1
|
Hong J, Medzikovic L, Sun W, Wong B, Ruffenach G, Rhodes CJ, Brownstein A, Liang LL, Aryan L, Li M, Vadgama A, Kurt Z, Schwantes-An TH, Mickler EA, Gräf S, Eyries M, Lutz KA, Pauciulo MW, Trembath RC, Perros F, Montani D, Morrell NW, Soubrier F, Wilkins MR, Nichols WC, Aldred MA, Desai AA, Trégouët DA, Umar S, Saggar R, Channick R, Tuder RM, Geraci MW, Stearman RS, Yang X, Eghbali M. Integrative Multiomics in the Lung Reveals a Protective Role of Asporin in Pulmonary Arterial Hypertension. Circulation 2024; 150:1268-1287. [PMID: 39167456 PMCID: PMC11473243 DOI: 10.1161/circulationaha.124.069864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 07/19/2024] [Indexed: 08/23/2024]
Abstract
BACKGROUND Integrative multiomics can elucidate pulmonary arterial hypertension (PAH) pathobiology, but procuring human PAH lung samples is rare. METHODS We leveraged transcriptomic profiling and deep phenotyping of the largest multicenter PAH lung biobank to date (96 disease and 52 control) by integration with clinicopathologic data, genome-wide association studies, Bayesian regulatory networks, single-cell transcriptomics, and pharmacotranscriptomics. RESULTS We identified 2 potentially protective gene network modules associated with vascular cells, and we validated ASPN, coding for asporin, as a key hub gene that is upregulated as a compensatory response to counteract PAH. We found that asporin is upregulated in lungs and plasma of multiple independent PAH cohorts and correlates with reduced PAH severity. We show that asporin inhibits proliferation and transforming growth factor-β/phosphorylated SMAD2/3 signaling in pulmonary artery smooth muscle cells from PAH lungs. We demonstrate in Sugen-hypoxia rats that ASPN knockdown exacerbated PAH and recombinant asporin attenuated PAH. CONCLUSIONS Our integrative systems biology approach to dissect the PAH lung transcriptome uncovered asporin as a novel protective target with therapeutic potential in PAH.
Collapse
Affiliation(s)
- Jason Hong
- Division of Pulmonary and Critical Care Medicine (J.H., B.W., A.B., L.L.L., A.V., R.S., R.C.), University of California, Los Angeles
| | - Lejla Medzikovic
- Departments of Anesthesiology & Perioperative Medicine (L.M., W.S., G.R., L.A., M.L., S.U., M. Eghbali), University of California, Los Angeles
| | - Wasila Sun
- Departments of Anesthesiology & Perioperative Medicine (L.M., W.S., G.R., L.A., M.L., S.U., M. Eghbali), University of California, Los Angeles
| | - Brenda Wong
- Division of Pulmonary and Critical Care Medicine (J.H., B.W., A.B., L.L.L., A.V., R.S., R.C.), University of California, Los Angeles
| | - Grégoire Ruffenach
- Departments of Anesthesiology & Perioperative Medicine (L.M., W.S., G.R., L.A., M.L., S.U., M. Eghbali), University of California, Los Angeles
| | | | - Adam Brownstein
- Division of Pulmonary and Critical Care Medicine (J.H., B.W., A.B., L.L.L., A.V., R.S., R.C.), University of California, Los Angeles
| | - Lloyd L Liang
- Division of Pulmonary and Critical Care Medicine (J.H., B.W., A.B., L.L.L., A.V., R.S., R.C.), University of California, Los Angeles
| | - Laila Aryan
- Departments of Anesthesiology & Perioperative Medicine (L.M., W.S., G.R., L.A., M.L., S.U., M. Eghbali), University of California, Los Angeles
| | - Min Li
- Departments of Anesthesiology & Perioperative Medicine (L.M., W.S., G.R., L.A., M.L., S.U., M. Eghbali), University of California, Los Angeles
| | - Arjun Vadgama
- Division of Pulmonary and Critical Care Medicine (J.H., B.W., A.B., L.L.L., A.V., R.S., R.C.), University of California, Los Angeles
| | - Zeyneb Kurt
- Northumbria University, Newcastle Upon Tyne, UK (Z.K.)
| | - Tae-Hwi Schwantes-An
- Department of Medicine, Indiana University, Indianapolis (T.-H.S.-A., E.A.M., M.A.A., A.A.D., R.S.S.)
| | - Elizabeth A Mickler
- Department of Medicine, Indiana University, Indianapolis (T.-H.S.-A., E.A.M., M.A.A., A.A.D., R.S.S.)
| | - Stefan Gräf
- Department of Medicine, Victor Phillip Dahdaleh Heart & Lung Research Institute, University of Cambridge, UK (S.G., N.W.M.)
| | - Mélanie Eyries
- Hôpital Pitié-Salpêtrière, AP-HP, Département de Génétique, Paris, France (M. Eyries)
| | - Katie A Lutz
- Department of Pediatrics, Division of Human Genetics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, OH (K.A.L., M.W.P., W.C.N.)
| | - Michael W Pauciulo
- Department of Pediatrics, Division of Human Genetics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, OH (K.A.L., M.W.P., W.C.N.)
| | - Richard C Trembath
- Department of Medical & Molecular Genetics, Faculty of Life Sciences & Medicine, King's College London, UK (R.C.T.)
| | - Frédéric Perros
- CarMeN Laboratory, INSERM U1060, INRAE U1397, Université Claude Bernard Lyon 1, Pierre-Bénite, France (F.P.)
| | - David Montani
- AP-HP, Service de Pneumologie, Hôpital Bicêtre, Le Kremlin Bicêtre, France (D.M.)
- Université Paris-Saclay, Le Kremlin Bicêtre, France (D.M.)
- UMR_S 999, Université Paris-Saclay, INSERM, Groupe Hospitalier Marie-Lannelongue-Saint Joseph, Le Plessis-Robinson, France (D.M.)
| | - Nicholas W Morrell
- Department of Medicine, Victor Phillip Dahdaleh Heart & Lung Research Institute, University of Cambridge, UK (S.G., N.W.M.)
| | | | - Martin R Wilkins
- National Heart and Lung Institute, Imperial College London, UK (C.J.R., M.R.W.)
| | - William C Nichols
- Department of Pediatrics, Division of Human Genetics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, OH (K.A.L., M.W.P., W.C.N.)
| | - Micheala A Aldred
- Department of Medicine, Indiana University, Indianapolis (T.-H.S.-A., E.A.M., M.A.A., A.A.D., R.S.S.)
| | - Ankit A Desai
- Department of Medicine, Indiana University, Indianapolis (T.-H.S.-A., E.A.M., M.A.A., A.A.D., R.S.S.)
| | | | - Soban Umar
- Departments of Anesthesiology & Perioperative Medicine (L.M., W.S., G.R., L.A., M.L., S.U., M. Eghbali), University of California, Los Angeles
| | - Rajan Saggar
- Division of Pulmonary and Critical Care Medicine (J.H., B.W., A.B., L.L.L., A.V., R.S., R.C.), University of California, Los Angeles
| | - Richard Channick
- Division of Pulmonary and Critical Care Medicine (J.H., B.W., A.B., L.L.L., A.V., R.S., R.C.), University of California, Los Angeles
| | - Rubin M Tuder
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora (R.M.T.)
| | - Mark W Geraci
- Department of Medicine, University of Pittsburgh, PA (M.W.G.)
| | - Robert S Stearman
- Department of Medicine, Indiana University, Indianapolis (T.-H.S.-A., E.A.M., M.A.A., A.A.D., R.S.S.)
| | - Xia Yang
- Integrative Biology and Physiology (X.Y.), University of California, Los Angeles
| | - Mansoureh Eghbali
- Departments of Anesthesiology & Perioperative Medicine (L.M., W.S., G.R., L.A., M.L., S.U., M. Eghbali), University of California, Los Angeles
| |
Collapse
|
2
|
Gaikwad AV, Eapen MS, Dey S, Bhattarai P, Shahzad AM, Chia C, Jaffar J, Westall G, Sutherland D, Singhera GK, Hackett TL, Lu W, Sohal SS. TGF-β1, pSmad-2/3, Smad-7, and β-Catenin Are Augmented in the Pulmonary Arteries from Patients with Idiopathic Pulmonary Fibrosis (IPF): Role in Driving Endothelial-to-Mesenchymal Transition (EndMT). J Clin Med 2024; 13:1160. [PMID: 38398472 PMCID: PMC10888973 DOI: 10.3390/jcm13041160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 02/15/2024] [Accepted: 02/17/2024] [Indexed: 02/25/2024] Open
Abstract
Background: We have previously reported that endothelial-to-mesenchymal transition (EndMT) is an active process in patients with idiopathic pulmonary fibrosis (IPF) contributing to arterial remodelling. Here, we aim to quantify drivers of EndMT in IPF patients compared to normal controls (NCs). Methods: Lung resections from thirteen IPF patients and eleven NCs were immunohistochemically stained for EndMT drivers, including TGF-β1, pSmad-2/3, Smad-7, and β-catenin. Intima, media, and adventitia were analysed for expression of each EndMT driver in pulmonary arteries. Computer- and microscope-assisted Image ProPlus7.0 image analysis software was used for quantifications. Results: Significant TGF-β1, pSmad-2/3, Smad-7, and β-catenin expression was apparent across all arterial sizes in IPF (p < 0.05). Intimal TGF-β1, pSmad-2/3, Smad-7, and β-catenin were augmented in the arterial range of 100-1000 μm (p < 0.001) compared to NC. Intimal TGF-β1 and β-catenin percentage expression showed a strong correlation with the percentage expression of intimal vimentin (r' = 0.54, p = 0.05 and r' = 0.61, p = 0.02, respectively) and intimal N-cadherin (r' = 0.62, p = 0.03 and r' = 0.70, p = 0.001, respectively). Intimal TGF-β1 and β-catenin expression were significantly correlated with increased intimal thickness as well (r' = 0.52, p = 0.04; r' = 0.052, p = 0.04, respectively). Moreover, intimal TGF-β1 expression was also significantly associated with increased intimal elastin deposition (r' = 0.79, p = 0.002). Furthermore, total TGF-β1 expression significantly impacted the percentage of DLCO (r' = -0.61, p = 0.03). Conclusions: This is the first study to illustrate the involvement of active TGF-β/Smad-2/3-dependent and β-catenin-dependent Wnt signalling pathways in driving EndMT and resultant pulmonary arterial remodelling in patients with IPF. EndMT is a potential therapeutic target for vascular remodelling and fibrosis in general in patients with IPF.
Collapse
Affiliation(s)
- Archana Vijay Gaikwad
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, TAS 7248, Australia
- National Health and Medical Research Council (NHMRC) Centre of Research Excellence (CRE) in Pulmonary Fibrosis, Respiratory Medicine and Sleep Unit, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
| | - Mathew Suji Eapen
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, TAS 7248, Australia
- National Health and Medical Research Council (NHMRC) Centre of Research Excellence (CRE) in Pulmonary Fibrosis, Respiratory Medicine and Sleep Unit, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
| | - Surajit Dey
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, TAS 7248, Australia
| | - Prem Bhattarai
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, TAS 7248, Australia
| | - Affan Mahmood Shahzad
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, TAS 7248, Australia
| | - Collin Chia
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, TAS 7248, Australia
- Launceston Respiratory and Sleep Centre, Launceston, TAS 7250, Australia
- Department of Respiratory Medicine, Launceston General Hospital, Launceston, TAS 7250, Australia
| | - Jade Jaffar
- Department of Allergy, Immunology and Respiratory Medicine, The Alfred Hospital, Melbourne, VIC 3004, Australia
- Department of Immunology and Pathology, Monash University, Melbourne, VIC 3004, Australia
| | - Glen Westall
- Department of Allergy, Immunology and Respiratory Medicine, The Alfred Hospital, Melbourne, VIC 3004, Australia
- Department of Immunology and Pathology, Monash University, Melbourne, VIC 3004, Australia
| | - Darren Sutherland
- Department of Anaesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, BC V6Z 1Y6, Canada
| | - Gurpreet Kaur Singhera
- Department of Anaesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, BC V6Z 1Y6, Canada
| | - Tillie-Louise Hackett
- Department of Anaesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, BC V6Z 1Y6, Canada
| | - Wenying Lu
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, TAS 7248, Australia
- National Health and Medical Research Council (NHMRC) Centre of Research Excellence (CRE) in Pulmonary Fibrosis, Respiratory Medicine and Sleep Unit, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
- Launceston Respiratory and Sleep Centre, Launceston, TAS 7250, Australia
| | - Sukhwinder Singh Sohal
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, TAS 7248, Australia
- National Health and Medical Research Council (NHMRC) Centre of Research Excellence (CRE) in Pulmonary Fibrosis, Respiratory Medicine and Sleep Unit, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
- Launceston Respiratory and Sleep Centre, Launceston, TAS 7250, Australia
| |
Collapse
|
3
|
Goncharova EA, Kudryashova TV, Pullamsetti SS. Too hot? Too cold? Wnt signalling in pulmonary arterial hypertension: can we treat it "just right"? Eur Respir J 2023; 61:2300504. [PMID: 37290809 DOI: 10.1183/13993003.00504-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 03/25/2023] [Indexed: 06/10/2023]
Affiliation(s)
- Elena A Goncharova
- Lung Center, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, Davis School of Medicine, Davis, CA, USA
| | - Tatiana V Kudryashova
- Lung Center, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, Davis School of Medicine, Davis, CA, USA
| | - Soni Savai Pullamsetti
- Max Planck Institute for Heart and Lung Research, Department of Lung Development and Remodeling, German Center for Lung Research (DZL), Bad Nauheim, Germany
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), DZL, Justus Liebig University, member of the German Center for Lung Research (DZL), Giessen, Germany
| |
Collapse
|
4
|
Devendran A, Kar S, Bailey R, Trivieri MG. The Role of Bone Morphogenetic Protein Receptor Type 2 ( BMPR2) and the Prospects of Utilizing Induced Pluripotent Stem Cells (iPSCs) in Pulmonary Arterial Hypertension Disease Modeling. Cells 2022; 11:3823. [PMID: 36497082 PMCID: PMC9741276 DOI: 10.3390/cells11233823] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 11/25/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease characterized by increased pulmonary vascular resistance (PVR), causing right ventricular hypertrophy and ultimately death from right heart failure. Heterozygous mutations in the bone morphogenetic protein receptor type 2 (BMPR2) are linked to approximately 80% of hereditary, and 20% of idiopathic PAH cases, respectively. While patients carrying a BMPR2 gene mutation are more prone to develop PAH than non-carriers, only 20% will develop the disease, whereas the majority will remain asymptomatic. PAH is characterized by extreme vascular remodeling that causes pulmonary arterial endothelial cell (PAEC) dysfunction, impaired apoptosis, and uncontrolled proliferation of the pulmonary arterial smooth muscle cells (PASMCs). To date, progress in understanding the pathophysiology of PAH has been hampered by limited access to human tissue samples and inadequacy of animal models to accurately mimic the pathogenesis of human disease. Along with the advent of induced pluripotent stem cell (iPSC) technology, there has been an increasing interest in using this tool to develop patient-specific cellular models that precisely replicate the pathogenesis of PAH. In this review, we summarize the currently available approaches in iPSC-based PAH disease modeling and explore how this technology could be harnessed for drug discovery and to widen our understanding of the pathophysiology of PAH.
