Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 157
Filter
Add more filters










Publication year range
1.
Exp Mol Pathol ; 71(1): 13-33, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11502094

ABSTRACT

The bronchiolar-alveolar epithelium (BAE) is a primary target site for inhaled agents that cause lung injury. These cells, consequently, release a broad range of mediators that influence other cell populations, including interstitial lung fibroblasts that are central to the development of interstitial pulmonary fibrosis (IPF). A number of peptide growth factors (GF) have been postulated to be essential in the pathogenesis of IPF. We demonstrate here that primary populations of mouse BAE and mesenchymal cells, maintained in culture, synthesize four potent GF. These are platelet-derived growth factor isoforms (PDGF) A and B, transforming growth factor beta-1 (TGF-beta(1)), and tumor necrosis factor alpha (TNF-alpha). A mouse lung epithelial cell isolation technique pioneered in this laboratory has been used to purify the BAE cells to greater than 85% (80 +/- 5.6% alveolar type II and 9 +/- 2.3% Clara cells) in culture. Northern analysis, RNase protection assay, and immunocytochemistry (ICC) were used to establish mRNA and protein expression of the GF over time in the cultured BAE and mesenchymal cells. We show for the first time in these primary mouse lung cells that treatment of both cell types with TNF-alpha upregulates expression of TGF-beta(1). The four GF are produced by both epithelial and mesenchymal cells but with different temporal patterns. TGF-beta(1) is expressed constitutively by BAE and mesenchymal cells, whereas TNF-alpha expression wanes over time. The findings by ICC were consistent with levels of mRNA expression in both cell types. As genetically defined and altered mouse strains are becoming increasingly valuable for modeling lung disease, studying the gene expression patterns of target cells from these animals in vitro would be useful in sorting out the complex responses by individual cell types of the lung and the interactions among the multitude of mediators that are released during lung cell injury.


Subject(s)
Bronchi/cytology , Gene Expression Regulation/immunology , Platelet-Derived Growth Factor/genetics , Pulmonary Alveoli/cytology , Respiratory Mucosa/immunology , Transforming Growth Factor beta/genetics , Tumor Necrosis Factor-alpha/genetics , Animals , Bronchi/immunology , Cell Culture Techniques/methods , Cell Survival/drug effects , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/immunology , Gene Expression Regulation/drug effects , Immunohistochemistry , Kinetics , Male , Mesoderm/cytology , Mesoderm/immunology , Mice , Mice, Inbred C57BL , Organ Specificity , Protein Biosynthesis , Pulmonary Alveoli/immunology , RNA, Messenger/genetics , Respiratory Mucosa/cytology , Time Factors , Transcription, Genetic , Transforming Growth Factor beta/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
2.
Am J Respir Cell Mol Biol ; 25(1): 3-7, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11472967

ABSTRACT

Tumor necrosis factor-alpha receptor knockout (TNF-alphaRKO) mice have homozygous deletions of the genes that code for both the 55- and 75-kD receptors. The mice are protected from the fibrogenic effects of bleomycin, silica, and inhaled asbestos. The asbestos-exposed animals exhibit reduced expression of other peptide growth factors such as transforming growth factor (TGF)-alpha, platelet-derived growth factors, and TGF-beta. In normal animals, these and other cytokines are elaborated at high levels during the development of fibroproliferative lung disease, but there is little information available that has allowed investigators to establish the role of the individual growth factors in disease pathogenesis. Here, we show that overexpression of TGF-beta(1) by means of a replication-deficient adenovirus vector induces fibrogenesis in the lungs of the fibrogenic-resistant TNF-alphaRKO mice. The fibrogenic lesions developed in both the KO and background controls within 7 d, and both types of animals exhibited similar incorporation of bromodeoxyuridine. Interestingly, airway epithelial cell proliferation appeared to be suppressed, perhaps due to the presence of the TGF-beta(1), a well-known inhibitor of epithelial mitogenesis. Before these experiments, there was no information available that would provide a basis for predicting whether or not TGF-beta(1) expression induces fibroproliferative lung disease in fibrogenic-resistant TNF-alphaRKO mice, an increasingly popular animal model.


