1
|
Wang J, Hu S, Xu Y, Wang T. Omega-6 polyunsaturated fatty acids and their metabolites: a potential targeted therapy for pulmonary hypertension. Respir Res 2025; 26:102. [PMID: 40089708 PMCID: PMC11909876 DOI: 10.1186/s12931-025-03172-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 02/26/2025] [Indexed: 03/17/2025] Open
Abstract
Pulmonary hypertension (PH) is a progressive and life-threatening cardiopulmonary disease that is not uncommon. The modulation of the pulmonary artery (PA) involves various fatty acids, including omega-6 polyunsaturated fatty acids (ω-6 PUFAs) and ω-6 PUFAs-derived oxylipins. These lipid mediators are produced through cyclooxygenase (COX), lipoxygenase (LOX), cytochrome P450 (CYP450), and non-enzymatic pathways. They play a crucial role in the occurrence and development of PH by regulating the function and phenotype of pulmonary artery endothelial cells (PAECs), pulmonary artery smooth muscle cells (PASMCs), pulmonary fibroblasts, alveolar macrophages, and inflammatory cells. The alterations in ω-6 PUFAs and oxylipins are pivotal in causing vasoconstriction, pulmonary remodeling, and ultimately leading to right heart failure in PH. Despite the limited understanding of the PH pathophysiology, there is potential for novel interventions through dietary and pharmacological approaches targeting ω-6 PUFAs and oxylipins. The aim of this review is to summarize the significant advances in clinical and basic research on omega-6 PUFAs and oxylipins in pulmonary vascular disease, particularly PH, and to propose a potential targeted therapeutic modality against omega-6 PUFAs.
Collapse
Affiliation(s)
- Jiayao Wang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
- The Center for Biomedical Research, Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, National Health Committee (NHC), Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Shunlian Hu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
- The Center for Biomedical Research, Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, National Health Committee (NHC), Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yahan Xu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
- The Center for Biomedical Research, Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, National Health Committee (NHC), Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Tao Wang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
- The Center for Biomedical Research, Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, National Health Committee (NHC), Huazhong University of Science and Technology, Wuhan, People's Republic of China.
| |
Collapse
|
2
|
Fayad FH, Sellke FW, Feng J. Pulmonary hypertension associated with cardiopulmonary bypass and cardiac surgery. J Card Surg 2022; 37:5269-5287. [PMID: 36378925 DOI: 10.1111/jocs.17160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/23/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND AIM Pulmonary hypertension (PH) is frequently associated with cardiovascular surgery and is a common complication that has been observed after surgery utilizing cardiopulmonary bypass (CPB). The purpose of this review is to explain the characteristics of PH, the mechanisms of PH induced by cardiac surgery and CPB, treatments for postoperative PH, and future directions in treating PH induced by cardiac surgery and CPB using up-to-date findings. METHODS The PubMed database was utilized to find published articles. RESULTS There are many mechanisms that contribute to PH after cardiac surgery and CPB which involve pulmonary vasomotor dysfunction, cyclooxygenase, the thromboxane A2 and prostacyclin pathway, the nitric oxide pathway, inflammation, and oxidative stress. Furthermore, there are several effective treatments for postoperative PH within different types of cardiac surgery. CONCLUSIONS By possessing a deep understanding of the mechanisms that contribute to PH after cardiac surgery and CPB, researchers can develop treatments for clinicians to use which target the mechanisms of PH and ultimately reduce and/or eliminate postoperative PH. Additionally, learning about the most up-to-date studies regarding treatments can allow clinicians to choose the best treatments for patients who are undergoing cardiac surgery and CPB.
Collapse
Affiliation(s)
- Fayez H Fayad
- The Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA.,Program in Liberal Medical Education, Brown University, Providence, Rhode Island, USA
| | - Frank W Sellke
- The Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA.,Division of Cardiothoracic Surgery, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Jun Feng
- The Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA.,Division of Cardiothoracic Surgery, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, Rhode Island, USA
| |
Collapse
|
3
|
Huang J, An Q, Zhang CL, He L, Wang L. Decreased low‑density lipoprotein and the presence of pulmonary arterial hypertension among newly diagnosed drug‑naïve patients with systemic lupus erythematosus: D‑dimer as a mediator. Exp Ther Med 2022; 24:595. [PMID: 35949327 PMCID: PMC9353521 DOI: 10.3892/etm.2022.11531] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/13/2022] [Indexed: 11/23/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is commonly associated with systemic lupus erythematosus (SLE). The present study investigated the relationship between coagulation and changes in lipid parameters in newly-diagnosed patients with SLE in the presence of PAH and whether the coagulation parameters were mediators between lipids and PAH presence. A total of 301 subjects scheduled for new-onset drug-naïve SLE were consecutively enrolled. Baseline data for patients without PAH and with PAH were gathered and compared. Coagulation and lipid parameters were compared across patients without lipid regulating and anticoagulation medications. Multivariable logistic regression model was applied to examine potential predictors of PAH in SLE. The relationships between them were examined using Spearman's correlation analysis. The relationship between coagulation index and lipids with SLE-PAH was evaluated using mediation analysis. Female patients accounted for 88.0% of the 301 subjects, and the average age was 32 years (range, 25-45 years). A total of 40 patients (13.3%) had PAH, and the average pulmonary artery systolic pressure (sPAP) was 55.825±26.67 mmHg. Patients with PAH were older and had higher levels of fibrin/fibrinogen degradation products (FDP), D-dimer, C-reactive protein, lower levels of complement 3, complement 4 and 25-hydroxy vitamin D3 compared with the non-PAH group. Multivariable logistic regression analysis showed that age and D-dimer were independent predictor factors for PAH. Among patients without lipid regulating and anticoagulation medications, patients in the PAH group had higher levels of D-dimer and FDP, and lower low-density lipoprotein (LDL) levels compared with patients without PAH. There was also a positive relationship between sPAP and D-dimer and FDP, and a negative relationship between sPAP and total cholesterol and LDL. Mediation analysis indicated that 25.61% of the effect of low LDL on PAH presence in systemic lupus erythematosus was mediated by D-dimer. Overall, the effect of low LDL on SLE-PAH appeared to be mediated by D-dimer, which mediated 25.61% of this effect.
Collapse
Affiliation(s)
- Jing Huang
- Department of Rheumatism and Immunology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Qi An
- Department of Rheumatism and Immunology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Cai-Lian Zhang
- Department of Pulmonary and Critical Care Medicine, Yan'an University Affiliated Hospital, Yan'an, Shaanxi 716000, P.R. China
| | - Lan He
- Department of Rheumatism and Immunology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Lei Wang
- Department of Pulmonary and Critical Care Medicine, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| |
Collapse
|
4
|
Bui CB, Kolodziej M, Lamanna E, Elgass K, Sehgal A, Rudloff I, Schwenke DO, Tsuchimochi H, Kroon MAGM, Cho SX, Maksimenko A, Cholewa M, Berger PJ, Young MJ, Bourke JE, Pearson JT, Nold MF, Nold-Petry CA. Interleukin-1 Receptor Antagonist Protects Newborn Mice Against Pulmonary Hypertension. Front Immunol 2019; 10:1480. [PMID: 31354700 PMCID: PMC6637286 DOI: 10.3389/fimmu.2019.01480] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 06/13/2019] [Indexed: 12/18/2022] Open
Abstract
Pulmonary hypertension secondary to bronchopulmonary dysplasia (BPD-PH) represents a major complication of BPD in extremely preterm infants for which there are currently no safe and effective interventions. The abundance of interleukin-1 (IL-1) is strongly correlated with the severity and long-term outcome of BPD infants and we have previously shown that IL-1 receptor antagonist (IL-1Ra) protects against murine BPD; therefore, we hypothesized that IL-1Ra may also be effective against BPD-PH. We employed daily injections of IL-1Ra in a murine model in which BPD/BPD-PH was induced by antenatal LPS and postnatal hyperoxia of 65% O2. Pups reared in hyperoxia for 28 days exhibited a BPD-PH-like disease accompanied by significant changes in pulmonary vascular morphology: micro-CT revealed an 84% reduction in small vessels (4-5 μm diameter) compared to room air controls; this change was prevented by IL-1Ra. Pulmonary vascular resistance, assessed at day 28 of life by echocardiography using the inversely-related surrogate marker time-to-peak-velocity/right ventricular ejection time (TPV/RVET), increased in hyperoxic mice (0.27 compared to 0.32 in air controls), and fell significantly with daily IL-1Ra treatment (0.31). Importantly, in vivo cine-angiography revealed that this protection afforded by IL-1Ra treatment for 28 days is maintained at day 60 of life. Despite an increased abundance of mediators of pulmonary angiogenesis in day 5 lung lysates, namely vascular endothelial growth factor (VEGF) and endothelin-1 (ET-1), no difference was detected in ex vivo pulmonary vascular reactivity between air and hyperoxia mice as measured in precision cut lung slices, or by immunohistochemistry in alpha-smooth muscle actin (α-SMA) and endothelin receptor type-A (ETA) at day 28. Further, on day 28 of life we observed cardiac fibrosis by Sirius Red staining, which was accompanied by an increase in mRNA expression of galectin-3 and CCL2 (chemokine (C-C motif) ligand 2) in whole hearts of hyperoxic pups, which improved with IL-1Ra. In summary, our findings suggest that daily administration of the anti-inflammatory IL-1Ra prevents the increase in pulmonary vascular resistance and the pulmonary dysangiogenesis of murine BPD-PH, thus pointing to IL-1Ra as a promising candidate for the treatment of both BPD and BPD-PH.
Collapse
Affiliation(s)
- Christine B Bui
- Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Paediatrics, Monash University, Clayton, VIC, Australia
| | | | - Emma Lamanna
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Kirstin Elgass
- Monash Micro Imaging, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Arvind Sehgal
- Department of Paediatrics, Monash University, Clayton, VIC, Australia.,Monash Newborn, Monash Children's Hospital, Melbourne, VIC, Australia
| | - Ina Rudloff
- Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Paediatrics, Monash University, Clayton, VIC, Australia
| | - Daryl O Schwenke
- Department of Physiology-Heart Otago, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Hirotsugu Tsuchimochi
- Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Maurice A G M Kroon
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.,Department of Pharmacy, Amsterdam UMC, Amsterdam, Netherlands
| | - Steven X Cho
- Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Paediatrics, Monash University, Clayton, VIC, Australia
| | - Anton Maksimenko
- Imaging and Medical Beamline, Australian Synchrotron, Clayton, VIC, Australia
| | - Marian Cholewa
- Centre for Innovation and Transfer of Natural Sciences and Engineering Knowledge, University of Rzeszow, Rzeszow, Poland
| | - Philip J Berger
- Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Paediatrics, Monash University, Clayton, VIC, Australia
| | - Morag J Young
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Jane E Bourke
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - James T Pearson
- Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan.,Department of Physiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Marcel F Nold
- Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Paediatrics, Monash University, Clayton, VIC, Australia
| | - Claudia A Nold-Petry
- Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Paediatrics, Monash University, Clayton, VIC, Australia
| |
Collapse
|
5
|
Wong MJ, Kantores C, Ivanovska J, Jain A, Jankov RP. Simvastatin prevents and reverses chronic pulmonary hypertension in newborn rats via pleiotropic inhibition of RhoA signaling. Am J Physiol Lung Cell Mol Physiol 2016; 311:L985-L999. [DOI: 10.1152/ajplung.00345.2016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 09/30/2016] [Indexed: 11/22/2022] Open
Abstract
Chronic neonatal pulmonary hypertension (PHT) frequently results in early death. Systemically administered Rho-kinase (ROCK) inhibitors prevent and reverse chronic PHT in neonatal rats, but at the cost of severe adverse effects, including systemic hypotension and growth restriction. Simvastatin has pleiotropic inhibitory effects on isoprenoid intermediates that may limit activity of RhoA, which signals upstream of ROCK. We therefore hypothesized that statin treatment would safely limit pulmonary vascular RhoA activity and prevent and reverse experimental chronic neonatal PHT via downstream inhibitory effects on pathological ROCK activity. Sprague-Dawley rats in normoxia (room air) or moderate normobaric hypoxia (13% O2) received simvastatin (2 mg·kg−1·day−1 ip) or vehicle from postnatal days 1–14 (prevention protocol) or from days 14–21 (rescue protocol). Chronic hypoxia increased RhoA and ROCK activity in lung tissue. Simvastatin reduced lung content of the isoprenoid intermediate farnesyl pyrophosphate and decreased RhoA/ROCK signaling in the hypoxia-exposed lung. Preventive or rescue treatment of chronic hypoxia-exposed animals with simvastatin decreased pulmonary vascular resistance, right ventricular hypertrophy, and pulmonary arterial remodeling. Preventive simvastatin treatment improved weight gain, did not lower systemic blood pressure, and did not cause apparent toxic effects on skeletal muscle, liver or brain. Rescue therapy with simvastatin improved exercise capacity. We conclude that simvastatin limits RhoA/ROCK activity in the chronic hypoxia-exposed lung, thus preventing or ameliorating hemodynamic and structural markers of chronic PHT and improving long-term outcome, without causing adverse effects.
