Skip to main content
Erschienen in: Respiratory Research 1/2002

Open Access 01.12.2002 | Research

Altered prostanoid production by fibroblasts cultured from the lungs of human subjects with idiopathic pulmonary fibrosis

verfasst von: Roberto Cruz-Gervis, Arlene A Stecenko, Ryszard Dworski, Kirk B Lane, James E Loyd, Richard Pierson, Gayle King, Kenneth L Brigham

Erschienen in: Respiratory Research | Ausgabe 1/2002

Abstract

Background

Prostanoids are known to participate in the process of fibrogenesis. Because lung fibroblasts produce prostanoids and are believed to play a central role in the pathogenesis of idiopathic pulmonary fibrosis (IPF), we hypothesized that fibroblasts (HF) cultured from the lungs of patients with IPF (HF-IPF) have an altered balance between profibrotic (thromboxane [TX]A2) and antifibrotic (prostacyclin [PGI2]) prostaglandins (PGs) when compared with normal human lung fibroblasts (HF-NL).

Methods

We measured inducible cyclooxygenase (COX)-2 gene and protein expression, and a profile of prostanoids at baseline and after IL-1β stimulation.

Results

In both HF-IPF and HF-NL COX-2 expression was undetectable at baseline, but was significantly upregulated by IL-1β. PGE2 was the predominant COX product in IL-1β-stimulated cells with no significant difference between HF-IPF and HF-NL (28.35 [9.09–89.09] vs. 17.12 [8.58–29.33] ng/106 cells/30 min, respectively; P = 0.25). TXB2 (the stable metabolite of TXA2) production was significantly higher in IL-1β-stimulated HF-IPF compared to HF-NL (1.92 [1.27–2.57] vs. 0.61 [0.21–1.64] ng/106 cells/30 min, respectively; P = 0.007) and the ratio of PGI2 (as measured by its stable metabolite 6-keto-PGF) to TXB2 was significantly lower at baseline in HF-IPF (0.08 [0.04–0.52] vs. 0.12 [0.11–0.89] in HF-NL; P = 0.028) and with IL-1β stimulation (0.24 [0.05–1.53] vs. 1.08 [0.51–3.79] in HF-NL; P = 0.09).

Conclusion

An alteration in the balance of profibrotic and antifibrotic PGs in HF-IPF may play a role in the pathogeneses of IPF.
Abkürzungen
COX
cyclooxygenase
HF
human fibroblasts
NL
normal lungs
IPF
idiopathic pulmonary fibrosis
IFN
interferon
IL
interleukin
PG
prostaglandin
TX
thromboxane
PGI2
prostacyclin

Introduction

The concept of lung fibroblasts as effector cells in the pathogenesis of idiopathic pulmonary fibrosis (IPF) has recently evolved [1, 2]. Lung fibroblasts respond, in vitro, to inflammatory cytokines by producing growth factors and collagen, resulting in fibroblast proliferation and extracellular matrix deposition [24]. In addition, activated lung fibroblasts have been shown to produce large amounts of inflammatory cytokines and chemokines, in vitro, and hence, these cells may also have a role as effector–inflammatory cells [1, 2]. This capacity to produce both inflammatory and fibrotic factors could mean that phenotypically altered lung fibroblasts act simultaneously as effector and target cells, via paracrine and autocrine mechanisms, perpetuating the fibrotic process [2].
Prostanoids are important regulators of fibroblast function [59]. Prostaglandin (PG)E2 is thought to have antifibrotic properties in vitro, but also can have proinflammatory effects both in vivo and in vitro[1012]. Thromboxane (TX)A2 increases proliferation, and DNA and RNA synthesis in several cell types, including fibroblasts and smooth muscle like glomerular mesangial cells [1316]. Conversely, prostacyclin (PGI2) decreases smooth muscle cell proliferation and collagen synthesis [17, 18].
Many cell types, including lung fibroblasts, contain cyclooxygenase (COX), a proximal enzyme in prostanoid production, and can generate prostanoids [19]. It has been previously reported that IPF lung fibroblasts have decreased COX-2 expression compared to normal lung fibroblasts and, hence, have decreased PGE2 production [12, 20, 21]. Because of these findings and the fact that PGs are important fibroblast regulators, we sought to investigate whether abnormalities in COX-2 expression could be associated with an altered balance between profibrotic and antifibrotic PGs. We hypothesized that fibroblasts from the lungs of patients with IPF (HF-IPF) have an altered PG balance compared to normal lung fibroblasts (HF-NL). This phenotypical abnormality could be an important factor in the pathogenesis of IPF.