Collapse
Affiliation(s)
- Anichavezhi Devendran
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sumanta Kar
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Rasheed Bailey
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Maria Giovanna Trivieri
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Medicine, Cardiology Unit, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
5
|
Azhdari MH, Goodarzi N, Doroudian M, MacLoughlin R. Molecular Insight into the Therapeutic Effects of Stem Cell-Derived Exosomes in Respiratory Diseases and the Potential for Pulmonary Delivery. Int J Mol Sci 2022; 23:ijms23116273. [PMID: 35682948 PMCID: PMC9181737 DOI: 10.3390/ijms23116273] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 05/31/2022] [Accepted: 06/01/2022] [Indexed: 02/07/2023] Open
Abstract
Respiratory diseases are the cause of millions of deaths annually around the world. Despite the recent growth of our understanding of underlying mechanisms contributing to the pathogenesis of lung diseases, most therapeutic approaches are still limited to symptomatic treatments and therapies that only delay disease progression. Several clinical and preclinical studies have suggested stem cell (SC) therapy as a promising approach for treating various lung diseases. However, challenges such as the potential tumorigenicity, the low survival rate of the SCs in the recipient body, and difficulties in cell culturing and storage have limited the applicability of SC therapy. SC-derived extracellular vesicles (SC-EVs), particularly SC-derived exosomes (SC-Exos), exhibit most therapeutic properties of stem cells without their potential drawbacks. Similar to SCs, SC-Exos exhibit immunomodulatory, anti-inflammatory, and antifibrotic properties with the potential to be employed in the treatment of various inflammatory and chronic respiratory diseases. Furthermore, recent studies have demonstrated that the microRNA (miRNA) content of SC-Exos may play a crucial role in the therapeutic potential of these exosomes. Several studies have investigated the administration of SC-Exos via the pulmonary route, and techniques for SCs and SC-Exos delivery to the lungs by intratracheal instillation or inhalation have been developed. Here, we review the literature discussing the therapeutic effects of SC-Exos against respiratory diseases and advances in the pulmonary route of delivery of these exosomes to the damaged tissues.
Collapse
Affiliation(s)
- Mohammad H. Azhdari
- Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran 15719-14911, Iran; (M.H.A.); (N.G.)
| | - Nima Goodarzi
- Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran 15719-14911, Iran; (M.H.A.); (N.G.)
| | - Mohammad Doroudian
- Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran 15719-14911, Iran; (M.H.A.); (N.G.)
- Correspondence: author: (M.D.); (R.M.)
| | - Ronan MacLoughlin
- Research and Development, Science and Emerging Technologies, Aerogen Limited, IDA Business Park, H91 HE94 Galway, Ireland
- School of Pharmacy, Royal College of Surgeons, D02 YN77 Dublin, Ireland
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, D02 PN40 Dublin, Ireland
- Correspondence: author: (M.D.); (R.M.)
| |
Collapse
|
6
|
Xu J, Strasburg GM, Reed KM, Velleman SG. Temperature and Growth Selection Effects on Proliferation, Differentiation, and Adipogenic Potential of Turkey Myogenic Satellite Cells Through Frizzled-7-Mediated Wnt Planar Cell Polarity Pathway. Front Physiol 2022; 13:892887. [PMID: 35677087 PMCID: PMC9167958 DOI: 10.3389/fphys.2022.892887] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/19/2022] [Indexed: 12/30/2022] Open
Abstract
Satellite cells (SCs) are a heterogeneous population of multipotential stem cells. During the first week after hatch, satellite cell function and fate are sensitive to temperature. Wingless-type mouse mammary tumor virus integration site family/planar cell polarity (Wnt/PCP) signaling pathway is significantly affected by thermal stress in turkey pectoralis major (p. major) muscle SCs. This pathway regulates the activity of SCs through a frizzled-7 (Fzd7) cell surface receptor and two intracellular effectors, rho-associated protein kinase (ROCK) and c-Jun. The objective of the present study was to determine the effects of thermal stress, growth selection, and the Fzd7-mediated Wnt/PCP pathway on proliferation, myogenic differentiation, lipid accumulation, and expression of myogenic and adipogenic regulatory genes. These effects were evaluated in SCs isolated from the p. major muscle of 1-week faster-growing modern commercial (NC) line of turkeys as compared to SCs of a slower-growing historic Randombred Control Line 2 (RBC2) turkey line. Heat stress (43°C) increased phosphorylation of both ROCK and c-Jun with greater increases observed in the RBC2 line. Cold stress (33°C) had an inhibitory effect on both ROCK and c-Jun phosphorylation with the NC line showing greater reductions. Knockdown of the expression of Fzd7 decreased proliferation, differentiation, and expression of myogenic regulatory genes: myoblast determination factor-1 and myogenin in both lines. Both lipid accumulation and expression of adipogenic regulatory genes: peroxisome proliferator-activated receptor-γ, CCAAT/enhancer-binding protein-β, and neuropeptide-Y were suppressed with the Fzd7 knockdown. The RBC2 line was more dependent on the Fzd7-mediated Wnt/PCP pathway for proliferation, differentiation, and lipid accumulation compared to the NC line. Thus, thermal stress may affect poultry breast muscle growth potential and protein to fat ratio by altering function and fate of SCs through the Fzd7-mediated Wnt/PCP pathway in a growth-dependent manner.
Collapse
Affiliation(s)
- Jiahui Xu
- Department of Animal Sciences, The Ohio State University, Wooster, OH, United States
| | - Gale M. Strasburg
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States
| | - Kent M. Reed
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Sandra G. Velleman
- Department of Animal Sciences, The Ohio State University, Wooster, OH, United States
| |
Collapse
|
7
|
Dierick F, Solinc J, Bignard J, Soubrier F, Nadaud S. Progenitor/Stem Cells in Vascular Remodeling during Pulmonary Arterial Hypertension. Cells 2021; 10:cells10061338. [PMID: 34071347 PMCID: PMC8226806 DOI: 10.3390/cells10061338] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/12/2021] [Accepted: 05/21/2021] [Indexed: 12/18/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterized by an important occlusive vascular remodeling with the production of new endothelial cells, smooth muscle cells, myofibroblasts, and fibroblasts. Identifying the cellular processes leading to vascular proliferation and dysfunction is a major goal in order to decipher the mechanisms leading to PAH development. In addition to in situ proliferation of vascular cells, studies from the past 20 years have unveiled the role of circulating and resident vascular in pulmonary vascular remodeling. This review aims at summarizing the current knowledge on the different progenitor and stem cells that have been shown to participate in pulmonary vascular lesions and on the pathways regulating their recruitment during PAH. Finally, this review also addresses the therapeutic potential of circulating endothelial progenitor cells and mesenchymal stem cells.
Collapse
Affiliation(s)
- France Dierick
- Lady Davis Institute for Medical Research, McGill University, Montréal, QC H3T 1E2, Canada;
| | - Julien Solinc
- UMR_S 1166, Faculté de Médecine Pitié-Salpêtrière, INSERM, Sorbonne Université, 75013 Paris, France; (J.S.); (J.B.); (F.S.)
| | - Juliette Bignard
- UMR_S 1166, Faculté de Médecine Pitié-Salpêtrière, INSERM, Sorbonne Université, 75013 Paris, France; (J.S.); (J.B.); (F.S.)
| | - Florent Soubrier
- UMR_S 1166, Faculté de Médecine Pitié-Salpêtrière, INSERM, Sorbonne Université, 75013 Paris, France; (J.S.); (J.B.); (F.S.)
| | - Sophie Nadaud
- UMR_S 1166, Faculté de Médecine Pitié-Salpêtrière, INSERM, Sorbonne Université, 75013 Paris, France; (J.S.); (J.B.); (F.S.)
- Correspondence:
| |
Collapse
|
8
|
Ueland T, Abraityte A, Norum H, Varathalingam S, Gullestad L, Aukrust P, Andreassen AK. Circulating regulators of the wingless pathway in precapillary pulmonary hypertension. Respirology 2021; 26:574-581. [PMID: 33830565 DOI: 10.1111/resp.14048] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 02/18/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND OBJECTIVE Dysregulated Wnt signalling has been implicated in pulmonary hypertension (PH). We hypothesized that plasma levels of secreted Wnt proteins would be increased in patients with precapillary PH, correlate with indices of vascular resistance and cardiac function and give information on long-term prognosis. METHODS We measured the Wnt ligand Wnt5a and secreted Wnt antagonists Dickkopf (DKK) DKK1, DKK3, secreted frizzled-related protein 3 (sFRP3), Wnt inhibitory factor-1 (WIF1) and sclerostin (SOST) in 106 patients with precapillary PH and 40 healthy controls. A second sample was obtained after a median of 4 months (n = 52). During a median of 90 months follow-up, 67 patients died. RESULTS Our main findings were (i) Precapillary PH is characterized by enhanced systemic Wnt activity as reflected by elevated plasma levels of Wnt5a and secreted antagonists irrespective of diagnostic subgroups. (ii) WIF1 and in particular Wnt5a correlated with pulmonary vascular resistance and cardiac dysfunction. (iii) High levels of Wnt5a, sFRP3, DKK3 and WIF1 were associated with poor prognosis in age- and sex-adjusted analysis (hazard ratios per log/SD change ~1.4) and for DKK3 after further adjustment with right arterial pressure, pulmonary oxygen saturation, cardiac index, N-terminal pro B-type natriuretic peptide and peak oxygen uptake (VO2 ). Finally, an elevation of Wnt5a and DKK3 during follow-up was independently associated with poor prognosis. CONCLUSION Our data indicate that Wnt signalling pathways could be implicated in the pathogenesis of precapillary PH, and that some of the Wnt-related molecules (i.e., Wnt5a and DKK3) should be further investigated in these patients.
Collapse
Affiliation(s)
- Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Faculty of Medicine, University of Oslo, Oslo, Norway.,K. G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, Tromsø, Norway
| | - Aurelija Abraityte
- Faculty of Medicine, University of Oslo, Oslo, Norway.,Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Hilde Norum
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Department of Research and Development, Division of Emergencies and Critical Care, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Sharanga Varathalingam
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Lars Gullestad
- Faculty of Medicine, University of Oslo, Oslo, Norway.,Center for Heart Failure Research, University of Oslo, Oslo, Norway.,Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway.,K. G. Jebsen Cardiac Research Center, University of Oslo, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Faculty of Medicine, University of Oslo, Oslo, Norway.,K. G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, Tromsø, Norway.,Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway.,K. G. Jebsen Inflammation Research Center, University of Oslo, Oslo, Norway
| | - Arne K Andreassen
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| |
Collapse
|
9
|
Hu Y, Ciminieri C, Hu Q, Lehmann M, Königshoff M, Gosens R. WNT Signalling in Lung Physiology and Pathology. Handb Exp Pharmacol 2021; 269:305-336. [PMID: 34463851 DOI: 10.1007/164_2021_521] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The main physiological function of the lung is gas exchange, mediated at the interface between the alveoli and the pulmonary microcapillary network and facilitated by conducting airway structures that regulate the transport of these gases from and to the alveoli. Exposure to microbial and environmental factors such as allergens, viruses, air pollution, and smoke contributes to the development of chronic lung diseases such as asthma, chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis (IPF), and lung cancer. Respiratory diseases as a cluster are the commonest cause of chronic disease and of hospitalization in children and are among the three most common causes of morbidity and mortality in the adult population worldwide. Many of these chronic respiratory diseases are associated with inflammation and structural remodelling of the airways and/or alveolar tissues. They can often only be treated symptomatically with no disease-modifying therapies that normalize the pathological tissue destruction driven by inflammation and remodelling. In search for novel therapeutic strategies for these diseases, several lines of evidence revealed the WNT pathway as an emerging target for regenerative strategies in the lung. WNT proteins, their receptors, and signalling effectors have central regulatory roles under (patho)physiological conditions underpinning lung function and (chronic) lung diseases and we summarize these roles and discuss how pharmacological targeting of the WNT pathway may be utilized for the treatment of chronic lung diseases.
Collapse
Affiliation(s)
- Yan Hu
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado, Aurora, CO, USA
| | - Chiara Ciminieri
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado, Aurora, CO, USA.,Department of Molecular Pharmacology, Groningen Research Institute for Asthma and COPD, University of Groningen, Groningen, The Netherlands
| | - Qianjiang Hu
- Lung Repair and Regeneration Unit, Helmholtz-Zentrum Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, Munich, Germany
| | - Mareike Lehmann
- Lung Repair and Regeneration Unit, Helmholtz-Zentrum Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, Munich, Germany
| | - Melanie Königshoff
- Lung Repair and Regeneration Unit, Helmholtz-Zentrum Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, Munich, Germany. .,Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Reinoud Gosens
- Department of Molecular Pharmacology, Groningen Research Institute for Asthma and COPD, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
10
|
Koçak A, Harmancı D, Güner Akdoğan G, Birlik M. Relationship of Wnt pathway activity and organ involvement in scleroderma types. Int J Rheum Dis 2020; 23:1558-1567. [PMID: 32996251 DOI: 10.1111/1756-185x.13973] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- Ayşe Koçak
- Kutahya Health Sciences University Kutahya Turkey
| | | | | | | |
Collapse
|
11
|
Mutgan AC, Jandl K, Kwapiszewska G. Endothelial Basement Membrane Components and Their Products, Matrikines: Active Drivers of Pulmonary Hypertension? Cells 2020; 9:cells9092029. [PMID: 32899187 PMCID: PMC7563239 DOI: 10.3390/cells9092029] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/27/2020] [Accepted: 08/29/2020] [Indexed: 12/19/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a vascular disease that is characterized by elevated pulmonary arterial pressure (PAP) due to progressive vascular remodeling. Extracellular matrix (ECM) deposition in pulmonary arteries (PA) is one of the key features of vascular remodeling. Emerging evidence indicates that the basement membrane (BM), a specialized cluster of ECM proteins underlying the endothelium, may be actively involved in the progression of vascular remodeling. The BM and its steady turnover are pivotal for maintaining appropriate vascular functions. However, the pathologically elevated turnover of BM components leads to an increased release of biologically active short fragments, which are called matrikines. Both BM components and their matrikines can interfere with pivotal biological processes, such as survival, proliferation, adhesion, and migration and thus may actively contribute to endothelial dysfunction. Therefore, in this review, we summarize the emerging role of the BM and its matrikines on the vascular endothelium and further discuss its implications on lung vascular remodeling in pulmonary hypertension.
Collapse
Affiliation(s)
- Ayse Ceren Mutgan
- Otto Loewi Research Center, Division of Physiology, Medical University of Graz, 8010 Graz, Austria;
| | - Katharina Jandl
- Ludwig Boltzmann Institute for Lung Vascular Research, 8010 Graz, Austria;
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria
| | - Grazyna Kwapiszewska
- Otto Loewi Research Center, Division of Physiology, Medical University of Graz, 8010 Graz, Austria;
- Ludwig Boltzmann Institute for Lung Vascular Research, 8010 Graz, Austria;
- Correspondence:
| |
Collapse
|
12
|
Maize adaptation across temperate climates was obtained via expression of two florigen genes. PLoS Genet 2020; 16:e1008882. [PMID: 32673315 DOI: 10.1371/journal.pgen.1008882] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 07/28/2020] [Accepted: 05/22/2020] [Indexed: 11/19/2022] Open
Abstract
Expansion of the maize growing area was central for food security in temperate regions. In addition to the suppression of the short-day requirement for floral induction, it required breeding for a large range of flowering time that compensates the effect of South-North gradients of temperatures. Here we show the role of a novel florigen gene, ZCN12, in the latter adaptation in cooperation with ZCN8. Strong eQTLs of ZCN8 and ZCN12, measured in 327 maize lines, accounted for most of the genetic variance of flowering time in platform and field experiments. ZCN12 had a strong effect on flowering time of transgenic Arabidopsis thaliana plants; a path analysis showed that it directly affected maize flowering time together with ZCN8. The allelic composition at ZCN QTLs showed clear signs of selection by breeders. This suggests that florigens played a central role in ensuring a large range of flowering time, necessary for adaptation to temperate areas.
Collapse
|
13
|
Harbaum L, Rhodes CJ, Otero-Núñez P, Wharton J, Wilkins MR. The application of 'omics' to pulmonary arterial hypertension. Br J Pharmacol 2020; 178:108-120. [PMID: 32201940 DOI: 10.1111/bph.15056] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 01/03/2020] [Accepted: 02/18/2020] [Indexed: 12/14/2022] Open
Abstract
Recent genome-wide analyses of rare and common sequence variations have brought greater clarity to the genetic architecture of pulmonary arterial hypertension and implicated novel genes in disease development. Transcriptional signatures have been reported in whole lung tissue, pulmonary vascular cells and peripheral circulating cells. High-throughput platforms for plasma proteomics and metabolomics have identified novel biomarkers associated with clinical outcomes and provided molecular instruments for risk assessment. There are methodological challenges to integrating these datasets, coupled to statistical power limitations inherent to the study of a rare disease, but the expectation is that this approach will reveal novel druggable targets and biomarkers that will open the way to personalized medicine. Here, we review the current state-of-the-art and future promise of 'omics' in the field of translational medicine in pulmonary arterial hypertension. LINKED ARTICLES: This article is part of a themed issue on Risk factors, comorbidities, and comedications in cardioprotection. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.1/issuetoc.
Collapse
Affiliation(s)
- Lars Harbaum
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Pablo Otero-Núñez
- National Heart and Lung Institute, Imperial College London, London, UK
| | - John Wharton
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Martin R Wilkins
- National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
14
|
Noncanonical Wnt planar cell polarity signaling in lung development and disease. Biochem Soc Trans 2020; 48:231-243. [PMID: 32096543 DOI: 10.1042/bst20190597] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/24/2020] [Accepted: 01/28/2020] [Indexed: 02/06/2023]
Abstract
The planar cell polarity (PCP) signaling pathway is a potent developmental regulator of directional cell behaviors such as migration, asymmetric division and morphological polarization that are critical for shaping the body axis and the complex three-dimensional architecture of tissues and organs. PCP is considered a noncanonical Wnt pathway due to the involvement of Wnt ligands and Frizzled family receptors in the absence of the beta-catenin driven gene expression observed in the canonical Wnt cascade. At the heart of the PCP mechanism are protein complexes capable of generating molecular asymmetries within cells along a tissue-wide axis that are translated into polarized actin and microtubule cytoskeletal dynamics. PCP has emerged as an important regulator of developmental, homeostatic and disease processes in the respiratory system. It acts along other signaling pathways to create the elaborately branched structure of the lung by controlling the directional protrusive movements of cells during branching morphogenesis. PCP operates in the airway epithelium to establish and maintain the orientation of respiratory cilia along the airway axis for anatomically directed mucociliary clearance. It also regulates the establishment of the pulmonary vasculature. In adult tissues, PCP dysfunction has been linked to a variety of chronic lung diseases such as cystic fibrosis, chronic obstructive pulmonary disease, and idiopathic pulmonary arterial hypertension, stemming chiefly from the breakdown of proper tissue structure and function and aberrant cell migration during regenerative wound healing. A better understanding of these (impaired) PCP mechanisms is needed to fully harness the therapeutic opportunities of targeting PCP in chronic lung diseases.
Collapse
|
15
|
Zhang S, Liu X, Ge LL, Li K, Sun Y, Wang F, Han Y, Sun C, Wang J, Jiang W, Xin Q, Xu C, Chen Y, Chen O, Zhang Z, Luan Y. Mesenchymal stromal cell-derived exosomes improve pulmonary hypertension through inhibition of pulmonary vascular remodeling. Respir Res 2020; 21:71. [PMID: 32192495 PMCID: PMC7082982 DOI: 10.1186/s12931-020-1331-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 02/28/2020] [Indexed: 12/15/2022] Open
Abstract
Background Pulmonary hypertension (PH) is a life-threatening disease characterized by pulmonary vascular remodeling, right ventricular hypertrophy and failure. So far no effective treatment exists for this disease; hence, novel approaches are urgently needed. The aim of the present research was to observe the treatment effect of mesenchymal stromal cell derived exosomes and reveal the mechanism. Methods Monocrotaline (MCT)-induced PH in rats and hypoxia-induced cell damage model were established, respectively. Exosomes derived from the supernatant of human umbilical cord mesenchymal stem cells (MSC-exo) were injected into MCT-PH model rat or added into the cells cultured medium. Immunohistochemistry, quantitative real-time polymerase chain reaction (qRT-PCR) and western blot methods were used in vivo and vitro. Results The results showed that MSC-exo could significantly attenuate right ventricular (RV) hypertrophy and pulmonary vascular remodelling in MCT-PH rats. In the cell culture experiments, we found that MSC-exo could significantly inhibit hypoxia-induced pulmonary arterial endothelial cell (PAEC) apoptosis and pulmonary arterial smooth muscle cells (PASMC) proliferation. Furthermore, the pulmonary arterioles endothelial-to-mesenchymal transition (EndMT) was obviously suppressed. Moreover, the present study suggest that MSC-exo can significantly upregulate the expression of Wnt5a in MCT-PH rats and hypoxic pulmonary vascular cells. Furthermore, with Wnt5a gene silencing, the therapeutic effect of MSC-exo against hypoxia injury was restrained. Conclusions Synthetically, our data provide a strong evidence for the therapeutic of MSC-exo on PH, more importantly, we confirmed that the mechanism was associated with up-regulation of the expression of Wnt5a. These results offer a theoretical basis for clinical prevention and treatment of PH.
Collapse
Affiliation(s)
- Shanshan Zhang
- The Second Hospital of Shandong University, No. 247, Beiyuan Dajie, Jinan, 250033, People's Republic of China
| | - Xiaoli Liu
- Department of Hematology, The Second Hospital of Shandong University, Jinan, People's Republic of China.,Institute of Biotherapy for Hematological Malignancies, The Second Hospital of Shandong University, Jinan, People's Republic of China
| | - Li Li Ge
- Department of Special Inspection, The Second Hospital of Shandong University, Jinan, People's Republic of China
| | - Kailin Li
- Institute of Medical Science, Central Research Laboratory, The Second Hospital of Shandong University, No. 247, Beiyuan Dajie, Jinan, 250033, People's Republic of China
| | - Yongchao Sun
- Department of Medicine, Jinan Vocational College of Nursing, Jinan, People's Republic of China
| | - Fang Wang
- Institute of Medical Science, Animal center, The Second Hospital of Shandong University, Jinan, People's Republic of China
| | - Ying Han
- Institute of Medical Science, Animal center, The Second Hospital of Shandong University, Jinan, People's Republic of China
| | - Chao Sun
- Institute of Medical Science, Central Research Laboratory, The Second Hospital of Shandong University, No. 247, Beiyuan Dajie, Jinan, 250033, People's Republic of China
| | - Jue Wang
- Institute of Medical Science, Central Research Laboratory, The Second Hospital of Shandong University, No. 247, Beiyuan Dajie, Jinan, 250033, People's Republic of China
| | - Wen Jiang
- Institute of Medical Science, Central Research Laboratory, The Second Hospital of Shandong University, No. 247, Beiyuan Dajie, Jinan, 250033, People's Republic of China
| | - Qian Xin
- Institute of Medical Science, Central Research Laboratory, The Second Hospital of Shandong University, No. 247, Beiyuan Dajie, Jinan, 250033, People's Republic of China
| | - Chaoyue Xu
- Department of Pediatrics, The Second Hospital of Shandong University, Jinan, People's Republic of China
| | - Yuan Chen
- Institute of Medical Science, Central Research Laboratory, The Second Hospital of Shandong University, No. 247, Beiyuan Dajie, Jinan, 250033, People's Republic of China
| | - Ou Chen
- School of nursing, Shandong University, Jinan, People's Republic of China
| | - Zhaohua Zhang
- Department of Pediatrics, The Second Hospital of Shandong University, Jinan, People's Republic of China
| | - Yun Luan
- Institute of Medical Science, Central Research Laboratory, The Second Hospital of Shandong University, No. 247, Beiyuan Dajie, Jinan, 250033, People's Republic of China.
| |
Collapse
|
16
|
Cui S, Wu Q, West J, Bai J. Machine learning-based microarray analyses indicate low-expression genes might collectively influence PAH disease. PLoS Comput Biol 2019; 15:e1007264. [PMID: 31404060 PMCID: PMC6705875 DOI: 10.1371/journal.pcbi.1007264] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 08/22/2019] [Accepted: 07/11/2019] [Indexed: 11/19/2022] Open
Abstract
Accurately predicting and testing the types of Pulmonary arterial hypertension (PAH) of each patient using cost-effective microarray-based expression data and machine learning algorithms could greatly help either identifying the most targeting medicine or adopting other therapeutic measures that could correct/restore defective genetic signaling at the early stage. Furthermore, the prediction model construction processes can also help identifying highly informative genes controlling PAH, leading to enhanced understanding of the disease etiology and molecular pathways. In this study, we used several different gene filtering methods based on microarray expression data obtained from a high-quality patient PAH dataset. Following that, we proposed a novel feature selection and refinement algorithm in conjunction with well-known machine learning methods to identify a small set of highly informative genes. Results indicated that clusters of small-expression genes could be extremely informative at predicting and differentiating different forms of PAH. Additionally, our proposed novel feature refinement algorithm could lead to significant enhancement in model performance. To summarize, integrated with state-of-the-art machine learning and novel feature refining algorithms, the most accurate models could provide near-perfect classification accuracies using very few (close to ten) low-expression genes. Pulmonary arterial hypertension (PAH) is a serious and progressive disease, with only a roughly 50% of 5-year survival rate even with best available therapies. Accurately detecting/differentiating different forms of PAH and developing drugs that could directly target at genes involved in PAH pathogenesis are essential. We proposed a computational approach using low-cost microarray data collected from a clinical trial and had accurately predicted each PAH group. In particular, we considered the fact that there might exist some low-expression genes that were usually discarded by researchers but might function collectively and significantly controlling the disease in each case. Therefore, we had developed different filtering algorithms that intentionally selected those low-expression genes for constructing prediction model. Using a few highly informative low-expression genes that had never been extensively investigated before, our systematic approach had produced models that could offer prefect accuracy in predicting PAH. Additionally, our analysis also found that the composition of gene factors controlling the PAH etiology under each form are quite different from each other.
Collapse
Affiliation(s)
- Song Cui
- College of Agronomy, Gansu Agricultural University, Lanzhou, Gansu, China
- School of Agriculture, Middle Tennessee State University, Murfreesboro, Tennessee, United States of America
| | - Qiang Wu
- Department of Mathematics, Middle Tennessee State University, Murfreesboro, Tennessee, United States of America
| | - James West
- Department of Medicine, Pulmonary Vascular Research Institute, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Jiangping Bai
- College of Agronomy, Gansu Agricultural University, Lanzhou, Gansu, China
- * E-mail:
| |
Collapse
|
17
|
Zhang L, Chen S, Zeng X, Lin D, Li Y, Gui L, Lin MJ. Revealing the pathogenic changes of PAH based on multiomics characteristics. J Transl Med 2019; 17:231. [PMID: 31331330 PMCID: PMC6647123 DOI: 10.1186/s12967-019-1981-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 07/12/2019] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Pulmonary artery hypertension (PAH), which is characterized by an increase in pulmonary circulation blood pressure, is a fatal disease, and its pathogenesis remains unclear. METHODS In this study, RNA sequencing (RNA-seq), tandem mass tags (TMT) and reduced representation bisulfite sequencing (RRBS) were performed to detect the levels of mRNA, protein, and DNA methylation in pulmonary arteries (PAs), respectively. To screen the possible pathways and proteins related to PAH, pathway enrichment analysis and protein-protein interaction (PPI) network analysis were performed. For selected genes, differential expression levels were confirmed at both the transcriptional and translational levels by real-time PCR and Western blot analyses, respectively. RESULTS A total of 362 differentially expressed genes (|Fold-change| > 1.5 and p < 0.05), 811 differentially expressed proteins (|Fold-change| > 1.2 and p < 0.05) and 76,562 differentially methylated regions (1000 bp slide windows, 500 bp overlap, p < 0.05, and |Fold-change| > 1.2) were identified when the PAH group (n = 15) was compared with the control group (n = 15). Through an integrated analysis of the characteristics of the three omic analyses, a multiomics table was constructed. Additionally, pathway enrichment analysis showed that the differentially expressed proteins were significantly enriched in five Kyoto Encyclopedia of Genes and Genomes (KEGG) biological pathways and ten Gene Ontology (GO) terms for the PAH group compared with the control group. Moreover, protein-protein interaction (PPI) networks were constructed to identify hub genes. Finally, according to the genes identified in the PPI and the protein expression fold-change, nine key genes and their associated proteins were verified by real-time PCR and Western blot analyses, including Col4a1, Itga5, Col2a1, Gstt1, Gstm3, Thbd, Mgst2, Kng1 and Fgg. CONCLUSIONS This study conducted multiomic characteristic profiling to identify genes that contribute to the hypoxia-induced PAH model, identifying new avenues for basic PAH research.
Collapse
Affiliation(s)
- Li Zhang
- Department of Physiology & Pathophysiology, Fujian Medical University, Fuzhou, China.,The Key Laboratory of Fujian Province University on Ion Channel and Signal Transduction in Cardiovascular Disease, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Shaokun Chen
- Department of Physiology & Pathophysiology, Fujian Medical University, Fuzhou, China.,The Key Laboratory of Fujian Province University on Ion Channel and Signal Transduction in Cardiovascular Disease, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Xixi Zeng
- Department of Physiology & Pathophysiology, Fujian Medical University, Fuzhou, China.,The Key Laboratory of Fujian Province University on Ion Channel and Signal Transduction in Cardiovascular Disease, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Dacen Lin
- Department of Physiology & Pathophysiology, Fujian Medical University, Fuzhou, China.,The Key Laboratory of Fujian Province University on Ion Channel and Signal Transduction in Cardiovascular Disease, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Yumei Li
- The Key Laboratory of Fujian Province University on Ion Channel and Signal Transduction in Cardiovascular Disease, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.,Fujian Center for Safety Evaluation of New Drug, Fujian Medical University, Fuzhou, China
| | - Longxin Gui
- The Key Laboratory of Fujian Province University on Ion Channel and Signal Transduction in Cardiovascular Disease, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Mo-Jun Lin
- Department of Physiology & Pathophysiology, Fujian Medical University, Fuzhou, China. .,The Key Laboratory of Fujian Province University on Ion Channel and Signal Transduction in Cardiovascular Disease, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
18
|
Hamid R, Austin ED. End Stage Takes Center Stage in Pulmonary Arterial Hypertension. Am J Respir Cell Mol Biol 2019; 60:607-608. [PMID: 30726110 PMCID: PMC6543743 DOI: 10.1165/rcmb.2019-0022ed] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Rizwan Hamid
- 1 Department of Pediatrics Vanderbilt University Medical Center Nashville, Tennessee
| | - Eric D Austin
- 1 Department of Pediatrics Vanderbilt University Medical Center Nashville, Tennessee
| |
Collapse
|
19
|
de Oliveira AR, da Silva FS, Bortolin RH, Marques DEDS, Ramos GV, Marqueti RC, da Silva NB, Medeiros KCDP, Corrêa MA, Lima JPMS, de Rezende AA, Ackermann PW, Abreu BJ, de Brito Vieira WH. Effect of photobiomodulation and exercise on early remodeling of the Achilles tendon in streptozotocin-induced diabetic rats. PLoS One 2019; 14:e0211643. [PMID: 30716140 PMCID: PMC6361457 DOI: 10.1371/journal.pone.0211643] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 01/17/2019] [Indexed: 02/06/2023] Open
Abstract
The aim of this study was to compare the treatment effects of laser photobiomodulation (LPBM) therapy and aerobic exercise on the biomechanical properties, tissue morphology and the expression of tendon matrix molecules during early remodeling of Achilles tendon (AT) injury in diabetic rats. Animals were randomly assigned to five groups: injured non diabetic (I, n = 15), injured diabetic (ID, n = 15), injured diabetic plus LPBM (IDL, n = 16), injured diabetic plus aerobic exercise (IDE, n = 16) and injured diabetic plus aerobic exercise and LPBM (IDEAL, n = 17). Type 1 diabetes was induced via a single intravenous injection of Streptozotocin at a dose of 40 mg/kg. A partial tenotomy was performed in the right AT. LPBM was performed with an indium-gallium-aluminum-phosphide 660 nm 10 mW laser device (spot size 0.04 cm2, power density 250 mW/cm2, irradiation duration 16 s, energy 0.16 J, energy density 4 J/cm2) on alternate days for a total of 9 sessions over 3 weeks (total energy 1.44 J), using a stationary contact technique to a single point over the dorsal aspect of the AT. Moderate aerobic exercise was performed on a motorized treadmill (velocity 9 m/min for 60 minutes). At 3 weeks post-injury, biomechanical analyzes as well as assessment of fibroblast number and orientation were performed. Collagen 1 (Col1) and 3 (Col3) and matrix metalloproteinases (MMPs) -3 and 13 protein distributions were studied by immunohistochemistry; while Col1 and Col3 and MMP-2 and 9 gene expression were assessed by quantitative RT-PCR (qRT-PCR). IDEAL exhibited significant increases in several biomechanical parameters in comparison to the other groups. Moreover, IDEAL presented stronger Col1 immunoreactivity when compared to ID, and weaker Col3 immunoreactivity than IDE. Both IDL and IDEAL demonstrated weaker expression of MMP-3 in comparison to I, while IDL presented no expression of MMP-13 when compared to ID. ID, IDL and IDE showed an increased number of fibroblasts in comparison to I, while IDEAL decreased the number of these cells in comparison to ID and IDE. IDL and IDEAL groups exhibited decreased angular dispersion among the fibroblasts when compared to I. The gene expression results showed that IDE demonstrated a downregulation in Col1 mRNA expression in comparison to I and ID. IDEAL demonstrated upregulation of Col1 mRNA expression when compared to IDL or IDE alone and increased MMP-2 expression when compared to IDL and IDE. MMP-9 expression was upregulated in IDEAL when compared to I, IDL and IDE. Our results suggest a beneficial interaction of combining both treatment strategies i.e., aerobic exercise and LPBM, on the biomechanical properties, tissue morphology and the expression of matrix molecules in diabetic tendons.
