51
|
Le Hiress M, Akagah B, Bernadat G, Tu L, Thuillet R, Huertas A, Phan C, Fadel E, Simonneau G, Humbert M, Jalce G, Guignabert C. Design, Synthesis, and Biological Activity of New N-(Phenylmethyl)-benzoxazol-2-thiones as Macrophage Migration Inhibitory Factor (MIF) Antagonists: Efficacies in Experimental Pulmonary Hypertension. J Med Chem 2018. [PMID: 29526099 DOI: 10.1021/acs.jmedchem.7b01312] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Macrophage migration inhibitory factor (MIF) is a key pleiotropic mediator and a promising therapeutic target in cancer as well as in several inflammatory and cardiovascular diseases including pulmonary arterial hypertension (PAH). Here, a novel series of N-(phenylmethyl)-benzoxazol-2-thiones 5-32 designed to target the MIF tautomerase active site was synthesized and evaluated for its effects on cell survival. Investigation of structure-activity relationship (SAR) particularly at the 5-position of the benzoxazole core led to the identification of 31 that potently inhibits cell survival in DU-145 prostate cancer cells and pulmonary endothelial cells derived from patients with idiopathic PAH (iPAH-ECs), two cell lines for which survival is MIF-dependent. Molecular docking studies helped to interpret initial SAR related to MIF tautomerase inhibition and propose preferred binding mode for 31 within the MIF tautomerase active site. Interestingly, daily treatment with 31 started 2 weeks after a subcutaneous monocrotaline injection regressed established pulmonary hypertension in rats.
Collapse
Affiliation(s)
- Morane Le Hiress
- INSERM UMR_S 999, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson , France.,Université Paris-Sud et Université Paris-Saclay , 94270 Le Kremlin-Bicêtre , France
| | - Bernardin Akagah
- MIFCARE , 24 rue du Faubourg Saint-Jacques , 75014 Paris , France
| | - Guillaume Bernadat
- BioCIS , Université Paris-Sud, CNRS, Université Paris-Saclay , 92290 Châtenay-Malabry , France
| | - Ly Tu
- INSERM UMR_S 999, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson , France.,Université Paris-Sud et Université Paris-Saclay , 94270 Le Kremlin-Bicêtre , France
| | - Raphaël Thuillet
- INSERM UMR_S 999, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson , France.,Université Paris-Sud et Université Paris-Saclay , 94270 Le Kremlin-Bicêtre , France
| | - Alice Huertas
- INSERM UMR_S 999, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson , France.,Université Paris-Sud et Université Paris-Saclay , 94270 Le Kremlin-Bicêtre , France.,AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre , France
| | - Carole Phan
- INSERM UMR_S 999, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson , France.,Université Paris-Sud et Université Paris-Saclay , 94270 Le Kremlin-Bicêtre , France
| | - Elie Fadel
- INSERM UMR_S 999, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson , France.,Université Paris-Sud et Université Paris-Saclay , 94270 Le Kremlin-Bicêtre , France
| | - Gérald Simonneau
- INSERM UMR_S 999, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson , France.,Université Paris-Sud et Université Paris-Saclay , 94270 Le Kremlin-Bicêtre , France.,AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre , France
| | - Marc Humbert
- INSERM UMR_S 999, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson , France.,Université Paris-Sud et Université Paris-Saclay , 94270 Le Kremlin-Bicêtre , France.,AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre , France
| | - Gaël Jalce
- MIFCARE , 24 rue du Faubourg Saint-Jacques , 75014 Paris , France
| | - Christophe Guignabert
- INSERM UMR_S 999, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson , France.,Université Paris-Sud et Université Paris-Saclay , 94270 Le Kremlin-Bicêtre , France
| |
Collapse
|
52
|
Folestad E, Kunath A, Wågsäter D. PDGF-C and PDGF-D signaling in vascular diseases and animal models. Mol Aspects Med 2018; 62:1-11. [PMID: 29410092 DOI: 10.1016/j.mam.2018.01.005] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 11/14/2017] [Accepted: 01/22/2018] [Indexed: 01/06/2023]
Abstract
Members of the platelet-derived growth factor (PDGF) family are well known to be involved in different pathological conditions. The cellular and molecular mechanisms induced by the PDGF signaling have been well studied. Nevertheless, there is much more to discover about their functions and some important questions to be answered. This review summarizes the known roles of two of the PDGFs, PDGF-C and PDGF-D, in vascular diseases. There are clear implications for these growth factors in several vascular diseases, such as atherosclerosis and stroke. The PDGF receptors are broadly expressed in the cardiovascular system in cells such as fibroblasts, smooth muscle cells and pericytes. Altered expression of the receptors and the ligands have been found in various cardiovascular diseases and current studies have shown important implications of PDGF-C and PDGF-D signaling in fibrosis, neovascularization, atherosclerosis and restenosis.
Collapse
Affiliation(s)
- Erika Folestad
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Anne Kunath
- Division of Drug Research, Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Dick Wågsäter
- Division of Drug Research, Department of Medical and Health Sciences, Linköping University, Linköping, Sweden.
| |
Collapse
|
53
|
Hensley MK, Levine A, Gladwin MT, Lai YC. Emerging therapeutics in pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2018; 314:L769-L781. [PMID: 29388467 DOI: 10.1152/ajplung.00259.2017] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Pulmonary hypertension (PH) is a progressive and often fatal illness presenting with nonspecific symptoms of dyspnea, lower extremity edema, and exercise intolerance. Pathologically, endothelial dysfunction leads to abnormal intimal and smooth muscle proliferation along with reduced apoptosis, resulting in increased pulmonary vascular resistance and elevated pulmonary pressures. PH is subdivided into five World Health Organization groups based on the disease pathology and specific cause. While there are Food and Drug Administration-approved medications for the treatment of pulmonary arterial hypertension (PAH; Group 1 PH), as well as for chronic thromboembolic PH (Group 4 PH), the morbidity and mortality remain high. Moreover, there are no approved therapies for other forms of PH (Groups 2, 3, and 5) at present. New research has identified molecular targets that mediate vasodilation, anti-inflammatory, and antifibrotic changes within the pulmonary vasculature. Given that PAH is the most commonly studied form of PH worldwide and because recent studies have led to better mechanistic understanding of this devastating disease, in this review we attempt to provide an updated overview of new therapeutic approaches under investigation for the treatment of PH, with a particular focus on PAH, as well as to offer guidelines for future investigations.
Collapse
Affiliation(s)
- Matthew K Hensley
- Division of Pulmonary and Critical Care Medicine, University of Michigan , Ann Arbor, Michigan
| | - Andrea Levine
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania.,Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Mark T Gladwin
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania.,Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Yen-Chun Lai
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania.,Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| |
Collapse
|
54
|
Yang K, Zhao M, Huang J, Zhang C, Zheng Q, Chen Y, Jiang H, Lu W, Wang J. Pharmacological activation of PPARγ inhibits hypoxia-induced proliferation through a caveolin-1-targeted and -dependent mechanism in PASMCs. Am J Physiol Cell Physiol 2018; 314:C428-C438. [PMID: 29351409 DOI: 10.1152/ajpcell.00143.2017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Previously, we and others have demonstrated that activation of peroxisome proliferator-activated receptor γ (PPARγ) by specific pharmacological agonists inhibits the pathogenesis of chronic hypoxia-induced pulmonary hypertension (CHPH) by suppressing the proliferation and migration in distal pulmonary arterial smooth muscle cells (PASMCs). Moreover, these beneficial effects of PPARγ are mediated by targeting the intracellular calcium homeostasis and store-operated calcium channel (SOCC) proteins, including the main caveolae component caveolin-1. However, other than the caveolin-1 targeted mechanism, in this study, we further uncovered a caveolin-1 dependent mechanism within the activation of PPARγ by the specific agonist GW1929. First, effective knockdown of caveolin-1 by small-interfering RNA (siRNA) markedly abolished the upregulation of GW1929 on PPARγ expression at both mRNA and protein levels; Then, in HEK293T, which has previously been reported with low endogenous caveolin-1 expression, exogenous expression of caveolin-1 significantly enhanced the upregulation of GW1929 on PPARγ expression compared with nontransfection control. In addition, inhibition of PPARγ by either siRNA or pharmacological inhibitor T0070907 led to increased phosphorylation of cellular mitogen-activated protein kinases ERK1/2 and p38. In parallel, GW1929 dramatically decreased the expression of the proliferative regulators (cyclin D1 and PCNA), whereas it increased the apoptotic factors (p21, p53, and mdm2) in hypoxic PASMCs. Furthermore, these effects of GW1929 could be partially reversed by recovery of the drug treatment. In combination, PPARγ activation by GW1929 reversibly drove the cell toward an antiproliferative and proapoptotic phenotype in a caveolin-1-dependent and -targeted mechanism.
Collapse
Affiliation(s)
- Kai Yang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University , Guangzhou, Guangdong , China
| | - Mingming Zhao
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health , Baltimore, Maryland
| | - Junyi Huang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University , Guangzhou, Guangdong , China
| | - Chenting Zhang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University , Guangzhou, Guangdong , China
| | - Qiuyu Zheng
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University , Guangzhou, Guangdong , China
| | - Yuqin Chen
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University , Guangzhou, Guangdong , China
| | - Haiyang Jiang
- Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine , Baltimore, Maryland
| | - Wenju Lu
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University , Guangzhou, Guangdong , China
| | - Jian Wang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University , Guangzhou, Guangdong , China.,Division of Translational and Regenerative Medicine, Department of Medicine, University of Arizona College of Medicine , Tucson, Arizona
| |
Collapse
|
55
|
Avouac J, Konstantinova I, Guignabert C, Pezet S, Sadoine J, Guilbert T, Cauvet A, Tu L, Luccarini JM, Junien JL, Broqua P, Allanore Y. Pan-PPAR agonist IVA337 is effective in experimental lung fibrosis and pulmonary hypertension. Ann Rheum Dis 2017; 76:1931-1940. [PMID: 28801346 DOI: 10.1136/annrheumdis-2016-210821] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Revised: 07/07/2017] [Accepted: 07/18/2017] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To evaluate the antifibrotic effects of the pan-peroxisome proliferator-activated receptor (PPAR) agonist IVA337 in preclinical mouse models of pulmonary fibrosis and related pulmonary hypertension (PH). METHODS IVA337 has been evaluated in the mouse model of bleomycin-induced pulmonary fibrosis and in Fra-2 transgenic mice, this latter being characterised by non-specific interstitial pneumonia and severe vascular remodelling of pulmonary arteries leading to PH. Mice received two doses of IVA337 (30 mg/kg or 100 mg/kg) or vehicle administered by daily oral gavage up to 4 weeks. RESULTS IVA337 demonstrated at a dose of 100 mg/kg a marked protection from the development of lung fibrosis in both mouse models compared with mice receiving 30 mg/kg of IVA337 or vehicle. Histological score was markedly reduced by 61% in the bleomycin model and by 50% in Fra-2 transgenic mice, and total lung hydroxyproline concentrations decreased by 28% and 48%, respectively, as compared with vehicle-treated mice. IVA337 at 100 mg/kg also significantly decreased levels of fibrogenic markers in lesional lungs of both mouse models. In addition, IVA337 substantially alleviated PH in Fra-2 transgenic mice by improving haemodynamic measurements and vascular remodelling. In primary human lung fibroblasts, IVA337 inhibited in a dose-dependent manner fibroblast to myofibroblasts transition induced by TGF-β and fibroblast proliferation mediated by PDGF. CONCLUSION We demonstrate that treatment with 100 mg/kg IVA337 prevents lung fibrosis in two complementary animal models and substantially attenuates PH in the Fra-2 mouse model. These findings confirm that the pan-PPAR agonist IVA337 is an appealing therapeutic candidate for these cardiopulmonary involvements.
Collapse
Affiliation(s)
- Jerome Avouac
- Université Paris Descartes, Sorbonne Paris Cité, INSERM U1016, Institut Cochin, CNRS UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité , Service de Rhumatologie A, Hôpital Cochin, Paris, France
| | | | - Christophe Guignabert
- INSERM UMR_S 999, Le Plessis-Robinson, France
- Université Paris-Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Sonia Pezet
- Université Paris Descartes, Sorbonne Paris Cité, INSERM U1016, Institut Cochin, CNRS UMR8104, Paris, France
| | - Jeremy Sadoine
- EA 2496 Pathologie, Imagerieet Biothérapies Orofaciales, UFR Odontologie, Université ParisDescartes and PIDV, PRES Sorbonne Paris Cité, Montrouge, France
| | - Thomas Guilbert
- Université Paris Descartes, Sorbonne Paris Cité, INSERM U1016, Institut Cochin, CNRS UMR8104, Paris, France
| | - Anne Cauvet
- Université Paris Descartes, Sorbonne Paris Cité, INSERM U1016, Institut Cochin, CNRS UMR8104, Paris, France
| | - Ly Tu
- INSERM UMR_S 999, Le Plessis-Robinson, France
- Université Paris-Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | | | | | | | - Yannick Allanore
- Université Paris Descartes, Sorbonne Paris Cité, INSERM U1016, Institut Cochin, CNRS UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité , Service de Rhumatologie A, Hôpital Cochin, Paris, France
- EA 2496 Pathologie, Imagerieet Biothérapies Orofaciales, UFR Odontologie, Université ParisDescartes and PIDV, PRES Sorbonne Paris Cité, Montrouge, France
| |
Collapse
|
56
|
Lecarpentier Y, Schussler O, Claes V, Vallée A. The Myofibroblast: TGFβ-1, A Conductor which Plays a Key Role in Fibrosis by Regulating the Balance between PPARγ and the Canonical WNT Pathway. NUCLEAR RECEPTOR RESEARCH 2017. [DOI: 10.11131/2017/101299] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l’Est Francilien (GHEP), Meaux, France
| | - Olivier Schussler
- Department of Cardiovascular Surgery, Cardiovascular Research Laboratory, HUG/CMU, Geneva, Switzerland
| | - Victor Claes
- Department of Pharmaceutical Sciences, University of Antwerp, Wilrijk, Belgium
| | - Alexandre Vallée
- Experimental and Clinical Neurosciences Laboratory, INSERM U1084, University of Poitiers, Poitiers, France
| |
Collapse
|
57
|
Avouac J, Guignabert C, Hoffmann-Vold AM, Ruiz B, Dorfmuller P, Pezet S, Amar O, Tu L, Van Wassenhove J, Sadoine J, Launay D, Elhai M, Cauvet A, Subramaniam A, Resnick R, Hachulla E, Molberg Ø, Kahan A, Humbert M, Allanore Y. Role of Stromelysin 2 (Matrix Metalloproteinase 10) as a Novel Mediator of Vascular Remodeling Underlying Pulmonary Hypertension Associated With Systemic Sclerosis. Arthritis Rheumatol 2017; 69:2209-2221. [DOI: 10.1002/art.40229] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 08/08/2017] [Indexed: 02/03/2023]
Affiliation(s)
- Jérôme Avouac
- Université Paris Descartes; Sorbonne Paris Cité; INSERM U1016 and CNRS UMR8104; Institut Cochin, and Université Paris Descartes; Sorbonne Paris Cité; Service de Rhumatologie A, Hôpital Cochin; Paris France
| | - Christophe Guignabert
- Inserm; UMR S 999, Plessis Robinson, France, and Université Paris-Sud, Université Paris-Saclay; Le Kremlin Bicêtre France
| | - Anna Maria Hoffmann-Vold
- Oslo University Hospital-Rikshospitalet and Institute of Clinical Medicine; University of Oslo; Oslo Norway
| | - Barbara Ruiz
- Université Paris Descartes; Sorbonne Paris Cité; INSERM U1016 and CNRS UMR8104; Institut Cochin; Paris France
| | - Peter Dorfmuller
- Inserm; UMR S 999, Plessis Robinson, France, and Université Paris-Sud, Université Paris-Saclay; Le Kremlin Bicêtre France
| | - Sonia Pezet
- Université Paris Descartes; Sorbonne Paris Cité; INSERM U1016 and CNRS UMR8104; Institut Cochin; Paris France
| | - Olivia Amar
- Université Paris Descartes; Sorbonne Paris Cité; INSERM U1016 and CNRS UMR8104; Institut Cochin; Paris France
| | - Ly Tu
- Inserm; UMR S 999, Plessis Robinson, France, and Université Paris-Sud, Université Paris-Saclay; Le Kremlin Bicêtre France
| | - Jérôme Van Wassenhove
- Université Paris Descartes; Sorbonne Paris Cité; INSERM U1016 and CNRS UMR8104; Institut Cochin; Paris France
| | - Jérémy Sadoine
- EA 2496 Pathologie; Imagerie et Biothérapies Orofaciales; UFR Odontologie; Université Paris Descartes and PIDV; PRES Sorbonne Paris Cité; Montrouge France
| | - David Launay
- Médecine Interne, Hopital Huriez; Université de Lille; Lille France
| | - Muriel Elhai
- Université Paris Descartes; Sorbonne Paris Cité; INSERM U1016 and CNRS UMR8104; Institut Cochin, and Université Paris Descartes; Sorbonne Paris Cité; Service de Rhumatologie A, Hôpital Cochin; Paris France
| | - Anne Cauvet
- Université Paris Descartes; Sorbonne Paris Cité; INSERM U1016 and CNRS UMR8104; Institut Cochin; Paris France
| | | | | | - Eric Hachulla
- Médecine Interne, Hopital Huriez; Université de Lille; Lille France
| | - Øyvind Molberg
- Oslo University Hospital-Rikshospitalet and Institute of Clinical Medicine; University of Oslo; Oslo Norway
| | - André Kahan
- Université Paris Descartes; Sorbonne Paris Cité; Service de Rhumatologie A, Hôpital Cochin; Paris France
| | - Marc Humbert
- Inserm UMR S 999; Plessis Robinson, France, and Université Paris-Sud; Université Paris-Saclay and AP-HP; Service de Pneumologie; Hôpital Bicêtre; Le Kremlin Bicêtre France
| | - Yannick Allanore
- Université Paris Descartes; Sorbonne Paris Cité; INSERM U1016 and CNRS UMR8104; Institut Cochin, and Université Paris Descartes; Sorbonne Paris Cité; Service de Rhumatologie A, Hôpital Cochin; Paris France
| |
Collapse
|
58
|
Dahan D, Hien TT, Tannenberg P, Ekman M, Rippe C, Boettger T, Braun T, Tran-Lundmark K, Tran PK, Swärd K, Albinsson S. MicroRNA-Dependent Control of Serotonin-Induced Pulmonary Arterial Contraction. J Vasc Res 2017; 54:246-256. [PMID: 28796998 DOI: 10.1159/000478013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 06/02/2017] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Serotonin (5-HT) is considered to play a role in pulmonary arterial hypertension by regulating vascular remodeling and smooth muscle contractility. Here, arteries from mice with inducible and smooth muscle-specific deletion of Dicer were used to address mechanisms by which microRNAs control 5-HT-induced contraction. METHODS Mice were used 5 weeks after Dicer deletion, and pulmonary artery contractility was analyzed by wire myography. RESULTS No change was seen in right ventricular systolic pressure following dicer deletion, but systemic blood pressure was reduced. Enhanced 5-HT-induced contraction in Dicer KO pulmonary arteries was associated with increased 5-HT2A receptor mRNA expression whereas 5-HT1B and 5-HT2B receptor mRNAs were unchanged. Contraction by the 5-HT2A agonist TCB-2 was increased in Dicer KO as was the response to the 5-HT2B agonist BW723C86. Effects of Src and protein kinase C inhibition were similar in control and KO arteries, but the effect of inhibition of Rho kinase was reduced. We identified miR-30c as a potential candidate for 5-HT2A receptor regulation as it repressed 5-HT2A mRNA and protein. CONCLUSION Our findings show that 5-HT receptor signaling in the arterial wall is subject to regulation by microRNAs and that this entails altered 5-HT2A receptor expression and signaling.
