Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters











Database
Language
Publication year range
1.
JCI Insight ; 2(22)2017 11 16.
Article in English | MEDLINE | ID: mdl-29202450

ABSTRACT

The neutrophil chemoattractant proline-glycine-proline (PGP) is generated from collagen by matrix metalloproteinase-8/9 (MMP-8/9) and prolyl endopeptidase (PE), and it is concomitantly degraded by extracellular leukotriene A4 hydrolase (LTA4H) to limit neutrophilia. Components of cigarette smoke can acetylate PGP, yielding a species (AcPGP) that is resistant to LTA4H-mediated degradation and can, thus, support a sustained neutrophilia. In this study, we sought to elucidate if an antiinflammatory system existed to degrade AcPGP that is analogous to the PGP-LTA4H axis. We demonstrate that AcPGP is degraded through a previously unidentified action of the enzyme angiotensin-converting enzyme (ACE). Pulmonary ACE is elevated during episodes of acute inflammation, as a consequence of enhanced vascular permeability, to ensure the efficient degradation of AcPGP. Conversely, we suggest that this pathway is aberrant in chronic obstructive pulmonary disease (COPD) enabling the accumulation of AcPGP. Consequently, we identify a potentially novel protective role for AcPGP in limiting pulmonary fibrosis and suggest the pathogenic function attributed to ACE in idiopathic pulmonary fibrosis (IPF) to be a consequence of overzealous AcPGP degradation. Thus, AcPGP seemingly has very divergent roles: it is pathogenic in its capacity to drive neutrophilic inflammation and matrix degradation in the context of COPD, but it is protective in its capacity to limit fibrosis in IPF.


Subject(s)
Inflammation/metabolism , Peptidyl-Dipeptidase A/metabolism , Pulmonary Fibrosis/metabolism , Adult , Aged , Aged, 80 and over , Animals , Female , Humans , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Mutant Strains , Middle Aged , Peptidyl-Dipeptidase A/blood , Pulmonary Disease, Chronic Obstructive/blood , Pulmonary Fibrosis/pathology , Smoke
2.
J Pharmacol Exp Ther ; 351(1): 87-95, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25047515

ABSTRACT

Myofibroblasts are effector cells in fibrotic disorders that synthesize and remodel the extracellular matrix (ECM). This study investigated the role of the Src kinase pathway in myofibroblast activation in vitro and fibrogenesis in vivo. The profibrotic cytokine, transforming growth factor ß1 (TGF-ß1), induced rapid activation of Src kinase, which led to myofibroblast differentiation of human lung fibroblasts. The Src kinase inhibitor AZD0530 (saracatinib) blocked TGF-ß1-induced Src kinase activation in a dose-dependent manner. Inhibition of Src kinase significantly reduced α-smooth muscle actin (α-SMA) expression, a marker of myofibroblast differentiation, in TGF-ß1-treated lung fibroblasts. In addition, the induced expression of collagen and fibronectin and three-dimensional collagen gel contraction were also significantly inhibited in AZD0530-treated fibroblasts. The therapeutic efficiency of Src kinase inhibition in vivo was tested in the bleomycin murine lung fibrosis model. Src kinase activation and collagen accumulation were significantly reduced in the lungs of AZD0530-treated mice when compared with controls. Furthermore, the total fibrotic area and expression of α-SMA and ECM proteins were significantly decreased in lungs of AZD0530-treated mice. These results indicate that Src kinase promotes myofibroblast differentiation and activation of lung fibroblasts. Additionally, these studies provide proof-of-concept for targeting the noncanonical TGF-ß signaling pathway involving Src kinase as an effective therapeutic strategy for lung fibrosis.


Subject(s)
Benzodioxoles/pharmacology , Cell Differentiation , Enzyme Inhibitors/pharmacology , Myofibroblasts/drug effects , Pulmonary Fibrosis/drug therapy , Quinazolines/pharmacology , src-Family Kinases/metabolism , Actins/genetics , Actins/metabolism , Animals , Benzodioxoles/therapeutic use , Cell Line , Cells, Cultured , Collagen/genetics , Collagen/metabolism , Enzyme Inhibitors/therapeutic use , Female , Humans , Male , Mice , Mice, Inbred C57BL , Myofibroblasts/cytology , Myofibroblasts/enzymology , Quinazolines/therapeutic use , Transforming Growth Factor beta/pharmacology , src-Family Kinases/antagonists & inhibitors
3.
Am J Respir Crit Care Med ; 190(1): 51-61, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24874071

ABSTRACT

RATIONALE: Chronic neutrophilic inflammation is a hallmark in the pathogenesis of chronic obstructive pulmonary disease (COPD) and persists after cigarette smoking has stopped. Mechanisms involved in this ongoing inflammatory response have not been delineated. OBJECTIVES: We investigated changes to the leukotriene A4 hydrolase (LTA4H)-proline-glycine-proline (PGP) pathway and chronic inflammation in the development of COPD. METHODS: A/J mice were exposed to air or cigarette smoke for 22 weeks followed by bronchoalveolar lavage and lung and cardiac tissue analysis. Two human cohorts were used to analyze changes to the LTA4H-PGP pathway in never smokers, control smokers, COPD smokers, and COPD former smokers. PGP/AcPGP and LTA4H aminopeptidase activity were detected by mass spectroscopy, LTA4H amounts were detected by ELISA, and acrolein was detected by Western blot. MEASUREMENTS AND MAIN RESULTS: Mice exposed to cigarette smoke developed emphysema with increased PGP, neutrophilic inflammation, and selective inhibition of LTA4H aminopeptidase, which ordinarily degrades PGP. We recapitulated these findings in smokers with and without COPD. PGP and AcPGP are closely associated with cigarette smoke use. Once chronic inflammation is established, changes to LTA4H aminopeptidase remain, even in the absence of ongoing cigarette use. Acrolein modifies LTA4H and inhibits aminopeptidase activity to the same extent as cigarette smoke. CONCLUSIONS: These results demonstrate a novel pathway of aberrant regulation of PGP/AcPGP, suggesting this inflammatory pathway may be intimately involved in disease progression in the absence of ongoing cigarette smoke exposure. We highlight a mechanism by which acrolein potentiates neutrophilic inflammation through selective inhibition of LTA4H aminopeptidase activity. Clinical trial registered with www.clinicaltrials.gov (NCT 00292552).


Subject(s)
Epoxide Hydrolases/immunology , Inflammation/physiopathology , Neutrophils/immunology , Pulmonary Disease, Chronic Obstructive/etiology , Smoking/adverse effects , Aged , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/immunology , Cohort Studies , Disease Models, Animal , Emphysema/etiology , Emphysema/immunology , Female , Glycine/metabolism , Humans , Inflammation/complications , Lung/immunology , Male , Mice , Middle Aged , Myocardium/immunology , Proline/metabolism , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/physiopathology , Smoking/immunology
4.
BMJ Open ; 3(12): e004140, 2013 Dec 23.
Article in English | MEDLINE | ID: mdl-24366582

ABSTRACT

RATIONALE: Proline-glycine-proline (PGP), a neutrophil chemoattractant derived from the enzymatic breakdown of collagen, is elevated in sputum of patients with chronic obstructive pulmonary disease (COPD) and may contribute to disease progression. Whether sputum levels of PGP respond to therapy for COPD or predict outcomes is unknown. OBJECTIVES: We conducted a study ancillary to a multicenter trial of the efficacy of azithromycin treatment for 1 year in preventing COPD exacerbations to test whether sputum levels of PGP were altered by treatment or associated with exacerbation frequency. METHODS: We collected remnant sputa from trial participants and assayed them in a blinded fashion for PGP, myeloperoxidase and matrix metalloproteinase (MMP)-9 and for the ability to generate PGP from collagen ex vivo. Once the parent trial was unblinded, the results were correlated with use of azithromycin or placebo and exacerbations in participants. RESULTS: Azithromycin treatment significantly reduced sputum levels of PGP and myeloperoxidase in patients with COPD, particularly with increased duration of therapy. We found no difference in sputum MMP-9 or PGP generation between participants taking azithromycin or placebo. Sputum PGP levels were highest around the time of an exacerbation and declined with successful treatment. CONCLUSIONS: These data support a role for PGP in the airway and parenchymal neutrophilic inflammation that drives COPD progression and exacerbations, and provide new information on the anti-inflammatory properties of macrolides. PGP may have potential as a target for novel anti-inflammatory therapies in COPD and as a biomarker for clinical trials.

5.
J Neuroimmunol ; 217(1-2): 51-4, 2009 Dec 10.
Article in English | MEDLINE | ID: mdl-19875179

ABSTRACT

Prolyl endopeptidase (PE), a protease that cleaves after proline residues in oligopeptides, is highly active in brain and degrades neuropeptides in vitro. We have recently demonstrated that PE, in concert with MMP's, can generate PGP (proline-glycine-proline), a novel, neutrophil chemoattractant, from collagen. In this study, we demonstrate that human peripheral blood neutrophils contain PE, which is constitutively active, and can generate PGP de novo from collagen after activation with LPS. This novel, pro-inflammatory role for PE raises the possibility of a self-sustaining pathway of neutrophilic inflammation and may provide biomarkers and therapeutic targets for diseases caused by chronic, neutrophilic inflammation.


Subject(s)
Collagen/chemistry , Neutrophils/metabolism , Oligopeptides/metabolism , Proline/analogs & derivatives , Serine Endopeptidases/metabolism , Adjuvants, Immunologic/pharmacology , Chromatography, Liquid/methods , Collagen/metabolism , Humans , Lipopolysaccharides/pharmacology , Neutrophil Activation , Neutrophils/drug effects , Oligopeptides/chemical synthesis , Oligopeptides/isolation & purification , Proline/chemical synthesis , Proline/isolation & purification , Proline/metabolism , Prolyl Oligopeptidases , Tandem Mass Spectrometry/methods
6.
J Immunol ; 180(8): 5662-9, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18390751

ABSTRACT

Chronic neutrophilic inflammation is a manifestation of a variety of lung diseases including cystic fibrosis (CF). There is increasing evidence that fragments of extracellular matrix proteins, such as collagen and elastin, play an important role in inflammatory cell recruitment to the lung in animal models of airway inflammation. Unfortunately, the association of these peptides with human disease and the identification of therapeutic targets directed toward these inflammatory pathways have remained elusive. In this study, we demonstrate that a novel extracellular matrix-derived neutrophil chemoattractant, proline-glycine-proline (PGP), acts through CXC receptors 1 and 2 on neutrophils, similar to N-acetylated proline-glycine-proline (N-alpha-PGP). We describe the specific multistep proteolytic pathway involved in PGP generation from collagen, involving matrix metalloproteases 8 and 9 and prolyl endopeptidase, a serine protease for which we identify a novel role in inflammation. PGP generation correlates closely with airway neutrophil counts after administration of proteases in vivo. Using CF as a model, we show that CF sputum has elevated levels of PGP peptides and that PGP levels decline during the course of CF inpatient therapy for acute pulmonary exacerbation, pointing to its role as a novel biomarker for this disease. Finally, we demonstrate that CF secretions are capable of generating PGP from collagen ex vivo and that this generation is significantly attenuated by the use of inhibitors directed toward matrix metalloprotease 8, matrix metalloprotease 9, or prolyl endopeptidase. These experiments highlight unique protease interactions with structural proteins regulating innate immunity and support a role for these peptides as novel biomarkers and therapeutic targets for chronic, neutrophilic lung diseases.


Subject(s)
Cystic Fibrosis/metabolism , Extracellular Matrix Proteins/metabolism , Inflammation/metabolism , Neutrophils/metabolism , Oligopeptides/metabolism , Proline/analogs & derivatives , Serine Endopeptidases/metabolism , Animals , Chemotactic Factors/immunology , Chemotactic Factors/metabolism , Chemotaxis, Leukocyte , Chronic Disease , Cystic Fibrosis/immunology , Extracellular Matrix/immunology , Extracellular Matrix/metabolism , Humans , Inflammation/immunology , Matrix Metalloproteinase 8/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Neutrophil Activation , Neutrophils/immunology , Proline/metabolism , Prolyl Oligopeptidases , Receptors, Interleukin-8A/metabolism , Receptors, Interleukin-8B/metabolism , Sputum/immunology , Sputum/metabolism
7.
Curr Opin Pharmacol ; 8(3): 242-8, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18346936

ABSTRACT

Chemoattractant properties of matrix proteins, like collagen and elastin, for neutrophils and monocytes in vitro have long been recognized. This activity often resides in fragments of these proteins. These peptides may play a role in diseases of the lung matrix, such as chronic obstructive pulmonary disease. Recent advances include the elucidation of the structure of chemotactic collagen fragments and the demonstration that their activity may reside in a structural relatedness to CXC chemokines. Collagen and elastin fragments have been demonstrated to have a role in in vivo lung pathophysiology and have been quantified in patients with chronic lung diseases where they may activate autoimmune pathways. Elucidation of these pathways may provide novel biomarkers and therapeutic targets for chronic lung diseases.


Subject(s)
Extracellular Matrix Proteins/metabolism , Inflammation/drug therapy , Lung Diseases/drug therapy , Animals , Chemokines, CXC/chemistry , Chemotactic Factors/metabolism , Collagen/metabolism , Elastin/metabolism , Humans , Inflammation/metabolism , Lung/metabolism , Matrix Metalloproteinase 9/physiology , Oligopeptides/biosynthesis , Oligopeptides/chemistry , Peptide Fragments/metabolism , Peptide Hydrolases/physiology , Pulmonary Disease, Chronic Obstructive/drug therapy
8.
Am J Respir Cell Mol Biol ; 28(4): 443-50, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12654633

ABSTRACT

Oxidative stress may impair alveolar macrophage function in patients with inflammatory lung diseases or those exposed to high concentrations of oxygen. We investigated putative mechanisms of injury to macrophages by oxidative stress, using RAW 264.7 cells exposed to 95% oxygen for 48 h. Hyperoxia-exposed macrophages were less able to phagocytose and kill Klebsiella pneumoniae than normoxic controls, despite increased production of nitric oxide, a free radical important in pathogen killing. Exposure of macrophages to hyperoxia had marked effects on the actin cytoskeleton, including increased actin polymerization, loss of cortical actin, formation of stress fibers, de novo synthesis of actin, and actin oxidation. Hyperoxia induced changes in cell morphology, with increased cell size and pseudopod formation. Exposure of macrophages to jasplakinolide, an agent that increases actin polymerization, also impaired their ability to phagocytose Klebsiella. Alveolar macrophages isolated from mice exposed to 100% oxygen for 84 h also demonstrated impaired phagocytic function, as well as similar effects on the actin cytoskeleton and cell morphology to macrophages exposed to hyperoxia in vitro. We conclude that oxidative stress in vitro and in vivo impairs macrophage antibacterial function through effects on actin.


Subject(s)
Actins/physiology , Macrophages, Alveolar/microbiology , Macrophages, Alveolar/physiology , Oxidative Stress/physiology , Phagocytosis/physiology , Adenosine Triphosphate/metabolism , Animals , Cell Line , Klebsiella pneumoniae , Macrophages, Alveolar/cytology , Mice , Microscopy, Fluorescence , Nitric Oxide/metabolism
9.
Chest ; 122(6 Suppl): 314S-320S, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12475808

ABSTRACT

ARDS is a disease process that is characterized by diffuse inflammation in the lung parenchyma. The involvement of inflammatory mediators in ARDS has been the subject of intense investigation, and oxidant-mediated tissue injury is likely to be important in the pathogenesis of ARDS. In response to various inflammatory stimuli, lung endothelial cells, alveolar cells, and airway epithelial cells, as well as activated alveolar macrophages, produce both nitric oxide and superoxide, which may react to form peroxynitrite, which can nitrate and oxidize key amino acids in various lung proteins, such as surfactant protein A, and inhibit their functions. The nitration and oxidation of a variety of crucial proteins present in the alveolar space have been shown to be associated with diminished function in vitro and also have been identified ex vivo in proteins sampled from patients with acute lung injury (ALI)/ARDS. Various enzymes and low-molecular-weight scavengers that are present in the lung tissue and alveolar lining fluid decreased the concentration of these toxic species. The purpose of this brief chapter is to review the results from various studies demonstrating increased levels of reactive oxygen-nitrogen intermediates in the alveolar spaces of patients with ALI/ARDS.


Subject(s)
Oxidative Stress/physiology , Respiratory Distress Syndrome/chemically induced , Respiratory Distress Syndrome/physiopathology , Antioxidants/metabolism , Bronchoalveolar Lavage Fluid/chemistry , Humans , Nitric Oxide/analysis , Reactive Nitrogen Species/adverse effects , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/adverse effects , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL