Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Am J Physiol ; 277(4): L737-42, 1999 10.
Article in English | MEDLINE | ID: mdl-10516214

ABSTRACT

Airway epithelial cell (AEC) proliferation is crucial to the maintenance of an intact airway surface and the preservation of host defenses. The factors that regulate AEC proliferation are not known. Keratinocyte growth factor (KGF), also known as FGF-7, is a member of the fibroblast growth factor family and a known epithelial cell mitogen. We studied the influence of KGF on the growth of cultured human bronchial epithelial cells and on bronchial cells of rats treated with KGF in vivo. First, we demonstrated the mRNA for the KGF receptor (KGFR) in both normal human bronchial epithelial (NHBE) cells and BEAS-2B cells (a human bronchial epithelial cell line). KGF caused a dose-dependent increase in DNA synthesis, as assessed by thymidine incorporation, in both cell types, with a maximal twofold increase in NHBE cells after 50 ng/ml KGF (P < 0.001). KGF also induced a doubling in NHBE cell number at 10 ng/ml (P < 0.001). Finally, we determined the effect of intratracheal administration of KGF to rats on proliferation of AEC in vivo. Measuring bromodeoxyuridine (BrdU) incorporation in AEC nuclei, KGF increased BrdU labeling of rat AEC in both large and small airways by approximately threefold compared with PBS-treated controls (P < 0.001). Thus KGF induces proliferation of bronchial epithelial cells both in vitro and in vivo.


Subject(s)
Bronchi/cytology , Fibroblast Growth Factors , Growth Substances/pharmacology , Receptors, Fibroblast Growth Factor , Animals , Bronchi/metabolism , Cell Division/drug effects , Cell Line, Transformed , Epithelial Cells/cytology , Epithelial Cells/metabolism , Fibroblast Growth Factor 10 , Fibroblast Growth Factor 7 , Humans , Male , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Fibroblast Growth Factor, Type 2 , Receptors, Growth Factor/genetics
2.
J Cell Sci ; 112 ( Pt 12): 2049-57, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10341222

ABSTRACT

Keratinocyte growth factor (KGF), also called fibroblast growth factor-7, is widely known as a paracrine growth and differentiation factor that is produced by mesenchymal cells and has been thought to act specifically on epithelial cells. Here it is shown to affect a new cell type, the microvascular endothelial cell. At subnanomolar concentrations KGF induced in vivo neovascularization in the rat cornea. In vitro it was not effective against endothelial cells cultured from large vessels, but did act directly on those cultured from small vessels, inducing chemotaxis with an ED50 of 0.02-0.05 ng/ml, stimulating proliferation and activating mitogen activated protein kinase (MAPK). KGF also helped to maintain the barrier function of monolayers of capillary but not aortic endothelial cells, protecting against hydrogen peroxide and vascular endothelial growth factor/vascular permeability factor (VEGF/VPF) induced increases in permeability with an ED50 of 0.2-0.5 ng/ml. These newfound abilities of KGF to induce angiogenesis and to stabilize endothelial barriers suggest that it functions in microvascular tissue as it does in epithelial tissues to protect them against mild insults and to speed their repair after major damage.


Subject(s)
Capillary Permeability/drug effects , Cornea/drug effects , Endothelium, Vascular/drug effects , Fibroblast Growth Factors , Growth Substances/pharmacology , Keratinocytes , Neovascularization, Physiologic/drug effects , Animals , Calcium-Calmodulin-Dependent Protein Kinases/drug effects , Cell Division/drug effects , Cell Movement/drug effects , Cells, Cultured , Cornea/cytology , Endothelium, Vascular/cytology , Female , Fibroblast Growth Factor 10 , Fibroblast Growth Factor 7 , Humans , Rats , Rats, Inbred F344 , Recombinant Proteins/pharmacology
3.
Am J Physiol ; 275(4): L780-7, 1998 10.
Article in English | MEDLINE | ID: mdl-9755111

ABSTRACT

Alveolar epithelial cell (AEC) injury and repair are important in the pathogenesis of oxidant-induced lung damage. Keratinocyte growth factor (KGF) prevents lung damage and mortality in animals exposed to various forms of oxidant stress, but the protective mechanisms are not yet established. Because DNA strand break (DNA-SB) formation is one of the earliest cellular changes that occurs after cells are exposed to an oxidant stress, we determined whether KGF reduces H2O2-induced pulmonary toxicity by attenuating AEC DNA damage. KGF (10-100 ng/ml) decreased H2O2 (0.05-0.5 mM)-induced DNA-SB formation in cultured A549 and rat alveolar type II cells measured by an alkaline unwinding, ethidium bromide fluorometric technique. The protective effects of KGF were independent of alterations in catalase, glutathione (GSH), or the expression of bcl-2 and bax, two protooncogenes known to regulate oxidant-induced apoptosis. Actinomycin D and cycloheximide abrogated protective effects of KGF. Furthermore, protection by KGF was completely blocked by 1) genistein, a tyrosine kinase inhibitor; 2) staurosporine and calphostin C, protein kinase C (PKC) inhibitors; and 3) aphidicolin, butylphenyl dGTP, and 2',3'-dideoxythymidine 5'-triphosphate, inhibitors of DNA polymerase. We conclude that KGF attenuates H2O2-induced DNA-SB formation in cultured AECs by mechanisms that involve tyrosine kinase, PKC, and DNA polymerases. These data suggest that the ability of KGF to protect against oxidant-induced lung injury is partly due to enhanced AEC DNA repair.


