Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 203
Filter
Add more filters










Publication year range
1.
Int Immunopharmacol ; 8(5): 679-87, 2008 May.
Article in English | MEDLINE | ID: mdl-18387510

ABSTRACT

Tumor necrosis factor-alpha (TNF-alpha) is a vital component of the inflammatory process and its aberrant over-expression has been linked to numerous disease states. New treatment strategies have sought to reduce circulating TNF-alpha, either with neutralizing anti-TNF-alpha binding proteins such as etanercept or via drugs that inhibit de novo TNF-alpha synthesis like pirfenidone. In the present study, we examined the effects of both classes of drugs on secreted and cell-associated TNF-alpha produced by THP-1 cells. All of the tested drugs significantly reduced secreted levels of bioactive TNF-alpha following stimulation with LPS as measured by bioassay. However, etanercept-treated cells had approximately six-fold higher levels of cell-associated TNF-alpha compared with that of the LPS-alone treatment group. Surprisingly, LPS+infliximab treated cells did not increase cell-associated TNF-alpha relative to the LPS-alone treatment. Pirfenidone significantly reduced both secreted and cell-associated TNF-alpha levels. These drug-related differences in cell-associated TNF-alpha may have broad implications in the future for the therapeutic uses of anti-TNF-alpha drugs in the management of TNF-alpha diseases.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents/pharmacology , Antibodies, Monoclonal/pharmacology , Immunoglobulin G/pharmacology , Pyridones/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Apoptosis/drug effects , Cell Survival/drug effects , Culture Media , Enzyme-Linked Immunosorbent Assay , Etanercept , Humans , Immunohistochemistry , Infliximab , Lipopolysaccharides/pharmacology , Receptors, Tumor Necrosis Factor , Tumor Necrosis Factor-alpha/biosynthesis
2.
J Vet Pharmacol Ther ; 27(5): 361-7, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15500575

ABSTRACT

Pirfenidone, a promising antifibrotic agent, was administered orally to dogs at 0, 40, 140, and 400 mg/kg/day. Serum was collected for pirfenidone assay at 0, 26 and 39 weeks of treatment. From the pirfenidone concentrations, pharmacokinetic parameters were determined for each dog at each treatment interval. The only significant differences because of gender were for concentration maxima. Unsurprisingly, there were many significant differences because of dose in concentration maximum and area under curve (AUC), and significant, positive linear correlations of both parameters with dose. There were few significant differences in time of maximal concentration and no correlation with dose. The mean +/- SE clearances were 1.99 +/- 0.13, 1.64 +/- 0.13 and 1.78 +/- 0.14 L/h/kg for doses of 40, 140, and 400 mg/kg, respectively, with no significant differences attributable to dose. There was an unexplained pattern in maximal concentration and AUC with regard to duration of treatment, with the parameters being highest at week 0, lowest at week 26, and intermediate at week 39. Clearance had the reverse pattern; time of maximal concentration had no pattern.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Dogs/metabolism , Pyridones/pharmacokinetics , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/blood , Area Under Curve , Drug Administration Schedule , Female , Male , Pyridones/administration & dosage , Pyridones/blood , Tumor Necrosis Factor-alpha/antagonists & inhibitors
3.
Res Commun Mol Pathol Pharmacol ; 115-116: 39-48, 2004.
Article in English | MEDLINE | ID: mdl-17564304

ABSTRACT

The antirheumatic effect of pirfenidone was compared with a positive control drug, oxyphenbutazone which is used in patients suffering from rheumatoid arthritis, in a double blind clinical trial in humans. The data collected in this pilot project revealed that pirfenidone was more effective (p < 0.025) than oxyphenbutazone in providing relief from arthritic pain. In addition, a greater number (p < 0.025) of patients reported favorable response to oral pirfenidone than oral oxyphenbutazone. However, there were no significant differences in the number of patients who dropped out from the trial and the number of patients who tolerated the drugs for 21 days of the trial between the pirfenidone and oxyphenbutazone groups. It was concluded from this pilot study that pirfenidone potentially offers a novel therapeutic modality for the management of rheumatoid arthritis with little or no adverse effects unlike steroidal and non-steroidal anti-inflammatory drugs which are frequently used for this chronic debilitating disease.


Subject(s)
Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Pyridones/therapeutic use , Adult , Antirheumatic Agents/administration & dosage , Antirheumatic Agents/adverse effects , Arthritis, Rheumatoid/physiopathology , Chi-Square Distribution , Double-Blind Method , Female , Humans , Male , Middle Aged , Oxyphenbutazone/administration & dosage , Oxyphenbutazone/therapeutic use , Patient Dropouts , Pilot Projects , Pyridones/administration & dosage , Pyridones/adverse effects , Treatment Outcome
4.
Nitric Oxide ; 7(2): 109-18, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12223180

