1
|
Tawa M, Okamura T. Factors influencing the soluble guanylate cyclase heme redox state in blood vessels. Vascul Pharmacol 2022; 145:107023. [PMID: 35718342 DOI: 10.1016/j.vph.2022.107023] [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: 03/11/2022] [Revised: 06/09/2022] [Accepted: 06/12/2022] [Indexed: 11/15/2022]
Abstract
Soluble guanylate cyclase (sGC) plays an important role in maintaining vascular homeostasis, as an acceptor for the biological messenger nitric oxide (NO). However, only reduced sGC (with a ferrous heme) can be activated by NO; oxidized (ferric heme) and apo (absent heme) sGC cannot. In addition, the proportions of reduced, oxidized, and apo sGC change under pathological conditions. Although diseased blood vessels often show decreased NO bioavailability in the vascular wall, a shift of sGC heme redox balance in favor of the oxidized/apo forms can also occur. Therefore, sGC is of growing interest as a drug target for various cardiovascular diseases. Notably, the balance between NO-sensitive reduced sGC and NO-insensitive oxidized/apo sGC in the body is regulated in a reversible manner by various biological molecules and proteins. Many studies have attempted to identify endogenous factors and determinants that influence this redox state. For example, various reactive nitrogen and oxygen species are capable of inducing the oxidation of sGC heme. Conversely, a heme reductase and some antioxidants reduce the ferric heme in sGC to the ferrous state. This review summarizes the factors and mechanisms identified by these studies that operate to regulate the sGC heme redox state.
Collapse
Affiliation(s)
- Masashi Tawa
- Department of Pathological and Molecular Pharmacology, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka 569-1094, Japan.
| | - Tomio Okamura
- Emeritus Professor, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| |
Collapse
|
2
|
Qin X, Gao A, Hou X, Xu X, Chen L, Sun L, Hao Y, Shi Y. Connexins may play a critical role in cigarette smoke-induced pulmonary hypertension. Arch Toxicol 2022; 96:1609-1621. [PMID: 35344070 DOI: 10.1007/s00204-022-03274-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 03/02/2022] [Indexed: 11/02/2022]
Abstract
Pulmonary hypertension (PH) is a chronic progressive disease characterized by pulmonary vasoconstriction and remodeling. It causes a gradual increase in pulmonary vascular resistance leading to right-sided heart failure, and may be fatal. Chronic exposure to cigarette smoke (CS) is an essential risk factor for PH group 3; however, smoking continues to be prevalent and smoking cessation is reported to be difficult. A majority of smokers exhibit PH, which leads to a concomitant increase in the risk of mortality. The current treatments for PH group 3 focus on vasodilation and long-term oxygen supplementation, and fail to stop or reverse PH-associated continuous vascular remodeling. Recent studies have suggested that pulmonary vascular endothelial dysfunction induced by CS exposure may be an initial event in the natural history of PH, which in turn may be associated with abnormal alterations in connexin (Cx) expression. The relationship between Cx and CS-induced PH development has not yet been directly investigated. Therefore, this review will describe the roles of CS and Cx in the development of PH and discuss the related downstream pathways. We also discuss the possible role of Cx in CS-induced PH. It is hoped that this review may provide new perspectives for early intervention.
Collapse
Affiliation(s)
- Xiaojiang Qin
- School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China.
- China Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China.
| | - Anqi Gao
- School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China
| | - Xiaomin Hou
- Department of Pharmacology, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China
- China Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China
| | - Xinrong Xu
- School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China
| | - Liangjin Chen
- School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China
| | - Lin Sun
- School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China
| | - Yuxuan Hao
- School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China
| | - Yiwei Shi
- Department of Respiratory and Critical Care Medicine, Shanxi Medical University Affiliated First Hospital, 85 Jiefang South Road, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
3
|
Friebe A, Englert N. NO-sensitive guanylyl cyclase in the lung. Br J Pharmacol 2020; 179:2328-2343. [PMID: 33332689 DOI: 10.1111/bph.15345] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 12/21/2022] Open
Abstract
In the late 1960s, several labatories identified guanylyl cyclase (GC) as the cGMP-producing enzyme. Subsequently, two different types of GC were described that differed in their cellular localization. Primarily found in the cytosol, nitric oxide (NO)-sensitive guanylyl cyclase (NO-GC) acts as receptor for the signalling molecule NO, in contrast the membrane-bound isoenzyme is activated by natriuretic peptides. The lung compared with other tissues exhibits the highest expression of NO-GC. The enzyme has been purified from lung for biochemical analysis. Although expressed in smooth muscle cells (SMCs) and in pericytes, the function of NO-GC in lung, especially in pericytes, is still not fully elucidated. However, pharmacological compounds that target NO-GC are available and have been implemented for the therapy of pulmonary arterial hypertension. In addition, NO-GC has been suggested as drug target for the therapy of asthma, acute respiratory distress syndrome and pulmonary fibrosis.
Collapse
Affiliation(s)
- Andreas Friebe
- Physiological Institute, Julius Maximilian University of Würzburg, Würzburg, Germany
| | - Nils Englert
- Physiological Institute, Julius Maximilian University of Würzburg, Würzburg, Germany
| |
Collapse
|
4
|
Donato M, Ferri N, Lupo MG, Faggin E, Rattazzi M. Current Evidence and Future Perspectives on Pharmacological Treatment of Calcific Aortic Valve Stenosis. Int J Mol Sci 2020; 21:ijms21218263. [PMID: 33158204 PMCID: PMC7663524 DOI: 10.3390/ijms21218263] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 10/31/2020] [Accepted: 11/02/2020] [Indexed: 02/07/2023] Open
Abstract
Calcific aortic valve stenosis (CAVS), the most common heart valve disease, is characterized by the slow progressive fibro-calcific remodeling of the valve leaflets, leading to progressive obstruction to the blood flow. CAVS is an increasing health care burden and the development of an effective medical treatment is a major medical need. To date, no effective pharmacological therapies have proven to halt or delay its progression to the severe symptomatic stage and aortic valve replacement represents the only available option to improve clinical outcomes and to increase survival. In the present report, the current knowledge and latest advances in the medical management of patients with CAVS are summarized, placing emphasis on lipid-lowering agents, vasoactive drugs, and anti-calcific treatments. In addition, novel potential therapeutic targets recently identified and currently under investigation are reported.
Collapse
Affiliation(s)
- Maristella Donato
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padova, Italy; (M.D.); (N.F.); (M.G.L.)
| | - Nicola Ferri
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padova, Italy; (M.D.); (N.F.); (M.G.L.)
| | - Maria Giovanna Lupo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padova, Italy; (M.D.); (N.F.); (M.G.L.)
| | - Elisabetta Faggin
- Department of Medicine—DIMED, University of Padova, 35122 Padova, Italy;
| | - Marcello Rattazzi
- Department of Medicine—DIMED, University of Padova, 35122 Padova, Italy;
- Correspondence: ; Tel.: +39-0498-211-867 or +39-0422-322-207
| |
Collapse
|
5
|
IRAG1 Deficient Mice Develop PKG1β Dependent Pulmonary Hypertension. Cells 2020; 9:cells9102280. [PMID: 33066124 PMCID: PMC7601978 DOI: 10.3390/cells9102280] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 10/09/2020] [Accepted: 10/11/2020] [Indexed: 02/07/2023] Open
Abstract
PKGs are serine/threonine kinases. PKG1 has two isoforms-PKG1α and β. Inositol trisphosphate receptor (IP3R)-associated cGMP-kinase substrate 1 (IRAG1) is a substrate for PKG1β. IRAG1 is also known to further interact with IP3RI, which mediates intracellular Ca2+ release. However, the role of IRAG1 in PH is not known. Herein, WT and IRAG1 KO mice were kept under normoxic or hypoxic (10% O2) conditions for five weeks. Animals were evaluated for echocardiographic variables and went through right heart catheterization. Animals were further sacrificed to prepare lungs and right ventricular (RV) for immunostaining, western blotting, and pulmonary artery smooth muscle cell (PASMC) isolation. IRAG1 is expressed in PASMCs and downregulated under hypoxic conditions. Genetic deletion of IRAG1 leads to RV hypertrophy, increase in RV systolic pressure, and RV dysfunction in mice. Absence of IRAG1 in lung and RV have direct impacts on PKG1β expression. Attenuated PKG1β expression in IRAG1 KO mice further dysregulates other downstream candidates of PKG1β in RV. IRAG1 KO mice develop PH spontaneously. Our results indicate that PKG1β signaling via IRAG1 is essential for the homeostasis of PASMCs and RV. Disturbing this signaling complex by deleting IRAG1 can lead to RV dysfunction and development of PH in mice.