Materials and methods

Primary lung fibroblasts

Fibroblasts from the lungs of seven patients (6 males) with IPF (HF-IPF) were harvested: a) from excised lung at the time of lung transplantation; b) during an autopsy performed within 4 hours from death; or c) during open or transbronchial lung biopsies at the time of diagnosis. Of the seven patients, five subjects had advanced lung fibrosis and were receiving prednisone ± immunosuppressive agents; 2 patients were at an earlier stage of their disease and were not receiving immunosuppressive drugs. The mean age of the patients was 59 (range 43–71)]. HF-NL were cultured from five human lungs that arrived at our transplant center with the intention of being used for transplantation, but for various reasons could not be transplanted; these were macroscopically and microscopically normal. The cells were harvested and cultured as per the protocol described by Kumar et al.[22]. Briefly, lung tissue sections were finely cut with sterile scissors and incubated with serum free DMEM containing trypsin, DNAse and collagenase for 30 min. The procedure was repeated twice, and the supernatants were pooled and cultured in one 100 mm plate and incubated at 37°C in a 5% CO2 humidified atmosphere. Culture medium (DMEM with 5% fetal bovine serum [FBS] and penicillin/streptomycin) was replaced three times per week and fibroblasts were passed (1:2 split) at the time they became confluent. On passage 4 the cells were resuspended in 1 ml of DMEM with 20% FBS and DMSO and frozen at -70°C. For each experiment described below the cells were thawed, cultured and passed at least once. All the experiments were conducted with cells at passages 6 to 8.

Inducible cyclooxygenase (COX)-2 expression and eicosanoid production

COX-2 activity was determined by measuring PGE2, 6-keto-PGF(stable PGI2 metabolite), TXB2 (stable TXA2 metabolite), and PGF production in stimulated fibroblasts. HF-IPF (n = 7) and HF-NL (n = 5) were brought to >90% confluency in 100mm plates and then placed on serum free DMEM for 24 hours to render them quiescent. Fibroblasts were then incubated in DMEM with 5% FBS alone or in the same medium with IL-1β (2.5 ng/ml) for 24 hours. At the end of the incubation period the supernatant was aspirated and fresh media containing 30 μM of arachidonic acid was added to the plates. After 30 min of incubation the supernatant was collected and saved at -70°C for later eicosanoid analysis. The cells were then resuspended and divided into two aliquots, which were used for RNA and protein extractions, respectively. The above experiments were repeated in HF-IPF (n = 2) and HF-NL (n = 2) using serum free media conditions.
Prostanoids were measured by modified stable isotope dilution assays that used gas chromatography-negative ion-chemical ionization mass spectrometry as previously described [23]. Briefly, deuterium-labeled internal standards of PGE2, PGF, TXB2, and 6-keto-PGF were added to the supernatants with isopropyl alcohol. Isopropyl alcohol was removed by evaporation under nitrogen. After acidification to pH 3.5, the samples were extracted on preconditioned C-18 PrepSep columns (Fisher Scientific, Fair Lawn, NJ), and eluted with ethyl acetate. The extract was then converted to a pentafluorobenzyl ester by treatment with a mixture of 12.5% pentafluorobenzyl bromide in acetonitrile and disopropylethylamine at room temperature for 30 min. After evaporation of reagents, the residue was subjected to TLC plates, using the solvent system chloroform/ethanol (93:7, vol/vol) for PGF and TXB2, and ethyl acetate/methanol (93:2, vol/vol) for 6-keto-PGF and PGE2. Then PGF was converted to trimethylsilyl ether derivative by treatment with N,O-bis (trimethylsilyl) trifluoroacetamide and dimethylformamide. The methoxime derivative of TXB2, PGE2 and 6-keto-PGF was made by treatment with 2% methoxamine hydrochloride in pyridine at 70°C for 60 min, followed by evaporation of the pyridine, addition of water, and extraction with ethyl acetate. Derivatization was completed by formation of the trimethylsilyl derivatives by treatment with N,O-bis (trimethylsilyl) trifluoroacetamide and pyridine. Eicosanoids were quantified by measuring the ratio of the intensity of ions m/z 569/573 for PGF, m/z 614/618 for TXB2 and 6-keto-PGF, and m/z 524/528 for PGE2. An analytical blank for each of these products was determined by measuring the amount of nondeuterated material, detected after extracting and analyzing 2 ml of saline to which the deuterium-labeled internal standards had been added.

Western analysis

After washing with PBS at pH 7.4, pellets were lyzed in solubilization buffer containing 50 mM TRIS at pH 8, 1% Tween 20, 10 mM phenylmethylsulphonyl fluoride, diethyldithiocarbamic acid, leupeptin and pepstatin A (all from Sigma Chemical), sonicated, boiled with gel loading buffer (62.5 mM TRIS-HCl, at pH 6.8, 10% glycerol, 2% SDS, 5% β-mercaptoethanol, and bromophenol blue), and centrifuged at 15,000 x g for 10 min. Equal amounts of protein (70 to 100 μg) were separated by electrophoresis. SDS-PAGE was performed using a 7.5% separating gel with a 4% stacking gel. The resolved proteins were transferred electrophoretically to nitrocellulose membranes (Hybond-ECL, Amersham Corp.). After transfer, the filters were incubated overnight at 4°C in a blocking solution (20 mM TRIS base, 137 mM sodium chloride at pH 7.6, 5% powdered milk, 3% BSA), and incubated with primary polyclonal rabbit antibodies against COX-2 at a dilution 1:1000 (Cayman Chemical, Ann Arbor, MI), for 1 hour at room temperature. The filters were washed (TBS-0.1% Tween 20 at pH 7.6) and incubated with horseradish peroxidase linked secondary antibodies at a dilution 1:4000 (Amersham). After washing, the membranes were incubated with luminol based chemiluminescence reagent (DuPont NEN Research Products, Boston, MA).

Northern analysis

Cell pellets were lyzed and RNA extracted using the RNeasy method® (Qiagen), following the manufacturer's instructions. RNA was quantified by determining light absorbance at 260 nm and then fractioned by electrophoresis (10 μg per lane) on a 1% agarose MOPS/formaldehyde gel. The RNA was denatured prior to loading by incubating the RNA at 65°C for 10 min in a loading buffer comprising 1X MOPS, 50% formamide, 6.5% formaldehyde, 5% glycerol, 0.1 mM EDTA, 0.025% bromophenol blue, 0.025% xylene cyanol. The RNA was transferred by gravity-assisted capillary method with 6X SSC to nylon hybridization membrane, and then fixed to the membrane by UV crosslinking (Stratalinker 1200 μj/cm2). Prehybridization and hybridization were performed at 42°C and using Quick Hyb® (Stratagene) as hybridization solution. The COX-2 probe was random primed following the directions of the manufacturer (Megaprime®, Amersham/Pharmacia). The membrane was then washed at a final stringency of 0.2X SSC, 0.1% SDS, at 60°C for 30 min. The membrane was wrapped in plastic wrap and exposed to Kodak XR film at -70°C with intensifier screen overnight.

Statistical methods

All results are presented as medians with their range. Comparisons between HF-IPF and HF-NL were done using the Mann-Whitney test. A P value of <0.05 was considered significant.

Results

Baseline and stimulated COX-2 activity in HF-IPF and HF-NL

Unstimulated eicosanoid production was similar in both HF-IPF and HF-NL (Fig. 1, a-d). When fibroblasts were stimulated with IL-1β there was a significant and similar upregulation of PGE2 production in both HF-IPF and HF-NL (28.35 [range: 9.09–89.09] versus 17.12 [8.58–29.33] ng/106cells/30 min, respectively; P = 0.25; [Fig. 1a]). IL-1β-stimulated production of TXB2 (stable metabolite of the active TXA2), PGF, and 6-keto-PGF (stable metabolite of PGI2) increased modestly in every case, except TXB2 production by HF-NL, which decreased (0.75 [0.15–2.58] ng/106 cells/30 min at baseline versus 0.61 [0.21–1.64] ng/106 cells/30 min with IL-1β stimulation) (Fig. 1b). Results of PGE2 production were similar when experiments were conducted in serum free media conditions (results not shown).
IL-1β stimulated TXB2 production was significantly greater in HF-IPF (1.92 [1.27–2.57] ng/106 cells/30 min) than in HF-NL (0.61 [0.21–1.64] ng/106 cells/30 min; P = 0.007) (Fig. 1b); baseline TXB2 production was not significantly different between the two cell groups (1.73 [0.77–2.53] versus 0.75 [0.15–2.58] ng/106 cells/30 min, in HF-IPF and HF-NL, respectively; P = 0.17 [Fig. 1b]). Because PGI2 and TXA2 have opposing effects in vivo, we calculated the ratio of their metabolites (6-keto-PGF:TXB2) and found a significantly lower ratio in HF-IPF at baseline (0.08 [0.04–0.52] versus 0.12 [0.11–0.89] in HF-IPF and HF-NL, respectively; P = 0.028) and a similar trend under stimulated conditions (0.24 [0.05–1.53] versus 1.08 [0.51–3.79] in HF-IPF and HF-NL, respectively; P = 0.09 [Fig. 2]).

