1
|
Noda K, Hattori Y, Murata H, Kokubo Y, Higashiyama A, Ihara M. Equol Nonproducing Status as an Independent Risk Factor for Acute Cardioembolic Stroke and Poor Functional Outcome. Nutrients 2024; 16:3377. [PMID: 39408343 PMCID: PMC11479244 DOI: 10.3390/nu16193377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 09/26/2024] [Accepted: 10/02/2024] [Indexed: 10/20/2024] Open
Abstract
Background/Objectives: Equol has protective effects against coronary artery disease and dementia by strongly binding to estrogen receptor beta, whereas the intake of soy isoflavone alone does not always confer such protective effects. Equol production is completely dependent on the existence of equol-producing gut microbiota. The effects of equol-producing status on the cerebrovascular diseases remain unclear. The current study was aimed to investigate the association of equol-producing status with the development of stroke and its neurological prognosis. Methods: Frequencies of equol producers were compared between healthy subjects (HS) registered in the Suita Study and patients with acute stroke admitted to our stroke center from September 2019 to October 2021 in a retrospective cohort study. Results: The proportion of HSs and patients with ischemic stroke who were equol producers did not significantly differ (50/103 [48.5%] vs. 60/140 [42.9%], p = 0.38). However, cardioembolic stroke was significantly associated with low a prevalence of equol producers (adjusted odds ratio [aOR] 0.46, 95% confidence interval [CI] 0.21-0.99, p = 0.05). A higher left atrial volume index was observed in equol nonproducers (46.3 ± 23.8 vs. 36.0 ± 11.6 mL/m2, p = 0.06). The equol nonproducers had a significantly higher prevalence of atrial fibrillation than the equol producers (27.5% vs. 13.3%, p = 0.04). Furthermore, the equol producers exhibited a significantly favorable functional outcome upon discharge (aOR 2.84, 95% CI 1.20-6.75, p = 0.02). Conclusions: Equol is a promising candidate for interventions aiming to reduce the risk of CES and atrial dysfunction, such as atrial fibrillation and improve neurological prognosis after ischemic stroke.
Collapse
Affiliation(s)
- Kotaro Noda
- Department of Neurology, National Cerebral and Cardiovascular Center, 6-1, Kishibe-shimmachi, Suita 564-8565, Japan
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Yorito Hattori
- Department of Neurology, Department of Preemptive Medicine for Dementia, National Cerebral and Cardiovascular Center, 6-1, Kishibe-shimmachi, Suita 564-8565, Japan
| | - Hiroaki Murata
- Department of Neurology, National Cerebral and Cardiovascular Center, 6-1, Kishibe-shimmachi, Suita 564-8565, Japan
| | - Yoshihiro Kokubo
- Department of Preventive Cardiology, National Cerebral and Cardiovascular Center, 6-1, Kishibe-shimmachi, Suita 564-8565, Japan
| | - Aya Higashiyama
- Department of Preventive Cardiology, National Cerebral and Cardiovascular Center, 6-1, Kishibe-shimmachi, Suita 564-8565, Japan
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Center, 6-1, Kishibe-shimmachi, Suita 564-8565, Japan
| |
Collapse
|
2
|
Correale M, Chirivì F, Bevere EML, Tricarico L, D’Alto M, Badagliacca R, Brunetti ND, Vizza CD, Ghio S. Endothelial Function in Pulmonary Arterial Hypertension: From Bench to Bedside. J Clin Med 2024; 13:2444. [PMID: 38673717 PMCID: PMC11051060 DOI: 10.3390/jcm13082444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/09/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
Pulmonary arterial hypertension is a complex pathology whose etiology is still not completely well clarified. The pathogenesis of pulmonary arterial hypertension involves different molecular mechanisms, with endothelial dysfunction playing a central role in disease progression. Both individual genetic predispositions and environmental factors seem to contribute to its onset. To further understand the complex relationship between endothelial and pulmonary hypertension and try to contribute to the development of future therapies, we report a comprehensive and updated review on endothelial function in pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Michele Correale
- Cardiothoracic Department, Policlinico Riuniti University Hospital, 71100 Foggia, Italy;
| | - Francesco Chirivì
- Department of Medical and Surgical Sciences, University of Foggia, 71100 Foggia, Italy; (F.C.); (E.M.L.B.); (N.D.B.)
| | - Ester Maria Lucia Bevere
- Department of Medical and Surgical Sciences, University of Foggia, 71100 Foggia, Italy; (F.C.); (E.M.L.B.); (N.D.B.)
| | - Lucia Tricarico
- Cardiothoracic Department, Policlinico Riuniti University Hospital, 71100 Foggia, Italy;
| | - Michele D’Alto
- Department of Cardiology, A.O.R.N. dei Colli, Monaldi Hospital, University of Campania L. ‘Vanvitelli’, 80133 Naples, Italy;
| | - Roberto Badagliacca
- Department of Clinical, Anesthesiological and Cardiovascular Sciences, I School of Medicine, Sapienza University of Rome, 00185 Rome, Italy; (R.B.); (C.D.V.)
| | - Natale D. Brunetti
- Department of Medical and Surgical Sciences, University of Foggia, 71100 Foggia, Italy; (F.C.); (E.M.L.B.); (N.D.B.)
| | - Carmine Dario Vizza
- Department of Clinical, Anesthesiological and Cardiovascular Sciences, I School of Medicine, Sapienza University of Rome, 00185 Rome, Italy; (R.B.); (C.D.V.)
| | - Stefano Ghio
- Division of Cardiology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| |
Collapse
|
3
|
Behringer EJ. Impact of aging on vascular ion channels: perspectives and knowledge gaps across major organ systems. Am J Physiol Heart Circ Physiol 2023; 325:H1012-H1038. [PMID: 37624095 PMCID: PMC10908410 DOI: 10.1152/ajpheart.00288.2023] [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: 05/16/2023] [Revised: 08/21/2023] [Accepted: 08/21/2023] [Indexed: 08/26/2023]
Abstract
Individuals aged ≥65 yr will comprise ∼20% of the global population by 2030. Cardiovascular disease remains the leading cause of death in the world with age-related endothelial "dysfunction" as a key risk factor. As an organ in and of itself, vascular endothelium courses throughout the mammalian body to coordinate blood flow to all other organs and tissues (e.g., brain, heart, lung, skeletal muscle, gut, kidney, skin) in accord with metabolic demand. In turn, emerging evidence demonstrates that vascular aging and its comorbidities (e.g., neurodegeneration, diabetes, hypertension, kidney disease, heart failure, and cancer) are "channelopathies" in large part. With an emphasis on distinct functional traits and common arrangements across major organs systems, the present literature review encompasses regulation of vascular ion channels that underlie blood flow control throughout the body. The regulation of myoendothelial coupling and local versus conducted signaling are discussed with new perspectives for aging and the development of chronic diseases. Although equipped with an awareness of knowledge gaps in the vascular aging field, a section has been included to encompass general feasibility, role of biological sex, and additional conceptual and experimental considerations (e.g., cell regression and proliferation, gene profile analyses). The ultimate goal is for the reader to see and understand major points of deterioration in vascular function while gaining the ability to think of potential mechanistic and therapeutic strategies to sustain organ perfusion and whole body health with aging.
Collapse
Affiliation(s)
- Erik J Behringer
- Basic Sciences, Loma Linda University, Loma Linda, California, United States
| |
Collapse
|
4
|
Ayon RJ, Yuan JXJ. Revisiting the Role of KCNA5 in Pulmonary Arterial Hypertension. Am J Respir Cell Mol Biol 2023; 69:123-125. [PMID: 37201951 PMCID: PMC10399140 DOI: 10.1165/rcmb.2023-0119ed] [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: 03/28/2023] [Accepted: 05/17/2023] [Indexed: 05/20/2023] Open
Affiliation(s)
- Ramon J Ayon
- Department of Molecular Physiology and Biological Physics University of Virginia Charlottesville, Virginia
| | - Jason X-J Yuan
- Department of Medicine University of California, San Diego La Jolla, California
| |
Collapse
|
5
|
Balistrieri A, Makino A, Yuan JXJ. Pathophysiology and pathogenic mechanisms of pulmonary hypertension: role of membrane receptors, ion channels, and Ca 2+ signaling. Physiol Rev 2023; 103:1827-1897. [PMID: 36422993 PMCID: PMC10110735 DOI: 10.1152/physrev.00030.2021] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/11/2022] [Accepted: 11/19/2022] [Indexed: 11/25/2022] Open
Abstract
The pulmonary circulation is a low-resistance, low-pressure, and high-compliance system that allows the lungs to receive the entire cardiac output. Pulmonary arterial pressure is a function of cardiac output and pulmonary vascular resistance, and pulmonary vascular resistance is inversely proportional to the fourth power of the intraluminal radius of the pulmonary artery. Therefore, a very small decrease of the pulmonary vascular lumen diameter results in a significant increase in pulmonary vascular resistance and pulmonary arterial pressure. Pulmonary arterial hypertension is a fatal and progressive disease with poor prognosis. Regardless of the initial pathogenic triggers, sustained pulmonary vasoconstriction, concentric vascular remodeling, occlusive intimal lesions, in situ thrombosis, and vascular wall stiffening are the major and direct causes for elevated pulmonary vascular resistance in patients with pulmonary arterial hypertension and other forms of precapillary pulmonary hypertension. In this review, we aim to discuss the basic principles and physiological mechanisms involved in the regulation of lung vascular hemodynamics and pulmonary vascular function, the changes in the pulmonary vasculature that contribute to the increased vascular resistance and arterial pressure, and the pathogenic mechanisms involved in the development and progression of pulmonary hypertension. We focus on reviewing the pathogenic roles of membrane receptors, ion channels, and intracellular Ca2+ signaling in pulmonary vascular smooth muscle cells in the development and progression of pulmonary hypertension.
Collapse
Affiliation(s)
- Angela Balistrieri
- Section of Physiology, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
- Harvard University, Cambridge, Massachusetts
| | - Ayako Makino
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Jason X-J Yuan
- Section of Physiology, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| |
Collapse
|
6
|
Benincasa G, Napoli C, Loscalzo J, Maron BA. Pursuing functional biomarkers in complex disease: Focus on pulmonary arterial hypertension. Am Heart J 2023; 258:96-113. [PMID: 36565787 DOI: 10.1016/j.ahj.2022.12.009] [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: 09/28/2022] [Revised: 12/12/2022] [Accepted: 12/19/2022] [Indexed: 05/11/2023]
Abstract
A major gap in diagnosis, classification, risk stratification, and prediction of therapeutic response exists in pulmonary arterial hypertension (PAH), driven in part by a lack of functional biomarkers that are also disease-specific. In this regard, leveraging big data-omics analyses using innovative approaches that integrate network medicine and machine learning correlated with clinically useful indices or risk stratification scores is an approach well-positioned to advance PAH precision medicine. For example, machine learning applied to a panel of 48 cytokines, chemokines, and growth factors could prognosticate PAH patients with immune-dominant subphenotypes at elevated or low-risk for mortality. Here, we discuss strengths and weaknesses of the most current studies evaluating omics-derived biomarkers in PAH. Progress in this field is offset by studies with small sample size, pervasive limitations in bioinformatics, and lack of standardized methods for data processing and interpretation. Future success in this field, in turn, is likely to hinge on mechanistic validation of data outputs in order to couple functional biomarker data with target-specific therapeutics in clinical practice.
Collapse
Affiliation(s)
- Giuditta Benincasa
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", Naples, Italy.
| | - Claudio Napoli
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Joseph Loscalzo
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA
| | - Bradley A Maron
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA.
| |
Collapse
|
7
|
Vera-Zambrano A, Baena-Nuevo M, Rinné S, Villegas-Esguevillas M, Barreira B, Telli G, de Benito-Bueno A, Blázquez JA, Climent B, Pérez-Vizcaino F, Valenzuela C, Decher N, Gonzalez T, Cogolludo A. Sigma-1 receptor modulation fine-tunes K V1.5 channels and impacts pulmonary vascular function. Pharmacol Res 2023; 189:106684. [PMID: 36740150 DOI: 10.1016/j.phrs.2023.106684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/23/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023]
Abstract
KV1.5 channels are key players in the regulation of vascular tone and atrial excitability and their impairment is associated with cardiovascular diseases including pulmonary arterial hypertension (PAH) and atrial fibrillation (AF). Unfortunately, pharmacological strategies to improve KV1.5 channel function are missing. Herein, we aimed to study whether the chaperone sigma-1 receptor (S1R) is able to regulate these channels and represent a new strategy to enhance their function. By using different electrophysiological and molecular techniques in X. laevis oocytes and HEK293 cells, we demonstrate that S1R physically interacts with KV1.5 channels and regulate their expression and function. S1R induced a bimodal regulation of KV1.5 channel expression/activity, increasing it at low concentrations and decreasing it at high concentrations. Of note, S1R agonists (PRE084 and SKF10047) increased, whereas the S1R antagonist BD1047 decreased, KV1.5 expression and activity. Moreover, PRE084 markedly increased KV1.5 currents in pulmonary artery smooth muscle cells and attenuated vasoconstriction and proliferation in pulmonary arteries. We also show that both KV1.5 channels and S1R, at mRNA and protein levels, are clearly downregulated in samples from PAH and AF patients. Moreover, the expression of both genes showed a positive correlation. Finally, the ability of PRE084 to increase KV1.5 function was preserved under sustained hypoxic conditions, as an in vitro PAH model. Our study provides insight into the key role of S1R in modulating the expression and activity of KV1.5 channels and highlights the potential role of this chaperone as a novel pharmacological target for pathological conditions associated with KV1.5 channel dysfunction.
