vascular endothelial growth factor (VEGF). Complicated parapneumonic pleural effusions (empyema) have a 19-fold higher VEGF level than pleural fluids secondary to congestive heart failure and a 4-fold higher level than pleural fluids secondary to uncomplicated parapneumonic effusions. We also analyzed the influence of live Staphylococcus aureus on mesothelial barrier function using a model of confluent mesothelial monolayers. There was a significant drop in electrical resistance across S. aureus-infected pleural mesothelial cell (PMC) monolayers. Recombinant VEGF also decreases PMC electrical resistance. Neutralizing antibodies to VEGF significantly inhibited the drop in PMC electrical resistance caused by S. aureus. S. aureus infection also caused a significant increase in protein leak across confluent mesothelial monolayers. Our results suggest that bacterial pathogens induce VEGF release in mesothelial cells and alter mesothelial permeability, leading to protein exudation in empyema.


Subject(s)
Pleura/metabolism , Staphylococcal Infections/metabolism , Staphylococcus aureus , Electric Impedance , Empyema, Pleural/complications , Endothelial Growth Factors/genetics , Endothelial Growth Factors/metabolism , Epithelial Cells/metabolism , Epithelial Cells/physiology , Heart Failure/complications , Humans , Lymphokines/genetics , Lymphokines/metabolism , Permeability , Pleura/pathology , Pleura/physiopathology , Pleural Effusion/etiology , Pleural Effusion/metabolism , Pneumonia/complications , RNA, Messenger/metabolism , Staphylococcal Infections/physiopathology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
8.
J Infect Dis ; 183(11): 1638-45, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11343213

ABSTRACT

Migration of polymorphonuclear neutrophils (PMNL) from the vascular compartment into the pleural space occurs rapidly during the development of parapneumonic effusions. This study investigated the polarized secretion of interleukin (IL)-8 in activated pleural mesothelial cells (PMC) and the migration of PMNL across resting, activated PMC monolayers. Results show that PMC produce IL-8 in a polar manner. When PMC were stimulated with Staphylococcus aureus or IL-1beta at the basal or at the apical surface, significantly (P< .05) more IL-8 was released toward the apical surface. This polarized production of IL-8 was confirmed by in situ hybridization. PMNL migration was higher from the basilar to apical than from the apical to basilar surface of PMC. Neutralizing antibodies against IL-8 and intercellular adhesion molecule (ICAM)-1 significantly (P< .001) blocked PMNL migration across activated monolayers. Thus, during pleural inflammation, PMC regulate the influx of PMNL into the pleural space by polar production of IL-8 and expression of ICAM-1.


Subject(s)
Epithelium/immunology , Intercellular Adhesion Molecule-1/physiology , Neutrophils/immunology , Cell Movement , Cells, Cultured , Epithelial Cells/immunology , Epithelial Cells/microbiology , Flow Cytometry , Humans , In Situ Hybridization , In Vitro Techniques , Intercellular Adhesion Molecule-1/analysis , Interleukin-8/analysis , Interleukin-8/pharmacology , Leukocytes, Mononuclear , Neutrophils/drug effects , Pleura/cytology , Staphylococcus aureus
9.
J Infect Dis ; 181(5): 1693-9, 2000 May.
Article in English | MEDLINE | ID: mdl-10823770

ABSTRACT

Bacterial empyema is a frequent complication of pneumonia in patients with acquired immunodeficiency syndrome (AIDS). A model of Staphylococcus aureus empyema was developed that closely resembles bacterial empyema in patients infected with human immunodeficiency virus (HIV). Results show a compartmentalized chemokine response in bacterial empyema. The chemokine levels were higher in the pleural compartment than in the peripheral circulation. Polymorphonuclear leukocyte counts, murine GRO-alpha (KC), and macrophage inflammatory protein-2 levels were significantly (P<.001) lower in CD4+ knockout (CD4 KO) mice pleural fluid than in CD4+ wild-type (CD4 WT) mice. The CD4 KO mice had poorer bacterial clearance than CD4 WT mice. During S. aureus infection, interleukin-10 levels increased in the CD4 KO mice, whereas interferon-gamma levels were increased in CD4 WT mice. CD4+ T cell depletion results in a decreased pleural chemokine response, decreased neutrophil influx into pleural space, and impaired bacterial clearance in empyema.