Collapse
Affiliation(s)
- Mathew J. Wong
- Physiology & Experimental Medicine Program, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Crystal Kantores
- Physiology & Experimental Medicine Program, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Julijana Ivanovska
- Physiology & Experimental Medicine Program, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Amish Jain
- Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, University of Toronto, Toronto, Ontario, Canada
- Department of Paediatrics, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; and
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Robert P. Jankov
- Physiology & Experimental Medicine Program, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
- Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, University of Toronto, Toronto, Ontario, Canada
- Department of Paediatrics, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; and
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
6
|
Sharma S, Ruffenach G, Umar S, Motayagheni N, Reddy ST, Eghbali M. Role of oxidized lipids in pulmonary arterial hypertension. Pulm Circ 2016; 6:261-73. [PMID: 27683603 DOI: 10.1086/687293] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a multifactorial disease characterized by interplay of many cellular, molecular, and genetic events that lead to excessive proliferation of pulmonary cells, including smooth muscle and endothelial cells; inflammation; and extracellular matrix remodeling. Abnormal vascular changes and structural remodeling associated with PAH culminate in vasoconstriction and obstruction of pulmonary arteries, contributing to increased pulmonary vascular resistance, pulmonary hypertension, and right ventricular failure. The complex molecular mechanisms involved in the pathobiology of PAH are the limiting factors in the development of potential therapeutic interventions for PAH. Over the years, our group and others have demonstrated the critical implication of lipids in the pathogenesis of PAH. This review specifically focuses on the current understanding of the role of oxidized lipids, lipid metabolism, peroxidation, and oxidative stress in the progression of PAH. This review also discusses the relevance of apolipoprotein A-I mimetic peptides and microRNA-193, which are known to regulate the levels of oxidized lipids, as potential therapeutics in PAH.
Collapse
Affiliation(s)
- Salil Sharma
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Grégoire Ruffenach
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Soban Umar
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Negar Motayagheni
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Srinivasa T Reddy
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Mansoureh Eghbali
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|
7
|
Ee MT, Kantores C, Ivanovska J, Wong MJ, Jain A, Jankov RP. Leukotriene B4 mediates macrophage influx and pulmonary hypertension in bleomycin-induced chronic neonatal lung injury. Am J Physiol Lung Cell Mol Physiol 2016; 311:L292-302. [PMID: 27317685 DOI: 10.1152/ajplung.00120.2016] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 06/13/2016] [Indexed: 11/22/2022] Open
Abstract
Systemically-administered bleomycin causes inflammation, arrested lung growth, and pulmonary hypertension (PHT) in the neonatal rat, similar to human infants with severe bronchopulmonary dysplasia (BPD). Leukotrienes (LTs) are inflammatory lipid mediators produced by multiple cell types in the lung. The major LTs, LTB4 and cysteinyl LTs, are suggested to contribute to BPD, but their specific roles remain largely unexplored in experimental models. We hypothesized that LTs are increased in bleomycin-induced BPD-like injury, and that inhibition of LT production would prevent inflammatory cell influx and thereby ameliorate lung injury. Rat pups were exposed to bleomycin (1 mg·kg(-1)·day(-1) ip) or vehicle (control) from postnatal days 1-14 and were treated with either zileuton (5-lipoxygenase inhibitor), montelukast (cysteinyl LT1 receptor antagonist), or SC57461A (LTA4 hydrolase inhibitor) 10 mg·kg(-1)·day(-1) ip. Bleomycin led to increased lung content of LTB4, but not cysteinyl LTs. Bleomycin-induced increases in tissue neutrophils and macrophages and lung contents of LTB4 and tumor necrosis factor-α were all prevented by treatment with zileuton. Treatment with zileuton or SC57461A also prevented the hemodynamic and structural markers of chronic PHT, including raised pulmonary vascular resistance, increased Fulton index, and arterial wall remodeling. However, neither treatment prevented impaired alveolarization or vascular hypoplasia secondary to bleomycin. Treatment with montelukast had no effect on macrophage influx, PHT, or on abnormal lung structure. We conclude that LTB4 plays a crucial role in lung inflammation and PHT in experimental BPD. Agents targeting LTB4 or LTB4-mediated signaling may have utility in infants at risk of developing BPD-associated PHT.
Collapse
Affiliation(s)
- Mong Tieng Ee
- Program in Physiology and Experimental Medicine, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada; Division of Neonatology, Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Crystal Kantores
- Program in Physiology and Experimental Medicine, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Julijana Ivanovska
- Program in Physiology and Experimental Medicine, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Mathew J Wong
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Amish Jain
- Division of Neonatology, Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Robert P Jankov
- Program in Physiology and Experimental Medicine, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada; Division of Neonatology, Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, Canada; Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, University of Toronto, Toronto, Ontario, Canada; and
| |
Collapse
|
8
|
Masood A, Yi M, Belcastro R, Li J, Lopez L, Kantores C, Jankov RP, Tanswell AK. Neutrophil elastase-induced elastin degradation mediates macrophage influx and lung injury in 60% O2-exposed neonatal rats. Am J Physiol Lung Cell Mol Physiol 2015; 309:L53-62. [DOI: 10.1152/ajplung.00298.2014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 04/30/2015] [Indexed: 12/20/2022] Open
Abstract
Neutrophil (PMNL) influx precedes lung macrophage (LM) influx into the lung following exposure of newborn pups to 60% O2. We hypothesized that PMNL were responsible for the signals leading to LM influx. This was confirmed when inhibition of PMNL influx with a CXC chemokine receptor-2 antagonist, SB-265610, also prevented the 60% O2-dependent LM influx, LM-derived nitrotyrosine formation, and pruning of small arterioles. Exposure to 60% O2was associated with increased lung contents of neutrophil elastase and α-elastin, a marker of denatured elastin, and a decrease in elastin fiber density. This led us to speculate that neutrophil elastase-induced elastin fragments were the chemokines that led to a LM influx into the 60% O2-exposed lung. Inhibition of neutrophil elastase with sivelestat or elafin attenuated the LM influx. Sivelestat also attenuated the 60% O2-induced decrease in elastin fiber density. Daily injections of pups with an antibody to α-elastin prevented the 60% O2-dependent LM influx, impaired alveologenesis, and impaired small vessel formation. This suggests that neutrophil elastase inhibitors may protect against neonatal lung injury not only by preventing structural elastin degradation, but also by blocking elastin fragment-induced LM influx, thus preventing tissue injury from LM-derived peroxynitrite formation.
Collapse
Affiliation(s)
- Azhar Masood
- Lung Biology Programme, Physiology and Experimental Medicine, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada; and
| | - Man Yi
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Rosetta Belcastro
- Lung Biology Programme, Physiology and Experimental Medicine, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Jun Li
- Lung Biology Programme, Physiology and Experimental Medicine, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Lianet Lopez
- Lung Biology Programme, Physiology and Experimental Medicine, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Crystal Kantores
- Lung Biology Programme, Physiology and Experimental Medicine, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Robert P. Jankov
- Lung Biology Programme, Physiology and Experimental Medicine, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
- Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada; and
- Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, University of Toronto, Toronto, Ontario, Canada
| | - A. Keith Tanswell
- Lung Biology Programme, Physiology and Experimental Medicine, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
- Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada; and
| |
Collapse
|
9
|
Belcastro R, Lopez L, Li J, Masood A, Tanswell AK. Chronic lung injury in the neonatal rat: up-regulation of TGFβ1 and nitration of IGF-R1 by peroxynitrite as likely contributors to impaired alveologenesis. Free Radic Biol Med 2015; 80:1-11. [PMID: 25514442 DOI: 10.1016/j.freeradbiomed.2014.12.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 11/22/2014] [Accepted: 12/05/2014] [Indexed: 10/24/2022]
Abstract
Postnatal alveolarization is regulated by a number of growth factors, including insulin-like growth factor-I (IGF-I) acting through the insulin-like growth factor receptor-1 (IGF-R1). Exposure of the neonatal rat lung to 60% O2 for 14 days results in impairments of lung cell proliferation, secondary crest formation, and alveologenesis. This lung injury is mediated by peroxynitrite and is prevented by treatment with a peroxynitrite decomposition catalyst. We hypothesized that one of the mechanisms by which peroxynitrite induces lung injury in 60% O2 is through nitration and inactivation of critical growth factors or their receptors. Increased nitration of both IGF-I and IGF-R1 was evident in 60% O2-exposed lungs, which was reversible by concurrent treatment with a peroxynitrite decomposition catalyst. Increased nitration of the IGF-R1 was associated with its reduced activation, as assessed by IGF-R1 phosphotyrosine content. IGF-I displacement binding plots were conducted in vitro using rat fetal lung distal epithelial cells which respond to IGF-I by an increase in DNA synthesis. When IGF-I was nitrated to a degree similar to that observed in vivo there was minimal, if any, effect on IGF-I displacement binding. In contrast, nitrating cell IGF-R1 to a similar degree to that observed in vivo completely prevented specific binding of IGF-I to the IGF-R1, and attenuated an IGF-I-mediated increase in DNA synthesis. Additionally, we hypothesized that peroxynitrite also impairs alveologenesis by being an upstream regulator of the growth inhibitor, TGFβ1. That 60% O2-induced impairment of alveologenesis was mediated in part by TGFβ1 was confirmed by demonstrating an improvement in secondary crest formation when 60% O2-exposed pups received concurrent treatment with the TGFß1 activin receptor-like kinase, SB 431542. That the increased TGFβ1 content in lungs of pups exposed to 60% O2 was regulated by peroxynitrite was confirmed by its attenuation by concurrent treatment with a peroxynitrite decomposition catalyst. We conclude that peroxynitrite contributes to the impaired alveologenesis observed following the exposure of neonatal rats to 60% O2 both by preventing binding of IGF-I to the IGF-R1, secondary to nitration of the IGF-R1, and by causing an up-regulation of the growth inhibitor, TGFβ1.
Collapse
Affiliation(s)
- Rosetta Belcastro
- Lung Biology Programme, Physiology & Experimental Medicine, Hospital for Sick Children Research Institute, Toronto, Ontario M5G 1X8
| | - Lianet Lopez
- Lung Biology Programme, Physiology & Experimental Medicine, Hospital for Sick Children Research Institute, Toronto, Ontario M5G 1X8
| | - Jun Li
- Lung Biology Programme, Physiology & Experimental Medicine, Hospital for Sick Children Research Institute, Toronto, Ontario M5G 1X8
| | - Azhar Masood
- Lung Biology Programme, Physiology & Experimental Medicine, Hospital for Sick Children Research Institute, Toronto, Ontario M5G 1X8; Department of Physiology, University of Toronto, Toronto, Ontario M5S 1A8
| | - A Keith Tanswell
- Lung Biology Programme, Physiology & Experimental Medicine, Hospital for Sick Children Research Institute, Toronto, Ontario M5G 1X8; Department of Physiology, University of Toronto, Toronto, Ontario M5S 1A8; Department of Paediatrics, University of Toronto, Toronto, Ontario M5G 1X8.
| |
Collapse
|
10
|
Abstract
Hypoxic pulmonary hypertension of the newborn is characterized by elevated pulmonary vascular resistance and pressure due to vascular remodeling and increased vessel tension secondary to chronic hypoxia during the fetal and newborn period. In comparison to the adult, the pulmonary vasculature of the fetus and the newborn undergoes tremendous developmental changes that increase susceptibility to a hypoxic insult. Substantial evidence indicates that chronic hypoxia alters the production and responsiveness of various vasoactive agents such as endothelium-derived nitric oxide, endothelin-1, prostanoids, platelet-activating factor, and reactive oxygen species, resulting in sustained vasoconstriction and vascular remodeling. These changes occur in most cell types within the vascular wall, particularly endothelial and smooth muscle cells. At the cellular level, suppressed nitric oxide-cGMP signaling and augmented RhoA-Rho kinase signaling appear to be critical to the development of hypoxic pulmonary hypertension of the newborn.
Collapse
Affiliation(s)
- Yuansheng Gao
- Department of Physiology and Pathophysiology, Peking University, Health Science Center, Beijing, China
| | | |
Collapse
|
11
|
Dick AS, Ivanovska J, Kantores C, Belcastro R, Keith Tanswell A, Jankov RP. Cyclic stretch stimulates nitric oxide synthase-1-dependent peroxynitrite formation by neonatal rat pulmonary artery smooth muscle. Free Radic Biol Med 2013; 61:310-9. [PMID: 23619128 DOI: 10.1016/j.freeradbiomed.2013.04.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 02/27/2013] [Accepted: 04/16/2013] [Indexed: 11/28/2022]
Abstract
Peroxynitrite, the reaction product of nitric oxide and superoxide, contributes to the pathogenesis of chronic pulmonary hypertension in immature animals by stimulating proliferation of pulmonary arterial smooth muscle cells (PASMCs). Pulmonary vasoconstriction, secondary to hypoxia and other stimuli, leads to enhanced pulsatile stretch of cells in the vascular wall, particularly in smooth muscle, which we hypothesized would cause increased peroxynitrite generation. Our objectives in this study were to determine whether cyclic mechanical stretch, at supraphysiologic levels, would cause increased production of reactive oxygen species (ROS), nitric oxide, and peroxynitrite in vitro. Early passage neonatal rat PASMCs were seeded and grown to subconfluence on collagen-coated elastomer-bottom plates and subjected to cyclic mechanical stretch (10% ("physiologic") or 20% ("supraphysiologic") at 0.5 Hz) for up to 24 h. Compared to nonstretched controls and to cells subjected to 10% stretch, 20% stretch increased H2O2 (stable marker of ROS) and nitrate/nitrite (stable marker of nitric oxide) in conditioned medium. These effects were accompanied by increased peroxynitrite, as evidenced by increased in situ dihydroethidium fluorescence and immunoreactive nitrotyrosine and by increased expression of nitric oxide synthase (NOS)-1 and NADPH oxidase 4 (NOX4), but not NOS-2. Stretch-induced H2O2 release and increased dihydroethidium fluorescence were prevented by pretreatment with a superoxide scavenger, nonspecific inhibitors of NADPH oxidase or NOS, or a peroxynitrite decomposition catalyst. Short-interfering RNA-mediated knockdown of NOS-1 or NOX4 attenuated increased nitric oxide and H2O2 content, respectively, in stretched-cell-conditioned medium. Knockdown of NOS-1 also attenuated increased immunoreactive nitrotyrosine content and stretch-induced proliferation, whereas knockdown of NOS-2 had no effect. We conclude that increased peroxynitrite generation by neonatal rat PASMCs subjected to supraphysiologic levels of cyclic stretch is NOS-1-dependent and that increased ROS production is predominantly mediated by NADPH oxidase, specifically NOX4.
Collapse
Affiliation(s)
- Andrew S Dick
- Physiology & Experimental Medicine Program, Hospital for Sick Children Research Institute, Toronto, ON, Canada M5G 1X8
| | - Julijana Ivanovska
- Physiology & Experimental Medicine Program, Hospital for Sick Children Research Institute, Toronto, ON, Canada M5G 1X8
| | - Crystal Kantores
- Physiology & Experimental Medicine Program, Hospital for Sick Children Research Institute, Toronto, ON, Canada M5G 1X8
| | - Rosetta Belcastro
- Physiology & Experimental Medicine Program, Hospital for Sick Children Research Institute, Toronto, ON, Canada M5G 1X8
| | - A Keith Tanswell
- Physiology & Experimental Medicine Program, Hospital for Sick Children Research Institute, Toronto, ON, Canada M5G 1X8; Department of Physiology, Division of Neonatology, Department of Paediatrics, and Heart and Stroke Richard Lewar Centre of Excellence, University of Toronto, Toronto, ON, Canada M5S 1A8; Division of Neonatology, Department of Paediatrics, and Heart and Stroke Richard Lewar Centre of Excellence, University of Toronto, Toronto, ON, Canada M5S 1A8
| | - Robert P Jankov
- Physiology & Experimental Medicine Program, Hospital for Sick Children Research Institute, Toronto, ON, Canada M5G 1X8; Department of Physiology, Division of Neonatology, Department of Paediatrics, and Heart and Stroke Richard Lewar Centre of Excellence, University of Toronto, Toronto, ON, Canada M5S 1A8; Division of Neonatology, Department of Paediatrics, and Heart and Stroke Richard Lewar Centre of Excellence, University of Toronto, Toronto, ON, Canada M5S 1A8; Heart and Stroke Richard Lewar Centre of Excellence, University of Toronto, Toronto, ON, Canada M5S 1A8.
| |
Collapse
|
12
|
Cabral JE, Belik J. Persistent pulmonary hypertension of the newborn: Recent advances in pathophysiology and treatment. JORNAL DE PEDIATRIA (VERSÃO EM PORTUGUÊS) 2013. [DOI: 10.1016/j.jpedp.2012.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
13
|
Cabral JEB, Belik J. Persistent pulmonary hypertension of the newborn: recent advances in pathophysiology and treatment. J Pediatr (Rio J) 2013; 89:226-42. [PMID: 23684454 DOI: 10.1016/j.jped.2012.11.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Accepted: 11/08/2012] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVES Although recognized for decades, little is known about the etiology, physiopathology, and prevention of persistent pulmonary hypertension of the newborn (PPHN), and its treatment remains a major challenge for neonatologists. In this review, the clinical features and physiopathology of the syndrome will be addressed, as well as its general and specific treatments. DATA SOURCE A review was carried out in PubMed, Cochrane Library, and MRei consult databases, searching for articles related to the syndrome and published between 1995 and 2011. DATA SYNTHESIS Risk factors and the physiopathological mechanisms of the syndrome are discussed. The clinical presentation depends on the different factors involved. These are related to the etiology and physiopathology of the different forms of the disease. In addition to the measures used to allow for the decrease in pulmonary vascular resistance after birth, in some instances pulmonary vasodilators will be required. Although inhaled nitric oxide has proved effective, other vasodilators have been recently used, but clinical evidence is still lacking to demonstrate their benefits in the treatment of PPHN. CONCLUSIONS Despite recent technological advances and new physiopathological knowledge of this disease, mortality associated with PPHN remains at 10%. More clinical research and evidence-based experimental results are needed to prevent, treat, and reduce the morbidity/mortality associated with this neonatal syndrome.
Collapse
|
14
|
Barrier M, Meloche J, Jacob MH, Courboulin A, Provencher S, Bonnet S. Today's and tomorrow's imaging and circulating biomarkers for pulmonary arterial hypertension. Cell Mol Life Sci 2012; 69:2805-31. [PMID: 22446747 PMCID: PMC11115077 DOI: 10.1007/s00018-012-0950-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Revised: 02/18/2012] [Accepted: 02/20/2012] [Indexed: 01/04/2023]
Abstract
The pathobiology of pulmonary arterial hypertension (PAH) involves a remodeling process in distal pulmonary arteries, as well as vasoconstriction and in situ thrombosis, leading to an increase in pulmonary vascular resistance, right heart failure and death. Its etiology may be idiopathic, but PAH is also frequently associated with underlying conditions such as connective tissue diseases. During the past decade, more than welcome novel therapies have been developed and are in development, including those increasingly targeting the remodeling process. These therapeutic options modestly increase the patients' long-term survival, now approaching 60% at 5 years. However, non-invasive tools for confirming PAH diagnosis, and assessing disease severity and response to therapy, are tragically lacking and would help to select the best treatment. After exclusion of other causes of pulmonary hypertension, a final diagnosis still relies on right heart catheterization, an invasive technique which cannot be repeated as often as an optimal follow-up might require. Similarly, other techniques and biomarkers used for assessing disease severity and response to treatment generally lack specificity and have significant limitations. In this review, imaging as well as current and future circulating biomarkers for diagnosis, prognosis, and follow-up are discussed.
Collapse
Affiliation(s)
- Marjorie Barrier
- Pulmonary Hypertension Research Group, Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, 2725 Chemin Ste-Foy, Québec, QC G1V 4G5 Canada
| | - Jolyane Meloche
- Pulmonary Hypertension Research Group, Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, 2725 Chemin Ste-Foy, Québec, QC G1V 4G5 Canada
| | - Maria Helena Jacob
- Pulmonary Hypertension Research Group, Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, 2725 Chemin Ste-Foy, Québec, QC G1V 4G5 Canada
| | - Audrey Courboulin
- Pulmonary Hypertension Research Group, Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, 2725 Chemin Ste-Foy, Québec, QC G1V 4G5 Canada
| | - Steeve Provencher
- Pulmonary Hypertension Research Group, Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, 2725 Chemin Ste-Foy, Québec, QC G1V 4G5 Canada
| | - Sébastien Bonnet
- Pulmonary Hypertension Research Group, Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, 2725 Chemin Ste-Foy, Québec, QC G1V 4G5 Canada
| |
Collapse
|
15
|
Chen JX, O’Mara PW, Poole SD, Brown N, Ehinger NJ, Slaughter JC, Paria BC, Aschner JL, Reese J. Isoprostanes as physiological mediators of transition to newborn life: novel mechanisms regulating patency of the term and preterm ductus arteriosus. Pediatr Res 2012; 72:122-8. [PMID: 22565502 PMCID: PMC3586272 DOI: 10.1038/pr.2012.58] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Increased oxygen tension at birth regulates physiologic events that are essential to postnatal survival, but the accompanying oxidative stress may also generate isoprostanes. We hypothesized that isoprostanes regulate ductus arteriosus (DA) function during postnatal vascular transition. METHODS Isoprostanes were measured by gas chromatography-mass spectrometry. DA tone was assessed by pressure myography. Gene expression was measured by quantitative PCR. RESULTS Oxygen exposure was associated with increased 8-iso-prostaglandin (PG)F2α in newborn mouse lungs. Both 8-iso-PGE2 and 8-iso-PGF2α induced concentration-dependent constriction of the isolated term DA, which was reversed by the thromboxane A2 (TxA2) receptor antagonist SQ29548. SQ29548 pretreatment unmasked an isoprostane-induced DA dilation mediated by the EP4 PG receptor. Exposure of the preterm DA to 8-iso-PGE2 caused unexpected DA relaxation that was reversed by EP4 antagonism. In contrast, exposure to 8-iso-PGF2α caused preterm DA constriction via TxA2 receptor activation. Further investigation revealed the predominance of the TxA2 receptor at term, whereas the EP4 receptor was expressed and functionally active from mid-gestation onward. CONCLUSION This study identifies a novel physiological role for isoprostanes during postnatal vascular transition and provide evidence that oxidative stress may act on membrane lipids to produce vasoactive mediators that stimulate physiological DA closure at birth or induce pathological patency of the preterm DA.
Collapse
Affiliation(s)
- Jian-Xiong Chen
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Patrick W. O’Mara
- Department of Pediatrics, Vanderbilt University, Nashville, Tennessee
| | - Stanley D. Poole
- Department of Pediatrics, Vanderbilt University, Nashville, Tennessee
| | - Naoko Brown
- Department of Pediatrics, Vanderbilt University, Nashville, Tennessee
| | - Noah J. Ehinger
- Department of Pediatrics, Vanderbilt University, Nashville, Tennessee
| | - James C. Slaughter
- Department of Biostatistics, Vanderbilt University, Nashville, Tennessee
| | - Bibhash C. Paria
- Department of Pediatrics, Vanderbilt University, Nashville, Tennessee
| | - Judy L. Aschner
- Department of Pediatrics, Vanderbilt University, Nashville, Tennessee
| | - Jeff Reese
- Department of Pediatrics, Vanderbilt University, Nashville, Tennessee;,Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
16
|
Sewing ACP, Kantores C, Ivanovska J, Lee AH, Masood A, Jain A, McNamara PJ, Tanswell AK, Jankov RP. Therapeutic hypercapnia prevents bleomycin-induced pulmonary hypertension in neonatal rats by limiting macrophage-derived tumor necrosis factor-α. Am J Physiol Lung Cell Mol Physiol 2012; 303:L75-87. [DOI: 10.1152/ajplung.00072.2012] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Bleomycin-induced lung injury is characterized in the neonatal rat by inflammation, arrested lung growth, and pulmonary hypertension (PHT), as observed in human infants with severe bronchopulmonary dysplasia. Inhalation of CO2 (therapeutic hypercapnia) has been described to limit cytokine production and to have anti-inflammatory effects on the injured lung; we therefore hypothesized that therapeutic hypercapnia would prevent bleomycin-induced lung injury. Spontaneously breathing rat pups were treated with bleomycin (1 mg/kg/d ip) or saline vehicle from postnatal days 1–14 while being continuously exposed to 5% CO2 (PaCO2 elevated by 15–20 mmHg), 7% CO2 (PaCO2 elevated by 35 mmHg), or normocapnia. Bleomycin-treated animals exposed to 7%, but not 5%, CO2, had significantly attenuated lung tissue macrophage influx and PHT, as evidenced by normalized pulmonary vascular resistance and right ventricular systolic function, decreased right ventricular hypertrophy, and attenuated remodeling of pulmonary resistance arteries. The level of CO2 neither prevented increased tissue neutrophil influx nor led to improvements in decreased lung weight, septal thinning, impaired alveolarization, or decreased numbers of peripheral arteries. Bleomycin led to increased expression and content of lung tumor necrosis factor (TNF)-α, which was found to colocalize with tissue macrophages and to be attenuated by exposure to 7% CO2. Inhibition of TNF-α signaling with the soluble TNF-2 receptor etanercept (0.4 mg/kg ip from days 1–14 on alternate days) prevented bleomycin-induced PHT without decreasing tissue macrophages and, similar to CO2, had no effect on arrested alveolar development. Our findings are consistent with a preventive effect of therapeutic hypercapnia with 7% CO2 on bleomycin-induced PHT via attenuation of macrophage-derived TNF-α. Neither tissue macrophages nor TNF-α appeared to contribute to arrested lung development induced by bleomycin. That 7% CO2 normalized pulmonary vascular resistance and right ventricular function without improving inhibited airway and vascular development suggests that vascular hypoplasia does not contribute significantly to functional changes of PHT in this model.
Collapse
Affiliation(s)
- A. Charlotte P. Sewing
- Physiology and Experimental Medicine Program, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Crystal Kantores
- Physiology and Experimental Medicine Program, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Julijana Ivanovska
- Physiology and Experimental Medicine Program, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Alvin H. Lee
- Physiology and Experimental Medicine Program, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Azhar Masood
- Physiology and Experimental Medicine Program, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
- Division of Neonatology, Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Amish Jain
- Physiology and Experimental Medicine Program, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
- Division of Neonatology, Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Patrick J. McNamara
- Physiology and Experimental Medicine Program, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
- Division of Neonatology, Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada
| | - A. Keith Tanswell
- Physiology and Experimental Medicine Program, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
- Division of Neonatology, Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Robert P. Jankov
- Physiology and Experimental Medicine Program, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
- Heart and Stroke Richard Lewar Centre of Excellence, University of Toronto, Toronto, Ontario, Canada
- Division of Neonatology, Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
17
|
Yi M, Masood A, Ziino A, Johnson BH, Belcastro R, Li J, Shek S, Kantores C, Jankov RP, Keith Tanswell A. Inhibition of apoptosis by 60% oxygen: a novel pathway contributing to lung injury in neonatal rats. Am J Physiol Lung Cell Mol Physiol 2011; 300:L319-29. [DOI: 10.1152/ajplung.00126.2010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
During early postnatal alveolar formation, the lung tissue of rat pups undergoes a physiological remodeling involving apoptosis of distal lung cells. Exposure of neonatal rats to severe hyperoxia (≥95% O2) both arrests lung growth and results in increased lung cell apoptosis. In contrast, exposure to moderate hyperoxia (60% O2) for 14 days does not completely arrest lung cell proliferation and is associated with parenchymal thickening. On the basis of similarities in lung architecture observed following either exposure to 60% O2, or pharmacological inhibition of physiological apoptosis, we hypothesized that exposure to 60% O2 would result in an inhibition of physiological lung cell apoptosis. Consistent with this hypothesis, we observed that the parenchymal thickening induced by exposure to 60% O2 was associated with decreased numbers of apoptotic cells, increased expressions of the antiapoptotic regulator Bcl-xL, and the putative antiapoptotic protein survivin, and decreased expressions of the proapoptotic cleaved caspases-3 and -7. In summary, exposure of the neonatal rat lung to moderate hyperoxia results in an inhibition of physiological apoptosis, which contributes to the parenchymal thickening observed in the resultant lung injury.
Collapse
Affiliation(s)
- Man Yi
- Lung Biology Programme, Physiology and Experimental Medicine, Hospital for Sick Children Research Institute, Toronto
| | - Azhar Masood
- Lung Biology Programme, Physiology and Experimental Medicine, Hospital for Sick Children Research Institute, Toronto
- The Departments of Paediatrics and Physiology, University of Toronto, Toronto; and
| | - Adrian Ziino
- Lung Biology Programme, Physiology and Experimental Medicine, Hospital for Sick Children Research Institute, Toronto
- The Departments of Paediatrics and Physiology, University of Toronto, Toronto; and
- Clinical Integrative Biology, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Ben-Hur Johnson
- Lung Biology Programme, Physiology and Experimental Medicine, Hospital for Sick Children Research Institute, Toronto
- The Departments of Paediatrics and Physiology, University of Toronto, Toronto; and
| | - Rosetta Belcastro
- Lung Biology Programme, Physiology and Experimental Medicine, Hospital for Sick Children Research Institute, Toronto
| | - Jun Li
- Lung Biology Programme, Physiology and Experimental Medicine, Hospital for Sick Children Research Institute, Toronto
| | - Samuel Shek
- Lung Biology Programme, Physiology and Experimental Medicine, Hospital for Sick Children Research Institute, Toronto
| | - Crystal Kantores
- Clinical Integrative Biology, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Robert P. Jankov
- The Departments of Paediatrics and Physiology, University of Toronto, Toronto; and
- Clinical Integrative Biology, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - A. Keith Tanswell
- Lung Biology Programme, Physiology and Experimental Medicine, Hospital for Sick Children Research Institute, Toronto
- The Departments of Paediatrics and Physiology, University of Toronto, Toronto; and
| |
Collapse
|
18
|
Xu EZ, Kantores C, Ivanovska J, Engelberts D, Kavanagh BP, McNamara PJ, Jankov RP. Rescue treatment with a Rho-kinase inhibitor normalizes right ventricular function and reverses remodeling in juvenile rats with chronic pulmonary hypertension. Am J Physiol Heart Circ Physiol 2010; 299:H1854-64. [PMID: 20889845 DOI: 10.1152/ajpheart.00595.2010] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Chronic pulmonary hypertension in infancy and childhood is characterized by a fixed and progressive increase in pulmonary arterial pressure and resistance, pulmonary arterial remodeling, and right ventricular hypertrophy and systolic dysfunction. These abnormalities are replicated in neonatal rats chronically exposed to hypoxia from birth in which increased activity of Rho-kinase (ROCK) is critical to injury, as evidenced by preventive effects of ROCK inhibitors. Our objective in the present study was to examine the reversing effects of a late or rescue approach to treatment with a ROCK inhibitor on the pulmonary and cardiac manifestations of established chronic hypoxic pulmonary hypertension. Rat pups were exposed to air or hypoxia (13% O(2)) from postnatal day 1 and were treated with Y-27632 (15 mg/kg) or saline vehicle by twice daily subcutaneous injection commencing on day 14, for up to 7 days. Treatment with Y-27632 significantly attenuated right ventricular hypertrophy, reversed arterial wall remodeling, and completely normalized right ventricular systolic function in hypoxia-exposed animals. Reversal of arterial wall remodeling was accompanied by increased apoptosis and attenuated content of endothelin (ET)-1 and ET(A) receptors. Treatment of primary cultured juvenile rat pulmonary artery smooth muscle cells with Y-27632 attenuated serum-stimulated ROCK activity and proliferation and increased apoptosis. Smooth muscle apoptosis was also induced by short interfering RNA-mediated knockdown of ROCK-II, but not of ROCK-I. We conclude that sustained rescue treatment with a ROCK inhibitor reversed both the hemodynamic and structural abnormalities of chronic hypoxic pulmonary hypertension in juvenile rats and normalized right ventricular systolic function. Attenuated expression and activity of ET-1 and its A-type receptor on pulmonary arterial smooth muscle was a likely contributor to the stimulatory effects of ROCK inhibition on apoptosis. In addition, our data suggest that ROCK-II may be dominant in enhancing survival of pulmonary arterial smooth muscle.
Collapse
Affiliation(s)
- Emily Z Xu
- Clinical Integrative Biology, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
During the development of the pulmonary vasculature in the fetus, many structural and functional changes occur to prepare the lung for the transition to air breathing. The development of the pulmonary circulation is genetically controlled by an array of mitogenic factors in a temporo-spatial order. With advancing gestation, pulmonary vessels acquire increased vasoreactivity. The fetal pulmonary vasculature is exposed to a low oxygen tension environment that promotes high intrinsic myogenic tone and high vasocontractility. At birth, a dramatic reduction in pulmonary arterial pressure and resistance occurs with an increase in oxygen tension and blood flow. The striking hemodynamic differences in the pulmonary circulation of the fetus and newborn are regulated by various factors and vasoactive agents. Among them, nitric oxide, endothelin-1, and prostaglandin I2 are mainly derived from endothelial cells and exert their effects via cGMP, cAMP, and Rho kinase signaling pathways. Alterations in these signaling pathways may lead to vascular remodeling, high vasocontractility, and persistent pulmonary hypertension of the newborn.
Collapse
Affiliation(s)
- Yuansheng Gao
- Department of Physiology and Pathophysiology, Peking University, Health Science Center, Beijing, China; and Department of Pediatrics, University of Illinois, College of Medicine at Chicago, Chicago, Illinois
| | - J. Usha Raj
- Department of Physiology and Pathophysiology, Peking University, Health Science Center, Beijing, China; and Department of Pediatrics, University of Illinois, College of Medicine at Chicago, Chicago, Illinois
| |
Collapse
|
20
|
Ziino AJA, Ivanovska J, Belcastro R, Kantores C, Xu EZ, Lau M, McNamara PJ, Tanswell AK, Jankov RP. Effects of rho-kinase inhibition on pulmonary hypertension, lung growth, and structure in neonatal rats chronically exposed to hypoxia. Pediatr Res 2010; 67:177-82. [PMID: 19858775 DOI: 10.1203/pdr.0b013e3181c6e5a7] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Rho-kinase (ROCK) inhibitors prevent pulmonary hypertension (PHT) in adult rodents, but little is known about their effects on the neonatal lung. Our objective was to examine the effects of ROCK inhibition on chronic hypoxia (CH)-induced PHT and abnormal lung structure in the neonatal rat. Pups were exposed to air or CH from postnatal d 1-14 while receiving Y-27632 (5 or 10 mg x kg(-1) x d(-1)), fasudil (20 mg x kg(-1) x d(-1)), or saline intraperitoneally. Relative to air, CH-exposed pups had increased pulmonary vascular resistance, right ventricular hypertrophy, arterial medial wall thickening, and abnormal distal airway morphology characterized by septal thinning and decreased secondary septation. Treatment with 10 mg/kg Y-27632 or fasudil attenuated the structural and hemodynamic changes of PHT while having no effect on septal thinning or inhibited secondary septation. In addition, Y-27632 (10 mg/kg) and fasudil augmented CH-induced somatic growth restriction. Pulmonary arteries of CH-exposed pups had increased ROCK activity, up-regulated expression of PDGF-BB and increased smooth muscle DNA synthesis, all of which were attenuated by treatment with 10 mg/kg Y-27632. Systemically administered ROCK inhibitors prevented PHT in the CH-exposed neonatal rat but at the cost of inhibited somatic growth. Limiting effects on vascular remodeling likely resulted, in major part, from attenuated vascular PDGF-BB/beta-receptor signaling.
Collapse
Affiliation(s)
- Adrian J A Ziino
- Clinical Integrative Biology, Sunnybrook Research Institute, Toronto, Ontario, Canada M4N 3M5
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Masood A, Yi M, Lau M, Belcastro R, Shek S, Pan J, Kantores C, McNamara PJ, Kavanagh BP, Belik J, Jankov RP, Tanswell AK. Therapeutic effects of hypercapnia on chronic lung injury and vascular remodeling in neonatal rats. Am J Physiol Lung Cell Mol Physiol 2009; 297:L920-30. [DOI: 10.1152/ajplung.00139.2009] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Permissive hypercapnia, achieved using low tidal volume ventilation, has been an effective protective strategy in patients with acute respiratory distress syndrome. To date, no such protective effect has been demonstrated for the chronic neonatal lung injury, bronchopulmonary dysplasia. The objective of our study was to determine whether evolving chronic neonatal lung injury, using a rat model, is resistant to the beneficial effects of hypercapnia or simply requires a less conservative approach to hypercapnia than that applied clinically to date. Neonatal rats inhaled air or 60% O2 for 14 days with or without 5.5% CO2. Lung parenchymal neutrophil and macrophage numbers were significantly increased by hyperoxia alone, which was associated with interstitial thickening and reduced secondary crest formation. The phagocyte influx, interstitial thickening, and impaired alveolar formation were significantly attenuated by concurrent hypercapnia. Hyperoxic pups that received 5.5% CO2 had a significant increase in alveolar number relative to air-exposed pups. Increased tyrosine nitration, a footprint for peroxynitrite-mediated reactions, arteriolar medial wall thickening, and both reduced small peripheral pulmonary vessel number and VEGF and angiopoietin-1 (Ang-1) expression, which were observed with hyperoxia, was attenuated by concurrent hypercapnia. We conclude that evolving chronic neonatal lung injury in a rat model is responsive to the beneficial effects of hypercapnia. Inhaled 5.5% CO2 provided a significant degree of protection against parenchymal and vascular injury in an animal model of chronic neonatal lung injury likely due, at least in part, to its inhibition of a phagocyte influx.
Collapse
Affiliation(s)
- Azhar Masood
- Canadian Institutes of Health Research Group in Lung Development, and
- Lung Biology Programme, Physiology and Experimental Medicine, Hospital for Sick Children Research Institute
- Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Man Yi
- Canadian Institutes of Health Research Group in Lung Development, and
- Lung Biology Programme, Physiology and Experimental Medicine, Hospital for Sick Children Research Institute
- Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Mandy Lau
- Canadian Institutes of Health Research Group in Lung Development, and
- Lung Biology Programme, Physiology and Experimental Medicine, Hospital for Sick Children Research Institute
- Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Rosetta Belcastro
- Canadian Institutes of Health Research Group in Lung Development, and
- Lung Biology Programme, Physiology and Experimental Medicine, Hospital for Sick Children Research Institute
| | - Samuel Shek
- Canadian Institutes of Health Research Group in Lung Development, and
- Lung Biology Programme, Physiology and Experimental Medicine, Hospital for Sick Children Research Institute
| | - Jingyi Pan
- Canadian Institutes of Health Research Group in Lung Development, and
- Lung Biology Programme, Physiology and Experimental Medicine, Hospital for Sick Children Research Institute
| | - Crystal Kantores
- Clinical Integrative Biology, Sunnybrook Research Institute; and
| | - Patrick J. McNamara
- Lung Biology Programme, Physiology and Experimental Medicine, Hospital for Sick Children Research Institute
- Paediatrics, and
| | - Brian P. Kavanagh
- Canadian Institutes of Health Research Group in Lung Development, and
- Lung Biology Programme, Physiology and Experimental Medicine, Hospital for Sick Children Research Institute
- Departments of 4Anaesthesia,
- Critical Care Medicine,
- Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Jaques Belik
- Canadian Institutes of Health Research Group in Lung Development, and
- Lung Biology Programme, Physiology and Experimental Medicine, Hospital for Sick Children Research Institute
- Paediatrics, and
- Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Robert P. Jankov
- Clinical Integrative Biology, Sunnybrook Research Institute; and
- Paediatrics, and
- Physiology, University of Toronto, Toronto, Ontario, Canada
| | - A. Keith Tanswell
- Canadian Institutes of Health Research Group in Lung Development, and
- Lung Biology Programme, Physiology and Experimental Medicine, Hospital for Sick Children Research Institute
- Paediatrics, and
- Physiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
22
|
|
23
|
McNamara PJ, Murthy P, Kantores C, Teixeira L, Engelberts D, van Vliet T, Kavanagh BP, Jankov RP. Acute vasodilator effects of Rho-kinase inhibitors in neonatal rats with pulmonary hypertension unresponsive to nitric oxide. Am J Physiol Lung Cell Mol Physiol 2008; 294:L205-13. [DOI: 10.1152/ajplung.00234.2007] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Pulmonary hypertension (PHT) in neonates is often refractory to the current best therapy, inhaled nitric oxide (NO). The utility of a new class of pulmonary vasodilators, Rho-kinase (ROCK) inhibitors, has not been examined in neonatal animals. Our objective was to examine the activity and expression of RhoA/ROCK in normal and injured pulmonary arteries and to determine the short-term pulmonary hemodynamic (assessed by pulse wave Doppler) effects of ROCK inhibitors (15 mg/kg ip Y-27632 or 30 mg/kg ip fasudil) in two neonatal rat models of chronic PHT with pulmonary vascular remodeling (chronic hypoxia, 0.13 FiO2, or 1 mg·kg−1·day−1 ip chronic bleomycin for 14 days from birth). Activity of the RhoA/ROCK pathway and ROCK expression were increased in hypoxia- and bleomycin-induced PHT. In both models, severe PHT [characterized by raised pulmonary vascular resistance (PVR) and impaired right ventricular (RV) performance] did not respond acutely to inhaled NO (20 ppm for 15 min) or to a single bolus of a NO donor, 3-morpholinosydnonimine hydrochloride (SIN-1; 2 μg/kg ip). In contrast, a single intraperitoneal bolus of either ROCK inhibitor (Y-27632 or fasudil) completely normalized PVR but had no acute effect on RV performance. ROCK-mediated vasoconstriction appears to play a key role in chronic PHT in our two neonatal rat models. Inhibitors of ROCK have potential as a testable therapy in neonates with PHT that is refractory to NO.
Collapse
|
24
|
Jankov RP, Kantores C, Pan J, Belik J. Contribution of xanthine oxidase-derived superoxide to chronic hypoxic pulmonary hypertension in neonatal rats. Am J Physiol Lung Cell Mol Physiol 2008; 294:L233-45. [DOI: 10.1152/ajplung.00166.2007] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Xanthine oxidase (XO)-derived reactive oxygen species (ROS) formation contributes to experimental chronic hypoxic pulmonary hypertension in adults, but its role in neonatal pulmonary hypertension has received little attention. In rats chronically exposed to hypoxia (13% O2) for 14 days from birth, we examined the effects of ROS scavengers (U74389G 10 mg·kg−1·day−1 or Tempol 100 mg·kg−1·day−1 ip) or a XO inhibitor, Allopurinol (50 mg·kg−1·day−1 ip). Both ROS scavengers limited oxidative stress in the lung and attenuated hypoxia-induced vascular remodeling, confirming a critical role for ROS in this model. However, both interventions also significantly inhibited somatic growth and normal cellular proliferation in distal air spaces. Hypoxia-exposed pups had evidence of increased serum and lung XO activity, increased vascular XO-derived superoxide production, and vascular nitrotyrosine formation. These changes were all prevented by treatment with Allopurinol, which also attenuated hypoxia-induced vascular remodeling and partially reversed inhibited endothelium-dependent arterial relaxation, without affecting normal growth and proliferation. Collectively, our findings suggest that XO-derived superoxide induces endothelial dysfunction, thus impairing pulmonary arterial relaxation, and contributes to vascular remodeling in hypoxia-exposed neonatal rats. Due to the potential for adverse effects on normal growth, targeting XO may represent a superior “antioxidant” strategy to ROS scavengers for neonates with pulmonary hypertension.
Collapse
|
25
|
Chicoine LG, Paffett ML, Girton MR, Metropoulus MJ, Joshi MS, Bauer JA, Nelin LD, Resta TC, Walker BR. Maturational changes in the regulation of pulmonary vascular tone by nitric oxide in neonatal rats. Am J Physiol Lung Cell Mol Physiol 2007; 293:L1261-70. [PMID: 17827249 DOI: 10.1152/ajplung.00235.2006] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Nitric oxide (NO) is an important regulator of vasomotor tone in the pulmonary circulation. We tested the hypothesis that the role NO plays in regulating vascular tone changes during early postnatal development. Isolated, perfused lungs from 7- and 14-day-old Sprague-Dawley rats were studied. Baseline total pulmonary vascular resistance (PVR) was not different between age groups. The addition of KCl to the perfusate caused a concentration-dependent increase in PVR that did not differ between age groups. However, the nitric oxide synthase (NOS) inhibitor N(omega)-nitro-L-arginine augmented the K(+)-induced increase in PVR in both groups, and the effect was greater in lungs from 14-day-old rats vs. 7-day-old rats. Lung levels of total endothelial, inducible, and neuronal NOS proteins were not different between groups; however, the production rate of exhaled NO was greater in lungs from 14-day-old rats compared with those of 7-day-old rats. Vasodilation to 0.1 microM of the NO donor spermine NONOate was greater in 14-day lungs than in 7-day lungs, and lung levels of both soluble guanylyl cyclase and cGMP were greater at 14 days than at 7 days. Vasodilation to 100 microM of the cGMP analog 8-(4-chlorophenylthio)guanosine-3',5'-cyclic monophosphate was greater in 7-day lungs than in 14-day lungs. Our results demonstrate that the pulmonary vascular bed depends more on NO production to modulate vascular tone at 14 days than at 7 days of age. The observed differences in NO sensitivity may be due to maturational increases in soluble guanylyl cyclase protein levels.
Collapse
Affiliation(s)
- Louis G Chicoine
- Center for Gene Therapy, Columbus Children's Research Institute, The Ohio State University, Columbus, Ohio, Columbus, OH 43205, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Yi SL, Kantores C, Belcastro R, Cabacungan J, Tanswell AK, Jankov RP. 8-Isoprostane-induced endothelin-1 production by infant rat pulmonary artery smooth muscle cells is mediated by Rho-kinase. Free Radic Biol Med 2006; 41:942-9. [PMID: 16934677 DOI: 10.1016/j.freeradbiomed.2006.05.035] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2005] [Revised: 05/26/2006] [Accepted: 05/31/2006] [Indexed: 11/20/2022]
Abstract
We have reported that 8-isoprostane stimulated the production of endothelin (ET)-1, a potent vasoconstrictor and critical mediator of chronic pulmonary hypertension, by infant rat pulmonary artery smooth muscle cells (PASMCs), through stimulation of the thromboxane A2 receptor. The aim of this study was to examine the contribution of putative downstream intracellular mediators of thromboxane A2 receptor stimulation to this effect. PASMCs from infant rats were treated with calcium ionophore (A23187), 8-isoprostane, or 8-isoprostane together with inhibitors of tyrosine kinase, protein kinase C, phosphatidylinositol 3-kinase, mitogen-activated protein kinases, or Rho-kinases (ROCK). A23187 had no effect on ET-1 production, excluding raised intracellular Ca2+ as a major contributor. Increased ET-1 production induced by 8-isoprostane was significantly attenuated by the ROCK inhibitors Y-27632 and hydroxyfasudil, but not by inhibitors of the other pathways. 8-Isoprostane also increased membrane binding of RhoA, a major determinant of ROCK activity, and ROCK-II expression through the protein kinase C pathway. These data indicate that the RhoA/ROCK pathway mediates increased ET-1 production by PASMCs, which we speculate may at least partly explain the beneficial effects of both antioxidants and ROCK inhibitors in animal models of chronic pulmonary hypertension.
Collapse
Affiliation(s)
- Soojin L Yi
- Clinical Integrative Biology, Sunnybrook Research Institute, Toronto, ON, Canada
| | | | | | | | | | | |
Collapse
|
27
|
Liao L, Ning Q, Li Y, Wang W, Wang A, Wei W, Liu X, Auten RL, Tanswell AK, Luo X. CXCR2 blockade reduces radical formation in hyperoxia-exposed newborn rat lung. Pediatr Res 2006; 60:299-303. [PMID: 16923948 DOI: 10.1203/01.pdr.0000233058.08200.d6] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Inflammation contributes greatly to the pathogenesis of bronchopulmonary dysplasia. In previous studies, we showed that blocking neutrophil influx by treatment with SB265610, a selective CXCR2 antagonist, could partly reduce superoxide accumulation and preserve alveolar development in 60% O(2)-exposed newborn rats. The purpose of this study was to further investigate the role of neutrophils in the formation of reactive oxygen and nitrogen species mediating hyperoxia-impaired lung development. We found that hydroxyl radical formation and lipid peroxidation in rat lungs were significantly increased during 60% O(2) exposure. These increases were attenuated by the administration of SB265610. In addition, SB265610 largely inhibited protein nitration induced by hyperoxia. SB265610 partly prevented the hyperoxia-enhanced bronchoalveolar lavage (BAL) protein content in 60% O(2)-exposed animals. Our results demonstrate that neutrophils have a pivotal role in hydroxyl radical formation, lipid peroxidation and protein nitration. Taken together with our previous studies, the present findings show that blocking neutrophil influx protects alveolar development and improves lung function in part by preventing reactive oxygen/nitrogen species accumulation.
Collapse
Affiliation(s)
- Lingjie Liao
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Jankov RP, Kantores C, Belcastro R, Yi M, Tanswell AK. Endothelin-1 inhibits apoptosis of pulmonary arterial smooth muscle in the neonatal rat. Pediatr Res 2006; 60:245-51. [PMID: 16857764 DOI: 10.1203/01.pdr.0000233056.37254.0b] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Vascular wall remodeling in pulmonary hypertension is contributed to by an aberration in the normal balance between proliferation and apoptosis of smooth muscle. We observed that endothelin (ET)-1 is a critical mediator of vascular remodeling in neonatal rats chronically exposed to 60% O(2), but has no direct proliferative effects on cultured neonatal rat pulmonary artery smooth muscle cells (PASMCs). These findings led us to hypothesize that ET-1 may modulate remodeling by inhibiting apoptosis of smooth muscle. ET-1 (0.1 microM) was found to significantly attenuate both Paclitaxel- and serum deprivation-induced PASMC apoptosis, likely through stimulation of the ET(A) receptor (ET(A)R). ET-1 also prevented Paclitaxel-induced up-regulation of pro-apoptotic Bax and cleaved (activated) caspase-3. In rat pups exposed from birth to 60% O(2) for 7 d, arterial wall expression of Bax was decreased and expression of both ET(A)R and anti-apoptotic Bcl-xL were increased. Furthermore, increased numbers of TUNEL-positive cells were evident in the walls of pulmonary arteries from 60% O(2)-exposed animals treated with a combined ET receptor antagonist, SB217242, relative to air-exposed and vehicle-treated groups. Together, these findings suggest that ET-1 mediates remodeling of neonatal rat pulmonary arteries by inhibiting smooth muscle apoptosis.
Collapse
Affiliation(s)
- Robert P Jankov
- Clinical Integrative Biology, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario M5S 1A8, Canada.
| | | | | | | | | |
Collapse
|
29
|
Kantores C, McNamara PJ, Teixeira L, Engelberts D, Murthy P, Kavanagh BP, Jankov RP. Therapeutic hypercapnia prevents chronic hypoxia-induced pulmonary hypertension in the newborn rat. Am J Physiol Lung Cell Mol Physiol 2006; 291:L912-22. [PMID: 16829630 DOI: 10.1152/ajplung.00480.2005] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Induction of hypercapnia by breathing high concentrations of carbon dioxide (CO(2)) may have beneficial effects on the pulmonary circulation. We tested the hypothesis that exposure to CO(2) would protect against chronic pulmonary hypertension in newborn rats. Atmospheric CO(2) was maintained at <0.5% (normocapnia), 5.5%, or 10% during exposure from birth for 14 days to normoxia (21% O(2)) or moderate hypoxia (13% O(2)). Pulmonary vascular and hemodynamic abnormalities in animals exposed to chronic hypoxia included increased pulmonary arterial resistance, right ventricular hypertrophy and dysfunction, medial thickening of pulmonary resistance arteries, and distal arterial muscularization. Exposure to 10% CO(2) (but not to 5.5% CO(2)) significantly attenuated pulmonary vascular remodeling and increased pulmonary arterial resistance in hypoxia-exposed animals (P < 0.05), whereas both concentrations of CO(2) normalized right ventricular performance. Exposure to 10% CO(2) attenuated increased oxidant stress induced by hypoxia, as quantified by 8-isoprostane content in the lung, and prevented upregulation of endothelin-1, a critical mediator of pulmonary vascular remodeling. We conclude that hypercapnic acidosis has beneficial effects on pulmonary hypertension and vascular remodeling induced by chronic hypoxia, which we speculate derives from antioxidant properties of CO(2) on the lung and consequent modulating effects on the endothelin pathway.
Collapse
Affiliation(s)
- Crystal Kantores
- Clinical Integrative Biology, Sunnybrook Research Institute, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
30
|
Jankov RP, Kantores C, Belcastro R, Yi S, Ridsdale RA, Post M, Tanswell AK. A role for platelet-derived growth factor beta-receptor in a newborn rat model of endothelin-mediated pulmonary vascular remodeling. Am J Physiol Lung Cell Mol Physiol 2005; 288:L1162-70. [PMID: 15722379 DOI: 10.1152/ajplung.00180.2004] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Newborn rats exposed to 60% O2 for 14 days develop endothelin (ET)-1-dependent pulmonary hypertension with vascular remodeling, characterized by increased smooth muscle cell (SMC) proliferation and medial thickening of pulmonary resistance arteries. Using immunohistochemistry and Western blot analyses, we examined the effect of exposure to 60% O2 on expression in the lung of receptors for the platelet-derived growth factors (PDGF), which are implicated in the pathogenesis of arterial smooth muscle hyperplasia. We observed a marked O2-induced upregulation of PDGF-alpha and -beta receptors (PDGF-alphaR and -betaR) on arterial smooth muscle. This led us to examine pulmonary vascular PDGF receptor expression in 60% O2-exposed rats given SB-217242, a combined ET receptor antagonist, which we found prevented the O2-induced upregulation of PDGF-betaR, but not PDGF-alphaR, on arterial smooth muscle. PDGF-BB, a major PDGF-betaR ligand, was found to be a potent in vitro inducer of hyperplasia and DNA synthesis in cultured pulmonary artery SMC from infant rats. A critical role for PDGF-betaR ligands in arterial SMC proliferation was confirmed in vivo using a truncated soluble PDGF-betaR intervention, which attenuated SMC proliferation induced by exposure to 60% O2. Collectively, these data are consistent with a major role for PDGF-betaR-mediated SMC proliferation, acting downstream of increased ET-1 in a newborn rat model of 60% O2-induced pulmonary hypertension.
Collapse
Affiliation(s)
- Robert P Jankov
- Clinical Integrative Biology, Sunnybrook & Women's Research Institute, Toronto, Ontario, Canada.
| | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
Isoprostanes were first recognized as convenient markers of oxidative stress, but their powerful effects on a variety of cell functions are now also being increasingly appreciated. This is particularly true of the lung, which is comprised of a wide variety of different cell types (smooth muscle, innervation, epithelium, lymphatics, etc.), all of which have been shown to respond to exogenously applied isoprostanes. In this review, we summarize these biological responses in the lung, and also consider the roles that isoprostanes might play in a range of pulmonary clinical disorders.
Collapse
Affiliation(s)
- Luke J Janssen
- Asthma Research Group, Father Sean O'Sullivan Research Center, Firestone Institute for Respiratory Health, St. Joseph's Hospital, Department of Medicine, McMaster University, Hamilton, Ontario, Canada.
| | | | | |
Collapse
|
32
|
Abstract
Isoprostanes are widely recognized as useful markers of membrane lipid peroxidation. It seems to be less well appreciated, however, that they also elicit important biological responses, even though this was first shown at the same time that they were introduced as markers of oxidative stress. The past several years have seen the list of cells/tissues which are sensitive to isoprostanes grow considerably: in fact, as we summarize here, there is now evidence that essentially every cell type in the lung responds in some pathologically relevant way to isoprostanes. In this sense, they might well be considered as not just markers of oxidative stress and inflammation, but also as a novel group of inflammatory mediators. Moreover, in addition to their pathological effects, we summarize here the evidence which has led us to hypothesize that isoprostanes could play an important role in vascular smooth muscle physiology as "endothelium-derived hyperpolarizing factors."
Collapse
Affiliation(s)
- L J Janssen
- Department of Medicine, Asthma Research Group, Father Sean O'Sullivan Research Center, Firestone Institute for Respiratory Health, St. Joseph's Hospital, McMaster University, Hamilton, Ont., Canada L8N4A6.
| |
Collapse
|
33
|
Affiliation(s)
- Robert P Jankov
- Canadian Institutes of Health Research (CIHR) Group in Lung Development and Lung Biology Programme, The Hospital for Sick Children Research Institute, Toronto, Ontario M5G 1X8, Canada
| | | |
Collapse
|
34
|
Belik J, Jankov RP, Pan J, Yi M, Chaudhry I, Tanswell AK. Chronic O2 exposure in the newborn rat results in decreased pulmonary arterial nitric oxide release and altered smooth muscle response to isoprostane. J Appl Physiol (1985) 2004; 96:725-30. [PMID: 14565964 DOI: 10.1152/japplphysiol.00825.2003] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Chronic oxygen exposure in the newborn rat results in lung isoprostane formation, which may contribute to the pulmonary hypertension evident in this animal model. The purpose of this study was to investigate the pulmonary arterial smooth muscle responses to 8-iso-prostaglandin F2α (8-iso-PGF2a) in newborn rats exposed to 60% O2 for 14 days. Because, in the adult rat, 8-iso-PGF2α may have a relaxant effect, mediated by nitric oxide (NO), we also sought to evaluate the pulmonary arterial NO synthase (NOS) protein content and NO release in the newborn exposed to chronic hyperoxia. Compared with air-exposed control animals, 8-iso-PGF2a induced a significantly greater force ( P < 0.01) and reduced ( P < 0.01) relaxation of precontracted pulmonary arteries in the 60% O2-treated animals. These changes were reproduced in control pulmonary arteries by NOS blockade by using NG-nitro-l-arginine methyl ester. Pulmonary arterial endothelial NOS was unaltered, but the inducible NOS protein content was significantly decreased ( P < 0.01) in the experimental group. Pulmonary ( P < 0.05) and aortic ( P < 0.01) tissue ex vivo NO accumulation was significantly reduced in the 60% O2-treated animals. We speculate that impaired pulmonary vascular tissue NO metabolism after chronic O2 exposure potentiates 8-iso-PGF2α-induced vasoconstriction in the newborn rat, thus contributing to pulmonary hypertension.
Collapse
Affiliation(s)
- J Belik
- Department of Paediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada M5G 1X8.
| | | | | | | | | | | |
Collapse
|
35
|
Spiropoulos K, Trakada G, Nikolaou E, Prodromakis E, Efremidis G, Pouli A, Koniavitou A. Endothelin-1 levels in the pathophysiology of chronic obstructive pulmonary disease and bronchial asthma. Respir Med 2003; 97:983-9. [PMID: 12924528 DOI: 10.1016/s0954-6111(03)00129-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
BACKGROUND Endothelin-1 (ET-1) has been implicated in the pathogenesis of asthma and chronic obstructive pulmonary disease (COPD). The ET-1 levels are elevated during exacerbations of asthma and COPD in bronchoalveolar lavage, serum, and sputum and fails with treatment of the exacerbations. Hypoxemia stimulates ET-1 secretion. OBJECTIVE The aim of this study was to examine the serum ET-1 levels in stable asthmatic and COPD patients. MATERIALS AND METHODS We examined 48 COPD and 26 asthmatic patients and 34 normal subjects. We collected arterial samples to measure baseline ET-1 levels in all patients and in the control group, during the day. All the patients underwent formal polysomnography (EEG, ECG, airflow, respiratory muscle movement, oximeter) to detect the presence of nocturnal, nonapneic, and oxyhemoglobin desaturation. Twelve of the COPD patients and six of the asthmatic patients were disqualified because of inadequate sleep or sleep apnea syndrome. Nineteen of the COPD patients desaturated below a baseline sleep saturation of 90% for 5 min or more, reaching a nadir saturation of at least 85%. We collected arterial samples to measure ET-1 levels, 5 min after the first period of desaturation in each of the 19 desaturators COPD patients. None of the 20 asthmatic patients exhibited oxyhemoglobin desaturation during sleep. RESULTS Baseline arterial ET-1 levels during the day were significantly higher in "desaturators" COPD patients (2.08+/-0.28 pg/ml) compared to "non-desaturators" COPD patients (1.38+/-0.16 pg/ml) (P<0.001) and to asthmatics (0.7+/-0.85 pg/ml) (P<0.001). ET-1 Levels in normal subjects were 1.221+/-0.02 pg/ml. In "desaturators" COPD patients ET-1 levels during the night, 5 min after the first oxyhemoglobin desaturation, were significantly higher (4.28+/-1.10 pg/ml) compared to those during the day (2.08+/-0.28 pg/ml) (P<0.001). A significant negative correlation was observed between ET-1 levels and degree of desaturation during the day (P=0.005, r=0.632) and during the night (P<0.001, r=0.930) in "desaturators" COPD patients. CONCLUSION According to our results: (1) ET-1 levels were significantly higher in "desaturators" COPD patients than in "non-desaturators" COPD and in asthmatics; (2) ET-1 levels were significantly higher during the night than during the day in "desaturators" COPD patients; (3) the degree of desaturation correlated negatively with the ET-1 levels in "desaturators" COPD patients, both during daytime and nighttime. These findings are consistent with the hypothesis that ET-1 is implicated in the pathophysiology of asthma and COPD, especially if nocturnal, nonapneic, oxyhemoglobin desaturation exists.
Collapse
Affiliation(s)
- K Spiropoulos
- Division of Pulmonology, Laboratory of Sleep, University of Patras Medical School, Patras 26 500, Greece.
| | | | | | | | | | | | | |
Collapse
|
36
|
Belik J, Jankov RP, Pan J, Tanswell AK. Chronic O2 exposure enhances vascular and airway smooth muscle contraction in the newborn but not adult rat. J Appl Physiol (1985) 2003; 94:2303-12. [PMID: 12562676 DOI: 10.1152/japplphysiol.00820.2002] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Neonatal rats exposed to 60% O(2) for 14 days develop lung changes compatible with human bronchopulmonary dysplasia and pulmonary hypertension. Our aim was to evaluate and compare the newborn and adult rat pulmonary vascular and airway smooth muscle force generation and relaxation potential after exposure to 60% O(2) for 14 days. Vascular and airway intrapulmonary rings 100 microm in diameter were mounted on a myograph and bathed in Krebs-Henseleit solution bubbled with air- 6% CO(2) at 37 degrees C. Significant age-dependent changes in intrapulmonary arteries and their neighboring airway muscle properties were observed. Whereas hyperoxia enhanced force in neonatal vascular and airway muscle, the opposite was seen in adult samples. No changes in endothelium-dependent vascular relaxation were observed at either age, but the dose response to an endothelium-independent NO donor was altered. In the newborn experimental animals, the relaxation was reduced, whereas, in their adult counterparts, it was enhanced. After O(2) exposure, the bronchial muscle relaxation response to epithelium-dependent and -independent stimulation was not altered in either age group, whereas the epithelium-dependent response was decreased only in the adult. The antioxidant Trolox, or an endothelin-A and -B receptor antagonist, reversed the vascular and airway muscle's hyperoxia-induced changes. We conclude that chronic O(2) exposure in the newborn rat results in enhanced lung vascular and airway muscle contraction potential via a mechanism involving reactive oxygen species and the endothelin pathway. The present findings also suggest that the newborn is more susceptible to airway hyperresponsiveness after chronic O(2) exposure.
Collapse
Affiliation(s)
- J Belik
- Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada M5G 1X8.
| | | | | | | |
Collapse
|
37
|
Abstract
During the fetal period, blood is oxygenated through the placenta, and most of the cardiac output bypasses the lung through the ductus arteriosus. At birth, pulmonary vascular resistance falls with the initiation of ventilation. Coincidentally, the ductus arteriosus constricts. Endothelin-1 (ET-1) appears to play an important role during that transition period and postnatally. ET-1 can dramatically increase resistance in the placental microcirculation and may be involved in blood flow redistribution with hypoxia. At birth, the increase in oxygen tension is important in triggering ductus vasoconstriction. It is proposed that oxygen triggers closure of the ductus arteriosus by activating a specific, cytochrome P450-linked reaction, which in turn stimulates the synthesis of ET-1. On the neonatal heart, ET-1 has a positive chronotropic but negative inotropic effect. In the newborn piglet and the fetal lamb, both term and preterm, ET-1 causes a potent, long-lasting pulmonary vasoconstriction. Furthermore, a transient dilator response has been identified, and it is ascribed to nitric oxide formation. ET receptors are abundant in the piglet pulmonary vasculature. They are predominantly of the ETA constrictor subtype, though ETB2 constrictor receptors may also be present in certain species. The dilator response is linked to the ETB1 receptor, and the number of ETB1 receptors is reduced in hypoxia-induced pulmonary hypertension. ET-1 appears to be a causative agent in the pathogenesis of hypoxia- and hyperoxia-induced pulmonary hypertension as demonstrated by reversal of hemodynamic and morphological changes with treatment with an ETA receptor antagonist. Findings are amenable to practical applications in the management of infants with pulmonary hypertension or requiring persistent patency of the ductus arteriosus.
Collapse
Affiliation(s)
- Thérèse Perreault
- Newborn Medicine, Department of Pediatrics, The Montreal Children's Hospital, McGill University, 2300 Tupper Street, Montreal, QC H3H 1P3, Canada.
| | | |
Collapse
|
38
|
Corl CM, Cao YZ, Cohen ZS, Sordillo LM. Oxidant stress enhances Lyso-PAF-AcT activity by modifying phospholipase D and phosphatidic acid in aortic endothelial cells. Biochem Biophys Res Commun 2003; 302:610-4. [PMID: 12615078 DOI: 10.1016/s0006-291x(03)00223-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Oxidant stress, as a consequence of selenium (Se) deficiency, alters production of vasoactive compounds including platelet-activating factor (PAF). Recent studies report that enhanced PAF production during Se deficiency is a consequence of increased lyso-PAF:acetyl-coenzyme A acetyltransferase (Lyso-PAF-AcT) activity. To elucidate the mechanism behind increased Lyso-PAF-AcT activity during oxidant stress, phospholipase D (PLD) activity and phosphatidic acid (PA) production were examined. Increased PLD activity and PA production were exhibited in bovine aortic endothelial cells using a Se-deficient model of oxidant stress. The direct effects of PLD and PA on Lyso-PAF-AcT activity were assessed using selective inhibitors and repletion experiments. Following the inhibition of PLD and addition of exogenous PA, Lyso-PAF-AcT activity significantly decreased and increased, respectively. Therefore, Se deficiency enhances Lyso-PAF-AcT activity in part by modifying PLD and PA. This suggests a novel link between Se status and PAF production, providing potential upstream therapeutic targets for PAF regulation under conditions of oxidant stress.
Collapse
Affiliation(s)
- C M Corl
- Department of Veterinary Science, The Pennsylvania State University, 115 Henning Building, University Park, PA 16802, USA
| | | | | | | |
Collapse
|
39
|
Jankov RP, Belcastro R, Ovcina E, Lee J, Massaeli H, Lye SJ, Tanswell AK. Thromboxane A(2) receptors mediate pulmonary hypertension in 60% oxygen-exposed newborn rats by a cyclooxygenase-independent mechanism. Am J Respir Crit Care Med 2002; 166:208-14. [PMID: 12119234 DOI: 10.1164/rccm.200112-124oc] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Endothelin-1 (ET-1) mediates the development of pulmonary hypertension (PHT) in newborn rats exposed to 60% O(2) for 14 days, a model for human chronic neonatal lung injury. ET-1 production by d-14 rat pulmonary artery smooth muscle cells in vitro was markedly increased by thromboxane (TX) A(2) receptor agonists and inhibited by a competitive antagonist. We hypothesized that stimulation of the TX A(2) receptor contributed to O(2)-mediated PHT in vivo. Newborn rat pups received daily intraperitoneal injections of L670596, a competitive TX A(2) receptor antagonist, or 5,5-dimethyl-3-(3-fluorophenyl)4-(4-methylsulfonyl)phenyl-2(5H)-furanone (DFU), a cyclooxygenase-2 inhibitor, during 14 days of 60% O(2) or air exposure. L670596, but not DFU, prevented 60% O(2)-mediated right ventricular and small pulmonary vessel smooth muscle hypertrophy. Lung ET-1 content was significantly reduced by L670596 in 60% O(2)-exposed animals. We conclude that TX A(2) receptor activation, though not by TX A(2), caused upregulation of ET-1 and PHT in this model. A likely mediator is the stable lipid peroxidation product, 8-iso-prostane, which acts as an incidental ligand of the TX A(2) receptor and is a potent inducer of ET-1 production by cultured d-14 rat pulmonary artery smooth muscle cells in vitro.
Collapse
MESH Headings
- 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology
- Animals
- Animals, Newborn
- Blotting, Western
- Carbazoles/pharmacology
- Cells, Cultured
- Cyclooxygenase Inhibitors/pharmacology
- Dinoprost/analogs & derivatives
- Endothelin-1/metabolism
- F2-Isoprostanes/metabolism
- F2-Isoprostanes/pharmacology
- Furans/pharmacology
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/physiopathology
- Hypertrophy, Right Ventricular/physiopathology
- Immunohistochemistry
- Lung/metabolism
- Muscle, Smooth, Vascular/metabolism
- Oxygen/physiology
- Oxygen/toxicity
- Prostaglandin Antagonists/pharmacology
- Prostaglandin-Endoperoxide Synthases/physiology
- Pulmonary Artery/metabolism
- Rats
- Receptors, Thromboxane/antagonists & inhibitors
- Receptors, Thromboxane/physiology
- Thromboxane B2/metabolism
- Up-Regulation
- Vasoconstrictor Agents/pharmacology
Collapse
Affiliation(s)
- Robert P Jankov
- Canadian Institutes of Health Research Group in Lung Development, Lung Biology Programme, Toronto, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
40
|
Janssen LJ, Tazzeo T. Involvement of TP and EP3 receptors in vasoconstrictor responses to isoprostanes in pulmonary vasculature. J Pharmacol Exp Ther 2002; 301:1060-6. [PMID: 12023538 DOI: 10.1124/jpet.301.3.1060] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Although isoprostanes generally act on smooth muscle via TXA(2)-selective prostanoid receptors (TPs), some suggest other prostanoid receptors or possibly even a novel isoprostane-selective receptor might be involved. We studied contractions to several isoprostanes in porcine pulmonary vasculature using organ bath techniques. 8-iso-prostaglandin E(2) (PGE(2)) was the most potent and efficacious of the isoprostanes, with a log EC(50) of -7.0 +/- 0.2 in the pulmonary artery and -6.8 +/- 0.2 in the pulmonary vein. The responses to all the isoprostanes were essentially completely blocked by the TP receptor antagonist ICI 192605 [4(Z)-6-[(2,4,5-cis)2-(2-chlorophenyl)-4-(2-hydroxyphenyl)1,3-dioxan-5-yl]hexenoic acid], and the equilibrium dissociation constants for ICI 192605 competing with U46619 or 8-iso-PGE(2) were both approximately 2 nM, indicating that isoprostane-evoked responses involve primarily TP receptors. Only 8-iso-PGE(2) was able to evoke substantial contractions in the presence of ICI 192605 and only in the pulmonary vein. The EC(50) of these ICI 192605-insensitive responses was -6.1 +/- 0.2. Using a variety of prostanoid agonists, we found the pulmonary vein lacked excitatory PGF(2alpha)-selective prostanoid receptor (FP) or PGD(2)-selective prostanoid receptor (DP) but expressed excitatory EP(3) receptors. The ICI 192605-insensitive responses to 8-iso-PGE(2) were unaffected by the EP(1) antagonist SC-19220 [8-chloro-debenz[b,f][1,4]oxazepine-10(11H)-carboxy-(2-acetyl) hydrazine; 10(-5) M] but were antagonized by the less selective DP/EP(1)/EP(2) antagonist AH6809 (6-isopropoxy-9-oxoxanthene-2-carboxylic acid; 10(-5) M) or by cyclopiazonic acid (10(-5) M; depletes the internal Ca(2+) store). Our data indicate that, whereas 8-iso-PGE(2) constricts pulmonary vasculature primarily through TP receptors, a substantial portion of this response is also directed through EP(3) receptors or possibly a novel isoprostane receptor.
Collapse
MESH Headings
- Animals
- Dinoprostone/analogs & derivatives
- Dinoprostone/pharmacology
- Dose-Response Relationship, Drug
- In Vitro Techniques
- Isoprostanes/pharmacology
- Isoprostanes/physiology
- Lung/blood supply
- Lung/drug effects
- Lung/physiology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/physiology
- Pulmonary Artery/drug effects
- Pulmonary Artery/physiology
- Pulmonary Veins/drug effects
- Pulmonary Veins/physiology
- Receptors, Prostaglandin E/agonists
- Receptors, Prostaglandin E/antagonists & inhibitors
- Receptors, Prostaglandin E/physiology
- Receptors, Prostaglandin E, EP3 Subtype
- Receptors, Thromboxane/agonists
- Receptors, Thromboxane/antagonists & inhibitors
- Receptors, Thromboxane/physiology
- Swine
- Vasoconstriction/drug effects
- Vasoconstriction/physiology
- Vasoconstrictor Agents/pharmacology
Collapse
Affiliation(s)
- Luke J Janssen
- Asthma Research Group, Department of Medicine, McMaster University, St. Joseph's Hospital, 50 Charlton Avenue East, Hamilton, Ontario, L8N 4A6, Canada.
| | | |
Collapse
|
41
|
Abstract
Reactive oxygen and nitrogen species are considered to play a major role in the pathogenesis of a wide range of human disorders. This may be a particularly important pathogenetic mechanism in the newborn nursery. The phrase "oxygen radical disease of prematurity" has been coined to collectively describe a wide range of neonatal disorders based on the belief that premature newborns are deficient in antioxidant defenses at a time when they are subjected to acute and chronic oxidant stresses. This belief has led to a number of clinical trials of antioxidant therapies being undertaken in neonatal patients. The realization that reactive oxygen species play a critical role in neonatal illnesses has only recently been paralleled by an increased understanding of their physiologic roles. A major concern is that effective scavenging of reactive oxygen species, to attenuate their toxic effects, will also inhibit essential cellular functions such as growth in potential target organs such as lung, brain, intestine, and retina.
Collapse
Affiliation(s)
- R P Jankov
- Canadian Institutes of Health Research Group in Lung Development and Lung Biology Programme, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | | | | |
Collapse
|
42
|
Jankov RP, Luo X, Belcastro R, Copland I, Frndova H, Lye SJ, Hoidal JR, Post M, Tanswell AK. Gadolinium chloride inhibits pulmonary macrophage influx and prevents O(2)-induced pulmonary hypertension in the neonatal rat. Pediatr Res 2001; 50:172-83. [PMID: 11477200 DOI: 10.1203/00006450-200108000-00003] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Newborn rats exposed to 60% O(2) for 14 d demonstrated a bronchopulmonary dysplasia-like lung morphology and pulmonary hypertension. A 21-aminosteroid antioxidant, U74389G, attenuated both pulmonary hypertension and macrophage accumulation in the O(2)-exposed lungs. To determine whether macrophage accumulation played an essential role in the development of pulmonary hypertension in this model, pups were treated with gadolinium chloride (GdCl(3)) to reduce lung macrophage content. Treatment of 60% O(2)-exposed animals with GdCl(3) prevented right ventricular hypertrophy (p < 0.05) and smooth muscle hyperplasia around pulmonary vessels, but had no effect on morphologic changes in the lung parenchyma. In addition, GdCl(3) inhibited 60% O(2)-mediated increases in endothelin-1, 8-isoprostane, and nitrotyrosine residues. Organotypic cultures of fetal rat distal lung cells were subjected to cyclical mechanical strain to assess the potential role of GdCl(3)-induced blockade of stretch-mediated cation channels in these effects. Mechanical strain caused a moderate increase of endothelin-1 (p < 0.05), which was unaffected by GdCl(3), but had no effect on 8-isoprostane or nitric oxide synthesis. A critical role for endothelin-1 in O(2)-mediated pulmonary hypertension was confirmed using the combined endothelin receptor antagonist SB217242. We concluded that pulmonary macrophage accumulation, in response to 60% O(2), mediated pulmonary hypertension through up-regulation of endothelin-1.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Bronchopulmonary Dysplasia/etiology
- Bronchopulmonary Dysplasia/pathology
- Cell Movement/drug effects
- Cells, Cultured
- Dinoprost/analogs & derivatives
- Dinoprost/metabolism
- Endothelin-1/metabolism
- F2-Isoprostanes
- Gadolinium/pharmacology
- Humans
- Hypertension, Pulmonary/etiology
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/prevention & control
- Hypertrophy, Right Ventricular/etiology
- Hypertrophy, Right Ventricular/pathology
- Infant, Newborn
- Macrophages, Alveolar/drug effects
- Macrophages, Alveolar/pathology
- Macrophages, Alveolar/physiology
- Oxygen/toxicity
- Rats
- Rats, Sprague-Dawley
- Tyrosine/analogs & derivatives
- Tyrosine/metabolism
Collapse
Affiliation(s)
- R P Jankov
- Canadian Institutes of Health Research Groups in Lung Development, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|