Collapse
Affiliation(s)
- Diana Dahan
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
59
|
Kang BY, Park K, Kleinhenz JM, Murphy TC, Sutliff RL, Archer D, Hart CM. Peroxisome Proliferator-Activated Receptor γ Regulates the V-Ets Avian Erythroblastosis Virus E26 Oncogene Homolog 1/microRNA-27a Axis to Reduce Endothelin-1 and Endothelial Dysfunction in the Sickle Cell Mouse Lung. Am J Respir Cell Mol Biol 2017; 56:131-144. [PMID: 27612006 DOI: 10.1165/rcmb.2016-0166oc] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Pulmonary hypertension (PH), a serious complication of sickle cell disease (SCD), causes significant morbidity and mortality. Although a recent study determined that hemin release during hemolysis triggers endothelial dysfunction in SCD, the pathogenesis of SCD-PH remains incompletely defined. This study examines peroxisome proliferator-activated receptor γ (PPARγ) regulation in SCD-PH and endothelial dysfunction. PH and right ventricular hypertrophy were studied in Townes humanized sickle cell (SS) and littermate control (AA) mice. In parallel studies, SS or AA mice were gavaged with the PPARγ agonist, rosiglitazone (RSG), 10 mg/kg/day, or vehicle for 10 days. In vitro, human pulmonary artery endothelial cells (HPAECs) were treated with vehicle or hemin for 72 hours, and selected HPAECs were treated with RSG. SS mice developed PH and right ventricular hypertrophy associated with reduced lung levels of PPARγ and increased levels of microRNA-27a (miR-27a), v-ets avian erythroblastosis virus E26 oncogene homolog 1 (ETS1), endothelin-1 (ET-1), and markers of endothelial dysfunction (platelet/endothelial cell adhesion molecule 1 and E selectin). HPAECs treated with hemin had increased ETS1, miR-27a, ET-1, and endothelial dysfunction and decreased PPARγ levels. These derangements were attenuated by ETS1 knockdown, inhibition of miR-27a, or PPARγ overexpression. In SS mouse lung or in hemin-treated HPAECs, activation of PPARγ with RSG attenuated reductions in PPARγ and increases in miR-27a, ET-1, and markers of endothelial dysfunction. In SCD-PH pathogenesis, ETS1 stimulates increases in miR-27a levels that reduce PPARγ and increase ET-1 and endothelial dysfunction. PPARγ activation attenuated SCD-associated signaling derangements, suggesting a novel therapeutic approach to attenuate SCD-PH pathogenesis.
Collapse
Affiliation(s)
- Bum-Yong Kang
- 1 Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Atlanta, Georgia; and
| | - Kathy Park
- 1 Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Atlanta, Georgia; and
| | - Jennifer M Kleinhenz
- 1 Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Atlanta, Georgia; and
| | - Tamara C Murphy
- 1 Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Atlanta, Georgia; and
| | - Roy L Sutliff
- 1 Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Atlanta, Georgia; and
| | - David Archer
- 2 Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - C Michael Hart
- 1 Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Atlanta, Georgia; and
| |
Collapse
|
60
|
Green DE, Murphy TC, Kang BY, Bedi B, Yuan Z, Sadikot RT, Hart CM. Peroxisome proliferator-activated receptor-γ enhances human pulmonary artery smooth muscle cell apoptosis through microRNA-21 and programmed cell death 4. Am J Physiol Lung Cell Mol Physiol 2017; 313:L371-L383. [PMID: 28522568 DOI: 10.1152/ajplung.00532.2016] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 05/01/2017] [Accepted: 05/11/2017] [Indexed: 02/06/2023] Open
Abstract
Pulmonary hypertension (PH) is a progressive disorder whose cellular pathogenesis involves enhanced smooth muscle cell (SMC) proliferation and resistance to apoptosis signals. Existing evidence demonstrates that the tumor suppressor programmed cell death 4 (PDCD4) affects patterns of cell growth and repair responses in the systemic vasculature following experimental injury. In the current study, the regulation PDCD4 and its functional effects on growth and apoptosis susceptibility in pulmonary artery smooth muscle cells were explored. We previously demonstrated that pharmacological activation of the nuclear transcription factor peroxisome proliferator-activated receptor-γ (PPARγ) attenuated hypoxia-induced proliferation of human pulmonary artery smooth muscle cells (HPASMCs) by inhibiting the expression and mitogenic functions of microRNA-21 (miR-21). In the current study, we hypothesize that PPARγ stimulates PDCD4 expression and HPASMC apoptosis by inhibiting miR-21. Our findings demonstrate that PDCD4 is reduced in the mouse lung upon exposure to chronic hypoxia (10% O2 for 3 wk) and in hypoxia-exposed HPASMCs (1% O2). HPASMC apoptosis was reduced by hypoxia, by miR-21 overexpression, or by siRNA-mediated PPARγ and PDCD4 depletion. Activation of PPARγ inhibited miR-21 expression and resultant proliferation, while restoring PDCD4 levels and apoptosis to baseline. Additionally, pharmacological activation of PPARγ with rosiglitazone enhanced PDCD4 protein expression and apoptosis in a dose-dependent manner as demonstrated by increased annexin V detection by flow cytometry. Collectively, these findings demonstrate that PPARγ confers growth-inhibitory signals in hypoxia-exposed HPASMCs through suppression of miR-21 and the accompanying derepression of PDCD4 that augments HPASMC susceptibility to undergo apoptosis.
Collapse
Affiliation(s)
- David E Green
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Atlanta Veterans Affairs Medical Center/Emory University, Atlanta, Georgia
| | - Tamara C Murphy
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Atlanta Veterans Affairs Medical Center/Emory University, Atlanta, Georgia
| | - Bum-Yong Kang
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Atlanta Veterans Affairs Medical Center/Emory University, Atlanta, Georgia
| | - Brahmchetna Bedi
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Atlanta Veterans Affairs Medical Center/Emory University, Atlanta, Georgia
| | - Zhihong Yuan
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Atlanta Veterans Affairs Medical Center/Emory University, Atlanta, Georgia
| | - Ruxana T Sadikot
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Atlanta Veterans Affairs Medical Center/Emory University, Atlanta, Georgia
| | - C Michael Hart
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Atlanta Veterans Affairs Medical Center/Emory University, Atlanta, Georgia
| |
Collapse
|
61
|
Xie X, Li S, Zhu Y, Liu L, Pan Y, Wang J, Shi W, Song Y, Yang L, Gao L, Zang W, Li M. MicroRNA-27a/b mediates endothelin-1-induced PPARγ reduction and proliferation of pulmonary artery smooth muscle cells. Cell Tissue Res 2017; 369:527-539. [PMID: 28484848 DOI: 10.1007/s00441-017-2625-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 04/05/2017] [Indexed: 11/27/2022]
Abstract
The down-regulation of peroxisome proliferator-activated receptor γ (PPARγ) expression has been found to correlate with the proliferation of pulmonary artery smooth muscle cells (PASMC), pulmonary vascular remodeling and pulmonary hypertension, while the molecular mechanisms underlying PPARγ reduction in PASMC remain largely unclear. The aim of the current study is to address this issue. Endothelin-1 (ET-1) dose- and time-dependently resulted in PPARγ reduction and proliferation of primary cultured rat PASMC, which was accompanied by the activation of nuclear factor-kappaB (NF-κB) and subsequent induction of microRNA-27a/b (miR-27a/b) expression. Chromatin immunoprecipitation assay revealed that NF-κB directly bound to the promoter regions of miR-27a/b. Luciferase reporter assay identified that miR-27a/b directly regulates the expression of PPARγ in PASMC. Further study indicated that the presence of either NF-κB inhibitor pyrrolidinedithiocarbamate or prior silencing miR-27a/b with anti-miRNA oligonucleotides suppressed ET-1-induced PPARγ reduction and proliferation of PASMC, while overexpression of miR-27a/b reduced PPARγ expression and enhanced PASMC proliferation. Taken together, our study demonstrates that ET-1 stimulates miR-27a/b expression by activation of the NF-κB pathway, which in turn results in PPARγ reduction and contributes to ET-1-induced PASMC proliferation.
Collapse
Affiliation(s)
- Xinming Xie
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, No.277, Yanta West Road, Xi'an, Shaanxi, People's Republic of China, 710061
| | - Shaojun Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, No.277, Yanta West Road, Xi'an, Shaanxi, People's Republic of China, 710061
| | - Yanting Zhu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, No.277, Yanta West Road, Xi'an, Shaanxi, People's Republic of China, 710061
| | - Lu Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, No.277, Yanta West Road, Xi'an, Shaanxi, People's Republic of China, 710061
| | - Yilin Pan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, No.277, Yanta West Road, Xi'an, Shaanxi, People's Republic of China, 710061
| | - Jian Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, No.277, Yanta West Road, Xi'an, Shaanxi, People's Republic of China, 710061
| | - Wenhua Shi
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, No.277, Yanta West Road, Xi'an, Shaanxi, People's Republic of China, 710061
| | - Yang Song
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, No.277, Yanta West Road, Xi'an, Shaanxi, People's Republic of China, 710061
| | - Lan Yang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, No.277, Yanta West Road, Xi'an, Shaanxi, People's Republic of China, 710061
| | - Li Gao
- Division of Allergy and Clinical Immunology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
| | - Weijin Zang
- Department of Pharmacology, School of Basic Medical Sciences, Xian Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China, 710061
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, No.277, Yanta West Road, Xi'an, Shaanxi, People's Republic of China, 710061.
| |
Collapse
|
62
|
Calvier L, Chouvarine P, Legchenko E, Hoffmann N, Geldner J, Borchert P, Jonigk D, Mozes MM, Hansmann G. PPARγ Links BMP2 and TGFβ1 Pathways in Vascular Smooth Muscle Cells, Regulating Cell Proliferation and Glucose Metabolism. Cell Metab 2017; 25:1118-1134.e7. [PMID: 28467929 DOI: 10.1016/j.cmet.2017.03.011] [Citation(s) in RCA: 176] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 12/21/2016] [Accepted: 03/20/2017] [Indexed: 01/24/2023]
Abstract
BMP2 and TGFβ1 are functional antagonists of pathological remodeling in the arteries, heart, and lung; however, the mechanisms in VSMCs, and their disturbance in pulmonary arterial hypertension (PAH), are unclear. We found a pro-proliferative TGFβ1-Stat3-FoxO1 axis in VSMCs, and PPARγ as inhibitory regulator of TGFβ1-Stat3-FoxO1 and TGFβ1-Smad3/4, by physically interacting with Stat3 and Smad3. TGFβ1 induces fibrosis-related genes and miR-130a/301b, suppressing PPARγ. Conversely, PPARγ inhibits TGFβ1-induced mitochondrial activation and VSMC proliferation, and regulates two glucose metabolism-related enzymes, platelet isoform of phosphofructokinase (PFKP, a PPARγ target, via miR-331-5p) and protein phosphatase 1 regulatory subunit 3G (PPP1R3G, a Smad3 target). PPARγ knockdown/deletion in VSMCs activates TGFβ1 signaling. The PPARγ agonist pioglitazone reverses PAH and inhibits the TGFβ1-Stat3-FoxO1 axis in TGFβ1-overexpressing mice. We identified PPARγ as a missing link between BMP2 and TGFβ1 pathways in VSMCs. PPARγ activation can be beneficial in TGFβ1-associated diseases, such as PAH, parenchymal lung diseases, and Marfan's syndrome.
Collapse
Affiliation(s)
- Laurent Calvier
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover 30625, Germany
| | - Philippe Chouvarine
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover 30625, Germany
| | - Ekaterina Legchenko
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover 30625, Germany
| | - Nadine Hoffmann
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover 30625, Germany
| | - Jonas Geldner
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover 30625, Germany
| | - Paul Borchert
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover 30625, Germany
| | - Danny Jonigk
- Institute of Pathology, Hannover Medical School, Hannover 30625, Germany
| | - Miklos M Mozes
- Department of Pathophysiology, Semmelweis University, Budapest 1089, Hungary
| | - Georg Hansmann
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover 30625, Germany.
| |
Collapse
|
63
|
Harvey LD, Chan SY. Emerging Metabolic Therapies in Pulmonary Arterial Hypertension. J Clin Med 2017; 6:jcm6040043. [PMID: 28375184 PMCID: PMC5406775 DOI: 10.3390/jcm6040043] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 03/28/2017] [Accepted: 03/29/2017] [Indexed: 12/26/2022] Open
Abstract
Pulmonary hypertension (PH) is an enigmatic vascular disorder characterized by pulmonary vascular remodeling and increased pulmonary vascular resistance, ultimately resulting in pressure overload, dysfunction, and failure of the right ventricle. Current medications for PH do not reverse or prevent disease progression, and current diagnostic strategies are suboptimal for detecting early-stage disease. Thus, there is a substantial need to develop new diagnostics and therapies that target the molecular origins of PH. Emerging investigations have defined metabolic aberrations as fundamental and early components of disease manifestation in both pulmonary vasculature and the right ventricle. As such, the elucidation of metabolic dysregulation in pulmonary hypertension allows for greater therapeutic insight into preventing, halting, or even reversing disease progression. This review will aim to discuss (1) the reprogramming and dysregulation of metabolic pathways in pulmonary hypertension; (2) the emerging therapeutic interventions targeting these metabolic pathways; and (3) further innovation needed to overcome barriers in the treatment of this devastating disease.
