Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 23(19)2022 Oct 08.
Article in English | MEDLINE | ID: mdl-36233248

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a progressive, chronic, interstitial lung disease with a poor prognosis. Although specific anti-fibrotic medications are now available, the median survival time following diagnosis remains very low, and new therapies are urgently needed. To uncover novel therapeutic targets, we examined how biochemical properties of the fibrotic lung are different from the healthy lung. Previous work identified lactate as a metabolite that is upregulated in IPF lung tissue. Importantly, inhibition of the enzyme responsible for lactate production prevents fibrosis in vivo. Further studies revealed that fibrotic lesions of the lung experience a significant decline in tissue pH, likely due to the overproduction of lactate. It is not entirely clear how cells in the lung respond to changes in extracellular pH, but a family of proton sensing G-protein coupled receptors has been shown to be activated by reductions in extracellular pH. This work examines the expression profiles of proton sensing GPCRs in non-fibrotic and IPF-derived primary human lung fibroblasts. We identify TDAG8 as a proton sensing GPCR that is upregulated in IPF fibroblasts and that knockdown of TDAG8 dampens myofibroblast differentiation. To our surprise, BTB, a proposed positive allosteric modulator of TDAG8, inhibits myofibroblast differentiation. Our data suggest that BTB does not require TDAG8 to inhibit myofibroblast differentiation, but rather inhibits myofibroblast differentiation through suppression of RhoA mediated signaling. Our work highlights the therapeutic potential of BTB as an anti-fibrotic treatment and expands upon the importance of RhoA-mediated signaling pathways in the context of myofibroblast differentiation. Furthermore, this works also suggests that TDAG8 inhibition may have therapeutic relevance in the treatment of IPF.


Subject(s)
Idiopathic Pulmonary Fibrosis , rhoA GTP-Binding Protein , Cell Differentiation/physiology , Fibroblasts/metabolism , Fibrosis , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Lactates/metabolism , Lung/pathology , Myofibroblasts/metabolism , Protons , rhoA GTP-Binding Protein/metabolism
2.
Sci Rep ; 11(1): 19436, 2021 09 30.
Article in English | MEDLINE | ID: mdl-34593881

ABSTRACT

Combustion related particulate matter air pollution (PM) is associated with an increased risk of respiratory infections in adults. The exact mechanism underlying this association has not been determined. We hypothesized that increased concentrations of combustion related PM would result in dysregulation of the innate immune system. This epidemiological study includes 111 adult patients hospitalized with respiratory infections who underwent transcriptional analysis of their peripheral blood. We examined the association between gene expression at the time of hospitalization and ambient measurements of particulate air pollutants in the 28 days prior to hospitalization. For each pollutant and time lag, gene-specific linear models adjusting for infection type were fit using LIMMA (Linear Models For Microarray Data), and pathway/gene set analyses were performed using the CAMERA (Correlation Adjusted Mean Rank) program. Comparing patients with viral and/or bacterial infection, the expression patterns associated with air pollution exposure differed. Adjusting for the type of infection, increased concentrations of Delta-C (a marker of biomass smoke) and other PM were associated with upregulation of iron homeostasis and protein folding. Increased concentrations of black carbon (BC) were associated with upregulation of viral related gene pathways and downregulation of pathways related to antigen presentation. The pollutant/pathway associations differed by lag time and by type of infection. This study suggests that the effect of air pollution on the pathogenesis of respiratory infection may be pollutant, timing, and infection specific.