Subject(s)
CD4 Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , Chemokines, CXC , Empyema, Pleural/immunology , Intercellular Signaling Peptides and Proteins , Staphylococcal Infections/immunology , Staphylococcus aureus , Animals , CD4 Antigens/genetics , Chemokine CXCL1 , Chemokine CXCL2 , Chemotactic Factors/analysis , Empyema, Pleural/microbiology , Female , Growth Substances/analysis , HIV Infections/complications , Humans , Inflammation , Leukocyte Count , Mice , Mice, Inbred C57BL , Mice, Knockout , Monokines/analysis , Neutrophils/physiology , Time Factors
10.
Am J Physiol Lung Cell Mol Physiol ; 278(3): L591-8, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10710532

ABSTRACT

Pleural injury results in the death of mesothelial cells and denudation of the mesothelial basement membrane. Repair of the mesothelium without fibrosis requires proliferation and migration of mesothelial cells into the injured area. We hypothesized that monocyte chemoattractant protein-1 (MCP-1) induces proliferative and haptotactic responses in pleural mesothelial cells (PMCs) and that the MCP-1 binding receptor CCR2 mediates the pleural repair process. We demonstrate that PMCs exhibited MCP-1-specific immunostaining on injury. MCP-1 induced proliferative and haptotactic responses in PMCs. PMCs express CCR2 in a time-dependent manner. Fluorescence-activated cell sorting analysis demonstrated that interleukin (IL)-2 upregulated CCR2 protein expression in PMCs, whereas lipopolysaccharide (LPS) downregulated the response at the initial period compared with that in resting PMCs. However, the inhibitory potential of LPS was lost after 12 h and showed a similar response at 24 and 48 h. Haptotactic migration was upregulated in PMCs that were cultured in the presence of IL-2. The increased haptotactic capacity of mesothelial cells in the presence of IL-2 correlated with increased CCR2 mRNA expression. PMCs cultured in the presence of LPS showed decreased haptotactic activity to MCP-1. Blocking the CCR2 with neutralizing antibodies decreased the haptotactic response of PMCs to MCP-1. These results suggest that the haptotactic migration of mesothelial cells in response to MCP-1 are mediated through CCR2, which may play a crucial role in reepithelialization of the denuded basement membrane at the site of pleural injury and may thus contribute to the regeneration of the mesothelium during the process of pleural repair.


Subject(s)
Chemokine CCL2/metabolism , Pleura/injuries , Receptors, Chemokine , Receptors, Cytokine/physiology , Wounds and Injuries/physiopathology , Cell Division/physiology , Cell Movement/physiology , Cells, Cultured , Epithelial Cells/physiology , Humans , Interleukin-2/pharmacology , Lipopolysaccharides/pharmacology , Pleura/metabolism , Pleura/pathology , Pleura/physiopathology , RNA, Messenger/metabolism , Receptors, CCR2 , Receptors, Cytokine/genetics , Receptors, Cytokine/metabolism , Wounds and Injuries/metabolism , Wounds and Injuries/pathology
11.
Am J Respir Crit Care Med ; 161(2 Pt 1): 595-600, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10673205

ABSTRACT

Pleurodesis with talc is an accepted method for the treatment of symptomatic pleural effusions secondary to mesotheliomas. Patients with mesothelioma who have talc-induced pleurodesis have a lower morbidity than do those who do not have pleurodesis. The mechanisms whereby talc mediated these effects were considered to be secondary to a decrease or absence of a pleural effusion. The possibility that talc may directly affect malignant cells was not considered. The present study was designed to evaluate if talc directly effects cell death of malignant mesothelioma cells (MMC) or normal pleural mesothelial cells (PMC). Three confluent MMC and PMC were exposed to talc for 24, 48, and 72 h. In parallel experiments, glass beads similar in size to talc were included as control. Apoptosis was determined by terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end labeling (TUNEL) and DNA electrophoresis. Our results demonstrated that talc at a therapeutically achievable concentration (6 microg/cm(2)) induces significant apoptosis in MMC. Talc-induced maximum apoptosis in MMC (39.50 +/- 2.55%, 31.87 +/- 4.69%, and 15.10 +/- 3.93% in CRL-2081, CRL-5820, and CRL-5915, respectively) at 48 h, which was significantly (p < 0.05) greater than that in control cells. Electrophoresis of DNA isolated from talc-exposed MMC demonstrated the typical ladder pattern of internucleosomal DNA cleavage. Talc did not induce apoptosis in PMC, and glass beads did not cause significant apoptosis in either MMC or PMC. The present study has demonstrated that talc induces apoptosis in MMC without affecting normal mesothelial cells of the pleura.


Subject(s)
Apoptosis/drug effects , Cell Survival/drug effects , Mesothelioma/pathology , Pleural Neoplasms/pathology , Pleurodesis , Talc/pharmacology , Tumor Cells, Cultured/drug effects , Humans , Pleura/drug effects , Pleura/pathology , Pleural Effusion, Malignant/pathology , Tumor Cells, Cultured/pathology
13.
Oncol Res ; 11(4): 187-94, 1999.
Article in English | MEDLINE | ID: mdl-10566617

ABSTRACT

Malignant pleural mesothelioma (MPM), despite current therapeutic strategies, is still an aggressive tumor with a very poor prognosis. Interleukin-8 (IL-8), a proinflammatory and angiogenic cytokine, has an important role in tumor-related neovascularization. IL-8 has also been described to function as an autocrine growth factor. The purpose of this study was to evaluate the effect of IL-8 antibody (IL-8 Ab) on progression of MPM in vivo. Athymic nude mice (n = 65) were injected intrapleurally with human MPM cells (CRL-2081), equally divided into three groups (IL-8 Ab, control Ab, untreated), and received IP injection of IL-8 Ab, control Ab, or no treatment, respectively, every 48 h up to 15 days. Pleural fluid and serum IL-8 levels, and tumor and body weight of mice were measured following 5, 10, and 15 days of tumor injection. We found that both pleural fluid and serum IL-8 levels were significantly (P < 0.0001) lower in mice that received IL-8 Ab when compared to the other groups. In this group, lower IL-8 levels were associated with a decreased rate of tumor growth. There was a significant and direct correlation between pleural fluid IL-8 levels and tumor weight of all animals enrolled in this study (P < 0.0001, r = 0.88). We demonstrate that antibody treatment against IL-8 decreased human MPM progression. Our results suggest that treatments targeting the decrease of MPM-associated IL-8 levels or the effects of this protein may inhibit mesothelioma growth.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Interleukin-8/antagonists & inhibitors , Mesothelioma/therapy , Pleural Neoplasms/therapy , Animals , Body Weight , Humans , Immunohistochemistry , Interleukin-8/blood , Leukocyte Count , Male , Mesothelioma/blood , Mice , Mice, Nude , Pleural Effusion/chemistry , Pleural Neoplasms/blood
14.
J Infect Dis ; 180(5): 1616-23, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10515824

ABSTRACT

The pleural mesothelium is a dynamic cellular membrane with multiple key functions. It plays a pivotal role in pleural inflammation through its release of several cytokines and the expression of cell-surface molecules. The expression of intercellular adhesion molecule (ICAM)-1 in the pleural mesothelium of patients with active pleural tuberculosis and the role of ICAM-1 in monocyte transmigration across pleural mesothelium during tuberculous inflammation was investigated. Results indicate pleural mesothelial cells (PMCs) express ICAM-1 in tuberculous pleuritis. When PMCs were stimulated with bacille Calmette-Guérin (BCG) in vitro, they expressed ICAM-1 in a time-dependent manner. Monocyte transmigration was higher across PMC monolayers that had been stimulated with BCG. Blocking ICAM-1 on BCG-activated PMC monolayers inhibited monocyte transmigration against chemotactic gradient generated by macrophage inflammatory protein 1-alpha or monocyte chemotactic protein-1. These results indicate that ICAM-1 expression in PMCs facilitates monocyte transmigration during tuberculous pleural inflammation.