Collapse
Affiliation(s)
- Lloyd D Harvey
- Medical Scientist Training Program, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA.
| | - Stephen Y Chan
- Division of Cardiology, Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA.
| |
Collapse
|
64
|
Huetsch JC, Suresh K, Bernier M, Shimoda LA. Update on novel targets and potential treatment avenues in pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2016; 311:L811-L831. [PMID: 27591245 PMCID: PMC5130539 DOI: 10.1152/ajplung.00302.2016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 08/29/2016] [Indexed: 02/08/2023] Open
Abstract
Pulmonary hypertension (PH) is a condition marked by a combination of constriction and remodeling within the pulmonary vasculature. It remains a disease without a cure, as current treatments were developed with a focus on vasodilatory properties but do not reverse the remodeling component. Numerous recent advances have been made in the understanding of cellular processes that drive pathologic remodeling in each layer of the vessel wall as well as the accompanying maladaptive changes in the right ventricle. In particular, the past few years have yielded much improved insight into the pathways that contribute to altered metabolism, mitochondrial function, and reactive oxygen species signaling and how these pathways promote the proproliferative, promigratory, and antiapoptotic phenotype of the vasculature during PH. Additionally, there have been significant advances in numerous other pathways linked to PH pathogenesis, such as sex hormones and perivascular inflammation. Novel insights into cellular pathology have suggested new avenues for the development of both biomarkers and therapies that will hopefully bring us closer to the elusive goal: a therapy leading to reversal of disease.
Collapse
Affiliation(s)
- John C Huetsch
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland; and
| | - Karthik Suresh
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland; and
| | - Meghan Bernier
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Larissa A Shimoda
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland; and
| |
Collapse
|
65
|
Sharma S, Ruffenach G, Umar S, Motayagheni N, Reddy ST, Eghbali M. Role of oxidized lipids in pulmonary arterial hypertension. Pulm Circ 2016; 6:261-73. [PMID: 27683603 DOI: 10.1086/687293] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a multifactorial disease characterized by interplay of many cellular, molecular, and genetic events that lead to excessive proliferation of pulmonary cells, including smooth muscle and endothelial cells; inflammation; and extracellular matrix remodeling. Abnormal vascular changes and structural remodeling associated with PAH culminate in vasoconstriction and obstruction of pulmonary arteries, contributing to increased pulmonary vascular resistance, pulmonary hypertension, and right ventricular failure. The complex molecular mechanisms involved in the pathobiology of PAH are the limiting factors in the development of potential therapeutic interventions for PAH. Over the years, our group and others have demonstrated the critical implication of lipids in the pathogenesis of PAH. This review specifically focuses on the current understanding of the role of oxidized lipids, lipid metabolism, peroxidation, and oxidative stress in the progression of PAH. This review also discusses the relevance of apolipoprotein A-I mimetic peptides and microRNA-193, which are known to regulate the levels of oxidized lipids, as potential therapeutics in PAH.
Collapse
Affiliation(s)
- Salil Sharma
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Grégoire Ruffenach
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Soban Umar
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Negar Motayagheni
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Srinivasa T Reddy
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Mansoureh Eghbali
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|
66
|
Guignabert C, Phan C, Seferian A, Huertas A, Tu L, Thuillet R, Sattler C, Le Hiress M, Tamura Y, Jutant EM, Chaumais MC, Bouchet S, Manéglier B, Molimard M, Rousselot P, Sitbon O, Simonneau G, Montani D, Humbert M. Dasatinib induces lung vascular toxicity and predisposes to pulmonary hypertension. J Clin Invest 2016; 126:3207-18. [PMID: 27482885 DOI: 10.1172/jci86249] [Citation(s) in RCA: 191] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 05/26/2016] [Indexed: 12/15/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a life-threatening disease that can be induced by dasatinib, a dual Src and BCR-ABL tyrosine kinase inhibitor that is used to treat chronic myelogenous leukemia (CML). Today, key questions remain regarding the mechanisms involved in the long-term development of dasatinib-induced PAH. Here, we demonstrated that chronic dasatinib therapy causes pulmonary endothelial damage in humans and rodents. We found that dasatinib treatment attenuated hypoxic pulmonary vasoconstriction responses and increased susceptibility to experimental pulmonary hypertension (PH) in rats, but these effects were absent in rats treated with imatinib, another BCR-ABL tyrosine kinase inhibitor. Furthermore, dasatinib treatment induced pulmonary endothelial cell apoptosis in a dose-dependent manner, while imatinib did not. Dasatinib treatment mediated endothelial cell dysfunction via increased production of ROS that was independent of Src family kinases. Consistent with these findings, we observed elevations in markers of endothelial dysfunction and vascular damage in the serum of CML patients who were treated with dasatinib, compared with CML patients treated with imatinib. Taken together, our findings indicate that dasatinib causes pulmonary vascular damage, induction of ER stress, and mitochondrial ROS production, which leads to increased susceptibility to PH development.
Collapse
|
67
|
Yeligar SM, Mehta AJ, Harris FL, Brown LAS, Hart CM. Peroxisome Proliferator-Activated Receptor γ Regulates Chronic Alcohol-Induced Alveolar Macrophage Dysfunction. Am J Respir Cell Mol Biol 2016; 55:35-46. [PMID: 26677910 PMCID: PMC4942203 DOI: 10.1165/rcmb.2015-0077oc] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 12/15/2015] [Indexed: 12/13/2022] Open
Abstract
Peroxisome proliferator-activated receptor (PPAR) γ is critical for alveolar macrophage (AM) function. Chronic alcohol abuse causes AM phagocytic dysfunction and susceptibility to respiratory infections by stimulating nicotinamide adenine dinucleotide oxidases (Nox), transforming growth factor-β1, and oxidative stress in the AM. Because PPARγ inhibits Nox expression, we hypothesized that alcohol reduces PPARγ, stimulating AM dysfunction. AMs were examined from: (1) patients with alcoholism or control patients; (2) a mouse model of chronic ethanol consumption; (3) PPARγ knockout mice; or (4) MH-S cells exposed to ethanol in vitro. Alcohol reduced AM PPARγ levels and increased Nox1, -2, and -4, transforming growth factor-β1, oxidative stress, and phagocytic dysfunction. Genetic loss of PPARγ recapitulated, whereas stimulating PPARγ activity attenuated alcohol-mediated alterations in gene expression and phagocytic function, supporting the importance of PPARγ in alcohol-induced AM derangements. Similarly, PPARγ activation in vivo reduced alcohol-mediated impairments in lung bacterial clearance. Alcohol increased levels of microRNA-130a/-301a, which bind to the PPARγ 3' untranslated region to reduce PPARγ expression. MicroRNA-130a/-301a inhibition attenuated alcohol-mediated PPARγ reductions and derangements in AM gene expression and function. Alcohol-induced Toll-like receptor 4 endocytosis was reversed by PPARγ activation. These findings demonstrate that targeting PPARγ provides a novel therapeutic approach for mitigating alcohol-induced AM derangements and susceptibility to lung infection.
Collapse
Affiliation(s)
- Samantha M. Yeligar
- Emory University, Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Emory and Children’s Healthcare of Atlanta Center for Developmental Lung Biology, Atlanta, Georgia
- Emory University, Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Decatur, Georgia
| | - Ashish J. Mehta
- Emory University, Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Decatur, Georgia
| | - Frank L. Harris
- Emory University, Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Emory and Children’s Healthcare of Atlanta Center for Developmental Lung Biology, Atlanta, Georgia
| | - Lou Ann S. Brown
- Emory University, Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Emory and Children’s Healthcare of Atlanta Center for Developmental Lung Biology, Atlanta, Georgia
| | - C. Michael Hart
- Emory University, Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Decatur, Georgia
| |
Collapse
|
68
|
Bijli KM, Kang BY, Sutliff RL, Hart CM. Proline-rich tyrosine kinase 2 downregulates peroxisome proliferator-activated receptor gamma to promote hypoxia-induced pulmonary artery smooth muscle cell proliferation. Pulm Circ 2016; 6:202-10. [PMID: 27252847 DOI: 10.1086/686012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Hypoxia stimulates pulmonary hypertension (PH), in part by increasing the proliferation of human pulmonary artery smooth muscle cells (HPASMCs) via sustained activation of mitogen-activated protein kinase, extracellular signal-regulated kinases 1 and 2 (ERK 1/2), and nuclear factor-kappa B (NF-κB); elevated expression of NADPH oxidase 4 (Nox4); and downregulation of peroxisome proliferator-activated receptor gamma (PPARγ) levels. However, the upstream mediators that control these responses remain largely unknown. We hypothesized that proline-rich tyrosine kinase 2 (Pyk2) plays a critical role in the mechanism of hypoxia-induced HPASMC proliferation. To test this hypothesis, HPASMCs were exposed to normoxia or hypoxia (1% O2) for 72 hours. Hypoxia activated Pyk2 (detected as Tyr402 phosphorylation), and inhibition of Pyk2 with small interfering RNA (siRNA) or tyrphostin A9 attenuated hypoxia-induced HPASMC proliferation. Pyk2 inhibition attenuated ERK 1/2 activation as early as 24 hours after the onset of hypoxia, suggesting a proximal role for Pyk2 in this response. Pyk2 inhibition also attenuated hypoxia-induced NF-κB activation, reduced HPASMC PPARγ messenger RNA levels and activity, and increased NF-κB-mediated Nox4 levels. The siRNA-mediated PPARγ knockdown enhanced Pyk2 activation, whereas PPARγ overexpression reduced Pyk2 activation in HPASMCs, confirming a reciprocal relationship between Pyk2 and PPARγ. Pyk2 depletion also attenuated hypoxia-induced NF-κB p65 activation and reduced PPARγ protein levels in human pulmonary artery endothelial cells. These in vitro findings suggest that Pyk2 plays a central role in the proliferative phenotype of pulmonary vascular wall cells under hypoxic conditions. Coupled with recent reports that hypoxia-induced PH is attenuated in Pyk2 knockout mice, these findings suggest that Pyk2 may represent a novel therapeutic target in PH.
Collapse
Affiliation(s)
- Kaiser M Bijli
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Atlanta, Georgia, USA
| | - Bum-Yong Kang
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Atlanta, Georgia, USA
| | - Roy L Sutliff
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Atlanta, Georgia, USA
| | - C Michael Hart
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Atlanta, Georgia, USA
| |
Collapse
|
69
|
Kang BY, Park KK, Kleinhenz JM, Murphy TC, Green DE, Bijli KM, Yeligar SM, Carthan KA, Searles CD, Sutliff RL, Hart CM. Peroxisome Proliferator-Activated Receptor γ and microRNA 98 in Hypoxia-Induced Endothelin-1 Signaling. Am J Respir Cell Mol Biol 2016; 54:136-46. [PMID: 26098770 DOI: 10.1165/rcmb.2014-0337oc] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Endothelin-1 (ET-1) plays a critical role in endothelial dysfunction and contributes to the pathogenesis of pulmonary hypertension (PH). We hypothesized that peroxisome proliferator-activated receptor γ (PPARγ) stimulates microRNAs that inhibit ET-1 and pulmonary artery endothelial cell (PAEC) proliferation. The objective of this study was to clarify molecular mechanisms by which PPARγ regulates ET-1 expression in vitro and in vivo. In PAECs isolated from patients with pulmonary arterial hypertension, microRNA (miR)-98 expression was reduced, and ET-1 protein levels and proliferation were increased. Similarly, hypoxia reduced miR-98 and increased ET-1 levels and PAEC proliferation in vitro. In vivo, hypoxia reduced miR-98 expression and increased ET-1 and proliferating cell nuclear antigen (PCNA) levels in mouse lung, derangements that were aggravated by treatment with the vascular endothelial growth factor receptor antagonist Sugen5416. Reporter assays confirmed that miR-98 binds directly to the ET-1 3'-untranslated region. Compared with littermate control mice, miR-98 levels were reduced and ET-1 and PCNA expression were increased in lungs from endothelial-targeted PPARγ knockout mice, whereas miR-98 levels were increased and ET-1 and PCNA expression was reduced in lungs from endothelial-targeted PPARγ-overexpression mice. Gain or loss of PPARγ function in PAECs in vitro confirmed that alterations in PPARγ were sufficient to regulate miR-98, ET-1, and PCNA expression. Finally, PPARγ activation with rosiglitazone regimens that attenuated hypoxia-induced PH in vivo and human PAEC proliferation in vitro restored miR-98 levels. The results of this study show that PPARγ regulates miR-98 to modulate ET-1 expression and PAEC proliferation. These results further clarify molecular mechanisms by which PPARγ participates in PH pathogenesis and therapy.
Collapse
Affiliation(s)
- Bum-Yong Kang
- Department of Medicine, Atlanta Veterans Affairs, and Emory University Medical Centers, Atlanta, Georgia
| | - Kathy K Park
- Department of Medicine, Atlanta Veterans Affairs, and Emory University Medical Centers, Atlanta, Georgia
| | - Jennifer M Kleinhenz
- Department of Medicine, Atlanta Veterans Affairs, and Emory University Medical Centers, Atlanta, Georgia
| | - Tamara C Murphy
- Department of Medicine, Atlanta Veterans Affairs, and Emory University Medical Centers, Atlanta, Georgia
| | - David E Green
- Department of Medicine, Atlanta Veterans Affairs, and Emory University Medical Centers, Atlanta, Georgia
| | - Kaiser M Bijli
- Department of Medicine, Atlanta Veterans Affairs, and Emory University Medical Centers, Atlanta, Georgia
| | - Samantha M Yeligar
- Department of Medicine, Atlanta Veterans Affairs, and Emory University Medical Centers, Atlanta, Georgia
| | - Kristal A Carthan
- Department of Medicine, Atlanta Veterans Affairs, and Emory University Medical Centers, Atlanta, Georgia
| | - Charles D Searles
- Department of Medicine, Atlanta Veterans Affairs, and Emory University Medical Centers, Atlanta, Georgia
| | - Roy L Sutliff
- Department of Medicine, Atlanta Veterans Affairs, and Emory University Medical Centers, Atlanta, Georgia
| | - C Michael Hart
- Department of Medicine, Atlanta Veterans Affairs, and Emory University Medical Centers, Atlanta, Georgia
| |
Collapse
|
70
|
Hopper RK, Moonen JRAJ, Diebold I, Cao A, Rhodes CJ, Tojais NF, Hennigs JK, Gu M, Wang L, Rabinovitch M. In Pulmonary Arterial Hypertension, Reduced BMPR2 Promotes Endothelial-to-Mesenchymal Transition via HMGA1 and Its Target Slug. Circulation 2016; 133:1783-94. [PMID: 27045138 DOI: 10.1161/circulationaha.115.020617] [Citation(s) in RCA: 187] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 03/11/2016] [Indexed: 02/05/2023]
Abstract
BACKGROUND We previously reported high-throughput RNA sequencing analyses that identified heightened expression of the chromatin architectural factor High Mobility Group AT-hook 1 (HMGA1) in pulmonary arterial endothelial cells (PAECs) from patients who had idiopathic pulmonary arterial hypertension (PAH) in comparison with controls. Because HMGA1 promotes epithelial-to-mesenchymal transition in cancer, we hypothesized that increased HMGA1 could induce transition of PAECs to a smooth muscle (SM)-like mesenchymal phenotype (endothelial-to-mesenchymal transition), explaining both dysregulation of PAEC function and possible cellular contribution to the occlusive remodeling that characterizes advanced idiopathic PAH. METHODS AND RESULTS We documented increased HMGA1 in PAECs cultured from idiopathic PAH versus donor control lungs. Confocal microscopy of lung explants localized the increase in HMGA1 consistently to pulmonary arterial endothelium, and identified many cells double-positive for HMGA1 and SM22α in occlusive and plexogenic lesions. Because decreased expression and function of bone morphogenetic protein receptor 2 (BMPR2) is observed in PAH, we reduced BMPR2 by small interfering RNA in control PAECs and documented an increase in HMGA1 protein. Consistent with transition of PAECs by HMGA1, we detected reduced platelet endothelial cell adhesion molecule 1 (CD31) and increased endothelial-to-mesenchymal transition markers, αSM actin, SM22α, calponin, phospho-vimentin, and Slug. The transition was associated with spindle SM-like morphology, and the increase in αSM actin was largely reversed by joint knockdown of BMPR2 and HMGA1 or Slug. Pulmonary endothelial cells from mice with endothelial cell-specific loss of Bmpr2 showed similar gene and protein changes. CONCLUSIONS Increased HMGA1 in PAECs resulting from dysfunctional BMPR2 signaling can transition endothelium to SM-like cells associated with PAH.