Subject(s)
DNA Damage , DNA Repair/drug effects , Enzyme Inhibitors/pharmacology , Fibroblast Growth Factors , Growth Substances/pharmacology , Hydrogen Peroxide/toxicity , Pulmonary Alveoli/physiology , Animals , Aphidicolin/pharmacology , Cell Death/drug effects , Cycloheximide/pharmacology , Dactinomycin/pharmacology , Fibroblast Growth Factor 10 , Fibroblast Growth Factor 7 , Genistein/pharmacology , Growth Substances/physiology , Humans , Kinetics , Lung Neoplasms , Naphthalenes/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Pulmonary Alveoli/cytology , Pulmonary Alveoli/drug effects , Rats , Staurosporine/pharmacology , Tumor Cells, Cultured
4.
Tumour Biol ; 19(1): 52-9, 1998.
Article in English | MEDLINE | ID: mdl-9422082

ABSTRACT

PKR (protein kinase, interferon-responsive) is a ribosomal-associated protein kinase found in all human cells. When activated by dsRNA or polyanionic substances, PKR efficiently inhibits cellular protein synthesis. PKR expression has been correlated with cellular differentiation in a number of tumor types, including squamous cell carcinoma of the head and neck region. Although transfection of PKR into mouse fibroblasts and yeast cells inhibits proliferation, it is not known if modulation of native PKR levels occurs during cellular proliferation and differentiation in human normal and neoplastic tissues. To determine whether PKR expression was inversely related to proliferative activity in vivo, we used double-label immunohistochemistry to colocalize PKR and the proliferation marker, proliferating cell nuclear antigen (PCNA), in a series of head and neck squamous cell carcinomas. Overall, neoplasms demonstrating high levels of PKR showed low levels of PCNA immunoreactivity; carcinomas with low levels of PKR expressed high levels of PCNA. Within individual tumors, PKR and PCNA showed an inverse regional distribution: PKR was located predominantly in the center of tumor nests, while PCNA was restricted to the periphery. Patients whose tumors expressed high levels of both PKR and PCNA had the longest mean disease-free survival. These findings support the hypothesis that PKR levels are modulated in cell proliferation and differentiation in head and neck squamous cell carcinoma. Further studies are needed to clarify the mechanisms underlying the antiproliferative activity of PKR.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Head and Neck Neoplasms/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Protein Serine-Threonine Kinases/metabolism , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal , Carcinoma, Squamous Cell/pathology , Disease-Free Survival , Female , Head and Neck Neoplasms/pathology , Humans , Immunoenzyme Techniques , Interferons/metabolism , Male , Middle Aged , Neoplasm Proteins/metabolism
5.
J Histochem Cytochem ; 45(8): 1165-70, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9267477

ABSTRACT

Incorporation of halogenated nucleotide analogues is often used to assess DNA synthesis and to quantitate cellular proliferation. Multiple antibodies have been developed to bromodeoxyuridine (BrdUrd) and it is the most frequently utilized substrate. Because the immunodetection of incorporated BrdUrd requires DNA denaturation or nuclease digestion, most of these antibodies are not reactive in tissues or cells fixed with crosslinking agents. Antigen retrieval techniques utilizing protease digestion restore BrdUrd antigenicity and permit the detection of BrdUrd in formalin-fixed tissue. However, during the development of a double label immunohistochemical protocol to quantitate proliferating alveolar Type II cells, we noted nucleus-specific staining in lung sections from animals that had not received BrdUrd. Therefore, we systematically analyzed the specificity of the immunohistochemical detection of incorporated BrdUrd in formalin-fixed tissue after protease digestion. Enzymatic antigen recovery diminished the specificity of the BrdUrd reaction product and caused false-positive staining with the BU-1, B44, and BR3 monoclonal antibodies. Staining was less prominent with Bu20a but was more specific. Protease antigen recovery may decrease the specificity of BrdUrd immunodetection. Appropriate controls are required when enzymatic digestion is used to detect incorporated BrdUrd in formalin-fixed tissue. The type and duration of fixation, antibody to BrdUrd, protease, and tissue may affect the specificity of the staining pattern.