Collapse
MESH Headings
- Achilles Tendon/metabolism
- Achilles Tendon/physiopathology
- Animals
- Collagen Type I/metabolism
- Collagen Type III/metabolism
- Diabetes Mellitus, Experimental/chemically induced
- Diabetes Mellitus, Experimental/etiology
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/physiopathology
- Diabetes Mellitus, Type 1/chemically induced
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/physiopathology
- Fibroblasts/metabolism
- Low-Level Light Therapy/methods
- Male
- Metalloendopeptidases/metabolism
- RNA, Messenger/metabolism
- Rats
- Rats, Wistar
- Streptozocin/pharmacology
- Tendon Injuries/etiology
- Tendon Injuries/metabolism
- Tendon Injuries/physiopathology
- Tendon Injuries/therapy
- Up-Regulation/physiology
- Wound Healing/physiology
Collapse
Affiliation(s)
| | - Flávio Santos da Silva
- Department of Health Sciences, Federal University of the Semiarid Region, Mossoró, Brazil
| | - Raul Hernandes Bortolin
- Department of Clinical and Toxicological Analysis, Federal University of Rio Grande do Norte, Natal, Brazil
| | | | | | | | | | | | | | | | - Adriana Augusto de Rezende
- Department of Clinical and Toxicological Analysis, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Paul W. Ackermann
- Department of Molecular Medicine and Surgery, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Bento J. Abreu
- Department of Morphology, Federal University of Rio Grande do Norte, Natal, Brazil
| | | |
Collapse
|
20
|
Huang J, Frid M, Gewitz MH, Fallon JT, Brown D, Krafsur G, Stenmark K, Mathew R. Hypoxia-induced pulmonary hypertension and chronic lung disease: caveolin-1 dysfunction an important underlying feature. Pulm Circ 2019; 9:2045894019837876. [PMID: 30806156 PMCID: PMC6434444 DOI: 10.1177/2045894019837876] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 02/22/2019] [Indexed: 12/30/2022] Open
Abstract
Caveolin-1 (cav-1) has been shown to play a significant role in the pathogenesis of pulmonary hypertension (PH). In the monocrotaline model of PH, the loss of endothelial cav-1 as well as reciprocal activation of proliferative and anti-apoptotic pathways initiate the disease process and facilitate its progression. In order to examine the role of cav-1 in hypoxia-induced PH, we exposed rats and neonatal calves to hypobaric hypoxia and obtained hemodynamic data and assessed the expression of cav-1 and related proteins eNOS, HSP90, PTEN, gp130, PY-STAT3, β-catenin, and Glut1 in the lung tissue. Chronic hypoxic exposure in rats (48 h-4 weeks) and calves (two weeks) did not alter the expression of cav-1, HSP90, or eNOS. PTEN expression was significantly decreased accompanied by PY-STAT3 activation and increased expression of gp130, Glut1, and β-catenin in hypoxic animals. We also examined cav-1 expression in the lung sections from steers with chronic hypoxic disease (Brisket disease) and from patients with chronic lung disease who underwent lung biopsy for medical reasons. There was no cav-1 loss in Brisket disease. In chronic lung disease cases, endothelial cav-1 expression was present, albeit with less intense staining in some cases. In conclusion, hypoxia did not alter the cav-1 expression in experimental models. The presence of cav-1, however, did not suppress hypoxia-induced activation of PY-STAT3 and β catenin, increased gp130 and Glut1 expression, or prevent the PTEN loss, indicating cav-1 dysfunction in hypoxia-induced PH.
Collapse
Affiliation(s)
- Jing Huang
- Department of Pediatrics, Maria Fareri Children’s Hospital at Westchester Medical Center, New York Medical College, Valhalla, NY, USA
| | - Maria Frid
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Michael H. Gewitz
- Department of Pediatrics, Maria Fareri Children’s Hospital at Westchester Medical Center, New York Medical College, Valhalla, NY, USA
| | - John T. Fallon
- Department of Pathology, New York Medical College, Valhalla, NY, USA
| | - Dale Brown
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Greta Krafsur
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kurt Stenmark
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Rajamma Mathew
- Department of Pediatrics, Maria Fareri Children’s Hospital at Westchester Medical Center, New York Medical College, Valhalla, NY, USA
- Department of Physiology, New York Medical College, Valhalla, NY, USA
| |
Collapse
|
21
|
Dieffenbach PB, Maracle M, Tschumperlin DJ, Fredenburgh LE. Mechanobiological Feedback in Pulmonary Vascular Disease. Front Physiol 2018; 9:951. [PMID: 30090065 PMCID: PMC6068271 DOI: 10.3389/fphys.2018.00951] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 06/28/2018] [Indexed: 01/06/2023] Open
Abstract
Vascular stiffening in the pulmonary arterial bed is increasingly recognized as an early disease marker and contributor to right ventricular workload in pulmonary hypertension. Changes in pulmonary artery stiffness throughout the pulmonary vascular tree lead to physiologic alterations in pressure and flow characteristics that may contribute to disease progression. These findings have led to a greater focus on the potential contributions of extracellular matrix remodeling and mechanical signaling to pulmonary hypertension pathogenesis. Several recent studies have demonstrated that the cellular response to vascular stiffness includes upregulation of signaling pathways that precipitate further vascular remodeling, a process known as mechanobiological feedback. The extracellular matrix modifiers, mechanosensors, and mechanotransducers responsible for this process have become increasingly well-recognized. In this review, we discuss the impact of vascular stiffening on pulmonary hypertension morbidity and mortality, evidence in favor of mechanobiological feedback in pulmonary hypertension pathogenesis, and the major contributors to mechanical signaling in the pulmonary vasculature.
Collapse
Affiliation(s)
- Paul B Dieffenbach
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States
| | - Marcy Maracle
- Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Daniel J Tschumperlin
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Laura E Fredenburgh
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States
| |
Collapse
|
22
|
Skronska-Wasek W, Gosens R, Königshoff M, Baarsma HA. WNT receptor signalling in lung physiology and pathology. Pharmacol Ther 2018; 187:150-166. [PMID: 29458107 DOI: 10.1016/j.pharmthera.2018.02.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The WNT signalling cascades have emerged as critical regulators of a wide variety of biological aspects involved in lung development as well as in physiological and pathophysiological processes in the adult lung. WNTs (secreted glycoproteins) interact with various transmembrane receptors and co-receptors to activate signalling pathways that regulate transcriptional as well as non-transcriptional responses within cells. In physiological conditions, the majority of WNT receptors and co-receptors can be detected in the adult lung. However, dysregulation of WNT signalling pathways contributes to the development and progression of chronic lung pathologies, including idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD), asthma and lung cancer. The interaction between a WNT and the (co-)receptor(s) present at the cell surface is the initial step in transducing an extracellular signal into an intracellular response. This proximal event in WNT signal transduction with (cell-specific) ligand-receptor interactions is of great interest as a potential target for pharmacological intervention. In this review we highlight the diverse expression of various WNT receptors and co-receptors in the aforementioned chronic lung diseases and discuss the currently available biologicals and pharmacological tools to modify proximal WNT signalling.
Collapse
Affiliation(s)
- Wioletta Skronska-Wasek
- Comprehensive Pneumology Center, Research Unit Lung Repair and Regeneration, Helmholtz Center Munich, Member of the German Center for Lung Research, Ludwig Maximilians University Munich, University Hospital Grosshadern, Munich, Germany
| | - Reinoud Gosens
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands; GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Melanie Königshoff
- Comprehensive Pneumology Center, Research Unit Lung Repair and Regeneration, Helmholtz Center Munich, Member of the German Center for Lung Research, Ludwig Maximilians University Munich, University Hospital Grosshadern, Munich, Germany; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA.
| | - Hoeke Abele Baarsma
- Comprehensive Pneumology Center, Research Unit Lung Repair and Regeneration, Helmholtz Center Munich, Member of the German Center for Lung Research, Ludwig Maximilians University Munich, University Hospital Grosshadern, Munich, Germany; GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
23
|
Foulquier S, Daskalopoulos EP, Lluri G, Hermans KCM, Deb A, Blankesteijn WM. WNT Signaling in Cardiac and Vascular Disease. Pharmacol Rev 2018; 70:68-141. [PMID: 29247129 PMCID: PMC6040091 DOI: 10.1124/pr.117.013896] [Citation(s) in RCA: 234] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
WNT signaling is an elaborate and complex collection of signal transduction pathways mediated by multiple signaling molecules. WNT signaling is critically important for developmental processes, including cell proliferation, differentiation and tissue patterning. Little WNT signaling activity is present in the cardiovascular system of healthy adults, but reactivation of the pathway is observed in many pathologies of heart and blood vessels. The high prevalence of these pathologies and their significant contribution to human disease burden has raised interest in WNT signaling as a potential target for therapeutic intervention. In this review, we first will focus on the constituents of the pathway and their regulation and the different signaling routes. Subsequently, the role of WNT signaling in cardiovascular development is addressed, followed by a detailed discussion of its involvement in vascular and cardiac disease. After highlighting the crosstalk between WNT, transforming growth factor-β and angiotensin II signaling, and the emerging role of WNT signaling in the regulation of stem cells, we provide an overview of drugs targeting the pathway at different levels. From the combined studies we conclude that, despite the sometimes conflicting experimental data, a general picture is emerging that excessive stimulation of WNT signaling adversely affects cardiovascular pathology. The rapidly increasing collection of drugs interfering at different levels of WNT signaling will allow the evaluation of therapeutic interventions in the pathway in relevant animal models of cardiovascular diseases and eventually in patients in the near future, translating the outcomes of the many preclinical studies into a clinically relevant context.
Collapse
Affiliation(s)
- Sébastien Foulquier
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands (S.F., K.C.M.H., W.M.B.); Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium (E.P.D.); Department of Medicine, Division of Cardiology, David Geffen School of Medicine (G.L., A.D.); and Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, Los Angeles, California (A.D.)
| | - Evangelos P Daskalopoulos
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands (S.F., K.C.M.H., W.M.B.); Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium (E.P.D.); Department of Medicine, Division of Cardiology, David Geffen School of Medicine (G.L., A.D.); and Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, Los Angeles, California (A.D.)
| | - Gentian Lluri
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands (S.F., K.C.M.H., W.M.B.); Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium (E.P.D.); Department of Medicine, Division of Cardiology, David Geffen School of Medicine (G.L., A.D.); and Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, Los Angeles, California (A.D.)
| | - Kevin C M Hermans
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands (S.F., K.C.M.H., W.M.B.); Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium (E.P.D.); Department of Medicine, Division of Cardiology, David Geffen School of Medicine (G.L., A.D.); and Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, Los Angeles, California (A.D.)
| | - Arjun Deb
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands (S.F., K.C.M.H., W.M.B.); Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium (E.P.D.); Department of Medicine, Division of Cardiology, David Geffen School of Medicine (G.L., A.D.); and Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, Los Angeles, California (A.D.)
| | - W Matthijs Blankesteijn
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands (S.F., K.C.M.H., W.M.B.); Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium (E.P.D.); Department of Medicine, Division of Cardiology, David Geffen School of Medicine (G.L., A.D.); and Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, Los Angeles, California (A.D.)
| |
Collapse
|
24
|
Lozano-Velasco E, Wangensteen R, Quesada A, Garcia-Padilla C, Osorio JA, Ruiz-Torres MD, Aranega A, Franco D. Hyperthyroidism, but not hypertension, impairs PITX2 expression leading to Wnt-microRNA-ion channel remodeling. PLoS One 2017; 12:e0188473. [PMID: 29194452 PMCID: PMC5711019 DOI: 10.1371/journal.pone.0188473] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 11/07/2017] [Indexed: 01/06/2023] Open
Abstract
PITX2 is a homeobox transcription factor involved in embryonic left/right signaling and more recently has been associated to cardiac arrhythmias. Genome wide association studies have pinpointed PITX2 as a major player underlying atrial fibrillation (AF). We have previously described that PITX2 expression is impaired in AF patients. Furthermore, distinct studies demonstrate that Pitx2 insufficiency leads to complex gene regulatory network remodeling, i.e. Wnt>microRNAs, leading to ion channel impairment and thus to arrhythmogenic events in mice. Whereas large body of evidences has been provided in recent years on PITX2 downstream signaling pathways, scarce information is available on upstream pathways influencing PITX2 in the context of AF. Multiple risk factors are associated to the onset of AF, such as e.g. hypertension (HTN), hyperthyroidism (HTD) and redox homeostasis impairment. In this study we have analyzed whether HTN, HTD and/or redox homeostasis impact on PITX2 and its downstream signaling pathways. Using rat models for spontaneous HTN (SHR) and experimentally-induced HTD we have observed that both cardiovascular risk factors lead to severe Pitx2 downregulation. Interesting HTD, but not SHR, leads to up-regulation of Wnt signaling as well as deregulation of multiple microRNAs and ion channels as previously described in Pitx2 insufficiency models. In addition, redox signaling is impaired in HTD but not SHR, in line with similar findings in atrial-specific Pitx2 deficient mice. In vitro cell culture analyses using gain- and loss-of-function strategies demonstrate that Pitx2, Zfhx3 and Wnt signaling influence redox homeostasis in cardiomyocytes. Thus, redox homeostasis seems to play a pivotal role in this setting, providing a regulatory feedback loop. Overall these data demonstrate that HTD, but not HTN, can impair Pitx2>>Wnt pathway providing thus a molecular link to AF.
Collapse
Affiliation(s)
- Estefanía Lozano-Velasco
- Cardiac and Skeletal Muscle Development Group, Department of Experimental Biology, University of Jaen, Jaen, Spain
| | | | - Andrés Quesada
- Department of Health Sciences, University of Jaen, Jaen, Spain
| | - Carlos Garcia-Padilla
- Cardiac and Skeletal Muscle Development Group, Department of Experimental Biology, University of Jaen, Jaen, Spain
| | - Julia A. Osorio
- Cardiac and Skeletal Muscle Development Group, Department of Experimental Biology, University of Jaen, Jaen, Spain
| | - María Dolores Ruiz-Torres
- Cardiac and Skeletal Muscle Development Group, Department of Experimental Biology, University of Jaen, Jaen, Spain
| | - Amelia Aranega
- Cardiac and Skeletal Muscle Development Group, Department of Experimental Biology, University of Jaen, Jaen, Spain
| | - Diego Franco
- Cardiac and Skeletal Muscle Development Group, Department of Experimental Biology, University of Jaen, Jaen, Spain
| |
Collapse
|
25
|
Kropski JA, Richmond BW, Gaskill CF, Foronjy RF, Majka SM. Deregulated angiogenesis in chronic lung diseases: a possible role for lung mesenchymal progenitor cells (2017 Grover Conference Series). Pulm Circ 2017; 8:2045893217739807. [PMID: 29040010 PMCID: PMC5731726 DOI: 10.1177/2045893217739807] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Chronic lung disease (CLD), including pulmonary fibrosis (PF) and chronic obstructive pulmonary disease (COPD), is the fourth leading cause of mortality worldwide. Both are debilitating pathologies that impede overall tissue function. A common co-morbidity in CLD is vasculopathy, characterized by deregulated angiogenesis, remodeling, and loss of microvessels. This substantially worsens prognosis and limits survival, with most current therapeutic strategies being largely palliative. The relevance of angiogenesis, both capillary and lymph, to the pathophysiology of CLD has not been resolved as conflicting evidence depicts angiogenesis as both reparative or pathologic. Therefore, we must begin to understand and model the underlying pathobiology of pulmonary vascular deregulation, alone and in response to injury induced disease, to define cell interactions necessary to maintain normal function and promote repair. Capillary and lymphangiogenesis are deregulated in both PF and COPD, although the mechanisms by which they co-regulate and underlie early pathogenesis of disease are unknown. The cell-specific mechanisms that regulate lung vascular homeostasis, repair, and remodeling represent a significant gap in knowledge, which presents an opportunity to develop targeted therapies. We have shown that that ABCG2pos multipotent adult mesenchymal stem or progenitor cells (MPC) influence the function of the capillary microvasculature as well as lymphangiogenesis. A balance of both is required for normal tissue homeostasis and repair. Our current models suggest that when lymph and capillary angiogenesis are out of balance, the non-equivalence appears to support the progression of disease and tissue remodeling. The angiogenic regulatory mechanisms underlying CLD likely impact other interstitial lung diseases, tuberous sclerosis, and lymphangioleiomyomatosis.
Collapse
Affiliation(s)
- Jonathan A Kropski
- 1 12328 Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bradley W Richmond
- 1 12328 Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Christa F Gaskill
- 1 12328 Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Robert F Foronjy
- 3 5718 Department of Medicine, Vanderbilt University, Nashville, TN, USA
| | - Susan M Majka
- 1 12328 Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.,2 74498 Department of Medicine, Division of Pulmonary and Critical Care Medicine, SUNY Downstate Medical Center, Brooklyn, NY, USA
| |
Collapse
|
26
|
Yun X, Jiang H, Lai N, Wang J, Shimoda LA. Aquaporin 1-mediated changes in pulmonary arterial smooth muscle cell migration and proliferation involve β-catenin. Am J Physiol Lung Cell Mol Physiol 2017; 313:L889-L898. [PMID: 28798257 DOI: 10.1152/ajplung.00247.2016] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 07/18/2017] [Accepted: 07/31/2017] [Indexed: 12/22/2022] Open
Abstract
Exposure to hypoxia induces migration and proliferation of pulmonary arterial smooth muscle cells (PASMCs), leading to vascular remodeling and contributing to the development of hypoxic pulmonary hypertension. The mechanisms controlling PASMC growth and motility are incompletely understood, although aquaporin 1 (AQP1) plays an important role. In tumor, kidney, and stem cells, AQP1 has been shown to interact with β-catenin, a dual function protein that activates the transcription of crucial target genes (i.e., c-Myc and cyclin D1) related to cell migration and proliferation. Thus the goal of this study was to examine mechanisms by which AQP1 mediates PASMC migration and proliferation, with a focus on β-catenin. Using primary rat PASMCs from resistance level pulmonary arteries infected with adenoviral constructs containing green fluorescent protein (control; AdGFP), wild-type AQP1 (AdAQP1), or AQP1 with the COOH-terminal tail deleted (AdAQP1M), we demonstrated that increasing AQP1 expression upregulated β-catenin protein levels and the expression (mRNA and protein) of the known β-catenin targets c-Myc and cyclin D1. In contrast, infection with AdAQP1M had no effect on any of these variables. Using silencing approaches to reduce β-catenin levels prevented both hypoxia- and AQP1-induced migration and proliferation of PASMCs, as well as induction of c-Myc and cyclin D1 by AQP1. Thus our results indicate that elevated AQP1 levels upregulate β-catenin protein levels, via a mechanism requiring the AQP1 COOH-terminal tail, enhancing expression of β-catenin targets and promoting PASMC proliferation and migration.
Collapse
Affiliation(s)
- Xin Yun
- Guangzhou Institute of Respiratory Diseases, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China; and.,Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Haiyang Jiang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Ning Lai
- Guangzhou Institute of Respiratory Diseases, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China; and
| | - Jian Wang
- Guangzhou Institute of Respiratory Diseases, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China; and.,Division of Pulmonary and Critical Care Medicine, Department of 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
| |
Collapse
|
27
|
Manichaikul A, Wang XQ, Sun L, Dupuis J, Borczuk AC, Nguyen JN, Raghu G, Hoffman EA, Onengut-Gumuscu S, Farber EA, Kaufman JD, Rabinowitz D, Stukovsky KDH, Kawut SM, Hunninghake GM, Washko GR, O'Connor GT, Rich SS, Barr RG, Lederer DJ. Genome-wide association study of subclinical interstitial lung disease in MESA. Respir Res 2017; 18:97. [PMID: 28521775 PMCID: PMC5437638 DOI: 10.1186/s12931-017-0581-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 05/10/2017] [Indexed: 12/14/2022] Open
Abstract
Background We conducted a genome-wide association study (GWAS) of subclinical interstitial lung disease (ILD), defined as high attenuation areas (HAA) on CT, in the population-based Multi-Ethnic Study of Atherosclerosis Study. Methods We measured the percentage of high attenuation areas (HAA) in the lung fields on cardiac CT scan defined as voxels with CT attenuation values between -600 and -250 HU. Genetic analyses were performed in MESA combined across race/ethnic groups: non-Hispanic White (n = 2,434), African American (n = 2,470), Hispanic (n = 2,065) and Chinese (n = 702), as well as stratified by race/ethnicity. Results Among 7,671 participants, regions at genome-wide significance were identified for basilar peel-core ratio of HAA in FLJ35282 downstream of ANRIL (rs7852363, P = 2.1x10−9) and within introns of SNAI3-AS1 (rs140142658, P = 9.6x10−9) and D21S2088E (rs3079677, P = 2.3x10−8). Within race/ethnic groups, 18 additional loci were identified at genome-wide significance, including genes related to development (FOXP4), cell adhesion (ALCAM) and glycosylation (GNPDA2, GYPC, GFPT1 and FUT10). Among these loci, SNP rs6844387 near GNPDA2 demonstrated nominal evidence of replication in analysis of n = 1,959 participants from the Framingham Heart Study (P = 0.029). FOXP4 region SNP rs2894439 demonstrated evidence of validation in analysis of n = 228 White ILD cases from the Columbia ILD Study compared to race/ethnicity-matched controls from MESA (one-sided P = 0.007). In lung tissue from 15 adults with idiopathic pulmonary fibrosis compared to 15 adults without lung disease. ANRIL (P = 0.001), ALCAM (P = 0.03) and FOXP4 (P = 0.046) were differentially expressed. Conclusions Our results suggest novel roles for protein glycosylation and cell cycle disinhibition by long non-coding RNA in the pathogenesis of ILD. Electronic supplementary material The online version of this article (doi:10.1186/s12931-017-0581-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ani Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA. .,Department of Public Health Sciences, Biostatistics Section, University of Virginia, Charlottesville, VA, USA. .,Center for Public Health Genomics, University of Virginia School of Medicine, West Complex Room 6115, Charlottesville, VA, 22903, USA.
| | - Xin-Qun Wang
- Department of Public Health Sciences, Biostatistics Section, University of Virginia, Charlottesville, VA, USA
| | - Li Sun
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Josée Dupuis
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA.,The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA, USA
| | - Alain C Borczuk
- Department of Pathology, Weill Cornell Medicine, New York, NY, USA
| | - Jennifer N Nguyen
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Ganesh Raghu
- University of Washington Center for Interstitial Lung Diseases, Seattle, WA, USA
| | - Eric A Hoffman
- Department of Radiology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Suna Onengut-Gumuscu
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Emily A Farber
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Joel D Kaufman
- Departmenst of Environmental & Occupational Health Sciences, Medicine, and Epidemiology, University of Washington, Seattle, WA, USA
| | - Dan Rabinowitz
- Department of Statistics, Columbia University, New York, NY, USA
| | | | - Steven M Kawut
- Department of Medicine and Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gary M Hunninghake
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - George R Washko
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - George T O'Connor
- The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA, USA.,Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - R Graham Barr
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA.,Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - David J Lederer
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA.,Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| |
Collapse
|
28
|
Baarsma HA, Königshoff M. 'WNT-er is coming': WNT signalling in chronic lung diseases. Thorax 2017; 72:746-759. [PMID: 28416592 PMCID: PMC5537530 DOI: 10.1136/thoraxjnl-2016-209753] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Revised: 03/01/2017] [Accepted: 03/16/2017] [Indexed: 02/06/2023]
Abstract
Chronic lung diseases represent a major public health problem with only limited therapeutic options. An important unmet need is to identify compounds and drugs that target key molecular pathways involved in the pathogenesis of chronic lung diseases. Over the last decade, there has been extensive interest in investigating Wingless/integrase-1 (WNT) signalling pathways; and WNT signal alterations have been linked to pulmonary disease pathogenesis and progression. Here, we comprehensively review the cumulative evidence for WNT pathway alterations in chronic lung pathologies, including idiopathic pulmonary fibrosis, pulmonary arterial hypertension, asthma and COPD. While many studies have focused on the canonical WNT/β-catenin signalling pathway, recent reports highlight that non-canonical WNT signalling may also significantly contribute to chronic lung pathologies; these studies will be particularly featured in this review. We further discuss recent advances uncovering the role of WNT signalling early in life, the potential of pharmaceutically modulating WNT signalling pathways and highlight (pre)clinical studies describing promising new therapies for chronic lung diseases.
Collapse
Affiliation(s)
- H A Baarsma
- Comprehensive Pneumology Center, Helmholtz Center Munich, Ludwig Maximilians University Munich, University Hospital Grosshadern, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - M Königshoff
- Comprehensive Pneumology Center, Helmholtz Center Munich, Ludwig Maximilians University Munich, University Hospital Grosshadern, Member of the German Center for Lung Research (DZL), Munich, Germany.,Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
29
|
Yanai S, Wakayama M, Nakayama H, Shinozaki M, Tsukuma H, Tochigi N, Nemoto T, Saji T, Shibuya K. Implication of overexpression of dishevelled-associated activator of morphogenesis 1 (Daam-1) for the pathogenesis of human Idiopathic Pulmonary Arterial Hypertension (IPAH). Diagn Pathol 2017; 12:25. [PMID: 28288669 PMCID: PMC5348773 DOI: 10.1186/s13000-017-0614-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 02/20/2017] [Indexed: 01/31/2023] Open
Abstract
Background Idiopathic pulmonary arterial hypertension (IPAH) is a rare, fatal disease of unknown pathogenesis. Evidence from our recent study suggests that IPAH pathogenesis is related to upregulation of the Wnt/planar cell polarity (Wnt/PCP) pathway. We used microscopic observation and immunohistochemical techniques to identify expression patterns of cascading proteins—namely Wnt-11, dishevelled-2 (Dvl-2), and dishevelled-associated activator of morphogenesis 1 (Daam-1)—in pulmonary arteries. Methods We analyzed sections of formalin-fixed and paraffin-embedded autopsied lung tissues obtained from 9 IPAH cases, 7 associated pulmonary arterial hypertension cases, and 16 age-matched controls without pulmonary arterial abnormalities. Results of microscopic observation were analyzed in relation to the cellular components and size of pulmonary arteries. Results Varying rates of positive reactivity to Dvl-2 and Daam-1 were confirmed in all cellular components of pulmonary arteries, namely, endothelial cells, myofibroblasts, and medial smooth muscle cells. In contrast, none of these components was reactive to Wnt-11. No specific expression patterns were observed for endothelial cells or myofibroblasts under any experimental conditions. However, marked expression of Dvl-2 and Daam-1 was confirmed in smooth muscle cells. In addition, Dvl-2 was depleted while Daam-1 expression was elevated in IPAH, in contrast with specimens from associated pulmonary arterial hypertension cases and controls. Conclusions High Daam-1 expression may upregulate the Wnt/PCP pathway and cause IPAH.
Collapse
Affiliation(s)
- Shun Yanai
- Department of Pediatrics, Toho University School of Medicine, 6-11-1 Omori-nishi, Ota-ku, Tokyo, 143-8541, Japan
| | - Megumi Wakayama
- Department of Surgical Pathology, Toho University School of Medicine, 6-11-1 Omori-nishi, Ota-ku, Tokyo, 143-8541, Japan.
| | - Haruo Nakayama
- Department of Neurosurgery, Toho University Ohashi Medical Center, 2-17-6 Ohashi, Meguro-ku, Tokyo, 153-8515, Japan
| | - Minoru Shinozaki
- Department of Surgical Pathology, Toho University School of Medicine, 6-11-1 Omori-nishi, Ota-ku, Tokyo, 143-8541, Japan
| | - Hisayuki Tsukuma
- Toho University School of Medicine, 5-21-16 Omori-nishi, Ota-ku, Tokyo, 143-8540, Japan
| | - Naobumi Tochigi
- Department of Surgical Pathology, Toho University School of Medicine, 6-11-1 Omori-nishi, Ota-ku, Tokyo, 143-8541, Japan
| | - Tetsuo Nemoto
- Department of Surgical Pathology, Toho University School of Medicine, 6-11-1 Omori-nishi, Ota-ku, Tokyo, 143-8541, Japan
| | - Tsutomu Saji
- Advanced and Integrated Cardiovascular Research Course in the Young and Adolescence, Toho University School of Medicine, 5-21-16 Omori-nishi, Ota-ku, Tokyo, 143-8540, Japan
| | - Kazutoshi Shibuya
- Department of Surgical Pathology, Toho University School of Medicine, 6-11-1 Omori-nishi, Ota-ku, Tokyo, 143-8541, Japan
| |
Collapse
|
30
|
Absence of the Adenosine A2A Receptor Confers Pulmonary Arterial Hypertension Through RhoA/ROCK Signaling Pathway in Mice. J Cardiovasc Pharmacol 2016; 66:569-75. [PMID: 26647014 DOI: 10.1097/fjc.0000000000000305] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Numerous evidence suggests that RhoA/Rho kinase (ROCK) signaling pathway plays an important role in the pathogenesis of pulmonary arterial hypertension (PAH), but little is known about its effects on the development of PAH in mice with absence of the adenosine A2A receptor (A2AR). Eight A2AR knockout (KO) and 8 wild-type mice were used. Morphometric analysis of pulmonary arterioles included right ventricle/left ventricle plus ventricular septum (Fulton index), vessel wall thickness/total vascular diameter (WT%), and vessel wall area/total vascular area (WA%). The expression of RhoA and ROCK1 mRNA was determined by real-time polymerase chain reaction. The expression of RhoA, ROCK1, and phosphorylation of myosin phosphatase target subunit 1 proteins in pulmonary tissue was tested by Western blot. The position of ROCK1 protein was evaluated by immunohistochemistry. Compared with wild-type mice, A2AR KO mice displayed (1) increased Fulton index, WT%, and WA% (P < 0.01); (2) increased mRNA expression of RhoA and ROCK1 (each P < 0.05); (3) increased protein expression of RhoA, ROCK1, and phosphorylation of myosin phosphatase target subunit 1 (each P < 0.01); (4) increased location of ROCK1 protein in endothelial and smooth muscle cells of pulmonary artery, bronchial, and alveolar epithelial cells. Activation of RhoA/ROCK signaling pathway may cause pulmonary vascular constriction, pulmonary artery remodeling, and PAH in adenosine A2A receptor KO mice.
Collapse
|
31
|
Awad KS, West JD, de Jesus Perez V, MacLean M. Novel signaling pathways in pulmonary arterial hypertension (2015 Grover Conference Series). Pulm Circ 2016; 6:285-94. [PMID: 27683605 PMCID: PMC5019081 DOI: 10.1086/688034] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 06/06/2016] [Indexed: 12/27/2022] Open
Abstract
The proliferative endothelial and smooth muscle cell phenotype, inflammation, and pulmonary vascular remodeling are prominent features of pulmonary arterial hypertension (PAH). Mutations in bone morphogenetic protein type 2 receptor (BMPR2) have been identified as the most common genetic cause of PAH and females with BMPR2 mutations are 2.5 times as likely to develop heritable forms of PAH than males. Higher levels of estrogen have also been observed in males with PAH, implicating sex hormones in PAH pathogenesis. Recently, the estrogen metabolite 16α-OHE1 (hydroxyestrone) was implicated in the regulation of miR29, a microRNA involved in modulating energy metabolism. In females, decreased miR96 enhances serotonin's effect by upregulating the 5-hydroxytryptamine 1B (5HT1B) receptor. Because PAH is characterized as a quasi-malignant disease, likely due to BMPR2 loss of function, altered signaling pathways that sustain this cancer-like phenotype are being explored. Extracellular signal-regulated kinases 1 and 2 and p38 mitogen-activated protein kinases (MAPKs) play a critical role in proliferation and cell motility, and dysregulated MAPK signaling is observed in various experimental models of PAH. Wnt signaling pathways preserve pulmonary vascular homeostasis, and dysregulation of this pathway could contribute to limited vascular regeneration in response to injury. In this review, we take a closer look at sex, sex hormones, and the interplay between sex hormones and microRNA regulation. We also focus on MAPK and Wnt signaling pathways in the emergence of a proproliferative, antiapoptotic endothelial phenotype, which then orchestrates an angioproliferative process of vascular remodeling, with the hope of developing novel therapies that could reverse the phenotype.
Collapse
Affiliation(s)
- Keytam S. Awad
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - James D. West
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - Margaret MacLean
- Research Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
32
|
Identifying microRNAs targeting Wnt/β-catenin pathway in end-stage idiopathic pulmonary arterial hypertension. J Mol Med (Berl) 2016; 94:875-85. [PMID: 27188753 DOI: 10.1007/s00109-016-1426-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 04/28/2016] [Accepted: 05/10/2016] [Indexed: 12/17/2022]
Abstract
UNLABELLED MicroRNAs (miRNAs) play important roles in the pathogenesis of pulmonary arterial hypertension (PAH). However, the pathways targeted by miRNAs in PAH have not been systematically investigated. We aim to identify dysregulated miRNAs for patients with idiopathic PAH (IPAH). miRNA profiling was performed on lung tissue total RNA from eight IPAH patients and eight control subjects. Real-time quantitative RT-PCR (qRT-PCR) was used for validation of miRNA and mRNA expression levels in 14 IPAH patients and 14 control subjects. Pathway enrichment analysis showed that Wnt/β-catenin signaling is among the top PAH-related pathways enriched in target genes of dysregulated miRNAs. We confirmed the significant increased expression levels of five miRNAs (let-7a-5p, miR-26b-5p, miR-27b-3p, miR-199a-3p and miR-656) targeting major PAH-related pathways. Moreover, qRT-PCR validation of Wnt/β-catenin pathway activation indicated multiple genes including receptors (FZD4, FZD5), core molecule (CTNNB1), and downstream targets (CCND1, VEGFA, and AXIN2) were significantly upregulated. The expression level of miR-199b-5p was positively correlated with patients' hemodynamics (PVR: r = 0.522, p = 0.038) and pulmonary vascular remodeling (muscularization: r = 0.540, p = 0.021). We confirmed overexpression of miR-199b-5p in hypoxic pulmonary arterial endothelial cells that negatively regulates GSK3B expression. In summary, miRNAs influence the pathogenesis of PAH by regulating major PAH-related pathways including Wnt/β-catenin in end-stage IPAH. KEY MESSAGE It is the first miRNA profiling study in lung tissue from end-stage idiopathic PAH. We identified dysregulated miRNAs and major pathways (e.g., Wnt signaling) in IPAH. Levels of miRNA expression were correlated with hemodynamics and pathological changes. We observed aberrant expression of target genes in the Wnt/β-catenin pathway. miRNAs influence the pathogenesis of PAH by regulating major PAH-related pathways.
Collapse
|
33
|
Hoffmann J, Wilhelm J, Olschewski A, Kwapiszewska G. Microarray analysis in pulmonary hypertension. Eur Respir J 2016; 48:229-41. [PMID: 27076594 PMCID: PMC5009873 DOI: 10.1183/13993003.02030-2015] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 02/15/2016] [Indexed: 12/21/2022]
Abstract
Microarrays are a powerful and effective tool that allows the detection of genome-wide gene expression differences between controls and disease conditions. They have been broadly applied to investigate the pathobiology of diverse forms of pulmonary hypertension, namely group 1, including patients with idiopathic pulmonary arterial hypertension, and group 3, including pulmonary hypertension associated with chronic lung diseases such as chronic obstructive pulmonary disease and idiopathic pulmonary fibrosis. To date, numerous human microarray studies have been conducted to analyse global (lung homogenate samples), compartment-specific (laser capture microdissection), cell type-specific (isolated primary cells) and circulating cell (peripheral blood) expression profiles. Combined, they provide important information on development, progression and the end-stage disease. In the future, system biology approaches, expression of noncoding RNAs that regulate coding RNAs, and direct comparison between animal models and human disease might be of importance. Comprehensive overview of compartment-specific microarray studies of material from pulmonary hypertension patientshttp://ow.ly/YEFO2
Collapse
Affiliation(s)
- Julia Hoffmann
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Jochen Wilhelm
- Dept of Internal Medicine, Justus-Liebig-University Giessen, Universities of Giessen and Marburg Lung Center, German Center for Lung Research, Giessen, Germany
| | - Andrea Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria Dept of Experimental Anaesthesiology, Medical University of Graz, Graz, Austria
| | - Grazyna Kwapiszewska
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria Dept of Experimental Anaesthesiology, Medical University of Graz, Graz, Austria
| |
Collapse
|
34
|
Mechanisms of Vessel Pruning and Regression. Dev Cell 2015; 34:5-17. [PMID: 26151903 DOI: 10.1016/j.devcel.2015.06.004] [Citation(s) in RCA: 198] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 05/26/2015] [Accepted: 06/03/2015] [Indexed: 01/27/2023]
Abstract
The field of angiogenesis research has primarily focused on the mechanisms of sprouting angiogenesis. Yet vascular networks formed by vessel sprouting subsequently undergo extensive vascular remodeling to form a functional and mature vasculature. This "trimming" includes distinct processes of vascular pruning, the regression of selected vascular branches. In some situations complete vascular networks may undergo physiological regression. Vessel regression is an understudied yet emerging field of research. This review summarizes the state-of-the-art of vessel pruning and regression with a focus on the cellular processes and the molecular regulators of vessel maintenance and regression.
Collapse
|
35
|
Takayasu H, Masumoto K, Hagiwara K, Sasaki T, Ono K, Jimbo T, Uesugi T, Gotoh C, Urita Y, Shinkai T, Tanaka H. Increased pulmonary RhoA expression in the nitrofen-induced congenital diaphragmatic hernia rat model. J Pediatr Surg 2015; 50:1467-71. [PMID: 25783350 DOI: 10.1016/j.jpedsurg.2015.02.063] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Revised: 02/05/2015] [Accepted: 02/24/2015] [Indexed: 10/24/2022]
Abstract
PURPOSE Persistent pulmonary hypertension remains a major cause of mortality and morbidity in cases of congenital diaphragmatic hernia (CDH). Recently, RhoA/Rho-kinase-mediated vasoconstriction has been reported to be important in the pathogenesis of pulmonary hypertension (PH). Several recent reports have described that fasudil, a potent Rho-kinase inhibitor and vasodilator, could represent a potential therapeutic option for PH. We designed this study to investigate the hypothesis that the expression level of RhoA is increased in the nitrofen-induced CDH rat model. The expression level of Wnt11, an activator of RhoA, was also evaluated. METHODS Pregnant rats were treated with or without nitrofen on gestational day 9 (D9). Fetuses were sacrificed on D17, D19 and D21 and were divided into control and CDH groups. Quantitative real-time polymerase chain reaction was performed to determine the pulmonary gene expression levels of both Wnt11 and RhoA. An immunofluorescence study was also performed to evaluate the expression and localization of RhoA. RESULTS The relative mRNA expression levels of pulmonary Wnt11 and RhoA on D21 were significantly increased in the CDH group compared with the control group (p=0.016 and p=0.008, respectively). The immunofluorescence study confirmed the overexpression of RhoA in the pulmonary vessels of CDH rats on D21. CONCLUSIONS Our results provide evidence that the RhoA/Rho-kinase-mediated pathway is involved in the pathogenesis of PH in the nitrofen-induced CDH rat model. Our data also suggest that the fasudil, a Rho-kinase inhibitor, could represent a therapeutic option for the treatment of PH in CDH.
Collapse
Affiliation(s)
- Hajime Takayasu
- Department of Pediatric Surgery, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennoudai, Tsukuba, Ibaraki 305-8575, Japan.
| | - Kouji Masumoto
- Department of Pediatric Surgery, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennoudai, Tsukuba, Ibaraki 305-8575, Japan
| | - Koki Hagiwara
- Department of Pediatric Surgery, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennoudai, Tsukuba, Ibaraki 305-8575, Japan
| | - Takato Sasaki
- Department of Pediatric Surgery, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennoudai, Tsukuba, Ibaraki 305-8575, Japan
| | - Kentaro Ono
- Department of Pediatric Surgery, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennoudai, Tsukuba, Ibaraki 305-8575, Japan
| | - Takahiro Jimbo
- Department of Pediatric Surgery, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennoudai, Tsukuba, Ibaraki 305-8575, Japan
| | - Toru Uesugi
- Department of Pediatric Surgery, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennoudai, Tsukuba, Ibaraki 305-8575, Japan
| | - Chikashi Gotoh
- Department of Pediatric Surgery, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennoudai, Tsukuba, Ibaraki 305-8575, Japan
| | - Yasuhisa Urita
- Department of Pediatric Surgery, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennoudai, Tsukuba, Ibaraki 305-8575, Japan
| | - Toko Shinkai
- Department of Pediatric Surgery, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennoudai, Tsukuba, Ibaraki 305-8575, Japan
| | - Hideaki Tanaka
- Department of Pediatric Surgery, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennoudai, Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
36
|
Li C, Bellusci S, Borok Z, Minoo P. Non-canonical WNT signalling in the lung. J Biochem 2015; 158:355-65. [PMID: 26261051 DOI: 10.1093/jb/mvv081] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 07/26/2015] [Indexed: 12/23/2022] Open
Abstract
The role of WNT signalling in metazoan organogenesis has been a topic of widespread interest. In the lung, while the role of canonical WNT signalling has been examined in some detail by multiple studies, the non-canonical WNT signalling has received limited attention. Reliable evidence shows that this important signalling mechanism constitutes a major regulatory pathway in lung development. In addition, accumulating evidence has also shown that the non-canonical WNT pathway is critical for maintaining lung homeostasis and that aberrant activation of this pathway may underlie several debilitating lung diseases. Functional analyses have further revealed that the non-canonical WNT pathway regulates multiple cellular activities in the lung that are dependent on the specific cellular context. In most cell types, non-canonical WNT signalling regulates canonical WNT activity, which is also critical for many aspects of lung biology. This review will summarize what is currently known about the role of non-canonical WNT signalling in lung development, homeostasis and pathogenesis of disease.
Collapse
Affiliation(s)
- Changgong Li
- Department of Pediatrics, Division of Newborn Medicine, Los Angeles County+University of Southern California Medical Center and Children's Hospital Los Angeles, Keck School of Medicine of USC, Los Angeles, CA 90033, USA;
| | - Saverio Bellusci
- Excellence Cluster Cardio-Pulmonary System (ECCPS), D-35392 Giessen, Hessen, Germany; Member of the German Center for Lung Research, Department of Internal Medicine II, Universities of Giessen and Marburg Lung Center (UGMLC), D-35390 Giessen, Hessen, Germany; Developmental Biology and Regenerative Medicine Program, Saban Research Institute of Childrens Hospital Los Angeles and University of Southern California, Los Angeles, CA 90027, USA; and
| | - Zea Borok
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Will Rogers Institute Pulmonary Research Center, Keck School of Medicine of USC, Los Angeles, CA 90033, USA
| | - Parviz Minoo
- Department of Pediatrics, Division of Newborn Medicine, Los Angeles County+University of Southern California Medical Center and Children's Hospital Los Angeles, Keck School of Medicine of USC, Los Angeles, CA 90033, USA
| |
Collapse
|
37
|
Hu J, Xu Q, McTiernan C, Lai YC, Osei-Hwedieh D, Gladwin M. Novel Targets of Drug Treatment for Pulmonary Hypertension. Am J Cardiovasc Drugs 2015; 15:225-34. [PMID: 26016608 DOI: 10.1007/s40256-015-0125-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Biomedical advances over the last decade have identified the central role of proliferative pulmonary arterial smooth muscle cells (PASMCs) in the development of pulmonary hypertension (PH). Furthermore, promoters of proliferation and apoptosis resistance in PASMCs and endothelial cells, such as aberrant signal pathways involving growth factors, G protein-coupled receptors, kinases, and microRNAs, have also been described. As a result of these discoveries, PH is currently divided into subgroups based on the underlying pathology, which allows focused and targeted treatment of the condition. The defining features of PH, which subsequently lead to vascular wall remodeling, are dysregulated proliferation of PASMCs, local inflammation, and apoptosis-resistant endothelial cells. Efforts to assess the relative contributions of these factors have generated several promising targets. This review discusses recent novel targets of therapies for PH that have been developed as a result of these advances, which are now in pre-clinical and clinical trials (e.g., imatinib [phase III]; nilotinib, AT-877ER, rituximab, tacrolimus, paroxetine, sertraline, fluoxetine, bardoxolone methyl [phase II]; and sorafenib, FK506, aviptadil, endothelial progenitor cells (EPCs) [phase I]). While substantial progress has been made in recent years in targeting key molecular pathways, PH still remains without a cure, and these novel therapies provide an important conceptual framework of categorizing patients on the basis of molecular phenotype(s) for effective treatment of the disease.
Collapse
|
38
|
Jin Y, Wang W, Chai S, Liu J, Yang T, Wang J. Wnt5a attenuates hypoxia-induced pulmonary arteriolar remodeling and right ventricular hypertrophy in mice. Exp Biol Med (Maywood) 2015; 240:1742-51. [PMID: 25956683 DOI: 10.1177/1535370215584889] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 02/24/2015] [Indexed: 12/14/2022] Open
Abstract
Hypoxic pulmonary hypertension (HPH), which is characterized by pulmonary arteriolar remodeling and right ventricular hypertrophy, is still a life-threatening disease with the current treatment strategies. The underlying molecular mechanisms of HPH remain unclear. Our previously published study showed that Wnt5a, one of the ligands in the Wnt family, was critically involved in the inhibition of hypoxia-induced pulmonary arterial smooth muscle cell proliferation by downregulation of β-catenin/cyclin D1 in vitro. In this study, we investigated the possible functions and mechanisms of Wnt5a in HPH in vivo. Recombinant mouse Wnt5a (rmWnt5a) or phosphate buffered saline (PBS) was administered to male C57/BL6 mice weekly from the first day to the end of the two or four weeks after exposed to hypoxia (10% O2). Hypoxia-induced pulmonary hypertension was associated with a marked increase in β-catenin/cyclin D1 expression in lungs. Right ventricular systolic pressure and right ventricular hypertrophy index were reduced in animals treated with rmWnt5a compared with PBS. Histology showed less pulmonary vascular remodeling and right ventricular hypertrophy in the group treated with rmWnt5a than with PBS. Treatment with rmWnt5a resulted in a concomitant reduction in β-catenin/cyclin D1 levels in lungs. These data demonstrate that Wnt5a exerts its beneficial effects on HPH by regulating pulmonary vascular remodeling and right ventricular hypertrophy in a manner that is associated with reduction in β-catenin/cyclin D1 signaling. A therapy targeting the β-catenin/cyclin D1 signaling pathway might be a potential strategy for HPH treatment.
Collapse
Affiliation(s)
- Yuling Jin
- Department of Physiology, Capital Medical University, Beijing 100069, P.R. China
| | - Wang Wang
- Department of Physiology, Capital Medical University, Beijing 100069, P.R. China
| | - Sanbao Chai
- Department of Physiology, Capital Medical University, Beijing 100069, P.R. China
| | - Jie Liu
- Department of Physiology, Capital Medical University, Beijing 100069, P.R. China
| | - Ting Yang
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100016, P.R. China
| | - Jun Wang
- Department of Physiology, Capital Medical University, Beijing 100069, P.R. China
| |
Collapse
|
39
|
Veith C, Zakrzewicz D, Dahal BK, Bálint Z, Murmann K, Wygrecka M, Seeger W, Schermuly RT, Weissmann N, Kwapiszewska G. Hypoxia- or PDGF-BB-dependent paxillin tyrosine phosphorylation in pulmonary hypertension is reversed by HIF-1α depletion or imatinib treatment. Thromb Haemost 2014; 112:1288-303. [PMID: 25231004 DOI: 10.1160/th13-12-1031] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 07/10/2014] [Indexed: 12/27/2022]
Abstract
Chronic exposure to hypoxia induces a pronounced remodelling of the pulmonary vasculature leading to pulmonary hypertension (PH). The remodelling process also entails increased proliferation and decreased apoptosis of pulmonary arterial smooth muscle cells (PASMC), processes regulated by the cytoskeletal protein paxillin. In this study, we aimed to examine the molecular mechanisms leading to deregulation of paxillin in PH. We detected a time-dependent increase in paxillin tyrosine 31 (Y31) and 118 (Y118) phosphorylation following hypoxic exposure (1 % O2) or platelet-derived growth factor (PDGF)-BB stimulation of primary human PASMC. In addition, both, hypoxia- and PDGF-BB increased the nuclear localisation of phospho-paxillin Y31 as indicated by immunofluorescence staining in human PASMC. Elevated paxillin tyrosine phosphorylation in human PASMC was attenuated by hypoxia-inducible factor (HIF)-1α depletion or by treatment with the PDGF-BB receptor antagonist, imatinib. Moreover, we observed elevated paxillin Y31 and Y118 phosphorylation in the pulmonary vasculature of chronic hypoxic mice (21 days, 10 % O2) which was reversible by imatinib-treatment. PDGF-BB-dependent PASMC proliferation was regulated via the paxillin-Erk1/2-cyclin D1 pathway. In conclusion, we suggest paxillin up-regulation and phosphorylation as an important mechanism of vascular remodelling underlying pulmonary hypertension.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - N Weissmann
- Norbert Weissmann, Excellence Cluster Cardio-Pulmonary System (ECCPS), Justus-Liebig-University Giessen, Aulweg 130, 35392 Giessen, Germany, Tel.: +49 641 99 46000, Fax: +49 641 99 42419, E-mail:
| | | |
Collapse
|
40
|
Hoffmann J, Wilhelm J, Marsh LM, Ghanim B, Klepetko W, Kovacs G, Olschewski H, Olschewski A, Kwapiszewska G. Distinct differences in gene expression patterns in pulmonary arteries of patients with chronic obstructive pulmonary disease and idiopathic pulmonary fibrosis with pulmonary hypertension. Am J Respir Crit Care Med 2014; 190:98-111. [PMID: 24918967 DOI: 10.1164/rccm.201401-0037oc] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
RATIONALE The development of pulmonary hypertension (PH) in patients with idiopathic pulmonary fibrosis (IPF) or chronic obstructive pulmonary disease (COPD) is associated with increased morbidity. OBJECTIVES To elucidate whether vascular remodeling in a well-characterized PH-COPD and PH-IPF patient cohort results from similar or divergent molecular changes. METHODS Vascular remodeling of donor, PH-COPD, and PH-IPF pulmonary arteries was assessed. Laser capture microdissected pulmonary artery profiles in combination with whole genome microarrays were performed. MEASUREMENTS AND MAIN RESULTS Pulmonary arteries from patients with COPD and IPF with PH exhibited remodeling of vascular layers and reduction of lumen area. Pathway analyses comparing normalized gene expression profiles obtained from patients with PH-IPF or PH-COPD revealed the retinol and extracellular matrix (ECM) receptor interaction to be the most perturbed processes. Within the ECM-receptor pathway, differential regulation of 5 out of the top 10 results (collagen, type III, α-1; tenascin C; collagen, type VI, α-3; thrombospondin 2; and von Willebrand factor) were verified by real-time polymerase chain reaction and immunohistochemical staining. CONCLUSIONS Despite clinical and histologic vascular remodeling in all patients with PH-COPD and PH-IPF, differential gene expression pattern was present in pulmonary artery profiles. Several genes involved in retinol metabolism and ECM receptor interaction enable discrimination of vascular remodeling in PH-IPF or PH-COPD. This suggests that pulmonary arterial remodeling in PH-COPD and PH-IPF is caused by different molecular mechanisms and may require specific therapeutic options.
Collapse
Affiliation(s)
- Julia Hoffmann
- 1 Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
West JD, Austin ED, Gaskill C, Marriott S, Baskir R, Bilousova G, Jean JC, Hemnes AR, Menon S, Bloodworth NC, Fessel JP, Kropski JA, Irwin D, Ware LB, Wheeler L, Hong CC, Meyrick B, Loyd JE, Bowman AB, Ess KC, Klemm DJ, Young PP, Merryman WD, Kotton D, Majka SM. Identification of a common Wnt-associated genetic signature across multiple cell types in pulmonary arterial hypertension. Am J Physiol Cell Physiol 2014; 307:C415-30. [PMID: 24871858 PMCID: PMC4154073 DOI: 10.1152/ajpcell.00057.2014] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 05/23/2014] [Indexed: 12/24/2022]
Abstract
Understanding differences in gene expression that increase risk for pulmonary arterial hypertension (PAH) is essential to understanding the molecular basis for disease. Previous studies on patient samples were limited by end-stage disease effects or by use of nonadherent cells, which are not ideal to model vascular cells in vivo. These studies addressed the hypothesis that pathological processes associated with PAH may be identified via a genetic signature common across multiple cell types. Expression array experiments were initially conducted to analyze cell types at different stages of vascular differentiation (mesenchymal stromal and endothelial) derived from PAH patient-specific induced pluripotent stem (iPS) cells. Molecular pathways that were altered in the PAH cell lines were then compared with those in fibroblasts from 21 patients, including those with idiopathic and heritable PAH. Wnt was identified as a target pathway and was validated in vitro using primary patient mesenchymal and endothelial cells. Taken together, our data suggest that the molecular lesions that cause PAH are present in all cell types evaluated, regardless of origin, and that stimulation of the Wnt signaling pathway was a common molecular defect in both heritable and idiopathic PAH.
Collapse
Affiliation(s)
- James D West
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee; Vanderbilt Vascular Biology Center, Nashville, Tennessee
| | - Eric D Austin
- Department of Pediatrics, Vanderbilt University, Nashville, Tennessee
| | - Christa Gaskill
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee
| | - Shennea Marriott
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee
| | - Rubin Baskir
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
| | - Ganna Bilousova
- Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado, Aurora, Colorado
| | | | - Anna R Hemnes
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee; Vanderbilt Vascular Biology Center, Nashville, Tennessee
| | - Swapna Menon
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee
| | | | - Joshua P Fessel
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee; Vanderbilt Vascular Biology Center, Nashville, Tennessee
| | - Johnathan A Kropski
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee
| | - David Irwin
- Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado, Aurora, Colorado
| | - Lorraine B Ware
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee; Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee
| | - Lisa Wheeler
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee
| | - Charles C Hong
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee; Veterans Administration Hospital, Nashville, Tennessee
| | - Barbara Meyrick
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee; Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee
| | - James E Loyd
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee
| | - Aaron B Bowman
- Department of Neurology, Vanderbilt Brain Institute, Nashville, Tennessee; Vanderbilt Center for Stem Cell Biology, Nashville, Tennessee
| | - Kevin C Ess
- Department of Pediatrics, Vanderbilt University, Nashville, Tennessee; Department of Neurology, Vanderbilt Brain Institute, Nashville, Tennessee; Vanderbilt Center for Stem Cell Biology, Nashville, Tennessee
| | - Dwight J Klemm
- Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado, Aurora, Colorado
| | - Pampee P Young
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee; Vanderbilt Center for Stem Cell Biology, Nashville, Tennessee
| | - W David Merryman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | | | - Susan M Majka
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee; Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee; Vanderbilt Center for Stem Cell Biology, Nashville, Tennessee; Vanderbilt Vascular Biology Center, Nashville, Tennessee; Pulmonary Vascular Research Institute, Kochi, and AnalyzeDat Consulting Services, Kerala, India; and
| |
Collapse
|
42
|
Guan S, Wang Z, Xin F, Xin H. Wnt5a is associated with the differentiation of bone marrow mesenchymal stem cells in vascular calcification by connecting with different receptors. Mol Med Rep 2014; 10:1985-91. [PMID: 25109262 DOI: 10.3892/mmr.2014.2449] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Accepted: 05/23/2014] [Indexed: 11/06/2022] Open
Abstract
Vascular calcification significantly affects the health of the elderly. Increasing evidence proved that vascular calcification is an actively regulated osteogenic process. The osteochondrocytic differentiation of mesenchymal stem cells (MSCs) is a significant step of osteogenic processes. The Wnt pathways has been identified as contributing to the regulation of osteogenic mineralization during development and disease. However, it remains unknown whether these MSCs in the vascular calcification differentiate into normal vascular smooth muscle cells (VSMCs) in vivo in order to treat damaged vascular tissue or into calcified VSMCs to aggravate calcification correlated to the Wnt pathways. Thus, it is necessary to analyze the mechanisms of MSC differentiation in detail. In the present study a cell‑cell co‑culturing in vitro system was used to observe MSCs that directly interact with normal or calcified VSMCs during calcification and to investigate the gene expression of the Wnt pathways during the process. Direct co‑cultures were established by seeding two different cell types, VSMCs or calcified VSMCs, or a mixture of both at ratios of 5,000:5,000 cells/1.7 cm2 onto either gelatin‑coated 1.7‑cm2 chamber slides for immunohistochemical analysis or gelatin‑coated 75‑cm2 tissue culture flasks for protein or RNA isolation. Osteoblastic differentiation was evaluated by examining the cell morphology and assessing the activity of alkaline phosphatase in the cell lysates by alkaline phosphatase staining. Additionally, the mRNA expression levels of the genes encoding for proteins involved in the Wnt signaling proteins, Wnt5A, LRP6, Ror2, c‑Jun‑N‑terminal kinase and β‑catenin, were assessed in each group. The present study demonstrated that Wnts are expressed in the progress of differentiation of MSCs during calcification. MSCs can differentiate into different cell phenotypes when there is direct cell‑cell contact with VSMCs or calcified VSMCs, and the Wnt5a/Ror2 signaling pathway may be associated with the determination of differentiation of MSCs in this process.
Collapse
Affiliation(s)
- Siming Guan
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Zhimin Wang
- Department of Neurology, The First People's Hospital of Taizhou, Taizhou, Zhejiang 318020, P.R. China
| | - Fang Xin
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Huaping Xin
- Department of Neurology, The First People's Hospital of Taizhou, Taizhou, Zhejiang 318020, P.R. China
| |
Collapse
|
43
|
Zaina S, Heyn H, Carmona FJ, Varol N, Sayols S, Condom E, Ramírez-Ruz J, Gomez A, Gonçalves I, Moran S, Esteller M. DNA methylation map of human atherosclerosis. ACTA ACUST UNITED AC 2014; 7:692-700. [PMID: 25091541 DOI: 10.1161/circgenetics.113.000441] [Citation(s) in RCA: 168] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Epigenetic alterations may contribute to the development of atherosclerosis. In particular, DNA methylation, a reversible and highly regulated DNA modification, could influence disease onset and progression because it functions as an effector for environmental influences, including diet and lifestyle, both of which are risk factors for cardiovascular diseases. METHODS AND RESULTS To address the role of DNA methylation changes in atherosclerosis, we compared a donor-matched healthy and atherosclerotic human aorta sample using whole-genome shotgun bisulfite sequencing. We observed that the atherosclerotic portion of the aorta was hypermethylated across many genomic loci in comparison with the matched healthy counterpart. Furthermore, we defined specific loci of differential DNA methylation using a set of donor-matched aortic samples and a high-density (>450 000 CpG sites) DNA methylation microarray. The functional importance in the disease was corroborated by crossing the DNA methylation signature with the corresponding expression data of the same samples. Among the differentially methylated CpGs associated with atherosclerosis onset, we identified genes participating in endothelial and smooth muscle functions. These findings provide new clues toward a better understanding of the molecular mechanisms of atherosclerosis. CONCLUSIONS Our data identify an atherosclerosis-specific DNA methylation profile that highlights the contribution of different genes and pathways to the disorder. Interestingly, the observed gain of DNA methylation in the atherosclerotic lesions justifies efforts to develop DNA demethylating agents for therapeutic benefit.
Collapse
Affiliation(s)
- Silvio Zaina
- From the Division of Health Sciences, Department of Medical Sciences, University of Guanajuato, León, Guanajuato, Mexico (S.Z.); Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Catalonia, Spain (H.H., F.J.C., N.V., S.S., A.G., S.M., M.E.); Department of Pathology, Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL) (E.C.), Barcelona, Spain; Department of Pathology and Experimental Therapeutics (E.C.), Department of Anatomy and Pathology, Hospital Clinic (J.R.-R.), Department of Physiological Sciences II, School of Medicine (M.E.), University of Barcelona, Barcelona, Catalonia, Spain; Experimental Cardiovascular Research, Lund University, Malmö, Sweden (I.G.); and Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain (M.E.).
| | - Holger Heyn
- From the Division of Health Sciences, Department of Medical Sciences, University of Guanajuato, León, Guanajuato, Mexico (S.Z.); Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Catalonia, Spain (H.H., F.J.C., N.V., S.S., A.G., S.M., M.E.); Department of Pathology, Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL) (E.C.), Barcelona, Spain; Department of Pathology and Experimental Therapeutics (E.C.), Department of Anatomy and Pathology, Hospital Clinic (J.R.-R.), Department of Physiological Sciences II, School of Medicine (M.E.), University of Barcelona, Barcelona, Catalonia, Spain; Experimental Cardiovascular Research, Lund University, Malmö, Sweden (I.G.); and Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain (M.E.)
| | - F Javier Carmona
- From the Division of Health Sciences, Department of Medical Sciences, University of Guanajuato, León, Guanajuato, Mexico (S.Z.); Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Catalonia, Spain (H.H., F.J.C., N.V., S.S., A.G., S.M., M.E.); Department of Pathology, Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL) (E.C.), Barcelona, Spain; Department of Pathology and Experimental Therapeutics (E.C.), Department of Anatomy and Pathology, Hospital Clinic (J.R.-R.), Department of Physiological Sciences II, School of Medicine (M.E.), University of Barcelona, Barcelona, Catalonia, Spain; Experimental Cardiovascular Research, Lund University, Malmö, Sweden (I.G.); and Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain (M.E.)
| | - Nuray Varol
- From the Division of Health Sciences, Department of Medical Sciences, University of Guanajuato, León, Guanajuato, Mexico (S.Z.); Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Catalonia, Spain (H.H., F.J.C., N.V., S.S., A.G., S.M., M.E.); Department of Pathology, Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL) (E.C.), Barcelona, Spain; Department of Pathology and Experimental Therapeutics (E.C.), Department of Anatomy and Pathology, Hospital Clinic (J.R.-R.), Department of Physiological Sciences II, School of Medicine (M.E.), University of Barcelona, Barcelona, Catalonia, Spain; Experimental Cardiovascular Research, Lund University, Malmö, Sweden (I.G.); and Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain (M.E.)
| | - Sergi Sayols
- From the Division of Health Sciences, Department of Medical Sciences, University of Guanajuato, León, Guanajuato, Mexico (S.Z.); Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Catalonia, Spain (H.H., F.J.C., N.V., S.S., A.G., S.M., M.E.); Department of Pathology, Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL) (E.C.), Barcelona, Spain; Department of Pathology and Experimental Therapeutics (E.C.), Department of Anatomy and Pathology, Hospital Clinic (J.R.-R.), Department of Physiological Sciences II, School of Medicine (M.E.), University of Barcelona, Barcelona, Catalonia, Spain; Experimental Cardiovascular Research, Lund University, Malmö, Sweden (I.G.); and Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain (M.E.)
| | - Enric Condom
- From the Division of Health Sciences, Department of Medical Sciences, University of Guanajuato, León, Guanajuato, Mexico (S.Z.); Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Catalonia, Spain (H.H., F.J.C., N.V., S.S., A.G., S.M., M.E.); Department of Pathology, Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL) (E.C.), Barcelona, Spain; Department of Pathology and Experimental Therapeutics (E.C.), Department of Anatomy and Pathology, Hospital Clinic (J.R.-R.), Department of Physiological Sciences II, School of Medicine (M.E.), University of Barcelona, Barcelona, Catalonia, Spain; Experimental Cardiovascular Research, Lund University, Malmö, Sweden (I.G.); and Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain (M.E.)
| | - José Ramírez-Ruz
- From the Division of Health Sciences, Department of Medical Sciences, University of Guanajuato, León, Guanajuato, Mexico (S.Z.); Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Catalonia, Spain (H.H., F.J.C., N.V., S.S., A.G., S.M., M.E.); Department of Pathology, Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL) (E.C.), Barcelona, Spain; Department of Pathology and Experimental Therapeutics (E.C.), Department of Anatomy and Pathology, Hospital Clinic (J.R.-R.), Department of Physiological Sciences II, School of Medicine (M.E.), University of Barcelona, Barcelona, Catalonia, Spain; Experimental Cardiovascular Research, Lund University, Malmö, Sweden (I.G.); and Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain (M.E.)
| | - Antonio Gomez
- From the Division of Health Sciences, Department of Medical Sciences, University of Guanajuato, León, Guanajuato, Mexico (S.Z.); Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Catalonia, Spain (H.H., F.J.C., N.V., S.S., A.G., S.M., M.E.); Department of Pathology, Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL) (E.C.), Barcelona, Spain; Department of Pathology and Experimental Therapeutics (E.C.), Department of Anatomy and Pathology, Hospital Clinic (J.R.-R.), Department of Physiological Sciences II, School of Medicine (M.E.), University of Barcelona, Barcelona, Catalonia, Spain; Experimental Cardiovascular Research, Lund University, Malmö, Sweden (I.G.); and Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain (M.E.)
| | - Isabel Gonçalves
- From the Division of Health Sciences, Department of Medical Sciences, University of Guanajuato, León, Guanajuato, Mexico (S.Z.); Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Catalonia, Spain (H.H., F.J.C., N.V., S.S., A.G., S.M., M.E.); Department of Pathology, Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL) (E.C.), Barcelona, Spain; Department of Pathology and Experimental Therapeutics (E.C.), Department of Anatomy and Pathology, Hospital Clinic (J.R.-R.), Department of Physiological Sciences II, School of Medicine (M.E.), University of Barcelona, Barcelona, Catalonia, Spain; Experimental Cardiovascular Research, Lund University, Malmö, Sweden (I.G.); and Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain (M.E.)
| | - Sebastian Moran
- From the Division of Health Sciences, Department of Medical Sciences, University of Guanajuato, León, Guanajuato, Mexico (S.Z.); Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Catalonia, Spain (H.H., F.J.C., N.V., S.S., A.G., S.M., M.E.); Department of Pathology, Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL) (E.C.), Barcelona, Spain; Department of Pathology and Experimental Therapeutics (E.C.), Department of Anatomy and Pathology, Hospital Clinic (J.R.-R.), Department of Physiological Sciences II, School of Medicine (M.E.), University of Barcelona, Barcelona, Catalonia, Spain; Experimental Cardiovascular Research, Lund University, Malmö, Sweden (I.G.); and Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain (M.E.)
| | - Manel Esteller
- From the Division of Health Sciences, Department of Medical Sciences, University of Guanajuato, León, Guanajuato, Mexico (S.Z.); Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Catalonia, Spain (H.H., F.J.C., N.V., S.S., A.G., S.M., M.E.); Department of Pathology, Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL) (E.C.), Barcelona, Spain; Department of Pathology and Experimental Therapeutics (E.C.), Department of Anatomy and Pathology, Hospital Clinic (J.R.-R.), Department of Physiological Sciences II, School of Medicine (M.E.), University of Barcelona, Barcelona, Catalonia, Spain; Experimental Cardiovascular Research, Lund University, Malmö, Sweden (I.G.); and Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain (M.E.).
| |
Collapse
|
44
|
Cottrill KA, Chan SY. Investigating pulmonary arterial hypertension from "stem" to stern. Focus on "Identification of a common Wnt-associated genetic signature across multiple cell types in pulmonary arterial hypertension". Am J Physiol Cell Physiol 2014; 307:C413-4. [PMID: 25031019 DOI: 10.1152/ajpcell.00242.2014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Katherine A Cottrill
- Divisions of Cardiovascular and Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Stephen Y Chan
- Divisions of Cardiovascular and Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
45
|
de Jesus Perez V, Yuan K, Alastalo TP, Spiekerkoetter E, Rabinovitch M. Targeting the Wnt signaling pathways in pulmonary arterial hypertension. Drug Discov Today 2014; 19:1270-6. [PMID: 24955837 DOI: 10.1016/j.drudis.2014.06.014] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 06/16/2014] [Indexed: 10/25/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a life-threatening disorder that is associated with elevated pulmonary pressures and right heart failure resulting from progressive loss and thickening of small pulmonary arteries. Despite their ability to improve symptoms, current therapies fail to prevent disease progression, leaving lung transplantation as the only therapy in end-stage PAH. To overcome the limitations of current therapies, there is an active search for disease-modifying agents capable of altering the natural history of, and improving clinical outcomes in, PAH. The Wnt signaling pathways have emerged as attractive treatment targets in PAH given their role in the preservation of pulmonary vascular homeostasis and the recent development of Wnt-specific compounds and biological therapies capable of modulating pathway activity. In this review, we summarize the literature describing the role of Wnt signaling in the pulmonary circulation and discuss promising advances in the field of Wnt therapeutics that could lead to novel clinical therapies capable of preventing and/or reversing pulmonary vascular pathology in patients with this devastating disease.
Collapse
Affiliation(s)
- Vinicio de Jesus Perez
- Division of Pulmonary and Critical Care Medicine, Stanford University Medical Center, 300 Pasteur Drive Grant S140B, Stanford, CA 94305, USA.
| | - Ke Yuan
- Division of Pulmonary and Critical Care Medicine, Stanford University Medical Center, 300 Pasteur Drive Grant S140B, Stanford, CA 94305, USA
| | - Tero-Pekka Alastalo
- Children's Hospital Helsinki, Tukholmankatu 8, FI-00290 Helsinki, Finland; Biomedicum Helsinski, Tukholmankatu 8, FI-00290 Helsinki, Finland Finland
| | - Edda Spiekerkoetter
- Division of Pulmonary and Critical Care Medicine, Stanford University Medical Center, 300 Pasteur Drive Grant S140B, Stanford, CA 94305, USA
| | - Marlene Rabinovitch
- Pediatric Cardiology, Stanford University Medical Center, 300 Pasteur Drive Grant S140B, Stanford, CA 94305, USA
| |
Collapse
|
46
|
Abstract
Study of RNA and proteins in cells of both normal and diseased tissues is providing researchers with new knowledge of disease pathologies. While still in its early stages, high-throughput expression analysis is improving our understanding of the pathogenesis of pulmonary arterial hypertension (PAH). While many studies have used microarray and proteomic analyses as "hypothesis-generating" tools, the technologies also have potential to identify and quantify biomarkers of disease. To date, many of the published studies have examined gene expression profiles of tissue biopsies, others have utilized cells from peripheral blood. Microarray technology has been employed successfully in the investigation of a diverse array of human diseases. The potential of high-throughput expression analysis to improve our understanding of the pathogenesis of PAH is highlighted in this review. Proteomic studies of PAH and pulmonary vascular diseases in general have been little utilized thus far. To date, such studies are few and no consistent biomarker has emerged from studies of either plasma or blood cells from idiopathic pulmonary arterial hypertension (IPAH) patients. The studies of both lung tissue and lymphocytes are perhaps more revealing and suggest that changes in the cytoskeletal machinery may play a role in the pathogenesis of idiopathic pulmonary arterial hypertension. The oncology literature has demonstrated the utility of gene microarray analysis to predict important outcomes such as response to therapy and survival. It is likely that in the near future, gene microarrays and proteomic analyses will also be employed in a pharmacogenomics approach in PAH, helping to identify the most appropriate therapies for individual patients.
Collapse
Affiliation(s)
- Mark Geraci
- Pulmonary Sciences and Critical Care Medicine Division, Pulmonary Hypertension Center, University of Colorado Denver, Denver, Colorado, USA
| | | |
Collapse
|
47
|
Patel NM, Kawut SM, Jelic S, Arcasoy SM, Lederer DJ, Borczuk AC. Pulmonary arteriole gene expression signature in idiopathic pulmonary fibrosis. Eur Respir J 2013; 41:1324-30. [PMID: 23728404 PMCID: PMC4720265 DOI: 10.1183/09031936.00084112] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
A third of patients with idiopathic pulmonary fibrosis (IPF) develop pulmonary hypertension (PH-IPF), which is associated with increased mortality. Whether an altered gene expression profile in the pulmonary vasculature precedes the clinical onset of PH-IPF is unknown. We compared gene expression in the pulmonary vasculature of IPF patients with and without PH with controls. Pulmonary arterioles were isolated using laser capture microdissection from 16 IPF patients: eight with PH (PH-IPF) and eight with no PH (NPH-IPF), and seven controls. Probe was prepared from extracted RNA, and hybridised to Affymetrix Hu133 2.0 Plus genechips. Biometric Research Branch array tools and Ingenuity Pathway Analysis software were used for analysis of the microarray data. Univariate analysis revealed 255 genes that distinguished IPF arterioles from controls (p<0.001). Mediators of vascular smooth muscle and endothelial cell proliferation, Wnt signalling and apoptosis were differentially expressed in IPF arterioles. Unsupervised and supervised clustering analyses revealed similar gene expression in PH-IPF and NPH-IPF arterioles. The pulmonary arteriolar gene expression profile is similar in IPF patients with and without coexistent PH. Pathways involved in vascular proliferation and aberrant apoptosis, which may contribute to pulmonary vascular remodelling, are activated in IPF patients.
Collapse
Affiliation(s)
- Nina M. Patel
- Division of Pulmonary, Allergy and Critical Care Medicine, Columbia University, New York, NY, USA,Interstitial Lung Disease Program, New York Presbyterian Hospital, New York, NY, USA
| | - Steven M. Kawut
- Dept of Medicine and the Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sanja Jelic
- Division of Pulmonary, Allergy and Critical Care Medicine, Columbia University, New York, NY, USA
| | - Selim M. Arcasoy
- Division of Pulmonary, Allergy and Critical Care Medicine, Columbia University, New York, NY, USA,Interstitial Lung Disease Program, New York Presbyterian Hospital, New York, NY, USA,Lung Transplantation Program, New York Presbyterian Hospital, New York, NY, USA
| | - David J. Lederer
- Division of Pulmonary, Allergy and Critical Care Medicine, Columbia University, New York, NY, USA,Interstitial Lung Disease Program, New York Presbyterian Hospital, New York, NY, USA,Lung Transplantation Program, New York Presbyterian Hospital, New York, NY, USA
| | - Alain C. Borczuk
- Dept of Pathology and Cell Biology, Columbia University, New York, NY, USA
| |
Collapse
|
48
|
Inhibition of LRP5/6-mediated Wnt/β-catenin signaling by Mesd attenuates hyperoxia-induced pulmonary hypertension in neonatal rats. Pediatr Res 2013; 73:719-25. [PMID: 23481549 DOI: 10.1038/pr.2013.42] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Hyperoxia-induced neonatal lung injury is associated with activation of Wnt/β-catenin signaling. Low-density lipoprotein receptor-related proteins 5 and 6 (LRP5/6) are Wnt coreceptors that bind to Wnt ligands and mediate canonical Wnt/β-catenin signaling. We hypothesized that inhibition of LRP5/6 by their universal inhibitor, Mesd, would attenuate hyperoxia-induced lung injury. METHODS Newborn rat pups were randomly exposed to normoxia or hyperoxia at 90% FiO2 and injected intraperitoneally with placebo or Mesd every other day for 14 d. On day 15, phosphorylation of LRP5/6 (pLRP5/6), expression of Wnt/β-catenin target genes, cyclin D1 and Wnt-induced signaling protein-1 (WISP-1), right-ventricular systolic pressure (RVSP), right-ventricular hypertrophy (RVH), pulmonary vascular remodeling, alveolarization, and vascularization were measured. RESULTS Hyperoxia exposure markedly induced pLRP5/6, cyclin D1, and WISP-1 expression in the lungs of placebo animals, but they were significantly attenuated by the administration of Mesd. Mesd also significantly attenuated hyperoxia-induced pulmonary hypertension (PH) and pulmonary vascular remodeling. However, there was no effect on alveolarization or vascularization after Mesd administration. CONCLUSION This study demonstrates that LRP5/6 mediates pulmonary vascular remodeling and PH in hyperoxia-induced neonatal lung injury, thereby suggesting a potential therapeutic target to alleviate PH in neonates with severe bronchopulmonary dysplasia.
Collapse
|
49
|
Al Alam D, Warburton D. Wingless: developmentally important genes that respond adversely to smoking. Thorax 2013; 68:703-4. [PMID: 23611881 DOI: 10.1136/thoraxjnl-2013-203249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
50
|
Storck EM, Wojciak-Stothard B. Rho GTPases in pulmonary vascular dysfunction. Vascul Pharmacol 2013; 58:202-10. [DOI: 10.1016/j.vph.2012.09.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Revised: 09/05/2012] [Accepted: 09/09/2012] [Indexed: 12/19/2022]
|