ABSTRACT

The effects of aminoguanidine (AG), a specific inhibitor of inducible nitric oxide synthase, on the bleomycin (BL)-induced lung fibrosis was evaluated in mice. The animals were placed into five groups: saline (SA)-instilled drinking water (SA+H(2)O), saline-instilled drinking water containing 0.5%AG (SA+0.5%AG), BL-instilled drinking water (BL+H(2)O), BL-instilled drinking water containing 0.2%AG (BL+0.2%AG), and BL-instilled drinking water containing 0.5%AG (BL+0.5%AG). The mice had free access to H(2)O or H(2)O containing AG and lab chow ad lib 2 days prior to intratracheal (IT) instillation of BL (0.07U/mouse/100 microL) or an equivalent volume of sterile isotonic saline. The mice in the SA+0.5%AG group consumed the greatest amount of AG without any ill effects than the mice in any other group. There were no differences in any of the measured biochemical determinants between the SA+H(2)O and SA+0.5%AG control groups. The IT instillation of BL in the BL+H(2)O group caused significant increases in the lipid peroxidation, hydroxyproline content, and prolyl hydroxylase activity of lungs and influx of inflammatory cells in the broncheoalveolar lavage fluid (BALF) as compared to both control groups. The intake of aminoguanidine by mice in the BL+0.5%AG group caused significant reductions in the BL-induced increases in all measured biochemical indices of lung fibrosis without any effects on the influx of inflammatory cells in the BALF. In fact, AG in both BL-treated groups additionally increased the total cell counts in the BALF from mice in the BL+0.2%AG and BL+0.5%AG groups as compared to the BL+H(2)O group. Histopathological evaluation of the lungs revealed that the mice in the BL+0.5%AG group had markedly fewer fibrotic lesions than mice in the BL+H(2)O group. These results demonstrate that aminoguanidine minimizes the BL-induced lung fibrosis at both the biochemical and the morphological level and support our earlier hypothesis that the production of nitric oxide plays a significant role in the pathogenesis lung fibrosis caused by BL.


Subject(s)
Bleomycin/pharmacology , Guanidines/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy , Animals , Bronchoalveolar Lavage Fluid/cytology , Cell Count , Disease Models, Animal , Guanidines/administration & dosage , Guanidines/therapeutic use , Hydroxyproline/analysis , Lipid Peroxidation/drug effects , Lung/chemistry , Lung/drug effects , Lung/enzymology , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type II , Procollagen-Proline Dioxygenase/metabolism , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Time Factors , Weight Gain/drug effects
5.
Biochem Pharmacol ; 60(12): 1949-58, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-11108812

ABSTRACT

Integrins are a family of transmembrane glycoproteins that can interact with components of the extracellular matrix. The alpha4beta1 and alpha4beta7 integrins are heterodimeric leukocyte cell surface molecules critical to their cell and matrix adhesive interactions. Evidence for a central role for the alpha4 integrins in leukocyte pathophysiology in the lung is well documented. In this study, we tested the hypothesis that neutralizing antibody for integrin alpha4 (PS2) may reduce bleomycin (BL)-induced lung fibrosis in vivo. Male C57BL/6 mice were injected intratracheally with saline (SA) or BL (0.08 U/mouse) followed by intraperitoneal injection of SA, isotype control antibody (1E6), or PS2 (100 microg) three times a week. Twenty-one days after the intratracheal instillation, mice were killed for bronchoalveolar lavage (BAL), biochemical, histopathological, and immunohistological analyses. Treatment with PS2 significantly reduced BL-induced increases in lung lipid peroxidation and hydroxyproline content. Lung histopathology also showed reduced fibrotic lesions in the BL-treated lungs by treatment with PS2. BL-treated mouse lungs also showed induction of cells with the myofibroblast phenotype, as indicated by the increased expression of alpha-smooth muscle actin (alphaSMA), whereas treatment with PS2 minimized the BL-induced alphaSMA expression. Furthermore, treatment with PS2 reduced the BL-induced increase in the BAL total cell number, and attenuated the BL-induced increase in the BAL protein level. It is concluded that integrin alpha4 may play an important role in BL-induced pulmonary fibrosis, and the use of anti-alpha4 antibody offers therapeutic antifibrotic potential in vivo.


Subject(s)
Antigens, CD/immunology , Antigens, CD/metabolism , Pulmonary Fibrosis/prevention & control , Actins/analysis , Animals , Anti-Bacterial Agents , Antibodies/immunology , Bleomycin , Bronchoalveolar Lavage Fluid/cytology , Cell Differentiation , Hydroxyproline/metabolism , Immunohistochemistry , Integrin alpha4 , Lipid Peroxidation , Male , Mice , Mice, Inbred C57BL , Muscle, Smooth/metabolism , Procollagen-Proline Dioxygenase/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/enzymology , Pulmonary Fibrosis/pathology
6.
Inflammation ; 24(5): 477-91, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10921510

ABSTRACT

We have previously reported the antifibrotic effects of pirfenidone (PD) in the bleomycin (BL)-hamster model of lung fibrosis. Since the development of fibrosis is generally preceded by acute lung inflammation, the present study was conducted to find out if dietary intake of PD (0.5%) has any effects on BL-induced lung inflammation. In this regard, we evaluated the effects of PD on BL-induced increased pulmonary vascular permeability, increased influx of inflammatory cells and increased levels of TGF-beta in the bronchoalveolar lavage fluid (BALF). Hamsters were intratracheally (IT) instilled with saline (SA) or BL (5.5 units/kg/5 ml). The animals were fed the control diet (CD) or the same diet containing 0.5% PD 2 days prior to IT instillation and throughout the study. The bronchoalveolar lavage was carried out at different times after IT instillation. Lavage fluid was used for total and differential cell counts and BALF-supernatant for measurement of total protein and TGF-beta. IT instillation of BL caused significant increases in total cells, neutrophils, macrophages and lymphocytes and in the levels of total protein and TGF-beta in BALF from hamsters in the BL + CD groups as compared to the corresponding SA + CD control groups. In contrast, treatment with pirfenidone in general, suppressed the BL-induced increases in the levels of proteins and TGF-beta and in the influx of neutrophils, macrophages and lymphocytes in BALF at the early time points in BL + PD groups. Based on the data reported in this study, we conclude that the anti-inflammatory effects of pirfenidone as evident by suppressions of BL-induced increased pulmonary vascular permeability and increased influx of inflammatory cells in the lung contribute additionally to its inherent anti-fibrotic effect.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Bleomycin/toxicity , Pulmonary Fibrosis/drug therapy , Pyridones/therapeutic use , Administration, Oral , Animal Feed , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Bronchoalveolar Lavage Fluid/chemistry , Capillary Permeability/drug effects , Cricetinae , Inflammation , Macrophages, Alveolar/drug effects , Male , Mesocricetus , Neutrophil Infiltration/drug effects , Pulmonary Fibrosis/pathology , Pyridones/administration & dosage , Pyridones/pharmacology , Transforming Growth Factor beta/analysis
7.
Nitric Oxide ; 4(4): 399-411, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10944425

ABSTRACT

The effects of taurine (T) and niacin (N) on the influx of inflammatory cells and nitric oxide (NO) levels in bronchoalveolar lavage fluid (BALF) and expression of inducible NO synthase (iNOS) mRNA and iNOS protein in lungs were evaluated in the bleomycin (BL)-mouse model of lung fibrosis. Mice were placed into four groups: saline-instilled (SA) with a control diet (CD) (SA + CD); saline-instilled with TN (1% taurine in water + 2.5% (w/w) niacin in diet) (SA + TN); BL-instilled with CD (BL + CD); and BL-instilled with TN treatment (BL + TN). There was no difference in differential cell counts in BALF between the SA + CD and SA + TN control groups. Intratracheal instillation (IT) of BL (0.1 U/mouse) in mice stimulated an early influx of neutrophils followed by an increase in lymphocytes and macrophages in the BL + CD group. Taurine and niacin treatment significantly reduced the numbers of neutrophils, lymphocytes, and macrophages in the BL + TN group and caused significant reductions in BL-induced increases in the lung hydroxyproline content at 14 and 21 days in the BL + TN group. The mice in the SA + CD and SA + TN control groups had low levels of NO in BALF, whereas mice in the BL + CD group as compared to the SA + CD control group had elevated levels of NO from day 3 through day 21. Taurine and niacin treatment caused significant reductions in BL-induced increases in NO levels in BALF from mice in the BL + TN group at 7, 14, and 21 days as compared to the corresponding BL + CD group. The increases in NO levels in BALF from the BL + CD group were associated with elevated levels of iNOS gene expression and protein in the lungs. RT-PCR analysis of total RNA isolated from the lungs indicated that taurine and niacin treatment suppressed the BL-induced increases in iNOS message and iNOS protein. The ability of taurine and niacin to suppress the BL-induced increased production of NO secondary to decreases in iNOS mRNA and protein appears to be one of the mechanisms for their anti-inflammatory and antifibrotic effects.


Subject(s)
Bleomycin/pharmacology , Lung/metabolism , Niacin/pharmacology , Nitric Oxide Synthase/metabolism , Nitric Oxide/metabolism , Pulmonary Fibrosis/metabolism , Taurine/pharmacology , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Disease Models, Animal , Gene Expression Regulation, Enzymologic/drug effects , Hydroxyproline/analysis , Lung/chemistry , Lung/enzymology , Male , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology , Random Allocation , Reverse Transcriptase Polymerase Chain Reaction
8.
Arch Biochem Biophys ; 379(1): 109-18, 2000 Jul 01.
Article in English | MEDLINE | ID: mdl-10864448

ABSTRACT

The ryanodine-sensitive calcium channels, also called ryanodine receptors, are intracellular Ca(2+)-release channels that have been shown to bind the neutral plant alkaloid ryanodine with nanomolar affinity. The activity of the skeletal muscle (RyR1), cardiac muscle (RyR2), and brain (RyR3) ryanodine receptor isoforms have been shown to be highly regulated by physiological factors including pH, temperature, and ionic strength; endogenous compounds including Ca(2+), Mg(2+), and adenosine triphosphate (ATP); and pharmacological agents including caffeine, ruthenium red, and neomycin. RyR3 is reportedly expressed in diverse tissues including lung; however, specific [(3)H]ryanodine binding sites in mammalian lung tissue have not been characterized. In this study, hamster lung ryanodine binding proteins were shown to specifically bind [(3)H]ryanodine with an affinity similar to that of RyR isoforms found in other tissues and this binding was shown to be sensitive to Ca(2+) concentration, stimulation by caffeine and spermine, and inhibition by Mg(2+), ruthenium red, and neomycin. The solubilized, intact ryanodine binding protein from hamster lung demonstrated approximately the same 30S sedimentation coefficient as RyR1 and RyR2, but a putative ryanodine receptor subunit from hamster lung was not found to cross-react with antibodies specific for the three known isoforms. We conclude that the hamster lung ryanodine binding protein demonstrates sedimentation and binding characteristics that are similar to those of the known RyR isoforms, but may exhibit antigenic dissimilarity from the typical RyR isoforms found in muscle and brain.


Subject(s)
Lung/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Ryanodine/metabolism , Animals , Antibodies/immunology , Binding Sites , Caffeine/pharmacology , Calcium/pharmacology , Centrifugation, Density Gradient , Cricetinae , Hydrogen-Ion Concentration , Kinetics , Magnesium/pharmacology , Neomycin/pharmacology , Osmolar Concentration , Protein Binding/drug effects , Protein Isoforms/immunology , Ruthenium Red/pharmacology , Ryanodine Receptor Calcium Release Channel/immunology , Ryanodine Receptor Calcium Release Channel/isolation & purification , Spermine/pharmacology , Tritium
9.
J Pharmacol Exp Ther ; 293(1): 82-90, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10734156

ABSTRACT

The effects of taurine (T) and niacin (N) on bleomycin (BL)-induced increased production of tumor necrosis factor-alpha (TNF-alpha), interleukin (IL)-1alpha, IL-6, and transforming growth factor-beta (TGF-beta) levels in the bronchoalveolar lavage fluid (BALF), and increased collagen content and nuclear factor-kappaB (NF-kappaB) activation in the lungs were investigated in mice. The mice were intratracheally instilled with saline (SA) or BL (0.1 U/mouse/50 microliter) under ketamine and xylazine anesthesia. They had ad libitum access to diet containing 2.5% niacin (w/w) or the same control diet (CD) and water with and without taurine (1%) 3 days before intratracheal instillation and throughout the study. The mice were sacrificed at different times for collecting BALF and lungs, which were appropriately processed for various measurements. Treatment with taurine and niacin attenuated the BL-induced increases in proinflammatory cytokines such as IL-1alpha, TNF-alpha, IL-6, and TGF-beta in BALF and lung hydroxyproline content of the mice in BL + TN groups. Reverse transcription-polymerase chain reaction analysis of total RNA from whole lung was performed to assess the induction of TNF-alpha and IL-1 mRNAs as markers of NF-kappaB activation. The NF-kappaB DNA-binding activity in whole-lung extract was evaluated by electrophoretic mobility shift assay. This revealed a progressive increase in NF-kappaB activation and IkBalpha depletion in lungs from mice in BL + CD groups from day 1 through day 21 compared with the corresponding SA + CD control groups. Treatment with taurine and niacin generally inhibited the BL-induced increases in the nuclear localization of NF-kappaB and preserved IkappaBalpha protein in BL + TN groups. This may be one of the mechanisms for the antifibrotic effect of taurine and niacin.


Subject(s)
Anti-Bacterial Agents/antagonists & inhibitors , Bleomycin/antagonists & inhibitors , Down-Regulation/genetics , Lung Diseases/prevention & control , NF-kappa B/genetics , Niacin/pharmacology , Pulmonary Fibrosis/prevention & control , Taurine/pharmacology , Animals , Anti-Bacterial Agents/toxicity , Bleomycin/toxicity , Blotting, Western , Bronchoalveolar Lavage Fluid/cytology , Down-Regulation/drug effects , Hydroxyproline/metabolism , Interleukin-1/physiology , Interleukin-6/physiology , Lung Diseases/chemically induced , Lung Diseases/pathology , Male , Mice , Mice, Inbred C57BL , NF-kappa B/drug effects , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta/physiology , Tumor Necrosis Factor-alpha/physiology
11.
J Pharmacol Exp Ther ; 291(1): 367-73, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10490926

ABSTRACT

This study was undertaken to investigate whether treatment with the antifibrotic drug pirfenidone (PD) down-regulates the bleomycin (BL)-induced overexpression of transforming growth factor (TGF)-beta gene in the lungs. Hamsters were intratracheally instilled with SA or BL (6.5 U/kg/4 ml) under anesthesia. They were fed a diet containing 0.5% PD or the same control diet (CD) without the drug 2 days before and throughout the study. After the animals were sacrificed, their lungs were appropriately processed. The BL treatment elevated the total influx of inflammatory cells, including macrophages, by severalfold at different days in bronchoalveolar lavage fluid (BALF) from hamsters in BL + CD groups, relative to the corresponding SA + CD control groups. Treatment with PD significantly (P

Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Down-Regulation/drug effects , Pulmonary Fibrosis/metabolism , Pyridones/pharmacology , Transforming Growth Factor beta/biosynthesis , Animals , Bleomycin/pharmacology , Bronchoalveolar Lavage Fluid/cytology , Cricetinae , Lung/drug effects , Lung/metabolism , Macrophages/pathology , Male , Mesocricetus , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Transcription, Genetic , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
12.
Thorax ; 54(9): 805-12, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10456973

ABSTRACT

BACKGROUND: Transforming growth factor beta (TGF-beta) is a key mediator of collagen synthesis in the development of lung fibrosis. It has previously been shown that the administration of TGF-beta antibody and TGF-beta binding proteoglycan, decorin, reduced bleomycin (BL) induced lung fibrosis in animals. The present study was carried out to investigate whether intratracheal instillation of TGF-beta soluble receptor (TR) would minimise the BL induced lung fibrosis in hamsters. METHODS: The effect of a recombinant TR (TGFbetaRII) on the lung collagen accumulation was evaluated in a BL hamster model of pulmonary fibrosis. Animals were divided into four groups and intratracheally injected with saline or BL at 6.5 U/4 ml/kg followed by intratracheal instillation of phosphate buffered saline (PBS) or 4 nmol TR in 0.3 ml twice a week. Twenty days after the first intratracheal instillation the hamsters were killed for bronchoalveolar lavage (BAL) fluid, biochemical, and histopathological analyses. RESULTS: Treatment of hamsters with TR after intratracheal instillation of BL significantly reduced BL induced lung fibrosis as shown by decreases in the lung hydroxyproline level and prolyl hydroxylase activity, although they were still significantly higher than those of the saline control. Histopathological examination showed a considerable decrease in BL induced fibrotic lesions by TR treatment. However, TR did not prevent the BL induced increases in total cells and protein in the BAL fluid. CONCLUSIONS: These results suggest that TR has antifibrotic potential in vivo and may be useful in the treatment of fibrotic diseases where increased TGF-beta is associated with excess collagen accumulation.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Bleomycin/adverse effects , Pulmonary Fibrosis/prevention & control , Receptors, Transforming Growth Factor beta/therapeutic use , Animals , Bronchoalveolar Lavage Fluid , Cell Division/drug effects , Collagen , Cricetinae , Drug Evaluation, Preclinical , Hydroxyproline/pharmacology , Male , Mesocricetus , Mixed Function Oxygenases/pharmacology , Pulmonary Fibrosis/chemically induced , Receptors, Transforming Growth Factor beta/administration & dosage
13.
J Pharmacol Exp Ther ; 289(1): 211-8, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10087006

ABSTRACT

A time course study was carried out to elucidate the mechanisms for antifibrotic effect of pirfenidone (PD). Hamsters were intratracheally (i.t.) instilled with saline (SA) or bleomycin (BL) (7.5 units/kg/5 ml). The animals were fed a diet containing 0.5% PD or the same control diet (CD) without the drug 2 days before and throughout the study. The animals were sacrificed at various times after instillation. The lung hydroxyproline level in BL + CD groups was gradually increased and peaked at 21 days to 181% of the SA + CD control. The BL + PD-treated groups showed a gradual decrease in their lung collagen content, showing a maximum reduction of 40% at day 21. The lung malondialdehyde levels of the BL + CD groups were increased by several-fold of the corresponding SA + CD groups at various times. The lung prolyl hydroxylase (PH) activities in the BL + CD groups were also increased by several-fold of the corresponding SA + CD groups at these time points. The hamsters in the BL + PD showed a gradual decrease in the lung malondialdehyde levels from 10 to 21days compared with their corresponding BL + CD groups. Treatment with PD also reduced the lung PH activities in the BL + PD groups compared with the corresponding BL + CD groups. However, PD failed to manifest any direct inhibitory effect on PH activity in vitro. BL treatment increased the lung procollagen I and III gene expressions in the BL + CD groups by several-fold at varying times compared with the corresponding SA + CD, and treatment with PD in the BL + PD groups significantly down-regulated the BL-induced overexpression of these genes. Studies evaluating the regulation of these genes at the transcriptional level revealed PD significantly reduced the transcription of PC I at 14 days. Our results indicate that the antifibrotic effect of PD was partly due to suppression of the BL-induced inflammatory events and partly due to down-regulation of BL-induced overexpression of lung procollagen I and III genes.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Gene Expression Regulation/drug effects , Procollagen/genetics , Pulmonary Fibrosis/metabolism , Pyridones/pharmacology , Animals , Bleomycin , Cricetinae , Hydroxyproline/biosynthesis , In Situ Hybridization , In Vitro Techniques , Kinetics , Lipid Peroxidation/drug effects , Lung/metabolism , Male , Malondialdehyde/metabolism , Mesocricetus , Procollagen/biosynthesis , Procollagen-Proline Dioxygenase/metabolism , Pulmonary Fibrosis/chemically induced , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Transcription, Genetic
14.
Am J Physiol ; 276(2): L311-8, 1999 02.
Article in English | MEDLINE | ID: mdl-9950894

ABSTRACT

Pirfenidone (PD) is known for its antifibrotic effects in the bleomycin (BL) hamster model of lung fibrosis. We evaluated whether pretreatment of hamsters with PD could influence the effects of BL-induced overexpression of platelet-derived growth factor (PDGF)-A and PDGF-B genes and proteins in the same model of lung fibrosis. We demonstrate elevated levels of PDGF-A and PDGF-B mRNAs in bronchoalveolar lavage (BAL) cells from lungs of BL-treated compared with saline control hamsters by RT-PCR analysis. However, these levels were not altered in BAL cells obtained from BL-treated hamsters on diets containing 0.5% PD. Western blot analysis of BAL fluid for PDGF isoforms demonstrated that PD treatment inhibited the synthesis of both PDGF-A and PDGF-B isoforms. PD treatment also decreased the mitogenic activity in the BAL fluid from BL-treated hamster lungs. Taken together, these data provide evidence that the protective effects of PD against BL-induced lung fibrosis may be mediated by a reduction in PDGF isoforms produced by lung macrophages.


Subject(s)
Platelet-Derived Growth Factor/antagonists & inhibitors , Platelet-Derived Growth Factor/genetics , Protein Biosynthesis/physiology , Pulmonary Fibrosis/metabolism , Pyridones/pharmacology , 3T3 Cells , Amino Acid Sequence/genetics , Animals , Bleomycin , Bronchoalveolar Lavage Fluid/chemistry , Cricetinae , Isomerism , Kinetics , Lung/metabolism , Male , Mesocricetus , Mice , Molecular Sequence Data , Platelet-Derived Growth Factor/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , RNA, Messenger/metabolism
15.
J Environ Pathol Toxicol Oncol ; 18(3): 169-77, 1999.
Article in English | MEDLINE | ID: mdl-15281229

ABSTRACT

Pirfenidone is a newly developed antifibrotic drug that has been reported to retard the progression of pulmonary fibrosis induced by bleomycin and cyclophosphamide in animal models of lung fibrosis. The present in vitro studies using noncellular and cellular systems evaluated the antioxidant and cytotoxic properties of this drug. The Fenton reaction [Fe(II) + H2O2 --> Fe(III) + *OH + OH-] and the xanthine/xanthine oxidase system were used as sources of hydroxyl (*OH) and superoxide anion (O2*-) radicals, respectively. Electron spin resonance spin trapping was used for free radical detection and measurement. The reaction rate of pirfenidone with *OH was found to be 1.63 x 10(10) M(-1) s(-1), which is comparable to several well-established antioxidants, such as ascorbate, glutathione, cysteine, azide, and lipoic acid. Compared to *OH radicals, the O2*- scavenging was less efficient 42.36 M(-1) s(-1) with pirfenidone. Pirfenidone was also effective in inhibiting zymosan-stimulated chemiluminescence. In a noncellular model of lipid peroxidation, pirfenidone inhibited crystalline silica-induced lipid peroxidation. The inhibition of crystalline silica-induced cytotoxic reactions and lipid peroxidation combined with the efficient antioxidant properties of pirfenidone indicate that this agent may express its antifibrotic effects partly through its ability to scavenge reactive oxygen species.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antioxidants/pharmacology , Macrophages, Alveolar/drug effects , Pyridones/pharmacology , Reactive Oxygen Species/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Antioxidants/metabolism , Cell Survival/drug effects , Dose-Response Relationship, Drug , Electron Spin Resonance Spectroscopy , Lipid Peroxidation/drug effects , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/pathology , Male , Pyridones/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/analysis , Specific Pathogen-Free Organisms , Spin Trapping
16.
Am J Respir Cell Mol Biol ; 18(3): 334-42, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9490651

ABSTRACT

We have reported that taurine (T) and niacin (N) inhibit the expression of procollagen type I and type III genes at the level of gene transcription in the bleomycin (BL) hamster model of lung fibrosis. In the present study, we have investigated the effects of TN in diet on the temporal expression of transforming growth factor-beta1 (TGF-beta1) mRNA and TGF-beta1 protein production in the same model of lung fibrosis to determine whether the decreased transcription of procollagen genes is associated with downregulation of TGF-beta1 mRNA. Our results demonstrate that expression of TGF-beta1 mRNA in lungs is increased in BL-treated hamsters in the BL + control diet (CD) group, compared to saline controls in the saline-instilled (SA) + CD group, by 3.5-, 2.5-, 4-, and 2-fold at 3, 7, 14, and 21 d, respectively, and TN treatment caused significant decreases in TGF-beta1 mRNA expression in BL-treated animals in the BL + TN group from Day 3 through Day 21. In addition, TN treatment also reduced TGF-beta1 protein in bronchoalveolar lavage fluid (BALF) from BL-treated animals in the BL + TN group. These decreases in TGF-beta1 mRNA and TGF-beta1 protein correlated with decreased lung collagen content in hamsters in the BL + TN group as demonstrated in our earlier study. To confirm that the TGF-beta1 activity observed in BALF is reflected at the transcriptional level, total RNA was isolated from lavaged cells. Reverse transcriptase-polymerase chain reaction analysis demonstrated maximal expression of TGF-beta1 mRNA transcripts in BL-treated lavaged cells from animals in the BL + CD group and only low levels were detected in both saline control groups, and in BL + TN-treated lavaged cells. Nuclear runoff analysis indicated that TN-mediated reduction of TGF-beta1 mRNA steady-state levels was a result of decreased gene transcription, suggesting a transcriptional downregulation mechanism. Our results indicate that the combined treatment with TN ameliorates BL-induced lung fibrosis, at least in part, via inhibition of TGF-beta1 mRNA expression.


Subject(s)
Niacin/pharmacology , Pulmonary Fibrosis/metabolism , Taurine/pharmacology , Transforming Growth Factor beta/biosynthesis , Animals , Bleomycin/pharmacology , Bronchoalveolar Lavage Fluid/cytology , Collagen/biosynthesis , Cricetinae , Diet , Disease Models, Animal , Gene Expression Regulation , Male , Mesocricetus , Pulmonary Fibrosis/chemically induced , Transforming Growth Factor beta/genetics
17.
Exp Lung Res ; 24(1): 119-32, 1998.
Article in English | MEDLINE | ID: mdl-9457473

ABSTRACT

Interstitial lung fibrosis (ILF) is a life-threatening disease which has no known drug for prevention and cure. In the present study, we evaluated the antifibrotic potential of pirfenidone (PD) (5-methyl-1-phenyl-2-(1H)-pyridone) in a three-dose bleomycin (BL)-hamster model of lung fibrosis. Hamsters were intratracheally (IT) instilled with three consecutive doses of bleomycin sulfate (2.5 U/kg/5mL, 2.0 U/kg/5mL, 1.5 U/kg/3.75 mL) or an equivalent volume of saline at weekly intervals. Hamsters were fed a diet after the second dose of BL containing 0.5% PD and hamsters in the control groups were fed the same diet without the drug. The four groups were saline-instilled fed control diet (SCD); saline-instilled fed the same diet containing PD (SPD); BL-instilled fed control diet (BCD); and BL-instilled fed the diet containing PD (BPD). Hamsters were sacrificed at 28 days after IT instillation of last dose of saline or BL and their lungs processed for various assays. Lung hydroxyproline, an index of fibrosis, in SCD, SPD, BCD and BPD were 830, 804, 1609, 1235 micrograms/lung, respectively. Lung prolyl hydroxylase activities in the SPD, BCD and BPD groups were 103%, 313%, 157% of the control SCD group (5.99 x 10(4) dpm/lung/30 min) respectively. Malondialdehyde equivalent levels and superoxide dismutase activity in the corresponding groups were 99, 79, 240 and 145 nmoles/lung and 412, 433, 538 and 410 units/lung respectively. Lung myeloperoxidase activities in the corresponding groups were 56%, 179%, and 116% of the control group (0.44 units/lung). It is concluded that PD is a novel antifibrotic drug that has therapeutic potential in arresting the progression of an ongoing fibrotic process in the lung.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Pulmonary Fibrosis/drug therapy , Pyridones/pharmacology , Animals , Bleomycin/toxicity , Body Weight/drug effects , Body Weight/physiology , Cricetinae , Diet , Disease Models, Animal , Hydroxyproline/metabolism , Lung/metabolism , Lung/pathology , Male , Malondialdehyde/metabolism , Mesocricetus , Peroxidase/metabolism , Procollagen-Proline Dioxygenase/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Superoxide Dismutase/metabolism
18.
Zentralbl Veterinarmed B ; 44(8): 449-60, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9394610

ABSTRACT

Three calves were sensitized with three doses of inactivated BTV-11 UC8 strain and then experimentally infected with the homologous virus. In addition, four BTV-seronegative heifers were also experimentally infected with BTV-11. Granulocyte rich fractions of peripheral blood leucocyte (PBL-GRF) cultures from BTV-sensitized/infected calves and from control unexposed cattle were exposed in vitro with BTV-11. Histamine, leukotriene (LT) C4 and prostaglandin (PG) D2 were assayed in supernatant fluids. Plasma histamine levels increased in BTV-infected heifers from 10.1 +/- 2 ng/ml at Day 0 to 23.1 +/- 6.6 ng/ml at Day 12 following virus exposure. In addition, in this experimental group the concentration of PGF2 alpha (mean 551.97 +/- 243.54 pg/ml) increased significantly (P < or = 0.05) compared with control cattle (mean 467.3 +/- 73.9 pg/ml). Bluetongue virus induced histamine and LTC4 release after in vitro infection of PBL-GRF. Release of LTC4 was significantly (P < or = 0.05) higher in PBL-GRF cultures from sensitized and control animals than in unexposed PBL-GRF cultures. In contrast to these results, PGD2 was not released after BTV infection of PBL-GRF in vitro. The histamine release caused by BTV was virus-specific and mainly mediated by an immunological reaction, since the release was significantly reduced by removal of cell surface immunoglobulins.


Subject(s)
Bluetongue/blood , Cattle Diseases/blood , Eicosanoids/blood , Histamine/blood , Leukocytes/metabolism , Animals , Bluetongue/metabolism , Bluetongue/pathology , Bluetongue virus/classification , Bluetongue virus/isolation & purification , Bluetongue virus/physiology , Cattle , Cattle Diseases/metabolism , Cattle Diseases/pathology , Cells, Cultured , Eicosanoids/metabolism , Female , Histamine/metabolism , Leukocytes/cytology , Leukocytes/virology
19.
Lipids ; 32(7): 759-67, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9252965

ABSTRACT

Although bleomycin (BLM), an antineoplastic drug, is used in the treatment of a variety of tumors, the mechanism(s) that contribute to its induced lung injury and fibrosis are not fully elucidated. Since alterations in the levels of certain fatty acid metabolites have been associated with BLM-induced lung injury, we tested the effects of dietary gamma-linolenic acid (GLA)-containing evening primrose oil on BLM-induced morphological alterations in the hamster lung, the marked elevation of tissue hydroxyproline (a marker for collagen synthesis), and elevated generation of arachidonic acid metabolites (marker of inflammatory mediators). Our data revealed that after 14 d of dietary GLA-containing oil (i) BLM-induced elevation of lung hydroxyproline was suppressed (P < 0.05), (ii) the marked BLM-induced elevation of lung leukotriene B4 (LTB4) (a marker of polymorphanuclear generation of proinflammatory LTB4) was significantly suppressed (P < 0.05). The decrease in LTB4 was accompanied by marked elevations (P < 0.05) of lung prostaglandin E1 (PGE1) and 15-hydroxyeicosatrienoic acid (15-HETrE), both with known antiinflammatory properties. Taken together, data from these studies suggest that dietary GLA-containing oil contributes to tissue elevation of PGE1 and 15-HETrE, which in vivo may attenuate lung inflammation and fibrosis.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Bleomycin/toxicity , Dietary Fats, Unsaturated/therapeutic use , Pulmonary Fibrosis/drug therapy , gamma-Linolenic Acid/therapeutic use , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cricetinae , Fatty Acids, Essential/pharmacology , Hydroxyproline/metabolism , Leukotriene B4/biosynthesis , Linoleic Acids , Lung/metabolism , Male , Mesocricetus , Oenothera biennis , Plant Oils , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/pathology
20.
J Pharmacol Exp Ther ; 281(3): 1047-58, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9190835

ABSTRACT

The different platelet-activating factor (PAF) receptor subtypes were identified in alveolar macrophages of hamster and guinea pig, based on the distinct characteristics of PAF-induced Ca++ responses and PAF antagonist potencies to these responses. PAF, but not lyso-PAF (inactive PAF), induced Ca++ release from intracellular Ca++ stores and the influx of extracellular Ca++ in a dose-dependent manner in both hamster and guinea pig alveolar macrophages. The potency for PAF-stimulated Ca++ release, however, was significantly different between the two species with EC50 values being 30- and 50-fold higher in Ca++ release and Ca++ influx responses in guinea pig than hamster, respectively. In addition, there were distinct differences in Ca++ influx characteristics between the two species; guinea pig macrophages exhibiting a rapid Ca++ extrusion and high sensitivity to thapsigargin (depletion of intracellular Ca++ store). The PAF-induced Ca++ response was sensitive to G-protein inhibitor pertussis toxin in hamster but not in guinea pig, suggesting the coupling of different types of G-proteins to PAF receptors. Pretreatment of macrophages with tyrosine kinase inhibitor, herbimycin A, caused a dose-dependent decrease in PAF-induced Ca++ response in guinea pig but surprisingly an increased response in hamster. These observations suggest the possibility of a dual mechanism, for G-protein and tyrosine kinase, in PAF-induced phospholipase C activation of macrophages from both species and thus Ca++ signaling in response to PAF-mediated receptor signal transduction cascade. The PAF-induced Ca++ response was desensitized by repetitive stimulation with PAF or pretreatment with protein kinase C activator, mitogen-activated protein kinase, which had a slightly greater potency in guinea pig than hamster. Importantly, three structurally distinct PAF antagonists, WEB2086, L659,989 and CL184005, blocked PAF-induced Ca++ responses in a dose-dependent manner with a markedly different potencies between the two species. The IC50 values for inhibiting PAF-induced Ca++ release were 2.5- (WEB2086), 650- (L659,989) and 120- (CL184005) fold less in hamster than in guinea pig. The relative potencies of these PAF antagonists in hamster macrophages were L659,989 > CL184005 > WEB2086. However, in guinea pig these three antagonists showed roughly the same potency. Interestingly, the opposite inhibitory effects of these antagonists on PAF-induced Ca++ influx were found in the two species, in which the IC50 were 15- (WEB2086) and 5- (CL184005) fold greater in hamster than in guinea pig but no difference in the IC50 value of L659,989 between the two species. Pretreatment of macrophages from both species with these antagonists had no effect on ATP-induced Ca++ response, suggesting that the antagonism is specific to PAF receptors. Based on our data, it was concluded that the alveolar macrophages isolated from the bronchoalveolar lavage of hamsters contain a distinct subtype PAF receptor that differs from that of guinea pigs in modulating a different signal transduction pathway.


Subject(s)
Calcium/metabolism , Macrophages/metabolism , Platelet Activating Factor/pharmacology , Platelet Membrane Glycoproteins/metabolism , Pulmonary Alveoli/metabolism , Receptors, Cell Surface , Receptors, G-Protein-Coupled , Animals , Cricetinae , Dose-Response Relationship, Drug , Guinea Pigs , Male , Platelet Membrane Glycoproteins/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...