Subject(s)
Particulate Matter/adverse effects , Respiratory Tract Infections/immunology , Smoke/adverse effects , Transcriptome , Adult , Environmental Exposure/adverse effects , Female , Humans , Immunity/genetics , Male , New York/epidemiology , Respiratory Tract Infections/etiology , Respiratory Tract Infections/genetics , Respiratory Tract Infections/metabolism , Soot/adverse effects
3.
Eur Respir J ; 56(5)2020 11.
Article in English | MEDLINE | ID: mdl-32943406

ABSTRACT

Pulmonary fibrosis is a devastating, progressive disease and carries a prognosis worse than most cancers. Despite ongoing research, the mechanisms that underlie disease pathogenesis remain only partially understood. However, the self-perpetuating nature of pulmonary fibrosis has led several researchers to propose the existence of pathological signalling loops. According to this hypothesis, the normal wound-healing process becomes corrupted and results in the progressive accumulation of scar tissue in the lung. In addition, several negative regulators of pulmonary fibrosis are downregulated and, therefore, are no longer capable of inhibiting these feed-forward loops. The combination of pathological signalling loops and loss of a checks and balances system ultimately culminates in a process of unregulated scar formation. This review details specific signalling pathways demonstrated to play a role in the pathogenesis of pulmonary fibrosis. The evidence of detrimental signalling loops is elucidated with regard to epithelial cell injury, cellular senescence and the activation of developmental and ageing pathways. We demonstrate where these loops intersect each other, as well as common mediators that may drive these responses and how the loss of pro-resolving mediators may contribute to the propagation of disease. By focusing on the overlapping signalling mediators among the many pro-fibrotic pathways, it is our hope that the pulmonary fibrosis community will be better equipped to design future trials that incorporate the redundant nature of these pathways as we move towards finding a cure for this unrelenting disease.


Subject(s)
Idiopathic Pulmonary Fibrosis , Pulmonary Fibrosis , Cellular Senescence , Epithelial Cells , Humans , Lung , Signal Transduction
4.
Curr Rheumatol Rev ; 15(4): 277-289, 2019.
Article in English | MEDLINE | ID: mdl-30652645

ABSTRACT

Interstitial Lung Disease (ILD) is a well-known complication of rheumatoid arthritis (RA) which often results in significant morbidity and mortality. It is often diagnosed late in the disease process via descriptive criteria. Multiple subtypes of RA-ILD exist as defined by chest CT and histopathology. In the absence of formal natural history studies and definitive diagnostics, a conventional dogma has emerged that there are two major subtypes of RA-ILD (nonspecific interstitial pneumonia (NSIP) and Usual Interstitial Pneumonia (UIP)). These subtypes are based on clinical experience and correlation studies. However, recent animal model data are incongruous with established paradigms of RA-ILD and beg reassessment of the clinical evidence in order to better understand etiology, pathogenesis, prognosis, and response to therapy. To this end, here we: 1) review the literature on epidemiology, radiology, histopathology and clinical outcomes of the various RAILD subtypes, existing animal models, and current theories on RA-ILD pathogenesis; 2) highlight the major gaps in our knowledge; and 3) propose future research to test an emerging theory of RAILD that posits initial rheumatic lung inflammation in the form of NSIP-like pathology transforms mesenchymal cells to derive chimeric disease, and subsequently develops into frank UIP-like fibrosis in some RA patients. Elucidation of the pathogenesis of RA-ILD is critical for the development of effective interventions for RA-ILD.


Subject(s)
Arthritis, Rheumatoid/complications , Lung Diseases, Interstitial/etiology , Animals , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Disease Models, Animal , Humans , Lung Diseases, Interstitial/diagnostic imaging , Lung Diseases, Interstitial/epidemiology , Tomography, X-Ray Computed
5.
Respir Med ; 145: 35-40, 2018 12.
Article in English | MEDLINE | ID: mdl-30509714