Subject(s)
Bromodeoxyuridine/analysis , Endopeptidases/pharmacology , Immunohistochemistry/methods , Tissue Fixation , Animals , Antigens/analysis , Antigens/drug effects , Bromodeoxyuridine/immunology , False Positive Reactions , Male , Rats , Rats, Sprague-Dawley , Sensitivity and Specificity , Testis/chemistry , Time Factors , Tissue Fixation/methods
6.
Am J Physiol ; 272(6 Pt 1): L1115-20, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9227512

ABSTRACT

Hepatocyte growth factor (HGF) can influence epithelial cell growth and differentiation. We examined the actions of recombinant human HGF (rhHGF) on the differentiation of human primary tracheal epithelial (HTE) cells cultured in vitro for up to 96 h. Basolateral, but not apical, treatment of confluent HTE cell sheets for 48 h with rhHGF led to increases in cell height, cell volume, cilia, and total protein content. Basolateral rhHGF treatment produced a decrease in HGF receptor (c-met) expression but had no effect on c-met mRNA levels. HTE cell sheets treated with rhHGF for 48 h showed a significant increase in mediator-induced Cl- secretion and a decrease in amiloride-sensitive sodium absorption. No effect on transepithelial resistance was observed with rhHGF treatment. The enhancement of short-circuit responses by basolateral rhHGF was dose dependent. Our results demonstrate that rhHGF has hypertrophic actions on, and can influence the differentiation of, human airway epithelia in vitro, presumably through the activation of c-met at the basolateral surface of these cells.


Subject(s)
Cell Differentiation/drug effects , Hepatocyte Growth Factor/pharmacology , Trachea/cytology , Trachea/physiology , Amiloride/pharmacology , Bradykinin/pharmacology , Cell Division/drug effects , Cell Membrane/drug effects , Cell Membrane/physiology , Cells, Cultured , Chlorides/metabolism , Cilia/drug effects , Cilia/ultrastructure , Epithelial Cells , Epithelium/drug effects , Histamine/pharmacology , Humans , Hypertrophy , Isoproterenol/pharmacology , Membrane Potentials/drug effects , Methacholine Chloride/pharmacology , Proto-Oncogene Proteins c-met , Receptor Protein-Tyrosine Kinases/biosynthesis , Recombinant Proteins/pharmacology
7.
Am J Physiol ; 272(6 Pt 1): L1174-80, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9227520

ABSTRACT

Administration of exogenous keratinocyte growth factor (KGF) prevents or attenuates several forms of oxidant-mediated lung injury. Because DNA damage in epithelial cells is a component of radiation pneumotoxicity, we determined whether KGF ameliorated DNA strand breaks in irradiated A549 cells. Cells were exposed to 137Cs gamma rays, and DNA damage was measured by alkaline unwinding and ethidium bromide fluorescence after a 30-min recovery period. Radiation induced a dose-dependent increase in DNA strand breaks. The percentage of double-stranded DNA after exposure to 30 Gy increased from 44.6 +/- 3.5% in untreated control cells to 61.6 +/- 5.0% in cells cultured with 100 ng/ml KGF for 24 h (P < 0.05). No reduction in DNA damage occurred when the cells were cultured with KGF but maintained at 0 degree C during and after irradiation. The sparing effect of KGF on radiation-induced DNA damage was blocked by aphidicolin, an inhibitor of DNA polymerases-alpha, -delta, and -epsilon and by butylphenyl dGTP, which blocks DNA polymerase-alpha strongly and polymerases-delta and -epsilon less effectively. However, dideoxythymidine triphosphate, a specific inhibitor of DNA polymerase-beta, did not abrogate the KGF effect. Thus KGF increases DNA repair capacity in irradiated pulmonary epithelial cells, an effect mediated at least in part by DNA polymerases-alpha, -delta, and -epsilon. Enhancement of DNA repair capability after cell damage may be one mechanism by which KGF is able to ameliorate oxidant-mediated alveolar epithelial injury.


Subject(s)
DNA Damage , DNA Repair/drug effects , Fibroblast Growth Factors , Growth Substances/pharmacology , Pulmonary Alveoli/radiation effects , Aphidicolin/pharmacology , Cell Line , Cesium Radioisotopes , DNA/biosynthesis , DNA/drug effects , DNA/radiation effects , DNA Polymerase I/antagonists & inhibitors , DNA Polymerase II/antagonists & inhibitors , Deoxyguanine Nucleotides/pharmacology , Dideoxynucleotides , Enzyme Inhibitors/pharmacology , Epithelium/drug effects , Epithelium/radiation effects , Fibroblast Growth Factor 10 , Fibroblast Growth Factor 7 , Gamma Rays , Humans , Thymine Nucleotides/pharmacology
8.
Am J Physiol ; 272(4 Pt 1): L681-9, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9142942

ABSTRACT

Keratinocyte growth factor (KGF) has recently been shown to protect rats from hyperoxia-induced lung injury. However, the mechanism of the protective effect of KGF remains unclear. To elucidate the mechanism of action of KGF, we determined the effect of KGF on the barrier function of epithelial monolayers exposed to H(2)O(2). Calu-3 (human airway epithelial cells) were grown on Transwell membranes, and the permeability to fluorescein isothiocyanate-albumin was measured. Exposure to 0.5 mM H(2)O(2) significantly increased permeability from 1.50 +/- 0.09 to 24.8 +/- 1.5 (mean +/- SE x 10(-6) cm/s; P < 0.001). Incubation of monolayers with 50 ng/ml KGF for 24 h significantly reduced basal albumin flux (0.85 +/- 0.09; P < 0.001), and pretreatment with KGF completely abolished the H(2)O(2)-induced permeability increase (1.08 +/- 0.09). The protective effect of KGF was dose dependent and was observed at concentrations as low as 1 ng/ml. Partial amelioration of the H(2)O(2)-induced permeability increase occurred after 1 h of exposure to KGF. Treatment of cells with calphostin C, an inhibitor of protein kinase C (PKC), had no effect on the permeability of control or H(2)O(2)-treated cells. Calphostin C abolished both the KGF-mediated decrease in basal albumin flux and the protective effect of KGF against H(2)O(2)-induced increases in permeability. KGF pretreatment also prevented H(2)O(2)-induced disruption of F-actin staining patterns, suggesting stabilization of the cytoskeleton. These studies demonstrate that KGF decreases albumin flux across airway epithelial monolayers and prevents H(2)O(2)-induced increases in permeability by a PKC-dependent process that may involve stabilization of the cytoskeleton.