Subject(s)
Intercellular Adhesion Molecule-1/physiology , Monocytes/physiology , Mycobacterium bovis/immunology , Pleura/immunology , Tuberculosis, Pleural/immunology , Animals , Cells, Cultured , Chemokine CCL3 , Chemokine CCL4 , Chemotaxis, Leukocyte , Epithelium/immunology , Flow Cytometry , Humans , Immunohistochemistry , Intercellular Adhesion Molecule-1/immunology , Macrophage Inflammatory Proteins/immunology , Macrophage Inflammatory Proteins/metabolism , Mice , Pleura/pathology
15.
Am J Respir Crit Care Med ; 159(5 Pt 1): 1653-9, 1999 May.
Article in English | MEDLINE | ID: mdl-10228140

ABSTRACT

The recruitment of leukocytes to an area of injury or inflammation site is one of the most fundamental host defenses. Pulmonary tuberculosis is characterized by granulomatous inflammation with an extensive infiltration of mononuclear cells. In tuberculous pleurisy pleural mesothelial cells are exposed to mycobacteria in the pleural space. In this study we demonstrate that mouse pleural mesothelial cells (PMCs), when stimulated with BCG or IFN-gamma, produced MIP-1alpha and MCP-1 in vitro. IFN-gamma enhanced the BCG-mediated MIP-1alpha and MCP-1 expression in a concentration-dependent manner. The RT-PCR studies also confirmed that both BCG and IFN-gamma induce chemokine expression. IL-4 inhibited the BCG-mediated MIP-1alpha and MCP-1 expression in a concentration-dependent manner. The lower concentrations of IL-4 were ineffective; however, at higher concentrations, the inhibitory effect of IL-4 persisted for 24 h and decreased thereafter. BCG stimulation resulted in an increase of IFN-gamma and IL-4 receptors on PMCs. Our results demonstrate that Th1 and Th2 cytokines may regulate the C-C chemokine expression in PMCs and thus play a biologically important role in mononuclear cell recruitment to the pleural space.


Subject(s)
Chemokine CCL2/metabolism , Cytokines/physiology , Macrophage Inflammatory Proteins/metabolism , Pleura/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , Animals , Chemokine CCL2/genetics , Chemokine CCL3 , Chemokine CCL4 , Cytokines/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Interleukin-4/pharmacology , Macrophage Inflammatory Proteins/genetics , Mice , Mice, Inbred C57BL , Mycobacterium bovis/physiology , Pleura/cytology , Pleura/drug effects , RNA, Messenger/metabolism , Receptors, Interleukin-4/metabolism
16.
Semin Respir Infect ; 14(1): 9-17, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10197393

ABSTRACT

The pleura responds to the presence of infecting organisms with a vigorous inflammatory response associated with an exudation of white blood cells and proteins. Changes in pleural permeability lead to formation of an exudative pleural effusion. The pleural mesothelial cell is the primary cell lining the pleural space and, when activated by the presence of organisms, initiates the inflammatory response by releasing a battery of chemokines and cytokines. Mesothelial cells are actively phagocytic and also release oxidants and proteases. The acute inflammatory process may resolve with appropriate antibiotic therapy and drainage leaving minimal fibrosis. However, under certain circumstances vigorous pleural fibrosis with scarring and loss of delineation of pleural surfaces can occur. Recognition of the stage of development of the empyema is an important clinical judgement that can affect outcome. The pathogenesis of infections of the pleural space and the role played by the various cell types is delineated in this article.


