Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Am J Respir Cell Mol Biol ; 50(4): 737-47, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24175906

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is characterized by progressive fibrotic destruction of normal lung architecture. Due to a lack of effective treatment options, new treatment approaches are needed. We previously identified transglutaminase (TG)2, a multifunctional protein expressed by human lung fibroblasts (HLFs), as a positive driver of fibrosis. TG2 catalyzes crosslinking of extracellular matrix proteins, enhances cell binding to fibronectin and integrin, and promotes fibronectin expression. We investigated whether the small electrophilic molecules 2-cyano-3,12-dioxoolean-1,9-dien-28-oic acid (CDDO) and 15-deoxy-delta-12,14-prostaglandin J2 (15d-PGJ2) inhibit the expression and profibrotic functions of TG2. CDDO and 15d-PGJ2 reduced expression of TG2 mRNA and protein in primary HLFs from control donors and donors with IPF. CDDO and 15d-PGJ2 also decreased the in vitro profibrotic effector functions of HLFs including collagen gel contraction and cell migration. The decrease in TG2 expression did not occur through activation of the peroxisome proliferator activated receptor γ or generation of reactive oxidative species. CDDO and 15d-PGJ2 inhibited the extracellular signal-regulated kinase pathway, resulting in the suppression of TG2 expression. This is the first study to show that small electrophilic compounds inhibit the expression and profibrotic effector functions of TG2, a key promoter of fibrosis. These studies identify new and important antifibrotic activities of these two small molecules, which could lead to new treatments for fibrotic lung disease.


Subject(s)
Enzyme Inhibitors/pharmacology , Idiopathic Pulmonary Fibrosis/enzymology , Lung/drug effects , Oleanolic Acid/analogs & derivatives , Prostaglandin D2/analogs & derivatives , Transglutaminases/antagonists & inhibitors , Case-Control Studies , Cell Movement/drug effects , Cell Shape/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Down-Regulation , Enzyme Inhibitors/chemistry , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/drug effects , Fibroblasts/enzymology , Fibroblasts/pathology , GTP-Binding Proteins , Humans , Idiopathic Pulmonary Fibrosis/pathology , Lung/enzymology , Lung/pathology , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System/drug effects , Molecular Targeted Therapy , Oleanolic Acid/chemistry , Oleanolic Acid/pharmacology , Phosphorylation , Prostaglandin D2/chemistry , Prostaglandin D2/pharmacology , Protein Glutamine gamma Glutamyltransferase 2 , Protein Kinase Inhibitors/pharmacology , Transglutaminases/metabolism
2.
PLoS One ; 8(5): e63798, 2013.
Article in English | MEDLINE | ID: mdl-23741300

ABSTRACT

Pulmonary Fibrosis (PF) is a devastating progressive disease in which normal lung structure and function is compromised by scarring. Lung fibrosis can be caused by thoracic radiation, injury from chemotherapy and systemic diseases such as rheumatoid arthritis that involve inflammatory responses. CDDO-Me (Methyl 2-cyano-3,12-dioxooleana-1,9(11)dien-28-oate, Bardoxolone methyl) is a novel triterpenoid with anti-fibrotic and anti-inflammatory properties as shown by our in vitro studies. Based on this evidence, we hypothesized that CDDO-Me would reduce lung inflammation, fibrosis and lung function impairment in a bleomycin model of lung injury and fibrosis. To test this hypothesis, mice received bleomycin via oropharyngeal aspiration (OA) on day zero and CDDO-Me during the inflammatory phase from days -1 to 9 every other day. Bronchoalveolar lavage fluid (BALF) and lung tissue were harvested on day 7 to evaluate inflammation, while fibrosis and lung function were evaluated on day 21. On day 7, CDDO-Me reduced total BALF protein by 50%, alveolar macrophage infiltration by 40%, neutrophil infiltration by 90% (p≤0.01), inhibited production of the inflammatory cytokines KC and IL-6 by over 90% (p≤0.001), and excess production of the pro-fibrotic cytokine TGFß by 50%. CDDO-Me also inhibited α-smooth muscle actin and fibronectin mRNA by 50% (p≤0.05). On day 21, CDDO-Me treatment reduced histological fibrosis, collagen deposition and αSMA production. Lung function was significantly improved at day 21 by treatment with CDDO-Me, as demonstrated by respiratory rate and dynamic compliance. These new findings reveal that CDDO-Me exhibits potent anti-fibrotic and anti-inflammatory properties in vivo. CDDO-Me is a potential new class of drugs to arrest inflammation and ameliorate fibrosis in patients who are predisposed to lung injury and fibrosis incited by cancer treatments (e.g. chemotherapy and radiation) and by systemic autoimmune diseases.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Macrophages, Alveolar/drug effects , Neutrophil Infiltration/drug effects , Oleanolic Acid/analogs & derivatives , Pulmonary Fibrosis/drug therapy , Actins/antagonists & inhibitors , Actins/genetics , Actins/metabolism , Administration, Inhalation , Animals , Bleomycin , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Collagen/antagonists & inhibitors , Collagen/genetics , Collagen/metabolism , Fibronectins/antagonists & inhibitors , Fibronectins/genetics , Fibronectins/metabolism , Gene Expression/drug effects , Humans , Macrophages, Alveolar/immunology , Macrophages, Alveolar/pathology , Male , Mice , Mice, Inbred C57BL , Oleanolic Acid/pharmacology , Pneumonia/chemically induced , Pneumonia/drug therapy , Pneumonia/immunology , Pneumonia/pathology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/pathology , Respiratory Function Tests , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism
3.
Am J Respir Crit Care Med ; 186(8): 740-51, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22923663

ABSTRACT

RATIONALE: Idiopathic pulmonary fibrosis (IPF) is a complex disease for which the pathogenesis is poorly understood. In this study, we identified lactic acid as a metabolite that is elevated in the lung tissue of patients with IPF. OBJECTIVES: This study examines the effect of lactic acid on myofibroblast differentiation and pulmonary fibrosis. METHODS: We used metabolomic analysis to examine cellular metabolism in lung tissue from patients with IPF and determined the effects of lactic acid and lactate dehydrogenase-5 (LDH5) overexpression on myofibroblast differentiation and transforming growth factor (TGF)-ß activation in vitro. MEASUREMENTS AND MAIN RESULTS: Lactic acid concentrations from healthy and IPF lung tissue were determined by nuclear magnetic resonance spectroscopy; α-smooth muscle actin, calponin, and LDH5 expression were assessed by Western blot of cell culture lysates. Lactic acid and LDH5 were significantly elevated in IPF lung tissue compared with controls. Physiologic concentrations of lactic acid induced myofibroblast differentiation via activation of TGF-ß. TGF-ß induced expression of LDH5 via hypoxia-inducible factor 1α (HIF1α). Importantly, overexpression of both HIF1α and LDH5 in human lung fibroblasts induced myofibroblast differentiation and synergized with low-dose TGF-ß to induce differentiation. Furthermore, inhibition of both HIF1α and LDH5 inhibited TGF-ß-induced myofibroblast differentiation. CONCLUSIONS: We have identified the metabolite lactic acid as an important mediator of myofibroblast differentiation via a pH-dependent activation of TGF-ß. We propose that the metabolic milieu of the lung, and potentially other tissues, is an important driving force behind myofibroblast differentiation and potentially the initiation and progression of fibrotic disorders.


Subject(s)
Cell Differentiation , Idiopathic Pulmonary Fibrosis/metabolism , Lactic Acid/metabolism , Myofibroblasts/metabolism , Transforming Growth Factor beta/metabolism , Case-Control Studies , Gene Expression Regulation, Enzymologic , Humans , Hydrogen-Ion Concentration , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Idiopathic Pulmonary Fibrosis/physiopathology , In Vitro Techniques , Isoenzymes/metabolism , L-Lactate Dehydrogenase/metabolism , Lactate Dehydrogenase 5 , Magnetic Resonance Spectroscopy , Up-Regulation
4.
PPAR Res ; 2012: 705352, 2012.
Article in English | MEDLINE | ID: mdl-22778711

ABSTRACT

Peroxisome proliferator activated receptor (PPAR)-γ is a nuclear hormone receptor that is activated by multiple agonists including thiazolidinediones, prostaglandins, and synthetic oleanolic acids. Many PPARγ ligands are under investigation as potential therapies for human diseases. These ligands modulate multiple cellular pathways via both PPARγ-dependent and PPARγ-independent mechanisms. Here, we review the role of PPARγ and PPARγ ligands in lung disease, with emphasis on PPARγ-independent effects. PPARγ ligands show great promise in moderating lung inflammation, as antiproliferative agents in combination to enhance standard chemotherapy in lung cancer and as treatments for pulmonary fibrosis, a progressive fatal disease with no effective therapy. Some of these effects occur when PPARγ is pharmaceutically antagonized or genetically PPARγ and are thus independent of classical PPARγ-dependent transcriptional control. Many PPARγ ligands demonstrate direct binding to transcription factors and other proteins, altering their function and contributing to PPARγ-independent inhibition of disease phenotypes. These PPARγ-independent mechanisms are of significant interest because they suggest new therapeutic uses for currently approved drugs and because they can be used as probes to identify novel proteins and pathways involved in the pathogenesis or treatment of disease, which can then be targeted for further investigation and drug development.

5.
Am J Respir Crit Care Med ; 184(6): 699-707, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21700912

ABSTRACT

RATIONALE: Idiopathic pulmonary fibrosis (IPF) is a deadly progressive disease with few treatment options. Transglutaminase 2 (TG2) is a multifunctional protein, but its function in pulmonary fibrosis is unknown. OBJECTIVES: To determine the role of TG2 in pulmonary fibrosis. METHODS: The fibrotic response to bleomycin was compared between wild-type and TG2 knockout mice. Transglutaminase and transglutaminase-catalyzed isopeptide bond expression was examined in formalin-fixed human lung biopsy sections by immunohistochemistry from patients with IPF. In addition, primary human lung fibroblasts were used to study TG2 function in vitro. MEASUREMENTS AND MAIN RESULTS: TG2 knockout mice developed significantly reduced fibrosis compared with wild-type mice as determined by hydroxyproline content and histologic fibrosis score (P < 0.05). TG2 expression and activity are increased in lung biopsy sections in humans with IPF compared with normal control subjects. In vitro overexpression of TG2 led to increased fibronectin deposition, whereas transglutaminase knockdown led to defects in contraction and adhesion. The profibrotic cytokine transforming growth factor-ß causes an increase in membrane-localized TG2, increasing its enzymatic activity. CONCLUSIONS: TG2 is involved in pulmonary fibrosis in a mouse model and in human disease and is important in normal fibroblast function. With continued research on TG2, it may offer a new therapeutic target.


Subject(s)
GTP-Binding Proteins/metabolism , Lung/enzymology , Pulmonary Fibrosis/enzymology , Transglutaminases/metabolism , Animals , Blotting, Western , Cells, Cultured , Disease Models, Animal , Fibroblasts/enzymology , Fibronectins/metabolism , Flow Cytometry , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Myofibroblasts/enzymology , Protein Glutamine gamma Glutamyltransferase 2
6.
Am J Pathol ; 178(4): 1556-67, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21406171

ABSTRACT

Fibrosis can occur in any human tissue when the normal wound healing response is amplified. Such amplification results in fibroblast proliferation, myofibroblast differentiation, and excessive extracellular matrix deposition. Occurrence of these sequelae in organs such as the eye or lung can result in severe consequences to health. Unfortunately, medical treatment of fibrosis is limited by a lack of safe and effective therapies. These therapies may be developed by identifying agents that inhibit critical steps in fibrotic progression; one such step is myofibroblast differentiation triggered by transforming growth factor-ß1 (TGFß1). In this study, we demonstrate that TGFß1-induced myofibroblast differentiation is blocked in human fibroblasts by a candidate endogenous aryl hydrocarbon receptor (AhR) ligand 2-(1'H-indole-3'-carbonyl)-thiazole-4-carboxylic acid methyl ester (ITE). Our data show that ITE disrupts TGFß1 signaling by inhibiting the nuclear translocation of Smad2/3/4. Although ITE functions as an AhR agonist, and biologically persistent AhR agonists, such as 2,3,7,8-tetrachlorodibenzo-p-dioxin, cause severe toxic effects, ITE exhibits no toxicity. Interestingly, ITE effectively inhibits TGFß1-driven myofibroblast differentiation in AhR(-/-) fibroblasts: Its ability to inhibit TGFß1 signaling is AhR independent. As supported by the results of this study, the small molecule ITE inhibits myofibroblast differentiation and may be useful clinically as an antiscarring agent.


Subject(s)
Indoles/pharmacology , Receptors, Aryl Hydrocarbon/chemistry , Thiazoles/pharmacology , Aryl Hydrocarbon Hydroxylases/metabolism , Cell Differentiation , Cytochrome P-450 CYP1B1 , Fibroblasts/metabolism , Fibronectins/metabolism , Humans , Ligands , Myofibroblasts/cytology , Orbit/cytology , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Smad4 Protein/metabolism , Transforming Growth Factor beta1/metabolism , Wound Healing
7.
PLoS One ; 6(1): e15909, 2011 Jan 06.
Article in English | MEDLINE | ID: mdl-21253589

ABSTRACT

Transforming growth factor beta (TGFß) induced differentiation of human lung fibroblasts to myofibroblasts is a key event in the pathogenesis of pulmonary fibrosis. Although the typical TGFß signaling pathway involves the Smad family of transcription factors, we have previously reported that peroxisome proliferator-activated receptor-γ (PPAR-γ) ligands inhibit TGFß-mediated differentiation of human lung fibroblasts to myofibroblasts via a Smad-independent pathway. TGFß also activates the phosphatidylinositol 3 kinase/protein kinase B (PI3K/Akt) pathway leading to phosphorylation of Akt(S473). Here, we report that PPAR-γ ligands, 2-cyano-3,12-dioxooleana-1,9-dien-28-oic acid (CDDO) and 15-deoxy-(12,14)-15d-prostaglandin J(2) (15d-PGJ(2)), inhibit human myofibroblast differentiation of normal and idiopathic pulmonary fibrotic (IPF) fibroblasts, by blocking Akt phosphorylation at Ser473 by a PPAR-γ-independent mechanism. The PI3K inhibitor LY294002 and a dominant-negative inactive kinase-domain mutant of Akt both inhibited TGFß-stimulated myofibroblast differentiation, as determined by Western blotting for α-smooth muscle actin and calponin. Prostaglandin A(1) (PGA(1)), a structural analogue of 15d-PGJ(2) with an electrophilic center, also reduced TGFß-driven phosphorylation of Akt, while CAY10410, another analogue that lacks an electrophilic center, did not; implying that the activity of 15d-PGJ(2) and CDDO is dependent on their electrophilic properties. PPAR-γ ligands inhibited TGFß-induced Akt phosphorylation via both post-translational and post-transcriptional mechanisms. This inhibition is independent of MAPK-p38 and PTEN but is dependent on TGFß-induced phosphorylation of FAK, a kinase that acts upstream of Akt. Thus, PPAR-γ ligands inhibit TGFß signaling by affecting two pro-survival pathways that culminate in myofibroblast differentiation. Further studies of PPAR-γ ligands and small electrophilic molecules may lead to a new generation of anti-fibrotic therapeutics.


Subject(s)
Cell Differentiation/drug effects , PPAR gamma/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta/pharmacology , Fibrosis/drug therapy , Humans , Ligands , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...