Subject(s)
Fibroblast Growth Factors , Growth Substances/pharmacology , Hydrogen Peroxide/pharmacology , Lung/metabolism , Cell Membrane Permeability/drug effects , Cytoskeleton/drug effects , Cytoskeleton/ultrastructure , Dose-Response Relationship, Drug , Epithelium/metabolism , Epithelium/pathology , Epithelium/ultrastructure , Fibroblast Growth Factor 10 , Fibroblast Growth Factor 7 , Humans , Lung/pathology , Lung/ultrastructure , Time Factors , Tumor Cells, Cultured
10.
Tohoku J Exp Med ; 183(4): 273-84, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9549827

ABSTRACT

We investigated the effects of pre-treatment with dibutyryl cAMP (db-cAMP) or cGMP on endotoxin-induced hemodynamic changes and pulmonary vascular permeability in isolated perfused rat lungs. Intraperitoneal injection of Salmonella enteritidis endotoxin (2 mg/kg) caused increases in pulmonary arterial resistance (Ra) after venous reservoir elevation, in pulmonary filtration coefficient (Kf) and in lung wet-to-dry (W/D) weight ratio. Pre-treatment with db-cAMP blocked endotoxin-induced increases in Ra, Kf and W/D weight ratio. Pre-treatment with cGMP attenuated only the increase in Ra caused by endotoxin. Moreover, administration of db-cAMP 2 hours after endotoxin injection attenuated the increase in Ra induced by endotoxin treatment. The increases in Kf and W/D weight ratio caused by endotoxin were not affected by post-treatment with db-cAMP. Since the increases in Ra, Kf and W/D weight ratio caused by endotoxin were blocked by pre-treatment with db-cAMP, agents that increase intracellular cAMP level may be useful to prevent acute pulmonary vascular injury.


Subject(s)
Bucladesine/pharmacology , Capillary Permeability/physiology , Endotoxins/antagonists & inhibitors , Pulmonary Circulation/physiology , Salmonella enteritidis , Vascular Resistance/physiology , Animals , Capillary Permeability/drug effects , Cyclic GMP/pharmacology , Endotoxins/toxicity , In Vitro Techniques , Male , Organ Size/drug effects , Organ Size/physiology , Pulmonary Circulation/drug effects , Rats , Rats, Sprague-Dawley , Vascular Resistance/drug effects
11.
Am J Respir Cell Mol Biol ; 15(5): 574-81, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8918364

ABSTRACT

Alveolar epithelial injury occurs universally in common respiratory illnesses associated with diffuse lung damage. After alveolar injury, type II cells proliferate and reestablish epithelial integrity, thereby restoring normal lung structure and function. However, the regulation of type II cell proliferation and alveolar epithelial repair is poorly understood. Hepatocyte growth factor/scatter factor (HGF/SF) is a heparin-binding growth factor that has been shown to be mitogenic for cultured alveolar type II cells. In this study, we determined the effect of intratracheal instillation of rhHGF/SF on type II cell proliferation in vivo. To quantify the alveolar type II cell proliferative response, we developed a double-label immunohistochemical technique to detect replicating alveolar type II cells in formalin-fixed lung sections that utilized the identification of proliferating cells by bromodeoxyuridine (BrdUrd) incorporation into DNA and alveolar type II cells by 3F9 immunoreactivity. BrdUrd detection was optimized by enzymatic antigen recovery and silver intensification of the horseradish peroxidase reaction product. Intratracheal instillation of rhHGF/SF induced a time- and dose-dependent increase in type II cell proliferation. The type II cell labeling index increased to 12.3 +/- 6.0% 48 h after 1.0 mg/kg rhHGF/SF administration, compared with 2.6 +/- 0.9% after PBS instillation. To compare the normal type II cell reparative response with the level of proliferation after exogenous rhHGF/SF administration, we measured the specific alveolar type II cell labeling index in rat lung sections obtained from animals exposed to hyperoxia for 50 h and then allowed to recover in room air. After 1 day of recovery, the alveolar type II cell labeling index was 0.45 +/- 0.2%. The specific labeling index increased to 5.4 +/- 1.3% at 2 days and then declined to 0.31 +/- 0.16% 5 days after hyperoxia exposure. In animals not exposed to hyperoxia, the alveolar type II cell labeling index was 0.6 +/- 0.14%. These studies demonstrated that intratracheal instillation of rhHGF/SF promoted alveolar type II cell proliferation in vivo. The maximal level of type II cell proliferation after rhHGF/SF administration was more than twice that reached during recovery from hyperoxia exposure. Thus, intratracheal instillation of HGF/SF may provide a potential strategy to promote type II cell proliferation and augment alveolar epithelial repair after lung injury.