Collapse
Affiliation(s)
- Alba Vera-Zambrano
- Department of Biochemistry, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain; Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Madrid, Spain; Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Madrid, Spain.
| | - Maria Baena-Nuevo
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Madrid, Spain
| | - Susanne Rinné
- Institute of Physiology and Pathophysiology, Vegetative Physiology, University of Marburg, 35043 Marburg, Germany
| | - Marta Villegas-Esguevillas
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Madrid, Spain; Ciber Enfermedades Respiratorias (CIBERES), Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Bianca Barreira
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Madrid, Spain; Ciber Enfermedades Respiratorias (CIBERES), Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Gokcen Telli
- Hacettepe University, Department of Pharmacology, Faculty of Pharmacy, Ankara, Turkey
| | | | | | - Belén Climent
- Department of Physiology, Faculty of Pharmacy, University Complutense of Madrid, Madrid, Spain
| | - Francisco Pérez-Vizcaino
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Madrid, Spain; Ciber Enfermedades Respiratorias (CIBERES), Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Carmen Valenzuela
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Madrid, Spain; Spanish Network for Biomedical Research in Cardiovascular Research (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Niels Decher
- Institute of Physiology and Pathophysiology, Vegetative Physiology, University of Marburg, 35043 Marburg, Germany
| | - Teresa Gonzalez
- Department of Biochemistry, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain; Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Madrid, Spain; Department of Physiology, Faculty of Pharmacy, University Complutense of Madrid, Madrid, Spain
| | - Angel Cogolludo
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Madrid, Spain; Ciber Enfermedades Respiratorias (CIBERES), Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.
| |
Collapse
|
8
|
Moreno-Domínguez A, Colinas O, Smani T, Ureña J, López-Barneo J. Acute oxygen sensing by vascular smooth muscle cells. Front Physiol 2023; 14:1142354. [PMID: 36935756 PMCID: PMC10020353 DOI: 10.3389/fphys.2023.1142354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 02/21/2023] [Indexed: 03/06/2023] Open
Abstract
An adequate supply of oxygen (O2) is essential for most life forms on earth, making the delivery of appropriate levels of O2 to tissues a fundamental physiological challenge. When O2 levels in the alveoli and/or blood are low, compensatory adaptive reflexes are produced that increase the uptake of O2 and its distribution to tissues within a few seconds. This paper analyzes the most important acute vasomotor responses to lack of O2 (hypoxia): hypoxic pulmonary vasoconstriction (HPV) and hypoxic vasodilation (HVD). HPV affects distal pulmonary (resistance) arteries, with its homeostatic role being to divert blood to well ventilated alveoli to thereby optimize the ventilation/perfusion ratio. HVD is produced in most systemic arteries, in particular in the skeletal muscle, coronary, and cerebral circulations, to increase blood supply to poorly oxygenated tissues. Although vasomotor responses to hypoxia are modulated by endothelial factors and autonomic innervation, it is well established that arterial smooth muscle cells contain an acute O2 sensing system capable of detecting changes in O2 tension and to signal membrane ion channels, which in turn regulate cytosolic Ca2+ levels and myocyte contraction. Here, we summarize current knowledge on the nature of O2 sensing and signaling systems underlying acute vasomotor responses to hypoxia. We also discuss similarities and differences existing in O2 sensors and effectors in the various arterial territories.
Collapse
Affiliation(s)
- Alejandro Moreno-Domínguez
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Olaia Colinas
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Tarik Smani
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
| | - Juan Ureña
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
| | - José López-Barneo
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- *Correspondence: José López-Barneo,
| |
Collapse
|
9
|
Redel-Traub G, Sampson KJ, Kass RS, Bohnen MS. Potassium Channels as Therapeutic Targets in Pulmonary Arterial Hypertension. Biomolecules 2022; 12:1341. [PMID: 36291551 PMCID: PMC9599705 DOI: 10.3390/biom12101341] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/16/2022] [Accepted: 09/18/2022] [Indexed: 12/08/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a devastating disease with high morbidity and mortality. Deleterious remodeling in the pulmonary arterial system leads to irreversible arterial constriction and elevated pulmonary arterial pressures, right heart failure, and eventually death. The difficulty in treating PAH stems in part from the complex nature of disease pathogenesis, with several signaling compounds known to be involved (e.g., endothelin-1, prostacyclins) which are indeed targets of PAH therapy. Over the last decade, potassium channelopathies were established as novel causes of PAH. More specifically, loss-of-function mutations in the KCNK3 gene that encodes the two-pore-domain potassium channel KCNK3 (or TASK-1) and loss-of-function mutations in the ABCC8 gene that encodes a key subunit, SUR1, of the ATP-sensitive potassium channel (KATP) were established as the first two potassium channelopathies in human cohorts with pulmonary arterial hypertension. Moreover, voltage-gated potassium channels (Kv) represent a third family of potassium channels with genetic changes observed in association with PAH. While other ion channel genes have since been reported in association with PAH, this review focuses on KCNK3, KATP, and Kv potassium channels as promising therapeutic targets in PAH, with recent experimental pharmacologic discoveries significantly advancing the field.
Collapse
Affiliation(s)
- Gabriel Redel-Traub
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Kevin J. Sampson
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Robert S. Kass
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Michael S. Bohnen
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
10
|
Moral-Sanz J, Lewis SA, MacMillan S, Meloni M, McClafferty H, Viollet B, Foretz M, Del-Pozo J, Mark Evans A. AMPK deficiency in smooth muscles causes persistent pulmonary hypertension of the new-born and premature death. Nat Commun 2022; 13:5034. [PMID: 36028487 PMCID: PMC9418192 DOI: 10.1038/s41467-022-32568-7] [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: 04/14/2021] [Accepted: 08/05/2022] [Indexed: 11/10/2022] Open
Abstract
AMPK has been reported to facilitate hypoxic pulmonary vasoconstriction but, paradoxically, its deficiency precipitates pulmonary hypertension. Here we show that AMPK-α1/α2 deficiency in smooth muscles promotes persistent pulmonary hypertension of the new-born. Accordingly, dual AMPK-α1/α2 deletion in smooth muscles causes premature death of mice after birth, associated with increased muscularisation and remodeling throughout the pulmonary arterial tree, reduced alveolar numbers and alveolar membrane thickening, but with no oedema. Spectral Doppler ultrasound indicates pulmonary hypertension and attenuated hypoxic pulmonary vasoconstriction. Age-dependent right ventricular pressure elevation, dilation and reduced cardiac output was also evident. KV1.5 potassium currents of pulmonary arterial myocytes were markedly smaller under normoxia, which is known to facilitate pulmonary hypertension. Mitochondrial fragmentation and reactive oxygen species accumulation was also evident. Importantly, there was no evidence of systemic vasculopathy or hypertension in these mice. Moreover, hypoxic pulmonary vasoconstriction was attenuated by AMPK-α1 or AMPK-α2 deletion without triggering pulmonary hypertension.
Collapse
Affiliation(s)
- Javier Moral-Sanz
- Centre for Discovery Brain Sciences and Cardiovascular Science, College of Medicine and Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Sophronia A Lewis
- Centre for Discovery Brain Sciences and Cardiovascular Science, College of Medicine and Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Sandy MacMillan
- Centre for Discovery Brain Sciences and Cardiovascular Science, College of Medicine and Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Marco Meloni
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Heather McClafferty
- Centre for Discovery Brain Sciences and Cardiovascular Science, College of Medicine and Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Benoit Viollet
- Université Paris Cité, CNRS, INSERM, Institut Cochin, F-75014, Paris, France
| | - Marc Foretz
- Université Paris Cité, CNRS, INSERM, Institut Cochin, F-75014, Paris, France
| | - Jorge Del-Pozo
- R(D)SVS, University of Edinburgh Easter Bush Campus, EH25 9RG, Roslin, Edinburgh, UK
| | - A Mark Evans
- Centre for Discovery Brain Sciences and Cardiovascular Science, College of Medicine and Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh, EH8 9XD, UK.
| |
Collapse
|
11
|
Welch CL, Chung WK. Channelopathy Genes in Pulmonary Arterial Hypertension. Biomolecules 2022; 12:265. [PMID: 35204766 PMCID: PMC8961593 DOI: 10.3390/biom12020265] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 01/22/2022] [Accepted: 01/25/2022] [Indexed: 02/07/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare, progressive vasculopathy with significant cardiopulmonary morbidity and mortality. The underlying pathogenetic mechanisms are heterogeneous and current therapies aim to decrease pulmonary vascular resistance but no curative treatments are available. Causal genetic variants can be identified in ~13% of adults and 43% of children with PAH. Knowledge of genetic diagnoses can inform clinical management of PAH, including multimodal medical treatment, surgical intervention and transplantation decisions, and screening for associated conditions, as well as risk stratification for family members. Roles for rare variants in three channelopathy genes-ABCC8, ATP13A3, and KCNK3-have been validated in multiple PAH cohorts, and in aggregate explain ~2.7% of PAH cases. Complete or partial loss of function has been demonstrated for PAH-associated variants in ABCC8 and KCNK3. Channels can be excellent targets for drugs, and knowledge of mechanisms for channel mutations may provide an opportunity for the development of PAH biomarkers and novel therapeutics for patients with hereditary PAH but also potentially more broadly for all patients with PAH.
Collapse
Affiliation(s)
- Carrie L. Welch
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY 10032, USA;
| | - Wendy K. Chung
- Department of Pediatrics, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Medicine, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
12
|
Borrego J, Feher A, Jost N, Panyi G, Varga Z, Papp F. Peptide Inhibitors of Kv1.5: An Option for the Treatment of Atrial Fibrillation. Pharmaceuticals (Basel) 2021; 14:1303. [PMID: 34959701 PMCID: PMC8704205 DOI: 10.3390/ph14121303] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/08/2021] [Accepted: 12/09/2021] [Indexed: 12/12/2022] Open
Abstract
The human voltage gated potassium channel Kv1.5 that conducts the IKur current is a key determinant of the atrial action potential. Its mutations have been linked to hereditary forms of atrial fibrillation (AF), and the channel is an attractive target for the management of AF. The development of IKur blockers to treat AF resulted in small molecule Kv1.5 inhibitors. The selectivity of the blocker for the target channel plays an important role in the potential therapeutic application of the drug candidate: the higher the selectivity, the lower the risk of side effects. In this respect, small molecule inhibitors of Kv1.5 are compromised due to their limited selectivity. A wide range of peptide toxins from venomous animals are targeting ion channels, including mammalian channels. These peptides usually have a much larger interacting surface with the ion channel compared to small molecule inhibitors and thus, generally confer higher selectivity to the peptide blockers. We found two peptides in the literature, which inhibited IKur: Ts6 and Osu1. Their affinity and selectivity for Kv1.5 can be improved by rational drug design in which their amino acid sequences could be modified in a targeted way guided by in silico docking experiments.
