101
|
Lawrie A. A report on the use of animal models and phenotyping methods in pulmonary hypertension research. Pulm Circ 2014; 4:2-9. [PMID: 25006416 DOI: 10.1086/674886] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 09/04/2013] [Indexed: 11/04/2022] Open
Abstract
The failure to translate positive results from preclinical studies into new clinical therapies is a major problem throughout medical research. Specifically, in pulmonary hypertension, numerous research studies have shown beneficial effects of new therapies in experimental models, but these have largely failed to translate into clinical benefit in human trials. This is undoubtedly due, at least in part, to inadequacies of the models, but while monogenic animal models will never fully recapitulate human disease, they do still provide the best platform on which to test novel therapeutic agents. In the postgenomic era, there is emphasis on a greater understanding of disease pathogenesis, which has subsequently led to the development of both new targets and new models in which to test them. The evolution of new technologies means that we are now better equipped to phenotype these models, but the level of detail provided varies dramatically throughout the literature. However, subtle variances in experimental methods can make comparing data/findings between research laboratories difficult and are a possible contributing factor to variance between preclinical and clinical data. The aim of this report was to capture information on current practice for use of the growing array of animal models, to help movement toward developing guidelines and standards for the "best" use of animal models of pulmonary hypertension.
Collapse
Affiliation(s)
- A Lawrie
- Department of Cardiovascular Science, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
102
|
Los patrones de vascularización pulmonar en la radiografía simple de tórax. RADIOLOGIA 2014; 56:346-56. [DOI: 10.1016/j.rx.2013.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 09/19/2013] [Accepted: 10/09/2013] [Indexed: 11/18/2022]
|
103
|
White K, Dempsie Y, Caruso P, Wallace E, McDonald RA, Stevens H, Hatley ME, Van Rooij E, Morrell NW, MacLean MR, Baker AH. Endothelial Apoptosis in Pulmonary Hypertension Is Controlled by a microRNA/Programmed Cell Death 4/Caspase-3 Axis. Hypertension 2014; 64:185-94. [DOI: 10.1161/hypertensionaha.113.03037] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Kevin White
- From the Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (K.W., Y.D., P.C., E.W., R.A.M., H.S., M.R.M., A.H.B.); Solid Tumor Division, St. Jude Children’s Research Hospital, Memphis, TN (M.E.H.); MiRagen Therapeutics, Boulder, CO (E.V.R.); and Division of Respiratory Medicine, Addenbrooke’s Hospital, University of Cambridge, School of Clinical Medicine, Cambridge, United Kingdom (N.W.M.)
| | - Yvonne Dempsie
- From the Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (K.W., Y.D., P.C., E.W., R.A.M., H.S., M.R.M., A.H.B.); Solid Tumor Division, St. Jude Children’s Research Hospital, Memphis, TN (M.E.H.); MiRagen Therapeutics, Boulder, CO (E.V.R.); and Division of Respiratory Medicine, Addenbrooke’s Hospital, University of Cambridge, School of Clinical Medicine, Cambridge, United Kingdom (N.W.M.)
| | - Paola Caruso
- From the Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (K.W., Y.D., P.C., E.W., R.A.M., H.S., M.R.M., A.H.B.); Solid Tumor Division, St. Jude Children’s Research Hospital, Memphis, TN (M.E.H.); MiRagen Therapeutics, Boulder, CO (E.V.R.); and Division of Respiratory Medicine, Addenbrooke’s Hospital, University of Cambridge, School of Clinical Medicine, Cambridge, United Kingdom (N.W.M.)
| | - Emma Wallace
- From the Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (K.W., Y.D., P.C., E.W., R.A.M., H.S., M.R.M., A.H.B.); Solid Tumor Division, St. Jude Children’s Research Hospital, Memphis, TN (M.E.H.); MiRagen Therapeutics, Boulder, CO (E.V.R.); and Division of Respiratory Medicine, Addenbrooke’s Hospital, University of Cambridge, School of Clinical Medicine, Cambridge, United Kingdom (N.W.M.)
| | - Robert A. McDonald
- From the Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (K.W., Y.D., P.C., E.W., R.A.M., H.S., M.R.M., A.H.B.); Solid Tumor Division, St. Jude Children’s Research Hospital, Memphis, TN (M.E.H.); MiRagen Therapeutics, Boulder, CO (E.V.R.); and Division of Respiratory Medicine, Addenbrooke’s Hospital, University of Cambridge, School of Clinical Medicine, Cambridge, United Kingdom (N.W.M.)
| | - Hannah Stevens
- From the Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (K.W., Y.D., P.C., E.W., R.A.M., H.S., M.R.M., A.H.B.); Solid Tumor Division, St. Jude Children’s Research Hospital, Memphis, TN (M.E.H.); MiRagen Therapeutics, Boulder, CO (E.V.R.); and Division of Respiratory Medicine, Addenbrooke’s Hospital, University of Cambridge, School of Clinical Medicine, Cambridge, United Kingdom (N.W.M.)
| | - Mark E. Hatley
- From the Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (K.W., Y.D., P.C., E.W., R.A.M., H.S., M.R.M., A.H.B.); Solid Tumor Division, St. Jude Children’s Research Hospital, Memphis, TN (M.E.H.); MiRagen Therapeutics, Boulder, CO (E.V.R.); and Division of Respiratory Medicine, Addenbrooke’s Hospital, University of Cambridge, School of Clinical Medicine, Cambridge, United Kingdom (N.W.M.)
| | - Eva Van Rooij
- From the Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (K.W., Y.D., P.C., E.W., R.A.M., H.S., M.R.M., A.H.B.); Solid Tumor Division, St. Jude Children’s Research Hospital, Memphis, TN (M.E.H.); MiRagen Therapeutics, Boulder, CO (E.V.R.); and Division of Respiratory Medicine, Addenbrooke’s Hospital, University of Cambridge, School of Clinical Medicine, Cambridge, United Kingdom (N.W.M.)
| | - Nicholas W. Morrell
- From the Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (K.W., Y.D., P.C., E.W., R.A.M., H.S., M.R.M., A.H.B.); Solid Tumor Division, St. Jude Children’s Research Hospital, Memphis, TN (M.E.H.); MiRagen Therapeutics, Boulder, CO (E.V.R.); and Division of Respiratory Medicine, Addenbrooke’s Hospital, University of Cambridge, School of Clinical Medicine, Cambridge, United Kingdom (N.W.M.)
| | - Margaret R. MacLean
- From the Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (K.W., Y.D., P.C., E.W., R.A.M., H.S., M.R.M., A.H.B.); Solid Tumor Division, St. Jude Children’s Research Hospital, Memphis, TN (M.E.H.); MiRagen Therapeutics, Boulder, CO (E.V.R.); and Division of Respiratory Medicine, Addenbrooke’s Hospital, University of Cambridge, School of Clinical Medicine, Cambridge, United Kingdom (N.W.M.)
| | - Andrew H. Baker
- From the Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (K.W., Y.D., P.C., E.W., R.A.M., H.S., M.R.M., A.H.B.); Solid Tumor Division, St. Jude Children’s Research Hospital, Memphis, TN (M.E.H.); MiRagen Therapeutics, Boulder, CO (E.V.R.); and Division of Respiratory Medicine, Addenbrooke’s Hospital, University of Cambridge, School of Clinical Medicine, Cambridge, United Kingdom (N.W.M.)
| |
Collapse
|
104
|
Wagenaar GTM, Sengers RMA, Laghmani EH, Chen X, Lindeboom MPHA, Roks AJM, Folkerts G, Walther FJ. Angiotensin II type 2 receptor ligand PD123319 attenuates hyperoxia-induced lung and heart injury at a low dose in newborn rats. Am J Physiol Lung Cell Mol Physiol 2014; 307:L261-72. [PMID: 24951776 DOI: 10.1152/ajplung.00345.2013] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Intervening in angiotensin (Ang)-II type 2 receptor (AT2) signaling may have therapeutic potential for bronchopulmonary dysplasia (BPD) by attenuating lung inflammation and preventing arterial hypertension (PAH)-induced right ventricular hypertrophy (RVH). We first investigated the role of AT2 inhibition with PD123319 (0.5 and 2 mg·kg(-1)·day(-1)) on the beneficial effect of AT2 agonist LP2-3 (5 μg/kg twice a day) on RVH in newborn rats with hyperoxia-induced BPD. Next we determined the cardiopulmonary effects of PD123319 (0.1 mg·kg(-1)·day(-1)) in two models: early treatment during continuous exposure to hyperoxia for 10 days and late treatment starting on day 6 in rat pups exposed postnatally to hyperoxia for 9 days, followed by a 9-day recovery period in room air. Parameters investigated included lung and heart histopathology, fibrin deposition, vascular leakage, and differential mRNA expression. Ten days of coadministration of LP2-3 and PD123319 abolished the beneficial effects of LP2-3 on RVH in experimental BPD. In the early treatment model PD123319 attenuated cardiopulmonary injury by reducing alveolar septal thickness, pulmonary influx of inflammatory cells, including macrophages and neutrophils, medial wall thickness of small arterioles, and extravascular collagen III deposition, and by preventing RVH. In the late treatment model PD123319 diminished PAH and RVH, demonstrating that PAH is reversible in the neonatal period. At high concentrations PD123319 blocks the beneficial effects of the AT2-agonist LP2-3 on RVH. At low concentrations PD123319 attenuates cardiopulmonary injury by reducing pulmonary inflammation and fibrosis and preventing PAH-induced RVH but does not affect alveolar and vascular development in newborn rats with experimental BPD.
Collapse
Affiliation(s)
- Gerry T M Wagenaar
- Department of Pediatrics, Division of Neonatology, Leiden University Medical Center, Leiden, The Netherlands;
| | - Rozemarijn M A Sengers
- Department of Pediatrics, Division of Neonatology, Leiden University Medical Center, Leiden, The Netherlands
| | - El Houari Laghmani
- Department of Pediatrics, Division of Neonatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Xueyu Chen
- Department of Pediatrics, Division of Neonatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Melissa P H A Lindeboom
- Department of Pediatrics, Division of Neonatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Anton J M Roks
- Division of Vascular Disease and Pharmacology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Gert Folkerts
- Department of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands; and
| | - Frans J Walther
- Department of Pediatrics, Division of Neonatology, Leiden University Medical Center, Leiden, The Netherlands; Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California
| |
Collapse
|
105
|
Ciuclan L, Sheppard K, Dong L, Sutton D, Duggan N, Hussey M, Simmons J, Morrell NW, Jarai G, Edwards M, Dubois G, Thomas M, Van Heeke G, England K. Treatment with anti-gremlin 1 antibody ameliorates chronic hypoxia/SU5416-induced pulmonary arterial hypertension in mice. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 183:1461-73. [PMID: 24160323 DOI: 10.1016/j.ajpath.2013.07.017] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 06/20/2013] [Accepted: 07/12/2013] [Indexed: 01/05/2023]
Abstract
The expression of the bone morphogenetic protein antagonist, Gremlin 1, was recently shown to be increased in the lungs of pulmonary arterial hypertension patients, and in response to hypoxia. Gremlin 1 released from the vascular endothelium may inhibit endogenous bone morphogenetic protein signaling and contribute to the development of pulmonary arterial hypertension. Here, we investigate the impact of Gremlin 1 inhibition in disease after exposure to chronic hypoxia/SU5416 in mice. We investigated the effects of an anti-Gremlin 1 monoclonal antibody in the chronic hypoxia/SU5416 murine model of pulmonary arterial hypertension. Chronic hypoxic/SU5416 exposure of mice induced upregulation of Gremlin 1 mRNA in lung and right ventricle tissue compared with normoxic controls. Prophylactic treatment with an anti-Gremlin 1 neutralizing mAb reduced the hypoxic/SU5416-dependent increase in pulmonary vascular remodeling and right ventricular hypertrophy. Importantly, therapeutic treatment with an anti-Gremlin 1 antibody also reduced pulmonary vascular remodeling and right ventricular hypertrophy indicating a role for Gremlin 1 in the progression of the disease. We conclude that Gremlin 1 plays a role in the development and progression of pulmonary arterial hypertension in the murine hypoxia/SU5416 model, and that Gremlin 1 is a potential therapeutic target for pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Loredana Ciuclan
- Respiratory Disease Area, Novartis Institutes for BioMedical Research, Horsham, West Sussex, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
106
|
Ciracì R, Tirone G, Scaglione F. The impact of drug–drug interactions on pulmonary arterial hypertension therapy. Pulm Pharmacol Ther 2014; 28:1-8. [DOI: 10.1016/j.pupt.2014.01.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 01/08/2014] [Accepted: 01/12/2014] [Indexed: 02/05/2023]
|
107
|
Jasińska-Stroschein M, Orszulak-Michalak D. The current approach into signaling pathways in pulmonary arterial hypertension and their implication in novel therapeutic strategies. Pharmacol Rep 2014; 66:552-64. [PMID: 24948054 DOI: 10.1016/j.pharep.2014.04.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 04/02/2014] [Accepted: 04/07/2014] [Indexed: 02/01/2023]
Abstract
Many mediators and signaling pathways, with their downstream effectors, have been implicated in the pathogenesis of pulmonary hypertension. Currently approved drugs, representing an option of specific therapy, target NO, prostacyclin or ET-1 pathways and provide a significant improvement in the symptomatic status of patients and a slower rate of clinical deterioration. However, despite such improvements in the treatment, PAH remains a chronic disease without a cure, the mortality associated with PAH remains high and effective therapeutic regimens are still required. Knowledge about the role of the pathways involved in PAH and their interactions provides a better understanding of the pathogenesis of the disease and may highlight directions for novel therapeutic strategies for PAH. This paper reviews some novel, promising PAH-associated signaling pathways, such as RAAS, RhoA/ROCK, PDGF, PPAR, and TGF, focusing also on their possible interactions with well-established ones such as NO, ET-1 and prostacyclin pathways.
Collapse
|
108
|
Zhao M, Austin ED, Hemnes AR, Loyd JE, Zhao Z. An evidence-based knowledgebase of pulmonary arterial hypertension to identify genes and pathways relevant to pathogenesis. MOLECULAR BIOSYSTEMS 2014; 10:732-40. [PMID: 24448676 PMCID: PMC3950334 DOI: 10.1039/c3mb70496c] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Accepted: 01/07/2014] [Indexed: 01/25/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a major progressive form of pulmonary hypertension (PH) with more than 4800 patients in the United States. In the last two decades, many studies have identified numerous genes associated with this disease. However, there is no comprehensive research resource for PAH or other PH types that integrates various genetic studies and their related biological information. Thus, the number of associated genes, and their strength of evidence, is unclear. In this study, we tested the hypothesis that a web-based knowledgebase could be used to develop a biological map of highly interrelated, functionally important genes in PAH. We developed the pulmonary arterial hypertension knowledgebase (PAHKB, ), a comprehensive database with a user-friendly web interface. PAHKB extracts genetic data from all available sources, including those from association studies, genetic mutation, gene expression, animal model, supporting literature, various genomic annotations, gene networks, cellular and regulatory pathways, as well as microRNAs. Moreover, PAHKB provides online tools for data browsing and searching, data integration, pathway graphical presentation, and gene ranking. In the current release, PAHKB contains 341 human PH-related genes (293 protein coding and 48 non-coding genes) curated from over 1000 PubMed abstracts. Based on the top 39 ranked PAH-related genes in PAHKB, we constructed a core biological map. This core map was enriched with the TGF-beta signaling pathway, focal adhesion, cytokine-cytokine receptor interaction, and MAPK signaling. In addition, the reconstructed map elucidates several novel cancer signaling pathways, which may provide clues to support the application of anti-cancer therapeutics to PAH. In summary, we have developed a system for the identification of core PH-related genes and identified critical signaling pathways that may be relevant to PAH pathogenesis. This system can be easily applied to other pulmonary diseases.
Collapse
Affiliation(s)
- Min Zhao
- Department of Biomedical Informatics , Vanderbilt University School of Medicine , Nashville , TN , USA .
| | - Eric D. Austin
- Department of Pediatrics , Vanderbilt University School of Medicine , Nashville , TN , USA
| | - Anna R. Hemnes
- Division of Allergy , Pulmonary and Critical Care Medicine , Vanderbilt University School of Medicine , Nashville , TN , USA
| | - James E. Loyd
- Department of Medicine , Vanderbilt University Medical Center , Nashville , TN , USA
| | - Zhongming Zhao
- Department of Biomedical Informatics , Vanderbilt University School of Medicine , Nashville , TN , USA .
- Department of Cancer Biology , Vanderbilt University School of Medicine , Nashville , TN , USA
- Department of Psychiatry , Vanderbilt University School of Medicine , Nashville , TN , USA
- Center for Quantitative Sciences , Vanderbilt University Medical Center , Nashville , TN , USA
| |
Collapse
|
109
|
Tuder RM, Archer SL, Dorfmüller P, Erzurum SC, Guignabert C, Michelakis E, Rabinovitch M, Schermuly R, Stenmark KR, Morrell NW. Relevant issues in the pathology and pathobiology of pulmonary hypertension. J Am Coll Cardiol 2014; 62:D4-12. [PMID: 24355640 DOI: 10.1016/j.jacc.2013.10.025] [Citation(s) in RCA: 402] [Impact Index Per Article: 40.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Accepted: 10/22/2013] [Indexed: 11/18/2022]
Abstract
Knowledge of the pathobiology of pulmonary hypertension (PH) continues to accelerate. However, fundamental gaps remain in our understanding of the underlying pathological changes in pulmonary arteries and veins in the different forms of this syndrome. Although PH primarily affects the arteries, venous disease is increasingly recognized as an important entity. Moreover, prognosis in PH is determined largely by the status of the right ventricle, rather than the levels of pulmonary artery pressures. It is increasingly clear that although vasospasm plays a role, PH is an obstructive lung panvasculopathy. Disordered metabolism and mitochondrial structure, inflammation, and dysregulation of growth factors lead to a proliferative, apoptosis-resistant state. These abnormalities may be acquired, genetically mediated as a result of mutations in bone morphogenetic protein receptor-2 or activin-like kinase-1, or epigenetically inherited (as a result of epigenetic silencing of genes such as superoxide dismutase-2). There is a pressing need to better understand how the pathobiology leads to severe disease in some patients versus mild PH in others. Recent recognition of a potential role of acquired abnormalities of mitochondrial metabolism in the right ventricular myocytes and pulmonary vascular cells suggests new therapeutic approaches, diagnostic modalities, and biomarkers. Finally, dissection of the role of pulmonary inflammation in the initiation and promotion of PH has revealed a complex yet fascinating interplay with pulmonary vascular remodeling, promising to lead to novel therapeutics and diagnostics. Emerging concepts are also relevant to the pathobiology of PH, including a role for bone marrow and circulating progenitor cells and microribonucleic acids. Continued interest in the interface of the genetic basis of PH and cellular and molecular pathogenetic links should further expand our understanding of the disease.
Collapse
Affiliation(s)
- Rubin M Tuder
- Program in Translational Lung Research, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado.
| | - Stephen L Archer
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Peter Dorfmüller
- Department of Pathology, Marie Lannelongue Hospital, University Paris-Sud, Le Plessis-Robinson, France
| | - Serpil C Erzurum
- Lerner Research Institute and Respiratory Institute, Cleveland Clinic, Cleveland, Ohio
| | - Christophe Guignabert
- INSERM UMR 999, LabEx LERMIT, Marie Lannelongue Hospital and University Paris-Sud, School of Medicine, Kremlin-Bicêtre, France
| | | | - Marlene Rabinovitch
- Cardiovascular Institute and Department of Pediatrics and The Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University School of Medicine, Stanford, California
| | - Ralph Schermuly
- Excellence Cluster Cardio-Pulmonary System, German Lung Center, Universities of Giessen and Marburg Lung Center, Justus-Liebig-University, Giessen, Germany
| | - Kurt R Stenmark
- Cardiovascular Pulmonary Laboratory, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - Nicholas W Morrell
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom.
| |
Collapse
|
110
|
Effects of vascular endothelial growth factor on endothelin-1 production by human lung microvascular endothelial cells in vitro. Life Sci 2014; 118:191-4. [PMID: 24607779 DOI: 10.1016/j.lfs.2014.02.032] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Revised: 02/18/2014] [Accepted: 02/22/2014] [Indexed: 11/23/2022]
Abstract
AIMS Increased endothelin-1 (ET-1) is a hallmark of pulmonary arterial hypertension (PAH), and contributes to its pathogenesis. The factors controlling ET-1 in PAH are poorly understood. Combined with other stimuli, vascular endothelial growth factor (VEGF) blockade results in PAH-like lesions in animal models, and has been associated with PAH in humans. The effects of VEGF on ET-1 production by human lung blood microvascular endothelial cells (HMVEC-LBl) are unknown. MAIN METHODS We exposed HMVEC-LBl in-vitro to human VEGF-121 (40 ng/mL) in serum-free medium for 7h, in the absence or presence of the VEGF receptor antagonist, SU5416 (3 and 10 μM). ET-1 production was measured in the supernatant. Phosphorylation of VEGF receptor 2 (VEGFR2) was measured by Western blotting after exposure to VEGF without or with SU5416 for 5 and 10 min. KEY FINDINGS VEGF effectively caused VEGFR2 phosphorylation, which was blocked by SU5416. VEGF decreased ET-1 production by at least 29%. In the absence of VEGF, SU5416 increased ET-1 production, by 16% at 10 μM, and SU5416 was able to completely abolish the VEGF effect on ET-1 production. SIGNIFICANCE VEGF may promote vascular health by decreasing ET-1 production in HVMEC-LBl. Blockade of VEGF signaling by SU5416 increases ET-1 levels. The role of VEGF in modulating endothelin production in PAH deserves further study.
Collapse
|
111
|
Xiao Y, Christou H, Liu L, Visner G, Mitsialis SA, Kourembanas S, Liu H. Endothelial indoleamine 2,3-dioxygenase protects against development of pulmonary hypertension. Am J Respir Crit Care Med 2014; 188:482-91. [PMID: 23822766 DOI: 10.1164/rccm.201304-0700oc] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
RATIONALE A proliferative and apoptosis-resistant phenotype in pulmonary arterial smooth muscle cells (PASMCs) is key to pathologic vascular remodeling in pulmonary hypertension (PH). Expression of indoleamine-2,3-dioxygenase (IDO) by vascular endothelium is a newly identified vasomotor-regulatory mechanism also involved in molecular signaling cascades governing vascular smooth muscle cell (vSMC) plasticity. OBJECTIVES To investigate the therapeutic potential of enhanced endothelial IDO in development of PH and its associated vascular remodeling. METHODS We used loss and gain of function in vivo studies to establish the role and determine the therapeutic effect of endothelial IDO in hypoxia-induced PH in mice and monocrotaline-induced PH in rats. We also studied PASMC phenotype in an IDO-high in vivo and in vitro tissue microenvironment. MEASUREMENTS AND MAIN RESULTS The endothelium was the primary site for endogenous IDO production within mouse lung, and the mice lacking this gene had exaggerated hypoxia-induced PH. Conversely, augmented pulmonary endothelial IDO expression, through a human IDO-encoding Sleeping Beauty (SB)-based nonviral gene-integrating approach, halted and attenuated the development of PH, right ventricular hypertrophy, and vascular remodeling in both preclinical models of PH. IDO derived from endothelial cells promoted apoptosis in PH-PASMCs through depolarization of mitochondrial transmembrane potential and down-regulated PH-PASMC proliferative/synthetic capacity through enhanced binding of myocardin to CArG box DNA sequences present within the promoters of vSMC differentiation-specific genes. CONCLUSIONS Enhanced endothelial IDO ameliorates PH and its associated vascular structural remodeling through paracrine phenotypic modulation of PH-PASMCs toward a proapoptotic and less proliferative/synthetic state.
Collapse
Affiliation(s)
- Yongguang Xiao
- Department of Surgery, Boston Children’s Hospital, Boston, MA, USA
| | | | | | | | | | | | | |
Collapse
|
112
|
Revermann M, Neofitidou S, Kirschning T, Schloss M, Brandes RP, Hofstetter C. Inhalation of the BK(Ca)-opener NS1619 attenuates right ventricular pressure and improves oxygenation in the rat monocrotaline model of pulmonary hypertension. PLoS One 2014; 9:e86636. [PMID: 24497961 PMCID: PMC3909005 DOI: 10.1371/journal.pone.0086636] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2013] [Accepted: 12/11/2013] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Right heart failure is a fatal consequence of chronic pulmonary hypertension (PH). The development of PH is characterized by increased proliferation of vascular cells, in particular pulmonary artery smooth muscle cells (PASMCs) and pulmonary artery endothelial cells. In the course of PH, an escalated right ventricular (RV) afterload occurs, which leads to increased perioperative morbidity and mortality. BK(Ca) channels are ubiquitously expressed in vascular smooth muscle cells and their opening induces cell membrane hyperpolarization followed by vasodilation. Moreover, BK activation induces anti-proliferative effects in a multitude of cell types. On this basis, we hypothesized that treatment with the nebulized BK channel opener NS1619 might be a therapy option for pulmonary hypertension and tested this in rats. METHODS (1) Rats received monocrotaline injection for PH induction. Twenty-four days later, rats were anesthetized and NS1619 or the solvent was administered by inhalation. Systemic hemodynamic parameters, RV hemodynamic parameters, and blood gas analyses were measured before as well as 30 and 120 minutes after inhalation. (2) Rat PASMCs were stimulated with PDGF-BB in the presence and absence of NS1619. AKT, ERK1 and ERK2 activation were investigated by western blot analyses, and relative cell number was determined 48 hours after stimulation. RESULTS Inhalation of a 12 µM and 100 µM NS1619 solution significantly reduced RV pressure without affecting systemic arterial pressure. Blood gas analyses demonstrated significantly reduced carbon dioxide and improved oxygenation in NS1619-treated animals pointing towards a considerable pulmonary shunt-reducing effect. In PASMC's, NS1619 (100 µM) significantly attenuated PASMC proliferation by a pathway independent of AKT and ERK1/2 activation. CONCLUSION NS1619 inhalation reduces RV pressure and improves oxygen supply and its application inhibits PASMC proliferation in vitro. Hence, BK opening might be a novel option for the treatment of pulmonary hypertension.
Collapse
MESH Headings
- Administration, Inhalation
- Animals
- Becaplermin
- Benzimidazoles/administration & dosage
- Benzimidazoles/pharmacology
- Blotting, Western
- Cell Proliferation/drug effects
- Cells, Cultured
- Hemodynamics/drug effects
- Hypertension, Pulmonary/chemically induced
- Hypertension, Pulmonary/physiopathology
- Hypertension, Pulmonary/prevention & control
- Large-Conductance Calcium-Activated Potassium Channels/metabolism
- Male
- Mitogen-Activated Protein Kinase 1/metabolism
- Mitogen-Activated Protein Kinase 3/metabolism
- Monocrotaline
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Oxygen/metabolism
- Proto-Oncogene Proteins c-akt/metabolism
- Proto-Oncogene Proteins c-sis/pharmacology
- Pulmonary Artery/cytology
- Rats
- Rats, Sprague-Dawley
- Ventricular Dysfunction, Right/physiopathology
- Ventricular Dysfunction, Right/prevention & control
- Ventricular Pressure/drug effects
Collapse
Affiliation(s)
- Marc Revermann
- Department of Anesthesiology and Critical Care Medicine, University Hospital Mannheim, Faculty of Medicine, University of Heidelberg, Heidelberg, Germany
- Institute for Cardiovascular Physiology, Medical Faculty of the Goethe-University Frankfurt, Frankfurt, Germany
| | - Skevi Neofitidou
- Department of Anesthesiology and Critical Care Medicine, University Hospital Mannheim, Faculty of Medicine, University of Heidelberg, Heidelberg, Germany
| | - Thomas Kirschning
- Department of Anesthesiology and Critical Care Medicine, University Hospital Mannheim, Faculty of Medicine, University of Heidelberg, Heidelberg, Germany
| | - Manuel Schloss
- Institute for Cardiovascular Physiology, Medical Faculty of the Goethe-University Frankfurt, Frankfurt, Germany
| | - Ralf P. Brandes
- Institute for Cardiovascular Physiology, Medical Faculty of the Goethe-University Frankfurt, Frankfurt, Germany
| | - Christian Hofstetter
- Department of Anesthesiology and Critical Care Medicine, University Hospital Mannheim, Faculty of Medicine, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
113
|
Fessel JP, Flynn CR, Robinson LJ, Penner NL, Gladson S, Kang CJ, Wasserman DH, Hemnes AR, West JD. Hyperoxia synergizes with mutant bone morphogenic protein receptor 2 to cause metabolic stress, oxidant injury, and pulmonary hypertension. Am J Respir Cell Mol Biol 2013; 49:778-87. [PMID: 23742019 DOI: 10.1165/rcmb.2012-0463oc] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) has been associated with a number of different but interrelated pathogenic mechanisms. Metabolic and oxidative stresses have been shown to play important pathogenic roles in a variety of model systems. However, many of these relationships remain at the level of association. We sought to establish a direct role for metabolic stress and oxidant injury in the pathogenesis of PAH. Mice that universally express a disease-causing mutation in bone morphogenic protein receptor 2 (Bmpr2) were exposed to room air or to brief daily hyperoxia (95% oxygen for 3 h) for 6 weeks, and were compared with wild-type animals undergoing identical exposures. In both murine tissues and cultured endothelial cells, the expression of mutant Bmpr2 was sufficient to cause oxidant injury that was particularly pronounced in mitochondrial membranes. With the enhancement of mitochondrial generation of reactive oxygen species by hyperoxia, oxidant injury was substantially enhanced in mitochondrial membranes, even in tissues distant from the lung. Hyperoxia, despite its vasodilatory actions in the pulmonary circulation, significantly worsened the PAH phenotype (elevated right ventricular systolic pressure, decreased cardiac output, and increased pulmonary vascular occlusion) in Bmpr2 mutant animals. These experiments demonstrate that oxidant injury and metabolic stress contribute directly to disease development, and provide further evidence for PAH as a systemic disease with life-limiting cardiopulmonary manifestations.
Collapse
Affiliation(s)
- Joshua P Fessel
- 1 Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine
| | | | | | | | | | | | | | | | | |
Collapse
|
114
|
Colvin KL, Cripe PJ, Ivy DD, Stenmark KR, Yeager ME. Bronchus-associated lymphoid tissue in pulmonary hypertension produces pathologic autoantibodies. Am J Respir Crit Care Med 2013; 188:1126-36. [PMID: 24093638 DOI: 10.1164/rccm.201302-0403oc] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
RATIONALE Autoimmunity has long been associated with pulmonary hypertension. Bronchus-associated lymphoid tissue plays important roles in antigen sampling and self-tolerance during infection and inflammation. OBJECTIVES We reasoned that activated bronchus-associated lymphoid tissue would be evident in rats with pulmonary hypertension, and that loss of self-tolerance would result in production of pathologic autoantibodies that drive vascular remodeling. METHODS We used animal models, histology, and gene expression assays to evaluate the role of bronchus-associated lymphoid tissue in pulmonary hypertension. MEASUREMENTS AND MAIN RESULTS Bronchus-associated lymphoid tissue was more numerous, larger, and more active in pulmonary hypertension compared with control animals. We found dendritic cells in and around lymphoid tissue, which were composed of CD3(+) T cells over a core of CD45RA(+) B cells. Antirat IgG and plasma from rats with pulmonary hypertension decorated B cells in lymphoid tissue, resistance vessels, and adventitia of large vessels. Lymphoid tissue in diseased rats was vascularized by aquaporin-1(+) high endothelial venules and vascular cell adhesion molecule-positive vessels. Autoantibodies are produced in bronchus-associated lymphoid tissue and, when bound to pulmonary adventitial fibroblasts, change their phenotype to one that may promote inflammation. Passive transfer of autoantibodies into rats caused pulmonary vascular remodeling and pulmonary hypertension. Diminution of lymphoid tissue reversed pulmonary hypertension, whereas immunologic blockade of CCR7 worsened pulmonary hypertension and hastened its onset. CONCLUSIONS Bronchus-associated lymphoid tissue expands in pulmonary hypertension and is autoimmunologically active. Loss of self-tolerance contributes to pulmonary vascular remodeling and pulmonary hypertension. Lymphoid tissue-directed therapies may be beneficial in treating pulmonary hypertension.
Collapse
|
115
|
Horvatits T, Fuhrmann V. Therapeutic options in pulmonary hepatic vascular diseases. Expert Rev Clin Pharmacol 2013; 7:31-42. [DOI: 10.1586/17512433.2014.857598] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
116
|
Abstract
Hypoxic pulmonary hypertension of the newborn is characterized by elevated pulmonary vascular resistance and pressure due to vascular remodeling and increased vessel tension secondary to chronic hypoxia during the fetal and newborn period. In comparison to the adult, the pulmonary vasculature of the fetus and the newborn undergoes tremendous developmental changes that increase susceptibility to a hypoxic insult. Substantial evidence indicates that chronic hypoxia alters the production and responsiveness of various vasoactive agents such as endothelium-derived nitric oxide, endothelin-1, prostanoids, platelet-activating factor, and reactive oxygen species, resulting in sustained vasoconstriction and vascular remodeling. These changes occur in most cell types within the vascular wall, particularly endothelial and smooth muscle cells. At the cellular level, suppressed nitric oxide-cGMP signaling and augmented RhoA-Rho kinase signaling appear to be critical to the development of hypoxic pulmonary hypertension of the newborn.
Collapse
Affiliation(s)
- Yuansheng Gao
- Department of Physiology and Pathophysiology, Peking University, Health Science Center, Beijing, China
| | | |
Collapse
|
117
|
Araz O, Ucar EY, Yalcin A, Pulur D, Acemoglu H, Tas H, Saglam L, Akgun M, Mirici A. The incidence and severity of pulmonary hypertension in obstructive sleep apnea with hypothyroidism. Med Sci Monit 2013; 19:883-7. [PMID: 24149072 PMCID: PMC3808256 DOI: 10.12659/msm.889619] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Accepted: 08/13/2013] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Hypothyroidism and obstructive sleep apnea (OSA) are both common health problems and can be seen together. Each of these 2 diseases can cause pulmonary hypertension (PH). We aimed to determine whether hypothyroidism with OSA has a significant effect on the frequency and severity of PH. MATERIAL AND METHODS A total of 236 patients were included in the study. Patients were divided into 3 groups: Group I, Obstructive Sleep Apnea (n=149); Group II, Hypothyroidism (n=56); and Group III, Obstructive Sleep Apnea-Hypothyroidism (n=31). All patients underwent polysomnography and echocardiography and serum levels of thyroid-stimulating hormone (TSH) and free thyroxine 4 (FT4) were analyzed. RESULTS There were 167 male and 69 female participants, and the mean age was 47.8 ± 11.5 (Group I: 81.9% male, 18.1% female; Group II: 44.6% male, 55.4% female; Group III: 64.6% male, 35.4% female). Distribution of mean pulmonary arterial pressure on echocardiography was statistically different among the 3 groups (x(2)=14.99, p=0.006). When adjusted according to the apnea-hypopnea index (AHI), age, and body mass index (BMI), a significant relation with PH was determined (p=0.002). CONCLUSIONS The combination of hypothyroidism with OSA is associated with an increased frequency and severity of PH. When PH is found out of line with the severity of OSA, thyroid dysfunction should be investigated.
Collapse
Affiliation(s)
- Omer Araz
- Department of Pulmonary Diseases, Ataturk University School of Medicine, Erzurum, Turkey
| | - Elif Yilmazel Ucar
- Department of Pulmonary Diseases, Ataturk University School of Medicine, Erzurum, Turkey
| | - Aslıhan Yalcin
- Department of Pulmonary Diseases, Marmara University, Istanbul, Turkey
| | - Didem Pulur
- Department of Pulmonary Diseases, Ataturk University School of Medicine, Erzurum, Turkey
| | - Hamit Acemoglu
- Department of Medical Education, Ataturk University School of Medicine, Erzurum, Turkey
| | - Hakan Tas
- Department of Cardiology, Ataturk University School of Medicine, Erzurum, Turkey
| | - Leyla Saglam
- Department of Pulmonary Diseases, Ataturk University School of Medicine, Erzurum, Turkey
| | - Metin Akgun
- Department of Pulmonary Diseases, Ataturk University School of Medicine, Erzurum, Turkey
| | - Arzu Mirici
- Department of Pulmonary Diseases, 18 Mart University School of Medicine, Canakkale, Turkey
| |
Collapse
|
118
|
Kolosionek E, King J, Rollinson D, Schermuly RT, Grimminger F, Graham BB, Morrell N, Butrous G. Schistosomiasis causes remodeling of pulmonary vessels in the lung in a heterogeneous localized manner: Detailed study. Pulm Circ 2013; 3:356-62. [PMID: 24015336 PMCID: PMC3757830 DOI: 10.4103/2045-8932.114764] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Schistosomiasis is a global parasitic disease with high impact on public health in tropical areas. Schistosomiasis is a well-described cause of pulmonary arterial hypertension (PAH). The exact pathogenesis is still unclear, though inflammatory mechanisms are suspected. Another unknown is whether the changes in the pulmonary vasculature are generalized or localized. We studied 13 mice infected with cercariae for 12 weeks compared with 10 control mice. In our model, we observed that the liver was a target during infection and was enlarged more than two-fold after infection. However, right heart hypertrophy as measured by RV/(LV + S) ratio was not observed at this time point. Moreover, we noticed that 72% of the sampled lobes (92% of the lungs) harvested from these animals costained evidence of granulomatous changes, secondary to egg deposition. We systemically mapped the distribution of granulomatous lesions in right lung lobes (n = 43) of infected mice. We observed that the distribution of the granulomatous lesions was heterogeneous. Remodeled pulmonary vessels were seen in 26% of the lobes (46% of the lungs) and were observed only in close proximity to the granuloma. No remodeling was observed in the absence of granulomas. These findings support the view that pulmonary vascular remodeling is caused by the local presence of granulomas in PAH associated with schistosomiasis. The heterogeneous nature of the remodeling partly explains why many patients with schistosomiasis do not develop pulmonary hypertension.
Collapse
|
119
|
Pulmonary hypertension after hematopoietic stem cell transplantation. Biol Blood Marrow Transplant 2013; 19:1546-56. [PMID: 23891748 DOI: 10.1016/j.bbmt.2013.07.017] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 07/16/2013] [Indexed: 12/17/2022]
Abstract
Pulmonary hypertension (PH) is a potentially fatal complication of hematopoietic stem cell transplantation (HSCT). Given its nonspecific clinical presentation, it is likely that this clinical entity is underdiagnosed after HSCT. Data describing the incidence, risk factors, and etiology of PH in HSCT recipients are minimal. Physicians caring for HSCT recipients should be aware of this severe post-transplant complication because timely diagnosis and treatment may allow improved clinical outcomes. We summarize the pathophysiology, clinical presentation, diagnosis, and management of PH in HSCT recipients.
Collapse
|
120
|
Wagenaar GTM, Laghmani EH, Fidder M, Sengers RMA, de Visser YP, de Vries L, Rink R, Roks AJM, Folkerts G, Walther FJ. Agonists of MAS oncogene and angiotensin II type 2 receptors attenuate cardiopulmonary disease in rats with neonatal hyperoxia-induced lung injury. Am J Physiol Lung Cell Mol Physiol 2013; 305:L341-51. [PMID: 23812633 DOI: 10.1152/ajplung.00360.2012] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Stimulation of MAS oncogene receptor (MAS) or angiotensin (Ang) receptor type 2 (AT2) may be novel therapeutic options for neonatal chronic lung disease (CLD) by counterbalancing the adverse effects of the potent vasoconstrictor angiotensin II, consisting of arterial hypertension (PAH)-induced right ventricular hypertrophy (RVH) and pulmonary inflammation. We determined the cardiopulmonary effects in neonatal rats with CLD of daily treatment during continuous exposure to 100% oxygen for 10 days with specific ligands for MAS [cyclic Ang-(1-7); 10-50 μg·kg(-1)·day(-1)] and AT2 [dKcAng-(1-7); 5-20 μg·kg(-1)·day(-1)]. Parameters investigated included lung and heart histopathology, fibrin deposition, vascular leakage, and differential mRNA expression in the lungs of key genes involved in the renin-angiotensin system, inflammation, coagulation, and alveolar development. We investigated the role of nitric oxide synthase inhibition with N(ω)-nitro-l-arginine methyl ester (25 mg·kg(-1)·day(-1)) during AT2 agonist treatment. Prophylactic treatment with agonists for MAS or AT2 for 10 days diminished cardiopulmonary injury by reducing alveolar septum thickness and medial wall thickness of small arterioles and preventing RVH. Both agonists attenuated the pulmonary influx of inflammatory cells, including macrophages (via AT2) and neutrophils (via MAS) but did not reduce alveolar enlargement and vascular alveolar leakage. The AT2 agonist attenuated hyperoxia-induced fibrin deposition. In conclusion, stimulation of MAS or AT2 attenuates cardiopulmonary injury by reducing pulmonary inflammation and preventing PAH-induced RVH but does not affect alveolar and vascular development in neonatal rats with experimental CLD. The beneficial effects of AT2 activation on experimental CLD were mediated via a NOS-independent mechanism.
Collapse
Affiliation(s)
- Gerry T M Wagenaar
- Department of Pediatrics, Division of Neonatology, Leiden University Medical Center, Leiden, the Netherlands.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
121
|
Mühlfeld C, Ochs M. Quantitative microscopy of the lung: a problem-based approach. Part 2: stereological parameters and study designs in various diseases of the respiratory tract. Am J Physiol Lung Cell Mol Physiol 2013; 305:L205-21. [PMID: 23709622 DOI: 10.1152/ajplung.00427.2012] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Design-based stereology provides efficient methods to obtain valuable quantitative information of the respiratory tract in various diseases. However, the choice of the most relevant parameters in a specific disease setting has to be deduced from the present pathobiological knowledge. Often it is difficult to express the pathological alterations by interpretable parameters in terms of volume, surface area, length, or number. In the second part of this companion review article, we analyze the present pathophysiological knowledge about acute lung injury, diffuse parenchymal lung diseases, emphysema, pulmonary hypertension, and asthma to come up with recommendations for the disease-specific application of stereological principles for obtaining relevant parameters. Worked examples with illustrative images are used to demonstrate the work flow, estimation procedure, and calculation and to facilitate the practical performance of equivalent analyses.
Collapse
Affiliation(s)
- Christian Mühlfeld
- Institute of Functional and Applied Anatomy, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany.
| | | |
Collapse
|
122
|
Abstract
Pulmonary hypertension (PH) is the remarkable hemodynamic consequence of widespread structural and functional changes within the pulmonary circulation. Elevated pulmonary vascular resistance leads to increased mean pulmonary arterial pressure and, ultimately, right ventricular dysfunction. PH carries a poor prognosis and warrants timely and accurate diagnosis for appropriate intervention. The 2008 Dana Point classification system provides the categorical framework currently guiding therapy and surveillance. Radiologic imaging is an essential tool in the detection and diagnostic evaluation of patients with PH. Echocardiography, ventilation-perfusion scintigraphy, multidetector computed tomography, and cardiac magnetic resonance imaging provide insights into vascular morphology, pulmonary parenchymal status, cardiac function, and underlying etiology of the disorder. Emerging techniques of functional pulmonary and cardiac imaging hold great promise for the assessment and monitoring of these patients in the future.
Collapse
Affiliation(s)
- Aletta Ann Frazier
- Department of Diagnostic Radiology, University of Maryland Medical System, Baltimore, MD 21201, USA.
| | | |
Collapse
|
123
|
Tonelli AR, Haserodt S, Aytekin M, Dweik RA. Nitric oxide deficiency in pulmonary hypertension: Pathobiology and implications for therapy. Pulm Circ 2013; 3:20-30. [PMID: 23662172 PMCID: PMC3641730 DOI: 10.4103/2045-8932.109911] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Nitric oxide (NO) is a diffusible gas with diverse roles in human physiology and disease. Significant progress in the understanding of its biological effects has taken place in recent years. This has led to a better understanding of the pathobiology of pulmonary hypertension (PH) and the development of new therapies. This article provides an overview of the NO physiology and its role in the pathobiology of lung diseases, particularly PH. We also discuss current and emerging specific treatments that target NO signaling pathways in PH.