Subject(s)
Empyema, Pleural/physiopathology , Pleural Effusion/physiopathology , Pleurisy/physiopathology , Empyema, Pleural/immunology , Empyema, Pleural/pathology , Fibrosis , Humans , Immunocompromised Host , Pleural Effusion/immunology , Pleural Effusion/pathology , Pleurisy/immunology , Pleurisy/pathology , Pneumonia, Bacterial/complications
17.
Cancer Res ; 59(2): 367-71, 1999 Jan 15.
Article in English | MEDLINE | ID: mdl-9927048

ABSTRACT

Interleukin 8 (IL-8) is a potent chemokine that also has a direct growth-potentiating effect on certain tumors. In the present study, we determined IL-8 levels in human malignant mesothelioma (MM) effusions and congestive heart failure pleural fluids. We also investigated antigenic IL-8 production by different MM cell lines, and we describe the role of IL-8 in the autocrine growth regulation of MMs. Mesothelial (CRL-9444 = MC) and MM (CRL-2081 = MM-1, CRL-5915 = MM-2, and CRL-5820 = MM-3) cell lines were grown using standard culture methods. The bioactive IL-8 levels were measured in supernatants of cultured cells by ELISA, and the expression of cell-associated immunoreactive IL-8 was observed by immunohistochemistry. The proliferative activity was determined by thymidine ([3H]thymidine) incorporation and also by direct cell counts after incubation with varying concentrations of IL-8 in the presence/absence of specific polyclonal IL-8 antibody. We found significantly higher levels of IL-8 in mesothelioma pleural fluids than congestive heart failure and a time-dependent increase in IL-8 levels in MM-1 and MM-2 cell supernatants during 96 h of incubation. IL-8 levels were nearly undetectable in MM-3 and MC cell line supernatants. In MM-1 and MM-2 cells, IL-8 caused a dose-dependent increase of [3H]thymidine incorporation to maximal levels of 46.3 +/- 3.6% and 12.3 +/- 1.6% (P < 0.001), respectively, when compared with serum-free medium as control. Neutralization of IL-8 significantly decreased proliferative activity of MM-1 and MM-2. IL-8 did not induce proliferative activity in MM-3 and MC cells. We conclude that IL-8 had a direct growth-potentiating activity in MMs.


Subject(s)
Growth Substances/physiology , Interleukin-8/physiology , Mesothelioma/pathology , Cell Division , Cell Line , Epithelial Cells/chemistry , Humans , Interleukin-8/analysis , Thymidine/metabolism
18.
J Infect Dis ; 178(5): 1450-6, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9780267

ABSTRACT

Pulmonary tuberculosis is characterized by granulomatous inflammation with an extensive infiltration of mononuclear phagocytes, but the mechanisms of phagocyte recruitment to the pleural space is unknown. In this study, pleural fluid from patients with tuberculosis contained significantly (P<.001) more biologically active MIP-1alpha and MCP-1 (C-C cytokines) than did effusions from patients with congestive heart failure. Antigenic MIP-1alpha and MCP-1 was detected by immunocytochemistry in pleural biopsy sections of patients with tuberculous pleurisy. In vitro, pleural mesothelial cells stimulated with bacille Calmette-Guérin (BCG) or interferon (IFN)-gamma produced MIP-1alpha and MCP-1. Reverse transcription-polymerase chain reaction studies confirmed that both BCG and IFN-gamma induced MIP-1alpha and MCP-1 expression in mesothelial cells, demonstrating that mesothelial cell-derived C-C chemokines play a biologically important role in the recruitment of mononuclear cells to the pleural space.


Subject(s)
Chemokine CCL2/biosynthesis , Epithelial Cells/metabolism , Macrophage Inflammatory Proteins/biosynthesis , Mycobacterium/metabolism , Pleura/metabolism , Tuberculosis, Pleural/metabolism , Biopsy , Chemokine CCL2/genetics , Chemokine CCL3 , Chemokine CCL4 , Chemotaxis, Leukocyte , Epithelial Cells/microbiology , Heart Failure/metabolism , Humans , Macrophage Inflammatory Proteins/genetics , Pleura/microbiology , Pleural Effusion/metabolism , RNA, Messenger/metabolism
19.
Am J Respir Crit Care Med ; 158(3): 971-8, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9731033