Collapse
Affiliation(s)
- Jesús Borrego
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem ter 1, H-4032 Debrecen, Hungary; (J.B.); (A.F.); (G.P.); (Z.V.)
| | - Adam Feher
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem ter 1, H-4032 Debrecen, Hungary; (J.B.); (A.F.); (G.P.); (Z.V.)
| | - Norbert Jost
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, 6725 Szeged, Hungary;
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, 6725 Szeged, Hungary
- ELKH-SZTE Research Group for Cardiovascular Pharmacology, Eötvös Loránd Research Network, 6725 Szeged, Hungary
| | - Gyorgy Panyi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem ter 1, H-4032 Debrecen, Hungary; (J.B.); (A.F.); (G.P.); (Z.V.)
| | - Zoltan Varga
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem ter 1, H-4032 Debrecen, Hungary; (J.B.); (A.F.); (G.P.); (Z.V.)
| | - Ferenc Papp
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem ter 1, H-4032 Debrecen, Hungary; (J.B.); (A.F.); (G.P.); (Z.V.)
| |
Collapse
|
13
|
West J, Rathinasabapathy A, Chen X, Shay S, Gladson S, Talati M. Overexpression of Msx1 in Mouse Lung Leads to Loss of Pulmonary Vessels Following Vascular Hypoxic Injury. Cells 2021; 10:cells10092306. [PMID: 34571956 PMCID: PMC8471093 DOI: 10.3390/cells10092306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/11/2021] [Accepted: 08/19/2021] [Indexed: 11/16/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive lung disease caused by thickening of the pulmonary arterial wall and luminal obliteration of the small peripheral arteries leading to increase in vascular resistance which elevates pulmonary artery pressure that eventually causes right heart failure and death. We have previously shown that transcription factor Msx1 (mainly expressed during embryogenesis) is strongly upregulated in transformed lymphocytes obtained from PAH patients, especially IPAH. Under pathological conditions, Msx1 overexpression can cause cell dedifferentiation or cell apoptosis. We hypothesized that Msx1 overexpression contributes to loss of small pulmonary vessels in PAH. In IPAH lung, MSX1 protein localization was strikingly increased in muscularized remodeled pulmonary vessels, whereas it was undetectable in control pulmonary arteries. We developed a transgenic mouse model overexpressing MSX1 (MSX1OE) by about 4-fold and exposed these mice to normoxic, sugen hypoxic (3 weeks) or hyperoxic (100% 02 for 3 weeks) conditions. Under normoxic conditions, compared to controls, MSX1OE mice demonstrated a 30-fold and 2-fold increase in lung Msx1 mRNA and protein expression, respectively. There was a significant retinal capillary dropout (p < 0.01) in MSX1OE mice, which was increased further (p < 0.03) with sugen hypoxia. At baseline, the number of pulmonary vessels in MSX1OE mice was similar to controls. In sugen-hypoxia-treated MSX1OE mice, the number of small (0-25 uM) and medium (25-50 uM) size muscularized vessels increased approximately 2-fold (p < 0.01) compared to baseline controls; however, they were strikingly lower (p < 0.001) in number than in sugen-hypoxia-treated control mice. In MSX1OE mouse lung, 104 genes were upregulated and 67 genes were downregulated compared to controls. Similarly, in PVECs, 156 genes were upregulated and 320 genes were downregulated from siRNA to MSX1OE, and in PVSMCs, 65 genes were upregulated and 321 genes were downregulated from siRNA to MSX1OE (with control in the middle). Many of the statistically significant GO groups associated with MSX1 expression in lung, PVECs, and PVSMCs were similar, and were involved in cell cycle, cytoskeletal and macromolecule organization, and programmed cell death. Overexpression of MSX1 suppresses many cell-cycle-related genes in PVSMCs but induces them in PVECs. In conclusion, overexpression of Msx1 leads to loss of pulmonary vessels, which is exacerbated by sugen hypoxia, and functional consequences of Msx1 overexpression are cell-dependent.
Collapse
Affiliation(s)
| | | | | | | | | | - Megha Talati
- Correspondence: ; Tel.: +1-615-322-8095; Fax: +1-615-343-7448
| |
Collapse
|
14
|
Ion channels as convergence points in the pathology of pulmonary arterial hypertension. Biochem Soc Trans 2021; 49:1855-1865. [PMID: 34346486 PMCID: PMC8421048 DOI: 10.1042/bst20210538] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 12/17/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a fatal disease of the cardiopulmonary system that lacks curative treatments. The main pathological event in PAH is elevated vascular resistance in the pulmonary circulation, caused by abnormal vasoconstriction and vascular remodelling. Ion channels are key determinants of vascular smooth muscle tone and homeostasis, and four PAH channelopathies (KCNK3, ABCC8, KCNA5, TRPC6) have been identified so far. However, the contribution of ion channels in other forms of PAH, which account for the majority of PAH patients, has been less well characterised. Here we reason that a variety of triggers of PAH (e.g. BMPR2 mutations, hypoxia, anorectic drugs) that impact channel function may contribute to the onset of the disease. We review the molecular mechanisms by which these ‘extrinsic’ factors converge on ion channels and provoke their dysregulation to promote the development of PAH. Ion channels of the pulmonary vasculature are therefore promising therapeutic targets because of the modulation they provide to both vasomotor tone and proliferation of arterial smooth muscle cells.
Collapse
|
15
|
Pyrrolizidine alkaloid-induced transcriptomic changes in rat lungs in a 28-day subacute feeding study. Arch Toxicol 2021; 95:2785-2796. [PMID: 34185104 PMCID: PMC8298252 DOI: 10.1007/s00204-021-03108-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/17/2021] [Indexed: 11/28/2022]
Abstract
Pyrrolizidine alkaloids (PAs) are secondary plant metabolites synthesized by a wide range of plants as protection against herbivores. These toxins are found worldwide and pose a threat to human health. PAs induce acute effects like hepatic sinusoidal obstruction syndrome and pulmonary arterial hypertension. Moreover, chronic exposure to low doses can induce cancer and liver cirrhosis in laboratory animals. The mechanisms causing hepatotoxicity have been investigated previously. However, toxic effects in the lung are less well understood, and especially data on the correlation effects with individual chemical structures of different PAs are lacking. The present study focuses on the identification of gene expression changes in vivo in rat lungs after exposure to six structurally different PAs (echimidine, heliotrine, lasiocarpine, senecionine, senkirkine, and platyphylline). Rats were treated by gavage with daily doses of 3.3 mg PA/kg bodyweight for 28 days and transcriptional changes in the lung and kidney were investigated by whole-genome microarray analysis. The results were compared with recently published data on gene regulation in the liver. Using bioinformatics data mining, we identified inflammatory responses as a predominant feature in rat lungs. By comparison, in liver, early molecular consequences to PAs were characterized by alterations in cell-cycle regulation and DNA damage response. Our results provide, for the first time, information about early molecular effects in lung tissue after subacute exposure to PAs, and demonstrates tissue-specificity of PA-induced molecular effects.
Collapse
|
16
|
Affiliation(s)
- Steven K Huang
- Division of Pulmonary and Critical Care Medicine University of Michigan Ann Arbor, Michigan
| |
Collapse
|
17
|
Egom EEA, Moyou-Somo R, Essame Oyono JL, Kamgang R. Identifying Potential Mutations Responsible for Cases of Pulmonary Arterial Hypertension. APPLICATION OF CLINICAL GENETICS 2021; 14:113-124. [PMID: 33732008 PMCID: PMC7958998 DOI: 10.2147/tacg.s260755] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 02/18/2021] [Indexed: 01/09/2023]
Abstract
Pulmonary Arterial Hypertension (PAH) is a progressive and devastating disease for which there is an escalating body of genetic and related pathophysiological information on disease pathobiology. Nevertheless, the success to date in identifying susceptibility genes, genetic variants and epigenetic processes has been limited due to PAH clinical multi-faceted variations. A number of germline gene candidates have been proposed but demonstrating consistently the association with PAH has been problematic, at least partly due to the reduced penetrance and variable expressivity. Although the data for bone morphogenetic protein receptor type 2 (BMPR2) and related genes remains undoubtedly the most extensive, recent advanced gene sequencing technologies have facilitated the discovery of further gene candidates with mutations among those with and without familial forms of PAH. An in depth understanding of the multitude of biologic variations associated with PAH may provide novel opportunities for therapeutic intervention in the coming years. This knowledge will irrevocably provide the opportunity for improved patient and family counseling as well as improved PAH diagnosis, risk assessment, and personalized treatment.
Collapse
Affiliation(s)
- Emmanuel Eroume-A Egom
- Institut du Savoir Montfort (ISM), Hôpital Montfort, Ottawa, ON, Canada.,Laboratory of Endocrinology and Radioisotopes, Institute of Medical Research and Medicinal Plants Studies (IMPM), Yaoundé, Cameroon.,Reflex Medical Centre Cardiac Diagnostics, Reflex Medical Centre, Mississauga, ON, Canada
| | - Roger Moyou-Somo
- Laboratory of Endocrinology and Radioisotopes, Institute of Medical Research and Medicinal Plants Studies (IMPM), Yaoundé, Cameroon
| | - Jean Louis Essame Oyono
- Laboratory of Endocrinology and Radioisotopes, Institute of Medical Research and Medicinal Plants Studies (IMPM), Yaoundé, Cameroon
| | - Rene Kamgang
- Laboratory of Endocrinology and Radioisotopes, Institute of Medical Research and Medicinal Plants Studies (IMPM), Yaoundé, Cameroon
| |
Collapse
|
18
|
Mondéjar-Parreño G, Cogolludo A, Perez-Vizcaino F. Potassium (K +) channels in the pulmonary vasculature: Implications in pulmonary hypertension Physiological, pathophysiological and pharmacological regulation. Pharmacol Ther 2021; 225:107835. [PMID: 33744261 DOI: 10.1016/j.pharmthera.2021.107835] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/26/2021] [Accepted: 03/02/2021] [Indexed: 02/06/2023]
Abstract
The large K+ channel functional diversity in the pulmonary vasculature results from the multitude of genes expressed encoding K+ channels, alternative RNA splicing, the post-transcriptional modifications, the presence of homomeric or heteromeric assemblies of the pore-forming α-subunits and the existence of accessory β-subunits modulating the functional properties of the channel. K+ channels can also be regulated at multiple levels by different factors controlling channel activity, trafficking, recycling and degradation. The activity of these channels is the primary determinant of membrane potential (Em) in pulmonary artery smooth muscle cells (PASMC), providing an essential regulatory mechanism to dilate or contract pulmonary arteries (PA). K+ channels are also expressed in pulmonary artery endothelial cells (PAEC) where they control resting Em, Ca2+ entry and the production of different vasoactive factors. The activity of K+ channels is also important in regulating the population and phenotype of PASMC in the pulmonary vasculature, since they are involved in cell apoptosis, survival and proliferation. Notably, K+ channels play a major role in the development of pulmonary hypertension (PH). Impaired K+ channel activity in PH results from: 1) loss of function mutations, 2) downregulation of its expression, which involves transcription factors and microRNAs, or 3) decreased channel current as a result of increased vasoactive factors (e.g., hypoxia, 5-HT, endothelin-1 or thromboxane), exposure to drugs with channel-blocking properties, or by a reduction in factors that positively regulate K+ channel activity (e.g., NO and prostacyclin). Restoring K+ channel expression, its intracellular trafficking and the channel activity is an attractive therapeutic strategy in PH.
Collapse
Affiliation(s)
- Gema Mondéjar-Parreño
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain; Ciber Enfermedades Respiratorias (CIBERES), Spain
| | - Angel Cogolludo
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain; Ciber Enfermedades Respiratorias (CIBERES), Spain
| | - Francisco Perez-Vizcaino
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain; Ciber Enfermedades Respiratorias (CIBERES), Spain.
| |
Collapse
|
19
|
Abstract
Atrial fibrillation is associated with aging, obesity, heart disease, diabetes, and/or hypertension. Recent evidence suggests that parenchymal and vascular lung diseases increase atrial fibrillation risk. We review the epidemiology, clinical features, pathophysiologic mechanisms, and treatment implications of atrial fibrillation associated with diseases of the lungs and their vasculature, especially pulmonary hypertension. We also consider other features of pulmonary disease-associated atrial fibrillation. A key mediator of these conditions is right heart disease and right atrial remodeling. We pay particular attention to the pathophysiology and treatment challenges in atrial fibrillation associated with right heart disease induced by pulmonary diseases, including pulmonary hypertension.
Collapse
Affiliation(s)
- Roddy Hiram
- Department of Medicine, Montreal Heart Institute (MHI), Université de Montréal, Montréal, Quebec, Canada.
| | - Steeve Provencher
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Quebec, Quebec, Canada; Department of medicine, Université Laval, 2325 rue de l'Universite, Montréal, Quebec G1V 0A6, Canada
| |
Collapse
|
20
|
Swietlik EM, Prapa M, Martin JM, Pandya D, Auckland K, Morrell NW, Gräf S. 'There and Back Again'-Forward Genetics and Reverse Phenotyping in Pulmonary Arterial Hypertension. Genes (Basel) 2020; 11:E1408. [PMID: 33256119 PMCID: PMC7760524 DOI: 10.3390/genes11121408] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/17/2020] [Accepted: 11/23/2020] [Indexed: 02/07/2023] Open
Abstract
Although the invention of right heart catheterisation in the 1950s enabled accurate clinical diagnosis of pulmonary arterial hypertension (PAH), it was not until 2000 when the landmark discovery of the causative role of bone morphogenetic protein receptor type II (BMPR2) mutations shed new light on the pathogenesis of PAH. Since then several genes have been discovered, which now account for around 25% of cases with the clinical diagnosis of idiopathic PAH. Despite the ongoing efforts, in the majority of patients the cause of the disease remains elusive, a phenomenon often referred to as "missing heritability". In this review, we discuss research approaches to uncover the genetic architecture of PAH starting with forward phenotyping, which in a research setting should focus on stable intermediate phenotypes, forward and reverse genetics, and finally reverse phenotyping. We then discuss potential sources of "missing heritability" and how functional genomics and multi-omics methods are employed to tackle this problem.
Collapse
Affiliation(s)
- Emilia M. Swietlik
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; (E.M.S.); (M.P.); (J.M.M.); (D.P.); (K.A.); (N.W.M.)
- Royal Papworth Hospital NHS Foundation Trust, Cambridge CB2 0AY, UK
- Addenbrooke’s Hospital NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - Matina Prapa
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; (E.M.S.); (M.P.); (J.M.M.); (D.P.); (K.A.); (N.W.M.)
- Addenbrooke’s Hospital NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - Jennifer M. Martin
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; (E.M.S.); (M.P.); (J.M.M.); (D.P.); (K.A.); (N.W.M.)
| | - Divya Pandya
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; (E.M.S.); (M.P.); (J.M.M.); (D.P.); (K.A.); (N.W.M.)
| | - Kathryn Auckland
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; (E.M.S.); (M.P.); (J.M.M.); (D.P.); (K.A.); (N.W.M.)
| | - Nicholas W. Morrell
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; (E.M.S.); (M.P.); (J.M.M.); (D.P.); (K.A.); (N.W.M.)
- Royal Papworth Hospital NHS Foundation Trust, Cambridge CB2 0AY, UK
- Addenbrooke’s Hospital NHS Foundation Trust, Cambridge CB2 0QQ, UK
- NIHR BioResource for Translational Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Stefan Gräf
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; (E.M.S.); (M.P.); (J.M.M.); (D.P.); (K.A.); (N.W.M.)
- NIHR BioResource for Translational Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
| |
Collapse
|
21
|
Le Ribeuz H, Capuano V, Girerd B, Humbert M, Montani D, Antigny F. Implication of Potassium Channels in the Pathophysiology of Pulmonary Arterial Hypertension. Biomolecules 2020; 10:biom10091261. [PMID: 32882918 PMCID: PMC7564204 DOI: 10.3390/biom10091261] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/24/2020] [Accepted: 08/27/2020] [Indexed: 02/06/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare and severe cardiopulmonary disease without curative treatments. PAH is a multifactorial disease that involves genetic predisposition, epigenetic factors, and environmental factors (drugs, toxins, viruses, hypoxia, and inflammation), which contribute to the initiation or development of irreversible remodeling of the pulmonary vessels. The recent identification of loss-of-function mutations in KCNK3 (KCNK3 or TASK-1) and ABCC8 (SUR1), or gain-of-function mutations in ABCC9 (SUR2), as well as polymorphisms in KCNA5 (Kv1.5), which encode two potassium (K+) channels and two K+ channel regulatory subunits, has revived the interest of ion channels in PAH. This review focuses on KCNK3, SUR1, SUR2, and Kv1.5 channels in pulmonary vasculature and discusses their pathophysiological contribution to and therapeutic potential in PAH.
Collapse
Affiliation(s)
- Hélène Le Ribeuz
- Faculté de Médecine, Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France; (H.L.R.); (V.C.); (B.G.); (M.H.); (D.M.)
- INSERM UMR_S 999, Hypertension pulmonaire, Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Assistance Publique—Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l’Hypertension Pulmonaire, Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre, France
| | - Véronique Capuano
- Faculté de Médecine, Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France; (H.L.R.); (V.C.); (B.G.); (M.H.); (D.M.)
- INSERM UMR_S 999, Hypertension pulmonaire, Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Assistance Publique—Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l’Hypertension Pulmonaire, Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre, France
| | - Barbara Girerd
- Faculté de Médecine, Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France; (H.L.R.); (V.C.); (B.G.); (M.H.); (D.M.)
- INSERM UMR_S 999, Hypertension pulmonaire, Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Assistance Publique—Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l’Hypertension Pulmonaire, Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre, France
| | - Marc Humbert
- Faculté de Médecine, Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France; (H.L.R.); (V.C.); (B.G.); (M.H.); (D.M.)
- INSERM UMR_S 999, Hypertension pulmonaire, Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Assistance Publique—Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l’Hypertension Pulmonaire, Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre, France
| | - David Montani
- Faculté de Médecine, Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France; (H.L.R.); (V.C.); (B.G.); (M.H.); (D.M.)
- INSERM UMR_S 999, Hypertension pulmonaire, Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Assistance Publique—Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l’Hypertension Pulmonaire, Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre, France
| | - Fabrice Antigny
- Faculté de Médecine, Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France; (H.L.R.); (V.C.); (B.G.); (M.H.); (D.M.)
- INSERM UMR_S 999, Hypertension pulmonaire, Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Assistance Publique—Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l’Hypertension Pulmonaire, Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre, France
- Correspondence: or ; Tel.: +33-1-40-94-22-99
| |
Collapse
|
22
|
Song S, Babicheva A, Zhao T, Ayon RJ, Rodriguez M, Rahimi S, Balistrieri F, Harrington A, Shyy JYJ, Thistlethwaite PA, Makino A, Yuan JXJ. Notch enhances Ca 2+ entry by activating calcium-sensing receptors and inhibiting voltage-gated K + channels. Am J Physiol Cell Physiol 2020; 318:C954-C968. [PMID: 32186932 DOI: 10.1152/ajpcell.00487.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The increase in cytosolic Ca2+ concentration ([Ca2+]cyt) and upregulation of calcium-sensing receptor (CaSR) and stromal interaction molecule 2 (STIM2) along with inhibition of voltage-gated K+ (KV) channels in pulmonary arterial smooth muscle cells (PASMC) have been implicated in the development of pulmonary arterial hypertension; however, the precise upstream mechanisms remain elusive. Activation of CaSR, a G protein-coupled receptor (GPCR), results in Ca2+ release from the endoplasmic/sarcoplasmic reticulum (ER/SR) and Ca2+ influx through receptor-operated and store-operated Ca2+ channels (SOC). Upon Ca2+ depletion from the SR, STIM forms clusters to mediate store-operated Ca2+ entry. Activity of KV channels, like KCNA5/KV1.5 and KCNA2/KV1.2, contributes to regulating membrane potential, and inhibition of KV channels results in membrane depolarization that increases [Ca2+]cyt by opening voltage-dependent Ca2+ channels. In this study, we show that activation of Notch by its ligand Jag-1 promotes the clustering of STIM2, and clustered STIM2 subsequently enhances the CaSR-induced Ca2+ influx through SOC channels. Extracellular Ca2+-mediated activation of CaSR increases [Ca2+]cyt in CASR-transfected HEK293 cells. Treatment of CASR-transfected cells with Jag-1 further enhances CaSR-mediated increase in [Ca2+]cyt. Moreover, CaSR-mediated increase in [Ca2+]cyt was significantly augmented in cells co-transfected with CASR and STIM2. CaSR activation results in STIM2 clustering in CASR/STIM2-cotransfected cells. Notch activation also induces significant clustering of STIM2. Furthermore, activation of Notch attenuates whole cell K+ currents in KCNA5- and KCNA2-transfected cells. Together, these results suggest that Notch activation enhances CaSR-mediated increases in [Ca2+]cyt by enhancing store-operated Ca2+ entry and inhibits KCNA5/KV1.5 and KCNA2/KV1.2, ultimately leading to voltage-activated Ca2+ entry.
Collapse
Affiliation(s)
| | - Aleksandra Babicheva
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Tengteng Zhao
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Ramon J Ayon
- Department of Physiology, University of Arizona, Tucson, Arizona
| | - Marisela Rodriguez
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Shamin Rahimi
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Francesca Balistrieri
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Angela Harrington
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - John Y-J Shyy
- Division of Cardiovascular Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| | | | - Ayako Makino
- Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, California
| | - Jason X-J Yuan
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| |
Collapse
|
23
|
Han L, Song N, Hu X, Zhu A, Wei X, Liu J, Yuan S, Mao W, Chen X. Inhibition of RELM-β prevents hypoxia-induced overproliferation of human pulmonary artery smooth muscle cells by reversing PLC-mediated KCNK3 decline. Life Sci 2020; 246:117419. [PMID: 32045592 DOI: 10.1016/j.lfs.2020.117419] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/29/2020] [Accepted: 02/07/2020] [Indexed: 02/09/2023]
Abstract
AIMS Although resistin-like molecule β (RELM-β) is involved in the pathological processes of various lung diseases, such as pulmonary inflammation, asthma and fibrosis, its potential roles in hypoxic pulmonary arterial hypertension (PAH) remain largely unknown. The study aims to investigate whether RELM-β contributes to hypoxia-induced excessive proliferation of human pulmonary artery smooth muscle cells (PASMCs) and to explore the potential mechanisms of this process. MAIN METHODS Human PASMCs were exposed to normoxia or hypoxia (1% O2) for 24 h. siRNA targeting RELM-β was transfected into cells. Protein levels of KCNK3, RELM-β, pSTAT3 and STAT3 were determined by immunoblotting. The translocation of NFATc2 and expression of KCNK3 were visualized by immunofluorescence. 5-ethynyl-2'-deoxyuridine assays and cell counting kit-8 assays were performed to assess the proliferation of PASMCs. KEY FINDINGS (1) Chronic hypoxia significantly decreased KCNK3 protein levels while upregulating RELM-β protein levels in human PASMCs, which was accompanied by excessive proliferation of cells. (2) RELM-β could promote human PASMCs proliferation and activate the STAT3/NFAT axis by downregulating KCNK3 protein under normoxia. (3) Inhibition of RELM-β expression effectively prevented KCNK3-mediated cell proliferation under hypoxia. (4) Phospholipase C (PLC) inhibitor U-73122 could not only prevent the hypoxia/RELM-β-induced decrease in KCNK3 protein, but also inhibit the enhanced cell viability caused by hypoxia/RELM-β. (5) Both hypoxia and RELM-β could downregulate membrane KCNK3 protein levels by enhancing endocytosis. SIGNIFICANCE RELM-β activation is responsible for hypoxia-induced excessive proliferation of human PASMCs. Interfering with RELM-β may alleviate the progression of hypoxic PAH by upregulating PLC-dependent KCNK3 expression.
Collapse
Affiliation(s)
- Linlin Han
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Nannan Song
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaomin Hu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Afang Zhu
- Department of Anesthesiology, Peking Union Medical College Hospital, CAMS&PUMC, Beijing, China
| | - Xin Wei
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jinmin Liu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shiying Yuan
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Weike Mao
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Xiangdong Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
24
|
Southgate L, Machado RD, Gräf S, Morrell NW. Molecular genetic framework underlying pulmonary arterial hypertension. Nat Rev Cardiol 2020; 17:85-95. [PMID: 31406341 DOI: 10.1038/s41569-019-0242-x] [Citation(s) in RCA: 181] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/12/2019] [Indexed: 02/02/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a rare, progressive disorder typified by occlusion of the pulmonary arterioles owing to endothelial dysfunction and uncontrolled proliferation of pulmonary artery smooth muscle cells and fibroblasts. Vascular occlusion can lead to increased pressure in the pulmonary arteries, often resulting in right ventricular failure with shortness of breath and syncope. Since the identification of BMPR2, which encodes a receptor in the transforming growth factor-β superfamily, the development of high-throughput sequencing approaches to identify novel causal genes has substantially advanced our understanding of the molecular genetics of PAH. In the past 6 years, additional pathways involved in PAH susceptibility have been described through the identification of deleterious genetic variants in potassium channels (KCNK3 and ABCC8) and transcription factors (TBX4 and SOX17), among others. Although familial PAH most often has an autosomal-dominant pattern of inheritance, cases of incomplete penetrance and evidence of genetic heterogeneity support a model of PAH as a Mendelian disorder with complex disease features. In this Review, we outline the latest advances in the detection of rare and common genetic variants underlying PAH susceptibility and disease progression. These findings have clinical implications for lung vascular function and can help to identify mechanistic pathways amenable to pharmacological intervention.
Collapse
Affiliation(s)
- Laura Southgate
- Molecular and Clinical Sciences Research Institute, St George's University of London, London, UK.,Department of Medical and Molecular Genetics, King's College London, London, UK
| | - Rajiv D Machado
- Molecular and Clinical Sciences Research Institute, St George's University of London, London, UK
| | - Stefan Gräf
- Department of Medicine, University of Cambridge, Cambridge, UK.,Department of Haematology, University of Cambridge, Cambridge, UK.,NIHR BioResource, Cambridge, UK
| | - Nicholas W Morrell
- Department of Medicine, University of Cambridge, Cambridge, UK. .,NIHR BioResource, Cambridge, UK.
| |
Collapse
|
25
|
Impact of Nutrition on Pulmonary Arterial Hypertension. Nutrients 2020; 12:nu12010169. [PMID: 31936113 PMCID: PMC7019983 DOI: 10.3390/nu12010169] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 12/26/2019] [Accepted: 01/03/2020] [Indexed: 02/07/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterized by sustained vasoconstriction, vascular remodeling, inflammation, and in situ thrombosis. Although there have been important advances in the knowledge of the pathophysiology of PAH, it remains a debilitating, limiting, and rapidly progressive disease. Vitamin D and iron deficiency are worldwide health problems of pandemic proportions. Notably, these nutritional alterations are largely more prevalent in PAH patients than in the general population and there are several pieces of evidence suggesting that they may trigger or aggravate disease progression. There are also several case reports associating scurvy, due to severe vitamin C deficiency, with PAH. Flavonoids such as quercetin, isoflavonoids such as genistein, and other dietary polyphenols including resveratrol slow the progression of the disease in animal models of PAH. Finally, the role of the gut microbiota and its interplay with the diet, host immune system, and energy metabolism is emerging in multiple cardiovascular diseases. The alteration of the gut microbiota has also been reported in animal models of PAH. It is thus possible that in the near future interventions targeting the nutritional status and the gut dysbiosis will improve the outcome of these patients.
Collapse
|
26
|
Ko EA, Kim YW, Lee D, Choi J, Kim S, Seo Y, Bang H, Kim JH, Ko JH. Expression of potassium channel genes predicts clinical outcome in lung cancer. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2019; 23:529-537. [PMID: 31680775 PMCID: PMC6819903 DOI: 10.4196/kjpp.2019.23.6.529] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 10/02/2019] [Accepted: 10/02/2019] [Indexed: 12/26/2022]
Abstract
Lung cancer is the most common cause of cancer deaths worldwide and several molecular signatures have been developed to predict survival in lung cancer. Increasing evidence suggests that proliferation and migration to promote tumor growth are associated with dysregulated ion channel expression. In this study, by analyzing high-throughput gene expression data, we identify the differentially expressed K+ channel genes in lung cancer. In total, we prioritize ten dysregulated K+ channel genes (5 up-regulated and 5 down-regulated genes, which were designated as K-10) in lung tumor tissue compared with normal tissue. A risk scoring system combined with the K-10 signature accurately predicts clinical outcome in lung cancer, which is independent of standard clinical and pathological prognostic factors including patient age, lymph node involvement, tumor size, and tumor grade. We further indicate that the K-10 potentially predicts clinical outcome in breast and colon cancers. Molecular signature discovered through K+ gene expression profiling may serve as a novel biomarker to assess the risk in lung cancer.
Collapse
Affiliation(s)
- Eun-A Ko
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, Nevada 89557, USA
| | - Young-Won Kim
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul 06974, Korea
| | - Donghee Lee
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul 06974, Korea
| | - Jeongyoon Choi
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul 06974, Korea
| | - Seongtae Kim
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul 06974, Korea
| | - Yelim Seo
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul 06974, Korea
| | - Hyoweon Bang
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul 06974, Korea
| | - Jung-Ha Kim
- Department of Family Medicine, Chung-Ang University Hospital, College of Medicine, Chung-Ang University, Seoul 06973, Korea
| | - Jae-Hong Ko
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul 06974, Korea
| |
Collapse
|
27
|
Mercurio V, Bianco A, Campi G, Cuomo A, Diab N, Mancini A, Parrella P, Petretta M, Hassoun PM, Bonaduce D. New Drugs, Therapeutic Strategies, and Future Direction for the Treatment of Pulmonary Arterial Hypertension. Curr Med Chem 2019; 26:2844-2864. [DOI: 10.2174/0929867325666180201095743] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 11/21/2017] [Accepted: 12/21/2017] [Indexed: 12/20/2022]
Abstract
Despite recent advances in Pulmonary Arterial Hypertension (PAH) treatment, this condition is still characterized by an extremely poor prognosis. In this review, we discuss the use of newly-approved drugs for PAH treatment with already known mechanisms of action (macitentan), innovative targets (riociguat and selexipag), and novel therapeutic approaches with initial up-front combination therapy. Secondly, we describe new potential signaling pathways and investigational drugs with promising role in the treatment of PAH.
Collapse
Affiliation(s)
- Valentina Mercurio
- Federico II University, Department of Translational Medical Sciences, Naples, Italy
| | - Anna Bianco
- Federico II University, Department of Translational Medical Sciences, Naples, Italy
| | - Giacomo Campi
- Federico II University, Department of Translational Medical Sciences, Naples, Italy
| | - Alessandra Cuomo
- Federico II University, Department of Translational Medical Sciences, Naples, Italy
| | - Nermin Diab
- University of Ottawa, Department of Medicine, Ottawa, ON, Canada
| | - Angela Mancini
- Federico II University, Department of Translational Medical Sciences, Naples, Italy
| | - Paolo Parrella
- Federico II University, Department of Translational Medical Sciences, Naples, Italy
| | - Mario Petretta
- Federico II University, Department of Translational Medical Sciences, Naples, Italy
| | - Paul M. Hassoun
- Johns Hopkins University, Division of Pulmonary and Critical Care Medicine, Baltimore, MD, United States
| | - Domenico Bonaduce
- Federico II University, Department of Translational Medical Sciences, Naples, Italy
| |
Collapse
|
28
|
Park MH, Park SI, Kim JH, Yu J, Lee EH, Seo SR, Jo SH. The acute effects of hydrocortisone on cardiac electrocardiography, action potentials, intracellular calcium, and contraction: The role of protein kinase C. Mol Cell Endocrinol 2019; 494:110488. [PMID: 31207272 DOI: 10.1016/j.mce.2019.110488] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 06/07/2019] [Accepted: 06/11/2019] [Indexed: 11/27/2022]
Abstract
Hydrocortisone exerts adverse effects on various organs, including the heart. This study investigated the still unclear effects of hydrocortisone on electrophysiological and biochemical aspects of cardiac excitation-contraction coupling. In guinea pigs' hearts, hydrocortisone administration reduced the QT interval of ECG and the action potential duration (APD). In guinea pig ventricular myocytes, hydrocortisone reduced contraction and Ca2+ transient amplitudes. These reductions and the effects on APD were prevented by pretreatment with the protein kinase C (PKC) inhibitor staurosporine. In an overexpression system of Xenopus oocytes, hydrocortisone increased hERG K+ currents and reduced Kv1.5 K+ currents; these effects were negated by pretreatment with staurosporine. Western blot analysis revealed dose- and time-dependent changes in PKCα/βII, PKCε, and PKCγ phosphorylation by hydrocortisone in guinea pig ventricular myocytes. Therefore, hydrocortisone can acutely affect cardiac excitation-contraction coupling, including ion channel activity, APD, ECG, Ca2+ transients, and contraction, possibly via biochemical changes in PKC.
Collapse
Affiliation(s)
- Mi-Hyeong Park
- Department of Physiology, Institute of Bioscience and Biotechnology, BK21 Plus Graduate Program, Kangwon National University College of Medicine, Chuncheon, 24341, South korea
| | - Seo-In Park
- Department of Physiology, Institute of Bioscience and Biotechnology, BK21 Plus Graduate Program, Kangwon National University College of Medicine, Chuncheon, 24341, South korea
| | - Jong-Hui Kim
- Department of Physiology, Institute of Bioscience and Biotechnology, BK21 Plus Graduate Program, Kangwon National University College of Medicine, Chuncheon, 24341, South korea
| | - Jing Yu
- Department of Physiology, Institute of Bioscience and Biotechnology, BK21 Plus Graduate Program, Kangwon National University College of Medicine, Chuncheon, 24341, South korea
| | - Eun Hye Lee
- Department of Molecular Bioscience, Institute of Bioscience and Biotechnology, Kangwon National University College of Biomedical Science, Chuncheon, 24341, South korea
| | - Su Ryeon Seo
- Department of Molecular Bioscience, Institute of Bioscience and Biotechnology, Kangwon National University College of Biomedical Science, Chuncheon, 24341, South korea.
| | - Su-Hyun Jo
- Department of Physiology, Institute of Bioscience and Biotechnology, BK21 Plus Graduate Program, Kangwon National University College of Medicine, Chuncheon, 24341, South korea.
| |
Collapse
|
29
|
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: 1.5] [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
|
30
|
Liu X, Mei M, Chen X, Lu Y, Dong X, Hu L, Hu X, Cheng G, Cao Y, Yang L, Zhou W. Identification of genetic factors underlying persistent pulmonary hypertension of newborns in a cohort of Chinese neonates. Respir Res 2019; 20:174. [PMID: 31382961 PMCID: PMC6683566 DOI: 10.1186/s12931-019-1148-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 07/29/2019] [Indexed: 01/02/2023] Open
Abstract
Background Persistent pulmonary hypertension of the newborn (PPHN) is a severe clinical problem among neonatal intensive care unit (NICU) patients. The genetic pathogenesis of PPHN is unclear. Only a few genetic polymorphisms have been identified in infants with PPHN. Our study aimed to investigate the potential genetic etiology of PPHN. Methods This study recruited PPHN patients admitted to the NICU of the Children’s Hospital of Fudan University from Jan 2016 to Dec 2017. Exome sequencing was performed for all patients. Variants in reported PPHN/pulmonary arterial hypertension (PAH)-related genes were assessed. Single nucleotide polymorphism (SNP) association and gene-level analyses were carried out in 74 PPHN cases and 115 non-PPHN controls with matched baseline characteristics. Results Among the patient cohort, 74 (64.3%) patients were late preterm and term infants (≥ 34 weeks gestation) and 41 (35.7%) were preterm infants (< 34 weeks gestation). Preterm infants with PPHN exhibited low birth weight and a high frequency of bronchopulmonary dysplasia, respiratory distress syndrome (RDS) and mortality. Nine patients (only one preterm infant) were identified as harboring genetic variants, including three with pathogenic/likely pathogenic variants in TBX4 and BMPR2 and six with variants of unknown significance in BMPR2, SMAD9, TGFB1, KCNA5 and TRPC6. Three SNPs (rs192759073, rs1047883 and rs2229589) in CPS1 and one SNP (rs1044008) in NOTCH3 were significantly associated with PPHN (p < 0.05). CPS1 and SMAD9 were identified as risk genes for PPHN (p < 0.05). Conclusions In this study, we identified genetic variants in PPHN patients, and we reported CPS1, NOTCH3 and SMAD9 as risk genes for late preterm and term PPHN in a single-center Chinese cohort. Our findings provide additional genetic evidence of the pathogenesis of PPHN and new insight into potential strategies for disease treatment. Electronic supplementary material The online version of this article (10.1186/s12931-019-1148-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xu Liu
- Clinical Genetic Center, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China.,Department of Neonatology, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China
| | - Mei Mei
- Department of Pulmonology, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China
| | - Xiang Chen
- Department of Neonatology, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China
| | - Yulan Lu
- Key Laboratory of Birth Defects, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China
| | - Xinran Dong
- Key Laboratory of Birth Defects, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China
| | - Liyuan Hu
- Department of Neonatology, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China
| | - Xiaojing Hu
- Department of Neonatology, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China
| | - Guoqiang Cheng
- Department of Neonatology, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China
| | - Yun Cao
- Department of Neonatology, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China
| | - Lin Yang
- Clinical Genetic Center, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China. .,Key Laboratory of Birth Defects, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China.
| | - Wenhao Zhou
- Clinical Genetic Center, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China. .,Department of Neonatology, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China. .,Key Laboratory of Birth Defects, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China.
| |
Collapse
|
31
|
Abstract
Background In genome-wide association studies (GWASs), meta-analysis has been widely used to improve statistical power by combining the results of different studies. Meta-analysis can detect phenotype associated variants that are failed to be detected in single studies. Especially, in biomedical sciences, meta-analysis is often necessary not only for improving statistical power, but also for reducing unavoidable limitation in data collection. As next-generation sequencing (NGS) technology has been developed, meta-analysis of rare variants is proceeding briskly along with meta-analysis of common variants in GWASs. However, meta-analysis on a single variant that is commonly used in common variant association test is improper for rare variants. A sparse signal of rare variant undermines the association signal and its large number causes multiple testing problem. To over-come these problems, we propose a meta-analysis method at the gene-level rather than variant level. Results Among many methods that have been developed, we used the unified quadratic tests (Q-tests); Q-test is more powerful than or as powerful as other tests such as Sequence Kernel Association Tests (SKAT). Since there are three different versions of Q-test (QTest1, QTest2, QTest3), each assumes different relationships among multiple rare variants, we extended them into meta-study accordingly. For meta-analysis, we consider two types of approaches, the one is to combine regression coefficients and the other is to combine test statistics from each single study. We extend the Q-test for meta-analysis, proposing Meta Quadratic Test (Meta-Qtest). Meta Q-test avoids the limitations of MetaSKAT. It does not only consider genetic heterogeneity among studies as MetaSKAT but also reflects diverse real situations; since we extend three different Q-tests into meta-analysis respectively, flexible Meta Q-test suggests way to deal with gene-level rare variant meta-analysis efficiently From the results of real data analysis of blood pressure trait, our meta-analysis could successfully discovered genes, KCNA5 and CABIN1 that are already well known for relevance with hypertension disease and they are not detected in MetaSKAT. Conclusion As exemplified by an application to T2D Genes projects data set, Meta-Qtest more effectively identified genes associated with hypertension disease than MetaSKAT did.
Collapse
Affiliation(s)
- Jieun Ka
- Department of Statistics, Seoul National University, Seoul, South Korea
| | - Jaehoon Lee
- Department of Statistics, Seoul National University, Seoul, South Korea
| | - Yongkang Kim
- Department of Statistics, Seoul National University, Seoul, South Korea
| | - Bermseok Oh
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul, South Korea
| | | | - Taesung Park
- Department of Statistics, Seoul National University, Seoul, South Korea. .,Interdisciplinary program in Bioinformatics, Seoul National University, Seoul, South Korea.
| |
Collapse
|
32
|
Mondéjar-Parreño G, Moral-Sanz J, Barreira B, De la Cruz A, Gonzalez T, Callejo M, Esquivel-Ruiz S, Morales-Cano D, Moreno L, Valenzuela C, Perez-Vizcaino F, Cogolludo A. Activation of K v 7 channels as a novel mechanism for NO/cGMP-induced pulmonary vasodilation. Br J Pharmacol 2019; 176:2131-2145. [PMID: 30883701 DOI: 10.1111/bph.14662] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 02/08/2019] [Accepted: 02/13/2019] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND AND PURPOSE The NO/cGMP pathway represents a major physiological signalling controlling tone in pulmonary arteries (PA), and drugs activating this pathway are used to treat pulmonary arterial hypertension. Kv channels expressed in PA smooth muscle cells (PASMCs) are key determinants of vascular tone. We aimed to analyse the contribution of Kv 1.5 and Kv 7 channels in the electrophysiological and vasodilating effects evoked by NO donors and the GC stimulator riociguat in PA. EXPERIMENTAL APPROACH Kv currents were recorded in isolated rat PASMCs using the patch-clamp technique. Vascular reactivity was assessed in a wire myograph. KEY RESULTS The NO donors diethylamine NONOate diethylammonium (DEA-NO) and sodium nitroprusside hyperpolarized the membrane potential and induced a bimodal effect on Kv currents (augmenting the current between -40 and -10 mV and decreasing it at more depolarized potentials). The hyperpolarization and the enhancement of the current were suppressed by Kv 7 channel inhibitors and by the GC inhibitor ODQ but preserved when Kv 1.5 channels were inhibited. Additionally, DEA-NO enhanced Kv 7.5 currents in COS7 cells expressing the KCNQ5 gene. Riociguat increased Kv currents at all potentials ≥-40 mV and induced membrane hyperpolarization. Both effects were prevented by Kv 7 inhibition. Likewise, PA relaxation induced by NO donors and riociguat was attenuated by Kv 7 inhibitors. CONCLUSIONS AND IMPLICATIONS NO donors and riociguat enhance Kv 7 currents, leading to PASMC hyperpolarization. This mechanism contributes to NO/cGMP-induced PA vasodilation. Our study identifies Kv 7 channels as a novel mechanism of action of vasodilator drugs used in the treatment of pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Gema Mondéjar-Parreño
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Javier Moral-Sanz
- Centres for Discovery Brain Sciences and Cardiovascular Science, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| | - Bianca Barreira
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Alicia De la Cruz
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Madrid, Spain.,CIBER Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Teresa Gonzalez
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Madrid, Spain.,CIBER Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.,Department of Biochemistry, School of Medicine, Universidad Autónoma de Madrid, Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Maria Callejo
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Sergio Esquivel-Ruiz
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Daniel Morales-Cano
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Laura Moreno
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Carmen Valenzuela
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Madrid, Spain.,CIBER Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Francisco Perez-Vizcaino
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Angel Cogolludo
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| |
Collapse
|
33
|
Cunningham KP, Holden RG, Escribano-Subias PM, Cogolludo A, Veale EL, Mathie A. Characterization and regulation of wild-type and mutant TASK-1 two pore domain potassium channels indicated in pulmonary arterial hypertension. J Physiol 2018; 597:1087-1101. [PMID: 30365877 PMCID: PMC6376074 DOI: 10.1113/jp277275] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 10/24/2018] [Indexed: 12/22/2022] Open
Abstract
Key points The TASK‐1 channel gene (KCNK3) has been identified as a possible disease‐causing gene in heritable pulmonary arterial hypertension (PAH). In the present study, we show that novel mutated TASK‐1 channels, seen in PAH patients, have a substantially reduced current compared to wild‐type TASK‐1 channels. These mutated TASK‐1 channels are located at the plasma membrane to the same degree as wild‐type TASK‐1 channels. ONO‐RS‐082 and alkaline pH 8.4 both activate TASK‐1 channels but do not recover current through mutant TASK‐1 channels. We show that the guanylate cyclase activator, riociguat, a novel treatment for PAH, enhances current through TASK‐1 channels but does not recover current through mutant TASK‐1 channels.
Abstract Pulmonary arterial hypertension (PAH) affects ∼15–50 people per million. KCNK3, the gene that encodes the two pore domain potassium channel TASK‐1 (K2P3.1), has been identified as a possible disease‐causing gene in heritable PAH. Recently, two new mutations have been identified in KCNK3 in PAH patients: G106R and L214R. The present study aimed to characterize the functional properties and regulation of wild‐type (WT) and mutated TASK‐1 channels and determine how these might contribute to PAH and its treatment. Currents through WT and mutated human TASK‐1 channels transiently expressed in tsA201 cells were measured using whole‐cell patch clamp electrophysiology. Localization of fluorescence‐tagged channels was visualized using confocal microscopy and quantified with in‐cell and on‐cell westerns. G106R or L214R mutated channels were located at the plasma membrane to the same degree as WT channels; however, their current was markedly reduced compared to WT TASK‐1 channels. Functional current through these mutated channels could not be restored using activators of WT TASK‐1 channels (pH 8.4, ONO‐RS‐082). The guanylate cyclase activator, riociguat, enhanced current through WT TASK‐1 channels; however, similar to the other activators investigated, riociguat did not have any effect on current through mutated TASK‐1 channels. Thus, novel mutations in TASK‐1 seen in PAH substantially alter the functional properties of these channels. Current through these channels could not be restored by activators of TASK‐1 channels. Riociguat enhancement of current through TASK‐1 channels could contribute to its therapeutic benefit in the treatment of PAH. The TASK‐1 channel gene (KCNK3) has been identified as a possible disease‐causing gene in heritable pulmonary arterial hypertension (PAH). In the present study, we show that novel mutated TASK‐1 channels, seen in PAH patients, have a substantially reduced current compared to wild‐type TASK‐1 channels. These mutated TASK‐1 channels are located at the plasma membrane to the same degree as wild‐type TASK‐1 channels. ONO‐RS‐082 and alkaline pH 8.4 both activate TASK‐1 channels but do not recover current through mutant TASK‐1 channels. We show that the guanylate cyclase activator, riociguat, a novel treatment for PAH, enhances current through TASK‐1 channels but does not recover current through mutant TASK‐1 channels.
Collapse
Affiliation(s)
- Kevin P Cunningham
- Medway School of Pharmacy, University of Kent and University of Greenwich, Chatham Maritime, Kent, UK
| | - Robyn G Holden
- Medway School of Pharmacy, University of Kent and University of Greenwich, Chatham Maritime, Kent, UK
| | | | - Angel Cogolludo
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Emma L Veale
- Medway School of Pharmacy, University of Kent and University of Greenwich, Chatham Maritime, Kent, UK
| | - Alistair Mathie
- Medway School of Pharmacy, University of Kent and University of Greenwich, Chatham Maritime, Kent, UK
| |
Collapse
|
34
|
Lambert M, Capuano V, Olschewski A, Sabourin J, Nagaraj C, Girerd B, Weatherald J, Humbert M, Antigny F. Ion Channels in Pulmonary Hypertension: A Therapeutic Interest? Int J Mol Sci 2018; 19:ijms19103162. [PMID: 30322215 PMCID: PMC6214085 DOI: 10.3390/ijms19103162] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 10/05/2018] [Accepted: 10/08/2018] [Indexed: 12/25/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a multifactorial and severe disease without curative therapies. PAH pathobiology involves altered pulmonary arterial tone, endothelial dysfunction, distal pulmonary vessel remodeling, and inflammation, which could all depend on ion channel activities (K⁺, Ca2+, Na⁺ and Cl-). This review focuses on ion channels in the pulmonary vasculature and discusses their pathophysiological contribution to PAH as well as their therapeutic potential in PAH.
Collapse
Affiliation(s)
- Mélanie Lambert
- Univ. Paris-Sud, Faculté de Médecine, 94270 Kremlin-Bicêtre, France.
- AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, 94270 Le Kremlin-Bicêtre, France.
- UMRS 999, INSERM and Univ. Paris⁻Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, 92350 Le Plessis Robinson, France.
| | - Véronique Capuano
- Univ. Paris-Sud, Faculté de Médecine, 94270 Kremlin-Bicêtre, France.
- AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, 94270 Le Kremlin-Bicêtre, France.
- UMRS 999, INSERM and Univ. Paris⁻Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, 92350 Le Plessis Robinson, France.
| | - Andrea Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Stiftingtalstrasse 24, Graz 8010, Austria.
- Department of Physiology, Medical University Graz, Neue Stiftingtalstraße 6, Graz 8010, Austria.
| | - Jessica Sabourin
- Signalisation et Physiopathologie Cardiovasculaire, UMRS 1180, Univ. Paris-Sud, INSERM, Université Paris-Saclay, 92296 Châtenay-Malabry, France.
| | - Chandran Nagaraj
- Ludwig Boltzmann Institute for Lung Vascular Research, Stiftingtalstrasse 24, Graz 8010, Austria.
| | - Barbara Girerd
- Univ. Paris-Sud, Faculté de Médecine, 94270 Kremlin-Bicêtre, France.
- AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, 94270 Le Kremlin-Bicêtre, France.
- UMRS 999, INSERM and Univ. Paris⁻Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, 92350 Le Plessis Robinson, France.
| | - Jason Weatherald
- Univ. Paris-Sud, Faculté de Médecine, 94270 Kremlin-Bicêtre, France.
- AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, 94270 Le Kremlin-Bicêtre, France.
- UMRS 999, INSERM and Univ. Paris⁻Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, 92350 Le Plessis Robinson, France.
- Division of Respirology, Department of Medicine, University of Calgary, Calgary, AB T1Y 6J4, Canada.
- Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB T1Y 6J4, Canada.
| | - Marc Humbert
- Univ. Paris-Sud, Faculté de Médecine, 94270 Kremlin-Bicêtre, France.
- AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, 94270 Le Kremlin-Bicêtre, France.
- UMRS 999, INSERM and Univ. Paris⁻Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, 92350 Le Plessis Robinson, France.
| | - Fabrice Antigny
- Univ. Paris-Sud, Faculté de Médecine, 94270 Kremlin-Bicêtre, France.
- AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, 94270 Le Kremlin-Bicêtre, France.
- UMRS 999, INSERM and Univ. Paris⁻Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, 92350 Le Plessis Robinson, France.
| |
Collapse
|
35
|
Rowan SC, Piouceau L, Cornwell J, Li L, McLoughlin P. EXPRESS: Gremlin1 blocks vascular endothelial growth factor signalling in the pulmonary microvascular endothelium. Pulm Circ 2018; 10:2045894018807205. [PMID: 30284507 PMCID: PMC7066471 DOI: 10.1177/2045894018807205] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 09/20/2018] [Indexed: 11/15/2022] Open
Abstract
The bone morphogenetic protein (BMP) antagonist gremlin 1 plays a central role in the pathogenesis of hypoxic pulmonary hypertension (HPH). Recently, non-canonical functions of gremlin 1 have been identified, including specific binding to the vascular endothelial growth factor receptor-2 (VEGFR2). We tested the hypothesis that gremlin 1 modulates VEGFR2 signaling in the pulmonary microvascular endothelium. We examined the effect of gremlin 1 haploinsufficiency on the expression of VEGF responsive genes and proteins in the hypoxic (10% O2) murine lung in vivo. Using human microvascular endothelial cells in vitro we examined the effect of gremlin 1 on VEGF signaling. Gremlin 1 haploinsufficiency (Grem1+/–) attenuated the hypoxia-induced increase in gremlin 1 observed in the wild-type mouse lung. Reduced gremlin 1 expression in hypoxic Grem1+/– mice restored VEGFR2 expression and endothelial nitric oxide synthase (eNOS) expression and activity to normoxic values. Recombinant monomeric gremlin 1 inhibited VEGFA-induced VEGFR2 activation, downstream signaling, and VEGF-induced increases in Bcl-2, cell number, and the anti-apoptotic effect of VEGFA in vitro. These results show that the monomeric form of gremlin 1 acts as an antagonist of VEGFR2 activation in the pulmonary microvascular endothelium. Given the previous demonstration that inhibition of VEGFR2 causes marked worsening of HPH, our results suggest that increased gremlin 1 in the hypoxic lung, in addition to blocking BMP receptor type-2 (BMPR2) signaling, contributes importantly to the development of PH by a non-canonical VEGFR2 blocking activity.
Collapse
Affiliation(s)
- Simon C. Rowan
- UCD School of Medicine and Conway Institute,
University
College Dublin, Dublin, Ireland
| | - Lucie Piouceau
- UCD School of Medicine and Conway Institute,
University
College Dublin, Dublin, Ireland
| | - Joanna Cornwell
- UCD School of Medicine and Conway Institute,
University
College Dublin, Dublin, Ireland
| | - Lili Li
- UCD School of Medicine and Conway Institute,
University
College Dublin, Dublin, Ireland
| | - Paul McLoughlin
- UCD School of Medicine and Conway Institute,
University
College Dublin, Dublin, Ireland
| |
Collapse
|
36
|
Abbasi Y, Jabbari J, Jabbari R, Glinge C, Izadyar S, Spiekerkoetter E, Zamanian RT, Carlsen J, Tfelt‐Hansen J. Exome data clouds the pathogenicity of genetic variants in Pulmonary Arterial Hypertension. Mol Genet Genomic Med 2018; 6:835-844. [PMID: 30084161 PMCID: PMC6160702 DOI: 10.1002/mgg3.452] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 04/25/2018] [Accepted: 06/03/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND We aimed to provide a set of previously reported PAH-associated missense and nonsense variants, and evaluate the pathogenicity of those variants. METHODS The Human Gene Mutation Database, PubMed, and Google Scholar were searched for previously reported PAH-associated genes and variants. Thereafter, both exome sequencing project and exome aggregation consortium as background population searched for previously reported PAH-associated missense and nonsense variants. The pathogenicity of previously reported PAH-associated missense variants evaluated by using four in silico prediction tools. RESULTS In total, 14 PAH-associated genes and 180 missense and nonsense variants were gathered. The BMPR2, the most frequent reported gene, encompasses 135 of 180 missense and nonsense variants. The exome sequencing project comprised 9, and the exome aggregation consortium counted 25 of 180 PAH-associated missense and nonsense variants. The TOPBP1 and ENG genes are unlikely to be the monogenic cause of PAH pathogenesis based on allele frequency in background population and prediction analysis. CONCLUSION This is the first evaluation of previously reported PAH-associated missense and nonsense variants. The BMPR2 identified as the major gene out of 14 PAH-associated genes. Based on findings, the ENG and TOPBP1 gene are not likely to be the monogenic cause of PAH.
Collapse
Affiliation(s)
- Yeganeh Abbasi
- Heart CentreDepartment of CardiologyCopenhagen University Hospital, RigshospitaletCopenhagenDenmark
- Department of CardiologySection for Pulmonary Hypertension and Right Heart FailureCopenhagen University Hospital, RigshospitaletCopenhagenDenmark
| | | | - Reza Jabbari
- Heart CentreDepartment of CardiologyCopenhagen University Hospital, RigshospitaletCopenhagenDenmark
- Department of CardiologySection for Pulmonary Hypertension and Right Heart FailureCopenhagen University Hospital, RigshospitaletCopenhagenDenmark
| | - Charlotte Glinge
- Heart CentreDepartment of CardiologyCopenhagen University Hospital, RigshospitaletCopenhagenDenmark
| | - Seyed Bahador Izadyar
- Heart CentreDepartment of CardiologyCopenhagen University Hospital, RigshospitaletCopenhagenDenmark
| | - Edda Spiekerkoetter
- Division of Pulmonary and Critical CareStanford University School of MedicineCalifornia
| | - Roham T. Zamanian
- Division of Pulmonary and Critical CareStanford University School of MedicineCalifornia
| | - Jørn Carlsen
- Heart CentreDepartment of CardiologyCopenhagen University Hospital, RigshospitaletCopenhagenDenmark
- Department of CardiologySection for Pulmonary Hypertension and Right Heart FailureCopenhagen University Hospital, RigshospitaletCopenhagenDenmark
| | - Jacob Tfelt‐Hansen
- Heart CentreDepartment of CardiologyCopenhagen University Hospital, RigshospitaletCopenhagenDenmark
- Department of CardiologySection for Pulmonary Hypertension and Right Heart FailureCopenhagen University Hospital, RigshospitaletCopenhagenDenmark
- Department of Forensic MedicineFaculty of Medical SciencesUniversity of CopenhagenDenmark
| |
Collapse
|
37
|
Yuan SM. Pulmonary artery hypertension in childhood: The transforming growth factor-β superfamily-related genes. Pediatr Neonatol 2018; 59:112-119. [PMID: 28967497 DOI: 10.1016/j.pedneo.2016.12.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 10/20/2016] [Accepted: 12/05/2016] [Indexed: 01/08/2023] Open
Abstract
Pulmonary artery hypertension (PAH) is very rare in childhood, and it can be divided into heritable, idiopathic drug- and toxin-induced and other disease (connective tissue disease, human immunodeficiency virus infection, portal hypertension, congenital heart disease, or schistosomiasis)-associated types. PAH could not be interpreted solely by pathophysiological theories. The impact of the transforming growth factor-β superfamily-related genes on the development of PAH in children remains to be clarified. Pertinent literature on the transforming growth factor-β superfamily-related genes in relation to PAH in children published after the year 2000 was reviewed and analyzed. Bone morphogenetic protein receptor type II gene mutation promotes cell division or prevents cell death, resulting in an overgrowth of cells in small arteries throughout the lungs. About 20% of individuals with a bone morphogenetic protein receptor type II gene mutation develop symptomatic PAH. In heritable PAH, bone morphogenetic protein receptor type II mutations may be absent; while mutations of other genes, such as type I receptor activin receptor-like kinase 1 and the type III receptor endoglin (both associated with hereditary hemorrhagic telangiectasia), caveolin-1 and KCNK3, the gene encoding potassium channel subfamily K, member 3, can be detected, instead. Gene mutations, environmental changes and acquired adjustment, etc. may explain the development of PAH. The researches on PAH rat model and familial PAH members may facilitate the elucidations of the mechanisms and further provide theories for prophylaxis and treatment of PAH.
Collapse
Affiliation(s)
- Shi-Min Yuan
- Department of Cardiothoracic Surgery, The First Hospital of Putian, Teaching Hospital, Fujian Medical University, Putian, Fujian Province, People's Republic of China.
| |
Collapse
|
38
|
Chan SY, Rubin LJ. Metabolic dysfunction in pulmonary hypertension: from basic science to clinical practice. Eur Respir Rev 2017; 26:26/146/170094. [PMID: 29263174 PMCID: PMC5842433 DOI: 10.1183/16000617.0094-2017] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 10/28/2017] [Indexed: 01/29/2023] Open
Abstract
Pulmonary hypertension (PH) is an often-fatal vascular disease of unclear molecular origins. The pulmonary vascular remodelling which occurs in PH is characterised by elevated vasomotor tone and a pro-proliferative state, ultimately leading to right ventricular dysfunction and heart failure. Guided in many respects by prior evidence from cancer biology, recent investigations have identified metabolic aberrations as crucial components of the disease process in both the pulmonary vessels and the right ventricle. Given the need for improved diagnostic and therapeutic options for PH, the development or repurposing of metabolic tracers and medications could provide an effective avenue for preventing or even reversing disease progression. In this review, we describe the metabolic mechanisms that are known to be dysregulated in PH; we explore the advancing diagnostic testing and imaging modalities that are being developed to improve diagnostic capability for this disease; and we discuss emerging drugs for PH which target these metabolic pathways. Understanding metabolic pathways in PH provides opportunities for improved diagnostic and therapeutic optionshttp://ow.ly/pFQb30guez6
Collapse
Affiliation(s)
- Stephen Y Chan
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Dept of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Lewis J Rubin
- University of California, San Diego School of Medicine, La Jolla, CA, USA
| |
Collapse
|
39
|
Tang H, Desai AA, Yuan JXJ. Genetic Insights into Pulmonary Arterial Hypertension. Application of Whole-Exome Sequencing to the Study of Pathogenic Mechanisms. Am J Respir Crit Care Med 2017; 194:393-7. [PMID: 27525458 DOI: 10.1164/rccm.201603-0577ed] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Haiyang Tang
- 1 Department of Medicine University of Arizona College of Medicine Tucson, Arizona
| | - Ankit A Desai
- 1 Department of Medicine University of Arizona College of Medicine Tucson, Arizona
| | - Jason X-J Yuan
- 1 Department of Medicine University of Arizona College of Medicine Tucson, Arizona
| |
Collapse
|
40
|
Olschewski A, Veale EL, Nagy BM, Nagaraj C, Kwapiszewska G, Antigny F, Lambert M, Humbert M, Czirják G, Enyedi P, Mathie A. TASK-1 (KCNK3) channels in the lung: from cell biology to clinical implications. Eur Respir J 2017; 50:50/5/1700754. [PMID: 29122916 DOI: 10.1183/13993003.00754-2017] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 08/05/2017] [Indexed: 12/18/2022]
Abstract
TWIK-related acid-sensitive potassium channel 1 (TASK-1 encoded by KCNK3) belongs to the family of two-pore domain potassium channels. This gene subfamily is constitutively active at physiological resting membrane potentials in excitable cells, including smooth muscle cells, and has been particularly linked to the human pulmonary circulation. TASK-1 channels are sensitive to a wide array of physiological and pharmacological mediators that affect their activity such as unsaturated fatty acids, extracellular pH, hypoxia, anaesthetics and intracellular signalling pathways. Recent studies show that modulation of TASK-1 channels, either directly or indirectly by targeting their regulatory mechanisms, has the potential to control pulmonary arterial tone in humans. Furthermore, mutations in KCNK3 have been identified as a rare cause of both familial and idiopathic pulmonary arterial hypertension. This review summarises our current state of knowledge of the functional role of TASK-1 channels in the pulmonary circulation in health and disease, with special emphasis on current advancements in the field.
Collapse
Affiliation(s)
- Andrea Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research Graz, Graz, Austria .,Institute of Physiology, Medical University of Graz, Graz, Austria
| | - Emma L Veale
- Medway School of Pharmacy, University of Kent, Central Avenue, Chatham Maritime, UK
| | - Bence M Nagy
- Institute of Physiology, Medical University of Graz, Graz, Austria
| | - Chandran Nagaraj
- Ludwig Boltzmann Institute for Lung Vascular Research Graz, Graz, Austria.,Institute of Physiology, Medical University of Graz, Graz, Austria
| | - Grazyna Kwapiszewska
- Ludwig Boltzmann Institute for Lung Vascular Research Graz, Graz, Austria.,Institute of Physiology, Medical University of Graz, Graz, Austria
| | - Fabrice Antigny
- Univ. Paris-Sud, Faculté de Médecine, Kremlin-Bicêtre, France.,AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, Le Kremlin-Bicêtre, France.,UMRS 999, INSERM and Univ. Paris-Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, Le Plessis Robinson, France
| | - Mélanie Lambert
- Univ. Paris-Sud, Faculté de Médecine, Kremlin-Bicêtre, France.,AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, Le Kremlin-Bicêtre, France.,UMRS 999, INSERM and Univ. Paris-Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, Le Plessis Robinson, France
| | - Marc Humbert
- Univ. Paris-Sud, Faculté de Médecine, Kremlin-Bicêtre, France.,AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, Le Kremlin-Bicêtre, France.,UMRS 999, INSERM and Univ. Paris-Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, Le Plessis Robinson, France
| | - Gábor Czirják
- Dept of Physiology, Semmelweis University, Budapest, Hungary
| | - Péter Enyedi
- Dept of Physiology, Semmelweis University, Budapest, Hungary
| | - Alistair Mathie
- Medway School of Pharmacy, University of Kent, Central Avenue, Chatham Maritime, UK
| |
Collapse
|
41
|
Al-Owais MM, Hettiarachchi NT, Boyle JP, Scragg JL, Elies J, Dallas ML, Lippiat JD, Steele DS, Peers C. Multiple mechanisms mediating carbon monoxide inhibition of the voltage-gated K + channel Kv1.5. Cell Death Dis 2017; 8:e3163. [PMID: 29095440 PMCID: PMC5775415 DOI: 10.1038/cddis.2017.568] [Citation(s) in RCA: 14] [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: 07/25/2017] [Revised: 09/19/2017] [Accepted: 09/21/2017] [Indexed: 12/13/2022]
Abstract
The voltage-gated K+ channel has key roles in the vasculature and in atrial excitability and contributes to apoptosis in various tissues. In this study, we have explored its regulation by carbon monoxide (CO), a product of the cytoprotective heme oxygenase enzymes, and a recognized toxin. CO inhibited recombinant Kv1.5 expressed in HEK293 cells in a concentration-dependent manner that involved multiple signalling pathways. CO inhibition was partially reversed by superoxide dismutase mimetics and by suppression of mitochondrial reactive oxygen species. CO also elevated intracellular nitric oxide (NO) levels. Prevention of NO formation also partially reversed CO inhibition of Kv1.5, as did inhibition of soluble guanylyl cyclase. CO also elevated intracellular peroxynitrite levels, and a peroxynitrite scavenger markedly attenuated the ability of CO to inhibit Kv1.5. CO caused nitrosylation of Kv1.5, an effect that was also observed in C331A and C346A mutant forms of the channel, which had previously been suggested as nitrosylation sites within Kv1.5. Augmentation of Kv1.5 via exposure to hydrogen peroxide was fully reversed by CO. Native Kv1.5 recorded in HL-1 murine atrial cells was also inhibited by CO. Action potentials recorded in HL-1 cells were increased in amplitude and duration by CO, an effect mimicked and occluded by pharmacological inhibition of Kv1.5. Our data indicate that Kv1.5 is a target for modulation by CO via multiple mechanisms. This regulation has important implications for diverse cellular functions, including excitability, contractility and apoptosis.
Collapse
Affiliation(s)
- Moza M Al-Owais
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Nishani T Hettiarachchi
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - John P Boyle
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Jason L Scragg
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Jacobo Elies
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Mark L Dallas
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Jon D Lippiat
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Derek S Steele
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Chris Peers
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
42
|
Effects of Mahuang ( Herba Ephedra Sinica ) and Wuweizi ( Fructus Schisandrae Chinensis ) medicated serum on chemotactic migration of alveolar macrophages and inters regions macrophages in rats. J TRADIT CHIN MED 2017. [DOI: 10.1016/s0254-6272(17)30313-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
43
|
Bohnen MS, Roman-Campos D, Terrenoire C, Jnani J, Sampson KJ, Chung WK, Kass RS. The Impact of Heterozygous KCNK3 Mutations Associated With Pulmonary Arterial Hypertension on Channel Function and Pharmacological Recovery. J Am Heart Assoc 2017; 6:JAHA.117.006465. [PMID: 28889099 PMCID: PMC5634293 DOI: 10.1161/jaha.117.006465] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Heterozygous loss of function mutations in the KCNK3 gene cause hereditary pulmonary arterial hypertension (PAH). KCNK3 encodes an acid-sensitive potassium channel, which contributes to the resting potential of human pulmonary artery smooth muscle cells. KCNK3 is widely expressed in the body, and dimerizes with other KCNK3 subunits, or the closely related, acid-sensitive KCNK9 channel. METHODS AND RESULTS We engineered homomeric and heterodimeric mutant and nonmutant KCNK3 channels associated with PAH. Using whole-cell patch-clamp electrophysiology in human pulmonary artery smooth muscle and COS7 cell lines, we determined that homomeric and heterodimeric mutant channels in heterozygous KCNK3 conditions lead to mutation-specific severity of channel dysfunction. Both wildtype and mutant KCNK3 channels were activated by ONO-RS-082 (10 μmol/L), causing cell hyperpolarization. We observed robust gene expression of KCNK3 in healthy and familial PAH patient lungs, but no quantifiable expression of KCNK9, and demonstrated in functional studies that KCNK9 minimizes the impact of select KCNK3 mutations when the 2 channel subunits co-assemble. CONCLUSIONS Heterozygous KCNK3 mutations in PAH lead to variable loss of channel function via distinct mechanisms. Homomeric and heterodimeric mutant KCNK3 channels represent novel therapeutic substrates in PAH. Pharmacological and pH-dependent activation of wildtype and mutant KCNK3 channels in pulmonary artery smooth muscle cells leads to membrane hyperpolarization. Co-assembly of KCNK3 with KCNK9 subunits may provide protection against KCNK3 loss of function in tissues where both KCNK9 and KCNK3 are expressed, contributing to the lung-specific phenotype observed clinically in patients with PAH because of KCNK3 mutations.
Collapse
Affiliation(s)
- Michael S Bohnen
- Department of Pharmacology, College of Physicians and Surgeons, Columbia University, New York, NY
| | | | - Cecile Terrenoire
- Department of Biophysics, Paulista School of Medicine, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - Jack Jnani
- Department of Pharmacology, College of Physicians and Surgeons, Columbia University, New York, NY
| | - Kevin J Sampson
- Department of Pharmacology, College of Physicians and Surgeons, Columbia University, New York, NY
| | - Wendy K Chung
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY
| | - Robert S Kass
- Department of Pharmacology, College of Physicians and Surgeons, Columbia University, New York, NY
| |
Collapse
|
44
|
|
45
|
Effects of equol on multiple K+ channels stably expressed in HEK 293 cells. PLoS One 2017; 12:e0183708. [PMID: 28832658 PMCID: PMC5568406 DOI: 10.1371/journal.pone.0183708] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 08/09/2017] [Indexed: 12/11/2022] Open
Abstract
The present study investigated the effects of equol on cardiovascular K+ channel currents. The cardiovascular K+ channel currents were determined in HEK 293 cells stably expressing cloned differential cardiovascular K+ channels with conventional whole-cell patch voltage-clamp technique. We found that equol inhibited hKv1.5 (IC50: 15.3 μM), hKv4.3 (IC50: 29.2 μM and 11.9 μM for hKv4.3 peak current and charge area, respectively), IKs (IC50: 24.7 μM) and IhERG (IC50: 31.6 and 56.5 μM for IhERG.tail and IhERG.step, respectively), but not hKir2.1 current, in a concentration-dependent manner. Interestingly, equol increased BKCa current with an EC50 of 0.1 μM. It had no significant effect on guinea pig ventricular action potentials at concentrations of ≤3 μM. These results demonstrate that equol inhibits several cardiac K+ currents at relatively high concentrations, whereas it increases BKCa current at very low concentrations, suggesting that equol is a safe drug candidate for treating patients with cerebral vascular disorders.
Collapse
|
46
|
Tanner MR, Tajhya RB, Huq R, Gehrmann EJ, Rodarte KE, Atik MA, Norton RS, Pennington MW, Beeton C. Prolonged immunomodulation in inflammatory arthritis using the selective Kv1.3 channel blocker HsTX1[R14A] and its PEGylated analog. Clin Immunol 2017; 180:45-57. [PMID: 28389388 PMCID: PMC5484050 DOI: 10.1016/j.clim.2017.03.014] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 02/27/2017] [Accepted: 03/28/2017] [Indexed: 12/31/2022]
Abstract
Effector memory T lymphocytes (TEM cells) that lack expression of CCR7 are major drivers of inflammation in a number of autoimmune diseases, including multiple sclerosis and rheumatoid arthritis. The Kv1.3 potassium channel is a key regulator of CCR7- TEM cell activation. Blocking Kv1.3 inhibits TEM cell activation and attenuates inflammation in autoimmunity, and as such, Kv1.3 has emerged as a promising target for the treatment of TEM cell-mediated autoimmune diseases. The scorpion venom-derived peptide HsTX1 and its analog HsTX1[R14A] are potent Kv1.3 blockers and HsTX1[R14A] is selective for Kv1.3 over closely-related Kv1 channels. PEGylation of HsTX1[R14A] to create a Kv1.3 blocker with a long circulating half-life reduced its affinity but not its selectivity for Kv1.3, dramatically reduced its adsorption to inert surfaces, and enhanced its circulating half-life in rats. PEG-HsTX1[R14A] is equipotent to HsTX1[R14A] in preferential inhibition of human and rat CCR7- TEM cell proliferation, leaving CCR7+ naïve and central memory T cells able to proliferate. It reduced inflammation in an active delayed-type hypersensitivity model and in the pristane-induced arthritis (PIA) model of rheumatoid arthritis (RA). Importantly, a single subcutaneous dose of PEG-HsTX1[R14A] reduced inflammation in PIA for a longer period of time than the non-PEGylated HsTX1[R14A]. Together, these data indicate that HsTX1[R14A] and PEG-HsTX1[R14A] are effective in a model of RA and are therefore potential therapeutics for TEM cell-mediated autoimmune diseases. PEG-HsTX1[R14A] has the additional advantages of reduced non-specific adsorption to inert surfaces and enhanced circulating half-life.
Collapse
Affiliation(s)
- Mark R Tanner
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX 77030, USA; Interdepartmental Graduate Program in Translational Biology & Molecular Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Rajeev B Tajhya
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Redwan Huq
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Elizabeth J Gehrmann
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kathia E Rodarte
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mustafa A Atik
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Raymond S Norton
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | | | - Christine Beeton
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX 77030, USA; Biology of Inflammation Center and Center for Drug Discovery, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
47
|
Zimmer J, Takahashi T, Hofmann AD, Puri P. Downregulation of KCNQ5 expression in the rat pulmonary vasculature of nitrofen-induced congenital diaphragmatic hernia. J Pediatr Surg 2017; 52:702-705. [PMID: 28189443 DOI: 10.1016/j.jpedsurg.2017.01.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 01/23/2017] [Indexed: 12/17/2022]
Abstract
PURPOSE Pulmonary hypertension (PH) is a common complication of congenital diaphragmatic hernia (CDH). Voltage-gated potassium channels KCNQ1, KCNQ4, and KCNQ5 are expressed by rodent pulmonary artery smooth muscle cells, contributing to their vascular tone. We hypothesized that KCNQ1, KCNQ4, and KCNQ5 expression is altered in the pulmonary vasculature of nitrofen-induced CDH rats. METHODS After ethical approval (REC913b), time-pregnant rats received nitrofen or vehicle on gestational day (D)9. D21 fetuses were divided into CDH and control group (n=22). QRT-PCR and western blotting were performed to determine gene and protein expression of KCNQ1, KCNQ4, and KCNQ5. Confocal microscopy was used to detect these proteins in the pulmonary vasculature. RESULTS Relative mRNA level of KCNQ5 (p=0.025) was significantly downregulated in CDH lungs compared to controls. KCNQ1 (p=0.052) and KCNQ4 (p=0.574) expression was not altered. Western blotting confirmed the decreased pulmonary KCNQ5 protein expression in CDH lungs. Confocal-microscopy detected a markedly diminished KCNQ5 expression in pulmonary vasculature of CDH fetuses. CONCLUSIONS Downregulated pulmonary expression of KCNQ5 in CDH lungs suggests that this potassium channel may play an important role in the development of PH in this model. KCNQ5 channel activator drugs may be a potential therapeutic target for the treatment of PH in CDH. LEVEL OF EVIDENCE 2b (Centre for Evidence-Based Medicine, Oxford).
Collapse
Affiliation(s)
- Julia Zimmer
- National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Gate 5, Dublin 12, Dublin, Ireland
| | - Toshiaki Takahashi
- National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Gate 5, Dublin 12, Dublin, Ireland
| | - Alejandro D Hofmann
- Department of Pediatric Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Prem Puri
- National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Gate 5, Dublin 12, Dublin, Ireland; School of Medicine and Medical Science and Conway Institute of Biomedical Research, University College Dublin, Belfield, Dublin 4, Dublin, Ireland.
| |
Collapse
|
48
|
Ohanyan V, Yin L, Bardakjian R, Kolz C, Enrick M, Hakobyan T, Luli J, Graham K, Khayata M, Logan S, Kmetz J, Chilian WM. Kv1.3 channels facilitate the connection between metabolism and blood flow in the heart. Microcirculation 2017; 24:10.1111/micc.12334. [PMID: 28504408 PMCID: PMC5433265 DOI: 10.1111/micc.12334] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Revised: 10/23/2016] [Accepted: 11/01/2016] [Indexed: 12/17/2022]
Abstract
The connection between metabolism and flow in the heart, metabolic dilation, is essential for cardiac function. We recently found redox-sensitive Kv1.5 channels play a role in coronary metabolic dilation; however, more than one ion channel likely plays a role in this process as animals null for these channels still showed limited coronary metabolic dilation. Accordingly, we examined the role of another Kv1 family channel, the energetically linked Kv1.3 channel, in coronary metabolic dilation. We measured myocardial blood flow (contrast echocardiography) during norepinephrine-induced increases in cardiac work (heart rate x mean arterial pressure) in WT, WT mice given correolide (preferential Kv1.3 antagonist), and Kv1.3-null mice (Kv1.3-/- ). We also measured relaxation of isolated small arteries mounted in a myograph. During increased cardiac work, myocardial blood flow was attenuated in Kv1.3-/- and in correolide-treated mice. In isolated vessels from Kv1.3-/- mice, relaxation to H2 O2 was impaired (vs WT), but responses to adenosine and acetylcholine were equivalent to WT. Correolide reduced dilation to adenosine and acetylcholine in WT and Kv1.3-/- , but had no effect on H2 O2 -dependent dilation in vessels from Kv1.3-/- mice. We conclude that Kv1.3 channels participate in the connection between myocardial blood flow and cardiac metabolism.
Collapse
Affiliation(s)
- Vahagn Ohanyan
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Liya Yin
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | | | - Christopher Kolz
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Molly Enrick
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Tatevik Hakobyan
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Jordan Luli
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Kathleen Graham
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | | | - Suzanna Logan
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - John Kmetz
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - William M Chilian
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| |
Collapse
|
49
|
Harvey LD, Chan SY. Emerging Metabolic Therapies in Pulmonary Arterial Hypertension. J Clin Med 2017; 6:jcm6040043. [PMID: 28375184 PMCID: PMC5406775 DOI: 10.3390/jcm6040043] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 03/28/2017] [Accepted: 03/29/2017] [Indexed: 12/26/2022] Open
Abstract
Pulmonary hypertension (PH) is an enigmatic vascular disorder characterized by pulmonary vascular remodeling and increased pulmonary vascular resistance, ultimately resulting in pressure overload, dysfunction, and failure of the right ventricle. Current medications for PH do not reverse or prevent disease progression, and current diagnostic strategies are suboptimal for detecting early-stage disease. Thus, there is a substantial need to develop new diagnostics and therapies that target the molecular origins of PH. Emerging investigations have defined metabolic aberrations as fundamental and early components of disease manifestation in both pulmonary vasculature and the right ventricle. As such, the elucidation of metabolic dysregulation in pulmonary hypertension allows for greater therapeutic insight into preventing, halting, or even reversing disease progression. This review will aim to discuss (1) the reprogramming and dysregulation of metabolic pathways in pulmonary hypertension; (2) the emerging therapeutic interventions targeting these metabolic pathways; and (3) further innovation needed to overcome barriers in the treatment of this devastating disease.
Collapse
Affiliation(s)
- Lloyd D Harvey
- Medical Scientist Training Program, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA.
| | - Stephen Y Chan
- Division of Cardiology, Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA.
| |
Collapse
|
50
|
Changes in gene expression profiles in patients with pulmonary arterial hypertension associated with scleroderma treated with tadalafil. Semin Arthritis Rheum 2017; 46:465-472. [DOI: 10.1016/j.semarthrit.2016.05.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 05/24/2016] [Accepted: 05/27/2016] [Indexed: 11/21/2022]
|