1
|
Aribindi K, Liu GY, Albertson TE. Emerging pharmacological options in the treatment of idiopathic pulmonary fibrosis (IPF). Expert Rev Clin Pharmacol 2024; 17:817-835. [PMID: 39192604 PMCID: PMC11441789 DOI: 10.1080/17512433.2024.2396121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 08/20/2024] [Indexed: 08/29/2024]
Abstract
INTRODUCTION Idiopathic pulmonary fibrosis (IPF) is a progressive-fibrosing lung disease with a median survival of less than 5 years. Currently, two agents, pirfenidone and nintedanib are approved for this disease, and both have been shown to reduce the rate of decline in lung function in patients with IPF. However, both have significant adverse effects and neither completely arrest the decline in lung function. AREAS COVERED Thirty experimental agents with unique mechanisms of action that are being evaluated for the treatment of IPF are discussed. These agents work through various mechanisms of action, these include inhibition of transcription nuclear factor k-B on fibroblasts, reduced expression of metalloproteinase 7, the generation of more lysophosphatidic acids, blocking the effects of transforming growth factor ß, and reducing reactive oxygen species as examples of some unique mechanisms of action of these agents. EXPERT OPINION New drug development has the potential to expand the treatment options available in the treatment of IPF patients. It is expected that the adverse drug effect profiles will be more favorable than current agents. It is further anticipated that these new agents or combinations of agents will arrest the fibrosis, not just slow the fibrotic process.
Collapse
Affiliation(s)
- Katyayini Aribindi
- Department of Internal Medicine, Division of Pulmonary, Critical Care & Sleep Medicine, University of California Davis, School of Medicine, Sacramento, CA, USA
- Department of Medicine, Department of Veterans Affairs Northern California Health Care System, Mather, CA, USA
| | - Gabrielle Y Liu
- Department of Internal Medicine, Division of Pulmonary, Critical Care & Sleep Medicine, University of California Davis, School of Medicine, Sacramento, CA, USA
| | - Timothy E Albertson
- Department of Internal Medicine, Division of Pulmonary, Critical Care & Sleep Medicine, University of California Davis, School of Medicine, Sacramento, CA, USA
- Department of Medicine, Department of Veterans Affairs Northern California Health Care System, Mather, CA, USA
| |
Collapse
|
2
|
Chen R, Dai J. Lipid metabolism in idiopathic pulmonary fibrosis: From pathogenesis to therapy. J Mol Med (Berl) 2023; 101:905-915. [PMID: 37289208 DOI: 10.1007/s00109-023-02336-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/26/2023] [Accepted: 05/29/2023] [Indexed: 06/09/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic irreversible interstitial lung disease characterized by a progressive decline in lung function. The etiology of IPF is unknown, which poses a significant challenge to the treatment of IPF. Recent studies have identified a strong association between lipid metabolism and the development of IPF. Qualitative and quantitative analysis of small molecule metabolites using lipidomics reveals that lipid metabolic reprogramming plays a role in the pathogenesis of IPF. Lipids such as fatty acids, cholesterol, arachidonic acid metabolites, and phospholipids are involved in the onset and progression of IPF by inducing endoplasmic reticulum stress, promoting cell apoptosis, and enhancing the expression of pro-fibrotic biomarkers. Therefore, targeting lipid metabolism can provide a promising therapeutic strategy for pulmonary fibrosis. This review focuses on lipid metabolism in the pathogenesis of pulmonary fibrosis.
Collapse
Affiliation(s)
- Ranxun Chen
- Department of Pulmonary and Critical Care Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, Jiangsu, China
| | - Jinghong Dai
- Department of Pulmonary and Critical Care Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, Jiangsu, China.
| |
Collapse
|
3
|
The Role and Regulation of Thromboxane A2 Signaling in Cancer-Trojan Horses and Misdirection. Molecules 2022; 27:molecules27196234. [PMID: 36234768 PMCID: PMC9573598 DOI: 10.3390/molecules27196234] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/09/2022] [Accepted: 09/09/2022] [Indexed: 11/24/2022] Open
Abstract
Over the last two decades, there has been an increasing awareness of the role of eicosanoids in the development and progression of several types of cancer, including breast, prostate, lung, and colorectal cancers. Several processes involved in cancer development, such as cell growth, migration, and angiogenesis, are regulated by the arachidonic acid derivative thromboxane A2 (TXA2). Higher levels of circulating TXA2 are observed in patients with multiple cancers, and this is accompanied by overexpression of TXA2 synthase (TBXAS1, TXA2S) and/or TXA2 receptors (TBXA2R, TP). Overexpression of TXA2S or TP in tumor cells is generally associated with poor prognosis, reduced survival, and metastatic disease. However, the role of TXA2 signaling in the stroma during oncogenesis has been underappreciated. TXA2 signaling regulates the tumor microenvironment by modulating angiogenic potential, tumor ECM stiffness, and host immune response. Moreover, the by-products of TXA2S are highly mutagenic and oncogenic, adding to the overall phenotype where TXA2 synthesis promotes tumor formation at various levels. The stability of synthetic enzymes and receptors in this pathway in most cancers (with few mutations reported) suggests that TXA2 signaling is a viable target for adjunct therapy in various tumors to reduce immune evasion, primary tumor growth, and metastasis.
Collapse
|
4
|
Suzuki T, Kropski JA, Chen J, Carrier EJ, Chen X, Sherrill TP, Winters NI, Camarata JE, Polosukhin VV, Han W, Rathinasabapathy A, Gutor S, Gulleman P, Sabusap C, Banovich NE, Tanjore H, Freeman ML, Tada Y, Young LR, Gokey JJ, Blackwell TS, West JD. Thromboxane-Prostanoid Receptor Signaling Drives Persistent Fibroblast Activation in Pulmonary Fibrosis. Am J Respir Crit Care Med 2022; 206:596-607. [PMID: 35728047 PMCID: PMC9716913 DOI: 10.1164/rccm.202106-1503oc] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 06/28/2022] [Indexed: 11/16/2022] Open
Abstract
Rationale: Although persistent fibroblast activation is a hallmark of idiopathic pulmonary fibrosis (IPF), mechanisms regulating persistent fibroblast activation in the lungs have not been fully elucidated. Objectives: On the basis of our observation that lung fibroblasts express TBXA2R (thromboxane-prostanoid receptor) during fibrosis, we investigated the role of TBXA2R signaling in fibrotic remodeling. Methods: We identified TBXA2R expression in lungs of patients with IPF and mice and studied primary mouse and human lung fibroblasts to determine the impact of TBXA2R signaling on fibroblast activation. We used TBXA2R-deficient mice and small-molecule inhibitors to investigate TBXA2R signaling in preclinical lung fibrosis models. Measurements and Main Results: TBXA2R expression was upregulated in fibroblasts in the lungs of patients with IPF and in mouse lungs during experimental lung fibrosis. Genetic deletion of TBXA2R, but not inhibition of thromboxane synthase, protected mice from bleomycin-induced lung fibrosis, thereby suggesting that an alternative ligand activates profibrotic TBXA2R signaling. In contrast to thromboxane, F2-isoprostanes, which are nonenzymatic products of arachidonic acid induced by reactive oxygen species, were persistently elevated during fibrosis. F2-isoprostanes induced TBXA2R signaling in fibroblasts and mediated a myofibroblast activation profile due, at least in part, to potentiation of TGF-β (transforming growth factor-β) signaling. In vivo treatment with the TBXA2R antagonist ifetroban reduced profibrotic signaling in the lungs, protected mice from lung fibrosis in three preclinical models (bleomycin, Hermansky-Pudlak mice, and radiation-induced fibrosis), and markedly enhanced fibrotic resolution after bleomycin treatment. Conclusions: TBXA2R links oxidative stress to fibroblast activation during lung fibrosis. TBXA2R antagonists could have utility in treating pulmonary fibrosis.
Collapse
Affiliation(s)
- Toshio Suzuki
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
- Department of Medical Oncology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Jonathan A. Kropski
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
- Department of Medicine, Department of Veterans Affairs Medical Center, Nashville, Tennessee
| | - Jingyuan Chen
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Erica J. Carrier
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Xinping Chen
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Taylor P. Sherrill
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Nichelle I. Winters
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Jane E. Camarata
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Vasiliy V. Polosukhin
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Wei Han
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
| | | | - Sergey Gutor
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Peter Gulleman
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Carleen Sabusap
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
| | | | - Harikrishna Tanjore
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Michael L. Freeman
- Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Yuji Tada
- Department of Pulmonary Medicine, School of Medicine, International University of Health and Welfare, Chiba, Japan; and
| | - Lisa R. Young
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
- Division of Pulmonary Medicine, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Jason J. Gokey
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Timothy S. Blackwell
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
- Department of Medicine, Department of Veterans Affairs Medical Center, Nashville, Tennessee
| | - James D. West
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
| |
Collapse
|
5
|
Thatcher TH, Peters-Golden M. From Biomarker to Mechanism? F2-isoprostanes in Pulmonary Fibrosis. Am J Respir Crit Care Med 2022; 206:530-532. [PMID: 35763804 DOI: 10.1164/rccm.202205-0914ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Thomas H Thatcher
- Virginia Commonwealth University, 6889, Pulmonary and Critical Care Medicine, Richmond, Virginia, United States;
| | - Marc Peters-Golden
- University of Michigan Medical School, 12266, Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Ann Arbor, Michigan, United States
| |
Collapse
|
6
|
Roberts MJ, May LT, Keen AC, Liu B, Lam T, Charlton SJ, Rosethorne EM, Halls ML. Inhibition of the Proliferation of Human Lung Fibroblasts by Prostacyclin Receptor Agonists is Linked to a Sustained cAMP Signal in the Nucleus. Front Pharmacol 2021; 12:669227. [PMID: 33995100 PMCID: PMC8116805 DOI: 10.3389/fphar.2021.669227] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 04/16/2021] [Indexed: 12/21/2022] Open
Abstract
Idiopathic pulmonary fibrosis is a chronic and progressive fibrotic lung disease, and current treatments are limited by their side effects. Proliferation of human lung fibroblasts in the pulmonary interstitial tissue is a hallmark of this disease and is driven by prolonged ERK signalling in the nucleus in response to growth factors such as platelet-derived growth factor (PDGF). Agents that increase cAMP have been suggested as alternative therapies, as this second messenger can inhibit the ERK cascade. We previously examined a panel of eight Gαs-cAMP-coupled G protein-coupled receptors (GPCRs) endogenously expressed in human lung fibroblasts. Although the cAMP response was important for the anti-fibrotic effects of GPCR agonists, the magnitude of the acute cAMP response was not predictive of anti-fibrotic efficacy. Here we examined the reason for this apparent disconnect by stimulating the Gαs-coupled prostacyclin receptor and measuring downstream signalling at a sub-cellular level. MRE-269 and treprostinil caused sustained cAMP signalling in the nucleus and complete inhibition of PDGF-induced nuclear ERK and fibroblast proliferation. In contrast, iloprost caused a transient increase in nuclear cAMP, there was no effect of iloprost on PDGF-induced ERK in the nucleus, and this agonist was much less effective at reversing PDGF-induced proliferation. This suggests that sustained elevation of cAMP in the nucleus is necessary for efficient inhibition of PDGF-induced nuclear ERK and fibroblast proliferation. This is an important first step towards understanding of the signalling events that drive GPCR inhibition of fibrosis.
Collapse
Affiliation(s)
- Maxine J Roberts
- Cell Signalling Research Group, School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom.,Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Vic, Australia
| | - Lauren T May
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Vic, Australia
| | - Alastair C Keen
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Vic, Australia
| | - Bonan Liu
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Vic, Australia
| | - Terrance Lam
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Vic, Australia
| | - Steven J Charlton
- Cell Signalling Research Group, School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom.,Excellerate Bioscience Ltd., BioCity, Nottingham, United Kingdom
| | - Elizabeth M Rosethorne
- Cell Signalling Research Group, School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom
| | - Michelle L Halls
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Vic, Australia
| |
Collapse
|
7
|
Pang J, Qi X, Luo Y, Li X, Shu T, Li B, Song M, Liu Y, Wei D, Chen J, Wang J, Wang C. Multi-omics study of silicosis reveals the potential therapeutic targets PGD 2 and TXA 2. Am J Cancer Res 2021; 11:2381-2394. [PMID: 33500731 PMCID: PMC7797695 DOI: 10.7150/thno.47627] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 11/15/2020] [Indexed: 02/06/2023] Open
Abstract
Rationale: Silicosis is a severe occupational lung disease. Current treatments for silicosis have highly limited availability (i.e., lung transplantation) or, do not effectively prolong patient survival time (i.e., lung lavage). There is thus an urgent clinical need for effective drugs to retard the progression of silicosis. Methods: To systematically characterize the molecular changes associated with silicosis and to discover potential therapeutic targets, we conducted a transcriptomics analysis of human lung tissues acquired during transplantation, which was integrated with transcriptomics and metabolomics analyses of silicosis mouse lungs. The results from the multi-omics analyses were then verified by qPCR, western blot, and immunohistochemistry. The effect of Ramatroban on the progression of silicosis was evaluated in a silica-induced mouse model. Results: Wide metabolic alterations were found in lungs from both human patients and mice with silicosis. Targeted metabolite quantification and validation of expression of their synthases revealed that arachidonic acid (AA) pathway metabolites, prostaglandin D2 (PGD2) and thromboxane A2 (TXA2), were significantly up-regulated in silicosis lungs. We further examined the effect of Ramatroban, a clinical antagonist of both PGD2 and TXA2 receptors, on treating silicosis using a mouse model. The results showed that Ramatroban significantly alleviated silica-induced pulmonary inflammation, fibrosis, and cardiopulmonary dysfunction compared with the control group. Conclusion: Our results revealed the importance of AA metabolic reprogramming, especially PGD2 and TXA2 in the progression of silicosis. By blocking the receptors of these two prostanoids, Ramatroban may be a novel potential therapeutic drug to inhibit the progression of silicosis.
Collapse
|
8
|
Nikitopoulou I, Manitsopoulos N, Kotanidou A, Tian X, Petrovic A, Magkou C, Ninou I, Aidinis V, Schermuly RT, Kosanovic D, Orfanos SE. Orotracheal treprostinil administration attenuates bleomycin-induced lung injury, vascular remodeling, and fibrosis in mice. Pulm Circ 2019; 9:2045894019881954. [PMID: 31819797 PMCID: PMC6883672 DOI: 10.1177/2045894019881954] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 09/04/2019] [Indexed: 12/26/2022] Open
Abstract
Pulmonary fibrosis is a progressive disease characterized by disruption of lung architecture and deregulation of the pulmonary function. Prostacyclin, a metabolite of arachidonic acid, is a potential disease mediator since it exerts anti-inflammatory and anti-fibrotic actions. We investigated the effect of treprostinil, a prostacyclin analogue, in bleomycin-induced experimental pulmonary fibrosis. Bleomycin sulfate or saline was administrated intratracheally to mice (n = 9-10/group) at day 0. Orotracheal aspiration of treprostinil or vehicle was administered daily and started 24 h prior to bleomycin challenge. Evaluation of lung pathology was performed in tissue samples and bronchoalveolar lavage fluid collected 7, 14 and 21 days after bleomycin exposure. Lung injury was achieved due to bleomycin exposure at all time points as indicated by impaired lung mechanics, pathologic lung architecture (from day 14), and cellular and protein accumulation in the alveolar space accompanied by a minor decrease in lung tissue VE-cadherin at day 14. Treprostinil preserved lung mechanics, and reduced lung inflammation, fibrosis, and vascular remodeling (day 21); reduced cellularity and protein content of bronchoalveolar lavage fluid were additionally observed with no significant effect on VE-cadherin expression. Bleomycin-induced collagen deposition was attenuated by treprostinil from day 14, while treprostinil involvement in regulating inflammatory processes appears mediated by NF-κB signaling. Overall, prophylactic administration of treprostinil, a stable prostacyclin analogue, maintained lung function, and prevented bleomycin-induced lung injury, and fibrosis, as well as vascular remodeling, a hallmark of pulmonary hypertension. This suggests potential therapeutic efficacy of treprostinil in pulmonary fibrosis and possibly in pulmonary hypertension related to chronic lung diseases.
Collapse
Affiliation(s)
- Ioanna Nikitopoulou
- GP Livanos and M Simou Laboratories,1st
Department of Critical Care & Pulmonary Services, Medical School, National &
Kapodistrian University of Athens, Evangelismos Hospital, Athens, Greece
| | - Nikolaos Manitsopoulos
- GP Livanos and M Simou Laboratories,1st
Department of Critical Care & Pulmonary Services, Medical School, National &
Kapodistrian University of Athens, Evangelismos Hospital, Athens, Greece
| | - Anastasia Kotanidou
- GP Livanos and M Simou Laboratories,1st
Department of Critical Care & Pulmonary Services, Medical School, National &
Kapodistrian University of Athens, Evangelismos Hospital, Athens, Greece
- 1st Department of Critical Care &
Pulmonary Services, Medical School, National & Kapodistrian University of
Athens, Evangelismos Hospital, Athens, Greece
| | - Xia Tian
- Universities of Giessen and Marburg Lung
Center, Member of the German Center for Lung Research (DZL), Justus-Liebig
University, Giessen, Germany
| | - Aleksandar Petrovic
- Universities of Giessen and Marburg Lung
Center, Member of the German Center for Lung Research (DZL), Justus-Liebig
University, Giessen, Germany
| | | | - Ioanna Ninou
- Institute of Immunology, Biomedical
Sciences Research Center Alexander Fleming, Athens, Greece
| | - Vassilis Aidinis
- Institute of Immunology, Biomedical
Sciences Research Center Alexander Fleming, Athens, Greece
| | - Ralph T. Schermuly
- Universities of Giessen and Marburg Lung
Center, Member of the German Center for Lung Research (DZL), Justus-Liebig
University, Giessen, Germany
| | - Djuro Kosanovic
- Universities of Giessen and Marburg Lung
Center, Member of the German Center for Lung Research (DZL), Justus-Liebig
University, Giessen, Germany
- Sechenov First Moscow State Medical
University
(Sechenov
University), Moscow, Russia
| | - Stylianos E. Orfanos
- GP Livanos and M Simou Laboratories,1st
Department of Critical Care & Pulmonary Services, Medical School, National &
Kapodistrian University of Athens, Evangelismos Hospital, Athens, Greece
- 1st Department of Critical Care &
Pulmonary Services, Medical School, National & Kapodistrian University of
Athens, Evangelismos Hospital, Athens, Greece
- 2nd Department of Critical Care, Medical
School, National & Kapodistrian University of Athens, “Attikon” Hospital,
Haidari, Athens, Greece
| |
Collapse
|
9
|
Mamazhakypov A, Schermuly RT, Schaefer L, Wygrecka M. Lipids - two sides of the same coin in lung fibrosis. Cell Signal 2019; 60:65-80. [PMID: 30998969 DOI: 10.1016/j.cellsig.2019.04.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 04/07/2019] [Accepted: 04/12/2019] [Indexed: 12/16/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is characterized by progressive extracellular matrix deposition in the lung parenchyma leading to the destruction of lung structure, respiratory failure and premature death. Recent studies revealed that the pathogenesis of IPF is associated with alterations in the synthesis and the activity of lipids, lipid regulating proteins and cell membrane lipid transporters and receptors in different lung cells. Furthermore, deregulated lipid metabolism was found to contribute to the profibrotic phenotypes of lung fibroblasts and alveolar epithelial cells. Consequently, several pharmacological agents, targeting lipids, lipid mediators, and lipoprotein receptors, was successfully tested in the animal models of lung fibrosis and entered early phase clinical trials. In this review, we highlight new therapeutic options to counteract disturbed lipid hemostasis in the maladaptive lung remodeling.
Collapse
Affiliation(s)
- Argen Mamazhakypov
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center, Giessen, Germany.
| | - Ralph T Schermuly
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center, Giessen, Germany.
| | - Liliana Schaefer
- Goethe University School of Medicine, Frankfurt am Main, Germany.
| | - Malgorzata Wygrecka
- Department of Biochemistry, Universities of Giessen and Marburg Lung Center, Giessen, Germany.
| |
Collapse
|
10
|
Zhu J, Chaki M, Lu D, Ren C, Wang SS, Rauhauser A, Li B, Zimmerman S, Jun B, Du Y, Vadnagara K, Wang H, Elhadi S, Quigg RJ, Topham MK, Mohan C, Ozaltin F, Zhou XJ, Marciano DK, Bazan NG, Attanasio M. Loss of diacylglycerol kinase epsilon in mice causes endothelial distress and impairs glomerular Cox-2 and PGE2 production. Am J Physiol Renal Physiol 2016; 310:F895-F908. [PMID: 26887830 PMCID: PMC4867310 DOI: 10.1152/ajprenal.00431.2015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 01/23/2016] [Indexed: 12/12/2022] Open
Abstract
Thrombotic microangiopathy (TMA) is a disorder characterized by microvascular occlusion that can lead to thrombocytopenia, hemolytic anemia, and glomerular damage. Complement activation is the central event in most cases of TMA. Primary forms of TMA are caused by mutations in genes encoding components of the complement or regulators of the complement cascade. Recently, we and others have described a genetic form of TMA caused by mutations in the gene diacylglycerol kinase-ε (DGKE) that encodes the lipid kinase DGKε (Lemaire M, Fremeaux-Bacchi V, Schaefer F, Choi MR, Tang WH, Le Quintrec M, Fakhouri F, Taque S, Nobili F, Martinez F, Ji WZ, Overton JD, Mane SM, Nurnberg G, Altmuller J, Thiele H, Morin D, Deschenes G, Baudouin V, Llanas B, Collard L, Majid MA, Simkova E, Nurnberg P, Rioux-Leclerc N, Moeckel GW, Gubler MC, Hwa J, Loirat C, Lifton RP. Nat Genet 45: 531-536, 2013; Ozaltin F, Li BH, Rauhauser A, An SW, Soylemezoglu O, Gonul II, Taskiran EZ, Ibsirlioglu T, Korkmaz E, Bilginer Y, Duzova A, Ozen S, Topaloglu R, Besbas N, Ashraf S, Du Y, Liang CY, Chen P, Lu DM, Vadnagara K, Arbuckle S, Lewis D, Wakeland B, Quigg RJ, Ransom RF, Wakeland EK, Topham MK, Bazan NG, Mohan C, Hildebrandt F, Bakkaloglu A, Huang CL, Attanasio M. J Am Soc Nephrol 24: 377-384, 2013). DGKε is unrelated to the complement pathway, which suggests that unidentified pathogenic mechanisms independent of complement dysregulation may result in TMA. Studying Dgke knockout mice may help to understand the pathogenesis of this disease, but no glomerular phenotype has been described in these animals so far. Here we report that Dgke null mice present subclinical microscopic anomalies of the glomerular endothelium and basal membrane that worsen with age and develop glomerular capillary occlusion when exposed to nephrotoxic serum. We found that induction of cyclooxygenase-2 and of the proangiogenic prostaglandin E2 are impaired in Dgke null kidneys and are associated with reduced expression of the antithrombotic cell adhesion molecule platelet endothelial cell adhesion molecule-1/CD31 in the glomerular endothelium. Notably, prostaglandin E2 supplementation was able to rescue motility defects of Dgke knockdown cells in vitro and to restore angiogenesis in a test in vivo. Our results unveil an unexpected role of Dgke in the induction of cyclooxygenase-2 and in the regulation of glomerular prostanoids synthesis under stress.
Collapse
Affiliation(s)
- Jili Zhu
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas; Department of Nephrology, Renmin Hospital, Wuhan University, Hubei, Wuhan, China
| | - Moumita Chaki
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas; Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Dongmei Lu
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Chongyu Ren
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Shan-Shan Wang
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Alysha Rauhauser
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Binghua Li
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Susan Zimmerman
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Bokkyoo Jun
- Department of Neuroscience, Louisiana State University, New Orleans, Louisiana
| | - Yong Du
- Biomedical Engineering, University of Houston, Houston, Texas
| | - Komal Vadnagara
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Hanquin Wang
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas; Institute of Basic Medical Sciences, Hubei University of Medicine, Hubei, Shiyan, China
| | - Sarah Elhadi
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas; Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Richard J Quigg
- Department of Medicine, University of Buffalo, Buffalo, New York
| | - Matthew K Topham
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Chandra Mohan
- Biomedical Engineering, University of Houston, Houston, Texas
| | - Fatih Ozaltin
- Department of Pediatric Nephrology, Faculty of Medicine, Hacettepe University, Ankara, Turkey; Nephrogenetics Laboratory, Department of Pediatric Nephrology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Xin J Zhou
- Renal Path Diagnostics, Pathologist BioMedical Laboratories and Department of Pathology, Baylor University Medical Center, Dallas, Texas; and
| | - Denise K Marciano
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Nicolas G Bazan
- Department of Neuroscience, Louisiana State University, New Orleans, Louisiana
| | - Massimo Attanasio
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas; Eugene McDermott Center for Growth and Development, The University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
11
|
Kandhare AD, Bodhankar SL, Mohan V, Thakurdesai PA. Effect of glycosides based standardized fenugreek seed extract in bleomycin-induced pulmonary fibrosis in rats: Decisive role of Bax, Nrf2, NF-κB, Muc5ac, TNF-α and IL-1β. Chem Biol Interact 2015; 237:151-65. [PMID: 26093215 DOI: 10.1016/j.cbi.2015.06.019] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 06/06/2015] [Accepted: 06/10/2015] [Indexed: 12/19/2022]
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a chronic progressive multifactorial disease with limited therapeutic options. Glycosides based standardized fenugreek seed extract (SFSE-G) possesses potent anti-inflammatory and anti-oxidant property. AIM To evaluate the efficacy of SFSE-G against bleomycin (BLM) induced pulmonary fibrosis by assessing behavioral, biochemical, molecular and ultrastructural changes in the laboratory rats. MATERIALS AND METHODS IPF was induced in male Sprague-Dawley rats by single intratracheal BLM (6IU/kg) injection followed by SFSE-G (5, 10, 20 and 40mg/kg, p.o.) or methylprednisolone (10mg/kg, p.o.) treatment for 28day. Various parameters were analyzed in lung and bronchoalveolar lavage fluid (BALF) after 14 and 28days of the drug treatment. RESULTS SFSE-G (20 and 40mg/kg, p.o.) administration significantly prevented the BLM induced alteration in body weight, lung index, lung function test and hematology. The altered total and differential cell count in BALF and blood was significantly prevented by SFSE-G treatment. The decreased peripheral blood oxygen content after BLM instillation was significantly increased by SFSE-G treatment. SFSE-G significantly enhanced the BALF and lung antioxidant status, through modulating the SOD, GSH, T-AOC, MDA, NO level and Nrf2, HO-1 mRNA expression. There was a significant reduction in lung 5-HT level by SFSE-G treatment. The altered mRNA expression of biomarkers of lung inflammation (TNF-α, IL-1β, IL-6 and IL-8), fibrosis (TGF-β, collagen-1, ET-1, Muc5ac, NF-κB, VEGF, Smad-3) and apoptosis (Bax, Bcl-2 and Caspase-3) were significantly prevented by SFSE-G treatment. BLM induced histological inflammatory and fibrotic insult in the lung were reduced by SFSE-G treatment. It also ameliorated BLM induced lung ultrastructural changes as observed by transmission electron microscopic studies. However, administration of SFSE-G (5mg/kg, p.o.) failed to show any protective effect against BLM-induced PF whereas SFSE-G (10mg/kg, p.o.) showed significant amelioration in BLM-induced PF except lung function test, BALF and lung antioxidant level. CONCLUSION SFSE-G showed anti-fibrotic efficacy executed through induction of Nrf2, which in turn may modulate anti-inflammatory molecules, inhibit fibrogenic molecules and decreased apoptosis to ameliorate BLM induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Amit D Kandhare
- Department of Pharmacology, Poona College of Pharmacy, Bharati Vidyapeeth Deemed University, Erandwane, Paud Road, Pune 411 038, India
| | - Subhash L Bodhankar
- Department of Pharmacology, Poona College of Pharmacy, Bharati Vidyapeeth Deemed University, Erandwane, Paud Road, Pune 411 038, India.
| | - Vishwaraman Mohan
- Indus Biotech Private Limited, 1, Rahul Residency, Off Salunke Vihar Road, Kondhwa, Pune 411 048, India
| | - Prasad A Thakurdesai
- Indus Biotech Private Limited, 1, Rahul Residency, Off Salunke Vihar Road, Kondhwa, Pune 411 048, India
| |
Collapse
|
12
|
Pulmonary vascular changes in asthma and COPD. Pulm Pharmacol Ther 2014; 29:144-55. [DOI: 10.1016/j.pupt.2014.09.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 09/10/2014] [Accepted: 09/12/2014] [Indexed: 12/11/2022]
|
13
|
Prostaglandin E2 switches from a stimulator to an inhibitor of cell migration after epithelial-to-mesenchymal transition. Prostaglandins Other Lipid Mediat 2014; 116-117:1-9. [PMID: 25460827 DOI: 10.1016/j.prostaglandins.2014.10.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 10/07/2014] [Accepted: 10/15/2014] [Indexed: 01/18/2023]
Abstract
Epithelial-mesenchymal transition (EMT) is critical for embryonic development, and this process is recapitulated in adults during wound healing, tissue regeneration, fibrosis and cancer progression. Cell migration is believed to play a key role in both normal wound repair and in abnormal tissue remodeling. Prostaglandin E2 (PGE2) inhibits fibroblast chemotaxis, but stimulates chemotaxis in airway epithelial cells. The current study was designed to explore the role of PGE2 and its four receptors on airway epithelial cell migration following EMT using both the Boyden blindwell chamber chemotaxis assay and the wound closure assay. EMT in human bronchial epithelial cells (HBECs) was induced by TGF-β1 and a mixture of cytokines (IL-1β, TNF-α, and IFN-γ). PGE2 and selective agonists for all four EP receptors stimulated chemotaxis and wound closure in HBECs. Following EMT, the EP1 and EP3 agonists were without effect, while the EP2 and EP4 agonists inhibited chemotaxis as did PGE2. The effects of the EP2 and EP4 receptors on HBEC and EMT cell migration were further confirmed by blocking the expected signaling pathways. Taken together, these results demonstrate that PGE2 switches from a stimulator to an inhibitor of cell migration following EMT of airway epithelial cells and that this inhibition is mediated by an altered effect of EP2 and EP4 signaling and an apparent loss of the stimulatory effects of EP1 and EP3. Change in the PGE2 modulation of chemotaxis may play a role in repair following injury.
Collapse
|
14
|
Prasad S, Hogaboam CM, Jarai G. Deficient repair response of IPF fibroblasts in a co-culture model of epithelial injury and repair. FIBROGENESIS & TISSUE REPAIR 2014; 7:7. [PMID: 24834127 PMCID: PMC4021590 DOI: 10.1186/1755-1536-7-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 04/17/2014] [Indexed: 12/28/2022]
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a progressive disorder marked by relentless fibrosis and damage of the lung architecture. A growing body of evidence now suggests that IPF progresses as a result of aberrant epithelial-fibroblast crosstalk. Injured epithelia are a major source of growth factors such as PDGF which guide resident fibroblasts to injury sites. RESULTS In this study, we utilized a novel co-culture system to investigate the effect of fibroblast phenotype on their response to epithelial injury. Fibroblasts from normal lungs (NHLF) responded to epithelial injury and populated the wound site forming a fibroblast plug/mechanical barrier which prevented epithelial wound closure. IPF fibroblasts were impaired in their response to epithelial injury. They also expressed reduced PDGFRα compared to NHLFs and were defective towards PDGF-AA mediated directional movement. Neutralization of PDGF-AA and pan-PDGF but not PDGF-BB reduced the injury response of NHLFs thereby preventing the formation of the mechanical barrier and promoting epithelial wound closure. Co-culture of epithelial cells with IPF fibroblasts led to marked increase in the levels of pro-fibrotic growth factors - bFGF and PDGF and significant depletion of anti-fibrotic HGF in the culture medium. Furthermore, IPF fibroblasts but not NHLFs induced a transient increase in mesenchymal marker expression in the wound lining epithelial cells. This was accompanied by increased migration and faster wound closure in co-cultures with IPF fibroblasts. CONCLUSIONS Our data demonstrate that the IPF fibroblasts have an aberrant repair response to epithelial injury.
Collapse
Affiliation(s)
- Sony Prasad
- Novartis Institutes of Biomedical Research, Respiratory Disease Area, Wimblehurst Road, Horsham, West Sussex RH12 5AB, UK
| | - Cory M Hogaboam
- Department of Medicine, Cedars Sinai Medical Center, AHSP Room A9108, 127 S. San Vicente Blvd, Los Angeles, CA 90048-3311, USA
| | - Gabor Jarai
- Novartis Institutes of Biomedical Research, Respiratory Disease Area, Wimblehurst Road, Horsham, West Sussex RH12 5AB, UK
| |
Collapse
|
15
|
Larsson-Callerfelt AK, Hallgren O, Andersson-Sjöland A, Thiman L, Björklund J, Kron J, Nihlberg K, Bjermer L, Löfdahl CG, Westergren-Thorsson G. Defective alterations in the collagen network to prostacyclin in COPD lung fibroblasts. Respir Res 2013; 14:21. [PMID: 23406566 PMCID: PMC3585859 DOI: 10.1186/1465-9921-14-21] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 02/11/2013] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Prostacyclin analogs are potent vasodilators and possess anti-inflammatory properties. However, the effect of prostacyclin on extracellular matrix (ECM) in COPD is not well known. Collagen fibrils and proteoglycans are essential ECM components in the lung and fibroblasts are key players in regulating the homeostasis of ECM proteins. The aim was to study the synthesis of prostacyclin and its effect on fibroblast activity and ECM production, and in particular collagen I and the collagen-associated proteoglycans biglycan and decorin. METHODS Parenchymal lung fibroblasts were isolated from lungs from COPD patients (GOLD stage IV) and from lungs and transbronchial biopsies from control subjects. The prostacyclin analog iloprost was used to study the effect of prostacyclin on ECM protein synthesis, migration, proliferation and contractile capacity of fibroblasts. RESULTS TGF-β1 stimulation significantly increased prostacyclin synthesis in fibroblasts from COPD patients (p < 0.01), but showed no effect on fibroblasts from control subjects. Collagen I synthesis was decreased by iloprost in both control and COPD fibroblasts (p < 0.05). Conversely, iloprost significantly altered biglycan and decorin synthesis in control fibroblasts, but iloprost displayed no effect on these proteoglycans in COPD fibroblasts. Proliferation rate was reduced (p < 0.05) and contractile capacity was increased in COPD fibroblasts (p < 0.05) compared to control fibroblasts. Iloprost decreased proliferative rate in control fibroblasts (p < 0.05), whereas iloprost attenuated contraction capacity in both COPD (p < 0.01) and control fibroblasts (p < 0.05). CONCLUSIONS Iloprost reduced collagen I synthesis and fibroblast contractility but did not affect the collagen-associated proteoglycans or proliferation rate in fibroblasts from COPD patients. Enhanced prostacyclin production could lead to improper collagen network fibrillogenesis and a more emphysematous lung structure in severe COPD patients.
Collapse
|
16
|
Aytemur ZA, Hacievliyagil SS, Iraz M, Samdanci E, Ozerol E, Kuku I, Nurkabulov Z, Yildiz K. Effects of iloprost on bleomycin-induced pulmonary fibrosis in rats compared with methyl-prednisolone. REVISTA PORTUGUESA DE PNEUMOLOGIA 2012; 18:272-7. [PMID: 22770817 DOI: 10.1016/j.rppneu.2012.04.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Revised: 04/17/2012] [Accepted: 04/29/2012] [Indexed: 01/29/2023] Open
Abstract
OBJECTIVE Prostacyclin (PGI2) has been shown to inhibit the expression of pro-inflammatory and pro-fibrotic mediators in pulmonary fibrosis. In this study, we aimed to test the preventive effects of intraperitoneally administered iloprost, a stable PGI2 analog, on bleomycin-induced pulmonary fibrosis in rats and to compare the effects of iloprost with the effects of methyl-prednisolone, a traditional therapy. METHODS Rats were randomly allocated into four groups: 1. Saline alone (n=6); 2. Bleomycin+placebo (n=7); 3. Bleomycin+methyl-prednisolone (n=7); 4. Bleomycin+iloprost (n=7). Fibrotic changes in the lungs were demonstrated by analyzing the cellular composition of bronchoalveolar lavage fluid, histological evaluation and lung hydroxyproline content. RESULTS Fibrosis was made in the lungs of rats by bleomycin experimentally. Fibrosis scores in the methyl-prednisolone and the iloprost groups were significantly lower than in the placebo group (p<0.05). Furthermore, the score of the iloprost group was significantly lower than the score of the methyl-prednisolone group. The hydroxyproline content was significantly less in the methyl-prednisolone and the iloprost groups (p<0.05). In the placebo group, the neutrophil percentage in bronchoalveolar lavage was significantly higher than in the other groups, whereas the macrophage percentage in placebo group was significantly lower (p<0.05). CONCLUSION Iloprost has protective effect on the pulmonary fibrosis induced by bleomycin and it may be more effective in decreasing fibrotic changes than methyl-prednisolone.
Collapse
Affiliation(s)
- Z A Aytemur
- Department of Pulmonary Disease, Inonu University Medical Faculty, Malatya, Turkey
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Moore BB, Peters-Golden M. Opposing roles of leukotrienes and prostaglandins in fibrotic lung disease. Expert Rev Clin Immunol 2010; 2:87-100. [PMID: 20477090 DOI: 10.1586/1744666x.2.1.87] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Lung fibrosis is a devastating disease that involves a variable degree of inflammation, alveolar epithelial injury, fibroblast hyperplasia and the deposition of extracellular matrix. Standard therapies that consist of corticosteroids and immunosuppressive agents offer little benefit and most patients experience a progressive deterioration in lung function which is ultimately fatal within 2-5 years of diagnosis. New pathogenetic insights and therapeutic approaches are badly needed. Eicosanoids are lipid mediators derived from arachidonic acid metabolism, the best studied of which are prostaglandins and leukotrienes. Although these mediators are primarily known for their roles in asthma, pain, fever and vascular responses, they also exert relevant effects on immune and inflammatory cells as well as structural cells such as epithelial cells and fibroblasts - cell types which participate in fibrogenesis. In general, leukotrienes promote while prostaglandin E(2) opposes fibrogenic responses. Lung fibrosis is associated with increased production of leukotrienes and decreased production of prostaglandin E(2). Furthermore, responses to prostaglandin E(2) are altered in fibrotic conditions. This review highlights the role of this leukotriene/prostaglandin imbalance in the evolution of fibrotic lung disease, offers insights into the mechanisms that underlie the dysregulated responses and discusses approaches for therapeutic targeting of eicosanoids in these conditions.
Collapse
Affiliation(s)
- Bethany B Moore
- Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School, 6220 MSRB III, 1150 W. Medical Center Drive, Ann Arbor, MI 48109-0642, USA.
| | | |
Collapse
|
18
|
Kurokawa S, Suda M, Okuda T, Miyake Y, Matsumura Y, Ishimura M, Saito R, Nakamura T. Effect of inhaled KP-496, a novel dual antagonist of the cysteinyl leukotriene and thromboxane A2 receptors, on a bleomycin-induced pulmonary fibrosis model in mice. Pulm Pharmacol Ther 2010; 23:425-31. [PMID: 20457270 DOI: 10.1016/j.pupt.2010.04.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2009] [Revised: 04/06/2010] [Accepted: 04/30/2010] [Indexed: 11/18/2022]
Abstract
Cysteinyl-leukotrienes (cysLTs) and thromboxane A(2) (TXA(2)) are important mediators in inflammatory lung diseases such as bronchial asthma and idiopathic pulmonary fibrosis (IPF). We examined the effects of inhaled KP-496, a novel dual antagonist of the cysLTs and TXA(2) receptors, on bleomycin-induced IPF in mice. Mice were intravenously injected bleomycin on day 0, and 0.5% of KP-496 was inhaled twice a day (30 min/time) for the entire experimental period. The effects of KP-496 were evaluated by the number of infiltrated cells in bronchoalveolar lavage fluid (BALF), hydroxyl-L-proline content in the lung, and histopathology. Analyses of BALF on days 7 and 21 revealed that inhaled KP-496 significantly decreased total cell numbers, macrophages, neutrophils, and eosinophils on both days. KP-496 significantly decreased hydroxyl-L-proline content in the lung on day 21. Histopathological analyses of lungs on day 21 demonstrated that KP-496 significantly suppressed inflammatory and fibrotic changes. Our results suggested that the suppression of cysLTs and TXA(2) pathways by KP-496 could control airway inflammation and pulmonary fibrosis, and that KP-496 could be a new therapeutic agent for lung diseases with inflammation and fibrogenesis such as IPF and chronic obstructive pulmonary disease.
Collapse
MESH Headings
- Administration, Inhalation
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/administration & dosage
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Benzoates/administration & dosage
- Benzoates/pharmacology
- Bleomycin
- Bronchoalveolar Lavage Fluid/cytology
- Disease Models, Animal
- Inflammation/metabolism
- Inflammation/pathology
- Inflammation/prevention & control
- Leukotriene Antagonists/administration & dosage
- Leukotriene Antagonists/pharmacology
- Male
- Mice
- Mice, Inbred ICR
- Pulmonary Fibrosis/chemically induced
- Pulmonary Fibrosis/prevention & control
- Receptors, Leukotriene/metabolism
- Receptors, Thromboxane A2, Prostaglandin H2/antagonists & inhibitors
- Receptors, Thromboxane A2, Prostaglandin H2/metabolism
- Thiazoles/administration & dosage
- Thiazoles/pharmacology
Collapse
Affiliation(s)
- Shigeo Kurokawa
- Pharmacology Department, Central Research Laboratories, Kaken Pharmaceutical Co., Ltd., Yamashina-ku, Kyoto, Japan
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Zhu Y, Liu Y, Zhou W, Xiang R, Jiang L, Huang K, Xiao Y, Guo Z, Gao J. A prostacyclin analogue, iloprost, protects from bleomycin-induced pulmonary fibrosis in mice. Respir Res 2010; 11:34. [PMID: 20302663 PMCID: PMC2848635 DOI: 10.1186/1465-9921-11-34] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2009] [Accepted: 03/20/2010] [Indexed: 12/16/2022] Open
Abstract
Background Metabolites of arachidonic acid such as prostacyclin (PGI2) have been shown to participate in the pathogenesis of pulmonary fibrosis by inhibiting the expression of pro-inflammatory and pro-fibrotic mediators. In this investigation, we examined whether iloprost, a stable PGI2 analogue, could prevent bleomycin-induced pulmonary inflammation and fibrosis in a mouse model. Methods Mice received a single intratracheal injection of bleomycin with or without intraperitoneal iloprost. Pulmonary inflammation and fibrosis were analysed by histological evaluation, cellular composition of bronchoalveolar lavage (BAL) fluid, and hydroxyproline content. Lung mechanics were measured. We also analysed the expression of inflammatory mediators in BAL fluid and lung tissue. Results Administration of iloprost significantly improved survival rate and reduced weight loss in the mice induced by bleomycin. The severe inflammatory response and fibrotic changes were significantly attenuated in the mice treated with iloprost as shown by reduction in infiltration of inflammatory cells into the airways and pulmonary parenchyma, diminution in interstitial collagen deposition, and lung hydroxyproline content. Iloprost significantly improved lung static compliance and tissue elastance. It increased the expression of IFNγ and CXCL10 in lung tissue measured by RT-PCR and their levels in BAL fluid as measured by ELISA. Levels of TNFα, IL-6 and TGFβ1 were lowered by iloprost. Conclusions Iloprost prevents bleomycin-induced pulmonary fibrosis, possibly by upregulating antifibrotic mediators (IFNγ and CXCL10) and downregulating pro-inflammatory and pro-fibrotic cytokines (TNFα, IL-6, and TGFβ1). Prostacyclin may represent a novel pharmacological agent for treating pulmonary fibrotic diseases.
Collapse
Affiliation(s)
- Yuanjue Zhu
- Department of Respiratory Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Li YJ, Wang XQ, Sato T, Kanaji N, Nakanishi M, Kim M, Michalski J, Nelson AJ, Sun JH, Farid M, Basma H, Patil A, Toews ML, Liu X, Rennard SI. Prostaglandin E₂ inhibits human lung fibroblast chemotaxis through disparate actions on different E-prostanoid receptors. Am J Respir Cell Mol Biol 2010; 44:99-107. [PMID: 20203295 DOI: 10.1165/rcmb.2009-0163oc] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The migration of fibroblasts is believed to play a key role in both normal wound repair and abnormal tissue remodeling. Prostaglandin E (PGE)(2), a mediator that can inhibit many fibroblast functions including chemotaxis, was reported to be mediated by the E-prostanoid (EP) receptor EP2. PGE(2), however, can act on four receptors. This study was designed to determine if EP receptors, in addition to EP2, can modulate fibroblast chemotaxis. Using human fetal lung fibroblasts, the expression of all four EP receptors was demonstrated by Western blotting. EP2-selective and EP4-selective agonists inhibited both chemotaxis toward fibronectin in the blindwell assay and migration in a wound-closure assay. In contrast, EP1-selective and EP3-selective agonists stimulated cell migration in both assay systems. These results were confirmed using EP-selective antagonists. The role of both EP2 and EP4 receptors in mediating the PGE(2) inhibition of chemotaxis was also confirmed by small interfering RNA suppression. Furthermore, the role of EP receptors was confirmed by blocking the expected signaling pathways. Taken together, these results demonstrate that PGE(2) can act on multiple EP receptors in human lung fibroblasts, to exert disparate effects. Alterations in EP receptor expression may have the potential to alter PGE(2) action. Targeting specific EP receptors may offer therapeutic opportunities in conditions characterized by abnormal tissue repair and remodeling.
Collapse
Affiliation(s)
- Ying-Ji Li
- Department of Hygiene and Public Health, Nippon Medical School, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Iyer A, Fenning A, Lim J, Le GT, Reid RC, Halili MA, Fairlie DP, Brown L. Antifibrotic activity of an inhibitor of histone deacetylases in DOCA-salt hypertensive rats. Br J Pharmacol 2010; 159:1408-17. [PMID: 20180942 DOI: 10.1111/j.1476-5381.2010.00637.x] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND AND PURPOSE Histone deacetylases (HDACs) silence genes by deacetylating lysine residues in histones and other proteins. HDAC inhibitors represent a new class of compounds with anti-inflammatory activity. This study investigated whether treatment with a broad spectrum HDAC inhibitor, suberoylanilide hydroxamic acid (SAHA), would prevent cardiac fibrosis, part of the cardiovascular remodelling in deoxycorticosterone acetate (DOCA)-salt rats. EXPERIMENTAL APPROACH Control and DOCA-salt rats were treated with SAHA (25 mg x kg(-1) x day(-1) s.c.) for 32 days. Changes in cardiovascular structure and function were assessed by blood pressure in vivo and in Langendorff perfused hearts, ventricular papillary muscle and in aortic rings in vitro. Left ventricular collagen deposition was assessed by histology. KEY RESULTS Administration of SAHA to DOCA-salt rats attenuated the following parameters: the increased concentration of over 20 pro-inflammatory cytokines in plasma, increased inflammatory cell infiltration and interstitial collagen deposition, increased passive diastolic stiffness in perfused hearts, prolongation of action potential duration at 20% and 90% of repolarization in papillary muscle, development of left ventricular hypertrophy, systolic hypertension and changes in vascular dysfunction. CONCLUSIONS AND IMPLICATIONS The HDAC inhibitor, SAHA, attenuated the cardiovascular remodelling associated with DOCA-salt hypertensive rats and improved cardiovascular structure and function, especially fibrosis, in the heart and blood vessels, possibly by suppressing inflammation. Control of cardiac histone or non-histone protein acetylation is a potential therapeutic approach to preventing cardiac remodelling, especially cardiac fibrosis.
Collapse
Affiliation(s)
- Abishek Iyer
- School of Biomedical Sciences, The University of Queensland, Brisbane, Qld, Australia
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Treprostinil to reverse pulmonary hypertension associated with idiopathic pulmonary fibrosis as a bridge to single-lung transplantation. J Heart Lung Transplant 2010; 28:964-7. [PMID: 19716051 DOI: 10.1016/j.healun.2009.05.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2008] [Revised: 05/09/2009] [Accepted: 05/09/2009] [Indexed: 11/23/2022] Open
Abstract
The available therapies and prognosis of idiopathic pulmonary fibrosis remain relatively poor, and concurrent pulmonary hypertension further increases the risk of death and complications after lung transplantation. Limited data exist for the treatment of pulmonary hypertension associated with idiopathic pulmonary fibrosis. We describe a case where intravenous treprostinil was used to bridge an elderly patient with idiopathic pulmonary fibrosis with severe pulmonary hypertension to successful single-lung transplantation.
Collapse
|
23
|
Murray DL, Johnson EN, Wang P, Gauthier J, Bing N, Vanderwall D, Xu Z, Jensen D, Cox RF, Kostura M, Sorensen SD. Functional antagonism of IL-1alpha induced gene expression profiles define the cAMP/PKA pathway as a unique regulator of IL-1alpha signaling networks. J Recept Signal Transduct Res 2009; 29:246-56. [PMID: 19627252 DOI: 10.1080/10799890903078473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Interleukin-1 (IL-1alpha) induced inflammatory and pro-fibrotic responses in human lung fibroblasts are mediated by activation of MAPK and NFkappaB pathways. The purpose of the present study was to broadly profile the activity of a variety of compounds which function as inhibitors of these key signaling pathways that may affect IL-1alpha mediated gene changes. A reference set of genes was derived from microarray analysis of IL-1alpha stimulated cells. The genes were chosen to provide a range of expression profiles which serve to represent the actions of the underlying signaling network. We show that G(s)-coupled receptor agonists have a unique pattern of activity as represented by their impact on IL-1alpha dependent gene changes. These effects were not mimicked by direct inhibitors of p38, JNK, MEK or IKK but were mimicked by forskolin and cAMP analogs. These findings indicate that cAMP/PKA serves as a point of convergence for regulation of IL-1alpha responses by multiple G(s)-coupled receptors and regulates IL-1alpha responses by a distinct mechanism that does not solely involve direct inhibition of p38, JNK, MEK or IKK. The data also point to a potentially useful paradigm wherein monitoring of a small subset of genes is sufficient to identify pathway activity of novel compounds.
Collapse
Affiliation(s)
- David L Murray
- Discovery Research and Chemoinformatics, GlaxoSmithKline, Research Triangle Park, NC 27709, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
KANAZAWA S, KINOSHITA Y, YAMAGUCHI K, NOMURA S. Gefitinib-induced autologous antitumor immunity. Asia Pac J Clin Oncol 2009. [DOI: 10.1111/j.1743-7563.2009.01201.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
25
|
Sokolova E, Hartig R, Reiser G. Downregulation of protease-activated receptor-1 in human lung fibroblasts is specifically mediated by the prostaglandin E receptor EP2 through cAMP elevation and protein kinase A. FEBS J 2008; 275:3669-79. [PMID: 18537828 DOI: 10.1111/j.1742-4658.2008.06511.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Many cellular functions of lung fibroblasts are controlled by protease-activated receptors (PARs). In fibrotic diseases, PAR-1 plays a major role in controlling fibroproliferative and inflammatory responses. Therefore, in these diseases, regulation of PAR-1 expression plays an important role. Using the selective prostaglandin EP2 receptor agonist butaprost and cAMP-elevating agents, we show here that prostaglandin (PG)E(2), via the prostanoid receptor EP2 and subsequent cAMP elevation, downregulates mRNA and protein levels of PAR-1 in human lung fibroblasts. Under these conditions, the functional response of PAR-1 in fibroblasts is reduced. These effects are specific for PGE(2). Activation of other receptors coupled to cAMP elevation, such as beta-adrenergic and adenosine receptors, does not reproduce the effects of PGE(2). PGE(2)-mediated downregulation of PAR-1 depends mainly on protein kinase A activity, but does not depend on another cAMP effector, the exchange protein activated by cAMP. PGE(2)-induced reduction of PAR-1 level is not due to a decrease of PAR-1 mRNA stability, but rather to transcriptional regulation. The present results provide further insights into the therapeutic potential of PGE(2) to specifically control fibroblast function in fibrotic diseases.
Collapse
Affiliation(s)
- Elena Sokolova
- Institut für Neurobiochemie, Medizinische Fakultät, Otto-von-Guericke-Universität Magdeburg, Germany
| | | | | |
Collapse
|
26
|
Togo S, Holz O, Liu X, Sugiura H, Kamio K, Wang X, Kawasaki S, Ahn Y, Fredriksson K, Skold CM, Mueller KC, Branscheid D, Welker L, Watz H, Magnussen H, Rennard SI. Lung fibroblast repair functions in patients with chronic obstructive pulmonary disease are altered by multiple mechanisms. Am J Respir Crit Care Med 2008; 178:248-60. [PMID: 18467512 PMCID: PMC2542423 DOI: 10.1164/rccm.200706-929oc] [Citation(s) in RCA: 154] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2007] [Accepted: 05/08/2008] [Indexed: 12/11/2022] Open
Abstract
RATIONALE Fibroblasts are believed to be the major cells responsible for the production and maintenance of extracellular matrix. Alterations in fibroblast functional capacity, therefore, could play a role in the pathogenesis of pulmonary emphysema, which is characterized by inadequate maintenance of tissue structure. OBJECTIVES To evaluate the hypothesis that deficient fibroblast repair characterizes cells obtained from individuals with chronic obstructive pulmonary disease (COPD) compared with control subjects. METHODS Fibroblasts were cultured from lung tissue obtained from individuals undergoing thoracotomy and were characterized in vitro. MEASUREMENTS AND MAIN RESULTS Fibroblasts from individuals with COPD, defined by reduced FEV(1), manifested reduced chemotaxis toward fibronectin and reduced contraction of three-dimensional collagen gels, two bioassays associated with fibroblast repair function. At least two mechanisms appear to account for these differences. Prostaglandin E (PGE), a known inhibitor of fibroblast repair functions, was produced in increased amount by fibroblasts from subjects with COPD, which also expressed increased amounts of the receptors EP2 and EP4, both of which signal through cyclic AMP. Incubation of fibroblasts with indomethacin or with the PKA inhibitor KT-5720 partially restored COPD subject fibroblast function. In addition, fibroblasts from subjects with COPD produced more transforming growth factor (TGF)-beta1, but manifested reduced response to TGF-beta1. The functional alterations in fibroblasts correlated with both lung function assessed by FEV(1) and, for the data available, with severity of emphysema assessed by Dl(CO). CONCLUSIONS Fibroblasts from individuals with COPD have reduced capability to sustain tissue repair, which suggests that this may be one mechanism that contributes to the development of emphysema.
Collapse
Affiliation(s)
- Shinsaku Togo
- University of Nebraska Medical Center, 985885 Nebraska Medical Center, Omaha, NE 68198-5885, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Arachidonic acid pathway activates multidrug resistance related protein in cultured human lung cells. Arch Toxicol 2007; 82:323-32. [PMID: 17943274 DOI: 10.1007/s00204-007-0251-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2007] [Accepted: 09/26/2007] [Indexed: 02/04/2023]
Abstract
Primary cultures of human lung cells can serve as a model system to study the mechanisms underlying the effects of irritants in air and to get a deeper insight into the (patho)physiological roles of the xenobiotic detoxification systems. For 99 human lung cancer cases the culture duration for bronchial epithelium and peripheral lung cells (PLC) are given in term of generations and weeks. Using this system, we investigated whether and how prostaglandins (PG) modify multidrug resistance related protein (MRP) function in normal human lung cells. PGF2alpha had no effect on MRP function, whereas PGE2 induced MRP activity in cultured NHBECs. The transport activity study of MRP in NHBEC, PLC, and A549 under the effect of exogenously supplied PGF2alpha (10 microM, 1 day) using single cell fluorimetry revealed no alteration in transport activity of MRP. PG concentrations were within the physiological range. COX I and II inhibitors indomethacin (5, 10 microM) and celecoxib (5, 10 microM) could substantially decrease the transport activity of MRP in NHBEC, PLC, and A549 in 1- and 4-day trials. Prostaglandin E2 did not change cadmium-induced caspase 3/7 activation in NHBECs and had no own effect on caspase 3/7 activity. Cadmium chloride (5, 10 microM) was an effective inducer of caspase 3/7 activation in NHBECs with a fivefold and ninefold rise of activity. In primary human lung cells arachidonic acid activates MRP transport function only in primary epithelial lung cells by prostaglandin E2 but not by F2alpha mediated pathways and this effect needs some time to develop.
Collapse
|
28
|
Hodges RJ, Jenkins RG, Wheeler-Jones CPD, Copeman DM, Bottoms SE, Bellingan GJ, Nanthakumar CB, Laurent GJ, Hart SL, Foster ML, McAnulty RJ. Severity of lung injury in cyclooxygenase-2-deficient mice is dependent on reduced prostaglandin E(2) production. THE AMERICAN JOURNAL OF PATHOLOGY 2004; 165:1663-76. [PMID: 15509536 PMCID: PMC1618657 DOI: 10.1016/s0002-9440(10)63423-2] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Levels of prostaglandin E(2) (PGE(2)), a potent inhibitor of fibroblast function, are decreased in the lungs of patients with pulmonary fibrosis, which has been shown to be because of limited expression of cyclooxygenase-2 (COX-2). To further investigate the relative importance of COX-2 and PGE(2) in the development of fibrosis we have used a selective COX-2 inhibitor and COX-2-deficient ((-/-) and (+/-)) mice in studies of bleomycin-induced lung fibrosis. We demonstrate in wild-type mice that bleomycin-induced lung PGE(2) production is predominantly COX-2 mediated. Furthermore, COX-2(+/-) mice show limited induction of PGE(2) and an enhanced fibrotic response with increased lung collagen content compared with wild-type mice after bleomycin injury (P < 0.001). In contrast, COX-2(-/-) mice show increased levels of lung PGE(2), compared with wild-type mice after injury (P < 0.05), because of compensatory up-regulation of COX-1, which appears to be associated with macrophage/monocytes but not fibroblasts derived from these mice. COX-2(-/-) mice show an enhanced and persistent inflammatory response to bleomycin, however the fibrotic response to injury was unaltered compared with wild-type animals. These data provide further direct evidence for the importance of up-regulating COX-2 and PGE(2) expression in protecting against the development of fibrosis after lung injury.
Collapse
Affiliation(s)
- Rebecca J Hodges
- Centre for Respiratory Research, University College London, Rayne Bldg., 5 University St., London WC1E 6JJ, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|