ABSTRACT

Treatment of symptomatic carcinomatous pleural effusions is primarily directed at local palliation with a wide variety of sclerosing agents, of which talc is considered to be the most successful. The mechanism whereby talc achieves this effect is unknown. The objective of this study was to investigate whether talc stimulates pleural mesothelial cells (PMC) to release C-X-C and/or C-C chemokines and express adhesion molecules that initiate and amplify the inflammatory process in the pleural space. When PMC were challenged with talc in vitro, interleukin-8 (IL-8) and monocyte chemotactic protein-1 (MCP-1) levels were increased (p < 0.001) both at the protein and the mRNA level as compared with unstimulated cultures. Talc-stimulated PMC culture supernatant showed chemotactic activity for neutrophils and monocytes. The chemotactic activity of PMC culture supernatant was blocked by 44.2% with IL-8-specific antibody and by 55.7% with MCP-1-specific antibody, demonstrating that PMC-derived chemokines are bioactive. Talc also enhanced intercellular adhesion molecule-1 (ICAM-1) expression in PMC. The data demonstrate that talc stimulates PMC to release chemokines and express adhesion molecules that may play a critical role in pleurodesis.


Subject(s)
Chemokines, CC/genetics , Chemokines, CXC/genetics , Intercellular Adhesion Molecule-1/genetics , Pleura/drug effects , Pleural Effusion/physiopathology , Sclerosing Solutions/pharmacology , Talc/pharmacology , Analysis of Variance , Antibodies , Cells, Cultured , Chemokine CCL2/genetics , Chemotaxis, Leukocyte , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Flow Cytometry , Humans , Intercellular Adhesion Molecule-1/drug effects , Interleukin-8/genetics , Monocytes/pathology , Neutrophils/pathology , Pleura/metabolism , Pleura/pathology , Pleural Effusion/metabolism , Pleural Effusion/pathology , Pleurodesis , RNA, Messenger/analysis , RNA, Messenger/genetics
20.
J Lab Clin Med ; 132(3): 202-9, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9735926

ABSTRACT

Parapneumonic pleural effusions are associated with the presence of a variety of inflammatory cells whose influx into the pleural space is attributed to the presence of inflammatory cytokines. Macrophage inflammatory protein-1alpha (MIP-1alpha), an important mononuclear chemokine, plays a critical role in pulmonary parenchymal inflammatory disease, but its role in the recruitment and activation of mononuclear phagocytes in the pleural space is unknown. In this study we demonstrate that complicated parapneumonic pleural effusions (empyema) and uncomplicated parapneumonic pleural effusions contain significantly (P < .001) higher levels of MIP-1alpha with higher numbers of mononuclear cells when compared with effusions resulting from malignancy and congestive heart failure. The MIP- 1alpha was biologically active and contributed 43% and 37% of the mononuclear chemotactic activity of complicated and uncomplicated parapneumonic pleural fluids, respectively. In vitro, human mesothelial cells, when stimulated with interleukin-1beta (IL-1beta), tumor necrosis factor-alpha (TNF-alpha), or bacterial lipopolysaccharide (LPS), produced MIP-1alpha. Northern blot analysis confirmed that both endogenous (IL-1beta or TNF-alpha) and exogenous (LPS) factors induce MIP-1alpha expression in mesothelial cells. Supernatants from activated mesothelial cells demonstrated chemotactic activity for mononuclear cells. This activity was blocked by MIP-1alpha antibody, indicating that the MIP-1alpha released was biologically active. We conclude that in parapneumonic pleural effusions, MIP-1alpha plays a major but not exclusive role in the recruitment of mononuclear leukocytes from the vascular compartment to the pleural space, and pleural mesothelial cells by production of MIP-1alpha actively participate in this process.


Subject(s)
Macrophage Inflammatory Proteins/metabolism , Pleural Effusion/metabolism , Pneumonia, Bacterial/metabolism , Cell Count , Cells, Cultured , Chemokine CCL3 , Chemokine CCL4 , Chemotaxis/drug effects , Chemotaxis/physiology , Culture Media, Conditioned/pharmacology , Epithelium/drug effects , Epithelium/metabolism , Heart Failure/metabolism , Heart Failure/pathology , Humans , Interleukin-1/pharmacology , Leukocytes, Mononuclear/drug effects , Lipopolysaccharides/pharmacology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Macrophage Inflammatory Proteins/genetics , Pleural Effusion/pathology , Pneumonia, Bacterial/pathology , RNA, Messenger/biosynthesis , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL