1
|
Xu W, Asosingh K, Janocha AJ, Madden E, Wanner N, Trotter D, Novotny MV, Mulya A, Farha S, Erzurum SC. Hypoxia responses in arginase 2 deficient mice enhance cardiovascular health. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.20.639297. [PMID: 40060685 PMCID: PMC11888215 DOI: 10.1101/2025.02.20.639297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/18/2025]
Abstract
RATIONALE Physiological responses to hypoxia involve adaptations in the hematopoietic and cardiovascular systems, which work together to ensure adequate oxygen delivery to tissues for energy production. The arginine/nitric oxide (NO) pathway regulates both systems through its effects on erythropoiesis and vasodilation. In Tibetan populations native to high-altitude hypoxia, increased NO production from arginine and decreased arginine metabolism by arginase contribute to these adaptive mechanisms. These metabolic changes enhance tissue oxygen delivery and reduce the risk of hypoxic pulmonary hypertension. Here, we hypothesize that genetic deletion of mitochondrial arginase 2 ( Arg2 ) in mice will enhance cardiovascular effects and mitigate hypoxia-induced pulmonary hypertension. METHODS Complete blood counts, bone marrow erythroid differentiation, plasma arginine and NO (measured as nitrite), right ventricular systolic pressure (RVSP), heart rate, heart weight, and blood pressure were measured in wild-type (WT) and Arg2 knockout ( Arg2 KO) mice exposed to short-term (6, 12, 48, or 72 hours) or long-term (3 weeks) hypoxia. RESULTS Under normoxic conditions, Arg2 KO and WT mice exhibit similar RBC counts, hemoglobin levels, hematocrit, heart rate, systolic and diastolic blood pressures, and heart weight (all P > 0.05). WT mice increase erythropoiesis at 12 hours of hypoxia, including proerythroblasts (stage I, P = 0.004), polychromatic erythroblasts (stage III, P = 0.0004), and orthochromatic erythroblasts (stage IV, P = 0.03), but Arg2 KO mice do not increase erythropoiesis. After 48 hours of hypoxia, Arg2 KO mice increase proerythroblasts (stage I, P = 0.0008), but levels remain significantly lower than in WT mice. Plasma arginine and NO levels increase under hypoxia. NO levels peak at 12 hours of hypoxia in WT mice, then decline rapidly. In contrast, NO levels in Arg2 KO mice are higher than in WT mice, with sustained elevations at 48 hours of hypoxia ( P = 0.03). Arg2 KO mice have significantly higher plasma arginine levels than WT at 6, 12, and 72 hours of hypoxia (all P < 0.05). Under chronic hypoxia, Arg2 KO and WT mice show similar RBC counts, hemoglobin levels, hematocrit, and NO levels. Unlike WT, Arg2 KO mice do not increase RVSP ( P = 0.4) and have lower mean arterial ( P = 0.03) and diastolic blood pressures ( P = 0.01), as well as much lower heart rates ( P < 0.0001). Additionally, small blood vessels increase in lungs of Arg2 KO mice (CD31, P = 0.02; vWF, P = 0.6). CONCLUSIONS Arginine metabolism in the mitochondria plays a key role in modulating adaptive responses to hypoxia. Deletion of Arg2 results in delayed erythropoiesis under acute hypoxia, but better cardiovascular health, as indicated by higher levels of nitrite and arginine, and lower RVSP, blood pressure, and heart rate with chronic hypoxia.
Collapse
|
2
|
Ogoshi T, Yatera K, Mukae H, Tsutsui M. Role of Nitric Oxide Synthases in Respiratory Health and Disease: Insights from Triple Nitric Oxide Synthases Knockout Mice. Int J Mol Sci 2024; 25:9317. [PMID: 39273265 PMCID: PMC11395504 DOI: 10.3390/ijms25179317] [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: 07/23/2024] [Revised: 08/26/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
The system of nitric oxide synthases (NOSs) is comprised of three isoforms: nNOS, iNOS, and eNOS. The roles of NOSs in respiratory diseases in vivo have been studied by using inhibitors of NOSs and NOS-knockout mice. Their exact roles remain uncertain, however, because of the non-specificity of inhibitors of NOSs and compensatory up-regulation of other NOSs in NOS-KO mice. We addressed this point in our triple-n/i/eNOSs-KO mice. Triple-n/i/eNOSs-KO mice spontaneously developed pulmonary emphysema and displayed exacerbation of bleomycin-induced pulmonary fibrosis as compared with wild-type (WT) mice. Triple-n/i/eNOSs-KO mice exhibited worsening of hypoxic pulmonary hypertension (PH), which was reversed by treatment with sodium nitrate, and WT mice that underwent triple-n/i/eNOSs-KO bone marrow transplantation (BMT) also showed aggravation of hypoxic PH compared with those that underwent WT BMT. Conversely, ovalbumin-evoked asthma was milder in triple-n/i/eNOSs-KO than WT mice. These results suggest that the roles of NOSs are different in different pathologic states, even in the same respiratory diseases, indicating the diversity of the roles of NOSs. In this review, we describe these previous studies and discuss the roles of NOSs in respiratory health and disease. We also explain the current state of development of inorganic nitrate as a new drug for respiratory diseases.
Collapse
Affiliation(s)
- Takaaki Ogoshi
- Department of Respiratory Medicine, Kokura Memorial Hospital, 1-1 Asano, Kokura-kita-ku, Kitakyushu 803-0802, Japan;
- Department of Respiratory Medicine, University of Occupational and Environmental Health, Japan, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu 807-8555, Japan;
| | - Kazuhiro Yatera
- Department of Respiratory Medicine, University of Occupational and Environmental Health, Japan, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu 807-8555, Japan;
| | - Hiroshi Mukae
- Department of Respiratory Disease, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan;
| | - Masato Tsutsui
- Department of Pharmacology, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Okinawa 903-0215, Japan
| |
Collapse
|
3
|
Knight H, Abis G, Kaur M, Green HL, Krasemann S, Hartmann K, Lynham S, Clark J, Zhao L, Ruppert C, Weiss A, Schermuly RT, Eaton P, Rudyk O. Cyclin D-CDK4 Disulfide Bond Attenuates Pulmonary Vascular Cell Proliferation. Circ Res 2023; 133:966-988. [PMID: 37955182 PMCID: PMC10699508 DOI: 10.1161/circresaha.122.321836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/31/2023] [Accepted: 11/02/2023] [Indexed: 11/14/2023]
Abstract
BACKGROUND Pulmonary hypertension (PH) is a chronic vascular disease characterized, among other abnormalities, by hyperproliferative smooth muscle cells and a perturbed cellular redox and metabolic balance. Oxidants induce cell cycle arrest to halt proliferation; however, little is known about the redox-regulated effector proteins that mediate these processes. Here, we report a novel kinase-inhibitory disulfide bond in cyclin D-CDK4 (cyclin-dependent kinase 4) and investigate its role in cell proliferation and PH. METHODS Oxidative modifications of cyclin D-CDK4 were detected in human pulmonary arterial smooth muscle cells and human pulmonary arterial endothelial cells. Site-directed mutagenesis, tandem mass-spectrometry, cell-based experiments, in vitro kinase activity assays, in silico structural modeling, and a novel redox-dead constitutive knock-in mouse were utilized to investigate the nature and definitively establish the importance of CDK4 cysteine modification in pulmonary vascular cell proliferation. Furthermore, the cyclin D-CDK4 oxidation was assessed in vivo in the pulmonary arteries and isolated human pulmonary arterial smooth muscle cells of patients with pulmonary arterial hypertension and in 3 preclinical models of PH. RESULTS Cyclin D-CDK4 forms a reversible oxidant-induced heterodimeric disulfide dimer between C7/8 and C135, respectively, in cells in vitro and in pulmonary arteries in vivo to inhibit cyclin D-CDK4 kinase activity, decrease Rb (retinoblastoma) protein phosphorylation, and induce cell cycle arrest. Mutation of CDK4 C135 causes a kinase-impaired phenotype, which decreases cell proliferation rate and alleviates disease phenotype in an experimental mouse PH model, suggesting this cysteine is indispensable for cyclin D-CDK4 kinase activity. Pulmonary arteries and human pulmonary arterial smooth muscle cells from patients with pulmonary arterial hypertension display a decreased level of CDK4 disulfide, consistent with CDK4 being hyperactive in human pulmonary arterial hypertension. Furthermore, auranofin treatment, which induces the cyclin D-CDK4 disulfide, attenuates disease severity in experimental PH models by mitigating pulmonary vascular remodeling. CONCLUSIONS A novel disulfide bond in cyclin D-CDK4 acts as a rapid switch to inhibit kinase activity and halt cell proliferation. This oxidative modification forms at a critical cysteine residue, which is unique to CDK4, offering the potential for the design of a selective covalent inhibitor predicted to be beneficial in PH.
Collapse
Affiliation(s)
- Hannah Knight
- School of Cardiovascular and Metabolic Medicine and Sciences, British Heart Foundation Centre of Research Excellence (H.K., M.K., H.L.H.G., J.C., O.R.), King’s College London, United Kingdom
| | - Giancarlo Abis
- Division of Biosciences, Institute of Structural and Molecular Biology, University College London, United Kingdom (G.A.)
| | - Manpreet Kaur
- School of Cardiovascular and Metabolic Medicine and Sciences, British Heart Foundation Centre of Research Excellence (H.K., M.K., H.L.H.G., J.C., O.R.), King’s College London, United Kingdom
| | - Hannah L.H. Green
- School of Cardiovascular and Metabolic Medicine and Sciences, British Heart Foundation Centre of Research Excellence (H.K., M.K., H.L.H.G., J.C., O.R.), King’s College London, United Kingdom
| | - Susanne Krasemann
- Institute of Neuropathology, University Medical Centre Hamburg-Eppendorf, Germany (S.K., K.H.)
| | - Kristin Hartmann
- Institute of Neuropathology, University Medical Centre Hamburg-Eppendorf, Germany (S.K., K.H.)
| | - Steven Lynham
- Proteomics Core Facility, Centre of Excellence for Mass Spectrometry (S.L.), King’s College London, United Kingdom
| | - James Clark
- School of Cardiovascular and Metabolic Medicine and Sciences, British Heart Foundation Centre of Research Excellence (H.K., M.K., H.L.H.G., J.C., O.R.), King’s College London, United Kingdom
| | - Lan Zhao
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, United Kingdom (L.Z.)
| | - Clemens Ruppert
- Universities of Giessen and Marburg Lung Center Giessen Biobank, Justus-Liebig-University Giessen, Germany (C.R.)
| | - Astrid Weiss
- Department of Internal Medicine, Justus-Liebig-University Giessen, Giessen, Member of the German Center for Lung Research (DZL), Germany (A.W., R.T.S.)
| | - Ralph T. Schermuly
- Department of Internal Medicine, Justus-Liebig-University Giessen, Giessen, Member of the German Center for Lung Research (DZL), Germany (A.W., R.T.S.)
| | - Philip Eaton
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom (P.E.)
| | - Olena Rudyk
- School of Cardiovascular and Metabolic Medicine and Sciences, British Heart Foundation Centre of Research Excellence (H.K., M.K., H.L.H.G., J.C., O.R.), King’s College London, United Kingdom
| |
Collapse
|
4
|
Sakarin S, Rungsipipat A, Surachetpong SD. Perivascular inflammatory cells and their association with pulmonary arterial remodelling in dogs with pulmonary hypertension due to myxomatous mitral valve disease. Vet Res Commun 2023; 47:1505-1521. [PMID: 36976445 DOI: 10.1007/s11259-023-10106-0] [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: 12/09/2022] [Accepted: 03/15/2023] [Indexed: 03/29/2023]
Abstract
Pulmonary hypertension (PH), an increase in pulmonary arterial pressure (PAP), may occur in dogs affected with myxomatous mitral valve disease (MMVD). Recent studies suggest that an accumulation of perivascular inflammatory cells may be involved with medial thickening which is a sign of the pulmonary artery remodelling in PH. The aim of this study was to characterise perivascular inflammatory cells in the surrounding pulmonary arteries of dogs with PH due to MMVD compared to MMVD dogs and healthy control dogs. Nineteen lung samples were collected from cadavers of small-breed dogs (control n = 5; MMVD n = 7; MMVD + PH n = 7). Toluidine blue stain and multiple IHC targeting α-SMA, vWF, CD20, CD68 and CD3 was performed to examine intimal and medial thickening, assess muscularisation of the small pulmonary arteries and characterise perivascular leucocytes. Medial thickening without intimal thickening of pulmonary arteries and muscularisation of normally non-muscularised small pulmonary arteries was observed in the MMVD and MMVD + PH groups compared with the control group. The perivascular numbers of B lymphocytes, T lymphocytes and macrophages was significantly increased in the MMVD + PH group compared with the MMVD and control groups. In contrast, the perivascular number of mast cells was significantly higher in the MMVD group compared with the MMVD + PH and control groups. This study suggested that pulmonary artery remodelling as medial thickening and muscularisation of the normally non-muscular small pulmonary arteries is accompanied by the accumulation of perivascular inflammatory cells.
Collapse
Affiliation(s)
- Siriwan Sakarin
- Department of Veterinary Medicine, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Anudep Rungsipipat
- Companion Animal Cancer Research Unit, Department of Veterinary Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Sirilak Disatian Surachetpong
- Department of Veterinary Medicine, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
5
|
Campos-Carraro C, Turck P, de Lima-Seolin BG, Teixeira RB, Zimmer A, Araujo ASDR, Belló-Klein A. Copaiba oil improves pulmonary nitric oxide bioavailability in monocrotaline-treated rats. Can J Physiol Pharmacol 2023; 101:447-454. [PMID: 37581356 DOI: 10.1139/cjpp-2023-0028] [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] [Indexed: 08/16/2023]
Abstract
Oxidative stress is involved in increased pulmonary vascular resistance (PVR) and right ventricular (RV) hypertrophy, characteristics of pulmonary arterial hypertension (PAH). Copaiba oil, an antioxidant compound, could attenuate PAH damage. This study's aim was to determine the effects of copaiba oil on lung oxidative stress, PVR, and mean pulmonary arterial pressure (mPAP) in the monocrotaline (MCT) model of PAH. Male Wistar rats (170 g, n = 7/group) were divided into four groups: control, MCT, copaiba oil, and MCT + copaiba oil (MCT-O). PAH was induced by MCT (60 mg/kg i.p.) and, after 1 week, the treatment with copaiba oil (400 mg/kg/day gavage) was started for 14 days. Echocardiographic and hemodynamic measurements were performed. RV was collected for morphometric evaluations and lungs and the pulmonary artery were used for biochemical analysis. Copaiba oil significantly reduced RV hypertrophy, PVR, mPAP, and antioxidant enzyme activities in the MCT-O group. Moreover, increased nitric oxide synthase and decreased NADPH oxidase activities were observed in the MCT-O group. In conclusion, copaiba oil was able to improve the balance between nitric oxide and reactive oxygen species in lungs and the pulmonary artery and to reduce PVR, which could explain a decrease in RV hypertrophy in this PAH model.
Collapse
Affiliation(s)
| | - Patrick Turck
- Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | | | | | - Alexsandra Zimmer
- Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | | | - Adriane Belló-Klein
- Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
6
|
Ormiston ML, Jankov RP, Stewart DJ. Oh NO! Loss of PHD2 leads to "radical" changes in the lung vasculature. Eur Respir J 2022; 60:2201776. [PMID: 36549689 DOI: 10.1183/13993003.01776-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 09/17/2022] [Indexed: 12/24/2022]
Affiliation(s)
- Mark L Ormiston
- Departments of Biomedical and Molecular Sciences, Medicine and Surgery, Queen's University, Kingston, ON, Canada
| | - Robert P Jankov
- Departments of Paediatrics and Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Duncan J Stewart
- Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute and Departments of Medicine and Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
7
|
Garcia SM, Yellowhair TR, Detweiler ND, Ahmadian R, Herbert LM, Gonzalez Bosc LV, Resta TC, Jernigan NL. Smooth muscle Acid-sensing ion channel 1a as a therapeutic target to reverse hypoxic pulmonary hypertension. Front Mol Biosci 2022; 9:989809. [PMID: 36275633 PMCID: PMC9581175 DOI: 10.3389/fmolb.2022.989809] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 09/15/2022] [Indexed: 11/17/2022] Open
Abstract
Acid-sensing ion channel 1a (ASIC1a) is a voltage-independent, non-selective cation channel that conducts both Na+ and Ca2+. Activation of ASIC1a elicits plasma membrane depolarization and stimulates intracellular Ca2+-dependent signaling pathways in multiple cell types, including vascular smooth muscle (SM) and endothelial cells (ECs). Previous studies have shown that increases in pulmonary vascular resistance accompanying chronic hypoxia (CH)-induced pulmonary hypertension requires ASIC1a to elicit enhanced pulmonary vasoconstriction and vascular remodeling. Both SM and EC dysfunction drive these processes; however, the involvement of ASIC1a within these different cell types is unknown. Using the Cre-LoxP system to generate cell-type-specific Asic1a knockout mice, we tested the hypothesis that SM-Asic1a contributes to CH-induced pulmonary hypertension and vascular remodeling, whereas EC-Asic1a opposes the development of CH-induced pulmonary hypertension. The severity of pulmonary hypertension was not altered in mice with specific deletion of EC-Asic1a (TekCre-Asic1afl/fl). However, similar to global Asic1a knockout (Asic1a−/-) mice, mice with specific deletion of SM-Asic1a (MHCCreER-Asic1afl/fl) were protected from the development of CH-induced pulmonary hypertension and right heart hypertrophy. Furthermore, pulmonary hypertension was reversed when deletion of SM-Asic1a was initiated in conditional MHCCreER-Asic1afl/fl mice with established pulmonary hypertension. CH-induced vascular remodeling was also significantly attenuated in pulmonary arteries from MHCCreER-Asic1afl/fl mice. These findings were additionally supported by decreased CH-induced proliferation and migration of pulmonary arterial smooth muscle cells (PASMCs) from Asic1a−/- mice. Together these data demonstrate that SM-, but not EC-Asic1a contributes to CH-induced pulmonary hypertension and vascular remodeling. Furthermore, these studies provide evidence for the therapeutic potential of ASIC1a inhibition to reverse pulmonary hypertension.
Collapse
|
8
|
Flores K, Siques P, Brito J, Arribas SM. AMPK and the Challenge of Treating Hypoxic Pulmonary Hypertension. Int J Mol Sci 2022; 23:ijms23116205. [PMID: 35682884 PMCID: PMC9181235 DOI: 10.3390/ijms23116205] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/29/2022] [Accepted: 04/30/2022] [Indexed: 02/01/2023] Open
Abstract
Hypoxic pulmonary hypertension (HPH) is characterized by sustained elevation of pulmonary artery pressure produced by vasoconstriction and hyperproliferative remodeling of the pulmonary artery and subsequent right ventricular hypertrophy (RVH). The search for therapeutic targets for cardiovascular pathophysiology has extended in many directions. However, studies focused on mitigating high-altitude pulmonary hypertension (HAPH) have been rare. Because AMP-activated protein kinase (AMPK) is involved in cardiovascular and metabolic pathology, AMPK is often studied as a potential therapeutic target. AMPK is best characterized as a sensor of cellular energy that can also restore cellular metabolic homeostasis. However, AMPK has been implicated in other pathways with vasculoprotective effects. Notably, cellular metabolic stress increases the intracellular ADP/ATP or AMP/ATP ratio, and AMPK activation restores ATP levels by activating energy-producing catabolic pathways and inhibiting energy-consuming anabolic pathways, such as cell growth and proliferation pathways, promoting cardiovascular protection. Thus, AMPK activation plays an important role in antiproliferative, antihypertrophic and antioxidant pathways in the pulmonary artery in HPH. However, AMPK plays contradictory roles in promoting HPH development. This review describes the main findings related to AMPK participation in HPH and its potential as a therapeutic target. It also extrapolates known AMPK functions to discuss the less-studied HAPH context.
Collapse
Affiliation(s)
- Karen Flores
- Institute of Health Studies, University Arturo Prat, Av. Arturo Prat 2120, Iquique 1110939, Chile; (P.S.); (J.B.)
- Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and Its Health Sequelae, 20251 Hamburg, Germany and Iquique 1100000, Chile
- Correspondence: ; Tel.: +56-572526392
| | - Patricia Siques
- Institute of Health Studies, University Arturo Prat, Av. Arturo Prat 2120, Iquique 1110939, Chile; (P.S.); (J.B.)
- Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and Its Health Sequelae, 20251 Hamburg, Germany and Iquique 1100000, Chile
| | - Julio Brito
- Institute of Health Studies, University Arturo Prat, Av. Arturo Prat 2120, Iquique 1110939, Chile; (P.S.); (J.B.)
- Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and Its Health Sequelae, 20251 Hamburg, Germany and Iquique 1100000, Chile
| | - Silvia M. Arribas
- Department of Physiology, University Autonoma of Madrid, 28049 Madrid, Spain;
| |
Collapse
|
9
|
Tobal R, Potjewijd J, van Empel VPM, Ysermans R, Schurgers LJ, Reutelingsperger CP, Damoiseaux JGMC, van Paassen P. Vascular Remodeling in Pulmonary Arterial Hypertension: The Potential Involvement of Innate and Adaptive Immunity. Front Med (Lausanne) 2022; 8:806899. [PMID: 35004784 PMCID: PMC8727487 DOI: 10.3389/fmed.2021.806899] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 12/02/2021] [Indexed: 11/30/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe disease with high morbidity and mortality. Current therapies are mainly focused on vasodilative agents to improve prognosis. However, recent literature has shown the important interaction between immune cells and stromal vascular cells in the pathogenic modifications of the pulmonary vasculature. The immunological pathogenesis of PAH is known as a complex interplay between immune cells and vascular stromal cells, via direct contacts and/or their production of extra-cellular/diffusible factors such as cytokines, chemokines, and growth factors. These include, the B-cell—mast-cell axis, endothelium mediated fibroblast activation and subsequent M2 macrophage polarization, anti-endothelial cell antibodies and the versatile role of IL-6 on vascular cells. This review aims to outline the major pathophysiological changes in vascular cells caused by immunological mechanisms, leading to vascular remodeling, increased pulmonary vascular resistance and eventually PAH. Considering the underlying immunological mechanisms, these mechanisms may be key to halt progression of disease.
Collapse
Affiliation(s)
- Rachid Tobal
- Division of Nephrology and Clinical and Experimental Immunology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, Netherlands
| | - Judith Potjewijd
- Division of Nephrology and Clinical and Experimental Immunology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, Netherlands
| | - Vanessa P M van Empel
- Department of Cardiology, Maastricht University Medical Center, Maastricht, Netherlands
| | - Renee Ysermans
- Division of Nephrology and Clinical and Experimental Immunology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, Netherlands
| | - Leon J Schurgers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht, Netherlands
| | - Chris P Reutelingsperger
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht, Netherlands
| | - Jan G M C Damoiseaux
- Central Diagnostic Laboratory, Maastricht University Medical Center, Maastricht, Netherlands
| | - Pieter van Paassen
- Division of Nephrology and Clinical and Experimental Immunology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, Netherlands
| |
Collapse
|
10
|
Kurakula K, Smolders VFED, Tura-Ceide O, Jukema JW, Quax PHA, Goumans MJ. Endothelial Dysfunction in Pulmonary Hypertension: Cause or Consequence? Biomedicines 2021; 9:biomedicines9010057. [PMID: 33435311 PMCID: PMC7827874 DOI: 10.3390/biomedicines9010057] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/30/2020] [Accepted: 01/03/2021] [Indexed: 12/11/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare, complex, and progressive disease that is characterized by the abnormal remodeling of the pulmonary arteries that leads to right ventricular failure and death. Although our understanding of the causes for abnormal vascular remodeling in PAH is limited, accumulating evidence indicates that endothelial cell (EC) dysfunction is one of the first triggers initiating this process. EC dysfunction leads to the activation of several cellular signalling pathways in the endothelium, resulting in the uncontrolled proliferation of ECs, pulmonary artery smooth muscle cells, and fibroblasts, and eventually leads to vascular remodelling and the occlusion of the pulmonary blood vessels. Other factors that are related to EC dysfunction in PAH are an increase in endothelial to mesenchymal transition, inflammation, apoptosis, and thrombus formation. In this review, we outline the latest advances on the role of EC dysfunction in PAH and other forms of pulmonary hypertension. We also elaborate on the molecular signals that orchestrate EC dysfunction in PAH. Understanding the role and mechanisms of EC dysfunction will unravel the therapeutic potential of targeting this process in PAH.
Collapse
Affiliation(s)
- Kondababu Kurakula
- Department of Cell and Chemical Biology, Laboratory for CardioVascular Cell Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
| | - Valérie F. E. D. Smolders
- Department of Surgery, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (V.F.E.D.S.); (P.H.A.Q.)
| | - Olga Tura-Ceide
- Department of Pulmonary Medicine, Hospital Clínic-Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain;
- Department of Pulmonary Medicine, Dr. Josep Trueta University Hospital de Girona, Santa Caterina Hospital de Salt and the Girona Biomedical Research Institut (IDIBGI), 17190 Girona, Catalonia, Spain
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), 28029 Madrid, Spain
| | - J. Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
| | - Paul H. A. Quax
- Department of Surgery, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (V.F.E.D.S.); (P.H.A.Q.)
| | - Marie-José Goumans
- Department of Cell and Chemical Biology, Laboratory for CardioVascular Cell Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
- Correspondence:
| |
Collapse
|
11
|
Otoupalova E, Smith S, Cheng G, Thannickal VJ. Oxidative Stress in Pulmonary Fibrosis. Compr Physiol 2020; 10:509-547. [PMID: 32163196 DOI: 10.1002/cphy.c190017] [Citation(s) in RCA: 163] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Oxidative stress has been linked to various disease states as well as physiological aging. The lungs are uniquely exposed to a highly oxidizing environment and have evolved several mechanisms to attenuate oxidative stress. Idiopathic pulmonary fibrosis (IPF) is a progressive age-related disorder that leads to architectural remodeling, impaired gas exchange, respiratory failure, and death. In this article, we discuss cellular sources of oxidant production, and antioxidant defenses, both enzymatic and nonenzymatic. We outline the current understanding of the pathogenesis of IPF and how oxidative stress contributes to fibrosis. Further, we link oxidative stress to the biology of aging that involves DNA damage responses, loss of proteostasis, and mitochondrial dysfunction. We discuss the recent findings on the role of reactive oxygen species (ROS) in specific fibrotic processes such as macrophage polarization and immunosenescence, alveolar epithelial cell apoptosis and senescence, myofibroblast differentiation and senescence, and alterations in the acellular extracellular matrix. Finally, we provide an overview of the current preclinical studies and clinical trials targeting oxidative stress in fibrosis and potential new strategies for future therapeutic interventions. © 2020 American Physiological Society. Compr Physiol 10:509-547, 2020.
Collapse
Affiliation(s)
- Eva Otoupalova
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Sam Smith
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Guangjie Cheng
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Victor J Thannickal
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
12
|
Ogoshi T, Tsutsui M, Kido T, Sakanashi M, Naito K, Oda K, Ishimoto H, Yamada S, Wang KY, Toyohira Y, Izumi H, Masuzaki H, Shimokawa H, Yanagihara N, Yatera K, Mukae H. Protective Role of Myelocytic Nitric Oxide Synthases against Hypoxic Pulmonary Hypertension in Mice. Am J Respir Crit Care Med 2019; 198:232-244. [PMID: 29480750 DOI: 10.1164/rccm.201709-1783oc] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
RATIONALE Nitric oxide (NO), synthesized by NOSs (NO synthases), plays a role in the development of pulmonary hypertension (PH). However, the role of NO/NOSs in bone marrow (BM) cells in PH remains elusive. OBJECTIVES To determine the role of NOSs in BM cells in PH. METHODS Experiments were performed on 36 patients with idiopathic pulmonary fibrosis and on wild-type (WT), nNOS (neuronal NOS)-/-, iNOS (inducible NOS)-/-, eNOS (endothelial NOS)-/-, and n/i/eNOSs-/- mice. MEASUREMENTS AND MAIN RESULTS In the patients, there was a significant correlation between higher pulmonary artery systolic pressure and lower nitrite plus nitrate levels in the BAL fluid. In the mice, hypoxia-induced PH deteriorated significantly in the n/i/eNOSs-/- genotype and, to a lesser extent, in the eNOS-/- genotype as compared with the WT genotype. In the n/i/eNOSs-/- genotype exposed to hypoxia, the number of circulating BM-derived vascular smooth muscle progenitor cells was significantly larger, and transplantation of green fluorescent protein-transgenic BM cells revealed the contribution of BM cells to pulmonary vascular remodeling. Importantly, n/i/eNOSs-/--BM transplantation significantly aggravated hypoxia-induced PH in the WT genotype, and WT-BM transplantation significantly ameliorated hypoxia-induced PH in the n/i/eNOSs-/- genotype. A total of 69 and 49 mRNAs related to immunity and inflammation, respectively, were significantly upregulated in the lungs of WT genotype mice transplanted with n/i/eNOSs-/--BM compared with those with WT-BM, suggesting the involvement of immune and inflammatory mechanisms in the exacerbation of hypoxia-induced PH caused by n/i/eNOSs-/--BM transplantation. CONCLUSIONS These results demonstrate that myelocytic n/i/eNOSs play an important protective role in the pathogenesis of PH.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Hiroshi Ishimoto
- 1 Department of Respiratory Medicine.,3 Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan; and
| | | | | | | | - Hiroto Izumi
- 7 Department of Occupational Pneumology, Institute of Industrial Ecological Sciences, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Hiroaki Masuzaki
- 8 Second Department of Internal Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Hiroaki Shimokawa
- 9 Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | - Hiroshi Mukae
- 1 Department of Respiratory Medicine.,3 Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan; and
| |
Collapse
|
13
|
Stenmark KR, Frid MG, Graham BB, Tuder RM. Dynamic and diverse changes in the functional properties of vascular smooth muscle cells in pulmonary hypertension. Cardiovasc Res 2019; 114:551-564. [PMID: 29385432 DOI: 10.1093/cvr/cvy004] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 01/26/2018] [Indexed: 12/21/2022] Open
Abstract
Pulmonary hypertension (PH) is the end result of interaction between pulmonary vascular tone and a complex series of cellular and molecular events termed 'vascular remodelling'. The remodelling process, which can involve the entirety of pulmonary arterial vasculature, almost universally involves medial thickening, driven by increased numbers and hypertrophy of its principal cellular constituent, smooth muscle cells (SMCs). It is noted, however that SMCs comprise heterogeneous populations of cells, which can exhibit markedly different proliferative, inflammatory, and extracellular matrix production changes during remodelling. We further consider that these functional changes in SMCs of different phenotype and their role in PH are dynamic and may undergo significant changes over time (which we will refer to as cellular plasticity); no single property can account for the complexity of the contribution of SMC to pulmonary vascular remodelling. Thus, the approaches used to pharmacologically manipulate PH by targeting the SMC phenotype(s) must take into account processes that underlie dominant phenotypes that drive the disease. We present evidence for time- and location-specific changes in SMC proliferation in various animal models of PH; we highlight the transient nature (rather than continuous) of SMC proliferation, emphasizing that the heterogenic SMC populations that reside in different locations along the pulmonary vascular tree exhibit distinct responses to the stresses associated with the development of PH. We also consider that cells that have often been termed 'SMCs' may arise from many origins, including endothelial cells, fibroblasts and resident or circulating progenitors, and thus may contribute via distinct signalling pathways to the remodelling process. Ultimately, PH is characterized by long-lived, apoptosis-resistant SMC. In line with this key pathogenic characteristic, we address the acquisition of a pro-inflammatory phenotype by SMC that is essential to the development of PH. We present evidence that metabolic alterations akin to those observed in cancer cells (cytoplasmic and mitochondrial) directly contribute to the phenotype of the SM and SM-like cells involved in PH. Finally, we raise the possibility that SMCs transition from a proliferative to a senescent, pro-inflammatory and metabolically active phenotype over time.
Collapse
Affiliation(s)
- Kurt R Stenmark
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, 12700 E. 19th Avenue, RC2, B131, Aurora, CO 80045, USA
| | - Maria G Frid
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, 12700 E. 19th Avenue, RC2, B131, Aurora, CO 80045, USA
| | - Brian B Graham
- Pulmonary and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, 12700 E. 19th Avenue, RC2, B131, Aurora, CO 80045, USA
| | - Rubin M Tuder
- Pulmonary and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, 12700 E. 19th Avenue, RC2, B131, Aurora, CO 80045, USA
| |
Collapse
|
14
|
Yeligar SM, Kang BY, Bijli KM, Kleinhenz JM, Murphy TC, Torres G, San Martin A, Sutliff RL, Hart CM. PPARγ Regulates Mitochondrial Structure and Function and Human Pulmonary Artery Smooth Muscle Cell Proliferation. Am J Respir Cell Mol Biol 2019; 58:648-657. [PMID: 29182484 DOI: 10.1165/rcmb.2016-0293oc] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Pulmonary hypertension (PH) is a progressive disorder that causes significant morbidity and mortality despite existing therapies. PH pathogenesis is characterized by metabolic derangements that increase pulmonary artery smooth muscle cell (PASMC) proliferation and vascular remodeling. PH-associated decreases in peroxisome proliferator-activated receptor γ (PPARγ) stimulate PASMC proliferation, and PPARγ in coordination with PPARγ coactivator 1α (PGC1α) regulates mitochondrial gene expression and biogenesis. To further examine the impact of decreases in PPARγ expression on human PASMC (HPASMC) mitochondrial function, we hypothesized that depletion of either PPARγ or PGC1α perturbs mitochondrial structure and function to stimulate PASMC proliferation. To test this hypothesis, HPASMCs were exposed to hypoxia and treated pharmacologically with the PPARγ antagonist GW9662 or with siRNA against PPARγ or PGC1α for 72 hours. HPASMC proliferation (cell counting), target mRNA levels (qRT-PCR), target protein levels (Western blotting), mitochondria-derived H2O2 (confocal immunofluorescence), mitochondrial mass and fragmentation, and mitochondrial bioenergetic profiling were determined. Hypoxia or knockdown of either PPARγ or PGC1α increased HPASMC proliferation, enhanced mitochondria-derived H2O2, decreased mitochondrial mass, stimulated mitochondrial fragmentation, and impaired mitochondrial bioenergetics. Taken together, these findings provide novel evidence that loss of PPARγ diminishes PGC1α and stimulates derangements in mitochondrial structure and function that cause PASMC proliferation. Overexpression of PGC1α reversed hypoxia-induced HPASMC derangements. This study identifies additional mechanistic underpinnings of PH, and provides support for the notion of activating PPARγ as a novel therapeutic strategy in PH.
Collapse
Affiliation(s)
- Samantha M Yeligar
- 1 Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Atlanta Veterans Affairs Medical Center, Decatur, Georgia.,2 Emory University, Atlanta, Georgia; and
| | - Bum-Yong Kang
- 1 Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Atlanta Veterans Affairs Medical Center, Decatur, Georgia.,2 Emory University, Atlanta, Georgia; and
| | - Kaiser M Bijli
- 1 Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Atlanta Veterans Affairs Medical Center, Decatur, Georgia.,2 Emory University, Atlanta, Georgia; and
| | - Jennifer M Kleinhenz
- 1 Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Atlanta Veterans Affairs Medical Center, Decatur, Georgia.,2 Emory University, Atlanta, Georgia; and
| | - Tamara C Murphy
- 1 Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Atlanta Veterans Affairs Medical Center, Decatur, Georgia.,2 Emory University, Atlanta, Georgia; and
| | - Gloria Torres
- 3 Division of Cardiology, Department of Medicine, Emory University, Atlanta, Georgia
| | - Alejandra San Martin
- 3 Division of Cardiology, Department of Medicine, Emory University, Atlanta, Georgia
| | - Roy L Sutliff
- 1 Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Atlanta Veterans Affairs Medical Center, Decatur, Georgia.,2 Emory University, Atlanta, Georgia; and
| | - C Michael Hart
- 1 Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Atlanta Veterans Affairs Medical Center, Decatur, Georgia.,2 Emory University, Atlanta, Georgia; and
| |
Collapse
|
15
|
Anti-hypoxic effect of dihydroartemisinin on pulmonary artery endothelial cells. Biochem Biophys Res Commun 2018; 506:840-846. [DOI: 10.1016/j.bbrc.2018.10.176] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 10/28/2018] [Indexed: 01/05/2023]
|
16
|
A Brief Overview of Nitric Oxide and Reactive Oxygen Species Signaling in Hypoxia-Induced Pulmonary Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 967:71-81. [PMID: 29047082 DOI: 10.1007/978-3-319-63245-2_6] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Pulmonary hypertension (PH) is characterized by increased vasoconstriction and smooth muscle cell hyperplasia driving pathological vascular remodeling of arterial vessels. In this short review, we discuss the primary source of reactive oxygen species (ROS) and nitric oxide (NO) relevant to PH and the mechanism by which dysregulation of their production contributes to PH. Specifically, hypoxia-induced PH is associated with diminished endothelial nitric oxide synthase (eNOS)-derived NO production and increased production of superoxide (O2•-) through eNOS uncoupling and defective mitochondrial respiration. This drives the inhibition of the NO/soluble guanylate cyclase (sGC) pathway and activation of the transcription factor hypoxia-inducible factor-1α (HIF-1α) with consequential dysregulation of the pulmonary vasculature. Therapeutics aimed at increasing NO or cGMP bioavailabilities are amenable to hypoxia disease-induced PH. Similarly, strategies targeting HIF-1α are now considered. Overall, pulmonary hypertension including hypoxia-induced PH offers unique opportunities for the rational development of therapeutics centered on modulating redox signaling.
Collapse
|
17
|
Frump A, Prewitt A, de Caestecker MP. BMPR2 mutations and endothelial dysfunction in pulmonary arterial hypertension (2017 Grover Conference Series). Pulm Circ 2018; 8:2045894018765840. [PMID: 29521190 PMCID: PMC5912278 DOI: 10.1177/2045894018765840] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 02/26/2018] [Indexed: 12/22/2022] Open
Abstract
Despite the discovery more than 15 years ago that patients with hereditary pulmonary arterial hypertension (HPAH) inherit BMP type 2 receptor ( BMPR2) mutations, it is still unclear how these mutations cause disease. In part, this is attributable to the rarity of HPAH and difficulty obtaining tissue samples from patients with early disease. However, in addition, limitations to the approaches used to study the effects of BMPR2 mutations on the pulmonary vasculature have restricted our ability to determine how individual mutations give rise to progressive pulmonary vascular pathology in HPAH. The importance of understanding the mechanisms by which BMPR2 mutations cause disease in patients with HPAH is underscored by evidence that there is reduced BMPR2 expression in patients with other, more common, non-hereditary form of PAH, and that restoration of BMPR2 expression reverses established disease in experimental models of pulmonary hypertension. In this paper, we focus on the effects on endothelial function. We discuss some of the controversies and challenges that have faced investigators exploring the role of BMPR2 mutations in HPAH, focusing specifically on the effects different BMPR2 mutation have on endothelial function, and whether there are qualitative differences between different BMPR2 mutations. We discuss evidence that BMPR2 signaling regulates a number of responses that may account for endothelial abnormalities in HPAH and summarize limitations of the models that are used to study these effects. Finally, we discuss evidence that BMPR2-dependent effects on endothelial metabolism provides a unifying explanation for the many of the BMPR2 mutation-dependent effects that have been described in patients with HPAH.
Collapse
Affiliation(s)
- Andrea Frump
- Division
of Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University
School of Medicine, Indianapolis, IN,
USA
| | | | - Mark P. de Caestecker
- Division
of Nephrology and Hypertension, Department of Medicine, Vanderbilt University
Medical center, Nashville, TN, USA
| |
Collapse
|
18
|
Telli G, Tel BC, Yersal N, Korkusuz P, Gumusel B. Effect of intermedin/adrenomedullin2 on the pulmonary vascular bed in hypoxia-induced pulmonary hypertensive rats. Life Sci 2018; 192:62-67. [DOI: 10.1016/j.lfs.2017.11.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 11/14/2017] [Accepted: 11/17/2017] [Indexed: 10/18/2022]
|
19
|
Kylhammar D, Rådegran G. The principal pathways involved in the in vivo modulation of hypoxic pulmonary vasoconstriction, pulmonary arterial remodelling and pulmonary hypertension. Acta Physiol (Oxf) 2017; 219:728-756. [PMID: 27381367 DOI: 10.1111/apha.12749] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 06/10/2016] [Accepted: 07/04/2016] [Indexed: 12/13/2022]
Abstract
Hypoxic pulmonary vasoconstriction (HPV) serves to optimize ventilation-perfusion matching in focal hypoxia and thereby enhances pulmonary gas exchange. During global hypoxia, however, HPV induces general pulmonary vasoconstriction, which may lead to pulmonary hypertension (PH), impaired exercise capacity, right-heart failure and pulmonary oedema at high altitude. In chronic hypoxia, generalized HPV together with hypoxic pulmonary arterial remodelling, contribute to the development of PH. The present article reviews the principal pathways in the in vivo modulation of HPV, hypoxic pulmonary arterial remodelling and PH with primary focus on the endothelin-1, nitric oxide, cyclooxygenase and adenine nucleotide pathways. In summary, endothelin-1 and thromboxane A2 may enhance, whereas nitric oxide and prostacyclin may moderate, HPV as well as hypoxic pulmonary arterial remodelling and PH. The production of prostacyclin seems to be coupled primarily to cyclooxygenase-1 in acute hypoxia, but to cyclooxygenase-2 in chronic hypoxia. The potential role of adenine nucleotides in modulating HPV is unclear, but warrants further study. Additional modulators of the pulmonary vascular responses to hypoxia may include angiotensin II, histamine, serotonin/5-hydroxytryptamine, leukotrienes and epoxyeicosatrienoic acids. Drugs targeting these pathways may reduce acute and/or chronic hypoxic PH. Endothelin receptor antagonists and phosphodiesterase-5 inhibitors may additionally improve exercise capacity in hypoxia. Importantly, the modulation of the pulmonary vascular responses to hypoxia varies between species and individuals, with hypoxic duration and age. The review also define how drugs targeting the endothelin-1, nitric oxide, cyclooxygenase and adenine nucleotide pathways may improve pulmonary haemodynamics, but also impair pulmonary gas exchange by interference with HPV in chronic lung diseases.
Collapse
Affiliation(s)
- D. Kylhammar
- Department of Clinical Sciences Lund, Cardiology; Faculty of Medicine; Lund University; Lund Sweden
- The Section for Heart Failure and Valvular Disease; VO Heart and Lung Medicine; Skåne University Hospital; Lund Sweden
| | - G. Rådegran
- Department of Clinical Sciences Lund, Cardiology; Faculty of Medicine; Lund University; Lund Sweden
- The Section for Heart Failure and Valvular Disease; VO Heart and Lung Medicine; Skåne University Hospital; Lund Sweden
| |
Collapse
|
20
|
Sang HY, Jin YL, Zhang WQ, Chen LB. Downregulation of microRNA-637 Increases Risk of Hypoxia-Induced Pulmonary Hypertension by Modulating Expression of Cyclin Dependent Kinase 6 (CDK6) in Pulmonary Smooth Muscle Cells. Med Sci Monit 2016; 22:4066-4072. [PMID: 27794186 PMCID: PMC5091202 DOI: 10.12659/msm.897254] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND The objective of this study was to investigate the molecular mechanism by which miR-637 interferes with the expression of CDK6, which contributes to the development of pulmonary hypertension (PH) with chronic obstructive pulmonary disease (COPD). MATERIAL AND METHODS We used an online miRNA database to identify CDK6 as a virtual target of miR-637, and validated the hypothesis using luciferase assay. Furthermore, we transfected SMCs with miR-637 mimics and inhibitor, and expression of CDK6 was determined using Western blot and real-time PCR. RESULTS In this study, we identified CDK6 as a target of miR-637 in smooth muscle cells (SMCs), and determined the expression of miR-637 in SMCs from PH patients with COPD and normal controls. We also identified the exact miR-637 binding site in the 3'UTR of CDK6 by using a luciferase reporter system. The mRNA and protein expression levels of CDK6 in SMCs from PH patients with COPD were clearly upregulated compared with the normal controls. Cells exposed to hypoxia also showed notably increased CKD6 mRNA and protein expression levels, and when treated with miR-637 or CDK6 siRNA, this increase in CKD6 expression was clearly attenuated. Additionally, cell viability and cell cycle analysis showed that hypoxia markedly increased viability of SMCs by causing an accumulation in S phase, which was relieved by the introduction of miR-637 or CDK6 siRNA. CONCLUSIONS Our study proved that the CDK6 gene is a target of miR-637, and demonstrated the regulatory association between miR-637 and CDK6, suggesting a possible therapeutic target for PH, especially in patients with COPD.
Collapse
Affiliation(s)
- Hai-Yan Sang
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Ying-Li Jin
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China (mainland)
| | - Wen-Qi Zhang
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Li-Bo Chen
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China (mainland)
| |
Collapse
|
21
|
Protective Roles of Endothelial AMP-Activated Protein Kinase Against Hypoxia-Induced Pulmonary Hypertension in Mice. Circ Res 2016; 119:197-209. [DOI: 10.1161/circresaha.115.308178] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 05/23/2016] [Indexed: 12/31/2022]
Abstract
Rationale:
Endothelial AMP-activated protein kinase (AMPK) plays an important role for vascular homeostasis, and its role is impaired by vascular inflammation. However, the role of endothelial AMPK in the pathogenesis of pulmonary arterial hypertension (PAH) remains to be elucidated.
Objective:
To determine the role of endothelial AMPK in the development of PAH.
Methods and Results:
Immunostaining showed that endothelial AMPK is downregulated in the pulmonary arteries of patients with PAH and hypoxia mouse model of pulmonary hypertension (PH). To elucidate the role of endothelial AMPK in PH, we used endothelial-specific AMPK-knockout mice (
eAMPK
–/–
), which were exposed to hypoxia. Under normoxic condition,
eAMPK
–/–
mice showed the normal morphology of pulmonary arteries compared with littermate controls (
eAMPK
flox/flox
). In contrast, development of hypoxia-induced PH was accelerated in
eAMPK
–/–
mice compared with controls. Furthermore, the exacerbation of PH in
eAMPK
–/–
mice was accompanied by reduced endothelial function, upregulation of growth factors, and increased proliferation of pulmonary artery smooth muscle cells. Importantly, conditioned medium from endothelial cells promoted pulmonary artery smooth muscle cell proliferation, which was further enhanced by the treatment with AMPK inhibitor. Serum levels of inflammatory cytokines, including tumor necrosis factor-α and interferon-γ were significantly increased in patients with PAH compared with healthy controls. Consistently, endothelial AMPK and cell proliferation were significantly reduced by the treatment with serum from patients with PAH compared with controls. Importantly, long-term treatment with metformin, an AMPK activator, significantly attenuated hypoxia-induced PH in mice.
Conclusions:
These results indicate that endothelial AMPK is a novel therapeutic target for the treatment of PAH.
Collapse
|
22
|
Dierick F, Héry T, Hoareau-Coudert B, Mougenot N, Monceau V, Claude C, Crisan M, Besson V, Dorfmüller P, Marodon G, Fadel E, Humbert M, Yaniz-Galende E, Hulot JS, Marazzi G, Sassoon D, Soubrier F, Nadaud S. Resident PW1+ Progenitor Cells Participate in Vascular Remodeling During Pulmonary Arterial Hypertension. Circ Res 2016; 118:822-33. [PMID: 26838788 DOI: 10.1161/circresaha.115.307035] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 01/12/2016] [Indexed: 12/20/2022]
Abstract
RATIONALE Pulmonary arterial hypertension is characterized by vascular remodeling and neomuscularization. PW1(+) progenitor cells can differentiate into smooth muscle cells (SMCs) in vitro. OBJECTIVE To determine the role of pulmonary PW1(+) progenitor cells in vascular remodeling characteristic of pulmonary arterial hypertension. METHODS AND RESULTS We investigated their contribution during chronic hypoxia-induced vascular remodeling in Pw1(nLacZ+/-) mouse expressing β-galactosidase in PW1(+) cells and in differentiated cells derived from PW1(+) cells. PW1(+) progenitor cells are present in the perivascular zone in rodent and human control lungs. Using progenitor markers, 3 distinct myogenic PW1(+) cell populations were isolated from the mouse lung of which 2 were significantly increased after 4 days of chronic hypoxia. The number of proliferating pulmonary PW1(+) cells and the proportion of β-gal(+) vascular SMC were increased, indicating a recruitment of PW1(+) cells and their differentiation into vascular SMC during early chronic hypoxia-induced neomuscularization. CXCR4 inhibition using AMD3100 prevented PW1(+) cells differentiation into SMC but did not inhibit their proliferation. Bone marrow transplantation experiments showed that the newly formed β-gal(+) SMC were not derived from circulating bone marrow-derived PW1(+) progenitor cells, confirming a resident origin of the recruited PW1(+) cells. The number of pulmonary PW1(+) cells was also increased in rats after monocrotaline injection. In lung from pulmonary arterial hypertension patients, PW1-expressing cells were observed in large numbers in remodeled vascular structures. CONCLUSIONS These results demonstrate the existence of a novel population of resident SMC progenitor cells expressing PW1 and participating in pulmonary hypertension-associated vascular remodeling.
Collapse
Affiliation(s)
- France Dierick
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Tiphaine Héry
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Bénédicte Hoareau-Coudert
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Nathalie Mougenot
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Virginie Monceau
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Caroline Claude
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Mihaela Crisan
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Vanessa Besson
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Peter Dorfmüller
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Gilles Marodon
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Elie Fadel
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Marc Humbert
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Elisa Yaniz-Galende
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Jean-Sébastien Hulot
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Giovanna Marazzi
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - David Sassoon
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Florent Soubrier
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Sophie Nadaud
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.).
| |
Collapse
|
23
|
Chai S, Wang W, Liu J, Guo H, Zhang Z, Wang C, Wang J. Leptin knockout attenuates hypoxia-induced pulmonary arterial hypertension by inhibiting proliferation of pulmonary arterial smooth muscle cells. Transl Res 2015; 166:772-82. [PMID: 26470682 DOI: 10.1016/j.trsl.2015.09.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 09/07/2015] [Accepted: 09/18/2015] [Indexed: 11/19/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a fatal disease characterized by excessive vascular smooth muscle cells proliferation in small pulmonary arteries, leading to elevation of pulmonary vascular resistance with consequent right ventricular (RV) failure and death. Recently, emerging evidence has shown that leptin signaling is involved in different cardiac pathologies; however, the role of leptin remains limited in the setting of PAH. Thus, in this study, we tested the hypothesis of direct involvement of leptin in the development of PAH. Our data show that leptin activity in plasma and protein level in the lung were higher in hypoxia- and monocrotaline-induced PAH models compared with control animals. Wild-type (WT) and C57BL/6J-Lep(ob) (ob/ob) male mice were exposed to normobaric hypoxia (10% O(2)) or normoxia (21% O(2)). After 2 and 4 weeks of chronic hypoxia exposure, WT mice developed PAH as reflected by the increased values of RV systolic pressure, RV hypertrophy index, the medial wall thickness of pulmonary arterioles, and muscularization of pulmonary arterioles. And, all these alterations were attenuated in ob/ob mice treated with hypoxia. Leptin could stimulate the proliferation of pulmonary arterial smooth muscle cells (PASMCs) by activating extracellular signal-regulated kinase (ERK), signal transducer and activator of transcription 3 (STAT3), and Akt pathways. These data suggest that the leptin signaling pathway is crucial for the development of PAH. Leptin activates ERK, STAT, and Akt pathways and subsequently PASMCs proliferation, providing new mechanistic information about hypoxia-induced PAH.
Collapse
Affiliation(s)
- SanBao Chai
- Department of Physiology, Capital Medical University, Beijing, P.R. China
| | - Wang Wang
- Department of Physiology, Capital Medical University, Beijing, P.R. China
| | - Jie Liu
- Department of Physiology, Capital Medical University, Beijing, P.R. China
| | - Huan Guo
- Department of Physiology, Capital Medical University, Beijing, P.R. China
| | - ZhiFei Zhang
- Department of Physiology, Capital Medical University, Beijing, P.R. China
| | - Chen Wang
- Department of Respiration, China-Japan Friendship Hospital, Beijing, P.R. China
| | - Jun Wang
- Department of Physiology, Capital Medical University, Beijing, P.R. China.
| |
Collapse
|
24
|
Xing Y, Zheng X, Li G, Liao L, Cao W, Xing H, Shen T, Sun L, Yang B, Zhu D. MicroRNA-30c contributes to the development of hypoxia pulmonary hypertension by inhibiting platelet-derived growth factor receptor β expression. Int J Biochem Cell Biol 2015; 64:155-66. [PMID: 25882492 DOI: 10.1016/j.biocel.2015.04.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 03/25/2015] [Accepted: 04/02/2015] [Indexed: 10/23/2022]
Abstract
Pulmonary arterial hypertension (PAH) is characterized by excessive proliferation and resistance to apoptosis of pulmonary artery smooth muscle cells (PASMCs). MicroRNAs have been implicated in the regulation of cell proliferation and might be implicated in the etiology of PAH. Data from in vivo and in vitro cell culture models showed that hypoxia inhibits microRNA-30c (miR-30c) expression in PASMCs. Inhibition of miR-30c by either hypoxia or AMO-30c results in PASMC proliferation (cell viability, 5-bromo-2-deoxyuridine (BrdU) incorporation, proliferating cell nuclear antigen, Ki67, and tubulin polymerization) and the inhibition of apoptosis (cell cycle progression, Cyclin A and Cyclin D, and TUNEL staining). Moreover, down-regulation of miR-30c also results in the phenotype switch from contractile to synthetic PASMC (SM22α and Calponin, osteopontin expression, and wound healing assay). In contrast, these effects were reversed by the application of an miR-30c mimetic under hypoxic conditions. Mechanically, miR-30c inhibited the platelet-derived growth factor receptor β (PDGFRβ) expression by directly binding to the 3' untranslated region of PDGFRβ mRNA (luciferase reporter assays, and PDGFRβ-masking antisense oligodeoxynucleotides). Pharmacological inhibition of PDGFR by AG-1296 displayed similar effects to the miR-30c mimetic. These data suggest that the down-regulation of miR-30c accounts for the up-regulation of PDGFRβ expression, and subsequent activation of PDGF signaling results in the hypoxia-induced PASMC proliferation and phenotype switching. Therefore, increasing miR-30c expression levels could be explored as a potential new therapy for hypoxia-induced PAH.
Collapse
Affiliation(s)
- Yan Xing
- Department of Pharmacology, College of Basic Medicine, Harbin Medical University (Daqing), Daqing 163319, China
| | - Xiaodong Zheng
- Department of Pathophysiology, College of Basic Medicine, Harbin Medical University (Daqing), Daqing 163319, China
| | - Guixia Li
- Department of Pharmacology, College of Basic Medicine, Harbin Medical University (Daqing), Daqing 163319, China
| | - Lin Liao
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University (Daqing), Daqing 163319, China; Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin 150081, China
| | - Weiwei Cao
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University (Daqing), Daqing 163319, China; Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin 150081, China
| | - Hao Xing
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University (Daqing), Daqing 163319, China; Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin 150081, China
| | - Tingting Shen
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University (Daqing), Daqing 163319, China; Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin 150081, China
| | - Lihua Sun
- Department of Pharmacology, Harbin Medical University (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin, 150081, China
| | - Baofeng Yang
- Department of Pharmacology, Harbin Medical University (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin, 150081, China
| | - Daling Zhu
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University (Daqing), Daqing 163319, China; Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin 150081, China.
| |
Collapse
|
25
|
Abstract
SIGNIFICANCE The pulmonary circulation is a low-pressure, low-resistance, highly compliant vasculature. In contrast to the systemic circulation, it is not primarily regulated by a central nervous control mechanism. The regulation of resting membrane potential due to ion channels is of integral importance in the physiology and pathophysiology of the pulmonary vasculature. RECENT ADVANCES Redox-driven ion conductance changes initiated by direct oxidation, nitration, and S-nitrosylation of the cysteine thiols and indirect phosphorylation of the threonine and serine residues directly affect pulmonary vascular tone. CRITICAL ISSUES Molecular mechanisms of changes in ion channel conductance, especially the identification of the sites of action, are still not fully elucidated. FUTURE DIRECTIONS Further investigation of the interaction between redox status and ion channel gating, especially the physiological significance of S-glutathionylation and S-nitrosylation, could result in a better understanding of the physiological and pathophysiological importance of these mediators in general and the implications of such modifications in cellular functions and related diseases and their importance for targeted treatment strategies.
Collapse
Affiliation(s)
- Andrea Olschewski
- 1 Ludwig Boltzmann Institute for Lung Vascular Research , Graz, Austria
| | | |
Collapse
|
26
|
Zhou S, Li M, Zeng D, Sun G, Zhou J, Wang R. Effects of basic fibroblast growth factor and cyclin D1 on cigarette smoke-induced pulmonary vascular remodeling in rats. Exp Ther Med 2014; 9:33-38. [PMID: 25452772 PMCID: PMC4247281 DOI: 10.3892/etm.2014.2044] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 07/31/2014] [Indexed: 01/10/2023] Open
Abstract
Cigarette smoking may contribute to pulmonary hypertension in chronic obstructive pulmonary disease by resulting in pulmonary vascular remodeling that involves pulmonary artery smooth muscle cell proliferation. This study investigated the effects of basic fibroblast growth factor (bFGF) and cyclin D1 on the pulmonary vascular remodeling in smoking-exposed rats. Twenty-four male Wistar rats were randomly divided into four groups. Three tobacco-exposed groups were exposed to the smoke produced by 20 cigarettes for 60 min, twice a day for two, four or eight weeks, and the control group were exposed to fresh air. The expression of bFGF and cyclin D1 in the pulmonary arterial smooth muscle cells were determined using immunohistochemistry. Quantitative polymerase chain reaction was conducted to determine the expression levels of bFGF and cyclin D1 mRNA. In addition, the expression of bFGF and cyclin D1 proteins was evaluated by western blotting. The expression of bFGF and cyclin D1 at the mRNA and protein levels was shown to increase with the duration of smoke exposure (P<0.05). The correlation analysis indicated the expression of bFGF and cyclin D1 was positively associated with the pulmonary vessel wall thickness. The expression of bFGF was positively associated with that of cyclin D1. Collectively, the data demonstrated that the upregulation of bFGF and cyclin D1 occurred in rats subjected to smoke exposure, which may be associated with the abnormal proliferation of the smooth muscle cells in the pulmonary arteries.
Collapse
Affiliation(s)
- Sijing Zhou
- Hefei Prevention and Treatment Center for Occupational Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China ; Department of Respiratory Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Min Li
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Daxiong Zeng
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Gengyun Sun
- Department of Respiratory Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Junsheng Zhou
- Hefei Prevention and Treatment Center for Occupational Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Ran Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| |
Collapse
|
27
|
|
28
|
Dubois M, Delannoy E, Duluc L, Closs E, Li H, Toussaint C, Gadeau AP, Gödecke A, Freund-Michel V, Courtois A, Marthan R, Savineau JP, Muller B. Biopterin metabolism and eNOS expression during hypoxic pulmonary hypertension in mice. PLoS One 2013; 8:e82594. [PMID: 24312428 PMCID: PMC3842263 DOI: 10.1371/journal.pone.0082594] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Accepted: 11/04/2013] [Indexed: 11/18/2022] Open
Abstract
Tetrahydrobiopterin (BH4), which fosters the formation of and stabilizes endothelial NO synthase (eNOS) as an active dimer, tightly regulates eNOS coupling / uncoupling. Moreover, studies conducted in genetically-modified models demonstrate that BH4 pulmonary deficiency is a key determinant in the pathogenesis of pulmonary hypertension. The present study thus investigates biopterin metabolism and eNOS expression, as well as the effect of sepiapterin (a precursor of BH4) and eNOS gene deletion, in a mice model of hypoxic pulmonary hypertension. In lungs, chronic hypoxia increased BH4 levels and eNOS expression, without modifying dihydrobiopterin (BH2, the oxidation product of BH4) levels, GTP cyclohydrolase-1 or dihydrofolate reductase expression (two key enzymes regulating BH4 availability). In intrapulmonary arteries, chronic hypoxia also increased expression of eNOS, but did not induce destabilisation of eNOS dimers into monomers. In hypoxic mice, sepiapterin prevented increase in right ventricular systolic pressure and right ventricular hypertrophy, whereas it modified neither remodelling nor alteration in vasomotor responses (hyper-responsiveness to phenylephrine, decrease in endothelium-dependent relaxation to acetylcholine) in intrapulmonary arteries. Finally, deletion of eNOS gene partially prevented hypoxia-induced increase in right ventricular systolic pressure, right ventricular hypertrophy and remodelling of intrapulmonary arteries. Collectively, these data demonstrate the absence of BH4/BH2 changes and eNOS dimer destabilisation, which may induce eNOS uncoupling during hypoxia-induced pulmonary hypertension. Thus, even though eNOS gene deletion and sepiapterin treatment exert protective effects on hypoxia-induced pulmonary vascular remodelling, increase on right ventricular pressure and / or right ventricular hypertrophy, these effects appear unrelated to biopterin-dependent eNOS uncoupling within pulmonary vasculature of hypoxic wild-type mice.
Collapse
Affiliation(s)
- Mathilde Dubois
- University Bordeaux, Centre de recherche Cardio-Thoracique de Bordeaux, U1045, Bordeaux, France
- INSERM, Centre de recherche Cardio-Thoracique de Bordeaux, U1045, Bordeaux, France
| | - Estelle Delannoy
- University Bordeaux, Centre de recherche Cardio-Thoracique de Bordeaux, U1045, Bordeaux, France
- INSERM, Centre de recherche Cardio-Thoracique de Bordeaux, U1045, Bordeaux, France
- CHU de, Bordeaux, Bordeaux, France
| | - Lucie Duluc
- University Bordeaux, Centre de recherche Cardio-Thoracique de Bordeaux, U1045, Bordeaux, France
- INSERM, Centre de recherche Cardio-Thoracique de Bordeaux, U1045, Bordeaux, France
| | - Ellen Closs
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Huige Li
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | | | | | - Axel Gödecke
- Institute of Cardiovascular Physiology, Heinrich-Heine University, Düsseldorf, Germany
| | - Véronique Freund-Michel
- University Bordeaux, Centre de recherche Cardio-Thoracique de Bordeaux, U1045, Bordeaux, France
- INSERM, Centre de recherche Cardio-Thoracique de Bordeaux, U1045, Bordeaux, France
| | - Arnaud Courtois
- University Bordeaux, Centre de recherche Cardio-Thoracique de Bordeaux, U1045, Bordeaux, France
- INSERM, Centre de recherche Cardio-Thoracique de Bordeaux, U1045, Bordeaux, France
| | - Roger Marthan
- University Bordeaux, Centre de recherche Cardio-Thoracique de Bordeaux, U1045, Bordeaux, France
- INSERM, Centre de recherche Cardio-Thoracique de Bordeaux, U1045, Bordeaux, France
- CHU de, Bordeaux, Bordeaux, France
| | - Jean-Pierre Savineau
- University Bordeaux, Centre de recherche Cardio-Thoracique de Bordeaux, U1045, Bordeaux, France
- INSERM, Centre de recherche Cardio-Thoracique de Bordeaux, U1045, Bordeaux, France
| | - Bernard Muller
- University Bordeaux, Centre de recherche Cardio-Thoracique de Bordeaux, U1045, Bordeaux, France
- INSERM, Centre de recherche Cardio-Thoracique de Bordeaux, U1045, Bordeaux, France
| |
Collapse
|
29
|
Tian L, Wang Z, Lakes RS, Chesler NC. Comparison of approaches to quantify arterial damping capacity from pressurization tests on mouse conduit arteries. J Biomech Eng 2013; 135:54504. [PMID: 24231965 DOI: 10.1115/1.4024135] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 04/04/2013] [Indexed: 11/08/2022]
Abstract
Large conduit arteries are not purely elastic, but viscoelastic, which affects not only the mechanical behavior but also the ventricular afterload. Different hysteresis loops such as pressure-diameter, pressure-luminal cross-sectional area (LCSA), and stress-strain have been used to estimate damping capacity, which is associated with the ratio of the dissipated energy to the stored energy. Typically, linearized methods are used to calculate the damping capacity of arteries despite the fact that arteries are nonlinearly viscoelastic. The differences in the calculated damping capacity between these hysteresis loops and the most common linear and correct nonlinear methods have not been fully examined. The purpose of this study was thus to examine these differences and to determine a preferred approach for arterial damping capacity estimation. Pressurization tests were performed on mouse extralobar pulmonary and carotid arteries in their physiological pressure ranges with pressure (P) and outer diameter (OD) measured. The P-inner diameter (ID), P-stretch, P-Almansi strain, P-Green strain, P-LCSA, and stress-strain loops (including the Cauchy and Piola-Kirchhoff stresses and Almansi and Green strains) were calculated using the P-OD data and arterial geometry. Then, the damping capacity was calculated from these loops with both linear and nonlinear methods. Our results demonstrate that the linear approach provides a reasonable approximation of damping capacity for all of the loops except the Cauchy stress-Almansi strain, for which the estimate of damping capacity was significantly smaller (22 ± 8% with the nonlinear method and 31 ± 10% with the linear method). Between healthy and diseased extralobar pulmonary arteries, both methods detected significant differences. However, the estimate of damping capacity provided by the linear method was significantly smaller (27 ± 11%) than that of the nonlinear method. We conclude that all loops except the Cauchy stress-Almansi strain loop can be used to estimate artery wall damping capacity in the physiological pressure range and the nonlinear method is recommended over the linear method.
Collapse
|
30
|
Klinger JR, Abman SH, Gladwin MT. Nitric oxide deficiency and endothelial dysfunction in pulmonary arterial hypertension. Am J Respir Crit Care Med 2013; 188:639-46. [PMID: 23822809 DOI: 10.1164/rccm.201304-0686pp] [Citation(s) in RCA: 155] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Nitric oxide (NO) signaling plays a major role in modulating vascular tone and remodeling in the pulmonary circulation, but its role in the pathogenesis of pulmonary vascular diseases is still not completely understood. Numerous abnormalities of NO synthesis and signaling have been identified in animal models of pulmonary vascular disease and in humans with pulmonary hypertension. Many of these abnormalities have become targets of new therapies for the treatment of pulmonary hypertension. However, it is unclear to what extent alterations in NO signaling contribute to pulmonary hypertensive responses or merely reflect abnormalities induced by the underlying disease. This perspective examines the current understanding of altered NO signaling in pulmonary hypertensive diseases and discusses how these alterations may contribute to the pathogenesis of pulmonary hypertension. The efficacy and limitations of presently available therapies for pulmonary hypertension that target NO signaling are reviewed along with an update on investigational therapies that use this pathway to reverse pulmonary hypertensive changes.
Collapse
Affiliation(s)
- James R Klinger
- 1 Division of Pulmonary, Sleep, and Critical Care Medicine, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island
| | | | | |
Collapse
|
31
|
Abstract
Genetically modified mouse models have unparalleled power to determine the mechanisms behind different processes involved in the molecular and physiologic etiology of various classes of human pulmonary hypertension (PH). Processes known to be involved in PH for which there are extensive mouse models available include the following: (1) Regulation of vascular tone through secreted vasoactive factors; (2) regulation of vascular tone through potassium and calcium channels; (3) regulation of vascular remodeling through alteration in metabolic processes, either through alteration in substrate usage or through circulating factors; (4) spontaneous vascular remodeling either before or after development of elevated pulmonary pressures; and (5) models in which changes in tone and remodeling are primarily driven by inflammation. PH development in mice is of necessity faster and with different physiologic ramifications than found in human disease, and so mice make poor models of natural history of PH. However, transgenic mouse models are a perfect tool for studying the processes involved in pulmonary vascular function and disease, and can effectively be used to test interventions designed against particular molecular pathways and processes involved in disease.
Collapse
Affiliation(s)
- Mita Das
- Department of Internal Medicine, University of Arkansas Medical Sciences, Little Rock, Arkansas, USA
| | | | | | | |
Collapse
|
32
|
Angelini DJ, Su Q, Yamaji-Kegan K, Fan C, Skinner JT, Poloczek A, El-Haddad H, Cheadle C, Johns RA. Hypoxia-induced mitogenic factor (HIMF/FIZZ1/RELMα) in chronic hypoxia- and antigen-mediated pulmonary vascular remodeling. Respir Res 2013; 14:1. [PMID: 23289668 PMCID: PMC3547770 DOI: 10.1186/1465-9921-14-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Accepted: 12/12/2012] [Indexed: 12/14/2022] Open
Abstract
Background Both chronic hypoxia and allergic inflammation induce vascular remodeling in the lung, but only chronic hypoxia appears to cause PH. We investigate the nature of the vascular remodeling and the expression and role of hypoxia-induced mitogenic factor (HIMF/FIZZ1/RELMα) in explaining this differential response. Methods We induced pulmonary vascular remodeling through either chronic hypoxia or antigen sensitization and challenge. Mice were evaluated for markers of PH and pulmonary vascular remodeling throughout the lung vascular bed as well as HIMF expression and genomic analysis of whole lung. Results Chronic hypoxia increased both mean pulmonary artery pressure (mPAP) and right ventricular (RV) hypertrophy; these changes were associated with increased muscularization and thickening of small pulmonary vessels throughout the lung vascular bed. Allergic inflammation, by contrast, had minimal effect on mPAP and produced no RV hypertrophy. Only peribronchial vessels were significantly thickened, and vessels within the lung periphery did not become muscularized. Genomic analysis revealed that HIMF was the most consistently upregulated gene in the lungs following both chronic hypoxia and antigen challenge. HIMF was upregulated in the airway epithelial and inflammatory cells in both models, but only chronic hypoxia induced HIMF upregulation in vascular tissue. Conclusions The results show that pulmonary vascular remodeling in mice induced by chronic hypoxia or antigen challenge is associated with marked increases in HIMF expression. The lack of HIMF expression in the vasculature of the lung and no vascular remodeling in the peripheral resistance vessels of the lung is likely to account for the failure to develop PH in the allergic inflammation model.
Collapse
Affiliation(s)
- Daniel J Angelini
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Cho WK, Lee CM, Kang MJ, Huang Y, Giordano FJ, Lee PJ, Trow TK, Homer RJ, Sessa WC, Elias JA, Lee CG. IL-13 receptor α2-arginase 2 pathway mediates IL-13-induced pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2012; 304:L112-24. [PMID: 23125252 DOI: 10.1152/ajplung.00101.2012] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Although previous literature suggests that interleukin (IL)-13, a T-helper type 2 cell effector cytokine, might be involved in the pathogenesis of pulmonary hypertension (PH), direct proof is lacking. Furthermore, a potential mechanism underlying IL-13-induced PH has never been explored. This study's goal was to investigate the role and mechanism of IL-13 in the pathogenesis of PH. Lung-specific IL-13-overexpressing transgenic (Tg) mice were examined for hemodynamic changes and pulmonary vascular remodeling. IL-13 Tg mice spontaneously developed PH phenotype by the age of 2 mo with increased expression and activity of arginase 2 (Arg2). The role of Arg2 in the development of IL-13-stimulated PH was further investigated using Arg2 and IL-13 receptor α2 (Rα2) null mutant mice and the small-interfering RNA (siRNA)-silencing approach in vivo and in vitro, respectively. IL-13-stimulated medial thickening of pulmonary arteries and right ventricle systolic pressure were significantly decreased in the IL-13 Tg mice with Arg2 null mutation. On the other hand, the production of nitric oxide was further increased in the lungs of these mice. In our in vitro evaluations, the recombinant IL-13 treatment significantly enhanced the proliferation of human pulmonary artery smooth muscle cells in an Arg2-dependent manner. The IL-13-stimulated cellular proliferation and the expression of Arg2 in hpaSMC were markedly decreased with IL-13Rα2 siRNA silencing. Our studies demonstrate that IL-13 contributes to the development of PH via an IL-13Rα2-Arg2-dependent pathway. The intervention of this pathway could be a potential therapeutic target in pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Won-Kyung Cho
- Yale University School of Medicine, Dept. of Internal Medicine, New Haven, CT 06520-8057, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
NOing the heart: role of nitric oxide synthase-3 in heart development. Differentiation 2012; 84:54-61. [PMID: 22579300 DOI: 10.1016/j.diff.2012.04.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Revised: 04/03/2012] [Accepted: 04/10/2012] [Indexed: 01/30/2023]
Abstract
Congenital heart disease is the most common birth defect in humans. Identifying factors that are critical to embryonic heart development could further our understanding of the disease and lead to new strategies of its prevention and treatment. Nitric oxide synthase-3 (NOS3) or endothelial nitric oxide synthase (eNOS) is known for many important biological functions including vasodilation, vascular homeostasis and angiogenesis. Over the past decade, studies from our lab and others have shown that NOS3 is required during heart development. More specifically, deficiency in NOS3 results in congenital septal defects, cardiac hypertrophy and postnatal heart failure. In addition, NOS3 is pivotal to the morphogenesis of major coronary arteries and myocardial capillary development. Interestingly, these effects of NOS3 are mediated through induction of transcription and growth factors that are crucial in the formation of coronary arteries. Finally, deficiency in NOS3 results in high incidences of bicuspid aortic valves, a disease in humans that often leads to complications with age including aortic valve stenosis or regurgitation, endocarditis, aortic aneurysm formation, and aortic dissection. In summary, these data suggest NOS3 plays a critical role in embryonic heart development and morphogenesis of coronary arteries and aortic valves.
Collapse
|
35
|
Ho JJD, Man HSJ, Marsden PA. Nitric oxide signaling in hypoxia. J Mol Med (Berl) 2012; 90:217-31. [DOI: 10.1007/s00109-012-0880-5] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Revised: 02/03/2012] [Accepted: 02/06/2012] [Indexed: 01/06/2023]
|
36
|
Rabieyousefi M, Soroosh P, Satoh K, Date F, Ishii N, Yamashita M, Oka M, McMurtry IF, Shimokawa H, Nose M, Sugamura K, Ono M. Indispensable roles of OX40L-derived signal and epistatic genetic effect in immune-mediated pathogenesis of spontaneous pulmonary hypertension. BMC Immunol 2011; 12:67. [PMID: 22171643 PMCID: PMC3269997 DOI: 10.1186/1471-2172-12-67] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Accepted: 12/15/2011] [Indexed: 12/20/2022] Open
Abstract
Background Pulmonary hypertension (PH) refers to a spectrum of diseases with elevated pulmonary artery pressure. Pulmonary arterial hypertension (PAH) is a disease category that clinically presents with severe PH and that is histopathologically characterized by the occlusion of pulmonary arterioles, medial muscular hypertrophy, and/or intimal fibrosis. PAH occurs with a secondary as well as a primary onset. Secondary PAH is known to be complicated with immunological disorders. The aim of the present study is to histopathologically and genetically characterize a new animal model of PAH and clarify the role of OX40 ligand in the pathogenesis of PAH. Results Spontaneous onset of PAH was stably identified in mice with immune abnormality because of overexpression of the tumor necrosis factor (TNF) family molecule OX40 ligand (OX40L). Histopathological and physical examinations revealed the onset of PAH-like disorders in the C57BL/6 (B6) strain of OX40L transgenic mice (B6.TgL). Comparative analysis performed using different strains of transgenic mice showed that this onset depends on the presence of OX40L in the B6 genetic background. Genetic analyses demonstrated a susceptibility locus of a B6 allele to this onset on chromosome 5. Immunological analyses revealed that the excessive OX40 signals in TgL mice attenuates expansion of regulatory T cells the B6 genetic background, suggesting an impact of the B6 genetic background on the differentiation of regulatory T cells. Conclusion Present findings suggest a role for the OX40L-derived immune response and epistatic genetic effect in immune-mediated pathogenesis of PAH.
Collapse
Affiliation(s)
- Moloud Rabieyousefi
- Department of Pathology, Tohoku University Graduate School of Medicine, 2-1 Seiryo, Aoba-ku, Sendai, Miyagi 980-8575 Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Hebbel RP. Reconstructing sickle cell disease: a data-based analysis of the "hyperhemolysis paradigm" for pulmonary hypertension from the perspective of evidence-based medicine. Am J Hematol 2011; 86:123-54. [PMID: 21264896 DOI: 10.1002/ajh.21952] [Citation(s) in RCA: 120] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The "hyperhemolytic paradigm" (HHP) posits that hemolysis in sickle disease sequentially and causally establishes increased cell-free plasma Hb, consumption of NO, a state of NO biodeficiency, endothelial dysfunction, and a high prevalence of pulmonary hypertension. The basic science underpinning this concept has added an important facet to the complexity of vascular pathobiology in sickle disease, and clinical research has identified worrisome clinical issues. However, this critique identifies and explains a number of significant concerns about the various HHP component tenets. In addressing these issues, this report presents: a very brief history of the HHP, an integrated synthesis of mechanisms underlying sickle hemolysis, a review of the evidentiary value of hemolysis biomarkers, an examination of evidence bearing on existence of a hyperhemolytic subgroup, and a series of questions that should naturally be applied to the HHP if it is examined using critical thinking skills, the fundamental basis of evidence-based medicine. The veracity of different HHP tenets is found to vary from true, to weakly supported, to demonstrably false. The thesis is developed that the HHP has misidentified the mechanism and clinical significance of its findings. The extant research questions identified by these analyses are delineated, and a conservative, evidence-based approach is suggested for application in clinical medicine.
Collapse
Affiliation(s)
- Robert P. Hebbel
- Department of Medicine, Division of Hematology‐Oncology‐Transplantation, Vascular Biology Center, University of Minnesota Medical School, Minneapolis, Minnesota
| |
Collapse
|
38
|
Rus A, Peinado MA, Castro L, Del Moral ML. Lung eNOS and iNOS are reoxygenation time-dependent upregulated after acute hypoxia. Anat Rec (Hoboken) 2010; 293:1089-98. [PMID: 20225207 DOI: 10.1002/ar.21141] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Nitric oxide plays a critical role in many physiological and physiopathological processes in the lung. Changes in the NO/NOS (Nitric Oxide/Nitric Oxide Synthase) system after hypoxia situations remain controversial in this organ, so that the aim of this work is to perform a complete study of this system in the hypoxic lung after different reoxygenation times ranging from 0 h to 5 days posthypoxia. This is a novel follow-up study carried out in Wistar rats submitted for 30 min to acute hypobaric hypoxia. We measured endothelial and inducible NOS (eNOS, iNOS) mRNA and protein expression, location, and in situ NOS activity as well as nitrated protein expression and location. In addition, NO levels were indirectly quantified (NOx) as well as the apoptosis level. Results showed an increase in eNOS mRNA, protein, activity as well as eNOS positive immunostaining at 0 h posthypoxia, coinciding with raised NOx levels. Contrary, iNOS, nitrated protein expression and apoptosis level augmented during the final reoxygenation times. The lung NO/NOS system provokes two responses to the hypoxia/reoxygenation processes: (i) eNOS is responsible of the immediate response, producing NO, which causes vasodilation and bronchodilation, and (ii) iNOS is related to the second late response, which seems to be involved in some of the deleterious consequences that hypoxia induces in the lung.
Collapse
Affiliation(s)
- Alma Rus
- Department of Experimental Biology, University of Jaén, Jaén, Spain
| | | | | | | |
Collapse
|
39
|
Angelini DJ, Su Q, Kolosova IA, Fan C, Skinner JT, Yamaji-Kegan K, Collector M, Sharkis SJ, Johns RA. Hypoxia-induced mitogenic factor (HIMF/FIZZ1/RELM alpha) recruits bone marrow-derived cells to the murine pulmonary vasculature. PLoS One 2010; 5:e11251. [PMID: 20582166 PMCID: PMC2889818 DOI: 10.1371/journal.pone.0011251] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2010] [Accepted: 05/25/2010] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Pulmonary hypertension (PH) is a disease of multiple etiologies with several common pathological features, including inflammation and pulmonary vascular remodeling. Recent evidence has suggested a potential role for the recruitment of bone marrow-derived (BMD) progenitor cells to this remodeling process. We recently demonstrated that hypoxia-induced mitogenic factor (HIMF/FIZZ1/RELM alpha) is chemotactic to murine bone marrow cells in vitro and involved in pulmonary vascular remodeling in vivo. METHODOLOGY/PRINCIPAL FINDINGS We used a mouse bone marrow transplant model in which lethally irradiated mice were rescued with bone marrow transplanted from green fluorescent protein (GFP)(+) transgenic mice to determine the role of HIMF in recruiting BMD cells to the lung vasculature during PH development. Exposure to chronic hypoxia and pulmonary gene transfer of HIMF were used to induce PH. Both models resulted in markedly increased numbers of BMD cells in and around the pulmonary vasculature; in several neomuscularized small (approximately 20 microm) capillary-like vessels, an entirely new medial wall was made up of these cells. We found these GFP(+) BMD cells to be positive for stem cell antigen-1 and c-kit, but negative for CD31 and CD34. Several of the GFP(+) cells that localized to the pulmonary vasculature were alpha-smooth muscle actin(+) and localized to the media layer of the vessels. This finding suggests that these cells are of mesenchymal origin and differentiate toward myofibroblast and vascular smooth muscle. Structural location in the media of small vessels suggests a functional role in the lung vasculature. To examine a potential mechanism for HIMF-dependent recruitment of mesenchymal stem cells to the pulmonary vasculature, we performed a cell migration assay using cultured human mesenchymal stem cells (HMSCs). The addition of recombinant HIMF induced migration of HMSCs in a phosphoinosotide-3-kinase-dependent manner. CONCLUSIONS/SIGNIFICANCE These results demonstrate HIMF-dependent recruitment of BMD mesenchymal-like cells to the remodeling pulmonary vasculature.
Collapse
Affiliation(s)
- Daniel J. Angelini
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Qingning Su
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Irina A. Kolosova
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Chunling Fan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - John T. Skinner
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Kazuyo Yamaji-Kegan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Michael Collector
- Department of Oncology and Cancer Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Saul J. Sharkis
- Department of Oncology and Cancer Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Roger A. Johns
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| |
Collapse
|
40
|
Anderson L, Lowery JW, Frank DB, Novitskaya T, Jones M, Mortlock DP, Chandler RL, de Caestecker MP. Bmp2 and Bmp4 exert opposing effects in hypoxic pulmonary hypertension. Am J Physiol Regul Integr Comp Physiol 2009; 298:R833-42. [PMID: 20042692 DOI: 10.1152/ajpregu.00534.2009] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The bone morphogenetic protein (BMP) type 2 receptor ligand, Bmp2, is upregulated in the peripheral pulmonary vasculature during hypoxia-induced pulmonary hypertension (PH). This contrasts with the expression of Bmp4, which is expressed in respiratory epithelia throughout the lung. Unlike heterozygous null Bmp4 mice (Bmp4(LacZ/+)), which are protected from the development of hypoxic PH, mice that are heterozygous null for Bmp2 (Bmp2(+/-)) develop more severe hypoxic PH than their wild-type littermates. This is associated with reduced endothelial nitric oxide synthase (eNOS) expression and activity in the pulmonary vasculature of hypoxic Bmp2(+/-) but not Bmp4(LacZ/+) mutant mice. Furthermore, exogenous BMP2 upregulates eNOS expression and activity in intrapulmonary artery and pulmonary endothelial cell preparations, indicating that eNOS is a target of Bmp2 signaling in the pulmonary vasculature. Together, these data demonstrate that Bmp2 and Bmp4 exert opposing roles in hypoxic PH and suggest that the protective effects of Bmp2 are mediated by increasing eNOS expression and activity in the hypoxic pulmonary vasculature.
Collapse
Affiliation(s)
- Lynda Anderson
- Department of Medicine, Vanderbilt Univ. Medical Center, Division of Nephrology, Nashville, TN 37232, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Weissmann N, Hackemack S, Dahal BK, Pullamsetti SS, Savai R, Mittal M, Fuchs B, Medebach T, Dumitrascu R, Eickels MV, Ghofrani HA, Seeger W, Grimminger F, Schermuly RT. The soluble guanylate cyclase activator HMR1766 reverses hypoxia-induced experimental pulmonary hypertension in mice. Am J Physiol Lung Cell Mol Physiol 2009; 297:L658-65. [PMID: 19617308 DOI: 10.1152/ajplung.00189.2009] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Severe pulmonary hypertension (PH) is a disabling disease with high mortality, characterized by pulmonary vascular remodeling and right heart hypertrophy. In mice with PH induced by chronic hypoxia, we examined the acute and chronic effects of the soluble guanylate cyclase (sGC) activator HMR1766 on hemodynamics and pulmonary vascular remodeling. In isolated perfused mouse lungs from control animals, HMR1766 dose-dependently inhibited the pressor response of acute hypoxia. This dose-response curve was shifted leftward when the effects of HMR1766 were investigated in isolated lungs from chronic hypoxic animals for 21 days at 10% oxygen. Mice exposed for 21 or 35 days to chronic hypoxia developed PH, right heart hypertrophy, and pulmonary vascular remodeling. Treatment with HMR1766 (10 mg x kg(-1) x day(-1)), after full establishment of PH from day 21 to day 35, significantly reduced PH, as measured continuously by telemetry. In addition, right ventricular (RV) hypertrophy and structural remodeling of the lung vasculature were reduced. Pharmacological activation of oxidized sGC partially reverses hemodynamic and structural changes in chronic hypoxia-induced experimental PH.
Collapse
Affiliation(s)
- Norbert Weissmann
- Univ. of Giessen Lung Center Medical Clinic II/V, Klinikstr. 36, 35392 Giessen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Ghofrani HA, Barst RJ, Benza RL, Champion HC, Fagan KA, Grimminger F, Humbert M, Simonneau G, Stewart DJ, Ventura C, Rubin LJ. Future perspectives for the treatment of pulmonary arterial hypertension. J Am Coll Cardiol 2009; 54:S108-S117. [PMID: 19555854 PMCID: PMC4883573 DOI: 10.1016/j.jacc.2009.04.014] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2009] [Accepted: 04/16/2009] [Indexed: 02/02/2023]
Abstract
Over the past 2 decades, pulmonary arterial hypertension has evolved from a uniformly fatal condition to a chronic, manageable disease in many cases, the result of unparalleled development of new therapies and advances in early diagnosis. However, none of the currently available therapies is curative, so the search for new treatment strategies continues. With a deeper understanding of the genetics and the molecular mechanisms of pulmonary vascular disorders, we are now at the threshold of entering a new therapeutic era. Our working group addressed what can be expected in the near future. The topics span the understanding of genetic variations, novel antiproliferative treatments, the role of stem cells, the right ventricle as a therapeutic target, and strategies and challenges for the translation of novel experimental findings into clinical practice.
Collapse
Affiliation(s)
- Hossein A Ghofrani
- University of Giessen Lung Center, University Hospital Giessen and Marburg GmbH, Giessen, Germany; Medical Clinic IV and V, University Hospital Giessen and Marburg GmbH, Giessen, Germany.
| | | | - Raymond L Benza
- Drexel University College of Medicine, Section of Heart Failure, Transplantation, and Pulmonary Hypertension Program, Allegheny General Hospital, Pittsburgh, Pennsylvania
| | - Hunter C Champion
- Division of Cardiology, Johns Hopkins University, Baltimore, Maryland
| | - Karen A Fagan
- Division of Pulmonary and Critical Care Medicine, University of South Alabama, Mobile, Alabama
| | - Friedrich Grimminger
- University of Giessen Lung Center, University Hospital Giessen and Marburg GmbH, Giessen, Germany; Medical Clinic IV and V, University Hospital Giessen and Marburg GmbH, Giessen, Germany
| | - Marc Humbert
- Université Paris-Sud, Service de Pneumologie, Hôpital Antoine-Béclère, Clamart, France
| | - Gérald Simonneau
- Université Paris-Sud, Service de Pneumologie, Hôpital Antoine-Béclère, Clamart, France
| | | | - Carlo Ventura
- University of Bologna, Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, Institute of Cardiology, S.Orsola-Malpighi Hospital, Bologna, Italy
| | - Lewis J Rubin
- University of California, San Diego, Medical Center, La Jolla, California
| |
Collapse
|
43
|
Tsai EJ, Kass DA. Cyclic GMP signaling in cardiovascular pathophysiology and therapeutics. Pharmacol Ther 2009; 122:216-38. [PMID: 19306895 PMCID: PMC2709600 DOI: 10.1016/j.pharmthera.2009.02.009] [Citation(s) in RCA: 316] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2009] [Accepted: 02/19/2009] [Indexed: 02/07/2023]
Abstract
Cyclic guanosine 3',5'-monophosphate (cGMP) mediates a wide spectrum of physiologic processes in multiple cell types within the cardiovascular system. Dysfunctional signaling at any step of the cascade - cGMP synthesis, effector activation, or catabolism - have been implicated in numerous cardiovascular diseases, ranging from hypertension to atherosclerosis to cardiac hypertrophy and heart failure. In this review, we outline each step of the cGMP signaling cascade and discuss its regulation and physiologic effects within the cardiovascular system. In addition, we illustrate how cGMP signaling becomes dysregulated in specific cardiovascular disease states. The ubiquitous role cGMP plays in cardiac physiology and pathophysiology presents great opportunities for pharmacologic modulation of the cGMP signal in the treatment of cardiovascular diseases. We detail the various therapeutic interventional strategies that have been developed or are in development, summarizing relevant preclinical and clinical studies.
Collapse
Affiliation(s)
- Emily J Tsai
- Division of Cardiology, Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, Maryland 21205, USA
| | | |
Collapse
|
44
|
Kimura S, Egashira K, Chen L, Nakano K, Iwata E, Miyagawa M, Tsujimoto H, Hara K, Morishita R, Sueishi K, Tominaga R, Sunagawa K. Nanoparticle-Mediated Delivery of Nuclear Factor κB Decoy Into Lungs Ameliorates Monocrotaline-Induced Pulmonary Arterial Hypertension. Hypertension 2009; 53:877-83. [DOI: 10.1161/hypertensionaha.108.121418] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Pulmonary arterial hypertension (PAH) is an intractable disease of the small pulmonary artery that involves multiple inflammatory factors. We hypothesized that a redox-sensitive transcription factor, nuclear factor κB (NF-κB), which regulates important inflammatory cytokines, plays a pivotal role in PAH. We investigated the activity of NF-κB in explanted lungs from patients with PAH and in a rat model of PAH. We also examined a nanotechnology-based therapeutic intervention in the rat model. Immunohistochemistry results indicated that the activity of NF-κB increased in small pulmonary arterial lesions and alveolar macrophages in lungs from patients with PAH compared with lungs from control patients. In a rat model of monocrotaline-induced PAH, single intratracheal instillation of polymeric nanoparticles (NPs) resulted in delivery of NPs into lungs for ≤14 days postinstillation. The NP-mediated NF-κB decoy delivery into lungs prevented monocrotaline-induced NF-κB activation. Blockade of NF-κB by NP-mediated delivery of the NF-κB decoy attenuated inflammation and proliferation and, thus, attenuated the development of PAH and pulmonary arterial remodeling induced by monocrotaline. Treatment with the NF-κB decoy NP 3 weeks after monocrotaline injection improved the survival rate as compared with vehicle administration. In conclusion, these data suggest that NF-κB plays a primary role in the pathogenesis of PAH and, thus, represent a new target for therapeutic intervention in PAH. This nanotechnology platform may be developed as a novel molecular approach for treatment of PAH in the future.
Collapse
Affiliation(s)
- Satoshi Kimura
- From the Departments of Surgery (S.K., R.T.), Cardiovascular Medicine (K.E., L.C., K.N., E.I., M.M., K. Sunagawa), and Pathology (K. Sueishi), Graduate School of Medical Science, Kyushu University, Fukuoka; Hosokawa Powder Technology Research Institute (H.T., K.H.), Osaka; and Division of Clinical Gene Therapy (R.M.), Osaka University Medical School, Osaka, Japan
| | - Kensuke Egashira
- From the Departments of Surgery (S.K., R.T.), Cardiovascular Medicine (K.E., L.C., K.N., E.I., M.M., K. Sunagawa), and Pathology (K. Sueishi), Graduate School of Medical Science, Kyushu University, Fukuoka; Hosokawa Powder Technology Research Institute (H.T., K.H.), Osaka; and Division of Clinical Gene Therapy (R.M.), Osaka University Medical School, Osaka, Japan
| | - Ling Chen
- From the Departments of Surgery (S.K., R.T.), Cardiovascular Medicine (K.E., L.C., K.N., E.I., M.M., K. Sunagawa), and Pathology (K. Sueishi), Graduate School of Medical Science, Kyushu University, Fukuoka; Hosokawa Powder Technology Research Institute (H.T., K.H.), Osaka; and Division of Clinical Gene Therapy (R.M.), Osaka University Medical School, Osaka, Japan
| | - Kaku Nakano
- From the Departments of Surgery (S.K., R.T.), Cardiovascular Medicine (K.E., L.C., K.N., E.I., M.M., K. Sunagawa), and Pathology (K. Sueishi), Graduate School of Medical Science, Kyushu University, Fukuoka; Hosokawa Powder Technology Research Institute (H.T., K.H.), Osaka; and Division of Clinical Gene Therapy (R.M.), Osaka University Medical School, Osaka, Japan
| | - Eiko Iwata
- From the Departments of Surgery (S.K., R.T.), Cardiovascular Medicine (K.E., L.C., K.N., E.I., M.M., K. Sunagawa), and Pathology (K. Sueishi), Graduate School of Medical Science, Kyushu University, Fukuoka; Hosokawa Powder Technology Research Institute (H.T., K.H.), Osaka; and Division of Clinical Gene Therapy (R.M.), Osaka University Medical School, Osaka, Japan
| | - Miho Miyagawa
- From the Departments of Surgery (S.K., R.T.), Cardiovascular Medicine (K.E., L.C., K.N., E.I., M.M., K. Sunagawa), and Pathology (K. Sueishi), Graduate School of Medical Science, Kyushu University, Fukuoka; Hosokawa Powder Technology Research Institute (H.T., K.H.), Osaka; and Division of Clinical Gene Therapy (R.M.), Osaka University Medical School, Osaka, Japan
| | - Hiroyuki Tsujimoto
- From the Departments of Surgery (S.K., R.T.), Cardiovascular Medicine (K.E., L.C., K.N., E.I., M.M., K. Sunagawa), and Pathology (K. Sueishi), Graduate School of Medical Science, Kyushu University, Fukuoka; Hosokawa Powder Technology Research Institute (H.T., K.H.), Osaka; and Division of Clinical Gene Therapy (R.M.), Osaka University Medical School, Osaka, Japan
| | - Kaori Hara
- From the Departments of Surgery (S.K., R.T.), Cardiovascular Medicine (K.E., L.C., K.N., E.I., M.M., K. Sunagawa), and Pathology (K. Sueishi), Graduate School of Medical Science, Kyushu University, Fukuoka; Hosokawa Powder Technology Research Institute (H.T., K.H.), Osaka; and Division of Clinical Gene Therapy (R.M.), Osaka University Medical School, Osaka, Japan
| | - Ryuichi Morishita
- From the Departments of Surgery (S.K., R.T.), Cardiovascular Medicine (K.E., L.C., K.N., E.I., M.M., K. Sunagawa), and Pathology (K. Sueishi), Graduate School of Medical Science, Kyushu University, Fukuoka; Hosokawa Powder Technology Research Institute (H.T., K.H.), Osaka; and Division of Clinical Gene Therapy (R.M.), Osaka University Medical School, Osaka, Japan
| | - Katsuo Sueishi
- From the Departments of Surgery (S.K., R.T.), Cardiovascular Medicine (K.E., L.C., K.N., E.I., M.M., K. Sunagawa), and Pathology (K. Sueishi), Graduate School of Medical Science, Kyushu University, Fukuoka; Hosokawa Powder Technology Research Institute (H.T., K.H.), Osaka; and Division of Clinical Gene Therapy (R.M.), Osaka University Medical School, Osaka, Japan
| | - Ryuji Tominaga
- From the Departments of Surgery (S.K., R.T.), Cardiovascular Medicine (K.E., L.C., K.N., E.I., M.M., K. Sunagawa), and Pathology (K. Sueishi), Graduate School of Medical Science, Kyushu University, Fukuoka; Hosokawa Powder Technology Research Institute (H.T., K.H.), Osaka; and Division of Clinical Gene Therapy (R.M.), Osaka University Medical School, Osaka, Japan
| | - Kenji Sunagawa
- From the Departments of Surgery (S.K., R.T.), Cardiovascular Medicine (K.E., L.C., K.N., E.I., M.M., K. Sunagawa), and Pathology (K. Sueishi), Graduate School of Medical Science, Kyushu University, Fukuoka; Hosokawa Powder Technology Research Institute (H.T., K.H.), Osaka; and Division of Clinical Gene Therapy (R.M.), Osaka University Medical School, Osaka, Japan
| |
Collapse
|
45
|
Yildiz P. Molecular mechanisms of pulmonary hypertension. Clin Chim Acta 2009; 403:9-16. [PMID: 19361468 DOI: 10.1016/j.cca.2009.01.018] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2008] [Revised: 01/18/2009] [Accepted: 01/23/2009] [Indexed: 12/11/2022]
Abstract
The pathogenesis of pulmonary arterial hypertension (PAH) is complex, involving multiple modulating genes and environmental factors. Multifactorial impairment of the physiologic balance can lead to vasoconstriction, vascular smooth muscle cell and endothelial cell proliferation/fibrosis, inflammation, remodeling and in-situ thrombosis. These are the likely mechanisms that lead to narrowing of the vessel followed by progressive increase in pulmonary vascular resistance and the clinical manifestations of pulmonary hypertension. Subsequently, major goal of the therapy is to avoid acute pulmonary vasoconstriction, halt the progression of vascular remodeling, and reverse the early vascular remodeling if possible. Recently published data addressing certain molecular mechanisms for pathogenesis of PAH have led to the successful therapeutic interventions. This review will focus on the common and critical molecular pathways including genetic basis of the development of PAH that on the whole may be new targets for therapeutic interventions.
Collapse
Affiliation(s)
- Pinar Yildiz
- Department of Pulmonology, Yedikule Chest Disease and Surgery Training and Research Hospital, Zeytinburnu Istanbul, Turkey.
| |
Collapse
|
46
|
Angelini DJ, Su Q, Yamaji-Kegan K, Fan C, Skinner JT, Champion HC, Crow MT, Johns RA. Hypoxia-induced mitogenic factor (HIMF/FIZZ1/RELMalpha) induces the vascular and hemodynamic changes of pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2009; 296:L582-93. [PMID: 19136574 DOI: 10.1152/ajplung.90526.2008] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Pulmonary hypertension (PH) is a serious disease of multiple etiologies mediated by hypoxia, immune stimuli, and elevated pulmonary pressure that leads to vascular thickening and eventual right heart failure. In a chronic hypoxia model of PH, we previously reported the induction of a novel pleiotropic cytokine, hypoxia-induced mitogenic factor (HIMF), that exhibits mitogenic, vasculogenic, contractile, and chemokine properties during PH-associated vascular remodeling. To examine the role of HIMF in hypoxia-induced vascular remodeling, we performed in vivo knockdown of HIMF using short hairpin RNA directed at rat HIMF in the chronic hypoxia model of PH. Knockdown of HIMF partially blocked increases in mean pulmonary artery pressure, pulmonary vascular resistance, right heart hypertrophy, and vascular remodeling caused by chronic hypoxia. To demonstrate a direct role for HIMF in the mechanism of PH development, we performed HIMF-gene transfer into the lungs of rats using a HIMF-expressing adeno-associated virus (AAV). AAV-HIMF alone caused development of PH similar to that of chronic hypoxia with increased mean pulmonary artery pressure and pulmonary vascular resistance, right heart hypertrophy, and neomuscularization and thickening of small pulmonary arterioles. The findings suggest that HIMF represents a critical cytokine-like growth factor in the development of PH.
Collapse
Affiliation(s)
- Daniel J Angelini
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Mehta VB, Zhou Y, Radulescu A, Besner GE. HB-EGF stimulates eNOS expression and nitric oxide production and promotes eNOS dependent angiogenesis. Growth Factors 2008; 26:301-15. [PMID: 18925469 DOI: 10.1080/08977190802393596] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Heparin-binding EGF-like growth factor (HB-EGF) is a member of the epidermal growth factor (EGF) family of ligands that is expressed by many cell types including endothelial cells. We have previously shown that HB-EGF stimulates angiogenesis in vitro in human umbilical vein endothelial cells (HUVEC). Nitric oxide (NO) derived from endothelial nitric oxide synthase (eNOS) is an important regulator of angiogenesis. However, the role of HB-EGF in regulation of eNOS has not yet been investigated. Whether HB-EGF-induced endothelial cell migration and vascular network formation are mediated via production of NO from eNOS is also unknown. To address these questions, we stimulated HUVEC with HB-EGF and evaluated the expression of eNOS at the mRNA and protein levels. HB-EGF significantly upregulated expression of eNOS mRNA, stimulated eNOS protein production, and increased NO release from HUVEC. HB-EGF phosphorylated eNOS in a phosphatidylinositol 3-kinase (PI3K) dependent fashion, and stimulated in vitro angiogenesis. eNOS siRNA inhibited HB-EGF-stimulated HUVEC migration in a scratch assay. NG-nitro-L-arginine-methyl-ester (L-NAME) and L-N5-(1-lminoethyl)ornithine,dihydochloride (L-NIO) (specific inhibitors of eNOS) also abolished HB-EGF-induced HUVEC migration and angiogenesis. More importantly, we found that HB-EGF also promotes angiogenesis in vivo in the Marigel plug assay. Lastly, inhibition of the p38 MAPK pathway enhanced HB-EGF-induced EC migration and angiogenesis. We conclude that HB-EGF, through its interaction with EGF receptors (EGFR), stimulates eNOS activation and NO production via a PI3K-dependent pathway. Thus, activation of eNOS appears to be one of the key signaling pathways necessary for HB-EGF mediated angiogenesis. These novel findings highlight an important role for HB-EGF as a regulator of endothelial cell function.
Collapse
Affiliation(s)
- Veela B Mehta
- Department of Pediatric Surgery, The Research Institute at Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, OH 43205, USA.
| | | | | | | |
Collapse
|
48
|
Abstract
Homeostasis in the pulmonary vasculature is maintained by the actions of vasoactive compounds, including nitric oxide (NO). NO is critical for normal development of the pulmonary vasculature and continues to mediate normal vasoregulation in adulthood. Loss of NO bioavailability is one component of the endothelial dysfunction and vascular pathology found in pulmonary hypertension (PH). A broad research effort continues to expand our understanding of the control of NO production and NO signaling and has generated novel theories on the importance of pulmonary NO production in the control of the systemic vasculature. This understanding has led to exciting developments in our ability to treat PH, including inhaled NO and phosphodiesterase inhibitors, and to several promising directions for future therapies using nitric oxide-donor compounds, stimulators of soluble guanylate cyclase, progenitor cells expressing NO synthase (NOS), and NOS gene manipulation.
Collapse
Affiliation(s)
- Matthew P Coggins
- Cardiology Division, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA.
| | | |
Collapse
|
49
|
Vermeersch P, Buys E, Pokreisz P, Marsboom G, Ichinose F, Sips P, Pellens M, Gillijns H, Swinnen M, Graveline A, Collen D, Dewerchin M, Brouckaert P, Bloch KD, Janssens S. Soluble guanylate cyclase-alpha1 deficiency selectively inhibits the pulmonary vasodilator response to nitric oxide and increases the pulmonary vascular remodeling response to chronic hypoxia. Circulation 2007; 116:936-43. [PMID: 17679618 DOI: 10.1161/circulationaha.106.677245] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Nitric oxide (NO) activates soluble guanylate cyclase (sGC), a heterodimer composed of alpha- and beta-subunits, to produce cGMP. NO reduces pulmonary vascular remodeling, but the role of sGC in vascular responses to acute and chronic hypoxia remains incompletely elucidated. We therefore studied pulmonary vascular responses to acute and chronic hypoxia in wild-type (WT) mice and mice with a nonfunctional alpha1-subunit (sGCalpha1-/-). METHODS AND RESULTS sGCalpha1-/- mice had significantly reduced lung sGC activity and vasodilator-stimulated phosphoprotein phosphorylation. Right ventricular systolic pressure did not differ between genotypes at baseline and increased similarly in WT (22+/-2 to 34+/-2 mm Hg) and sGCalpha1-/- (23+/-2 to 34+/-1 mm Hg) mice in response to acute hypoxia. Inhaled NO (40 ppm) blunted the increase in right ventricular systolic pressure in WT mice (22+/-2 to 24+/-2 mm Hg, P<0.01 versus hypoxia without NO) but not in sGCalpha1-/- mice (22+/-1 to 33+/-1 mm Hg) and was accompanied by a significant rise in lung cGMP content only in WT mice. In contrast, the NO-donor sodium nitroprusside (1.5 mg/kg) decreased systemic blood pressure similarly in awake WT and sGCalpha1-/- mice as measured by telemetry (-37+/-2 versus -42+/-4 mm Hg). After 3 weeks of hypoxia, the increases in right ventricular systolic pressure, right ventricular hypertrophy, and muscularization of intra-acinar pulmonary vessels were 43%, 135%, and 46% greater, respectively, in sGCalpha1-/- than in WT mice (P<0.01). Increased remodeling in sGCalpha1-/- mice was associated with an increased frequency of 5'-bromo-deoxyuridine-positive vessels after 1 and 3 weeks (P<0.01 versus WT). CONCLUSIONS Deficiency of sGCalpha1 does not alter hypoxic pulmonary vasoconstriction. sGCalpha1 is essential for NO-mediated pulmonary vasodilation and limits chronic hypoxia-induced pulmonary vascular remodeling.
Collapse
Affiliation(s)
- Pieter Vermeersch
- Center for Transgene Technology and Gene Therapy, VIB, Leuven, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Sehgal PB, Mukhopadhyay S. Pulmonary arterial hypertension: a disease of tethers, SNAREs and SNAPs? Am J Physiol Heart Circ Physiol 2007; 293:H77-85. [PMID: 17416597 DOI: 10.1152/ajpheart.01386.2006] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Histological and electron microscopic studies over the past four decades have highlighted "plump," "enlarged" endothelial, smooth muscle, and fibroblastic cellular elements with increased endoplasmic reticulum, Golgi stacks, and vacuolation in pulmonary arterial lesions in human and in experimental (hypoxia and monocrotaline) pulmonary arterial hypertension. However, the contribution of disrupted intracellular membrane trafficking in the pathobiology of this disease has received insufficient attention. Recent studies suggest a pathogenetic role of the disruption of intracellular trafficking of vasorelevant proteins and cell-surface receptors in the development of this disease. The purpose of this essay is to highlight the molecular regulation of vesicular trafficking by membrane tethers, SNAREs and SNAPs, and to suggest how their dysfunction, directly and/or indirectly, might contribute to development of pulmonary arterial hypertension in experimental models and in humans, including that due to mutations in bone morphogenetic receptor type 2.
Collapse
Affiliation(s)
- Pravin B Sehgal
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY 10595, USA.
| | | |
Collapse
|