Collapse
Affiliation(s)
- Rachel K Hopper
- From Department of Pediatrics, the Vera Moulton Wall Center for Pulmonary Vascular Disease and the Cardiovascular Institute, Stanford University School of Medicine, CA (R.K.H., J.-R.A.J.M., A.C., C.J.R., N.F.T., J.K.H., M.G., L.W., M.R.); Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania and Children's Hospital of Philadelphia (R.K.H.); Center for Congenital Heart Diseases, Pediatric Cardiology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, The Netherlands (J.-R.A.J.M.)
| | - Jan-Renier A J Moonen
- From Department of Pediatrics, the Vera Moulton Wall Center for Pulmonary Vascular Disease and the Cardiovascular Institute, Stanford University School of Medicine, CA (R.K.H., J.-R.A.J.M., A.C., C.J.R., N.F.T., J.K.H., M.G., L.W., M.R.); Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania and Children's Hospital of Philadelphia (R.K.H.); Center for Congenital Heart Diseases, Pediatric Cardiology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, The Netherlands (J.-R.A.J.M.)
| | - Isabel Diebold
- From Department of Pediatrics, the Vera Moulton Wall Center for Pulmonary Vascular Disease and the Cardiovascular Institute, Stanford University School of Medicine, CA (R.K.H., J.-R.A.J.M., A.C., C.J.R., N.F.T., J.K.H., M.G., L.W., M.R.); Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania and Children's Hospital of Philadelphia (R.K.H.); Center for Congenital Heart Diseases, Pediatric Cardiology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, The Netherlands (J.-R.A.J.M.)
| | - Aiqin Cao
- From Department of Pediatrics, the Vera Moulton Wall Center for Pulmonary Vascular Disease and the Cardiovascular Institute, Stanford University School of Medicine, CA (R.K.H., J.-R.A.J.M., A.C., C.J.R., N.F.T., J.K.H., M.G., L.W., M.R.); Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania and Children's Hospital of Philadelphia (R.K.H.); Center for Congenital Heart Diseases, Pediatric Cardiology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, The Netherlands (J.-R.A.J.M.)
| | - Christopher J Rhodes
- From Department of Pediatrics, the Vera Moulton Wall Center for Pulmonary Vascular Disease and the Cardiovascular Institute, Stanford University School of Medicine, CA (R.K.H., J.-R.A.J.M., A.C., C.J.R., N.F.T., J.K.H., M.G., L.W., M.R.); Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania and Children's Hospital of Philadelphia (R.K.H.); Center for Congenital Heart Diseases, Pediatric Cardiology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, The Netherlands (J.-R.A.J.M.)
| | - Nancy F Tojais
- From Department of Pediatrics, the Vera Moulton Wall Center for Pulmonary Vascular Disease and the Cardiovascular Institute, Stanford University School of Medicine, CA (R.K.H., J.-R.A.J.M., A.C., C.J.R., N.F.T., J.K.H., M.G., L.W., M.R.); Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania and Children's Hospital of Philadelphia (R.K.H.); Center for Congenital Heart Diseases, Pediatric Cardiology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, The Netherlands (J.-R.A.J.M.)
| | - Jan K Hennigs
- From Department of Pediatrics, the Vera Moulton Wall Center for Pulmonary Vascular Disease and the Cardiovascular Institute, Stanford University School of Medicine, CA (R.K.H., J.-R.A.J.M., A.C., C.J.R., N.F.T., J.K.H., M.G., L.W., M.R.); Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania and Children's Hospital of Philadelphia (R.K.H.); Center for Congenital Heart Diseases, Pediatric Cardiology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, The Netherlands (J.-R.A.J.M.)
| | - Mingxia Gu
- From Department of Pediatrics, the Vera Moulton Wall Center for Pulmonary Vascular Disease and the Cardiovascular Institute, Stanford University School of Medicine, CA (R.K.H., J.-R.A.J.M., A.C., C.J.R., N.F.T., J.K.H., M.G., L.W., M.R.); Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania and Children's Hospital of Philadelphia (R.K.H.); Center for Congenital Heart Diseases, Pediatric Cardiology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, The Netherlands (J.-R.A.J.M.)
| | - Lingli Wang
- From Department of Pediatrics, the Vera Moulton Wall Center for Pulmonary Vascular Disease and the Cardiovascular Institute, Stanford University School of Medicine, CA (R.K.H., J.-R.A.J.M., A.C., C.J.R., N.F.T., J.K.H., M.G., L.W., M.R.); Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania and Children's Hospital of Philadelphia (R.K.H.); Center for Congenital Heart Diseases, Pediatric Cardiology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, The Netherlands (J.-R.A.J.M.)
| | - Marlene Rabinovitch
- From Department of Pediatrics, the Vera Moulton Wall Center for Pulmonary Vascular Disease and the Cardiovascular Institute, Stanford University School of Medicine, CA (R.K.H., J.-R.A.J.M., A.C., C.J.R., N.F.T., J.K.H., M.G., L.W., M.R.); Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania and Children's Hospital of Philadelphia (R.K.H.); Center for Congenital Heart Diseases, Pediatric Cardiology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, The Netherlands (J.-R.A.J.M.).
| |
Collapse
|
71
|
Werner F, Kojonazarov B, Gaßner B, Abeßer M, Schuh K, Völker K, Baba HA, Dahal BK, Schermuly RT, Kuhn M. Endothelial actions of atrial natriuretic peptide prevent pulmonary hypertension in mice. Basic Res Cardiol 2016; 111:22. [PMID: 26909880 PMCID: PMC4766231 DOI: 10.1007/s00395-016-0541-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 02/16/2016] [Indexed: 11/30/2022]
Abstract
The cardiac hormone atrial natriuretic peptide (ANP) regulates systemic and pulmonary arterial blood pressure by activation of its cyclic GMP-producing guanylyl cyclase-A (GC-A) receptor. In the lung, these hypotensive effects were mainly attributed to smooth muscle-mediated vasodilatation. It is unknown whether pulmonary endothelial cells participate in the homeostatic actions of ANP. Therefore, we analyzed GC-A/cGMP signalling in lung endothelial cells and the cause and functional impact of lung endothelial GC-A dysfunction. Western blot and cGMP determinations showed that cultured human and murine pulmonary endothelial cells exhibit prominent GC-A expression and activity which were markedly blunted by hypoxia, a condition known to trigger pulmonary hypertension (PH). To elucidate the consequences of impaired endothelial ANP signalling, we studied mice with genetic endothelial cell-restricted ablation of the GC-A receptor (EC GC-A KO). Notably, EC GC-A KO mice exhibit PH already under resting, normoxic conditions, with enhanced muscularization of small arteries and perivascular infiltration of inflammatory cells. These alterations were aggravated on exposure of mice to chronic hypoxia. Lung endothelial GC-A dysfunction was associated with enhanced expression of angiotensin converting enzyme (ACE) and increased pulmonary levels of Angiotensin II. Angiotensin II/AT1-blockade with losartan reversed pulmonary vascular remodelling and perivascular inflammation of EC GC-A KO mice, and prevented their increment by chronic hypoxia. This experimental study indicates that endothelial effects of ANP are critical to prevent pulmonary vascular remodelling and PH. Chronic endothelial ANP/GC-A dysfunction, e.g. provoked by hypoxia, is associated with activation of the ACE-angiotensin pathway in the lung and PH.
Collapse
Affiliation(s)
- Franziska Werner
- Physiologisches Institut der Universität Würzburg, Röntgenring 9, 97070, Würzburg, Germany
| | - Baktybek Kojonazarov
- Department of Internal Medicine, University of Gießen and Marburg Lung Center (UGMLC), Justus-Liebig University Gießen, Giessen, Germany.,German Center for Lung Research, Heidelberg, Germany
| | - Birgit Gaßner
- Physiologisches Institut der Universität Würzburg, Röntgenring 9, 97070, Würzburg, Germany
| | - Marco Abeßer
- Physiologisches Institut der Universität Würzburg, Röntgenring 9, 97070, Würzburg, Germany
| | - Kai Schuh
- Physiologisches Institut der Universität Würzburg, Röntgenring 9, 97070, Würzburg, Germany
| | - Katharina Völker
- Physiologisches Institut der Universität Würzburg, Röntgenring 9, 97070, Würzburg, Germany
| | - Hideo A Baba
- Institute of Pathology, University Hospital of Essen, University of Duisburg-Essen, Essen, Germany
| | - Bhola K Dahal
- Department of Internal Medicine, University of Gießen and Marburg Lung Center (UGMLC), Justus-Liebig University Gießen, Giessen, Germany.,German Center for Lung Research, Heidelberg, Germany
| | - Ralph T Schermuly
- Department of Internal Medicine, University of Gießen and Marburg Lung Center (UGMLC), Justus-Liebig University Gießen, Giessen, Germany.,German Center for Lung Research, Heidelberg, Germany
| | - Michaela Kuhn
- Physiologisches Institut der Universität Würzburg, Röntgenring 9, 97070, Würzburg, Germany.
| |
Collapse
|
72
|
Le Hiress M, Tu L, Ricard N, Phan C, Thuillet R, Fadel E, Dorfmüller P, Montani D, de Man F, Humbert M, Huertas A, Guignabert C. Proinflammatory Signature of the Dysfunctional Endothelium in Pulmonary Hypertension. Role of the Macrophage Migration Inhibitory Factor/CD74 Complex. Am J Respir Crit Care Med 2016. [PMID: 26203495 DOI: 10.1164/rccm.201402-0322oc] [Citation(s) in RCA: 142] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Inflammation and endothelial dysfunction are considered two primary instigators of pulmonary arterial hypertension (PAH). CD74 is a receptor for the proinflammatory cytokine macrophage migration inhibitory factor (MIF). This ligand/receptor complex initiates survival pathways and cell proliferation, and it triggers the synthesis and secretion of major proinflammatory factors and cell adhesion molecules. OBJECTIVES We hypothesized that the MIF/CD74 signaling pathway is overexpressed in idiopathic PAH (iPAH) and contributes to a proinflammatory endothelial cell (EC) phenotype. METHODS Primary early passage cultures of human ECs isolated from lung tissues obtained from patients with iPAH and controls were examined for their ability to secrete proinflammatory mediators and bind inflammatory cells with or without modulation of the functional activities of the MIF/CD74 complex. In addition, we tested the efficacies of curative treatments with either the MIF antagonist ISO-1 or anti-CD74 neutralizing antibodies on the aberrant proinflammatory EC phenotype in vitro and in vivo and on the progression of monocrotaline-induced pulmonary hypertension. MEASUREMENTS AND MAIN RESULTS In human lung tissues, intercellular adhesion molecule-1, vascular cell adhesion molecule-1, and E-selectin expressions are markedly up-regulated in the endothelium of distal iPAH pulmonary arteries. Circulating MIF levels are increased in the serum of patients with PAH compared with control subjects, and T-cell lymphocytes represent a source of this overabundance. In addition, CD74 is highly expressed in the endothelium of muscularized pulmonary arterioles and in cultured pulmonary ECs from iPAH, contributing to an exaggerated recruitment of peripheral blood mononuclear cells to pulmonary iPAH ECs. Finally, we found that curative treatments with the MIF antagonist ISO-1 or anti-CD74 neutralizing antibodies partially reversed development of pulmonary hypertension in rats and substantially reduced inflammatory cell infiltration. CONCLUSIONS We report here that CD74 and MIF are markedly increased and activated in patients with iPAH, contributing to the abnormal proinflammatory phenotype of pulmonary ECs in iPAH.
Collapse
Affiliation(s)
- Morane Le Hiress
- 1 INSERM UMR_S 999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France.,2 Université Paris-Sud and Université Paris-Saclay, School of Médecine, Kremlin-Bicêtre, France
| | - Ly Tu
- 1 INSERM UMR_S 999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France.,2 Université Paris-Sud and Université Paris-Saclay, School of Médecine, Kremlin-Bicêtre, France
| | - Nicolas Ricard
- 1 INSERM UMR_S 999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France.,2 Université Paris-Sud and Université Paris-Saclay, School of Médecine, Kremlin-Bicêtre, France
| | - Carole Phan
- 1 INSERM UMR_S 999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France.,2 Université Paris-Sud and Université Paris-Saclay, School of Médecine, Kremlin-Bicêtre, France
| | - Raphaël Thuillet
- 1 INSERM UMR_S 999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France.,2 Université Paris-Sud and Université Paris-Saclay, School of Médecine, Kremlin-Bicêtre, France
| | - Elie Fadel
- 1 INSERM UMR_S 999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France.,2 Université Paris-Sud and Université Paris-Saclay, School of Médecine, Kremlin-Bicêtre, France
| | - Peter Dorfmüller
- 1 INSERM UMR_S 999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France.,2 Université Paris-Sud and Université Paris-Saclay, School of Médecine, Kremlin-Bicêtre, France
| | - David Montani
- 1 INSERM UMR_S 999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France.,2 Université Paris-Sud and Université Paris-Saclay, School of Médecine, Kremlin-Bicêtre, France.,3 AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital de Bicêtre, Kremlin-Bicêtre, France; and
| | - Frances de Man
- 4 Department of Pulmonology, VU University Medical Center/Institute of Cardiovascular Research, Amsterdam, the Netherlands
| | - Marc Humbert
- 1 INSERM UMR_S 999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France.,2 Université Paris-Sud and Université Paris-Saclay, School of Médecine, Kremlin-Bicêtre, France.,3 AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital de Bicêtre, Kremlin-Bicêtre, France; and
| | - Alice Huertas
- 1 INSERM UMR_S 999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France.,2 Université Paris-Sud and Université Paris-Saclay, School of Médecine, Kremlin-Bicêtre, France.,3 AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital de Bicêtre, Kremlin-Bicêtre, France; and
| | - Christophe Guignabert
- 1 INSERM UMR_S 999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France.,2 Université Paris-Sud and Université Paris-Saclay, School of Médecine, Kremlin-Bicêtre, France
| |
Collapse
|
73
|
Vattulainen-Collanus S, Akinrinade O, Li M, Koskenvuo M, Li CG, Rao SP, de Jesus Perez V, Yuan K, Sawada H, Koskenvuo JW, Alvira C, Rabinovitch M, Alastalo TP. Loss of PPARγ in endothelial cells leads to impaired angiogenesis. J Cell Sci 2016; 129:693-705. [PMID: 26743080 DOI: 10.1242/jcs.169011] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 12/30/2015] [Indexed: 12/21/2022] Open
Abstract
Tie2-promoter-mediated loss of peroxisome proliferator-activated receptor gamma (PPARγ, also known as PPARG) in mice leads to osteopetrosis and pulmonary arterial hypertension. Vascular disease is associated with loss of PPARγ in pulmonary microvascular endothelial cells (PMVEC); we evaluated the role of PPARγ in PMVEC functions, such as angiogenesis and migration. The role of PPARγ in angiogenesis was evaluated in Tie2CrePPARγ(flox/flox) and wild-type mice, and in mouse and human PMVECs. RNA sequencing and bioinformatic approaches were utilized to reveal angiogenesis-associated targets for PPARγ. Tie2CrePPARγ(flox/flox) mice showed an impaired angiogenic capacity. Analysis of endothelial progenitor-like cells using bone marrow transplantation combined with evaluation of isolated PMVECs revealed that loss of PPARγ attenuates the migration and angiogenic capacity of mature PMVECs. PPARγ-deficient human PMVECs showed a similar migration defect in culture. Bioinformatic and experimental analyses newly revealed E2F1 as a target of PPARγ in the regulation of PMVEC migration. Disruption of the PPARγ-E2F1 axis was associated with a dysregulated Wnt pathway related to the GSK3B interacting protein (GSKIP). In conclusion, PPARγ plays an important role in sustaining angiogenic potential in mature PMVECs through E2F1-mediated gene regulation.
Collapse
Affiliation(s)
- Sanna Vattulainen-Collanus
- Children's Hospital Helsinki, Pediatric Cardiology, University of Helsinki and Helsinki University Central Hospital, Helsinki 00290, Finland
| | - Oyediran Akinrinade
- Children's Hospital Helsinki, Pediatric Cardiology, University of Helsinki and Helsinki University Central Hospital, Helsinki 00290, Finland Institute of Biomedicine, University of Helsinki, Helsinki 00290, Finland
| | - Molong Li
- The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA Research Center of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku 20520, Finland
| | - Minna Koskenvuo
- Children's Hospital Helsinki, Division of Hematology-Oncology and Stem Cell Transplantation, University of Helsinki and Helsinki University Central Hospital, 00290 Helsinki, Finland
| | - Caiyun Grace Li
- Department of Pediatrics, Wall Center for Pulmonary Vascular Disease, Cardiovascular Institute Stanford University, Stanford, CA 94305, USA
| | - Shailaja P Rao
- Department of Pediatrics, Wall Center for Pulmonary Vascular Disease, Cardiovascular Institute Stanford University, Stanford, CA 94305, USA
| | - Vinicio de Jesus Perez
- Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, CA 94305, USA
| | - Ke Yuan
- Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, CA 94305, USA
| | - Hirofumi Sawada
- Department of Pediatrics, Wall Center for Pulmonary Vascular Disease, Cardiovascular Institute Stanford University, Stanford, CA 94305, USA Department of Pediatrics, Mie University Graduate School of Medicine, Mie 5148507, Japan
| | - Juha W Koskenvuo
- Research Center of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku 20520, Finland Department of Clinical Physiology and Nuclear Medicine, HUS Medical Imaging Center, Helsinki University Central Hospital and University of Helsinki, 00290 Helsinki, Finland
| | - Cristina Alvira
- Department of Pediatrics, Wall Center for Pulmonary Vascular Disease, Cardiovascular Institute Stanford University, Stanford, CA 94305, USA
| | - Marlene Rabinovitch
- Department of Pediatrics, Wall Center for Pulmonary Vascular Disease, Cardiovascular Institute Stanford University, Stanford, CA 94305, USA
| | - Tero-Pekka Alastalo
- Children's Hospital Helsinki, Pediatric Cardiology, University of Helsinki and Helsinki University Central Hospital, Helsinki 00290, Finland
| |
Collapse
|
74
|
Hu W, Huang Y. Targeting the platelet-derived growth factor signalling in cardiovascular disease. Clin Exp Pharmacol Physiol 2015; 42:1221-4. [DOI: 10.1111/1440-1681.12478] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 08/03/2015] [Accepted: 08/08/2015] [Indexed: 01/08/2023]
Affiliation(s)
- Weining Hu
- Shenzhen Research Institute; Institute of Vascular Medicine and Li Ka Shing Institute of Health Sciences; Chinese University of Hong Kong; Hong Kong China
| | - Yu Huang
- Shenzhen Research Institute; Institute of Vascular Medicine and Li Ka Shing Institute of Health Sciences; Chinese University of Hong Kong; Hong Kong China
| |
Collapse
|
75
|
Hou Y, Liu M, Husted C, Chen C, Thiagarajan K, Johns JL, Rao SP, Alvira CM. Activation of the nuclear factor-κB pathway during postnatal lung inflammation preserves alveolarization by suppressing macrophage inflammatory protein-2. Am J Physiol Lung Cell Mol Physiol 2015; 309:L593-604. [PMID: 26163511 DOI: 10.1152/ajplung.00029.2015] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 07/06/2015] [Indexed: 11/22/2022] Open
Abstract
A significant portion of lung development is completed postnatally during alveolarization, rendering the immature lung vulnerable to inflammatory stimuli that can disrupt lung structure and function. Although the NF-κB pathway has well-recognized pro-inflammatory functions, novel anti-inflammatory and developmental roles for NF-κB have recently been described. Thus, to determine how NF-κB modulates alveolarization during inflammation, we exposed postnatal day 6 mice to vehicle (PBS), systemic lipopolysaccharide (LPS), or the combination of LPS and the global NF-κB pathway inhibitor BAY 11-7082 (LPS + BAY). LPS impaired alveolarization, decreased lung cell proliferation, and reduced epithelial growth factor expression. BAY exaggerated these detrimental effects of LPS, further suppressing proliferation and disrupting pulmonary angiogenesis, an essential component of alveolarization. The more severe pathology induced by LPS + BAY was associated with marked increases in lung and plasma levels of macrophage inflammatory protein-2 (MIP-2). Experiments using primary neonatal pulmonary endothelial cells (PEC) demonstrated that MIP-2 directly impaired neonatal PEC migration in vitro; and neutralization of MIP-2 in vivo preserved lung cell proliferation and pulmonary angiogenesis and prevented the more severe alveolar disruption induced by the combined treatment of LPS + BAY. Taken together, these studies demonstrate a key anti-inflammatory function of the NF-κB pathway in the early alveolar lung that functions to mitigate the detrimental effects of inflammation on pulmonary angiogenesis and alveolarization. Furthermore, these data suggest that neutralization of MIP-2 may represent a novel therapeutic target that could be beneficial in preserving lung growth in premature infants exposed to inflammatory stress.
Collapse
Affiliation(s)
- Yanli Hou
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Min Liu
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Cristiana Husted
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California; Department of Biochemistry, Faculty of Medicine, University of Nevada/Reno, Reno, Nevada; and
| | - Chihhsin Chen
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Kavitha Thiagarajan
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Jennifer L Johns
- Department of Comparative Medicine, Stanford University School of Medicine, Stanford, California
| | - Shailaja P Rao
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Cristina M Alvira
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California; Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California;
| |
Collapse
|
76
|
Clapp LH, Gurung R. The mechanistic basis of prostacyclin and its stable analogues in pulmonary arterial hypertension: Role of membrane versus nuclear receptors. Prostaglandins Other Lipid Mediat 2015; 120:56-71. [PMID: 25917921 DOI: 10.1016/j.prostaglandins.2015.04.007] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 04/13/2015] [Indexed: 12/22/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease of distal pulmonary arteries in which patients suffer from elevated pulmonary arterial pressure, extensive vascular remodelling and right ventricular failure. To date prostacyclin (PGI2) therapy remains the most efficacious treatment for PAH and is the only approved monotherapy to have a positive impact on long-term survival. A key thing to note is that improvement exceeds that predicted from vasodilator testing strongly suggesting that additional mechanisms contribute to the therapeutic benefit of prostacyclins in PAH. Given these agents have potent antiproliferative, anti-inflammatory and endothelial regenerating properties suggests therapeutic benefit might result from a slowing, stabilization or even some reversal of vascular remodelling in vivo. This review discusses evidence that the pharmacology of each prostacyclin (IP) receptor agonist so far developed is distinct, with non-IP receptor targets clearly contributing to the therapeutic and side effect profile of PGI2 (EP3), iloprost (EP1), treprostinil (EP2, DP1) along with a family of nuclear receptors known as peroxisome proliferator-activated receptors (PPARs), to which PGI2 and some analogues directly bind. These targets are functionally expressed to varying degrees in arteries, veins, platelets, fibroblasts and inflammatory cells and are likely to be involved in the biological actions of prostacylins. Recently, a highly selective IP agonist, selexipag has been developed for PAH. This agent should prove useful in distinguishing IP from other prostanoid receptors or PPAR binding effects in human tissue. It remains to be determined whether selectivity for the IP receptor gives rise to a superior or inferior clinical benefit in PAH.
Collapse
Affiliation(s)
- Lucie H Clapp
- Department of Medicine, UCL, Rayne Building, London WC1E 6JF, UK.
| | - Rijan Gurung
- Department of Medicine, UCL, Rayne Building, London WC1E 6JF, UK
| |
Collapse
|
77
|
Stoynev N, Dimova I, Rukova B, Hadjidekova S, Nikolova D, Toncheva D, Tankova T. Gene expression in peripheral blood of patients with hypertension and patients with type 2 diabetes. J Cardiovasc Med (Hagerstown) 2015; 15:702-9. [PMID: 23337395 DOI: 10.2459/jcm.0b013e32835dbcc8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
AIM To evaluate the expression of atherosclerosis-associated genes in patients with hypertension and type 2 diabetes mellitus. MATERIAL AND METHODS Twenty-seven patients (14 men, 13 women), mean age 43.26 ± 11.69 years, were included in the study, which was divided into three groups: group 1 - patients with newly diagnosed hypertension and normal glucose tolerance (n = 9), group 2 - normotensive individuals with newly diagnosed type 2 diabetes (n = 9), and control group - normotensive individuals with normal glucose tolerance (n = 9). Gene expression analysis was performed with Human Atherosclerosis RT2 Profiler PCR Array. RESULTS In patients with hypertension, we found eight genes with increased expression - FABP3, FAS, FN1, IL1R2, LPL, SERPINE1, TGFB1, and VCAM1. Decreased expression was observed for two genes - SELPLG and SERPINEB2. In patients with type 2 diabetes we found seven up-regulated genes - APOE, BAX, MMP1, NFKB1, PDGFB, SPP1, and TGFB2, whereas no specifically down-regulated genes were observed. Three genes - KLF2, PDGFRB, and PPARD were found to be expressed only in groups 1 and 2. CONCLUSION Hypertension is associated with increased expression of FABP3, FAS, FN1, IL1R2, LPL, SERPINE1, TGFB1, and VCAM1 and decreased expression of SELPLG and SERPINEB2. The up-regulation of FAS, FN1, SERPINE1, TGFB1, and VCAM1 might be associated with an increased cardiovascular risk. Type 2 diabetes is associated with increased expression of APOE, BAX, MMP1, NFKB1, PDGFB, SPP1, and TGFB2. KLF2 and PPARD might be part of protective mechanisms that limit target organ damage in both disease conditions. Expression of PDGFRB might play an important role in the pathogenesis of both hypertension and type 2 diabetes.
Collapse
Affiliation(s)
- Nikolay Stoynev
- aDepartment of Diabetology, Clinical Center of Endocrinology bDepartment of Medical Genetics cDepartment of Physiology, Medical University Sofia, Bulgaria
| | | | | | | | | | | | | |
Collapse
|
78
|
Diebold I, Hennigs JK, Miyagawa K, Li CG, Nickel NP, Kaschwich M, Cao A, Wang L, Reddy S, Chen PI, Nakahira K, Alcazar MAA, Hopper RK, Ji L, Feldman BJ, Rabinovitch M. BMPR2 preserves mitochondrial function and DNA during reoxygenation to promote endothelial cell survival and reverse pulmonary hypertension. Cell Metab 2015; 21:596-608. [PMID: 25863249 PMCID: PMC4394191 DOI: 10.1016/j.cmet.2015.03.010] [Citation(s) in RCA: 178] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 12/19/2014] [Accepted: 03/19/2015] [Indexed: 01/17/2023]
Abstract
Mitochondrial dysfunction, inflammation, and mutant bone morphogenetic protein receptor 2 (BMPR2) are associated with pulmonary arterial hypertension (PAH), an incurable disease characterized by pulmonary arterial (PA) endothelial cell (EC) apoptosis, decreased microvessels, and occlusive vascular remodeling. We hypothesized that reduced BMPR2 induces PAEC mitochondrial dysfunction, promoting a pro-inflammatory or pro-apoptotic state. Mice with EC deletion of BMPR2 develop hypoxia-induced pulmonary hypertension that, in contrast to non-transgenic littermates, does not reverse upon reoxygenation and is associated with reduced PA microvessels and lung EC p53, PGC1α and TFAM, regulators of mitochondrial biogenesis, and mitochondrial DNA. Decreasing PAEC BMPR2 by siRNA during reoxygenation represses p53, PGC1α, NRF2, TFAM, mitochondrial membrane potential, and ATP and induces mitochondrial DNA deletion and apoptosis. Reducing PAEC BMPR2 in normoxia increases p53, PGC1α, TFAM, mitochondrial membrane potential, ATP production, and glycolysis, and induces mitochondrial fission and a pro-inflammatory state. These features are recapitulated in PAECs from PAH patients with mutant BMPR2.
Collapse
Affiliation(s)
- Isabel Diebold
- Department of Pediatrics and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; The Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jan K Hennigs
- Department of Pediatrics and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; The Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kazuya Miyagawa
- Department of Pediatrics and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; The Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Caiyun G Li
- Department of Pediatrics and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; The Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nils P Nickel
- Department of Pediatrics and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; The Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mark Kaschwich
- Department of Pediatrics and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; The Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Aiqin Cao
- Department of Pediatrics and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; The Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lingli Wang
- Department of Pediatrics and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; The Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sushma Reddy
- Department of Pediatrics and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Pin-I Chen
- Department of Pediatrics and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; The Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kiichi Nakahira
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Miguel A Alejandre Alcazar
- Department of Pediatrics and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; The Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Rachel K Hopper
- Department of Pediatrics and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; The Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lijuan Ji
- Department of Pediatrics and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Brian J Feldman
- Department of Pediatrics and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Marlene Rabinovitch
- Department of Pediatrics and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; The Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
79
|
Bijli KM, Kleinhenz JM, Murphy TC, Kang BY, Adesina SE, Sutliff RL, Hart CM. Peroxisome proliferator-activated receptor gamma depletion stimulates Nox4 expression and human pulmonary artery smooth muscle cell proliferation. Free Radic Biol Med 2015; 80:111-20. [PMID: 25557278 PMCID: PMC4355175 DOI: 10.1016/j.freeradbiomed.2014.12.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 11/25/2014] [Accepted: 12/18/2014] [Indexed: 10/24/2022]
Abstract
Hypoxia stimulates pulmonary hypertension (PH) in part by increasing the proliferation of pulmonary vascular wall cells. Recent evidence suggests that signaling events involved in hypoxia-induced cell proliferation include sustained nuclear factor-kappaB (NF-κB) activation, increased NADPH oxidase 4 (Nox4) expression, and downregulation of peroxisome proliferator-activated receptor gamma (PPARγ) levels. To further understand the role of reduced PPARγ levels associated with PH pathobiology, siRNA was employed to reduce PPARγ levels in human pulmonary artery smooth muscle cells (HPASMC) in vitro under normoxic conditions. PPARγ protein levels were reduced to levels comparable to those observed under hypoxic conditions. Depletion of PPARγ for 24-72 h activated mitogen-activated protein kinase, ERK 1/2, and NF-κB. Inhibition of ERK 1/2 prevented NF-κB activation caused by PPARγ depletion, indicating that ERK 1/2 lies upstream of NF-κB activation. Depletion of PPARγ for 72 h increased NF-κB-dependent Nox4 expression and H2O2 production. Inhibition of NF-κB or Nox4 attenuated PPARγ depletion-induced HPASMC proliferation. Degradation of PPARγ depletion-induced H2O2 by PEG-catalase prevented HPASMC proliferation and also ERK 1/2 and NF-κB activation and Nox4 expression, indicating that H2O2 participates in feed-forward activation of the above signaling events. Contrary to the effects of PPARγ depletion, HPASMC PPARγ overexpression reduced ERK 1/2 and NF-κB activation, Nox4 expression, and cell proliferation. Taken together these findings provide novel evidence that PPARγ plays a central role in the regulation of the ERK1/2-NF-κB-Nox4-H2O2 signaling axis in HPASMC. These results indicate that reductions in PPARγ caused by pathophysiological stimuli such as prolonged hypoxia exposure are sufficient to promote the proliferation of pulmonary vascular smooth muscle cells observed in PH pathobiology.
Collapse
Affiliation(s)
- Kaiser M Bijli
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Atlanta, GA 30033, USA
| | - Jennifer M Kleinhenz
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Atlanta, GA 30033, USA
| | - Tamara C Murphy
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Atlanta, GA 30033, USA
| | - Bum-Yong Kang
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Atlanta, GA 30033, USA
| | - Sherry E Adesina
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Atlanta, GA 30033, USA
| | - Roy L Sutliff
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Atlanta, GA 30033, USA
| | - C Michael Hart
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Atlanta, GA 30033, USA.
| |
Collapse
|
80
|
Yang XH, Li P, Yin YL, Tu JH, Dai W, Liu LY, Wang SX. Rosiglitazone via PPARγ-dependent suppression of oxidative stress attenuates endothelial dysfunction in rats fed homocysteine thiolactone. J Cell Mol Med 2015; 19:826-35. [PMID: 25656735 PMCID: PMC4395197 DOI: 10.1111/jcmm.12510] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Accepted: 11/17/2014] [Indexed: 02/03/2023] Open
Abstract
To explore whether rosiglitazone (RSG), a selective peroxisome proliferator-activated receptor γ (PPARγ) agonist, exerts beneficial effects on endothelial dysfunction induced by homocysteine thiolactone (HTL) and to investigate the potential mechanisms. Incubation of cultured human umbilical vein endothelial cells with HTL (1 mM) for 24 hrs significantly reduced cell viabilities assayed by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide, as well as enhanced productions of reactive oxygen species, activation of nuclear factor kappa B, and increased intercellular cell adhesion molecule-1 secretion. Pre-treatment of cells with RSG (0.001–0.1 mM), pyrollidine dithiocarbamate (PDTC, 0.1 mM) or apocynin (0.1 mM) for 1 hr reversed these effects induced by HTL. Furthermore, co-incubation with GW9662 (0.01 mM) abolished the protective effects of RSG on HTL-treated cells. In ex vivo experiments, exposure of isolated aortic rings from. rats to HTL (1 mM) for 1 hr dramatically impaired acetylcholine-induced endothelium-dependent relaxation, reduced release of nitric oxide and activity of superoxide dismutase, and increased malondialdehyde content in aortic tissues. Preincubation of aortic rings with RSG (0.1, 0.3, 1 mM), PDTC or apocynin normalized the disorders induced by HTL. In vivo analysis indicated that administration of RSG (20 mg/kg/d) remarkably suppressed oxidative stress and prevented endothelial dysfunction in rats fed HTL (50 mg/kg/d) for 8 weeks. RSG improves endothelial functions in rats fed HTL, which is related to PPARγ-dependent suppression of oxidative stress.
Collapse
Affiliation(s)
- Xu-Hong Yang
- Department of Pharmacology, Pharmaceutical College, Central South University, Changsha, China
| | | | | | | | | | | | | |
Collapse
|
81
|
Guignabert C, Tu L, Girerd B, Ricard N, Huertas A, Montani D, Humbert M. New Molecular Targets of Pulmonary Vascular Remodeling in Pulmonary Arterial Hypertension. Chest 2015; 147:529-537. [DOI: 10.1378/chest.14-0862] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
82
|
Bertero T, Cottrill K, Krauszman A, Lu Y, Annis S, Hale A, Bhat B, Waxman AB, Chau BN, Kuebler WM, Chan SY. The microRNA-130/301 family controls vasoconstriction in pulmonary hypertension. J Biol Chem 2015; 290:2069-85. [PMID: 25505270 PMCID: PMC4303661 DOI: 10.1074/jbc.m114.617845] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 11/26/2014] [Indexed: 12/19/2022] Open
Abstract
Pulmonary hypertension (PH) is a complex disorder, spanning several known vascular cell types. Recently, we identified the microRNA-130/301 (miR-130/301) family as a regulator of multiple pro-proliferative pathways in PH, but the true breadth of influence of the miR-130/301 family across cell types in PH may be even more extensive. Here, we employed targeted network theory to identify additional pathogenic pathways regulated by miR-130/301, including those involving vasomotor tone. Guided by these predictions, we demonstrated, via gain- and loss-of-function experimentation in vitro and in vivo, that miR-130/301-specific control of the peroxisome proliferator-activated receptor γ regulates a panel of vasoactive factors communicating between diseased pulmonary vascular endothelial and smooth muscle cells. Of these, the vasoconstrictive factor endothelin-1 serves as an integral point of communication between the miR-130/301-peroxisome proliferator-activated receptor γ axis in endothelial cells and contractile function in smooth muscle cells. Thus, resulting from an in silico analysis of the architecture of the PH disease gene network coupled with molecular experimentation in vivo, these findings clarify the expanded role of the miR-130/301 family in the global regulation of PH. They further emphasize the importance of molecular cross-talk among the diverse cellular populations involved in PH.
Collapse
Affiliation(s)
- Thomas Bertero
- From the Divisions of Cardiovascular and Network Medicine and
| | | | - Adrienn Krauszman
- the Keenan Research Centre for Biomedical Science of St. Michael's, University of Toronto, Toronto, Ontario M5R 0A3, Canada
| | - Yu Lu
- From the Divisions of Cardiovascular and Network Medicine and
| | - Sofia Annis
- From the Divisions of Cardiovascular and Network Medicine and
| | - Andrew Hale
- From the Divisions of Cardiovascular and Network Medicine and
| | | | - Aaron B Waxman
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - B Nelson Chau
- Regulus Therapeutics, San Diego, California 92121, and
| | - Wolfgang M Kuebler
- the Keenan Research Centre for Biomedical Science of St. Michael's, University of Toronto, Toronto, Ontario M5R 0A3, Canada, the Department of Physiology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Stephen Y Chan
- From the Divisions of Cardiovascular and Network Medicine and
| |
Collapse
|
83
|
Vaillancourt M, Ruffenach G, Meloche J, Bonnet S. Adaptation and remodelling of the pulmonary circulation in pulmonary hypertension. Can J Cardiol 2014; 31:407-15. [PMID: 25630876 DOI: 10.1016/j.cjca.2014.10.023] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Revised: 10/06/2014] [Accepted: 10/20/2014] [Indexed: 01/22/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterized by remodelling of pulmonary arteries caused by a proliferation/apoptosis imbalance within the vascular wall. This pathological phenotype seems to be triggered by different environmental stress and injury events such as increased inflammation, DNA damage, and epigenetic deregulation. It appears that one of the first hit to occur is endothelial cells (ECs) injury and apoptosis, which leads to paracrine signalling to other ECs, pulmonary artery smooth muscle cells (PASMCs), and fibroblasts. These signals promote a phenotypic change of surviving ECs by disturbing different signalling pathways leading to sustained vasoconstriction, proproliferative and antiapoptotic phenotype, deregulated angiogenesis, and formation of plexiform lesions. EC signalling also recruits proinflammatory cells, leading to pulmonary infiltration of lymphocytes, macrophages, and dendritic cells, sustaining the inflammatory environment and autoimmune response. Finally, EC signalling promotes proliferative and antiapoptotic PAH-PASMC phenotypes, which acquire migratory capacities, resulting in increased vascular wall thickness and muscularization of small pulmonary arterioles. Adaptation and remodelling of pulmonary circulation also involves epigenetic components, such as microRNA deregulation, DNA methylation, and histone modification. This review will focus on the different cellular and epigenetic aspects including EC stress response, molecular mechanisms contributing to PAH-PASMC and PAEC proliferation and resistance to apoptosis, as well as epigenetic control involved in adaptation and remodelling of the pulmonary circulation in PAH.
Collapse
Affiliation(s)
- Mylène Vaillancourt
- Pulmonary Hypertension Research Group of The Quebec Heart And Lung Institute Research Centre, Québec City, Québec, Canada
| | - Grégoire Ruffenach
- Pulmonary Hypertension Research Group of The Quebec Heart And Lung Institute Research Centre, Québec City, Québec, Canada
| | - Jolyane Meloche
- Pulmonary Hypertension Research Group of The Quebec Heart And Lung Institute Research Centre, Québec City, Québec, Canada.
| | - Sébastien Bonnet
- Pulmonary Hypertension Research Group of The Quebec Heart And Lung Institute Research Centre, Québec City, Québec, Canada.
| |
Collapse
|
84
|
Ricard N, Tu L, Le Hiress M, Huertas A, Phan C, Thuillet R, Sattler C, Fadel E, Seferian A, Montani D, Dorfmüller P, Humbert M, Guignabert C. Increased Pericyte Coverage Mediated by Endothelial-Derived Fibroblast Growth Factor-2 and Interleukin-6 Is a Source of Smooth Muscle–Like Cells in Pulmonary Hypertension. Circulation 2014; 129:1586-97. [DOI: 10.1161/circulationaha.113.007469] [Citation(s) in RCA: 136] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Background—
Pericytes and their crosstalk with endothelial cells are critical for the development of a functional microvasculature and vascular remodeling. It is also known that pulmonary endothelial dysfunction is intertwined with the initiation and progression of pulmonary arterial hypertension (PAH). We hypothesized that pulmonary endothelial dysfunction, characterized by abnormal fibroblast growth factor-2 and interleukin-6 signaling, leads to abnormal microvascular pericyte coverage causing pulmonary arterial medial thickening.
Methods and Results—
In human lung tissues, numbers of pericytes are substantially increased (up to 2-fold) in distal PAH pulmonary arteries compared with controls. Interestingly, human pulmonary pericytes exhibit, in vitro, an accentuated proliferative and migratory response to conditioned media from human idiopathic PAH endothelial cells compared with conditioned media from control cells. Importantly, by using an anti–fibroblast growth factor-2 neutralizing antibody, we attenuated these proliferative and migratory responses, whereas by using an anti–interleukin-6 neutralizing antibody, we decreased the migratory response without affecting the proliferative response. Furthermore, in our murine retinal angiogenesis model, both fibroblast growth factor-2 and interleukin-6 administration increased pericyte coverage. Finally, using idiopathic PAH human and NG2DsRedBAC mouse lung tissues, we demonstrated that this increased pericyte coverage contributes to pulmonary vascular remodeling as a source of smooth muscle–like cells. Furthermore, we found that transforming growth factor-β, in contrast to fibroblast growth factor-2 and interleukin-6, promotes human pulmonary pericyte differentiation into contractile smooth muscle–like cells.
Conclusions—
To the best of our knowledge, this is the first report of excessive pericyte coverage in distal pulmonary arteries in human PAH. We also show that this phenomenon is directly linked with pulmonary endothelial dysfunction.
Collapse
Affiliation(s)
- Nicolas Ricard
- From the National Institute of Health and Medical Research, Unit 999, LabEx Laboratory of Excellence in Research on Medication and Innovative Therapeutics, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (N.R., L.T., M.L.H., A.H., C.P., R.T., C.S., E.F., A.S., D.M., P.D., M.H., C.G.); University Paris-Sud, School of Medicine, Kremlin-Bicêtre, France (N.R., L.T., M.L.H., A.H., C.P., R.T., C.S., E.F., A.S., D.M., P.D., M.H., C.G.); and Public Hospitals of Paris, Pneumology Service,
| | - Ly Tu
- From the National Institute of Health and Medical Research, Unit 999, LabEx Laboratory of Excellence in Research on Medication and Innovative Therapeutics, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (N.R., L.T., M.L.H., A.H., C.P., R.T., C.S., E.F., A.S., D.M., P.D., M.H., C.G.); University Paris-Sud, School of Medicine, Kremlin-Bicêtre, France (N.R., L.T., M.L.H., A.H., C.P., R.T., C.S., E.F., A.S., D.M., P.D., M.H., C.G.); and Public Hospitals of Paris, Pneumology Service,
| | - Morane Le Hiress
- From the National Institute of Health and Medical Research, Unit 999, LabEx Laboratory of Excellence in Research on Medication and Innovative Therapeutics, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (N.R., L.T., M.L.H., A.H., C.P., R.T., C.S., E.F., A.S., D.M., P.D., M.H., C.G.); University Paris-Sud, School of Medicine, Kremlin-Bicêtre, France (N.R., L.T., M.L.H., A.H., C.P., R.T., C.S., E.F., A.S., D.M., P.D., M.H., C.G.); and Public Hospitals of Paris, Pneumology Service,
| | - Alice Huertas
- From the National Institute of Health and Medical Research, Unit 999, LabEx Laboratory of Excellence in Research on Medication and Innovative Therapeutics, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (N.R., L.T., M.L.H., A.H., C.P., R.T., C.S., E.F., A.S., D.M., P.D., M.H., C.G.); University Paris-Sud, School of Medicine, Kremlin-Bicêtre, France (N.R., L.T., M.L.H., A.H., C.P., R.T., C.S., E.F., A.S., D.M., P.D., M.H., C.G.); and Public Hospitals of Paris, Pneumology Service,
| | - Carole Phan
- From the National Institute of Health and Medical Research, Unit 999, LabEx Laboratory of Excellence in Research on Medication and Innovative Therapeutics, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (N.R., L.T., M.L.H., A.H., C.P., R.T., C.S., E.F., A.S., D.M., P.D., M.H., C.G.); University Paris-Sud, School of Medicine, Kremlin-Bicêtre, France (N.R., L.T., M.L.H., A.H., C.P., R.T., C.S., E.F., A.S., D.M., P.D., M.H., C.G.); and Public Hospitals of Paris, Pneumology Service,
| | - Raphaël Thuillet
- From the National Institute of Health and Medical Research, Unit 999, LabEx Laboratory of Excellence in Research on Medication and Innovative Therapeutics, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (N.R., L.T., M.L.H., A.H., C.P., R.T., C.S., E.F., A.S., D.M., P.D., M.H., C.G.); University Paris-Sud, School of Medicine, Kremlin-Bicêtre, France (N.R., L.T., M.L.H., A.H., C.P., R.T., C.S., E.F., A.S., D.M., P.D., M.H., C.G.); and Public Hospitals of Paris, Pneumology Service,
| | - Caroline Sattler
- From the National Institute of Health and Medical Research, Unit 999, LabEx Laboratory of Excellence in Research on Medication and Innovative Therapeutics, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (N.R., L.T., M.L.H., A.H., C.P., R.T., C.S., E.F., A.S., D.M., P.D., M.H., C.G.); University Paris-Sud, School of Medicine, Kremlin-Bicêtre, France (N.R., L.T., M.L.H., A.H., C.P., R.T., C.S., E.F., A.S., D.M., P.D., M.H., C.G.); and Public Hospitals of Paris, Pneumology Service,
| | - Elie Fadel
- From the National Institute of Health and Medical Research, Unit 999, LabEx Laboratory of Excellence in Research on Medication and Innovative Therapeutics, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (N.R., L.T., M.L.H., A.H., C.P., R.T., C.S., E.F., A.S., D.M., P.D., M.H., C.G.); University Paris-Sud, School of Medicine, Kremlin-Bicêtre, France (N.R., L.T., M.L.H., A.H., C.P., R.T., C.S., E.F., A.S., D.M., P.D., M.H., C.G.); and Public Hospitals of Paris, Pneumology Service,
| | - Andrei Seferian
- From the National Institute of Health and Medical Research, Unit 999, LabEx Laboratory of Excellence in Research on Medication and Innovative Therapeutics, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (N.R., L.T., M.L.H., A.H., C.P., R.T., C.S., E.F., A.S., D.M., P.D., M.H., C.G.); University Paris-Sud, School of Medicine, Kremlin-Bicêtre, France (N.R., L.T., M.L.H., A.H., C.P., R.T., C.S., E.F., A.S., D.M., P.D., M.H., C.G.); and Public Hospitals of Paris, Pneumology Service,
| | - David Montani
- From the National Institute of Health and Medical Research, Unit 999, LabEx Laboratory of Excellence in Research on Medication and Innovative Therapeutics, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (N.R., L.T., M.L.H., A.H., C.P., R.T., C.S., E.F., A.S., D.M., P.D., M.H., C.G.); University Paris-Sud, School of Medicine, Kremlin-Bicêtre, France (N.R., L.T., M.L.H., A.H., C.P., R.T., C.S., E.F., A.S., D.M., P.D., M.H., C.G.); and Public Hospitals of Paris, Pneumology Service,
| | - Peter Dorfmüller
- From the National Institute of Health and Medical Research, Unit 999, LabEx Laboratory of Excellence in Research on Medication and Innovative Therapeutics, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (N.R., L.T., M.L.H., A.H., C.P., R.T., C.S., E.F., A.S., D.M., P.D., M.H., C.G.); University Paris-Sud, School of Medicine, Kremlin-Bicêtre, France (N.R., L.T., M.L.H., A.H., C.P., R.T., C.S., E.F., A.S., D.M., P.D., M.H., C.G.); and Public Hospitals of Paris, Pneumology Service,
| | - Marc Humbert
- From the National Institute of Health and Medical Research, Unit 999, LabEx Laboratory of Excellence in Research on Medication and Innovative Therapeutics, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (N.R., L.T., M.L.H., A.H., C.P., R.T., C.S., E.F., A.S., D.M., P.D., M.H., C.G.); University Paris-Sud, School of Medicine, Kremlin-Bicêtre, France (N.R., L.T., M.L.H., A.H., C.P., R.T., C.S., E.F., A.S., D.M., P.D., M.H., C.G.); and Public Hospitals of Paris, Pneumology Service,
| | - Christophe Guignabert
- From the National Institute of Health and Medical Research, Unit 999, LabEx Laboratory of Excellence in Research on Medication and Innovative Therapeutics, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (N.R., L.T., M.L.H., A.H., C.P., R.T., C.S., E.F., A.S., D.M., P.D., M.H., C.G.); University Paris-Sud, School of Medicine, Kremlin-Bicêtre, France (N.R., L.T., M.L.H., A.H., C.P., R.T., C.S., E.F., A.S., D.M., P.D., M.H., C.G.); and Public Hospitals of Paris, Pneumology Service,
| |
Collapse
|
85
|
Koyama M, Furuhashi M, Ishimura S, Mita T, Fuseya T, Okazaki Y, Yoshida H, Tsuchihashi K, Miura T. Reduction of endoplasmic reticulum stress by 4-phenylbutyric acid prevents the development of hypoxia-induced pulmonary arterial hypertension. Am J Physiol Heart Circ Physiol 2014; 306:H1314-23. [PMID: 24610918 DOI: 10.1152/ajpheart.00869.2013] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Pulmonary arterial hypertension (PAH) is characterized by vasoconstriction and vascular remodeling of the pulmonary artery (PA). Recently, endoplasmic reticulum (ER) stress and inappropriate adaptation through the unfolded protein response (UPR) have been disclosed in various types of diseases. Here we examined whether ER stress is involved in the pathogenesis of PAH. Four weeks of chronic normobaric hypoxia increased right ventricular (RV) systolic pressure by 63% compared with that in normoxic controls and induced RV hypertrophy and medial thickening of the PA in C57BL/6J mice. Treatment with 4-phenylbutyric acid (4-PBA), a chemical chaperone, significantly reduced RV systolic pressure by 30%, attenuated RV hypertrophy and PA muscularization, and increased total running distance in a treadmill test by 70% in hypoxic mice. The beneficial effects of 4-PBA were associated with suppressed expression of inflammatory cytokines and ER stress markers, including Grp78 and Grp94 in the activating transcription factor-6 branch, sXbp1 and Pdi in the inositol-requiring enzyme-1 branch and Atf4 in the PKR-like ER kinase branch, and reduced phosphorylation of c-Jun NH2-terminal kinase and eukaryotic translation initiation factor-2α in the lung. The pattern of changes in ER stress and inflammatory markers by 4-PBA in the lung of the PAH model was reproduced in PA smooth muscle cells by chronic stimulation of platelet-derived growth factor-BB or hypoxia. Furthermore, knockdown of each UPR branch sensor activated other branches and promoted proliferation of PA smooth muscle cells. The findings indicate that activation of all branches of the UPR and accompanying inflammation play a major role in the pathogenesis of PAH, and that chemical chaperones are potentially therapeutic agents for PAH.
Collapse
Affiliation(s)
- Masayuki Koyama
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan; and
| | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Activation of PPAR-γ ameliorates pulmonary arterial hypertension via inducing heme oxygenase-1 and p21WAF1: An in vivo study in rats. Life Sci 2014; 98:39-43. [DOI: 10.1016/j.lfs.2013.12.208] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2013] [Revised: 12/08/2013] [Accepted: 12/26/2013] [Indexed: 11/19/2022]
|
87
|
Sawada H, Saito T, Nickel NP, Alastalo TP, Glotzbach JP, Chan R, Haghighat L, Fuchs G, Januszyk M, Cao A, Lai YJ, Perez VDJ, Kim YM, Wang L, Chen PI, Spiekerkoetter E, Mitani Y, Gurtner GC, Sarnow P, Rabinovitch M. Reduced BMPR2 expression induces GM-CSF translation and macrophage recruitment in humans and mice to exacerbate pulmonary hypertension. ACTA ACUST UNITED AC 2014; 211:263-80. [PMID: 24446489 PMCID: PMC3920564 DOI: 10.1084/jem.20111741] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Idiopathic pulmonary arterial hypertension (PAH [IPAH]) is an insidious and potentially fatal disease linked to a mutation or reduced expression of bone morphogenetic protein receptor 2 (BMPR2). Because intravascular inflammatory cells are recruited in IPAH pathogenesis, we hypothesized that reduced BMPR2 enhances production of the potent chemokine granulocyte macrophage colony-stimulating factor (GM-CSF) in response to an inflammatory perturbation. When human pulmonary artery (PA) endothelial cells deficient in BMPR2 were stimulated with tumor necrosis factor (TNF), a twofold increase in GM-CSF was observed and related to enhanced messenger RNA (mRNA) translation. The mechanism was associated with disruption of stress granule formation. Specifically, loss of BMPR2 induced prolonged phospho-p38 mitogen-activated protein kinase (MAPK) in response to TNF, and this increased GADD34-PP1 phosphatase activity, dephosphorylating eukaryotic translation initiation factor (eIF2α), and derepressing GM-CSF mRNA translation. Lungs from IPAH patients versus unused donor controls revealed heightened PA expression of GM-CSF co-distributing with increased TNF and expanded populations of hematopoietic and endothelial GM-CSF receptor α (GM-CSFRα)-positive cells. Moreover, a 3-wk infusion of GM-CSF in mice increased hypoxia-induced PAH, in association with increased perivascular macrophages and muscularized distal arteries, whereas blockade of GM-CSF repressed these features. Thus, reduced BMPR2 can subvert a stress granule response, heighten GM-CSF mRNA translation, increase inflammatory cell recruitment, and exacerbate PAH.
Collapse
Affiliation(s)
- Hirofumi Sawada
- The Vera Moulton Wall Center for Pulmonary Vascular Disease, 2 Department of Pediatrics, 3 Department of Surgery, 4 Department of Microbiology and Immunology, 5 Department of Medicine, and 6 Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Kang BY, Park KK, Green DE, Bijli KM, Searles CD, Sutliff RL, Hart CM. Hypoxia mediates mutual repression between microRNA-27a and PPARγ in the pulmonary vasculature. PLoS One 2013; 8:e79503. [PMID: 24244514 PMCID: PMC3828382 DOI: 10.1371/journal.pone.0079503] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 09/22/2013] [Indexed: 01/02/2023] Open
Abstract
Pulmonary hypertension (PH) is a serious disorder that causes significant morbidity and mortality. The pathogenesis of PH involves complex derangements in multiple pathways including reductions in peroxisome proliferator-activated receptor gamma (PPARγ). Hypoxia, a common PH stimulus, reduces PPARγ in experimental models. In contrast, activating PPARγ attenuates hypoxia-induced PH and endothelin 1 (ET-1) expression. To further explore mechanisms of hypoxia-induced PH and reductions in PPARγ, we examined the effects of hypoxia on selected microRNA (miRNA or miR) levels that might reduce PPARγ expression leading to increased ET-1 expression and PH. Our results demonstrate that exposure to hypoxia (10% O2) for 3-weeks increased levels of miR-27a and ET-1 in the lungs of C57BL/6 mice and reduced PPARγ levels. Hypoxia-induced increases in miR-27a were attenuated in mice treated with the PPARγ ligand, rosiglitazone (RSG, 10 mg/kg/d) by gavage for the final 10 d of exposure. In parallel studies, human pulmonary artery endothelial cells (HPAECs) were exposed to control (21% O2) or hypoxic (1% O2) conditions for 72 h. Hypoxia increased HPAEC proliferation, miR-27a and ET-1 expression, and reduced PPARγ expression. These alterations were attenuated by treatment with RSG (10 µM) during the last 24 h of hypoxia exposure. Overexpression of miR-27a or PPARγ knockdown increased HPAEC proliferation and ET-1 expression and decreased PPARγ levels, whereas these effects were reversed by miR-27a inhibition. Further, compared to lungs from littermate control mice, miR-27a levels were upregulated in lungs from endothelial-targeted PPARγ knockout (ePPARγ KO) mice. Knockdown of either SP1 or EGR1 was sufficient to significantly attenuate miR-27a expression in HPAECs. Collectively, these studies provide novel evidence that miR-27a and PPARγ mediate mutually repressive actions in hypoxic pulmonary vasculature and that targeting PPARγ may represent a novel therapeutic approach in PH to attenuate proliferative mediators that stimulate proliferation of pulmonary vascular cells.
Collapse
Affiliation(s)
- Bum-Yong Kang
- Departments of Medicine, Atlanta Veterans Affairs Medical Centers and Emory University, Atlanta, Georgia, United States of America
| | - Kathy K. Park
- Departments of Medicine, Atlanta Veterans Affairs Medical Centers and Emory University, Atlanta, Georgia, United States of America
| | - David E. Green
- Departments of Medicine, Atlanta Veterans Affairs Medical Centers and Emory University, Atlanta, Georgia, United States of America
| | - Kaiser M. Bijli
- Departments of Medicine, Atlanta Veterans Affairs Medical Centers and Emory University, Atlanta, Georgia, United States of America
| | - Charles D. Searles
- Departments of Medicine, Atlanta Veterans Affairs Medical Centers and Emory University, Atlanta, Georgia, United States of America
| | - Roy L. Sutliff
- Departments of Medicine, Atlanta Veterans Affairs Medical Centers and Emory University, Atlanta, Georgia, United States of America
| | - C. Michael Hart
- Departments of Medicine, Atlanta Veterans Affairs Medical Centers and Emory University, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
89
|
Tajsic T, Morrell NW. Smooth muscle cell hypertrophy, proliferation, migration and apoptosis in pulmonary hypertension. Compr Physiol 2013; 1:295-317. [PMID: 23737174 DOI: 10.1002/cphy.c100026] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Pulmonary hypertension is a multifactorial disease characterized by sustained elevation of pulmonary vascular resistance (PVR) and pulmonary arterial pressure (PAP). Central to the pathobiology of this disease is the process of vascular remodelling. This process involves structural and functional changes to the normal architecture of the walls of pulmonary arteries (PAs) that lead to increased muscularization of the muscular PAs, muscularization of the peripheral, previously nonmuscular, arteries of the respiratory acinus, formation of neointima, and formation of plexiform lesions. Underlying or contributing to the development of these lesions is hypertrophy, proliferation, migration, and resistance to apoptosis of medial cells and this article is concerned with the cellular and molecular mechanisms of these processes. In the first part of the article we focus on the concept of smooth muscle cell phenotype and the difficulties surrounding the identification and characterization of the cell/cells involved in the remodelling of the vessel media and we review the general mechanisms of cell hypertrophy, proliferation, migration and apoptosis. Then, in the larger part of the article, we review the factors identified thus far to be involved in PH intiation and/or progression and review and discuss their effects on pulmonary artery smooth muscle cells (PASMCs) the predominant cells in the tunica media of PAs.
Collapse
Affiliation(s)
- Tamara Tajsic
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | | |
Collapse
|
90
|
Wang J, Yang K, Xu L, Zhang Y, Lai N, Jiang H, Zhang Y, Zhong N, Ran P, Lu W. Sildenafil inhibits hypoxia-induced transient receptor potential canonical protein expression in pulmonary arterial smooth muscle via cGMP-PKG-PPARγ axis. Am J Respir Cell Mol Biol 2013; 49:231-40. [PMID: 23526219 DOI: 10.1165/rcmb.2012-0185oc] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Transient receptor potential canonical (TRPC) proteins play important roles in chronically hypoxic pulmonary hypertension (CHPH). Previous results indicated that sildenafil inhibited TRPC1 and TRPC6 expression in rat distal pulmonary arteries (PAs). However, the underlying mechanisms remain unknown. We undertook this study to investigate the downstream signaling of sildenafil's regulation on TRPC1 and TRPC6 expression in pulmonary arterial smooth muscle cells (PASMCs). Hypoxia-exposed rats (10% O2 for 21 d) and rat distal PASMCs (4% O2 for 60 h) were taken as models to mimic CHPH. Real-time PCR, Western blotting, and Fura-2-based fluorescent microscopy were performed for mRNA, protein, and Ca(2+) measurements, respectively. The cellular cyclic guanosine monophosphate (cGMP) analogue 8-(4-chlorophenylthio)-guanosine 3',5'-cyclic monophosphate sodium salt (CPT-cGMP) (100 μM) inhibited TRPC1 and TRPC6 expression, store-operated Ca(2+) entry (SOCE), and the proliferation and migration of PASMCs exposed to prolonged hypoxia. The inhibition of CPT-cGMP on TRPC1 and TRPC6 expression in PASMCs was relieved by either the inhibition or knockdown of cGMP-dependent protein kinase (PKG) and peroxisome proliferator-activated receptor γ (PPARγ) expression. Under hypoxic conditions, CPT-cGMP increased PPARγ expression. This increase was abolished by the PKG antagonists Rp8 or KT5823. PPARγ agonist GW1929 significantly decreased TRPC1 and TRPC6 expression in PASMCs. Moreover, hypoxia exposure decreased, whereas sildenafil treatment increased, PKG and PPARγ expression in PASMCs ex vivo, and in rat distal PAs in vivo. The suppressive effects of sildenafil on TRPC1 and TRPC6 in rat distal PAs and on the hemodynamic parameters of CHPH were inhibited by treatment with the PPARγ antagonist T0070907. We conclude that sildenafil inhibits TRPC1 and TRPC6 expression in PASMCs via cGMP-PKG-PPARγ-dependent signaling during CHPH.
Collapse
Affiliation(s)
- Jian Wang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Ardelean DS, Jerkic M, Yin M, Peter M, Ngan B, Kerbel RS, Foster FS, Letarte M. Endoglin and activin receptor-like kinase 1 heterozygous mice have a distinct pulmonary and hepatic angiogenic profile and response to anti-VEGF treatment. Angiogenesis 2013; 17:129-46. [PMID: 24061911 DOI: 10.1007/s10456-013-9383-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 09/09/2013] [Indexed: 02/08/2023]
Abstract
Hereditary hemorrhagic telangiectasia (HHT) is a vascular dysplasia associated with dysregulated angiogenesis and arteriovascular malformations. The disease is caused by mutations in endoglin (ENG; HHT1) or activin receptor-like kinase 1 (ALK1; HHT2) genes, coding for transforming growth factor β (TGF-β) superfamily receptors. Vascular endothelial growth factor (VEGF) has been implicated in HHT and beneficial effects of anti-VEGF treatment were recently reported in HHT patients. To investigate the systemic angiogenic phenotype of Endoglin and Alk1 mutant mice and their response to anti-VEGF therapy, we assessed microvessel density (MVD) in multiple organs after treatment with an antibody to mouse VEGF or vehicle. Lungs were the only organ showing an angiogenic defect, with reduced peripheral MVD and secondary right ventricular hypertrophy (RVH), yet distinctly associated with a fourfold increase in thrombospondin-1 (TSP-1) in Eng (+/-) versus a rise in angiopoietin-2 (Ang-2) in Alk1 (+/-) mice. Anti-VEGF treatment did reduce lung VEGF levels but interestingly, led to an increase in peripheral pulmonary MVD and attenuation of RVH; it also normalized TSP-1 and Ang-2 expression. Hepatic MVD, unaffected in mutant mice, was reduced by anti-VEGF therapy in heterozygous and wild type mice, indicating a liver-specific effect of treatment. Contrast-enhanced micro-ultrasound demonstrated a reduction in hepatic microvascular perfusion after anti-VEGF treatment only in Eng (+/-) mice. Our findings indicate that the mechanisms responsible for the angiogenic imbalance and the response to anti-VEGF therapy differ between Eng and Alk1 heterozygous mice and raise the need for systemic monitoring of anti-angiogenic therapy effects in HHT patients.
Collapse
MESH Headings
- Activin Receptors, Type I/genetics
- Activin Receptors, Type I/metabolism
- Activin Receptors, Type II
- Animals
- Antibodies, Monoclonal/pharmacology
- Endoglin
- Heterozygote
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Liver/blood supply
- Liver/metabolism
- Liver/pathology
- Lung/blood supply
- Lung/metabolism
- Lung/pathology
- Mice
- Mice, Mutant Strains
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Ribonuclease, Pancreatic/genetics
- Ribonuclease, Pancreatic/metabolism
- Telangiectasia, Hereditary Hemorrhagic/drug therapy
- Telangiectasia, Hereditary Hemorrhagic/genetics
- Telangiectasia, Hereditary Hemorrhagic/metabolism
- Telangiectasia, Hereditary Hemorrhagic/pathology
- Vascular Endothelial Growth Factor A/antagonists & inhibitors
- Vascular Endothelial Growth Factor A/genetics
- Vascular Endothelial Growth Factor A/metabolism
Collapse
Affiliation(s)
- Daniela S Ardelean
- Molecular Structure and Function Program, Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
| | | | | | | | | | | | | | | |
Collapse
|
92
|
Morrell NW, Archer SL, Defelice A, Evans S, Fiszman M, Martin T, Saulnier M, Rabinovitch M, Schermuly R, Stewart D, Truebel H, Walker G, Stenmark KR. Anticipated classes of new medications and molecular targets for pulmonary arterial hypertension. Pulm Circ 2013; 3:226-44. [PMID: 23662201 PMCID: PMC3641734 DOI: 10.4103/2045-8932.109940] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) remains a life-limiting condition with a major impact on the ability to lead a normal life. Although existing therapies may improve the outlook in some patients there remains a major unmet need to develop more effective therapies in this condition. There have been significant advances in our understanding of the genetic, cell and molecular basis of PAH over the last few years. This research has identified important new targets that could be explored as potential therapies for PAH. In this review we discuss whether further exploitation of vasoactive agents could bring additional benefits over existing approaches. Approaches to enhance smooth muscle cell apotosis and the potential of receptor tyrosine kinase inhibition are summarised. We evaluate the role of inflammation, epigenetic changes and altered glycolytic metabolism as potential targets for therapy, and whether inherited genetic mutations in PAH have revealed druggable targets. The potential of cell based therapies and gene therapy are also discussed. Potential candidate pathways that could be explored in the context of experimental medicine are identified.
Collapse
|
93
|
Relaxation of human pulmonary arteries by PPARγ agonists. Naunyn Schmiedebergs Arch Pharmacol 2013; 386:445-53. [PMID: 23483194 PMCID: PMC3622741 DOI: 10.1007/s00210-013-0846-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 02/28/2013] [Indexed: 12/14/2022]
Abstract
It has been suggested that activation of nuclear peroxisome proliferator-activated receptors γ (PPARγ) may represent a new strategy for the treatment of pulmonary arterial hypertension. It has been demonstrated that PPARγ activation relaxed the isolated mouse pulmonary artery. The aims of the present study were to examine whether and to which extent the two PPARγ agonists rosiglitazone and pioglitazone relax the isolated human pulmonary artery and to investigate the underlying mechanism(s). Isolated human pulmonary arteries were obtained from patients without clinical evidence of pulmonary hypertension during resection of lung carcinoma. Vasodilatory effects of PPARγ agonists were examined on endothelium-intact or endothelium-denuded vessels preconstricted with the thromboxane prostanoid receptor agonist U-46619. Rosiglitazone and pioglitazone (0.01–100 μM) caused a concentration- and/or time-dependent full relaxation of U-46619-preconstricted vessels. The rosiglitazone-induced relaxation was attenuated by the PPARγ antagonist GW9662 1 μM, endothelium denudation, the nitric oxide synthase inhibitor L-NAME 300 μM, the cyclooxygenase inhibitor indomethacin 10 μM, and the KATP channel blocker glibenclamide 10 μM. The prostacyclin IP receptor antagonist RO1138452 1 μM shifted the concentration–response curve for rosiglitazone to the right. The PPARγ agonists pioglitazone and rosiglitazone relax human pulmonary arteries. The rosiglitazone-induced vasorelaxation is partially endothelium-dependent and involves PPARγ receptors, arachidonic acid degradation products, nitric oxide, and KATP channels. Thus, the relaxant effect of PPARγ agonists in human pulmonary arteries may represent a new therapeutic target in pulmonary arterial hypertension.
Collapse
|
94
|
Karoor V, Oka M, Walchak SJ, Hersh LB, Miller YE, Dempsey EC. Neprilysin regulates pulmonary artery smooth muscle cell phenotype through a platelet-derived growth factor receptor-dependent mechanism. Hypertension 2013; 61:921-30. [PMID: 23381789 DOI: 10.1161/hypertensionaha.111.199588] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Reduced neprilysin (NEP), a cell surface metallopeptidase, which cleaves and inactivates proinflammatory and vasoactive peptides, predisposes the lung vasculature to exaggerated remodeling in response to hypoxia. We hypothesize that loss of NEP in pulmonary artery smooth muscle cells results in increased migration and proliferation. Pulmonary artery smooth muscle cells isolated from NEP(-/-) mice exhibited enhanced migration and proliferation in response to serum and platelet-derived growth factor, which was attenuated by NEP replacement. Inhibition of NEP by overexpression of a peptidase dead mutant or knockdown by small interfering RNA in NEP(+/+) cells increased migration and proliferation. Loss of NEP led to an increase in Src kinase activity and phosphorylation of PTEN, resulting in activation of the platelet-derived growth factor receptor (PDGFR). Knockdown of Src kinase with small interfering RNA or inhibition with PP2, a src kinase inhibitor, decreased PDGFR(Y751) phosphorylation and attenuated migration and proliferation in NEP(-/-) smooth muscle cells. NEP substrates, endothelin 1 or fibroblast growth factor 2, increased activation of Src and PDGFR in NEP(+/+) cells, which was decreased by an endothelin A receptor antagonist, neutralizing antibody to fibroblast growth factor 2 and Src inhibitor. Similar to the observations in pulmonary artery smooth muscle cells, levels of phosphorylated PDGFR, Src, and PTEN were elevated in NEP(-/-) lungs. Endothelin A receptor antagonist also attenuated the enhanced responses in NEP(-/-) pulmonary artery smooth muscle cells and lungs. Taken together our results suggest a novel mechanism for the regulation of PDGFR signaling by NEP substrates involving Src and PTEN. Strategies that increase lung NEP activity/expression or target key downstream effectors, like Src, PTEN, or PDGFR, may be of therapeutic benefit in pulmonary vascular disease.
Collapse
Affiliation(s)
- Vijaya Karoor
- Cardiovascular Pulmonary Research Laboratory, RC-2 Room 8118, University of Colorado Anschutz Medical Campus, 12700 E 19th Ave, RC-2, Aurora, CO 80045, USA.
| | | | | | | | | | | |
Collapse
|
95
|
Abstract
Genetically modified mouse models have unparalleled power to determine the mechanisms behind different processes involved in the molecular and physiologic etiology of various classes of human pulmonary hypertension (PH). Processes known to be involved in PH for which there are extensive mouse models available include the following: (1) Regulation of vascular tone through secreted vasoactive factors; (2) regulation of vascular tone through potassium and calcium channels; (3) regulation of vascular remodeling through alteration in metabolic processes, either through alteration in substrate usage or through circulating factors; (4) spontaneous vascular remodeling either before or after development of elevated pulmonary pressures; and (5) models in which changes in tone and remodeling are primarily driven by inflammation. PH development in mice is of necessity faster and with different physiologic ramifications than found in human disease, and so mice make poor models of natural history of PH. However, transgenic mouse models are a perfect tool for studying the processes involved in pulmonary vascular function and disease, and can effectively be used to test interventions designed against particular molecular pathways and processes involved in disease.
Collapse
Affiliation(s)
- Mita Das
- Department of Internal Medicine, University of Arkansas Medical Sciences, Little Rock, Arkansas, USA
| | | | | | | |
Collapse
|
96
|
Chen WC, Park SH, Hoffman C, Philip C, Robinson L, West J, Grunig G. Right ventricular systolic pressure measurements in combination with harvest of lung and immune tissue samples in mice. J Vis Exp 2013:e50023. [PMID: 23354416 DOI: 10.3791/50023] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The function of the right heart is to pump blood through the lungs, thus linking right heart physiology and pulmonary vascular physiology. Inflammation is a common modifier of heart and lung function, by elaborating cellular infiltration, production of cytokines and growth factors, and by initiating remodeling processes. Compared to the left ventricle, the right ventricle is a low-pressure pump that operates in a relatively narrow zone of pressure changes. Increased pulmonary artery pressures are associated with increased pressure in the lung vascular bed and pulmonary hypertension. Pulmonary hypertension is often associated with inflammatory lung diseases, for example chronic obstructive pulmonary disease, or autoimmune diseases. Because pulmonary hypertension confers a bad prognosis for quality of life and life expectancy, much research is directed towards understanding the mechanisms that might be targets for pharmaceutical intervention. The main challenge for the development of effective management tools for pulmonary hypertension remains the complexity of the simultaneous understanding of molecular and cellular changes in the right heart, the lungs and the immune system. Here, we present a procedural workflow for the rapid and precise measurement of pressure changes in the right heart of mice and the simultaneous harvest of samples from heart, lungs and immune tissues. The method is based on the direct catheterization of the right ventricle via the jugular vein in close-chested mice, first developed in the late 1990s as surrogate measure of pressures in the pulmonary artery. The organized team-approach facilitates a very rapid right heart catheterization technique. This makes it possible to perform the measurements in mice that spontaneously breathe room air. The organization of the work-flow in distinct work-areas reduces time delay and opens the possibility to simultaneously perform physiology experiments and harvest immune, heart and lung tissues. The procedural workflow outlined here can be adapted for a wide variety of laboratory settings and study designs, from small, targeted experiments, to large drug screening assays. The simultaneous acquisition of cardiac physiology data that can be expanded to include echocardiography and harvest of heart, lung and immune tissues reduces the number of animals needed to obtain data that move the scientific knowledge basis forward. The procedural workflow presented here also provides an ideal basis for gaining knowledge of the networks that link immune, lung and heart function. The same principles outlined here can be adapted to study other or additional organs as needed.
Collapse
Affiliation(s)
- Wen-Chi Chen
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, USA
| | | | | | | | | | | | | |
Collapse
|
97
|
Sharma S, Barton J, Rafikov R, Aggarwal S, Kuo HC, Oishi PE, Datar SA, Fineman JR, Black SM. Chronic inhibition of PPAR-γ signaling induces endothelial dysfunction in the juvenile lamb. Pulm Pharmacol Ther 2012; 26:271-80. [PMID: 23257346 DOI: 10.1016/j.pupt.2012.12.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 11/21/2012] [Accepted: 12/07/2012] [Indexed: 01/19/2023]
Abstract
We have recently shown that the development of endothelial dysfunction in lambs with increased pulmonary blood flow (PBF) correlates with a decrease in peroxisome proliferator activated receptor-γ (PPAR-γ) signaling. Thus, in this study we determined if the loss of PPAR-γ signaling is necessary and sufficient to induce endothelial dysfunction by exposing lambs with normal PBF to the PPAR-γ antagonist, GW9662. Two-weeks of exposure to GW9662 significantly decreased both PPAR-γ protein and activity. In addition, although eNOS protein and nitric oxide metabolites (NO(x)) were significantly increased, endothelial dependent pulmonary vasodilation in response to acetylcholine was attenuated, indicative of endothelial dysfunction. To elucidate whether downstream mediators of vasodilation were impaired we examined soluble guanylate cyclase (sGC)-α and β subunit protein, cGMP levels, and phosphodiesterase 5 (PDE5) protein and activity, but we found no significant changes. However, we found that peroxynitrite levels were significantly increased in GW9662-treated lambs and this correlated with a significant increase in protein kinase G-1α (PKG-1α) nitration and a reduction in PKG activity. Peroxynitrite is formed by the interaction of NO with superoxide and we found that there was a significant increase in superoxide generation in GW9662-treated lambs. Further, we identified dysfunctional mitochondria as the primary source of the increased superoxide. Finally, we found that the mitochondrial dysfunction was due to a disruption in carnitine metabolism. We conclude that loss of PPAR-γ signaling is sufficient to induce endothelial dysfunction confirming its important role in maintaining a healthy vasculature.
Collapse
Affiliation(s)
- Shruti Sharma
- Vascular Biology Center, Georgia Health Sciences University, 1459 Laney Walker Blvd, CB3210B, Augusta, GA 30912, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Abstract
Recent clinical and experimental studies are redefining the cellular and molecular bases of pulmonary arterial hypertension (PAH). The genetic abnormalities first identified in association with the idiopathic form of PAH--together with a vast increase in our understanding of cell signaling, cell transformation, and cell-cell interactions; gene expression; microRNA processing; and mitochondrial and ion channel function--have helped explain the abnormal response of vascular cells to injury. Experimental and clinical studies now converge on the intersection and interactions between a genetic predisposition involving the BMPR2 signaling pathway and an impaired metabolic and chronic inflammatory state in the vessel wall. These deranged processes culminate in an exuberant proliferative response that occludes the pulmonary arterial (PA) lumen and obliterates the most distal intraacinar vessels. Here, we describe emerging therapies based on preclinical studies that address these converging pathways.
Collapse
Affiliation(s)
- Marlene Rabinovitch
- Stanford University School of Medicine, Stanford, California 94305-5162, USA.
| |
Collapse
|
99
|
Liu Y, Tian XY, Mao G, Fang X, Fung ML, Shyy JYJ, Huang Y, Wang N. Peroxisome proliferator-activated receptor-γ ameliorates pulmonary arterial hypertension by inhibiting 5-hydroxytryptamine 2B receptor. Hypertension 2012; 60:1471-8. [PMID: 23108648 DOI: 10.1161/hypertensionaha.112.198887] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
An elevated plasma level of 5-hydroxytryptamine (5-HT) or upregulation of 5-HT receptor signaling or both is implicated in vascular contraction and remodeling in pulmonary arterial hypertension (PAH). Recently, peroxisome proliferator-activated receptor-γ (PPARγ) agonists have been shown to ameliorate PAH. However, their effects on the 5-HT-induced contraction of pulmonary arteries remain unknown. Here, we examined the role of PPARγ in inhibiting 5-HT2B receptor (5-HT2BR) to ameliorate PAH. Pulmonary arteries from PAH rats induced by monocrotaline or chronic hypoxia showed an enhanced vasoconstriction in response to BW723C86, a specific agonist for 5-HT2BR. Expression of 5-HT2BR was also increased in pulmonary arteries from the PAH rats, accompanied by vascular remodeling and right ventricular hypertrophy. Treatment with the PPARγ agonist rosiglitazone in vivo reversed the expression and the vasocontractive effect of 5-HT2BR as well as the thickening of pulmonary arteries. In pulmonary artery smooth muscle cells, 5-HT induced the gene expression of 5-HT2BR, which was inhibited by rosiglitazone, pioglitazone, or adenovirus-mediated overexpression of constitutively activated PPARγ. The pharmacological effect of PPARγ was through the suppression of the 5-HT-induced activator protein-1 activity. These results demonstrated the beneficial effect of PPARγ on 5-HT2BR-mediated vasocontraction, providing a new mechanism for the potential use of PPARγ agonists in PAH.
Collapse
Affiliation(s)
- Yahan Liu
- Institute of Cardiovascular Science, Peking University, Beijing 100191, China
| | | | | | | | | | | | | | | |
Collapse
|
100
|
Hameed AG, Arnold ND, Chamberlain J, Pickworth JA, Paiva C, Dawson S, Cross S, Long L, Zhao L, Morrell NW, Crossman DC, Newman CMH, Kiely DG, Francis SE, Lawrie A. Inhibition of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) reverses experimental pulmonary hypertension. ACTA ACUST UNITED AC 2012; 209:1919-35. [PMID: 23071256 PMCID: PMC3478928 DOI: 10.1084/jem.20112716] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Genetic deletion of TRAIL or antibody blockade prevents the development of pulmonary arterial hypertension and can reverse vascular remodeling in established disease. Pulmonary arterial hypertension (PAH) is a life-threatening disease characterized by the progressive narrowing and occlusion of small pulmonary arteries. Current therapies fail to fully reverse this vascular remodeling. Identifying key pathways in disease pathogenesis is therefore required for the development of new-targeted therapeutics. We have previously reported tumor necrosis factor–related apoptosis-inducing ligand (TRAIL) immunoreactivity within pulmonary vascular lesions from patients with idiopathic PAH and animal models. Because TRAIL can induce both endothelial cell apoptosis and smooth muscle cell proliferation in the systemic circulation, we hypothesized that TRAIL is an important mediator in the pathogenesis of PAH. We demonstrate for the first time that TRAIL is a potent stimulus for pulmonary vascular remodeling in human cells and rodent models. Furthermore, antibody blockade or genetic deletion of TRAIL prevents the development of PAH in three independent rodent models. Finally, anti-TRAIL antibody treatment of rodents with established PAH reverses pulmonary vascular remodeling by reducing proliferation and inducing apoptosis, improves hemodynamic indices, and significantly increases survival. These preclinical investigations are the first to demonstrate the importance of TRAIL in PAH pathogenesis and highlight its potential as a novel therapeutic target to direct future translational therapies.
Collapse
Affiliation(s)
- Abdul G Hameed
- Department of Cardiovascular Science, 2 Department of Neuroscience, University of Sheffield, S10 2RX Sheffield, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|