Subject(s)
Hepatocyte Growth Factor/administration & dosage , Immunoenzyme Techniques , Plant Lectins , Pulmonary Alveoli/drug effects , Animals , Antigens/analysis , Bromodeoxyuridine/analysis , Cell Division/drug effects , DNA/biosynthesis , Hyperoxia , Instillation, Drug , Lectins , Male , Pulmonary Alveoli/chemistry , Pulmonary Alveoli/cytology , Rats , Rats, Sprague-Dawley , Trachea
12.
Arthritis Rheum ; 39(9): 1566-75, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8814069

ABSTRACT

OBJECTIVE: Angiogenesis is an integral component of the vasculoproliferative phase of rheumatoid arthritis (RA). Recently, a heparin-binding cytokine termed hepatocyte growth factor (HGF), or scatter factor (due to its ability to disperse cohesive epithelial colonies), was described. We conducted this study to investigate the hypothesis that this cytokine was present in the milieu of the inflamed joint, and that it contributed to the chemotaxis of endothelial cells in the synovial tissue. METHODS: We examined synovial fluid, synovial tissue, and peripheral blood from 91 patients with RA and other arthritides. We used 83 total samples in an enzyme-linked immunosorbent assay to quantitate the HGF in synovial fluids and peripheral blood. To determine whether the HGF was biologically active, an epithelial scatter factor assay was performed. Immunohistochemical analysis was used to determine localization in synovial tissues. To define a function for synovial HGF, we preincubated rheumatoid synovial fluids with neutralizing anti-HGF and measured the ability of these synovial fluids to induce endothelial chemotaxis. RESULTS: Synovial fluid from patients with RA contained a mean +/- SEM HGF concentration of 2.0 +/- 0.3 ng/ml, while synovial fluid from patients with other arthritides (including inflammatory arthritis) contained 2.4 +/- 0.7 ng/ml HGF. Osteoarthritis (OA) patient samples contained the smallest quantities of synovial fluid HGF at 0.9 +/- 0.1 ng/ml. RA synovial fluid contained significantly more HGF than did RA peripheral blood (1.1 +/- 0.2 ng/ml) (P < 0.05). Rheumatoid synovial fluids induced more scattering of cells than did OA synovial fluids, suggesting a role for this cytokine in rheumatoid joint destruction. Interleukin-1 beta induced expression of rheumatoid synovial tissue fibroblast antigenic HGF and scatter factor activity. Immunohistochemically, HGF, as well as the HGF receptor (the met gene product), localized to significantly more rheumatoid synovial tissue lining cells than normal lining cells (P < 0.05). Both HGF and its receptor immunolocalized to subsynovial macrophages as well. Levels of synovial tissue immunoreactive HGF correlated positively with the number of synovial tissue blood vessels. Anti-HGF neutralized a mean of 24% of the chemotactic activity for endothelial cells found in 10 rheumatoid synovial fluid samples. CONCLUSION: These results indicate that synovial HGF may contribute to the vasculoproliferative phase of inflammatory arthritides such as RA, by inducing HGF-mediated synovial neovascularization. These findings point to a newly described role for HGF in the fibroproliferative phase of RA-associated synovitis.


Subject(s)
Arthritis, Rheumatoid/blood , Hepatocyte Growth Factor/analysis , Hepatocyte Growth Factor/physiology , Osteoarthritis/blood , Receptor Protein-Tyrosine Kinases/analysis , Synovial Fluid/chemistry , Synovial Membrane/chemistry , Chemotaxis , Endothelium, Vascular/pathology , Fibroblasts/metabolism , Hepatocyte Growth Factor/blood , Humans , Proto-Oncogene Proteins c-met , Synovial Fluid/metabolism , Synovial Membrane/metabolism , Synovial Membrane/pathology
13.
Ann Clin Lab Sci ; 26(5): 437-50, 1996.
Article in English | MEDLINE | ID: mdl-8879362

ABSTRACT

Positive pressure ventilation with hyperdistention of the lungs (PPVHDL) causes microscopic lung injury in rats and in mice. This study compared lung lavage and serum levels of lactate dehydrogenase (LDH), aspartate aminotransferase (AST), creatinine phosphokinase (CPK), lung lavage and plasma endothelin-1 (ET-1) concentration, lung tissue ET-1 mRNA expression, angiotensin converting enzyme (ACE) activity of lung homogenates, and histology of the lung structure in control and PPVHDL rats. Rats were anesthetized with pentobarbital. While control rats were breathing spontaneously, the PPVHDL rats were ventilated with a rodent ventilator delivering 30 percent oxygen, a tidal volume of 18.6 +/- 4.5 ml/kg, and a respiratory rate of 55 to 60 per minute. End-tidal CO2 was maintained at 38-40 mm Hg. After seven hours, rats were killed and the lungs were lavaged. Red blood cells were present in the sediment of lavage fluid in PPVHDL rats and their lung structure showed severe congestion, alveolar septa filled with red cells, and extravasation of red blood cells and inflammatory cells into the alveolar space. Lung lavage fluid AST and LDH were significantly higher in the PPVHDL compared with the control group (P < 0.03 and P < 0.001, respectively). Electrophoresis of the lung lavage LDH showed increased peak-5 in the PPVHDL group. Serum LDH, CPK, AST, and potassium concentrations [K]+ were significantly higher in the PPVHDL rats whereas their serum total protein level was significantly lower than the control group (P < 0.001). Electrophoretic patterns of serum and lung lavage protein were similar in both groups indicating a transmural passage of serum protein from the intravascular to the intra-alveolar space. No significant difference was found in lung tissue ET-1 mRNA expression and lung protein concentration between the two groups. Lung ACE activity, in contrast, was significantly lower in PPVHDL rats. This study demonstrated that moderate alveolar hyperdistention caused significant structural lung damage accompanied by decreased ACE activity after seven hours of mechanical ventilation and that elevated lung lavage and serum LDH and AST levels in lung lavage and in serum might be early markers of ventilator-induced lung injury in this rat model.