Collapse
Affiliation(s)
- Adriano R Tonelli
- Department of Pulmonary, Allergy and Critical Care Medicine, Respiratory Institute, Cleveland, Ohio, USA
| | | | | | | |
Collapse
|
124
|
Bi LQ, Zhu R, Kong H, Wu SL, Li N, Zuo XR, Zhou SM, Kou JP, Yu BY, Wang H, Xie WP. Ruscogenin attenuates monocrotaline-induced pulmonary hypertension in rats. Int Immunopharmacol 2013; 16:7-16. [DOI: 10.1016/j.intimp.2013.03.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Revised: 01/24/2013] [Accepted: 03/11/2013] [Indexed: 01/28/2023]
|
125
|
Pugh ME, Buchowski MS, Robbins IM, Newman JH, Hemnes AR. Physical activity limitation as measured by accelerometry in pulmonary arterial hypertension. Chest 2013; 142:1391-1398. [PMID: 22576635 DOI: 10.1378/chest.12-0150] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND The 6-min walk test, commonly used to assess exercise capacity and response to therapy in pulmonary arterial hypertension (PAH), has many well-described limitations. Sedentary time is associated with adverse cardiovascular outcomes and reduced quality of life, and measuring sedentary time and physical activity using accelerometry is another potential way to quantify exercise capacity in PAH. Whether sedentary time is different in patients with PAH vs control subjects is unknown. METHODS Physical activity was measured in 20 patients with PAH and 30 matched healthy control subjects using accelerometry for 7 consecutive days. Patients with PAH completed standard 6-min walk testing, and baseline demographics were recorded for all study participants. Total daily activity counts, sedentary time, and proportion of time at various activity levels were compared between groups. RESULTS Sedentary time was significantly higher in patients with PAH (mean, 92.1% daily activity; 95% CI, 89.5-94.8%) than in control subjects (mean, 79.9% daily activity; 95% CI, 76.4%-83.5%; P < .001), and all levels of physical activity were reduced in the PAH group compared with the control group ( P < .01 for all). Daily moderate to vigorous physical activity was reduced in the PAH group (7.5 min; 95% CI; 0.8-15.6 min) compared with the control group (mean, 64.7 min; 95% CI, 51.1-78.2 min; P < .001). Activity counts correlated with 6-min walk distance in the PAH group (Spearman rank correlation 5 0.72, P < .001). CONCLUSIONS Sedentary time is increased in patients with PAH and may lead to increased risk for metabolic and cardiovascular morbidity. Quantitation of daily activity and sedentary time using accelerometry may be a novel end point for PAH management and clinical trials.
Collapse
Affiliation(s)
- Meredith E Pugh
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN.
| | - Maciej S Buchowski
- Division of Gastroenterology, Vanderbilt University Medical Center, Nashville, TN
| | - Ivan M Robbins
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - John H Newman
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Anna R Hemnes
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
126
|
Austin ED, Lahm T, West J, Tofovic SP, Johansen AK, MacLean MR, Alzoubi A, Oka M. Gender, sex hormones and pulmonary hypertension. Pulm Circ 2013; 3:294-314. [PMID: 24015330 PMCID: PMC3757824 DOI: 10.4103/2045-8932.114756] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Most subtypes of pulmonary arterial hypertension (PAH) are characterized by a greater susceptibility to disease among females, although females with PAH appear to live longer after diagnosis. While this "estrogen paradoxȍ of enhanced female survival despite increased female susceptibility remains a mystery, recent progress has begun to shed light upon the interplay of sex hormones, the pathogenesis of pulmonary hypertension, and the right ventricular response to stress. For example, emerging data in humans and experimental models suggest that estrogens or differential sex hormone metabolism may modify disease risk among susceptible subjects, and that estrogens may interact with additional local factors such as serotonin to enhance the potentially damaging chronic effects of estrogens on the pulmonary vasculature. Regardless, it remains unclear why not all estrogenic compounds behave equally, nor why estrogens appear to be protective in certain settings but detrimental in others. The contribution of androgens and other compounds, such as dehydroepiandrosterone, to pathogenesis and possibly treatment must be considered as well. In this review, we will discuss the recent understandings on how estrogens, estrogen metabolism, dehydroepiandrosterone, and additional susceptibility factors may all contribute to the pathogenesis or potentially to the treatment of pulmonary hypertension, by evaluating current human, cell-based, and experimental model data.
Collapse
Affiliation(s)
- Eric D. Austin
- Department of Pediatrics, Division of Allergy, Immunology, and Pulmonary Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Tim Lahm
- Division of Pulmonary, Allergy, Critical Care, Occupational, and Sleep Medicine and Richard L. Roudebush Veterans Affairs Medical Center, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - James West
- Department of Medicine, Division of Allergy, Immunology, and Pulmonary Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Stevan P. Tofovic
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Anne Katrine Johansen
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, USA
| | - Margaret R. MacLean
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, USA
| | - Abdallah Alzoubi
- Department of Medicine and Pharmacology and Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA
| | - Masahiko Oka
- Department of Medicine and Pharmacology and Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA
| |
Collapse
|
127
|
Farkas L, Kolb M. Vascular repair and regeneration as a therapeutic target for pulmonary arterial hypertension. ACTA ACUST UNITED AC 2013; 85:355-64. [PMID: 23594605 DOI: 10.1159/000350177] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The last decade has seen substantial changes in our understanding of the pathobiology of pulmonary arterial hypertension (PAH), a severe and devastating disease without curative treatment. It is now accepted that injury to the endothelial cells of the pulmonary arteries is central for the subsequent development of lumen-obliterative lung vascular lesions. A variety of circulating and lung-resident progenitor and stem cells likely contribute to vascular integrity, and evidence for the presence of cells expressing stem and progenitor cell markers is found inside and in the immediate vicinity of pulmonary vascular lesions in PAH. The currently available vasodilator therapies mainly target enhanced vasoconstriction in the lung circulation and help to maintain or improve right ventricular function, but do not treat pulmonary vascular remodeling, the underlying cause of the disease. Vascular gene therapy and cell therapy with progenitor and stem cells is a progressing field in the context of the development of novel treatment options for PAH, but the majority of the studies are currently performed at the level of preclinical studies in animal models. The current review provides an overview of the current knowledge on cell- and gene therapy-based approaches for vascular repair and regeneration in PAH.
Collapse
Affiliation(s)
- Laszlo Farkas
- Division of Pulmonary Disease and Critical Care Medicine, Department of Internal Medicine, Victoria Johnson Center for Obstructive Lung Disease, Virginia Commonwealth University, Richmond, VA 23298-0456, USA. lfarkas @ vcu.edu
| | | |
Collapse
|
128
|
Role of Rho-kinase and its inhibitors in pulmonary hypertension. Pharmacol Ther 2013; 137:352-64. [DOI: 10.1016/j.pharmthera.2012.12.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Accepted: 10/27/2012] [Indexed: 11/20/2022]
|
129
|
Bauer EM, Shapiro R, Zheng H, Ahmad F, Ishizawar D, Comhair SA, Erzurum SC, Billiar TR, Bauer PM. High mobility group box 1 contributes to the pathogenesis of experimental pulmonary hypertension via activation of Toll-like receptor 4. Mol Med 2013; 18:1509-18. [PMID: 23269975 DOI: 10.2119/molmed.2012.00283] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 07/16/2012] [Indexed: 01/24/2023] Open
Abstract
Survival rates for patients with pulmonary hypertension (PH) remain low, and our understanding of the mechanisms involved are incomplete. Here we show in a mouse model of chronic hypoxia (CH)-induced PH that the nuclear protein and damage-associate molecular pattern molecule (DAMP) high mobility group box 1 (HMGB1) contributes to PH via a Toll-like receptor 4 (TLR4)-dependent mechanism. We demonstrate extranuclear HMGB1 in pulmonary vascular lesions and increased serum HMGB1 in patients with idiopathic pulmonary arterial hypertension. The increase in circulating HMGB1 correlated with mean pulmonary artery pressure. In mice, we similarly detected the translocation and release of HMGB1 after exposure to CH. HMGB1-neutralizing antibody attenuated the development of CH-induced PH, as assessed by measurement of right ventricular systolic pressure, right ventricular hypertrophy, pulmonary vascular remodeling and endothelial activation and inflammation. Genetic deletion of the pattern recognition receptor TLR4, but not the receptor for advanced glycation end products, likewise attenuated CH-induced PH. Finally, daily treatment of mice with recombinant human HMGB1 exacerbated CH-induced PH in wild-type (WT) but not Tlr4(-/-) mice. These data demonstrate that HMGB1-mediated activation of TLR4 promotes experimental PH and identify HMGB1 and/or TLR4 as potential therapeutic targets for the treatment of PH.
Collapse
Affiliation(s)
- Eileen M Bauer
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, United States of America
| | | | | | | | | | | | | | | | | |
Collapse
|
130
|
Wagenaar GTM, Laghmani EH, de Visser YP, Sengers RMA, Steendijk P, Baelde HJ, Walther FJ. Ambrisentan reduces pulmonary arterial hypertension but does not stimulate alveolar and vascular development in neonatal rats with hyperoxic lung injury. Am J Physiol Lung Cell Mol Physiol 2013; 304:L264-75. [PMID: 23292811 DOI: 10.1152/ajplung.00073.2012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Ambrisentan, an endothelin receptor type A antagonist, may be a novel therapeutic agent in neonatal chronic lung disease (CLD) by blocking the adverse effects of the vasoconstrictor endothelin-1, especially pulmonary arterial hypertension (PAH)-induced right ventricular hypertrophy (RVH). We determined the cardiopulmonary effects of ambrisentan treatment (1-20 mg·kg(-1)·day(-1)) in neonatal rats with CLD in 2 models: early treatment during continuous exposure to hyperoxia for 10 days and late treatment starting on day 6 in rat pups exposed postnatally to hyperoxia for 9 days, followed by a 9-day recovery period in room air. Parameters investigated included survival, lung and heart histopathology, right ventricular function, fibrin deposition, and differential mRNA expression in the lungs. In the early treatment model, we investigated the role of nitric oxide synthase (NOS) inhibition with N(ω)-nitro-L-arginine methyl ester (L-NAME; 25 mg·kg(-1)·day(-1)) during ambrisentan treatment. In the early treatment model, ambrisentan improved survival with reduced lung fibrin and collagen III deposition, arterial medial wall thickness, and RVH. These changes were not affected by L-NAME administration. Ambrisentan did not reduce the influx of macrophages and neutrophils or prevent reduced irregular elastin expression. In the late treatment model, ambrisentan diminished PAH, RVH, and right ventricular peak pressure, demonstrating that RVH is reversible in the neonatal period. Alveolarization and vascularization were not affected by ambrisentan. In conclusion, ambrisentan prolongs survival and reduces lung injury, PAH, and RVH via a NOS-independent mechanism but does not affect inflammation and alveolar and vascular development in neonatal rats with CLD.
Collapse
Affiliation(s)
- Gerry T M Wagenaar
- Department of Pediatrics, Division of Neonatology, Leiden University Medical Center, Leiden, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
131
|
Gammella E, Leuenberger C, Gassmann M, Ostergaard L. Evidence of synergistic/additive effects of sildenafil and erythropoietin in enhancing survival and migration of hypoxic endothelial cells. Am J Physiol Lung Cell Mol Physiol 2012. [PMID: 23204066 DOI: 10.1152/ajplung.00112.2012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Endothelial cell dysfunction is a common event to several pathologies including pulmonary hypertension, which is often associated with hypoxia. As the endothelium plays an essential role in regulating the dynamic interaction between pulmonary vasodilatation and vasoconstriction, this cell type is fundamental in the development of vascular remodeling and increased vascular resistance. We investigated the protective effects of sildenafil, a phosphodiesterase type 5 inhibitor, given in combination with erythropoietin (Epo), as it has been demonstrated that both drugs have antiapoptotic effects on several cell types. Specifically, we examined the viability and angiogenic properties of rat pulmonary artery endothelial cells upon exposure to either 21% or 1% oxygen, in presence of sildenafil (1 and 100 nM) and Epo (5 and 20 U/ml) alone or in combination (1 nM and 20 U/ml). Cell proliferation and viability were analyzed by Trypan blue staining, MTT assay, and Annexin V/propidium iodide stainings. In all assays, the ability of the combination treatment in improving cell viability was superior to that of either drug alone. The angiogenic properties were studied using a migration and a 3D collagen assay, and the results revealed increases in the migration potential of endothelial cells as well as the ability to form tube-like structures in response to sildenafil and the combination treatment. We therefore conclude that both drugs exert protective effects on endothelial cells on hypoxia and that sildenafil enhances the migratory and angiogenic properties, especially in hypoxic conditions. Furthermore, we present evidence of possible additive or synergistic effects of both drugs.
Collapse
Affiliation(s)
- Elena Gammella
- Department of Human Morphology and Biomedical Sciences, Città Studi, Università degli Studi di Milano, Milan, Italy
| | | | | | | |
Collapse
|
132
|
ALK2 and BMPR2 knockdown and endothelin-1 production by pulmonary microvascular endothelial cells. Microvasc Res 2012; 85:46-53. [PMID: 23142694 DOI: 10.1016/j.mvr.2012.10.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 10/08/2012] [Indexed: 01/15/2023]
Abstract
BACKGROUND Many cases of pulmonary arterial hypertension (PAH) are heritable and related to gene mutations in bone morphogenic receptor-2 (BMPR2). These patients consequently may have a signaling imbalance within the transforming growth factor beta (TGFβ) receptor superfamily. The causes of increased endothelin-1 (ET-1), which contributes to PAH, are unknown, and we therefore studied the contribution of various BMPs and their receptors on ET-1 production in vitro, after knockdown of BMPR2 in human pulmonary microvascular endothelial cells (HMVEC-LBl). METHODOLOGY/PRINCIPAL FINDINGS Receptor knockdown in HMVEC-LBl was performed using siRNA to BMPR2, and activin like-kinases 1 and 2 (ALK1, ALK2). ET-1 and TGFβ levels in the medium were measured by ELISA. In some experiments, cells were exposed to TGFβ or BMP7 or FK506 (tacrolimus). Using Western blotting, levels of BMPR2, endothelin ET(B) receptor, phosphorylated SMAD 2 (pSMAD 2), phosphorylated SMAD 1,5 (pSMAD 1,5), ALK1, ALK2, ALK5, TGFβ receptor 2, plasminogen activator inhibitor-1 (PAI-1) and ID1 were measured. BMPR2 knockdown significantly increased ET-1 levels. It did not affect ET(B) receptor or TGFβ levels. TGFβ increased ET-1 levels, with or without BMPR2 knockdown. BMPR2 knockdown did not affect TGFβ (pSMAD 2 and PAI-1) signaling. BMP7 increased ET-1 levels after BMPR2 knockdown but this was prevented by ALK2 knockdown as was the increase in ID1 caused by BMPR2 knockdown. FK506, which interacts with ALK2, increased ET-1 levels and ID1 levels, and this was blocked by ALK2 knockdown. CONCLUSION/SIGNIFICANCE ALK2 may be an important receptor in ET-1 production during BMPR2 knockdown.
Collapse
|
133
|
Yeager ME, Colvin KL, Everett AD, Stenmark KR, Ivy DD. Plasma proteomics of differential outcome to long-term therapy in children with idiopathic pulmonary arterial hypertension. Proteomics Clin Appl 2012; 6:257-67. [PMID: 22653875 DOI: 10.1002/prca.201100078] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
PURPOSE The prognosis for children with IPAH unresponsive to therapy is poor. We investigated the plasma proteome for a molecular basis of good versus poor outcome to long-term vasodilator therapy. EXPERIMENTAL DESIGN Plasma was collected at baseline or shortly after therapy initiation and following chronic vasodilator therapy, then divided into those with good outcome (n = 8), and those with a poor outcome (n = 7). To identify proteins unique to either outcome, we used differential gel electrophoresis and mass spectrometry. Results were confirmed by commercial enzyme-linked immunosorbent assay. RESULTS Before and after therapy, SAA-4 was 4-fold lower in those with good outcome compared to those with poor outcome, while serum paraoxonase/arylesterase-1 was increased 2-fold in those with good outcome versus poor outcome. After therapy, haptoglobin and hemopexin were 1.45- and 1.8-fold lower, respectively, in those with a good versus poor outcome. Among those with a good outcome, SAP was 1.3-fold lower prior to therapy. CONCLUSIONS AND CLINICAL RELEVANCE SAP and SAA-4 regulate circulating mononuclear phagocytes. As such, they may contribute to the differential response to chronic vasodilator therapy in the context of inflammation in IPAH.
Collapse
Affiliation(s)
- Michael E Yeager
- Department of Pediatric Critical Care, University of Colorado Denver, Denver, CO 80045, USA.
| | | | | | | | | |
Collapse
|
134
|
Giusca S, Jurcut R, Coman IM, Ghiorghiu I, Catrina D, Popescu BA, Dima L, Ginghina C. Right Ventricular Function Predicts Clinical Response to Specific Vasodilator Therapy in Patients with Pulmonary Hypertension. Echocardiography 2012; 30:17-26. [DOI: 10.1111/j.1540-8175.2012.01809.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Affiliation(s)
- Sorin Giusca
- Cardiology Department; Institute for Emergencies in Cardiovascular Diseases “C. C. Iliescu; Bucharest; Romania
| | | | | | - Ioana Ghiorghiu
- Cardiology Department; Institute for Emergencies in Cardiovascular Diseases “C. C. Iliescu; Bucharest; Romania
| | - Daniela Catrina
- Cardiology Department; Institute for Emergencies in Cardiovascular Diseases “C. C. Iliescu; Bucharest; Romania
| | - Bogdan A. Popescu
- University of Medicine and Pharmacy “Carol Davila; Bucharest; Romania
| | - Laura Dima
- Cardiology Department; Institute for Emergencies in Cardiovascular Diseases “C. C. Iliescu; Bucharest; Romania
| | | |
Collapse
|
135
|
Abstract
INTRODUCTION Pulmonary arterial hypertension (PAH) encompasses a rare potentially lethal group of diseases characterized by vasoconstriction, in situ thrombosis and vascular remodeling. Most of the existing therapies including endothelin receptor antagonists, prostacyclin and derivatives, or phsophodiesterase-5 inhibitors tackle mainly the endothelial dysfunction, leaving the remodeling suboptimally inhibited. This explains the disease progression that occurs even with combined therapies and the need for other therapies able to adequately inhibit the vascular remodeling. AREAS COVERED Platelet-derived growth factor (PDGF) signaling pathway was demonstrated to be involved in the vascular remodeling in PAH, and therefore, it might be a desirable therapeutic target in this setting. This review discusses the pathogenic role of this pathway in PAH and its potential inhibitory approaches, focusing on imatinib as well as on the existing preclinical data on this compound. EXPERT OPINION Preclinical studies demonstrated that PDGF inhibition with receptor antagonists such as imatinib reduces vascular remodeling. Therefore, PDGF might represent a plausible therapeutic target in this disease. However, compounds able to block this pathway via different mechanisms might also become potential PAH therapies.
Collapse
Affiliation(s)
- Sabina Antonela Antoniu
- Grigore T Popa, University of Medicine and Pharmacy Iaşi, Department of Medicine II -Pulmonary Disease, Pulmonary Disease University Hospital, 30 Dr I Cihac Str, 700115 Iasi, Romania.
| |
Collapse
|
136
|
Abstract
Pulmonary arterial hypertension (PAH) is a rapidly progressive and fatal disease for which there is an ever-expanding body of genetic and related pathophysiological information on disease pathogenesis. The most common single culprit gene known is BMPR2, and animal models of the disease in several forms exist. There is a wealth of genetic data regarding modifiers of disease expression, penetrance, and severity. Despite the rapid accumulation of data in the last decade, a complete picture of the molecular pathogenesis of PAH leading to novel therapies is lacking. In this review, we attempt to summarize the current understanding of PAH from the genetic perspective. The most recent PAH demographics are discussed. Heritable PAH in the post-BMPR2 era is examined in detail as the most robust model of PAH genetics in both animal models and human pedigrees. Important downstream molecular pathways and modifiers of disease expression are reviewed in light of what is known about PAH pathogenesis. Current and emerging therapies are examined in light of genetic data. The role of genetic testing in PAH in the post-BMPR2 era is discussed. Finally, directions for future investigations that ideally will fulfill the promise of novel therapeutic or preventive strategies are discussed.
Collapse
Affiliation(s)
- Joshua P Fessel
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University, Nashville, Tennessee, US
| | | | | |
Collapse
|
137
|
Goncharova NS, Kazymly AV, Naimushin AV, Moiseeva OM. Contemporary treatment of pulmonary arterial hypertension: the North-West Registry data analysis. КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2012. [DOI: 10.15829/1728-8800-2012-4-79-84] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Aim. Using the prospective Registry data, to assess the effects of conventional and specific therapy on the clinical course and survival of the patients with pulmonary arterial hypertension (PAH). Material and methods. The study included 124 patients (mean age 38,2±13,7 years; 34 men and 78 women): 31 with idiopathic PAH (IPAH), 52 with Eisenmenger syndrome, 17 with inoperable chronic thromboembolic pulmonary hypertension, 9 with PAH and corrected congenital heart disease, 6 with PAH and systemic scleroderma, and 6 with PAH and HIV infection. Results. The cumulative one-year and three-year survival rates were 94% and 75%, respectively. Irrespective of the absence of right heart catheterisation and vasoreactive testing, 42,7% of the patients were treated with calcium antagonists. PAH-specific therapy was administered to 40,3% of the participants (64,5% and 21% of those with IPAH and Eisenmenger syndrome, respectively). PAH-specific therapy was associated with an increase in survival time. Conclusion. In PAH patients, the prognosis is linked to early administration of specific monotherapy and possible combination therapy. Developing a national registry of pulmonary hypertension will facilitate the assessment of the real-world demand for specific therapy and the related costs.