Collapse
|
6
|
Yarsagumba is a Promising Therapeutic Option for Treatment of Pulmonary Hypertension due to the Potent Anti-Proliferative and Vasorelaxant Properties. ACTA ACUST UNITED AC 2020; 56:medicina56030131. [PMID: 32188043 PMCID: PMC7142425 DOI: 10.3390/medicina56030131] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 03/12/2020] [Accepted: 03/13/2020] [Indexed: 02/04/2023]
Abstract
Background and objectives: Pulmonary hypertension (PH) is characterized by the vasoconstriction and abnormally proliferative vascular cells. The available allopathic treatment options for PH are still not able to cure the disease. Alternative medicine is becoming popular and drawing the attention of the general public and scientific communities. The entomogenous fungus Yarsagumba (Cordyceps sinensis) and its biologically active ingredient cordycepin may represent the therapeutic option for this incurable disease, owing to their anti-inflammatory, vasodilatory and anti-oxidative effects. Methods: In this study, we investigated whether Yarsagumba extract and cordycepin possess anti-proliferative and vasorelaxant properties in the context of PH, using 5-bromo-2’-deoxyuridine assay and isolated mice lungs, respectively. Results: Our results revealed that Yarsagumba extract and its bioactive compound cordycepin significantly attenuated the proliferation of human pulmonary artery smooth muscle cells derived from donor and PH subjects. In isolated murine lungs, only Yarsagumba extract, but not cordycepin, resulted in vasodilatation, indicating the probable existence of other bioactive metabolites present in Yarsagumba that may be responsible for this outcome. Conclusion: Future comprehensive in vivo and in vitro research is crucially needed to discover the profound mechanistic insights with regard to this promising therapeutic potency of Yarsagumba extract and to provide further evidence as to whether it can be used as a strategy for the treatment of PH.
Collapse
|
7
|
de Wijs‐Meijler DPM, Duncker DJ, Danser AHJ, Reiss IKM, Merkus D. Changes in the nitric oxide pathway of the pulmonary vasculature after exposure to hypoxia in swine model of neonatal pulmonary vascular disease. Physiol Rep 2018; 6:e13889. [PMID: 30375198 PMCID: PMC6205946 DOI: 10.14814/phy2.13889] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 09/12/2018] [Accepted: 09/16/2018] [Indexed: 11/24/2022] Open
Abstract
Neonatal pulmonary vascular disease (PVD) is increasingly recognized as a disease that complicates the cardiopulmonary adaptations after birth and predisposes to long-term cardiopulmonary disease. There is growing evidence that PVD is associated with disruptions in the nitric oxide (NO)-cGMP-phosphodiesterase 5 (PDE5) pathway. Examination of the functionality of different parts of this pathway is required for better understanding of the pathogenesis of neonatal PVD. For this purpose, the role of the NO-cGMP-PDE5 pathway in regulation of pulmonary vascular function was investigated in vivo, both at rest and during exercise, and in isolated pulmonary small arteries in vitro, in a neonatal swine model with hypoxia-induced PVD. Endothelium-dependent vasodilatation was impaired in piglets with hypoxia-induced PVD both in vivo at rest and in vitro. Moreover, the responsiveness to the NO-donor SNP was reduced in hypoxia-exposed piglets in vivo, while the relaxation to SNP and 8-bromo-cyclicGMP in vitro were unaltered. Finally, PDE5 inhibition-induced pulmonary vasodilatation was impaired in hypoxia-exposed piglets both in vitro and in vivo at rest. During exercise, however, the pulmonary vasodilator effect of PDE5 inhibition was significantly larger in hypoxia-exposed as compared to normoxia-exposed piglets. In conclusion, the impaired endothelium-dependent vasodilatation in piglets with hypoxia-induced PVD was accompanied by reduced responsiveness to NO, potentially caused by altered sensitivity and/or activity of soluble guanylyl cyclase (sGC), resulting in an impaired cGMP production. Our findings in a newborn animal model for neonatal PVD suggests that sGC stimulators/activators may be a novel treatment strategy to alleviate neonatal PVD.
Collapse
Affiliation(s)
- Daphne P. M. de Wijs‐Meijler
- Division of Experimental CardiologyDepartment of CardiologyUniversity Medical Center RotterdamErasmus MCRotterdamThe Netherlands
- Division of NeonatologyDepartment of PediatricsSophia Children's HospitalErasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Dirk J. Duncker
- Division of Experimental CardiologyDepartment of CardiologyUniversity Medical Center RotterdamErasmus MCRotterdamThe Netherlands
| | - A. H. Jan Danser
- Division of PharmacologyDepartment of Internal MedicineErasmus MC University Medical Center RotterdamRotterdamThe Netherlands
| | - Irwin K. M. Reiss
- Division of NeonatologyDepartment of PediatricsSophia Children's HospitalErasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Daphne Merkus
- Division of Experimental CardiologyDepartment of CardiologyUniversity Medical Center RotterdamErasmus MCRotterdamThe Netherlands
| |
Collapse
|
8
|
Amirjanians M, Egemnazarov B, Sydykov A, Kojonazarov B, Brandes R, Luitel H, Pradhan K, Stasch JP, Redlich G, Weissmann N, Grimminger F, Seeger W, Ghofrani H, Schermuly R. Chronic intratracheal application of the soluble guanylyl cyclase stimulator BAY 41-8543 ameliorates experimental pulmonary hypertension. Oncotarget 2018; 8:29613-29624. [PMID: 28410199 PMCID: PMC5444690 DOI: 10.18632/oncotarget.16769] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 03/08/2017] [Indexed: 12/26/2022] Open
Abstract
Dysfunction of the NO/sGC/cGMP signaling pathway has been implicated in the pathogenesis of pulmonary hypertension (PH). Therefore, agents stimulating cGMP synthesis via sGC are important therapeutic options for treatment of PH patients. An unwanted effect of this novel class of drugs is their systemic hypotensive effect. We tested the hypothesis that aerosolized intra-tracheal delivery of the sGC stimulator BAY41-8543 could diminish its systemic vasodilating effect.Pharmacodynamics and -kinetics of BAY41-8543 after single intra-tracheal delivery was tested in healthy rats. Four weeks after a single injection of monocrotaline (MCT, 60 mg/kg s.c.), rats were randomized to a two-week treatment with either placebo, BAY 41-8543 (10 mg/kg per os (PO)) or intra-tracheal (IT) instillation (3 mg/kg or 1 mg/kg).Circulating concentrations of the drug 10 mg/kg PO and 3 mg/kg IT were comparable. BAY 41-8543 was detected in the lung tissue and broncho-alveolar fluid after IT delivery at higher concentrations than after PO administration. Systemic arterial pressure transiently decreased after oral BAY 41-8543 and was unaffected by intratracheal instillation of the drug. PO 10 mg/kg and IT 3 mg/kg regimens partially reversed pulmonary hypertension and improved heart function in MCT-injected rats. Minor efficacy was noted in rats treated IT with 1 mg/kg. The degree of pulmonary vascular remodeling was largely reversed in all treatment groups.Intratracheal administration of BAY 41-8543 reverses PAH and vascular structural remodeling in MCT-treated rats. Local lung delivery is not associated with systemic blood pressure lowering and represents thus a further development of PH treatment with sGC stimulators.
Collapse
Affiliation(s)
| | | | | | | | - Ralf Brandes
- Institute for Cardiovascular Physiology, J.W. Goethe University, Frankfurt, Germany
| | - Himal Luitel
- University of Giessen Lung Center, Giessen, Germany
| | | | - Johannes-Peter Stasch
- Cardiology Research, Pharmaceuticals, Bayer AG, Wuppertal, Germany.,Institute of Pharmacy, Martin Luther University of Halle Wittenberg, Halle, Germany
| | - Gorden Redlich
- Research Pharmacokinetics, Pharmaceuticals, Bayer AG, Wuppertal, Germany
| | | | | | - Werner Seeger
- University of Giessen Lung Center, Giessen, Germany.,Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | | | - Ralph Schermuly
- University of Giessen Lung Center, Giessen, Germany.,Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| |
Collapse
|
9
|
Rain M, Chaudhary H, Kukreti R, Thinlas T, Mohammad G, Pasha Q. Elevated Vasodilatory Cyclases and Shorter Telomere Length Contribute to High-Altitude Pulmonary Edema. High Alt Med Biol 2018; 19:60-68. [PMID: 29443612 DOI: 10.1089/ham.2017.0136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Rain, Manjari, Himanshi Chaudhary, Ritushree Kukreti, Tashi Thinlas, Ghulam Mohammad, and Qadar Pasha. Elevated vasodilatory cyclases and shorter telomere length contribute to high-altitude pulmonary edema. High Alt Med Biol. 19:60-68, 2018. AIM High-altitude (HA) genetics is complex with respect to health and disease (HA pulmonary edema i.e., HAPE). Based on the widely recognized fact that oxidative stress is a major trigger of several physiological processes, this study was designed to establish the significance of vasodilatory cyclases and telomere length in HA physiology. The study was performed in three groups, namely HAPE-free sojourners (HAPE-f, n = 150), HAPE patients (HAPE-p, n = 150), and healthy highland natives or highlanders (HLs, n = 150). Variations in soluble guanylyl cyclase β1-subunit (GUCY1B3) and adenylyl cyclase type 6 (ADCY6) were genotyped by the SNaPshot method and/or Fluidigm SNP type genotyping. Plasma GUCY1B3 and ADCY6 levels were estimated using ELISA, and relative telomere length was estimated by qRT-PCR. RESULTS The rs7638AA genotype was over-represented in HLs compared with HAPE-f and HAPE-p (p = 0.035 and p = 0.012, respectively). Similarly, the rs7638A allele was prevalent in HLs compared with both groups, but significance was attained against HAPE-p (p = 0.012). Significantly elevated plasma levels of GUCY1B3 and ADCY6 were obtained in HAPE-p compared with HAPE-f (p = 0.001 and p = 0.006, respectively) and HLs (p = 3.31E-05 and p = 0.05, respectively). Shorter telomere length was observed in HAPE-p compared with HAPE-f (p > 0.05) and HLs (p = 0.017). CONCLUSION Elevated cyclases and shorter telomere length associate with HAPE pathophysiology.
Collapse
Affiliation(s)
- Manjari Rain
- 1 CSIR-Institute of Genomics and Integrative Biology , Delhi, India .,2 Academy of Scientific and Innovative Research , New Delhi, India
| | | | - Ritushree Kukreti
- 1 CSIR-Institute of Genomics and Integrative Biology , Delhi, India .,2 Academy of Scientific and Innovative Research , New Delhi, India
| | - Tashi Thinlas
- 3 Department of Medicine, Sonam Norboo Memorial Hospital , Leh, Ladakh, Jammu and Kashmir, India
| | - Ghulam Mohammad
- 3 Department of Medicine, Sonam Norboo Memorial Hospital , Leh, Ladakh, Jammu and Kashmir, India
| | - Qadar Pasha
- 1 CSIR-Institute of Genomics and Integrative Biology , Delhi, India .,4 Indian Council of Medical Research , New Delhi, India
| |
Collapse
|
10
|
Thin Air Resulting in High Pressure: Mountain Sickness and Hypoxia-Induced Pulmonary Hypertension. Can Respir J 2017; 2017:8381653. [PMID: 28522921 PMCID: PMC5385916 DOI: 10.1155/2017/8381653] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 02/15/2017] [Accepted: 02/28/2017] [Indexed: 12/31/2022] Open
Abstract
With rising altitude the partial pressure of oxygen falls. This phenomenon leads to hypobaric hypoxia at high altitude. Since more than 140 million people permanently live at heights above 2500 m and more than 35 million travel to these heights each year, understanding the mechanisms resulting in acute or chronic maladaptation of the human body to these circumstances is crucial. This review summarizes current knowledge of the body's acute response to these circumstances, possible complications and their treatment, and health care issues resulting from long-term exposure to high altitude. It furthermore describes the characteristic mechanisms of adaptation to life in hypobaric hypoxia expressed by the three major ethnic groups permanently dwelling at high altitude. We additionally summarize current knowledge regarding possible treatment options for hypoxia-induced pulmonary hypertension by reviewing in vitro, rodent, and human studies in this area of research.
Collapse
|
11
|
Jernigan NL, Resta TC, Gonzalez Bosc LV. Altered Redox Balance in the Development of Chronic Hypoxia-induced Pulmonary Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 967:83-103. [PMID: 29047083 DOI: 10.1007/978-3-319-63245-2_7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Normally, the pulmonary circulation is maintained in a low-pressure, low-resistance state with little resting tone. Pulmonary arteries are thin-walled and rely heavily on pulmonary arterial distension and recruitment for reducing pulmonary vascular resistance when cardiac output is elevated. Under pathophysiological conditions, however, active vasoconstriction and vascular remodeling lead to enhanced pulmonary vascular resistance and subsequent pulmonary hypertension (PH). Chronic hypoxia is a critical pathological factor associated with the development of PH resulting from airway obstruction (COPD, sleep apnea), diffusion impairment (interstitial lung disease), developmental lung abnormalities, or high altitude exposure (World Health Organization [WHO]; Group III). The rise in pulmonary vascular resistance increases right heart afterload causing right ventricular hypertrophy that can ultimately lead to right heart failure in patients with chronic lung disease. PH is typically characterized by diminished paracrine release of vasodilators, antimitogenic factors, and antithrombotic factors (e.g., nitric oxide and protacyclin) and enhanced production of vasoconstrictors and mitogenic factors (e.g., reactive oxygen species and endothelin-1) from the endothelium and lung parenchyma. In addition, phenotypic changes to pulmonary arterial smooth muscle cells (PASMC), including alterations in Ca2+ homeostasis, Ca2+ sensitivity, and activation of transcription factors are thought to play prominent roles in the development of both vasoconstrictor and arterial remodeling components of hypoxia-associated PH. These changes in PASMC function are briefly reviewed in Sect. 1 and the influence of altered reactive oxygen species homeostasis on PASMC function discussed in Sects. 2-4.
Collapse
Affiliation(s)
- Nikki L Jernigan
- Department Cell Biology and Physiology, Vascular Physiology Group, University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Thomas C Resta
- Department Cell Biology and Physiology, Vascular Physiology Group, University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Laura V Gonzalez Bosc
- Department Cell Biology and Physiology, Vascular Physiology Group, University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, NM, 87131, USA.
| |
Collapse
|
12
|
Brockhoff B, Schreckenberg R, Forst S, Heger J, Bencsik P, Kiss K, Ferdinandy P, Schulz R, Schlüter K. Effect of nitric oxide deficiency on the pulmonary PTHrP system. J Cell Mol Med 2017; 21:96-106. [PMID: 27581501 PMCID: PMC5192877 DOI: 10.1111/jcmm.12942] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 07/04/2016] [Indexed: 01/20/2023] Open
Abstract
Nitric oxide (NO) deficiency is common in pulmonary diseases, but its effect on pulmonary remodelling is still controversial. As pulmonary parathyroid hormone-related protein (PTHrP) expression is a key regulator of pulmonary fibrosis and development, the effect of chronic NO deficiency on the pulmonary PTHrP system and its relationship with oxidative stress was addressed. NO bioavailability in adult rats was reduced by systemic administration of L-NAME via tap water. To clarify the role of NO synthase (NOS)-3-derived NO on pulmonary expression of PTHrP, NOS-3-deficient mice were used. Captopril and hydralazine were used to reduce the hypertensive effect of L-NAME treatment and to interfere with the pulmonary renin-angiotensin system (RAS). Quantitative RT-PCR and immunoblot techniques were used to characterize the expression of key proteins involved in pulmonary remodelling. L-NAME administration significantly reduced pulmonary NO concentration and caused oxidative stress as characterized by increased pulmonary nitrite concentration and increased expression of NOX2, p47phox and p67phox. Furthermore, L-NAME induced the pulmonary expression of PTHrP and of its corresponding receptor, PTH-1R. Expression of PTHrP and PTH-1R correlated with the expression of two well-established PTHrP downstream targets, ADRP and PPARγ, suggesting an activation of the pulmonary PTHrP system by NO deficiency. Captopril reduced the expression of PTHrP, profibrotic markers and ornithine decarboxylase, but neither that of PTH-1R nor that of ADRP and PPARγ. All transcriptional changes were confirmed in NOS-3-deficient mice. In conclusion, NOS-3-derived NO suppresses pulmonary PTHrP and PTH-1R expression, thereby modifying pulmonary remodelling.
Collapse
Affiliation(s)
- Bastian Brockhoff
- Physiologisches InstitutJustus‐Liebig‐Universität GießenGießenGermany
| | | | - Svenja Forst
- Physiologisches InstitutJustus‐Liebig‐Universität GießenGießenGermany
| | - Jacqueline Heger
- Physiologisches InstitutJustus‐Liebig‐Universität GießenGießenGermany
| | - Péter Bencsik
- Pharmahungary GroupSzegedHungary
- Cardiovascular Research GroupDepartment of BiochemistryUniversity of SzegedSzegedHungary
| | - Krisztina Kiss
- Pharmahungary GroupSzegedHungary
- Cardiovascular Research GroupDepartment of BiochemistryUniversity of SzegedSzegedHungary
| | - Peter Ferdinandy
- Pharmahungary GroupSzegedHungary
- Department of Pharmacology and PharmacotherapySemmelweis UniversityBudapestHungary
| | - Rainer Schulz
- Physiologisches InstitutJustus‐Liebig‐Universität GießenGießenGermany
| | | |
Collapse
|
13
|
Baldissera L, Squebola-Cola DM, Calixto MC, Lima-Barbosa AP, Rennó AL, Anhê GF, Condino-Neto A, De Nucci G, Antunes E. The soluble guanylyl cyclase activator BAY 60-2770 inhibits murine allergic airways inflammation and human eosinophil chemotaxis. Pulm Pharmacol Ther 2016; 41:86-95. [PMID: 27816773 DOI: 10.1016/j.pupt.2016.11.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 09/21/2016] [Accepted: 11/01/2016] [Indexed: 02/08/2023]
Abstract
OBJECTIVES Activators of soluble guanylyl cyclase (sGC) act preferentially in conditions of enzyme oxidation or haem group removal. This study was designed to investigate the effects of the sGC activator BAY 60-2770 in murine airways inflammation and human eosinophil chemotaxis. METHODS C57Bl/6 mice treated or not with BAY 60-2770 (1 mg/kg/day, 14 days) were intranasally challenged with ovalbumin (OVA). At 48 h, bronchoalveolar lavage fluid (BALF) was performed, and circulating blood, bone marrow and lungs were obtained. Human eosinophils purified from peripheral blood were used to evaluate the cell chemotaxis. RESULTS OVA-challenge promoted marked increases in eosinophil number in BAL, lung tissue, circulating blood and bone marrow, all of which were significantly reduced by BAY 60-2770. The IL-4 and IL-5 levels in BALF were significantly reduced by BAY 60-2770. Increased protein expression of iNOS, along with decreases of expression of sGC (α1 and β1 subunits) and cGMP levels were detected in lung tissue of OVA-challenged mice. BAY 60-2770 fully restored to baseline the iNOS and sGC subunit expressions, and cGMP levels. In human isolated eosinophils, BAY 60-2770 (1-5 μM) had no effects on the cGMP levels and eotaxin-induced chemotaxis; however, prior incubation with ODQ (10 μM) markedly elevated the BAY 60-2770-induced cyclic GMP production, further inhibiting the eosinophil chemotaxis. CONCLUSIONS BAY 60-2770 reduces airway eosinophilic inflammation and rescue the sGC levels. In human eosinophils under oxidized conditions, BAY 60-2770 elevates the cGMP levels causing cell chemotaxis inhibition. BAY 60-2770 may reveal a novel therapeutic target for asthma treatment.
Collapse
Affiliation(s)
- Lineu Baldissera
- Department of Pharmacology, Faculty of Medical Science, State University of Campinas (UNICAMP), Campinas, 13084-971, SP, Brazil
| | - Dalize M Squebola-Cola
- Department of Pharmacology, Faculty of Medical Science, State University of Campinas (UNICAMP), Campinas, 13084-971, SP, Brazil
| | - Marina C Calixto
- Department of Pharmacology, Faculty of Medical Science, State University of Campinas (UNICAMP), Campinas, 13084-971, SP, Brazil
| | - Ana P Lima-Barbosa
- Department of Pharmacology, Faculty of Medical Science, State University of Campinas (UNICAMP), Campinas, 13084-971, SP, Brazil
| | - André L Rennó
- Department of Pharmacology, Faculty of Medical Science, State University of Campinas (UNICAMP), Campinas, 13084-971, SP, Brazil
| | - Gabriel F Anhê
- Department of Pharmacology, Faculty of Medical Science, State University of Campinas (UNICAMP), Campinas, 13084-971, SP, Brazil
| | - Antonio Condino-Neto
- Department of Pharmacology, Faculty of Medical Science, State University of Campinas (UNICAMP), Campinas, 13084-971, SP, Brazil
| | - Gilberto De Nucci
- Department of Pharmacology, Faculty of Medical Science, State University of Campinas (UNICAMP), Campinas, 13084-971, SP, Brazil
| | - Edson Antunes
- Department of Pharmacology, Faculty of Medical Science, State University of Campinas (UNICAMP), Campinas, 13084-971, SP, Brazil.
| |
Collapse
|
14
|
Papapetropoulos A, Hobbs AJ, Topouzis S. Extending the translational potential of targeting NO/cGMP-regulated pathways in the CVS. Br J Pharmacol 2015; 172:1397-414. [PMID: 25302549 DOI: 10.1111/bph.12980] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 09/08/2014] [Accepted: 10/05/2014] [Indexed: 02/06/2023] Open
Abstract
The discovery of NO as both an endogenous signalling molecule and as a mediator of the cardiovascular effects of organic nitrates was acknowledged in 1998 by the Nobel Prize in Physiology/Medicine. The characterization of its downstream signalling, mediated through stimulation of soluble GC (sGC) and cGMP generation, initiated significant translational interest, but until recently this was almost exclusively embodied by the use of PDE5 inhibitors in erectile dysfunction. Since then, research progress in two areas has contributed to an impressive expansion of the therapeutic targeting of the NO-sGC-cGMP axis: first, an increased understanding of the molecular events operating within this complex pathway and second, a better insight into its dys-regulation and uncoupling in human disease. Already-approved PDE5 inhibitors and novel, first-in-class molecules, which up-regulate the activity of sGC independently of NO and/or of the enzyme's haem prosthetic group, are undergoing clinical evaluation to treat pulmonary hypertension and myocardial failure. These molecules, as well as combinations or second-generation compounds, are also being assessed in additional experimental disease models and in patients in a wide spectrum of novel indications, such as endotoxic shock, diabetic cardiomyopathy and Becker's muscular dystrophy. There is well-founded optimism that the modulation of the NO-sGC-cGMP pathway will sustain the development of an increasing number of successful clinical candidates for years to come.
Collapse
|
15
|
Dasgupta A, Bowman L, D'Arsigny CL, Archer SL. Soluble guanylate cyclase: a new therapeutic target for pulmonary arterial hypertension and chronic thromboembolic pulmonary hypertension. Clin Pharmacol Ther 2014; 97:88-102. [PMID: 25670386 DOI: 10.1002/cpt.10] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 10/03/2014] [Indexed: 01/08/2023]
Abstract
Nitric oxide (NO) activates soluble guanylate cyclase (sGC) by binding its prosthetic heme group, thereby catalyzing cyclic guanosine monophosphate (cGMP) synthesis. cGMP causes vasodilation and may inhibit smooth muscle cell proliferation and platelet aggregation. The NO-sGC-cGMP pathway is disordered in pulmonary arterial hypertension (PAH), a syndrome in which pulmonary vascular obstruction, inflammation, thrombosis, and constriction ultimately lead to death from right heart failure. Expression of sGC is increased in PAH but its function is reduced by decreased NO bioavailability, sGC oxidation and the related loss of sGC's heme group. Two classes of sGC modulators offer promise in PAH. sGC stimulators (e.g., riociguat) require heme-containing sGC to catalyze cGMP production, whereas sGC activators (e.g., cinaciguat) activate heme-free sGC. Riociguat is approved for PAH and yields functional and hemodynamic benefits similar to other therapies. Its main serious adverse effect is dose-dependent hypotension. Riociguat is also approved for inoperable chronic thromboembolic pulmonary hypertension.
Collapse
Affiliation(s)
- A Dasgupta
- Department of Medicine, Queen's University, Etherington Hall, Kingston, Ontario, Canada
| | | | | | | |
Collapse
|
16
|
Silva FH, Leiria LO, Alexandre EC, Davel APC, Mónica FZ, De Nucci G, Antunes E. Prolonged therapy with the soluble guanylyl cyclase activator BAY 60-2770 restores the erectile function in obese mice. J Sex Med 2014; 11:2661-70. [PMID: 25196910 DOI: 10.1111/jsm.12682] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
INTRODUCTION Cardiovascular and endocrine-metabolic diseases associated with increased oxidative stress such as obesity lead to erectile dysfunction (ED). Activators of soluble guanylyl cyclase (sGC) such as BAY 60-2770 reactivate the heme-oxidized sGC in vascular diseases. AIM This study aimed to evaluate the effects of 2-week oral intake with BAY 60-2270 on a murine model of obesity-associated ED. METHODS C57BL/6 male mice were fed for 12 weeks with standard chow or high-fat diet. Lean and obese mice were treated with BAY 60-2770 (1 mg/kg/day, 2 weeks). MAIN OUTCOME MEASURES Measurements of intracavernosal pressure (ICP), along with acetylcholine (10(-9) to 10(-5) M) and electrical field stimulation (EFS; 4-10 Hz)-induced corpus cavernosum relaxations in vitro, were obtained. Levels of cyclic guanosine monophosphate (cGMP), reactive oxygen species (ROS), and sGC protein expressions in cavernosal tissues were measured. RESULTS Cavernous nerve stimulation caused frequency-dependent ICP increases, which were significantly lower in obese compared with lean mice (P < 0.05). Two-week therapy with BAY 60-2770 fully reversed the decreased ICP in obese group. Acetylcholine-induced cavernosal relaxations were 45% lower (P < 0.001) in obese mice, which were fully restored by BAY 60-2770 treatment. Likewise, the EFS-induced relaxations in obese mice were restored by BAY 60-2770. Basal cGMP content in erectile tissue was 68% lower (P < 0.05) in obese mice, an effect normalized by BAY 60-2770. Levels of ROS were 52% higher (P < 0.05) whereas protein expression of α1 sGC subunit was reduced in cavernosal tissue of obese mice, both of which were normalized by BAY 60-2770. In lean group, BAY 60-2770 did not significantly affect any functional, biochemical, or molecular parameter analyzed. CONCLUSIONS Two-week therapy with BAY 60-2770 restores the erectile function in obese mice that is associated with reduced ROS levels, up-regulation of α1 sGC subunit, and increased cGMP levels in the erectile tissue.
Collapse
Affiliation(s)
- Fábio H Silva
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas, Campinas, Brazil
| | | | | | | | | | | | | |
Collapse
|
17
|
Weissmann N, Lobo B, Pichl A, Parajuli N, Seimetz M, Puig-Pey R, Ferrer E, Peinado VI, Domínguez-Fandos D, Fysikopoulos A, Stasch JP, Ghofrani HA, Coll-Bonfill N, Frey R, Schermuly RT, García-Lucio J, Blanco I, Bednorz M, Tura-Ceide O, Tadele E, Brandes RP, Grimminger J, Klepetko W, Jaksch P, Rodriguez-Roisin R, Seeger W, Grimminger F, Barberà JA. Stimulation of soluble guanylate cyclase prevents cigarette smoke-induced pulmonary hypertension and emphysema. Am J Respir Crit Care Med 2014; 189:1359-73. [PMID: 24738736 DOI: 10.1164/rccm.201311-2037oc] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
RATIONALE Chronic obstructive pulmonary disease (COPD) is a major cause of death worldwide. No therapy stopping progress of the disease is available. OBJECTIVES To investigate the role of the soluble guanylate cyclase (sGC)-cGMP axis in development of lung emphysema and pulmonary hypertension (PH) and to test whether the sGC-cGMP axis is a treatment target for these conditions. METHODS Investigations were performed in human lung tissue from patients with COPD, healthy donors, mice, and guinea pigs. Mice were exposed to cigarette smoke (CS) for 6 hours per day, 5 days per week for up to 6 months and treated with BAY 63-2521. Guinea pigs were exposed to CS from six cigarettes per day for 3 months, 5 days per week and treated with BAY 41-2272. Both BAY compounds are sGC stimulators. Gene and protein expression analysis were performed by quantitative real-time polymerase chain reaction and Western blotting. Lung compliance, hemodynamics, right ventricular heart mass alterations, and alveolar and vascular morphometry were performed, as well as inflammatory cell infiltrate assessment. In vitro assays of cell adhesion, proliferation, and apoptosis have been done. MEASUREMENTS AND MAIN RESULTS The functionally essential sGC β1-subunit was down-regulated in patients with COPD and in CS-exposed mice. sGC stimulators prevented the development of PH and emphysema in the two different CS-exposed animal models. sGC stimulation prevented peroxynitrite-induced apoptosis of alveolar and endothelial cells, reduced CS-induced inflammatory cell infiltrate in lung parenchyma, and inhibited adhesion of CS-stimulated neutrophils. CONCLUSIONS The sGC-cGMP axis is perturbed by chronic exposure to CS. Treatment of COPD animal models with sGC stimulators can prevent CS-induced PH and emphysema.
Collapse
Affiliation(s)
- Norbert Weissmann
- 1 Justus-Liebig University, Excellence Cluster Cardiopulmonary System, Universities of Giessen and Marburg Lung Center (UGMLC), DZL, Giessen, Marburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
Oxidative stress has many implications in the pathogenesis of lung diseases. In this review, we provide an overview of Reactive Oxygen Species (ROS) and nitrogen (RNS) species and antioxidants, how they relate to normal physiological function and the pathophysiology of different lung diseases, and therapeutic strategies. The production of ROS/RNS from endogenous and exogenous sources is first discussed, followed by antioxidant systems that restore oxidative balance and cellular homeostasis. The contribution of oxidant/antioxidant imbalance in lung disease pathogenesis is also discussed. An overview of therapeutic strategies is provided, such as augmenting NO bioactivity, blocking the production of ROS/RNS and replacement of deficient antioxidants. The limitations of current strategies and failures of clinical trials are then addressed, followed by discussion of novel experimental approaches for the development of improved antioxidant therapies.
Collapse
|
19
|
The Soluble Guanylyl Cyclase Activator BAY 60-2770 Ameliorates Overactive Bladder in Obese Mice. J Urol 2014; 191:539-47. [DOI: 10.1016/j.juro.2013.09.020] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2013] [Indexed: 11/20/2022]
|
20
|
Schwappacher R, Kilic A, Kojonazarov B, Lang M, Diep T, Zhuang S, Gawlowski T, Schermuly RT, Pfeifer A, Boss GR, Pilz RB. A molecular mechanism for therapeutic effects of cGMP-elevating agents in pulmonary arterial hypertension. J Biol Chem 2013; 288:16557-16566. [PMID: 23612967 DOI: 10.1074/jbc.m113.458729] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive, usually fatal disease with abnormal vascular remodeling. Pulmonary artery smooth muscle cells (PASMCs) from PAH patients are hyperproliferative and apoptosis-resistant and demonstrate decreased signaling in response to bone morphogenetic proteins (BMPs). Cyclic GMP-elevating agents are beneficial in PAH, but their mechanism(s) of action are incompletely understood. Here we show that BMP signaling via Smad1/5/8 requires cGMP-dependent protein kinase isotype I (PKGI) to maintain PASMCs in a differentiated, low proliferative state. BMP cooperation with cGMP/PKGI was crucial for transcription of contractile genes and suppression of pro-proliferative and anti-apoptotic genes. Lungs from mice with low or absent PKGI (Prkg1(+/-) and Prkg1(-/-) mice) exhibited impaired BMP signaling, decreased contractile gene expression, and abnormal vascular remodeling. Conversely, cGMP stimulation of PKGI restored defective BMP signaling in rats with hypoxia-induced PAH, consistent with cGMP-elevating agents reversing vascular remodeling in this PAH model. Our results provide a mechanism for the therapeutic effects of cGMP-elevating agents in PAH and suggest that combining them with BMP mimetics may provide a novel, disease-modifying approach to PAH therapy.
Collapse
Affiliation(s)
- Raphaela Schwappacher
- Department of Medicine, University of California San Diego, La Jolla, California 92093.
| | - Ana Kilic
- Institute for Pharmacology and Toxicology, University of Bonn, 53113 Bonn, Germany
| | | | - Michaela Lang
- University of Giessen and Marburg Lung Center, 35392 Giessen, Germany
| | - Thuan Diep
- Department of Medicine, University of California San Diego, La Jolla, California 92093
| | - Shunhui Zhuang
- Department of Medicine, University of California San Diego, La Jolla, California 92093
| | - Thomas Gawlowski
- Department of Medicine, University of California San Diego, La Jolla, California 92093
| | - Ralph T Schermuly
- University of Giessen and Marburg Lung Center, 35392 Giessen, Germany
| | - Alexander Pfeifer
- Institute for Pharmacology and Toxicology, University of Bonn, 53113 Bonn, Germany
| | - Gerry R Boss
- Department of Medicine, University of California San Diego, La Jolla, California 92093
| | - Renate B Pilz
- Department of Medicine, University of California San Diego, La Jolla, California 92093
| |
Collapse
|
21
|
Ronisz A, Delcroix M, Quarck R. Measurement of right ventricular pressure by telemetry in conscious moving rabbits. Lab Anim 2013; 47:175-183. [PMID: 23579323 DOI: 10.1177/0023677213483725] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Implantable radiotelemetry methodology has been used to continuously monitor pulmonary hemodynamics including right ventricular pressure (RVP) or pulmonary arterial pressure (PAP) in conscious, untethered and freely moving animals such as mice and rats. The use of implantable radiotelemetry to monitor RVP or PAP has never previously been described in rabbits. The aim of the present study was to use implantable radiotelemetry to continuously monitor RVP in conscious adult rabbits. Telemetry transmitters were implanted in 44 adult male New Zealand rabbits using a trans-diaphragm approach for the catheter placement. RVP, heart rate (HR) and activity were monitored every 15 min for 20 s. Body mass was recorded once a week. A total of 39 (88%) rabbits were successfully implanted. Thirty rabbits survived the surgical procedure resulting in an overall survival rate of 73%. RVP, HR and activity were long-term monitored in 17 rabbits for an average period of 103 ± 15 days. Weekly body mass follow-up showed that implantable radiotelemetry did not impair the normal development of the animal. Twenty-four-hour period monitoring of RVP, HR and activity showed concomitant changes in RVP, HR and activity according to the dark/light cycle applied to the rabbits. To conclude, implantable radiotelemetry methodology can be safely used to continuously monitor RVP in conscious rabbits.
Collapse
Affiliation(s)
- Alicja Ronisz
- Respiratory Division, Department of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium
| | | | | |
Collapse
|
22
|
Mendes-Silverio CB, Leiria LOS, Morganti RP, Anhê GF, Marcondes S, Mónica FZ, De Nucci G, Antunes E. Activation of haem-oxidized soluble guanylyl cyclase with BAY 60-2770 in human platelets lead to overstimulation of the cyclic GMP signaling pathway. PLoS One 2012; 7:e47223. [PMID: 23144808 PMCID: PMC3493568 DOI: 10.1371/journal.pone.0047223] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Accepted: 09/12/2012] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND AND AIMS Nitric oxide-independent soluble guanylyl cyclase (sGC) activators reactivate the haem-oxidized enzyme in vascular diseases. This study was undertaken to investigate the anti-platelet mechanisms of the haem-independent sGC activator BAY 60-2770 in human washed platelets. The hypothesis that sGC oxidation potentiates the anti-platelet activities of BAY 60-2770 has been tested. METHODS Human washed platelet aggregation and adhesion assays, as well as flow cytometry for α(IIb)β(3) integrin activation and Western blot for α1 and β1 sGC subunits were performed. Intracellular calcium levels were monitored in platelets loaded with a fluorogenic calcium-binding dye (FluoForte). RESULTS BAY 60-2770 (0.001-10 µM) produced significant inhibition of collagen (2 µg/ml)- and thrombin (0.1 U/ml)-induced platelet aggregation that was markedly potentiated by the sGC inhibitor ODQ (10 µM). In fibrinogen-coated plates, BAY 60-2770 significantly inhibited platelet adhesion, an effect potentiated by ODQ. BAY 60-2770 increased the cGMP levels and reduced the intracellular Ca(2+) levels, both of which were potentiated by ODQ. The cell-permeable cGMP analogue 8-Br-cGMP (100 µM) inhibited platelet aggregation and Ca(2+) levels in an ODQ-insensitive manner. The cAMP levels remained unchanged by BAY 60-2770. Collagen- and thrombin-induced α(IIb)β(3) activation was markedly inhibited by BAY 60-2770 that was further inhibited by ODQ. The effects of sodium nitroprusside (3 µM) were all prevented by ODQ. Incubation with ODQ (10 µM) significantly reduced the protein levels of α1 and β1 sGC subunits, which were prevented by BAY 60-2770. CONCLUSION The inhibitory effects of BAY 60-2770 on aggregation, adhesion, intracellular Ca(2+) levels and α(IIb)β(3) activation are all potentiated in haem-oxidizing conditions. BAY 60-2770 prevents ODQ-induced decrease in sGC protein levels. BAY 60-2770 could be of therapeutic interest in cardiovascular diseases associated with thrombotic complications.
Collapse
Affiliation(s)
- Camila B. Mendes-Silverio
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), São Paulo, Brazil
| | - Luiz O. S. Leiria
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), São Paulo, Brazil
| | - Rafael P. Morganti
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), São Paulo, Brazil
| | - Gabriel F. Anhê
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), São Paulo, Brazil
| | - Sisi Marcondes
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), São Paulo, Brazil
| | - Fabíola Z. Mónica
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), São Paulo, Brazil
| | - Gilberto De Nucci
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), São Paulo, Brazil
| | - Edson Antunes
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), São Paulo, Brazil
- * E-mail:
| |
Collapse
|
23
|
Lundgren J, Kylhammar D, Hedelin P, Rådegran G. sGC stimulation totally reverses hypoxia-induced pulmonary vasoconstriction alone and combined with dual endothelin-receptor blockade in a porcine model. Acta Physiol (Oxf) 2012; 206:178-94. [PMID: 22682645 DOI: 10.1111/j.1748-1716.2012.02445.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Revised: 12/21/2011] [Accepted: 04/12/2012] [Indexed: 11/30/2022]
Abstract
AIM Stimulation of soluble guanylate cyclase (sGC) with BAY 41-8543 was hypothesized to attenuate acute hypoxic pulmonary vasoconstriction alone and combined with dual endothelin (ET)-receptor antagonist tezosentan. METHODS Measurements were taken in 18 anaesthetized pigs with a mean ± SEM weight of 31.1 ± 0.4 kg, in normoxia (FiO(2)~0.21) and hypoxia (FiO(2)~0.10) without (control protocol, n = 6), and with right atrial infusion of BAY 41-8543 at 1, 3, 6, 9 and 12 μg min(-1) per kg (protocol 2, n = 6) or tezosentan at 5 mg kg(-1) followed by BAY 41-8543 at 1, 3 and 6 μg min(-1) per kg (protocol 3, n = 6). RESULTS Hypoxia (n = 18) increased (P < 0.001) mean pulmonary artery pressure (MPAP) and pulmonary vascular resistance (PVR) by 14.2 ± 0.6 mmHg and 2.8 ± 0.3 WU respectively. During sustained hypoxia without treatment, MPAP and PVR remained stable. BAY 41-8543 (n = 6) dose-dependently decreased (P < 0.001) MPAP and PVR by 15.0 ± 1.2 mmHg and 4.7 ± 0.7 WU respectively. Tezosentan (n = 6) decreased (P < 0.001) MPAP and PVR by 11.8 ± 1.2 mmHg and 2.0 ± 0.2 WU, respectively, whereafter BAY 41-8543 (n = 6) further decreased (P < 0.001) MPAP and PVR by 6.6 ± 0.9 mmHg and 1.9 ± 0.4 WU respectively. Both BAY 41-8543 and tezosentan decreased (P < 0.001) systemic arterial pressure and systemic vascular resistance. Blood-O(2) consumption remained unaltered (P = ns) during all interventions. CONCLUSION BAY 41-8543 totally reverses the effects of acute hypoxia-induced pulmonary vasoconstriction, and enhances the attenuating effects of tezosentan, without affecting oxygenation. Thus, sGC stimulation, alone or combined with dual ET-receptor blockade, could offer a means to treat pulmonary hypertension related to hypoxia and potentially other causes.
Collapse
Affiliation(s)
- J Lundgren
- The Öresund Cardiovascular Research Collaboration, The Clinic for Heart Failure and Valvular Disease, Skåne University Hospital, Lund, Sweden
| | | | | | | |
Collapse
|
24
|
Cattano D, Doursout MF. Pulmonary hypertension: have we learned enough yet? Intern Emerg Med 2012; 7:395-7. [PMID: 22903538 DOI: 10.1007/s11739-012-0840-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Accepted: 08/04/2012] [Indexed: 10/28/2022]
Affiliation(s)
- Davide Cattano
- Department of Anesthesiology, The University of Texas, Medical School at Houston, Houston, TX, USA.
| | | |
Collapse
|
25
|
Tabima DM, Roldan-Alzate A, Wang Z, Hacker TA, Molthen RC, Chesler NC. Persistent vascular collagen accumulation alters hemodynamic recovery from chronic hypoxia. J Biomech 2011; 45:799-804. [PMID: 22183202 DOI: 10.1016/j.jbiomech.2011.11.020] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2011] [Indexed: 01/04/2023]
Abstract
Pulmonary arterial hypertension (PAH) is caused by narrowing and stiffening of the pulmonary arteries that increase pulmonary vascular impedance (PVZ). In particular, small arteries narrow and large arteries stiffen. Large pulmonary artery (PA) stiffness is the best current predictor of mortality from PAH. We have previously shown that collagen accumulation leads to extralobar PA stiffening at high strain (Ooi et al. 2010). We hypothesized that collagen accumulation would increase PVZ, including total pulmonary vascular resistance (Z(0)), characteristic impedance (Z(C)), pulse wave velocity (PWV) and index of global wave reflections (P(b)/P(f)), which contribute to increased right ventricular afterload. We tested this hypothesis by exposing mice unable to degrade type I collagen (Col1a1(R/R)) to 21 days of hypoxia (hypoxia), some of which were allowed to recover for 42 days (recovery). Littermate wild-type mice (Col1a1(+/+)) were used as controls. In response to hypoxia, mean PA pressure (mPAP) increased in both mouse genotypes with no changes in cardiac output (CO) or PA inner diameter (ID); as a consequence, Z(0) (mPAP/CO) increased by ~100% in both genotypes (p<0.05). Contrary to our expectations, Z(C), PWV and P(b)/P(f) did not change. However, with recovery, Z(C) and PWV decreased in the Col1a1(+/+) mice and remained unchanged in the Col1a1(R/R) mice. Z(0) decreased with recovery in both genotypes. Microcomputed tomography measurements of large PAs did not show evidence of stiffness changes as a function of hypoxia exposure or genotype. We conclude that hypoxia-induced PA collagen accumulation does not affect the pulsatile components of pulmonary hemodynamics but that excessive collagen accumulation does prevent normal hemodynamic recovery, which may have important consequences for right ventricular function.
Collapse
Affiliation(s)
- Diana M Tabima
- Department of Biomedical Engineering, University of Wisconsin-Madison, WI 53706, USA
| | | | | | | | | | | |
Collapse
|
26
|
Tate MK, Lawrence WS, Gourley RL, Zavala DL, Weaver LE, Moen ST, Peterson JW. Telemetric left ventricular monitoring using wireless telemetry in the rabbit model. BMC Res Notes 2011; 4:320. [PMID: 21892949 PMCID: PMC3228511 DOI: 10.1186/1756-0500-4-320] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2010] [Accepted: 09/05/2011] [Indexed: 11/26/2022] Open
Abstract
Background Heart failure is a critical condition that affects many people and often results from left ventricular dysfunction. Numerous studies investigating this condition have been performed using various model systems. To do so, investigators must be able to accurately measure myocardial performance in order to determine the degree of left ventricular function. In this model development study, we employ a wireless telemetry system purchased from Data Sciences International to continuously assess left ventricular function in the rabbit model. Findings We surgically implanted pressure-sensitive catheters fitted to wireless radio-transmitters into the left ventricle of Dutch-belted rabbits. Following recovery of the animals, we continuously recorded indices of cardiac contractility and ventricular relaxation at baseline for a given time period. The telemetry system allowed us to continuously record baseline left ventricular parameters for the entire recording period. During this time, the animals were unrestrained and fully conscious. The values we recorded are similar to those obtained using other reported methods. Conclusions The wireless telemetry system can continuously measure left ventricular pressure, cardiac contractility, and cardiac relaxation in the rabbit model. These results, which were obtained just as baseline levels, substantiate the need for further validation in this model system of left ventricular assessment.
Collapse
Affiliation(s)
- Mallory K Tate
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, USA.
| | | | | | | | | | | | | |
Collapse
|
27
|
Baliga RS, MacAllister RJ, Hobbs AJ. New perspectives for the treatment of pulmonary hypertension. Br J Pharmacol 2011; 163:125-40. [PMID: 21175577 PMCID: PMC3085874 DOI: 10.1111/j.1476-5381.2010.01164.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Revised: 11/12/2010] [Accepted: 11/16/2010] [Indexed: 11/30/2022] Open
Abstract
Pulmonary hypertension (PH) is a debilitating disease with a poor prognosis. Therapeutic options remain limited despite the introduction of prostacyclin analogues, endothelin receptor antagonists and phosphodiesterase 5 inhibitors within the last 15 years; these interventions address predominantly the endothelial and vascular dysfunctionS associated with the condition, but simply delay progression of the disease rather than offer a cure. In an attempt to improve efficacy, emerging approaches have focused on targeting the pro-proliferative phenotype that underpins the pulmonary vascular remodelling in the lung and contributes to the impaired circulation and right heart failure. Many novel targets have been investigated and validated in animal models of PH, including modulation of guanylate cyclases, phosphodiesterases, tyrosine kinases, Rho kinase, bone morphogenetic proteins signalling, 5-HT, peroxisome proliferator activator receptors and ion channels. In addition, there is hope that combinations of such treatments, harnessing and optimizing vasodilator and anti-proliferative properties, will provide a further, possibly synergistic, increase in efficacy; therapies directed at the right heart may also offer an additional benefit. This overview highlights current therapeutic options, promising new therapies, and provides the rationale for a combination approach to treat the disease.
Collapse
|
28
|
Lasker GF, Maley JH, Pankey EA, Kadowitz PJ. Targeting soluble guanylate cyclase for the treatment of pulmonary hypertension. Expert Rev Respir Med 2011; 5:153-61. [PMID: 21510726 PMCID: PMC3108035 DOI: 10.1586/ers.11.9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Pulmonary arterial hypertension is a disease characterized by a sustained increase in pulmonary arterial pressure leading to right heart failure. Current treatments focus on endothelial dysfunction and an aberrant regulatory pathway for vascular tone. Unfortunately, a large proportion of patients are unresponsive to conventional vasodilator therapy. Investigations are ongoing into the effects of experimental therapies targeting the signal transduction pathway that mediates vasodilation. Here, we briefly discuss the pathophysiology of pulmonary hypertension and endothelial dysfunction, along with current treatments. We then present a focused review of recent animal studies and human trials examining the use of activators and stimulators of soluble guanylate cyclase for the treatment of pulmonary arterial hypertension and chronic thromboembolic pulmonary hypertension.
Collapse
Affiliation(s)
- George F Lasker
- Department of Pharmacology, Tulane University School of Medicine, 1430 Tulane Avenue, SL83, New Orleans, LA 70112-72699, USA
| | - Jason H Maley
- Department of Pharmacology, Tulane University School of Medicine, 1430 Tulane Avenue, SL83, New Orleans, LA 70112-72699, USA
| | - Edward A Pankey
- Department of Pharmacology, Tulane University School of Medicine, 1430 Tulane Avenue, SL83, New Orleans, LA 70112-72699, USA
| | - Philip J Kadowitz
- Department of Pharmacology, Tulane University School of Medicine, 1430 Tulane Avenue, SL83, New Orleans, LA 70112-72699, USA
| |
Collapse
|
29
|
Abstract
Since the discovery of nitric oxide (NO), which is released from endothelial cells as the main mediator of vasodilation, its target, the soluble guanylyl cyclase (sGC), has become a focus of interest for the treatment of diseases associated with endothelial dysfunction. NO donors were developed to suppress NO deficiency; however, tolerance to organic nitrates was reported. Non-NO-based drugs targeting sGC were developed to overcome the problem of tolerance. In this review, we briefly describe the process of sGC activation by its main physiological activator NO and the advances in the development of drugs capable of activating sGC in a NO-independent manner. sGC stimulators, as some of these drugs are called, require the integrity of the reduced heme moiety of the prosthetic group within the sGC and therefore are called heme-dependent stimulators. Other drugs are able to activate sGC independent of heme moiety and are hence called heme-independent activators. Because pathologic conditions modulate sGC and oxidize the heme moiety, the heme-independent sGC activators could potentially become drugs of choice because of their higher affinity to the oxidized enzyme. However, these drugs are still undergoing clinical trials and are not available for clinical use.
Collapse
|
30
|
Pankey EA, Bhartiya M, Badejo AM, Haider U, Stasch JP, Murthy SN, Nossaman BD, Kadowitz PJ. Pulmonary and systemic vasodilator responses to the soluble guanylyl cyclase activator, BAY 60-2770, are not dependent on endogenous nitric oxide or reduced heme. Am J Physiol Heart Circ Physiol 2011; 300:H792-802. [PMID: 21217076 DOI: 10.1152/ajpheart.00953.2010] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
4-({(4-Carboxybutyl)[2-(5-fluoro-2-{[4'-(trifluoromethyl)biphenyl-4-yl]methoxy}phenyl)ethyl]amino}methyl)benzoic acid (BAY 60-2770) is a nitric oxide (NO)-independent activator of soluble guanylyl cyclase (sGC) that increases the catalytic activity of the heme-oxidized or heme-free form of the enzyme. In this study, responses to intravenous injections of the sGC activator BAY 60-2770 were investigated under baseline and elevated tone conditions induced by the thromboxane mimic U-46619 when NO synthesis was inhibited by N(ω)-nitro-L-arginine methyl ester hydrochloride (L-NAME), when sGC activity was inhibited by 1H-[1,2,4]-oxadizaolo[4,3]quinoxaline-1-one (ODQ), an agent that oxidizes sGC, and in animals with monocrotaline-induced pulmonary hypertension. The intravenous injections of BAY 60-2770 under baseline conditions caused small decreases in pulmonary arterial pressure, larger decreases in systemic arterial pressure, and no change or small increases in cardiac output. Under elevated tone conditions during infusion of U-46619, intravenous injections of BAY 60-2770 caused larger decreases in pulmonary arterial pressure, smaller decreases in systemic arterial pressure, and increases in cardiac output. Pulmonary vasodilator responses to BAY 60-2770 were enhanced by L-NAME or by ODQ in a dose that attenuated responses to the NO donor sodium nitroprusside. ODQ had no significant effect on baseline pressures and attenuated pulmonary and systemic vasodilator responses to the sGC stimulator BAY 41-8543 2-{1-[2-(fluorophenyl)methyl]-1H-pyrazolo[3,4-b]pyridin-3-yl}-5(4-morpholinyl)-4,6-pyrimidinediamine. BAY 60-2770 and sodium nitroprusside decreased pulmonary and systemic arterial pressures in monocrotaline-treated rats in a nonselective manner. The present data show that BAY 60-2770 has vasodilator activity in the pulmonary and systemic vascular beds that is enhanced by ODQ and NOS inhibition, suggesting that the heme-oxidized form of sGC can be activated in vivo in an NO-independent manner to promote vasodilation. These results show that BAY 60-2770 and sodium nitroprusside decreased pulmonary and systemic arterial pressures in monocrotaline-treated rats, suggesting that BAY 60-2770 does not have selective pulmonary vasodilator activity in animals with monocrotaline-induced pulmonary hypertension.
Collapse
Affiliation(s)
- Edward A Pankey
- Department of Pharmacology, Tulane University School of Medicine, New Orleans 70112-2699, USA
| | | | | | | | | | | | | | | |
Collapse
|
31
|
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: 2.9] [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
|
32
|
Harrington LS, Moreno L, Reed A, Wort SJ, Desvergne B, Garland C, Zhao L, Mitchell JA. The PPARbeta/delta agonist GW0742 relaxes pulmonary vessels and limits right heart hypertrophy in rats with hypoxia-induced pulmonary hypertension. PLoS One 2010; 5:e9526. [PMID: 20209098 PMCID: PMC2831997 DOI: 10.1371/journal.pone.0009526] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2009] [Accepted: 12/28/2009] [Indexed: 01/29/2023] Open
Abstract
Background Pulmonary vascular diseases are increasingly recognised as important clinical conditions. Pulmonary hypertension associated with a range of aetiologies is difficult to treat and associated with progressive morbidity and mortality. Current therapies for pulmonary hypertension include phosphodiesterase type 5 inhibitors, endothelin receptor antagonists, or prostacyclin mimetics. However, none of these provide a cure and the clinical benefits of these drugs individually decline over time. There is, therefore, an urgent need to identify new treatment strategies for pulmonary hypertension. Methodology/Principal Findings Here we show that the PPARβ/δ agonist GW0742 induces vasorelaxation in systemic and pulmonary vessels. Using tissue from genetically modified mice, we show that the dilator effects of GW0742 are independent of the target receptor PPARβ/δ or cell surface prostacyclin (IP) receptors. In aortic tissue, vascular relaxant effects of GW0742 were not associated with increases in cGMP, cAMP or hyperpolarisation, but were attributed to inhibition of RhoA activity. In a rat model of hypoxia-induced pulmonary hypertension, daily oral dosing of animals with GW0742 (30 mg/kg) for 3 weeks significantly reduced the associated right heart hypertrophy and right ventricular systolic pressure. GW0742 had no effect on vascular remodelling induced by hypoxia in this model. Conclusions/Significance These observations are the first to show a therapeutic benefit of ‘PPARβ/δ’ agonists in experimental pulmonary arterial hypertension and provide pre-clinical evidence to favour clinical trials in man.
Collapse
Affiliation(s)
| | - Laura Moreno
- Cardiothoracic Pharmacology, NHLI, Imperial College London, United Kingdom
| | - Anna Reed
- Cardiothoracic Pharmacology, NHLI, Imperial College London, United Kingdom
| | - Stephen J. Wort
- Critical Care Medicine, NHLI, Royal Brompton Hospital, London, United Kingdom
| | - Béatrice Desvergne
- Center for Integrative Genomics, University of Lausanne, Genopode, Lausanne, Switzerland
| | | | - Lan Zhao
- Experimental Medicine and Toxicology, Hammersmith Hospital, Imperial College London, London, United Kingdom
| | - Jane A. Mitchell
- Cardiothoracic Pharmacology, NHLI, Imperial College London, United Kingdom
- * E-mail:
| |
Collapse
|
33
|
Abstract
Pulmonary hypertension is an orphan disease that until recently has received limited attention within the wider medical community. This has changed distinctly in the last 10 years with the advent of new classes of therapy and a renewed interest in mechanisms of pathogenesis. This review utilized information gathered from recent conferences, and a review of the literature was conducted using MedLine and Pubmed. Accepted mechanisms of pathogenesis and currently available treatments are presented. We will discuss interesting new concepts in pathogenesis, including the importance of genetic forms of the disease and in particular the transforming growth factor receptor superfamily and the evolving evidence of the contribution of dysregulated immunity. Areas of research may yield therapeutic benefits in the not-too-distant future, including anti-proliferative therapies and stem cell therapy.
Collapse
Affiliation(s)
- M Toshner
- Department of Medicine, University of Cambridge, Cambridge CB2 2QQ, UK
| | | | | |
Collapse
|