Baseline and stimulated COX-2 expression

Western blot in unstimulated fibroblasts showed no detectable COX-2 protein in either group of cells, while IL-1β significantly induced COX-2 to a similar degree in IPF and normal lung fibroblasts (Fig. 3). Northern blot showed minimal COX-2 mRNA in unstimulated cells and significant upregulation of COX-2 mRNA expression when stimulated with IL-1β in both HF-IPF and HF-NL (Fig. 4).

Discussion

Several factors modulate fibroblast proliferation and collagen production, including mitogenic cytokines (e.g., transforming growth factor β [TGFβ], platelet-derived growth factor [PDGF], basic fibroblast growth factor [bFGF]), eicosanoids (i.e., PGE2, TXB2, and PGI2), and antifibrogenic cytokines (e.g. IFN-γ) [13]. It is very likely that a complex interaction among these factors exists in the tissue repair process, and it is possible that pathologic fibrosis, as in IPF, results from phenotypical alterations in fibroblasts that affect the "normal" interaction of these factors.
Our results show that stimulation of primary cultures of human lung fibroblasts with the proximal cytokine IL-1β upregulates COX-2 protein and mRNA expression to a similar degree in normal and IPF fibroblasts. TXA2 production tended to be greater in IPF than in normal fibroblasts at baseline; when stimulated with IL-1β this difference became statistically significant. The ratio of PGI2 to TXA2 metabolites was lower in IPF fibroblasts at baseline and with IL-1β stimulation. The above results suggest that a decreased PGI2:TXA2 ratio could be a phenotypic alteration present in IPF fibroblasts, resulting in a loss of their capacity to autoregulate proliferation and extracellular matrix production.
The effects of PGs on cell proliferation and collagen production have been widely studied in different cell types [1317, 26]. TXA2 has been studied extensively because of its apparent role in atherosclerosis, due to its prothrombotic and mitogenic activities on vascular smooth muscle cells [15, 16]. These mitogenic effects are potentiated by growth factors [15, 16, 27, 28]. In vascular smooth muscle cells TXA2 stimulates synthesis of bFGF and increases the expression of the proto-oncogenes c-fos, c-myc, and egr-1, which are associated with entry into the cell growth cycle [15]. In addition, TXA2 increases proliferation of fibroblasts [13] and smooth muscle-like glomerular mesangial cells [14].
On the other hand, PGI2 decreases vascular smooth muscle cell proliferation and collagen and glycosaminoglycane synthesis, via activation of adenylyl cyclase and subsequent production of cAMP [17]. Betaprost, an analog of PGI2, decreases procollagen I and III mRNA expression in cardiac fibroblasts [18]. These effects may counteract the profibrotic effects seen with TXA2 and it is possible that an alteration of a "normal" physiologic balance between PGI2 and TXA2 could increase tendency towards fibrogenesis.
It is important to mention that our experiments were conducted at similar passage levels (passage 6 to 8) in both groups, since senescence of fibroblasts is associated with a shift from the biosynthesis of PGI2 to TXA2[24, 25]. It is possible that the difference seen in our study between HF-IPF and HF-NL could result from comparing fibroblasts of different ages. HF-IPF might have been harvested from fibrotic lesions where fibroblasts had previously undergone a greater number of cell divisions than HF-NL, obtained from nonfibrotic lungs. Although this is a possibility, the age-related shift in PG production has only been shown at very high cell passages and has not been documented in vivo.
We also found that both HF-IPF and HF-NL had similar PGE2 production at baseline, and a similar increase when stimulated with IL-1β. PGE2 can decrease fibroblast proliferation and collagen synthesis, and increase collagen degradation [58].
Recent reports suggesting decreased COX-2 expression and PGE2 production in IPF fibroblasts have received significant attention [12, 20, 21]. In our study we found that both COX-2 protein expression and PGE2 production were upregulated to a similar degree in IPF and normal lung fibroblasts. We believe that differences in methodology and patient selection may explain the discrepancies with other studies. Vancheri and collaborators [20] found that TNF-α-stimulated fibrotic lung fibroblasts had decreased COX-2 expression and PGE2 production, but they further showed that these findings were a result of decreased expression of TNF-α receptors. The latter finding would argue against a primary defect in COX-2 expression, since no other stimulus, other than TNF-α, was tested. In another study, Keerthisingam et al.[21] reported that fibrotic lung fibroblasts had decreased COX-2 expression and PGE2 production in response to TGFβ stimulation. This study differed from ours in that a different stimulus was used. Of significance is the fact that the COX-2 gene is known to be NF-κB dependent, and IL-1β, but not TGFβ, is a potent inducer of NF-κB activation. Hence, the pathway involved in the induction of the COX-2 gene by IL-1β and TGFβ may be different. Furthermore, a significant proportion of the fibroblasts used in the study by Keerthisingam et al.[21] were obtained from patients with systemic sclerosis, which makes their fibroblast population more heterogeneous.
Wilborn et al.[12] also reported a decreased production of PGE2 by IL-1β-stimulated IPF fibroblasts, due to decreased COX-2 expression [12]. There is a possibility that patient selection may have differed between the two studies. However, we feel certain that the diagnostic accuracy of our patient population was high, due to the fact that 5 out of a total of 7 IPF subjects included in our study underwent lung transplantation with confirmatory pathology results consistent with IPF. The other 2 subjects had biopsy-proven IPF. In addition, our results were similar when comparing lung fibroblasts obtained from 5 subjects with advanced stage IPF with those of 2 subjects at an earlier stage of their disease, who had received no therapy. Although the reasons for our different results are unclear, the fact that we found similar COX-2 expression and PGE2 production in normal and IPF lung fibroblasts suggests that loss of COX-2 expression is not a universal characteristic of fibroblasts cultured from the lungs of subjects with IPF.

Conclusion

We have found that fibroblasts cultured from normal and IPF human lungs have a significant and similar induction of the COX-2 enzyme when stimulated with IL-1β, but that IPF fibroblasts produced more thromboxane and had a significantly lower prostacyclin:thromboxane ratio. We hypothesize that the lower PGI2:TXA2 ratio seen in HF-IPF may be a phenotypic alteration that plays a role in the pathogenesis of IPF.

Acknowledgements

Supported by NIH grants KO8 HL04402 (Dr. Cruz-Gervis), RO1 HL63204 (Dr. Brigham), and training grant HL07123.
Literatur
2.
Zurück zum Zitat Tremblay GM, Jordana M, Gauldie J, Sarnstrand B: Fibroblasts as effector cells in fibrosis. In Pulmonary Fibrosis; Lung Biology in Health and Disease, (Edited by: Phan SH, Thrall RS). New York: Marcel Dekker 1995, 80:541–577. Tremblay GM, Jordana M, Gauldie J, Sarnstrand B: Fibroblasts as effector cells in fibrosis. In Pulmonary Fibrosis; Lung Biology in Health and Disease, (Edited by: Phan SH, Thrall RS). New York: Marcel Dekker 1995, 80:541–577.
3.
Zurück zum Zitat Crystal RG, Ferrans VJ, Basset F: Biologic basis of pulmonary fibrosis. In The Lung: Scientific Foundations. (Edited by: Crystal RG, West JB). New York: Raven Press 1991, 2031–2046. Crystal RG, Ferrans VJ, Basset F: Biologic basis of pulmonary fibrosis. In The Lung: Scientific Foundations. (Edited by: Crystal RG, West JB). New York: Raven Press 1991, 2031–2046.
4.
Zurück zum Zitat Strieter RM, Keane MP, Standiford TJ, Kunkel SL: Cytokine biology and the pathogenesis of interstitial lung disease. In Interstitial Lung Disease. (Edited by: Schwarz MI, King TE). Hamilton: B.C. Decker 1998, 181–205. Strieter RM, Keane MP, Standiford TJ, Kunkel SL: Cytokine biology and the pathogenesis of interstitial lung disease. In Interstitial Lung Disease. (Edited by: Schwarz MI, King TE). Hamilton: B.C. Decker 1998, 181–205.
5.
Zurück zum Zitat Elias J: Tumor necrosis factor interacts with interleukin-1 and interferons to inhibit fibroblast proliferation via prostaglandin-dependent and independent mechanisms. Am Rev Respir Dis 1988, 131:94–99. Elias J: Tumor necrosis factor interacts with interleukin-1 and interferons to inhibit fibroblast proliferation via prostaglandin-dependent and independent mechanisms. Am Rev Respir Dis 1988, 131:94–99.
6.
Zurück zum Zitat Goldstein R, Polgar P: The effect and interaction of bradykinin and prostaglandins on protein and collagen production by lung fibroblasts. J Biol Chem 1982, 257:8630–8633.PubMed Goldstein R, Polgar P: The effect and interaction of bradykinin and prostaglandins on protein and collagen production by lung fibroblasts. J Biol Chem 1982, 257:8630–8633.PubMed
7.
Zurück zum Zitat Korn J, Halushka P, Leroy E: Mononuclear cell modulation of connective tissue function: suppression of fibroblast growth by stimulation of endogenous prostaglandin production. J Clin Invest 1980, 65:543–554.CrossRefPubMedPubMedCentral Korn J, Halushka P, Leroy E: Mononuclear cell modulation of connective tissue function: suppression of fibroblast growth by stimulation of endogenous prostaglandin production. J Clin Invest 1980, 65:543–554.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Baum B, Moss S, Breul R, Crystal R: Effect of cyclic AMP on the intracellular degradation of newly synthesized collagen. J Biol Chem 1980, 255:2843–2847.PubMed Baum B, Moss S, Breul R, Crystal R: Effect of cyclic AMP on the intracellular degradation of newly synthesized collagen. J Biol Chem 1980, 255:2843–2847.PubMed
9.
Zurück zum Zitat Baud L, Perez J, Denis M, Ardaillou R: Modulation of fibroblast proliferation by sulfidopeptide leukotrienes: effect of indomethacin. J Immunol 1987, 138:1190–1195.PubMed Baud L, Perez J, Denis M, Ardaillou R: Modulation of fibroblast proliferation by sulfidopeptide leukotrienes: effect of indomethacin. J Immunol 1987, 138:1190–1195.PubMed
10.
Zurück zum Zitat Agro A, Langdon C, Smith F, Richards CD: Prostaglandin E 2 enhances interleukin 8 (IL-8) and IL-6 but inhibits GMCSF production by IL-1 stimulated human synovial fibroblasts in vitro. J Rheumatol 1996, 23:862–868.PubMed Agro A, Langdon C, Smith F, Richards CD: Prostaglandin E 2 enhances interleukin 8 (IL-8) and IL-6 but inhibits GMCSF production by IL-1 stimulated human synovial fibroblasts in vitro. J Rheumatol 1996, 23:862–868.PubMed
11.
Zurück zum Zitat Hinson RM, Williams JA, Shacter E: Elevated interleukin 6 is induced by prostaglandin E 2 in a murine model of inflammation: possible role of cyclooxygenase-2. Proc Natl Acad Sci USA 1996, 93:4885–4890.CrossRefPubMedPubMedCentral Hinson RM, Williams JA, Shacter E: Elevated interleukin 6 is induced by prostaglandin E 2 in a murine model of inflammation: possible role of cyclooxygenase-2. Proc Natl Acad Sci USA 1996, 93:4885–4890.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Wilborn J, Crofford L, Burdick MD, Kunkel SL, Streiter RM, Peters-Golden M: Cultured lung fibroblasts isolated from patients with idiopathic pulmonary fibrosis have diminished capacity to synthesize prostaglandin E 2 and to express cyclooxygenase-2. J Clin Invest 1995, 95:1861–1868.CrossRefPubMedPubMedCentral Wilborn J, Crofford L, Burdick MD, Kunkel SL, Streiter RM, Peters-Golden M: Cultured lung fibroblasts isolated from patients with idiopathic pulmonary fibrosis have diminished capacity to synthesize prostaglandin E 2 and to express cyclooxygenase-2. J Clin Invest 1995, 95:1861–1868.CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Murota SI, Morita I, Abe M: The effects of Thromboxane B 2 and 6-ketoprostaglandin F 1α on cultured fibroblasts. Biochim Biophys Acta 1977, 479:122–125.CrossRefPubMed Murota SI, Morita I, Abe M: The effects of Thromboxane B 2 and 6-ketoprostaglandin F on cultured fibroblasts. Biochim Biophys Acta 1977, 479:122–125.CrossRefPubMed
14.
Zurück zum Zitat Mene P, Abboud HE, Dunn MJ: Regulation of human mesangial cell growth in culture by thromboxane A 2 and prostacyclin. Kidney Int 1990, 38:232–239.CrossRefPubMed Mene P, Abboud HE, Dunn MJ: Regulation of human mesangial cell growth in culture by thromboxane A 2 and prostacyclin. Kidney Int 1990, 38:232–239.CrossRefPubMed
15.
Zurück zum Zitat Dorn GW II: Role of thromboxane A 2 in mitogenesis of vascular smooth muscle cells. Agents Actions 1997, 48:43–61. Dorn GW II: Role of thromboxane A 2 in mitogenesis of vascular smooth muscle cells. Agents Actions 1997, 48:43–61.
16.
Zurück zum Zitat Pakala R, Willerson JT, Benedict CR: Effect of serotonin, thromboxane A 2 , and specific receptor antagonists on vascular smooth cell proliferation. Circulation 1997, 96:2280–2286.CrossRefPubMed Pakala R, Willerson JT, Benedict CR: Effect of serotonin, thromboxane A 2 , and specific receptor antagonists on vascular smooth cell proliferation. Circulation 1997, 96:2280–2286.CrossRefPubMed
17.
Zurück zum Zitat Sinzinger H, Fitscha P, Kritz H: Antimitotic actions of vasodilatory prostaglandins-clinical aspects. Agents Actions 1997, 48:93–106. Sinzinger H, Fitscha P, Kritz H: Antimitotic actions of vasodilatory prostaglandins-clinical aspects. Agents Actions 1997, 48:93–106.
18.
Zurück zum Zitat Gallagher AM, Yu H, Printz MP: Bradykinin-induced reductions in collagen gene expression involve prostacyclin. Hypertension 1998, 32:84–88.CrossRefPubMed Gallagher AM, Yu H, Printz MP: Bradykinin-induced reductions in collagen gene expression involve prostacyclin. Hypertension 1998, 32:84–88.CrossRefPubMed
19.
Zurück zum Zitat Funk CD: Molecular biology in the eicosanoid field. Progress Nucleic Acid Res and Mol Biol 1993, 45:67–98.CrossRef Funk CD: Molecular biology in the eicosanoid field. Progress Nucleic Acid Res and Mol Biol 1993, 45:67–98.CrossRef
20.
Zurück zum Zitat Vancheri C, Sortino MA, Tomaselli V, Mastruzzo C, Condorelli F, Bellistri G, Pistorio MP, Canonico PL, Crimi N: Different expression of TNF-α receptors and prostaglandin E 2 production in normal and fibrotic lung fibroblasts: potential implications for the evolution of the inflammatory process. Am J Respir Cell Mol Biol 2000, 22:628–634.CrossRefPubMed Vancheri C, Sortino MA, Tomaselli V, Mastruzzo C, Condorelli F, Bellistri G, Pistorio MP, Canonico PL, Crimi N: Different expression of TNF-α receptors and prostaglandin E 2 production in normal and fibrotic lung fibroblasts: potential implications for the evolution of the inflammatory process. Am J Respir Cell Mol Biol 2000, 22:628–634.CrossRefPubMed
21.
Zurück zum Zitat Keerthisingam CB, Jenkins RG, Harrison NK, Hernandez-Rodriguez NA, Booth H, Laurent GJ, Hart SL, Foster ML, McAnulty RJ: Cyclooxygenase-2 deficiency results in a loss of the anti-proliferative response to transforming growth factor-β in human fibrotic lung fibroblasts and promotes bleomycin-induced pulmonary fibrosis in mice. Am J Pathol 2001, 158:1411–1422.CrossRefPubMedPubMedCentral Keerthisingam CB, Jenkins RG, Harrison NK, Hernandez-Rodriguez NA, Booth H, Laurent GJ, Hart SL, Foster ML, McAnulty RJ: Cyclooxygenase-2 deficiency results in a loss of the anti-proliferative response to transforming growth factor-β in human fibrotic lung fibroblasts and promotes bleomycin-induced pulmonary fibrosis in mice. Am J Pathol 2001, 158:1411–1422.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Kumar RK, Bennett RA, Brody AR: A homologue of platelet-derived growth factor produced by rat alveolar macrophages. FASEB J 1988, 2:2272–2277.PubMed Kumar RK, Bennett RA, Brody AR: A homologue of platelet-derived growth factor produced by rat alveolar macrophages. FASEB J 1988, 2:2272–2277.PubMed
23.
Zurück zum Zitat Dworski R, Sheller JR, Wickersham NE, Oates JA, Brigham KL, Roberts LJ II, Fitzgerald GA: Allergen-stimulated release of mediators into sheep bronchoalveolar lavage fluid; effect of cyclooxygenase inhibition. Am Rev Respir Dis 1989, 139:46–51.CrossRefPubMed Dworski R, Sheller JR, Wickersham NE, Oates JA, Brigham KL, Roberts LJ II, Fitzgerald GA: Allergen-stimulated release of mediators into sheep bronchoalveolar lavage fluid; effect of cyclooxygenase inhibition. Am Rev Respir Dis 1989, 139:46–51.CrossRefPubMed
24.
Zurück zum Zitat Fitzsimmons C, Proudfoot D, Bowyer DE: Monocyte prostaglandins inhibit procollagen secretion by human vascular smooth muscle cells: implications for plaque stability. Atherosclerosis 1999, 142:287–293.CrossRefPubMed Fitzsimmons C, Proudfoot D, Bowyer DE: Monocyte prostaglandins inhibit procollagen secretion by human vascular smooth muscle cells: implications for plaque stability. Atherosclerosis 1999, 142:287–293.CrossRefPubMed
25.
Zurück zum Zitat Zucker TP, Bonisch D, Muck S, Weber AA, Bretschneider E, Glusa E, Schror K: Thrombin-induced mitogenesis in coronary artery smooth muscle cells is potentiated by thromboxane A 2 and involves upregulation of thromboxane receptor mRNA. Circulation 1998, 97:589–595.CrossRefPubMed Zucker TP, Bonisch D, Muck S, Weber AA, Bretschneider E, Glusa E, Schror K: Thrombin-induced mitogenesis in coronary artery smooth muscle cells is potentiated by thromboxane A 2 and involves upregulation of thromboxane receptor mRNA. Circulation 1998, 97:589–595.CrossRefPubMed
26.
Zurück zum Zitat Grosser T, Zucker TP, Webber AA, Schulte K, Sachinidis A, Vetter H, Schror K: Thromboxane A 2 induces cell signaling but requires platelet-derived growth factor to act as a mitogen. Eur J Pharmacol 1997, 319:327–332.CrossRefPubMed Grosser T, Zucker TP, Webber AA, Schulte K, Sachinidis A, Vetter H, Schror K: Thromboxane A 2 induces cell signaling but requires platelet-derived growth factor to act as a mitogen. Eur J Pharmacol 1997, 319:327–332.CrossRefPubMed
27.
Zurück zum Zitat Polgar P, Taylor L: Alterations in prostaglandin synthesis during senescence of human lung fibroblasts. Mech Ageing Dev 1980, 12:305–310.CrossRefPubMed Polgar P, Taylor L: Alterations in prostaglandin synthesis during senescence of human lung fibroblasts. Mech Ageing Dev 1980, 12:305–310.CrossRefPubMed
28.
Zurück zum Zitat Murota S, Matsuoka K, Mitsui Y, Morita I, Kurata M: Age-related shift from the biosynthesis of prostacyclin to thromboxane A 2 and change in cyclic AMP production in response to prostacyclin in human diploid fibroblasts. Adv Prostaglandin Thromboxane Leukot Res 1983, 12:319–325.PubMed Murota S, Matsuoka K, Mitsui Y, Morita I, Kurata M: Age-related shift from the biosynthesis of prostacyclin to thromboxane A 2 and change in cyclic AMP production in response to prostacyclin in human diploid fibroblasts. Adv Prostaglandin Thromboxane Leukot Res 1983, 12:319–325.PubMed
Metadaten
Titel
Altered prostanoid production by fibroblasts cultured from the lungs of human subjects with idiopathic pulmonary fibrosis
verfasst von
Roberto Cruz-Gervis
Arlene A Stecenko
Ryszard Dworski
Kirk B Lane
James E Loyd
Richard Pierson
Gayle King
Kenneth L Brigham
Publikationsdatum
01.12.2002
Verlag
BioMed Central
Erschienen in
Respiratory Research / Ausgabe 1/2002
Elektronische ISSN: 1465-993X
DOI
https://doi.org/10.1186/rr166

Weitere Artikel der Ausgabe 1/2002

Respiratory Research 1/2002 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Reizdarmsyndrom: Diäten wirksamer als Medikamente

29.04.2024 Reizdarmsyndrom Nachrichten

Bei Reizdarmsyndrom scheinen Diäten, wie etwa die FODMAP-arme oder die kohlenhydratreduzierte Ernährung, effektiver als eine medikamentöse Therapie zu sein. Das hat eine Studie aus Schweden ergeben, die die drei Therapieoptionen im direkten Vergleich analysierte.

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.