Subject(s)
Lung Diseases/etiology , Positive-Pressure Respiration/adverse effects , Animals , Biomarkers , Blood/metabolism , Bronchoalveolar Lavage Fluid/chemistry , Electrophoresis, Polyacrylamide Gel , Lung/metabolism , Lung/pathology , Lung Diseases/metabolism , Lung Diseases/pathology , Male , Rats , Rats, Sprague-Dawley , Time Factors
14.
Clin Chest Med ; 17(1): 151-69, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8665787

ABSTRACT

Multiple mediators have been implicated in the interactions between the liver and the lungs in various disease states. The best characterized mediator of liver-lung interaction is alpha 1-antitrypsin. Several cytokines and mediators may be involved in the pathogenesis of the hepatopulmonary syndrome and in the cytokine cascades that are activated in systemic inflammatory states such as acute respiratory distress syndrome. Hepatocyte growth factor or scatter factor is a recently described peptide with a broad range of biologic effects that may mediate lung-liver interactions.


Subject(s)
Cytokines/metabolism , Growth Substances/metabolism , Inflammation Mediators/metabolism , Liver Diseases/metabolism , Lung Diseases/metabolism , Humans , Liver Diseases/complications , Liver Diseases/physiopathology , Lung Diseases/complications , Lung Diseases/physiopathology
15.
Am J Physiol ; 270(1 Pt 1): L101-9, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8772532

ABSTRACT

Increased production of endothelin-1 (ET-1) has been detected in lungs of fawn-hooded rats (FHR) with idiopathic pulmonary hypertension. Accelerated pulmonary artery (PA) smooth muscle cell (SMC) proliferation contributes to vascular remodeling in these rats. We hypothesized that PA SMC would be an important site of enhanced ET-1 expression in FHR lung, that these SMC would have increased growth compared with cells from a normotensive strain, and that this locally produced ET-1 would contribute to the increased growth of these cells. We found that isolated FHR PASMC overexpressed preproET-1 mRNA and produced more ET-1 peptide compared with cells from normotensive Sprague-Dawley control rats (SDR). PA SMC from FHR had increased growth compared with control cells under conditions of serum withdrawal (0.1%), submaximal serum stimulation (0.3%; a condition previously found to be required for detection of growth in response to the comitogen, ET-1), and maximal serum stimulation (10%). Enhanced growth of FHR PA SMC in the presence of 0.3% serum, but not under the other test conditions, was inhibited by the ETA receptor antagonist, BQ-123. In summary, PA SMC from rats with idiopathic pulmonary hypertension overproduce ET-1. This overproduction contributes to the enhanced growth of FHR PA SMC in the presence of 0.3% serum. These cells also possess other unique growth characteristics that are independent of ET-1. Together, these ET-1-dependent and -independent growth properties likely contribute to the hyperplasia of FHR PA SMC found in vivo.


Subject(s)
Endothelin-1/metabolism , Hypertension, Pulmonary/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , Animals , Blood Physiological Phenomena , Cell Division , Endothelins/genetics , Female , Hypertension, Pulmonary/pathology , Protein Precursors/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred Strains/genetics , Rats, Sprague-Dawley , Thymidine/metabolism
16.
Respiration ; 63(6): 346-51, 1996.
Article in English | MEDLINE | ID: mdl-8933652

ABSTRACT

We investigated the influence of low oxygen ventilation, air-bubble infusion into the pulmonary artery and their synergistic effect on pulmonary hemodynamics and microvascular permeability in isolated perfused rat lungs. Pulmonary arterial pressure was significantly increased by 70 min of ventilation with 3% O2 (hypoxia, group H); by 0.2-ml air-bubble infusion (pulmonary air embolism, group AE), and by 0.2-ml air-bubble infusion and 70 min of 3% O2 ventilation (hypoxia and pulmonary air embolism, group H & AE) compared with that of a control group (0.2 ml saline infusion, group C). Neither total (TPR) nor arterial (Ra) pulmonary vascular resistance in group H showed any difference compared to control values. TPR and Ra in groups AE and H & AE were significantly higher than those in group C. However, there was no significant difference in TPR or Ra between groups AE and H & AE. The pulmonary capillary fluid filtration coefficient, dry lung to wet lung weight ratio and white blood cell count in the perfusate of group H were not changed, while those of the groups AE and H & AE were significantly increased compared to those of controls. However, there was no significant difference in these values between groups AE and H & AE. Since hypoxia did not damage isolated perfused rat lungs, as determined by hemodynamics and permeability, nor enhance lung injury caused by air embolism, it was suggested that air embolism contributed more to high-altitude lung injury than low oxygen.


Subject(s)
Embolism, Air/physiopathology , Hypoxia/physiopathology , Lung/physiopathology , Pulmonary Embolism/physiopathology , Analysis of Variance , Animals , Capillary Permeability , Embolism, Air/complications , Hemodynamics , Hypertension, Pulmonary/etiology , Hypoxia/complications , Lung/anatomy & histology , Male , Organ Size , Pulmonary Embolism/complications , Rats , Rats, Sprague-Dawley
17.
Toxicol Pathol ; 23(6): 689-95, 1995.
Article in English | MEDLINE | ID: mdl-8772254

ABSTRACT

Reactive oxygen species, especially iron-catalyzed hydroxyl radicals (.OH) are implicated in the pathogenesis of asbestos-induced pulmonary toxicity. We previously demonstrated that phytic acid, an iron chelator, reduces amosite asbestos-induced .OH generation, DNA strand break formation, and injury to cultured pulmonary epithelial cells (268[1995, Am. J. Physiol.(Lung Cell. Mol. Physiol.) 12:L471-480]). To determine whether phytic acid diminishes pulmonary inflammation and fibrosis in rats after a single intratracheal (it) instillation of amosite asbestos, Sprague-Dawley rats were given either saline (1 ml), amosite asbestos (5 mg; 1 ml saline), or amosite treated with phytic acid (500 microM) for 24 hr and then instilled. At various times after asbestos exposure, the rats were euthanized and the lungs were lavaged and examined histologically. A fibrosis score was determined from trichrome-stained specimens. As compared to controls, asbestos elicited a significant pulmonary inflammatory response, as evidence by an increase (approximately 2-fold) in bronchoalveolar lavage (BAL) cell counts at 1 wk and the percentage of BAL neutrophils (PMNs) and giant cells at 2 wk (0.1 vs 6.5% and 1.3 vs 6.1%, respectively; p < 0.05). Asbestos significantly increased the fibrosis score at 2 wk (0 +/- 0 vs 5 +/- 1; p < 0.05). The inflammatory and fibrotic changes were, as expected, observed in the respiratory bronchioles and terminal alveolar duct bifurcations. The increased percentage of BAl PMNs and giant cells persisted at 4 wk, as did the fibrotic changes. Compared to asbestos alone, phytic acid-treated asbestos elicited significantly less BAL PMNs (6.5 vs 1.0%; p < 0.05) and giant cells (6.1 vs 0.2%; p < 0.05) and caused significantly less fibrosis (5 vs 0.8; p < 0.05) 2 wk after exposure. We conclude that asbestos causes pulmonary inflammation and fibrosis in rats after it instillation and that phytic acid reduces these effects. These data support the role of iron-catalyzed free radicals in causing pulmonary toxicity from asbestos in vivo.


Subject(s)
Asbestos, Amosite/toxicity , Asbestosis/pathology , Asbestosis/prevention & control , Iron Chelating Agents/therapeutic use , Phytic Acid/therapeutic use , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/prevention & control , Animals , Asbestos, Amosite/administration & dosage , Asbestosis/metabolism , Drug Administration Schedule , Free Radicals/metabolism , Free Radicals/toxicity , Intubation, Intratracheal , Male , Pulmonary Fibrosis/metabolism , Rats , Rats, Sprague-Dawley
18.
J Clin Invest ; 96(4): 2026-33, 1995 Oct.
Article in English | MEDLINE | ID: mdl-7560096

ABSTRACT

Alveolar type II cell proliferation occurs after many forms of lung injury and is thought to play a critical role in alveolar epithelial repair. Keratinocyte growth factor/fibroblast growth factor 7 (KGF) has been shown to promote alveolar type II cell growth in primary culture and alveolar epithelial hyperplasia in vivo. In this study, we used immunohistochemical analysis to determine the intrapulmonary distribution and cellular localization of recombinant human KGF (rhKGF) instilled into the trachea of rats. 6 h after administration, immunoreactive KGF was observed within the lung parenchyma and along alveolar epithelial cell membranes. By 18-24 h, KGF was detected intracellularly in alveolar epithelial cells and intraalveolar macrophages. Immunoreactive KGF was not demonstrable 48 h after delivery or in lung sections from PBS-treated animals. Intratracheal instillation of 5 mg/kg rhKGF stimulated a marked, time-dependent increase in the alveolar type II cell specific labeling index to a maximum level of 33 +/- 3% 48 h after rhKGF administration compared with 1.3 +/- 0.3% after PBS instillation. In addition, this increase in type II cell proliferation in vivo was documented by flow cytometric analysis of isolated type II cells which revealed a nearly fivefold increase in the proportion of cells traversing through the S and G2/M phases of the cell cycle. To test the hypothesis that KGFs effects on type II cells in vivo might affect the response to lung injury, rats were treated with rhKGF and exposed to hyperoxia. Animals that received 1 or 5 mg/kg rhKGF exhibited dramatically reduced mortality (P < 0.001, for both doses). Survival for animals treated with 0.1 mg/kg rhKGF was not significantly different from either untreated rats or animals treated with heat-denatured rhKGF. The lungs of rhKGF-treated animals that survived hyperoxia exposure had minimal hemorrhage and no exudate within the intraalveolar space. These experiments established that intratracheal administration of rhKGF stimulated alveolar type II cell proliferation in vivo and reduced hyperoxia-induced lung injury in rats. Directed delivery of KGF to the lungs may provide a therapeutic strategy to preserve or restore the alveolar epithelium during exposure to hyperoxia or other injurious agents.


Subject(s)
Fibroblast Growth Factors , Growth Substances/pharmacology , Oxygen/toxicity , Pulmonary Alveoli/drug effects , Animals , Cell Division/drug effects , Fibroblast Growth Factor 10 , Fibroblast Growth Factor 7 , Growth Substances/administration & dosage , Humans , Male , Pulmonary Alveoli/cytology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Superoxide Dismutase/metabolism , Trachea/drug effects
19.
Compr Ther ; 20(5): 289-93, 1994.
Article in English | MEDLINE | ID: mdl-8045086

ABSTRACT

IPF is one of the most common causes of ILD of unknown etiology. Although the pathogenesis of IPF is not well understood, the clinical, radiographic, and physiologic findings in this disease have been clearly described. Most patients are middle aged and present with progressive breathlessness and dyspnea on exertion. The most sensitive chest imaging technique is HRCT. Pulmonary function testing often reveals restriction and hypoxemia at rest or with exertion. Therapy for IPF usually involves corticosteroids or cytotoxic agents, such as cyclophosphamide or azathioprine, to decrease pulmonary inflammation and retard the fibrotic process. Unfortunately, only 20-30% of patients objectively respond to treatment. Lung transplantation offers the only potential for the restoration of pulmonary function.


Subject(s)
Pulmonary Fibrosis/therapy , Female , Humans , Male , Middle Aged , Pulmonary Fibrosis/diagnostic imaging , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/physiopathology , Radiography
20.
J Clin Invest ; 92(2): 969-77, 1993 Aug.
Article in English | MEDLINE | ID: mdl-7688769

ABSTRACT

Epithelial-mesenchymal interactions mediate aspects of normal lung growth and development and are important in the restoration of normal alveolar architecture after lung injury. To determine if fibroblasts are a source of soluble growth factors for alveolar type II cells, we investigated the effect of fibroblast-conditioned medium (CM) on alveolar type II cell DNA synthesis. Serum-free CM from confluent adult human lung fibroblasts was concentrated fivefold by lyophilization. Type II cells were isolated from adult rats by elastase dissociation and incubated with [3H]thymidine and varying dilutions of concentrated CM and serum from day 1 to 3 of culture. Stimulation of type II cell DNA synthesis by fibroblast-CM was maximal after 48 h of conditioning and required the presence of serum. The activity of the CM was eliminated by boiling and by treatment with trypsin, pepsin, or dithiothreitol and was additive with saturating concentrations of acidic fibroblast growth factor, epidermal growth factor, and insulin. The growth factor activity bound to heparin-Sepharose and was eluted with 0.6 and 1.0 M NaCl. Neutralizing antibody studies demonstrated that the primary mitogens isolated in the 0.6 and 1.0 M NaCl fractions were keratinocyte growth factor (KGF, fibroblast growth factor 7) and hepatocyte growth factor/scatter factor (HGF/SF), respectively. HGF/SF was demonstrated in the crude CM and KGF was detected in the 0.6 M NaCl eluent by immunoblotting. Northern blot analysis confirmed that the lung fibroblasts expressed both KGF and HGF/SF transcripts. Human recombinant KGF and HGF/SF induced a concentration- and serum-dependent increase in rat alveolar type II cell DNA synthesis. We conclude that adult human lung fibroblasts produce at least two soluble heparin-binding growth factors, KGF and HGF/SF, which promote DNA synthesis and proliferation of rat alveolar type II cells in primary culture. KGF and HGF/SF may be important stimuli for alveolar type II cell proliferation during lung growth and after lung injury.


Subject(s)
Fibroblast Growth Factors , Fibroblasts/physiology , Growth Substances/metabolism , Growth Substances/pharmacology , Hepatocyte Growth Factor/metabolism , Lung/physiology , Pulmonary Alveoli/metabolism , Animals , Cell Communication , Cell Division , Cells, Cultured , Culture Media, Conditioned , Culture Media, Serum-Free , DNA/biosynthesis , Epidermal Growth Factor/pharmacology , Fibroblast Growth Factor 1/pharmacology , Fibroblast Growth Factor 10 , Fibroblast Growth Factor 7 , Fibroblasts/cytology , Growth Substances/biosynthesis , Hepatocyte Growth Factor/biosynthesis , Hepatocyte Growth Factor/pharmacology , Humans , Insulin/pharmacology , Kinetics , Lung/cytology , Pulmonary Alveoli/cytology , Pulmonary Alveoli/drug effects , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...