Collapse
Affiliation(s)
- N. S. Goncharova
- V. A. Almazov Federal Centre of Heart, Blood, and Endocrinology, St. Petersburg
| | - A. V. Kazymly
- V. A. Almazov Federal Centre of Heart, Blood, and Endocrinology, St. Petersburg
| | - A. V. Naimushin
- V. A. Almazov Federal Centre of Heart, Blood, and Endocrinology, St. Petersburg
| | - O. M. Moiseeva
- V. A. Almazov Federal Centre of Heart, Blood, and Endocrinology, St. Petersburg
| |
Collapse
|
138
|
Desai AA, Zhou T, Ahmad H, Zhang W, Mu W, Trevino S, Wade MS, Raghavachari N, Kato GJ, Peters-Lawrence MH, Thiruvoipati T, Turner K, Artz N, Huang Y, Patel AR, Yuan JXJ, Gordeuk VR, Lang RM, Garcia JGN, Machado RF. A novel molecular signature for elevated tricuspid regurgitation velocity in sickle cell disease. Am J Respir Crit Care Med 2012; 186:359-68. [PMID: 22679008 PMCID: PMC3443809 DOI: 10.1164/rccm.201201-0057oc] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Accepted: 05/24/2012] [Indexed: 01/14/2023] Open
Abstract
RATIONALE An increased tricuspid regurgitation jet velocity (TRV > 2.5 m/s) and pulmonary hypertension defined by right heart catheterization both independently confer increased mortality in sickle cell disease (SCD). OBJECTIVES We explored the usefulness of peripheral blood mononuclear cell-derived gene signatures as biomarkers for an elevated TRV in SCD. METHODS Twenty-seven patients with SCD underwent echocardiography and peripheral blood mononuclear cell isolation for expression profiling and 112 patients with SCD were genotyped for single-nucleotide polymorphisms. MEASUREMENTS AND MAIN RESULTS Genome-wide gene and miRNA expression profiles were correlated against TRV, yielding 631 transcripts and 12 miRNAs. Support vector machine analysis identified a 10-gene signature including GALNT13 (encoding polypeptide N-acetylgalactosaminyltransferase 13) that discriminates patients with and without increased TRV with 100% accuracy. This finding was then validated in a cohort of patients with SCD without (n = 10) and with pulmonary hypertension (n = 10, 90% accuracy). Increased TRV-related miRNAs revealed strong in silico binding predictions of miR-301a to GALNT13 corroborated by microarray analyses demonstrating an inverse correlation between their expression. A genetic association study comparing patients with an elevated (n = 49) versus normal (n = 63) TRV revealed five significant single-nucleotide polymorphisms within GALNT13 (P < 0.005), four trans-acting (P < 2.1 × 10(-7)) and one cis-acting (P = 0.6 × 10(-4)) expression quantitative trait locus upstream of the adenosine-A2B receptor gene (ADORA2B). CONCLUSIONS These studies validate the clinical usefulness of genomic signatures as potential biomarkers and highlight ADORA2B and GALNT13 as potential candidate genes in SCD-associated elevated TRV.
Collapse
Affiliation(s)
- Ankit A. Desai
- Institute for Personalized Respiratory Medicine
- Section of Cardiology
| | - Tong Zhou
- Institute for Personalized Respiratory Medicine
| | - Homaa Ahmad
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Wei Zhang
- Department of Pediatrics, Institute of Human Genetics, Cancer Center
| | - Wenbo Mu
- Department of Pediatrics, Institute of Human Genetics, Cancer Center
| | - Sharon Trevino
- Institute for Personalized Respiratory Medicine
- Section of Pulmonary, Critical Care, Sleep, and Allergy, and
| | | | | | - Gregory J. Kato
- Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland; and
| | - Marlene H. Peters-Lawrence
- Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland; and
| | | | - Kristin Turner
- Department of Medicine, Loyola University, Maywood, Illinois
| | - Nicole Artz
- Department of Medicine, Loyola University, Maywood, Illinois
| | - Yong Huang
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Amit R. Patel
- Department of Medicine, University of Chicago, Chicago, Illinois
| | | | - Victor R. Gordeuk
- Comprehensive Sickle Cell Center, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Roberto M. Lang
- Department of Medicine, University of Chicago, Chicago, Illinois
| | | | - Roberto F. Machado
- Institute for Personalized Respiratory Medicine
- Section of Pulmonary, Critical Care, Sleep, and Allergy, and
| |
Collapse
|
139
|
Hamal KR, Erf GF, Anthony NB, Wideman RF. Immunohistochemical examination of plexiform-like complex vascular lesions in the lungs of broiler chickens selected for susceptibility to idiopathic pulmonary arterial hypertension. Avian Pathol 2012; 41:211-9. [PMID: 22515539 DOI: 10.1080/03079457.2012.663077] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Idiopathic pulmonary arterial hypertension (IPAH) is a disease of unknown cause that is characterized by elevated pulmonary arterial pressure and pulmonary vascular resistance, and by extensive vascular remodelling. In human IPAH patients, remodelling of the pulmonary vasculature results in the formation of plexiform lesions in the terminal pulmonary arterioles. Various molecules are expressed in the human plexiform lesions, including alpha smooth muscle actin, von Willebrand factor, vascular endothelial growth factor, vascular endothelial growth factor receptor type 2, hypoxia inducible factor-1α, survivin, tenascin, collagen, fibronectin, and various immune/inflammatory cells such as, cytotoxic lymphocytes, B lymphocytes, MHC class II cells, and monocytes/macrophages are also present. Plexiform lesions rarely develop in the lungs of laboratory animals, but plexiform-like complex vascular lesions (CVL) do develop spontaneously in the lungs of broiler chickens from an IPAH-susceptible line. To examine angioproliferative and immune-system-related activities associated with CVL in broiler lungs, paraformaldehyde-fixed, paraffin-embedded lung sections from 8-week-old to 24-week-old broiler chickens were stained immunohistochemically using monoclonal or polyclonal antibodies specific for angioproliferative molecules and immune/inflammatory cells. The CVL in the lungs of broiler chickens exhibited positive staining for both angioproliferative molecules and immune/inflammatory cells. These observations combined with the close histological resemblance of broiler CVL to the plexiform lesions of human IPAH patients further validates chickens from our IPAH-susceptible line as an excellent animal model of spontaneous plexogenic arteriopathy.
Collapse
Affiliation(s)
- Krishna R Hamal
- Department of Poultry Science, University of Arkansas, 1260 West Maple, Fayetteville, AR 72701, USA.
| | | | | | | |
Collapse
|
140
|
Dorfmüller P, Humbert M. Progress in Pulmonary Arterial Hypertension Pathology: Relighting a Torch Inside the Tunnel. Am J Respir Crit Care Med 2012; 186:210-2. [DOI: 10.1164/rccm.201206-1049ed] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
141
|
Oudiz RJ, Brundage BH, Galiè N, Ghofrani HA, Simonneau G, Botros FT, Chan M, Beardsworth A, Barst RJ. Tadalafil for the treatment of pulmonary arterial hypertension: a double-blind 52-week uncontrolled extension study. J Am Coll Cardiol 2012; 60:768-74. [PMID: 22818063 DOI: 10.1016/j.jacc.2012.05.004] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Revised: 04/24/2012] [Accepted: 05/15/2012] [Indexed: 10/28/2022]
Abstract
OBJECTIVES The aim of this study was to evaluate the long-term safety and durability of efficacy of tadalafil for pulmonary arterial hypertension. BACKGROUND Tadalafil is an oral phosphodiesterase-5 inhibitor approved for PAH treatment. In the multicenter, placebo-controlled, randomized, 16-week PHIRST (Pulmonary Arterial Hypertension and Response to Tadalafil) study, tadalafil 40 mg improved exercise capacity and delayed clinical worsening. METHODS Eligible patients from PHIRST received once-daily tadalafil 20 mg (T20 mg) or 40 mg (T40 mg) (n = 357) in the double-blind, 52-week, uncontrolled extension study (PHIRST-2); 293 patients completed PHIRST-2. Durability of efficacy was explored using the 6-min walk distance (6MWD) test. Clinical worsening and changes in World Health Organization functional class were evaluated. RESULTS The safety profile of tadalafil in PHIRST-2 was similar to that in PHIRST, with typical phosphodiesterase-5 inhibitor adverse events. The 6MWDs achieved in PHIRST for the subset of patients receiving T20 mg and T40 mg in both PHIRST and PHIRST-2 (406 ± 67 m [n = 52] and 413 ± 81 m [n = 59] at PHIRST-2 enrollment, respectively) were maintained at PHIRST-2 completion (415 ± 80 m [n = 51] and 410 ± 78 m [n = 59], respectively). Numerically fewer patients who were on T40 mg in PHIRST and PHIRST-2 experienced World Health Organization functional class deterioration (6% [n = 5]) compared with those randomized to T20 mg (9% [n = 7]) across both studies. Post hoc analyses showed that background bosentan use and higher 6MWD at PHIRST baseline were associated with fewer clinical worsening events. CONCLUSIONS Long-term treatment with tadalafil was well tolerated in patients with pulmonary arterial hypertension. In patients receiving either T20 mg or T40 mg, the improvements in 6MWD demonstrated in the 16-week PHIRST study appeared sustained for up to 52 additional weeks of treatment in PHIRST-2. (Pulmonary Arterial Hypertension and Response to Tadalafil Study; NCT00549302).
Collapse
Affiliation(s)
- Ronald J Oudiz
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
142
|
Etiopathogenetic mechanisms of pulmonary hypertension in sleep-related breathing disorders. Pulm Med 2012; 2012:273591. [PMID: 22848814 PMCID: PMC3401569 DOI: 10.1155/2012/273591] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Accepted: 06/04/2012] [Indexed: 12/29/2022] Open
Abstract
Obstructive sleep apnea syndrome is a common disorder with significant health consequences and is on the rise in consonance with the obesity pandemic. In view of the association between sleep-disordered breathing and pulmonary hypertension as depicted by multiple studies, current clinical practice guidelines categorize obstructive sleep apnea as a risk factor for pulmonary hypertension and recommend an assessment for sleep disordered breathing in evaluating patients with pulmonary hypertension. The dysregulatory mechanisms associated with hypoxemic episodes observed in sleep related breathing disorders contribute to the onset of pulmonary hypertension and identification of these potentially treatable factors might help in the reduction of overall cardiovascular mortality.
Collapse
|
143
|
Recomendações da ESC para o tratamento da cardiopatia congénita no adulto (nova versão de 2010). Rev Port Cardiol 2012. [DOI: 10.1016/j.repc.2012.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
144
|
Zhao L, Chen CN, Hajji N, Oliver E, Cotroneo E, Wharton J, Wang D, Li M, McKinsey TA, Stenmark KR, Wilkins MR. Histone deacetylation inhibition in pulmonary hypertension: therapeutic potential of valproic acid and suberoylanilide hydroxamic acid. Circulation 2012; 126:455-67. [PMID: 22711276 DOI: 10.1161/circulationaha.112.103176] [Citation(s) in RCA: 180] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Epigenetic programming, dynamically regulated by histone acetylation, is a key mechanism regulating cell proliferation and survival. Little is known about the contribution of histone deacetylase (HDAC) activity to the development of pulmonary arterial hypertension, a condition characterized by profound structural remodeling of pulmonary arteries and arterioles. METHODS AND RESULTS HDAC1 and HDAC5 protein levels were elevated in lungs from human idiopathic pulmonary arterial hypertension and in lungs and right ventricles from rats exposed to hypoxia. Immunohistochemistry localized increased expression to remodeled vessels in the lung. Both valproic acid, a class I HDAC inhibitor, and suberoylanilide hydroxamic acid (vorinostat), an inhibitor of class I, II, and IV HDACs, mitigated the development of and reduced established hypoxia-induced pulmonary hypertension in the rat. Both valproic acid and suberoylanilide hydroxamic acid inhibited the imprinted highly proliferative phenotype of fibroblasts and R-cells from pulmonary hypertensive bovine vessels and platelet-derived growth factor-stimulated growth of human vascular smooth muscle cells in culture. Exposure to valproic acid and suberoylanilide hydroxamic acid was associated with increased levels of p21 and FOXO3 and reduced expression of survivin. The significantly higher levels of expression of cKIT, monocyte chemoattractant protein-1, interleukin-6, stromal-derived factor-1, platelet-derived growth factor-b, and S100A4 in R-cells were downregulated by valproic acid and suberoylanilide hydroxamic acid treatment. CONCLUSIONS Increased HDAC activity contributes to the vascular pathology of pulmonary hypertension. The effectiveness of HDAC inhibitors, valproic acid, and suberoylanilide hydroxamic acid, in models of pulmonary arterial hypertension supports a therapeutic strategy based on HDAC inhibition in pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Lan Zhao
- Centre for Pharmacology and Therapeutics, Experimental Medicine, Imperial College London, Du Cane Rd, London W12 ONN, UK.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
145
|
Corte TJ, McDonagh TA, Wort SJ. Pulmonary hypertension in left heart disease: A review. Int J Cardiol 2012; 156:253-8. [DOI: 10.1016/j.ijcard.2011.06.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2010] [Revised: 04/26/2011] [Accepted: 06/03/2011] [Indexed: 11/26/2022]
|
146
|
Schwaiblmair M, Faul C, von Scheidt W, Berghaus TM. Differences of cardiac output measurements by open-circuit acetylene uptake in pulmonary arterial hypertension and chronic thromboembolic pulmonary hypertension: a cohort study. Respir Res 2012; 13:18. [PMID: 22409387 PMCID: PMC3323432 DOI: 10.1186/1465-9921-13-18] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Accepted: 03/12/2012] [Indexed: 11/22/2022] Open
Abstract
Background As differences in gas exchange between pulmonary arterial hypertension (PAH) and chronic thromboembolic pulmonary hypertension (CTEPH) have been demonstrated, we asked if cardiac output measurements determined by acetylene (C2H2) uptake significantly differed in these diseases when compared to the thermodilution technique. Method Single-breath open-circuit C2H2 uptake, thermodilution, and cardiopulmonary exercise testing were performed in 72 PAH and 32 CTEPH patients. Results In PAH patients the results for cardiac output obtained by the two methods showed an acceptable agreement with a mean difference of -0.16 L/min (95% CI -2.64 to 2.32 L/min). In contrast, the agreement was poorer in the CTEPH group with the difference being -0.56 L/min (95% CI -4.96 to 3.84 L/min). Functional dead space ventilation (44.5 ± 1.6 vs. 32.2 ± 1.4%, p < 0.001) and the mean arterial to end-tidal CO2 gradient (9.9 ± 0.8 vs. 4.1 ± 0.5 mmHg, p < 0.001) were significantly elevated among CTEPH patients. Conclusion Cardiac output evaluation by the C2H2 technique should be interpreted with caution in CTEPH, as ventilation to perfusion mismatching might be more relevant than in PAH.
Collapse
Affiliation(s)
- Martin Schwaiblmair
- Department of Internal Medicine, Klinikum Augsburg, Ludwig-Maximilians-University Munich, Stenglinstrasse 2, D-86156 Augsburg, Germany.
| | | | | | | |
Collapse
|
147
|
Gerasimovskaya E, Kratzer A, Sidiakova A, Salys J, Zamora M, Taraseviciene-Stewart L. Interplay of macrophages and T cells in the lung vasculature. Am J Physiol Lung Cell Mol Physiol 2012; 302:L1014-22. [PMID: 22387295 DOI: 10.1152/ajplung.00357.2011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In severe pulmonary arterial hypertension (PAH), vascular lesions are composed of phenotypically altered vascular and inflammatory cells that form clusters or tumorlets. Because macrophages are found in increased numbers in intravascular and perivascular space in human PAH, here we address the question whether macrophages play a role in pulmonary vascular remodeling and whether accumulation of macrophages in the lung vasculature could be compromised by the immune system. We used the mouse macrophage cell line RAW 264.7 because these cells are resistant to apoptosis, have high proliferative capacity, and resemble cells in the plexiform lesions that tend to pile up instead of maintaining a monolayer. Cells were characterized by immunocytochemistry with cell surface markers (Lycopersicon Esculentum Lectin, CD117, CD133, FVIII, CD31, VEGFR-2, and S100). Activated, but not quiescent, T cells were able to suppress RAW 264.7 cell proliferative and migration activity in vitro. The carboxyfluorescein diacetate-labeled RAW 264.7 cells were injected into the naïve Sprague Dawley (SD) rat and athymic nude rat. Twelve days later, cells were found in the lung vasculature of athymic nude rats that lack functional T cells, contributing to vascular remodeling. No labeled RAW 264.7 cells were detected in the lungs of immune-competent SD rats. Our data demonstrate that T cells can inhibit in vitro migration and in vivo accumulation of macrophage-like cells.
Collapse
Affiliation(s)
- Evgenia Gerasimovskaya
- Department of Pediatrics, University of Colorado Denver, 12700 E. 19th Ave, Aurora, CO 80045, USA
| | | | | | | | | | | |
Collapse
|
148
|
Gomez-Arroyo J, Saleem SJ, Mizuno S, Syed AA, Bogaard HJ, Abbate A, Taraseviciene-Stewart L, Sung Y, Kraskauskas D, Farkas D, Conrad DH, Nicolls MR, Voelkel NF. A brief overview of mouse models of pulmonary arterial hypertension: problems and prospects. Am J Physiol Lung Cell Mol Physiol 2012; 302:L977-91. [PMID: 22307907 DOI: 10.1152/ajplung.00362.2011] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Many chronic pulmonary diseases are associated with pulmonary hypertension (PH) and pulmonary vascular remodeling, which is a term that continues to be used to describe a wide spectrum of vascular abnormalities. Pulmonary vascular structural changes frequently increase pulmonary vascular resistance, causing PH and right heart failure. Although rat models had been standard models of PH research, in more recent years the availability of genetically engineered mice has made this species attractive for many investigators. Here we review a large amount of data derived from experimental PH reports published since 1996. These studies using wild-type and genetically designed mice illustrate the challenges and opportunities provided by these models. Hemodynamic measurements are difficult to obtain in mice, and right heart failure has not been investigated in mice. Anatomical, cellular, and genetic differences distinguish mice and rats, and pharmacogenomics may explain the degree of PH and the particular mode of pulmonary vascular adaptation and also the response of the right ventricle.
Collapse
Affiliation(s)
- Jose Gomez-Arroyo
- Victoria Johnson Center for Obstructive Lung Disease Research, Virginia Commonwealth University, 1220 E. Broad St., Richmond, VA 23298, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
149
|
Fairfax KC, Amiel E, King IL, Freitas TC, Mohrs M, Pearce EJ. IL-10R blockade during chronic schistosomiasis mansoni results in the loss of B cells from the liver and the development of severe pulmonary disease. PLoS Pathog 2012; 8:e1002490. [PMID: 22291593 PMCID: PMC3266936 DOI: 10.1371/journal.ppat.1002490] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Accepted: 12/04/2011] [Indexed: 12/17/2022] Open
Abstract
In schistosomiasis patients, parasite eggs trapped in hepatic sinusoids become foci for CD4+ T cell-orchestrated granulomatous cellular infiltrates. Since the immune response is unable to clear the infection, the liver is subjected to ongoing cycles of focal inflammation and healing that lead to vascular obstruction and tissue fibrosis. This is mitigated by regulatory mechanisms that develop over time and which minimize the inflammatory response to newly deposited eggs. Exploring changes in the hepatic inflammatory infiltrate over time in infected mice, we found an accumulation of schistosome egg antigen-specific IgG1-secreting plasma cells during chronic infection. This population was significantly diminished by blockade of the receptor for IL-10, a cytokine implicated in plasma cell development. Strikingly, IL-10R blockade precipitated the development of portal hypertension and the accumulation of parasite eggs in the lungs and heart. This did not reflect more aggressive Th2 cell responsiveness, increased hepatic fibrosis, or the emergence of Th1 or Th17 responses. Rather, a role for antibody in the prevention of severe disease was suggested by the finding that pulmonary involvement was also apparent in mice unable to secrete class switched antibody. A major effect of anti-IL-10R treatment was the loss of a myeloid population that stained positively for surface IgG1, and which exhibited characteristics of regulatory/anti-inflammatory macrophages. This finding suggests that antibody may promote protective effects within the liver through local interactions with macrophages. In summary, our data describe a role for IL-10-dependent B cell responses in the regulation of tissue damage during a chronic helminth infection.
Collapse
MESH Headings
- Animals
- Antibodies, Helminth/genetics
- Antibodies, Helminth/immunology
- Antibodies, Helminth/metabolism
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD4-Positive T-Lymphocytes/pathology
- Chronic Disease
- Immunoglobulin G/genetics
- Immunoglobulin G/immunology
- Immunoglobulin G/metabolism
- Interleukin-10/genetics
- Interleukin-10/immunology
- Interleukin-10/metabolism
- Liver/immunology
- Liver/metabolism
- Liver/parasitology
- Liver/pathology
- Liver Cirrhosis/genetics
- Liver Cirrhosis/immunology
- Liver Cirrhosis/metabolism
- Liver Cirrhosis/parasitology
- Lung Diseases, Parasitic/genetics
- Lung Diseases, Parasitic/immunology
- Lung Diseases, Parasitic/metabolism
- Lung Diseases, Parasitic/parasitology
- Lung Diseases, Parasitic/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Plasma Cells/immunology
- Plasma Cells/metabolism
- Plasma Cells/pathology
- Receptors, Interleukin-10/antagonists & inhibitors
- Receptors, Interleukin-10/genetics
- Receptors, Interleukin-10/immunology
- Receptors, Interleukin-10/metabolism
- Schistosoma mansoni
- Schistosomiasis mansoni/genetics
- Schistosomiasis mansoni/immunology
- Schistosomiasis mansoni/metabolism
- Schistosomiasis mansoni/pathology
Collapse
Affiliation(s)
- Keke C. Fairfax
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Trudeau Institute, Saranac Lake, New York, United States of America
| | - Eyal Amiel
- Trudeau Institute, Saranac Lake, New York, United States of America
| | - Irah L. King
- Trudeau Institute, Saranac Lake, New York, United States of America
| | - Tori C. Freitas
- Trudeau Institute, Saranac Lake, New York, United States of America
| | - Markus Mohrs
- Trudeau Institute, Saranac Lake, New York, United States of America
| | - Edward J. Pearce
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Trudeau Institute, Saranac Lake, New York, United States of America
- * E-mail:
| |
Collapse
|
150
|
Lammers S, Scott D, Hunter K, Tan W, Shandas R, Stenmark KR. Mechanics and Function of the Pulmonary Vasculature: Implications for Pulmonary Vascular Disease and Right Ventricular Function. Compr Physiol 2012; 2:295-319. [PMID: 23487595 DOI: 10.1002/cphy.c100070] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The relationship between cardiac function and the afterload against which the heart muscle must work to circulate blood throughout the pulmonary circulation is defined by a complex interaction between many coupled system parameters. These parameters range broadly and incorporate system effects originating primarily from three distinct locations: input power from the heart, hydraulic impedance from the large conduit pulmonary arteries, and hydraulic resistance from the more distal microcirculation. These organ systems are not independent, but rather, form a coupled system in which a change to any individual parameter affects all other system parameters. The result is a highly nonlinear system which requires not only detailed study of each specific component and the effect of disease on their specific function, but also requires study of the interconnected relationship between the microcirculation, the conduit arteries, and the heart in response to age and disease. Here, we investigate systems-level changes associated with pulmonary hypertensive disease progression in an effort to better understand this coupled relationship.
Collapse
Affiliation(s)
- Steven Lammers
- Department of Cardiovascular Pulmonary Research, University of Colorado Denver, Aurora, Colorado ; Department of Bioengineering, University of Colorado Denver, Aurora, Colorado
| | | | | | | | | | | |
Collapse
|