Subject(s)
Pulmonary Fibrosis/genetics , Receptors, Tumor Necrosis Factor/physiology , Transforming Growth Factor beta/metabolism , Adenoviridae/genetics , Animals , Bromodeoxyuridine , Genetic Vectors , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Tumor Necrosis Factor/genetics , Transduction, Genetic , Transforming Growth Factor beta/genetics
5.
J Environ Pathol Toxicol Oncol ; 20(2): 97-108, 2001.
Article in English | MEDLINE | ID: mdl-11394717

ABSTRACT

Inhalation of numerous fibrogenic agents causes interstitial pulmonary fibrosis (IPF) in humans and in a number of animal models. Several of these models provide evidence that certain peptide growth factors (GF) are playing a role in the disease process. Transforming growth factor beta 1 (TGF-beta1) is a potent inducer of extracellular matrix production by mesenchymal cells, and we have shown that this peptide is produced in the lung after asbestos exposure. We used in situ hybridization to demonstrate that the mRNA for TGF-beta1 is rapidly expressed post-exposure at sites of initial asbestos-induced lung injury in both rats and mice. The TGF-beta1 is expressed by bronchiolar-alveolar epithelial cells as well as by mesenchymal cells and lung macrophages in exposed animals. Normal rats and mice express little TGF-beta1, as we have demonstrated previously for PDGF-A and -B, TGF-alpha, and TNF-alpha. TGF-beta1 expression is accompanied by collagen and fibronectin production in asbestos-exposed animals. Most interesting, TGF-beta1 expression is largely absent in the lungs of TNF-alpha receptor knockout mice that fail to develop asbestos-induced IPE We have shown previously that the mRNAs and cognate peptides of PDGF-A and -B and TGF-alpha, but not TNF-alpha, are reduced in the fibrosis-resistant knockout mice. In this article, we show that TGF-beta1 is included in this group of cytokines, supporting the postulate that TNF-alpha is necessary for the expression of other, more downstream growth factors, and the consequent development of idiopathic pulmonary fibrosis (IPF).


Subject(s)
Asbestosis/metabolism , Lung/metabolism , Pulmonary Fibrosis/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Transforming Growth Factor beta/metabolism , Administration, Inhalation , Animals , Asbestos/toxicity , Asbestosis/pathology , Collagen/metabolism , Fibronectins/metabolism , Immunoenzyme Techniques , In Situ Hybridization , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Fibrosis/pathology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Tumor Necrosis Factor/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta1
7.
Am J Respir Crit Care Med ; 163(1): 244-52, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11208652

ABSTRACT

Murine exposure to silica is associated with enhanced tumor necrosis factor alpha (TNF-alpha) expression and matrix deposition. The regulation of TNF is mediated through TNF receptor (TNFR) activation of transcription factors. In the present work we have studied the importance of the individual TNFR in silica-induced lung inflammation and matrix deposition in mice. We studied RNA expression of TNF, alpha1(I) collagen, interstitial collagenase (MMP-13), and its inhibitor (TIMP-1) in the lungs of silica-treated mice. Furthermore, we correlated MMP-13/TIMP-1 RNA abundance with activation of the transcription factors AP-1 and NF-kappaB in the lungs of C57BL/6 mice, and of mice deficient in one of the two types of TNFR (p55(-/-) or p75(-/-)), exposed to silica (0.2 g/kg) or saline by intratracheal instillation. Animals were killed 28 d after exposure and lung hydroxyproline (HP), TNF, alpha1(I) collagen, MMP-13, and TIMP-1 RNA abundance was measured. AP-1 and NF-kappaB activation was studied by gel-shift assays. Compared with C57BL/6 mice, p55(-/-) and p75(-/-) mice significantly (*p < 0.05) decreased lung HP accumulation in response to silica. All murine strains enhanced TNF and alpha1(I) collagen mRNA in response to silica. Enhanced (p < 0.05) MMP-13 RNA expression was also observed in all murine strains in response to silica. Enhanced (p < 0.05) TIMP-1 RNA expression was observed in C57BL/6 mice, but not in p55(-/-) or p75(-/-) mice, in response to silica. NF-kappaB activation was observed in all murine strains, whereas AP-1 activation was observed only in C57BL/6 mice after silica treatment. These data suggest that TNFR deletion modifies MMP-13/ TIMP-1 expression in favor of matrix degradation.