ABSTRACT

INTRODUCTION: Pulmonary infiltrates in immunosuppressed patients are common. Yields from bronchoscopy with bronchoalveolar lavage (BAL) has been reported to be between 31 and 65%. The clinical impact of pneumocystis and viral Polymerase chain reaction (PCR) testing on BAL has not been extensively evaluated in a mixed immunosuppressed patient population. METHODS: We performed a retrospective chart review of immunosuppressed adults with pulmonary infiltrates who underwent BAL at the University of Rochester Medical Center. Only one BAL per patient was included. We compared the rate of positive PCR testing to conventional testing. We then investigated factors associated with positive PCR testing. Finally, we assessed for changes in antimicrobial therapy after bronchoscopy. RESULTS: Three hundred and fifty-nine patients underwent BAL with 249 patients having pneumocystis PCR testing and 142 having viral PCR testing. Pneumocystis identification occurred in 43 patients and viral species identification occurred in 56 patients. PCR testing increased pneumocystis identification compared to microscopy, 14% vs. 5%, p = 0.01, and viral identification compared to culture, 25% vs. 6%, p = 0.0001. Of the patients with positive pneumocystis PCR testing 49% had antibiotics stopped, 66% were started on anti-pneumocystis therapy, and only 6% did not receive treatment. There was no difference in the number of patients with antibiotics stopped based on viral PCR testing results. DISCUSSION: PCR testing increases BAL yield in immunosuppressed patients compared to conventional testing. Pneumocystis identified by PCR only may cause a self-limited infection and may not require antimicrobial therapy. PCR testing should be included in the evaluation of pulmonary infiltrates in immunosuppressed patients.


Subject(s)
Bronchoalveolar Lavage Fluid/microbiology , Immunocompromised Host , Pneumocystis Infections/diagnosis , Pneumocystis Infections/microbiology , Pneumocystis/isolation & purification , Polymerase Chain Reaction/methods , Adult , Aged , Aged, 80 and over , Anti-Bacterial Agents/administration & dosage , Antifungal Agents/administration & dosage , Antiviral Agents/administration & dosage , Bronchoalveolar Lavage Fluid/virology , Female , Humans , Male , Middle Aged , Pneumocystis Infections/drug therapy , Retrospective Studies , Young Adult
6.
J Chromatogr B Analyt Technol Biomed Life Sci ; 1061-1062: 468-473, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28822945

ABSTRACT

Exhaled breath condensate (EBC) is easily obtained for clinical diagnosis and prognosis for pulmonary diseases and has gained much interest in biomarker discovery research and studies. Lactate, a physiological material, is found in EBC and has been demonstrated to be a potential indicator of chronic obstructive pulmonary disease and other lung diseases. Several assays are available to detect lactate in human body fluids, and yet none is suitable for detecting lactate in EBC. Due to the very low concentration of lactate in EBC and low volume of EBC, it is very important to develop an assay to measure lactate with high sensitivity, accuracy and easy sample processing. We report here a novel LC-MS/MS based assay to measure lactate using HILIC column separation. Sample preparation was simple and straightforward through a "dilute and shoot" approach with a separation of 4min. The limit of quantification was determined to be 0.5µM. This assay was linear from 0.5µM to at least 100µM. The inter- and intra- day precision at the levels of 1µM, 10µM, and 100µM were less than 3% with recovery within 5.4% of expected values. There was no ion suppression for the assay, and no carry-over was observed up to 500µM. Furthermore, we discovered that lactate is ubiquitously present in the lab environment, which can create significant challenges for accurate detection of lactate at low concentrations. We provided practical approaches in this paper to overcome the challenges and ensure the accuracy of the assay. In summary, this article presents an accurate and sensitive method using LC-MS/MS for measuring lactate in EBC, and this method is suitable for measuring lactate concentrations for non-invasive monitoring of pulmonary functions.


Subject(s)
Breath Tests/methods , Chromatography, Liquid/methods , Lactic Acid/analysis , Tandem Mass Spectrometry/methods , Biomarkers/analysis , Exhalation , Female , Humans , Linear Models , Male , Reproducibility of Results , Sensitivity and Specificity
7.
Radiat Res ; 188(1): 35-43, 2017 07.
Article in English | MEDLINE | ID: mdl-28463588

ABSTRACT

Exposure of the lung to ionizing radiation that occurs in radiotherapy, as well as after accidental or intentional mass casualty incident can result in pulmonary fibrosis, which has few treatment options. Pulmonary fibrosis is characterized by an accumulation of extracellular matrix proteins that create scar tissue. Although the mechanisms leading to radiation-induced pulmonary fibrosis remain poorly understood, one frequent observation is the activation of the profibrotic cytokine transforming growth factor-beta (TGF-ß). Our laboratory has shown that the metabolite lactate activates latent TGF-ß by a reduction in extracellular pH. We recently demonstrated that lactate dehydrogenase-A (LDHA), the enzyme that produces lactate, is upregulated in patients with radiation-induced pulmonary fibrosis. Furthermore, genetic silencing of LDHA or pharmacologic inhibition using the LDHA inhibitor gossypol prevented radiation-induced extracellular matrix secretion in vitro through inhibition of TGF-ß activation. In the current study, we hypothesized that LDHA inhibition in vivo prevents radiation-induced pulmonary fibrosis. To test this hypothesis, C57BL/6 mice received 5 Gy total-body irradiation plus 10 Gy thoracic irradiation from a 137Cs source to induce pulmonary fibrosis. Starting at 4 weeks postirradiation, mice were treated with 5 mg/kg of the LDHA inhibitor gossypol or vehicle daily until sacrifice at 26 weeks postirradiation. Exposure to radiation resulted in pulmonary fibrosis, characterized by an increase in collagen content, fibrosis area, extracellular matrix gene expression and TGF-ß activation. Irradiated mice treated with gossypol had significantly reduced fibrosis outcomes, including reduced collagen content in the lungs, reduced expression of active TGF-ß, LDHA and the transcription factor hypoxia-inducible factor-1 alpha (HIF-1α). These findings suggest that inhibition of LDHA protects against radiation-induced pulmonary fibrosis, and may be a novel therapeutic strategy for radiation-induced pulmonary fibrosis.


Subject(s)
Gossypol/administration & dosage , L-Lactate Dehydrogenase/antagonists & inhibitors , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/prevention & control , Radiation Pneumonitis/immunology , Radiation Pneumonitis/prevention & control , Animals , Cytokines/immunology , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred C57BL , Pulmonary Fibrosis/pathology , Radiation Dosage , Radiation Pneumonitis/pathology , Radiation Protection/methods , Radiation Tolerance/drug effects , Radiation-Protective Agents/administration & dosage , Treatment Outcome
8.
Am J Respir Cell Mol Biol ; 56(4): 453-464, 2017 04.
Article in English | MEDLINE | ID: mdl-27967234

ABSTRACT

An aberrant oxygen environment at birth increases the severity of respiratory viral infections later in life through poorly understood mechanisms. Here, we show that alveolar epithelial cell (AEC) 2 cells (AEC2s), progenitors for AEC1 cells, are depleted in adult mice exposed to neonatal hypoxia or hyperoxia. Airway cells expressing surfactant protein (SP)-C and ATP binding cassette subfamily A member 3, alveolar pod cells expressing keratin (KRT) 5, and pulmonary fibrosis were observed when these mice were infected with a sublethal dose of HKx31, H3N2 influenza A virus. This was not seen in infected siblings birthed into room air. Genetic lineage tracing studies in mice exposed to neonatal hypoxia or hyperoxia revealed pre-existing secretoglobin 1a1+ cells produced airway cells expressing SP-C and ATP binding cassette subfamily A member 3. Pre-existing Kr5+ progenitor cells produced squamous alveolar cells expressing receptor for advanced glycation endproducts, aquaporin 5, and T1α in alveoli devoid of AEC2s. They were not the source of KRT5+ alveolar pod cells. These oxygen-dependent changes in epithelial cell regeneration and fibrosis could be recapitulated by conditionally depleting AEC2s in mice using diphtheria A toxin and then infecting with influenza A virus. Likewise, airway cells expressing SP-C and alveolar cells expressing KRT5 were observed in human idiopathic pulmonary fibrosis. These findings suggest that alternative progenitor lineages are mobilized to regenerate the alveolar epithelium when AEC2s are severely injured or depleted by previous insults, such as an adverse oxygen environment at birth. Because these lineages regenerate AECs in spatially distinct compartments of a lung undergoing fibrosis, they may not be sufficient to prevent disease.


Subject(s)
Aging/metabolism , Alveolar Epithelial Cells/cytology , Cell Lineage , Stem Cells/cytology , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Animals , Animals, Newborn , Cell Lineage/drug effects , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Keratin-5/metabolism , Mice , Models, Biological , Oxygen/pharmacology , Pulmonary Surfactant-Associated Protein C/metabolism , Regeneration/drug effects , Stem Cells/drug effects , Stem Cells/metabolism , Uteroglobin/metabolism
9.
Am J Physiol Lung Cell Mol Physiol ; 309(8): L888-901, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26301452

ABSTRACT

Inflammation is a protective response to injury, but it can become chronic, leading to tissue damage and disease. Cigarette smoke causes multiple inflammatory diseases, which account for thousands of deaths and cost billions of dollars annually. Cigarette smoke disrupts the function of immune cells, such as macrophages, by prolonging inflammatory signaling, promoting oxidative stress, and impairing phagocytosis, contributing to increased incidence of infections. Recently, new families of lipid-derived mediators, "specialized proresolving mediators" (SPMs), were identified. SPMs play a critical role in the active resolution of inflammation by counterregulating proinflammatory signaling and promoting resolution pathways. We have identified dysregulated concentrations of lipid mediators in exhaled breath condensate, bronchoalveolar lavage fluid, and serum from patients with chronic obstructive pulmonary disease (COPD). In human alveolar macrophages from COPD and non-COPD patients, D-series resolvins decreased inflammatory cytokines and enhanced phagocytosis. To further investigate the actions of resolvins on human cells, macrophages were differentiated from human blood monocytes and treated with D-series resolvins and then exposed to cigarette smoke extract. Resolvins significantly suppressed macrophage production of proinflammatory cytokines, enzymes, and lipid mediators. Resolvins also increased anti-inflammatory cytokines, promoted an M2 macrophage phenotype, and restored cigarette smoke-induced defects in phagocytosis, highlighting the proresolving functions of these molecules. These actions were receptor-dependent and involved modulation of canonical and noncanonical NF-κB expression, with the first evidence for SPM action on alternative NF-κB signaling. These data show that resolvins act on human macrophages to attenuate cigarette smoke-induced inflammatory effects through proresolving mechanisms and provide new evidence of the therapeutic potential of SPMs.


Subject(s)
Docosahexaenoic Acids/pharmacology , Inflammation/prevention & control , Macrophages, Alveolar/drug effects , Smoking/adverse effects , Cells, Cultured , Cytokines/biosynthesis , Humans , Inflammation/pathology , Inflammation/physiopathology , Inflammation Mediators/metabolism , Lipid Metabolism/drug effects , Macrophages, Alveolar/pathology , Macrophages, Alveolar/physiology , Oxidative Stress/drug effects , Phagocytosis/drug effects , Pulmonary Disease, Chronic Obstructive/pathology , Pulmonary Disease, Chronic Obstructive/physiopathology , Signal Transduction/drug effects , Smoke/adverse effects
10.
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
11.
Am J Respir Cell Mol Biol ; 46(5): 687-94, 2012 May.
Article in English | MEDLINE | ID: mdl-22246864

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a progressive scarring disorder characterized by the proliferation of interstitial fibroblasts and the deposition of extracellular matrix causing impaired gas exchange. Spiruchostatin A (SpA) is a histone deacetylase inhibitor (HDI) with selectivity toward Class I enzymes, which distinguishes it from other nonspecific HDIs that are reported to inhibit (myo)fibroblast proliferation and differentiation. Because the selectivity of HDIs may be important clinically, we postulated that SpA inhibits the proliferation and differentiation of IPF fibroblasts. Primary fibroblasts were grown from lung biopsy explants obtained from patients with IPF or from normal control subjects, using two-dimensional or three-dimensional culture models. The effect of SpA on fibroproliferation in serum-containing medium ± transforming growth factor (TGF)-ß(1) was quantified by methylene blue binding. The acetylation of histone H3, the expression of the cell-cycle inhibitor p21(waf1), and the myofibroblast markers α-smooth muscle actin (α-SMA) and collagens I and III were determined by Western blotting, quantitative RT-PCR, immunofluorescent staining, or colorimetry. SpA inhibited the proliferation of IPF or normal fibroblasts in a time-dependent and concentration-dependent manner (concentration required to achieve 50% inhibition = 3.8 ± 0.4 nM versus 7.8 ± 0.2 nM, respectively; P < 0.05), with little cytotoxicity. Western blot analyses revealed that SpA caused a concentration-dependent increase in histone H3 acetylation, paralleling its antiproliferative effect. SpA also increased p21(waf1) expression, suggesting that direct cell-cycle regulation was the mechanism of inhibiting proliferation. Although treatment with TGF-ß(1) induced myofibroblast differentiation associated with increased expression of α-SMA, collagen I and collagen III and soluble collagen release, these responses were potently inhibited by SpA. These data support the concept that bicyclic tetrapeptide HDIs merit further investigation as potential treatments for IPF.


Subject(s)
Cell Differentiation/drug effects , Cell Proliferation/drug effects , Histone Deacetylase Inhibitors/pharmacology , Peptides, Cyclic/pharmacology , Pulmonary Fibrosis/pathology , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/drug effects , Fluorescent Antibody Technique, Indirect , Humans , Polymerase Chain Reaction
13.
PPAR Res ; 2007: 71323, 2007.
Article in English | MEDLINE | ID: mdl-17710235

ABSTRACT

Pulmonary fibrosis is a group of disorders characterized by accumulation of scar tissue in the lung interstitium, resulting in loss of alveolar function, destruction of normal lung architecture, and respiratory distress. Some types of fibrosis respond to corticosteroids, but for many there are no effective treatments. Prognosis varies but can be poor. For example, patients with idiopathic pulmonary fibrosis (IPF) have a median survival of only 2.9 years. Prognosis may be better in patients with some other types of pulmonary fibrosis, and there is variability in survival even among individuals with biopsy-proven IPF. Evidence is accumulating that the peroxisome proliferator-activated receptors (PPARs) play important roles in regulating processes related to fibrogenesis, including cellular differentiation, inflammation, and wound healing. PPARalpha agonists, including the hypolidipemic fibrate drugs, inhibit the production of collagen by hepatic stellate cells and inhibit liver, kidney, and cardiac fibrosis in animal models. In the mouse model of lung fibrosis induced by bleomycin, a PPARalpha agonist significantly inhibited the fibrotic response, while PPARalpha knockout mice developed more serious fibrosis. PPARbeta/delta appears to play a critical role in regulating the transition from inflammation to wound healing. PPARbeta/delta agonists inhibit lung fibroblast proliferation and enhance the antifibrotic properties of PPARgamma agonists. PPARgamma ligands oppose the profibrotic effect of TGF-beta, which induces differentiation of fibroblasts to myofibroblasts, a critical effector cell in fibrosis. PPARgamma ligands, including the thiazolidinedione class of antidiabetic drugs, effectively inhibit lung fibrosis in vitro and in animal models. The clinical availability of potent and selective PPARalpha and PPARgamma agonists should facilitate rapid development of successful treatment strategies based on current and ongoing research.

SELECTION OF CITATIONS
SEARCH DETAIL
...