Subject(s)
Collagenases/metabolism , Matrix Metalloproteinase 1/biosynthesis , Receptors, Tumor Necrosis Factor/deficiency , Silicosis/enzymology , Animals , Collagen/analysis , Female , Lung/chemistry , Matrix Metalloproteinase 13 , Mice , Mice, Inbred C57BL
8.
Exp Lung Res ; 27(8): 639-53, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11768716

ABSTRACT

Lung fibroblasts are activated to proliferate and produce connective tissue during the development of lung fibrosis. The 129 mouse strain does not develop asbestos-induced fibrogenesis, whereas several other inbred strains rapidly respond to inhaled fibers. Thus, in the experiments presented here, we have compared the responses of primary lung fibroblasts isolated from 129 and C57BL/6 mice. The 129 and C57BL/6 mouse lung fibroblasts (MLFs) proliferated similarly in 10% fetal bovine serum (FBS), but after quiescence, the 129 MLFs grew more slowly in serum and responded less to the BB isoform of platelet-derived growth factor. This is consistent with our finding that the mRNA for the PDGF-a receptor exhibits reduced expression by the 129 MLFs compared to those from C57BL/6 mice. Fibroblasts from the SJL mouse strain, from a C57BL/6-129 hybrid, and from the 3T3 cell line all proliferated more vigorously than MLFs from the 129 mice. In addition, the 129 MLFs exhibited reduced expression of alpha1 procollagen mRNA consequent to treatment with tumor necrosisfactor alpha. Based on these new findings, we suggest that the reduced fibrogenesis in asbestos-exposed 129 mice is due to an intrinsic difference in the ability of the lung fibroblasts to respond to peptide growth factors.


Subject(s)
Lung/drug effects , Platelet-Derived Growth Factor/pharmacology , 3T3 Cells , Animals , Biomarkers/analysis , Blotting, Northern , Cell Count , Cell Division/drug effects , Collagen Type I/genetics , Collagen Type I/metabolism , Dose-Response Relationship, Drug , Fibroblasts/cytology , Fibroblasts/drug effects , Immunohistochemistry , Lung/cytology , Mice , Mice, Inbred C57BL , Platelet-Derived Growth Factor/genetics , RNA, Messenger/metabolism , Receptor, Platelet-Derived Growth Factor alpha/biosynthesis , Receptor, Platelet-Derived Growth Factor alpha/genetics , Species Specificity , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects , Up-Regulation/genetics
9.
Environ Health Perspect ; 108 Suppl 4: 751-62, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10931794

ABSTRACT

Interstitial pulmonary fibrosis (IPF) is scarring of the lung caused by a variety of inhaled agents including mineral particles, organic dusts, and oxidant gases. The disease afflicts millions of individuals worldwide, and there are no effective therapeutic approaches. A major reason for this lack of useful treatments is that few of the molecular mechanisms of disease have been defined sufficiently to design appropriate targets for therapy. Our laboratory has focused on the molecular mechanisms through which three selected peptide growth factors could play a role in the development of IPF. Hundreds of growth factors and cytokines could be involved in the complex disease process. We are studying platelet-derived growth factor because it is the most potent mesenchymal cell mitogen yet described, transforming growth factor beta because it is a powerful inducer of extracellular matrix (scar tissue) components by mesenchymal cells, and tumor necrosis factor alpha because it is a pleiotropic cytokine that we and others have shown is essential for the development of IPF in animal models. This review describes some of the evidence from studies in humans, in animal models, and in vitro, that supports the growth factor hypothesis. The use of modern molecular and transgenic technologies could elucidate those targets that will allow effective therapeutic approaches.


Subject(s)
Air Pollutants/adverse effects , Growth Substances/metabolism , Inhalation Exposure/adverse effects , Lung Diseases, Interstitial/etiology , Animals , Humans , Lung Diseases, Interstitial/immunology , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/immunology
10.
Inhal Toxicol ; 12 Suppl 3: 245-50, 2000 Jan.
Article in English | MEDLINE | ID: mdl-26368622

ABSTRACT

It would be impossible to completely review this broad topic in the space and time allowed, lust as we must make choices when focusing on a research topic, this brief overview of particles and fibrosis deals with a small corner of our current thinking on the molecular mechanisms through which inhaled particles mediate fibroproliferative lung disease. At some point, there must be a common pathway of action because the fundamental process after any exposure involves fibroblast proliferation and production of extracellular matrix, that is, scar tissue. Thus, if we can understand how to control the fibroblast cell cycle and those genes that code for matrix components, it may be possible to develop effective therapeutic modalities for pulmonary fibrosis where none now exist. To control cell cycle and matrix genes that are expressed due to particle exposure, investigators have focused on certain peptide growth factors. The genes that code for these factors as well as the genes and oncogenes that are expressed when the peptides bind to their cognate receptors offer the best opportunities for understanding the molecular mechanisms of particle-induced lung fibrosis. As an example, mice with the genes knocked out for both the 55- and 75-kD receptors for tumor necrosis factor-alpha (TNF-α) are protected from the fibrogenic effects of inhaled asbestos and silica. Expression of other peptide growth factors in these animals is reduced, leading to a central working hypothesis, that TNF-α is a "master switch" that controls the expression of other more downstream factors that mediate components of the fibrogenic process. There is ample evidence of TNF-α expression in animal models and in populations of dust-exposed workers to support this postulate. In addition, it appears that transforming growth factor beta (TGF-ß) is the primary peptide that controls production of extracellular matrix. Blocking of TGF-ß expression in animal models prevents fibrosis, and the use of specific antisense RNA blocks both TGF-ß and collagen gene expression in primary lung fibroblasts. Conversely, an adenovirus vector that transduces expression of TGF-ß to the bronchiolar-alveolar epithelium induces diffuse fibrogenesis in rats and mice. Thus, particle-induced pulmonary fibrosis is a complex process that will be understood only after we dissect and elucidate the signal transduction pathways that control growth factor and matrix gene expression. The emerging technologies for developing transgenic and knockout mice, viral vectors for gene transduction, antisense approaches, and microarray gene analysis will finally allow us to accomplish these goals.

13.
Am J Pathol ; 154(6): 1763-75, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10362801

ABSTRACT

Because of its expression pattern and its potent effects on mesenchymal cells, platelet-derived growth factor (PDGF) has been implicated as an important factor in epithelial-mesenchymal cell interactions during normal lung development and in the pathogenesis of fibrotic lung disease. To further explore the role of PDGF in these processes, we have developed transgenic mice that express the PDGF-B gene from the lung-specific surfactant protein C (SPC) promoter. Adult SPC-PDGFB transgenic mice exhibited lung pathology characterized by enlarged airspaces, inflammation, and fibrosis. Emphysematous changes frequently occurred throughout the lung, but inflammation and fibrotic lesions were usually confined to focal areas. The severity of this phenotype varied significantly among individual mice within the same SPC-PDGFB transgenic lineage. A pathology similar to that observed in adult mice was noted in lungs from transgenic mice as young as 1 week of age. Neonatal transgenic mice exhibited enlarged saccules and thickened primary septa. Results of these studies indicated that overexpression of PDGF-B induced distinct abnormalities in the developing and adult lung and led to a complex phenotype that encompassed aspects of both emphysema and fibrotic lung disease.


Subject(s)
Lung Diseases/pathology , Platelet-Derived Growth Factor/biosynthesis , Platelet-Derived Growth Factor/genetics , Pulmonary Emphysema/pathology , Pulmonary Fibrosis/pathology , Age Factors , Animals , Animals, Newborn , Immunoenzyme Techniques , Immunohistochemistry , Inflammation/pathology , Lung/abnormalities , Mice , Mice, Inbred C57BL , Mice, Transgenic/genetics , Proteolipids/genetics , Pulmonary Surfactants/genetics , RNA, Messenger/metabolism , Transgenes/genetics
14.
Am J Respir Cell Mol Biol ; 20(4): 825-33, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10101016

ABSTRACT

We have investigated a potential role for tumor necrosis factor (TNF)-alpha and its two receptors (p55 and p75) in lung injury. We used several varieties of mice exposed endotracheally to two fibrogenic agents, silica (0.2 g/kg) and bleomycin (4 U/kg). The lungs were analyzed at 14 and 28 d after exposure to bleomycin or silica, respectively, for TNF and TNF receptor (TNFR) messenger RNA (mRNA), hydroxyproline content, and histopathology. Silica induced increased (over saline-treated animals) expression of TNF mRNA in double TNFR knockout (Ko), C57BL/6, BALB/c, and 129/J mice. In contrast, bleomycin increased expression in all but BALB/c mice, which are resistant to the fibrogenic effects of this drug. mRNA expression of both receptors was constitutively expressed in all of the normal murine strains. Silica upregulated expression of the p75 receptor, but not the p55 receptor, in the C57BL/6, BALB/c, and 129/J mice. In comparison, bleomycin had little effect on either receptor in the bleomycin-resistant BALB/c mice. Hydroxyproline content of the lungs after treatment followed this same pattern, with significant increases caused by silica in the C57BL/6, BALB/c, and 129/J mice, whereas bleomycin caused no apparent increases in the BALB/c mice. Even though silica and bleomycin induced increases in TNF in the TNFR Ko mice, the mice were protected from the fibrogenic effects of these agents. This study supports the concept that TNF is a central mediator of interstitial pulmonary fibrosis.


Subject(s)
Antigens, CD/genetics , Antigens, CD/metabolism , Bleomycin/toxicity , Lung/pathology , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Silicon Dioxide/toxicity , Transcription, Genetic/drug effects , Animals , Antigens, CD/physiology , Crosses, Genetic , Female , Gene Expression Regulation/drug effects , Lung/drug effects , Lung/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mutagenesis, Insertional , RNA, Messenger/genetics , Receptors, Tumor Necrosis Factor/physiology , Receptors, Tumor Necrosis Factor, Type I , Receptors, Tumor Necrosis Factor, Type II , Recombination, Genetic , Up-Regulation/drug effects
15.
Am J Pathol ; 154(3): 853-62, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10079263

ABSTRACT

Tumor necrosis factor (TNF)-alpha and transforming growth factor (TGF)-beta mRNA and protein expression and the degree of fibroproliferative response to inhaled asbestos fibers are clearly reduced in the 129 inbred mouse strain as compared with typical fibrogenesis observed in the C57BL/6 inbred strain. The C57BL/6 mice showed prominent lesions at bronchiolar-alveolar duct (BAD) junctions where asbestos fibers deposit and responding macrophages accumulate. The 129 mice, however, were generally indistinguishable from controls even though the numbers of asbestos fibers deposited in the lungs of all exposed animals were the same. Quantitative morphometry of H&E-stained lung sections comparing the C57BL/6 and 129 mice showed significantly less mean cross-sectional area of the BAD junctions in the 129 animals, apparent at both 48 hours and 4 weeks after exposure. In addition, fewer macrophages had accumulated at these sites in the 129 mice. Nuclear bromodeoxyuridine immunostaining demonstrated that the number of proliferating cells at first alveolar duct bifurcations and in adjacent terminal bronchioles was significantly reduced in the 129 strain compared with C57BL/6 mice at 48 hours after exposure (P < 0.01). TNF-alpha and TGF-beta1 gene expression, as measured by in situ hybridization, was reduced in the 129 mice at 48 hours after exposure, and expression of TNF-alpha and TGF-beta1 protein, as measured by immunohistochemistry, was similarly reduced or absent in the 129 animals. We postulate that the protection afforded the 129 mice is related to reduction of growth factor expression by the bronchiolar-alveolar epithelium and lung macrophages.


Subject(s)
Asbestos , Lung/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Count , Cell Division/physiology , Lung/pathology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron, Scanning , Pulmonary Fibrosis/pathology , RNA, Messenger/metabolism , Transforming Growth Factor beta/genetics , Tumor Necrosis Factor-alpha/genetics
16.
Am J Physiol ; 275(6): L1208-18, 1998 12.
Article in English | MEDLINE | ID: mdl-9843859

ABSTRACT

Apoptosis is considered to be a protective mechanism that limits lung injury. However, apoptosis might contribute to the inflammatory burden present in the injured lung. The exposure of mice to bleomycin (BLM) is a well-established model for the study of lung injury. BLM exposure induces DNA damage and enhances tumor necrosis factor (TNF)-alpha expression in the lung. To evaluate the importance of alveolar macrophage (AM) apoptosis in the pathogenesis of lung injury, we exposed BLM-sensitive (C57BL/6) and BLM-resistant (BALB/c) mice to BLM (120 mg/kg) and studied the induction of apoptosis [by light-microscopy changes (2, 8, 12, 24, 48, and 72 h) and annexin V uptake by flow cytometry (24 h)], the secretion of TNF-alpha (measured by ELISA), and the expression of p53 (by immunoblotting) in AM retrieved from these mice. BLM, but not vehicle, induced apoptosis in AM from both murine strains. The numbers of apoptotic AM were significantly greater (P < 0.001) in C57BL/6 mice (52.9%) compared with BALB/c mice (40.8%) as demonstrated by annexin V uptake. BLM induction of apoptosis in AM was preceded by an increased secretion of TNF-alpha in C57BL/6 but not in BALB/c mice. Furthermore, double TNF-alpha receptor-deficient mice, developed on a C57BL/6 background, demonstrated significantly (P < 0.001) lower numbers of apoptotic AM compared with C57BL/6 and BALB/c mice. BLM also enhanced p53 expression in AM from both murine strains. However, p53-deficient mice developed BLM-induced lung injury, exhibited similar lung cell proliferation (measured as proliferating cell nuclear antigen immunostaining), and accumulated similar amounts of lung hydroxyproline (65 +/- 6.9 microgram/lung) as did C57BL/6 (62 +/- 6.5 microgram/lung) mice. Therefore, AM apoptosis is occurring during BLM-induced lung injury in a manner that correlates with murine strain sensitivity to BLM. Furthermore, TNF-alpha secretion rather than p53 expression contributes to the difference in murine strain response to BLM.tumor necrosis factor; strain susceptibility


Subject(s)
Apoptosis/physiology , Bleomycin/pharmacology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/physiology , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/drug effects , Female , Hydroxyproline/metabolism , Immunoblotting , Immunohistochemistry , Macrophages, Alveolar/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Proliferating Cell Nuclear Antigen/metabolism , Receptors, Tumor Necrosis Factor/deficiency
17.
Am J Pathol ; 153(6): 1839-47, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9846974

ABSTRACT

We have demonstrated that C57BL/6-129 hybrid mice with genes for both the 55kd and 75kd receptors for TNF-alpha knocked out (TNF-alphaRKO) fail to develop fibroproliferative lesions after asbestos exposure. There is good evidence that TNF-alpha plays a major role in mediating interstitial pulmonary fibrosis. Our findings support this view and we present here new data obtained by in situ hybridization showing that expression of the genes coding for transforming growth factor alpha (TGF-alpha) and platelet-derived growth factor A-chain (PDGF-A) is reduced in the TNF-alphaRKO mice compared with control animals. In accordance with this observation, data on bromodeoxyuridine (BrdU) incorporation in the lungs of the TNF-alphaRKO mice show no increases over unexposed control animals. In contrast, wild-type control mice exposed to asbestos exhibit 15- to 20-fold increases in BrdU uptake and consequently develop fibrogenic lesions. Even though the levels of TNF-alpha gene expression and protein production were increased in the asbestos-exposed TNF-alphaRKO mice, the lack of receptor signaling protected the mice from developing fibroproliferative lesions. We agree with the view that TNF-alpha is essential for the development of interstitial pulmonary fibrosis and postulate that TNF-alpha mediates its effects through activation of other growth factors such as PDGF and TGF-alpha that control cell growth and matrix production.


Subject(s)
Asbestos/adverse effects , Pulmonary Fibrosis/pathology , Receptors, Tumor Necrosis Factor/genetics , Tumor Necrosis Factor-alpha/physiology , Animals , Cell Division , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet-Derived Growth Factor/metabolism , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/metabolism , RNA, Messenger/metabolism , Transforming Growth Factor alpha/metabolism , Tumor Necrosis Factor-alpha/metabolism
18.
Exp Lung Res ; 24(6): 721-43, 1998.
Article in English | MEDLINE | ID: mdl-9839161

ABSTRACT

Bleomycin (BLM) induction of lung fibrosis in mice is an established model to study the mechanism of pulmonary fibrosis. Cytokine secretion has been implicated as a fundamental component of the lung fibrotic process observed in response to BLM. Among the cytokines implicated in lung fibrosis, Tumor necrosis factor (TNF) alpha has been considered to play a fundamental role. In the present study, we characterized the cellular sources of TNF during BLM-induced lung injury and examined the importance of TNF receptors in this process. To characterize the expression of TNF, we utilized two strains of mice, one sensitive (C57BL/6) and one resistant (BALB/c) to BLM-induced lung injury. Mice received BLM (120 mg/kg total) or saline, as control, by multiple subcutaneous injections. BLM induced the development of inflammation in subpleural areas only in the lungs of BLM-sensitive mice. These subpleural areas were characterized by infiltration of CD68-positive macrophages and increased collagen deposition. BLM enhanced the expression of TNF mRNA in BLM-sensitive, but not in BLM-resistant, mice. In situ hybridization studies localized the expression of TNF in the areas of BLM-induced inflammation in 6% and 27% of macrophages at 14 and 21 days post BLM treatment. In addition to TNF, BLM exposure resulted in the upregulated expression of transforming growth factor (TGF)-beta 1, but not interleukin (IL)-1, mRNA in the lungs of both murine strains at 14 and 21 days. This upregulated expression of TGF-beta 1 mRNA was greater in the lungs of BLM-sensitive mice. In separate experiments, double TNF receptor knockout mice were exposed to BLM. These animals demonstrated an increased expression of TNF, but not TGF-beta 1, mRNA in response to BLM and did not exhibit histologic evidence of lung injury following BLM exposure. In summary, the upregulation of TNF mRNA in macrophages correlated with the appearance of inflammation following BLM exposure and was limited to the BLM-sensitive strain. Furthermore, in addition to the release of the TNF ligand, it appears that the presence of TNF receptors is necessary for the development of BLM-induced lung injury, and signaling through these receptors may contribute to the regulation of the TGF-beta 1 mRNA expression observed in response to bleomycin. These results provide further support for a role of macrophages and TNF in the induction of lung inflammation.


Subject(s)
Pulmonary Fibrosis/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Bleomycin/toxicity , DNA Primers/chemistry , Female , Genotype , Hydroxyproline/metabolism , Immunoenzyme Techniques , In Situ Hybridization , Interleukin-1/metabolism , Lung/drug effects , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mutation , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology , RNA, Messenger/biosynthesis , Receptors, Tumor Necrosis Factor/deficiency , Receptors, Tumor Necrosis Factor/genetics , Specific Pathogen-Free Organisms , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/genetics
20.
Am J Respir Crit Care Med ; 157(5 Pt 1): 1652-7, 1998 May.
Article in English | MEDLINE | ID: mdl-9603151

ABSTRACT

Platelet-derived growth factor-AA (PDGF-AA) and its matching alpha receptor (PDGF-R alpha) are upregulated in rat lung fibroblasts (RLFs) after exposure to chrysotile asbestos fibers in vitro, which results in asbestos-induced RLF proliferation. We now report our in vivo observations, which show an increase in the expression of PDGF-R alpha mRNA, but not PDGF-beta receptor mRNA, in asbestos-exposed rat lungs when compared with RNA from air-exposed (sham) and iron-exposed lungs. Western analysis of membrane preparations confirmed the observations on mRNA expression by demonstrating an increase in PDGF-R alpha peptide expression in the asbestos-exposed rat lungs, compared with that in the air-exposed lungs. Immunohistochemistry for the PDGF-R alpha was performed on air- and asbestos-exposed rat lungs and revealed a clear increase in staining within interstitial and subepithelial compartments in the exposed animals. These observations, along with our previous report demonstrating an increase in the PDGF-AA isoform expression immediately after asbestos-exposure, suggest a scenario in which a potent lung mesenchymal cell mitogen, PDGF-AA, and its alpha-receptor are upregulated prior to the development of a fibroproliferative lung lesion, and thus may play a central role in the pathogenesis of asbestos-induced lung fibrosis.


Subject(s)
Asbestosis/metabolism , Lung/metabolism , Pulmonary Fibrosis/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , Up-Regulation , Animals , Asbestosis/complications , Blotting, Northern , Blotting, Western , Immunohistochemistry , Male , Pulmonary Fibrosis/etiology , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Receptor, Platelet-Derived Growth Factor alpha , Receptor, Platelet-Derived Growth Factor beta
SELECTION OF CITATIONS
SEARCH DETAIL
...