1
|
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: 17] [Impact Index Per Article: 17.0] [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
|
2
|
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
|
3
|
Breault NM, Wu D, Dasgupta A, Chen KH, Archer SL. Acquired disorders of mitochondrial metabolism and dynamics in pulmonary arterial hypertension. Front Cell Dev Biol 2023; 11:1105565. [PMID: 36819102 PMCID: PMC9933518 DOI: 10.3389/fcell.2023.1105565] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 01/09/2023] [Indexed: 02/05/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is an orphan disease of the cardiopulmonary unit that reflects an obstructive pulmonary vasculopathy and presents with hypertrophy, inflammation, fibrosis, and ultimately failure of the right ventricle (RVF). Despite treatment using pulmonary hypertension (PH)-targeted therapies, persistent functional impairment reduces the quality of life for people with PAH and death from RVF occurs in approximately 40% of patients within 5 years of diagnosis. PH-targeted therapeutics are primarily vasodilators and none, alone or in combination, are curative. This highlights a need to therapeutically explore molecular targets in other pathways that are involved in the pathogenesis of PAH. Several candidate pathways in PAH involve acquired mitochondrial dysfunction. These mitochondrial disorders include: 1) a shift in metabolism related to increased expression of pyruvate dehydrogenase kinase and pyruvate kinase, which together increase uncoupled glycolysis (Warburg metabolism); 2) disruption of oxygen-sensing related to increased expression of hypoxia-inducible factor 1α, resulting in a state of pseudohypoxia; 3) altered mitochondrial calcium homeostasis related to impaired function of the mitochondrial calcium uniporter complex, which elevates cytosolic calcium and reduces intramitochondrial calcium; and 4) abnormal mitochondrial dynamics related to increased expression of dynamin-related protein 1 and its binding partners, such as mitochondrial dynamics proteins of 49 kDa and 51 kDa, and depressed expression of mitofusin 2, resulting in increased mitotic fission. These acquired mitochondrial abnormalities increase proliferation and impair apoptosis in most pulmonary vascular cells (including endothelial cells, smooth muscle cells and fibroblasts). In the RV, Warburg metabolism and induction of glutaminolysis impairs bioenergetics and promotes hypokinesis, hypertrophy, and fibrosis. This review will explore our current knowledge of the causes and consequences of disordered mitochondrial function in PAH.
Collapse
Affiliation(s)
- Nolan M. Breault
- Department of Medicine, Queen’s University, Kingston, ON, Canada
| | - Danchen Wu
- Department of Medicine, Queen’s University, Kingston, ON, Canada,*Correspondence: Danchen Wu, ; Stephen L. Archer,
| | - Asish Dasgupta
- Department of Medicine, Queen’s University, Kingston, ON, Canada
| | - Kuang-Hueih Chen
- Department of Medicine, Queen’s University, Kingston, ON, Canada
| | - Stephen L. Archer
- Department of Medicine, Queen’s University, Kingston, ON, Canada,Queen’s Cardiopulmonary Unit (QCPU), Translational Institute of Medicine (TIME), Department of Medicine, Queen’s University, Kingston, ON, Canada,*Correspondence: Danchen Wu, ; Stephen L. Archer,
| |
Collapse
|
4
|
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: 2.5] [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
|
5
|
Christou H, Khalil RA. Mechanisms of pulmonary vascular dysfunction in pulmonary hypertension and implications for novel therapies. Am J Physiol Heart Circ Physiol 2022; 322:H702-H724. [PMID: 35213243 PMCID: PMC8977136 DOI: 10.1152/ajpheart.00021.2022] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/22/2022] [Accepted: 02/22/2022] [Indexed: 12/21/2022]
Abstract
Pulmonary hypertension (PH) is a serious disease characterized by various degrees of pulmonary vasoconstriction and progressive fibroproliferative remodeling and inflammation of the pulmonary arterioles that lead to increased pulmonary vascular resistance, right ventricular hypertrophy, and failure. Pulmonary vascular tone is regulated by a balance between vasoconstrictor and vasodilator mediators, and a shift in this balance to vasoconstriction is an important component of PH pathology, Therefore, the mainstay of current pharmacological therapies centers on pulmonary vasodilation methodologies that either enhance vasodilator mechanisms such as the NO-cGMP and prostacyclin-cAMP pathways and/or inhibit vasoconstrictor mechanisms such as the endothelin-1, cytosolic Ca2+, and Rho-kinase pathways. However, in addition to the increased vascular tone, many patients have a "fixed" component in their disease that involves altered biology of various cells in the pulmonary vascular wall, excessive pulmonary artery remodeling, and perivascular fibrosis and inflammation. Pulmonary arterial smooth muscle cell (PASMC) phenotypic switch from a contractile to a synthetic and proliferative phenotype is an important factor in pulmonary artery remodeling. Although current vasodilator therapies also have some antiproliferative effects on PASMCs, they are not universally successful in halting PH progression and increasing survival. Mild acidification and other novel approaches that aim to reverse the resident pulmonary vascular pathology and structural remodeling and restore a contractile PASMC phenotype could ameliorate vascular remodeling and enhance the responsiveness of PH to vasodilator therapies.
Collapse
Affiliation(s)
- Helen Christou
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Raouf A Khalil
- Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
6
|
Role of Ion Channel Remodeling in Endothelial Dysfunction Induced by Pulmonary Arterial Hypertension. Biomolecules 2022; 12:biom12040484. [PMID: 35454073 PMCID: PMC9031742 DOI: 10.3390/biom12040484] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 12/12/2022] Open
Abstract
Endothelial dysfunction is a key player in advancing vascular pathology in pulmonary arterial hypertension (PAH), a disease essentially characterized by intense remodeling of the pulmonary vasculature, vasoconstriction, endothelial dysfunction, inflammation, oxidative stress, and thrombosis in situ. These vascular features culminate in an increase in pulmonary vascular resistance, subsequent right heart failure, and premature death. Over the past years, there has been a great development in our understanding of pulmonary endothelial biology related to the genetic and molecular mechanisms that modulate the endothelial response to direct or indirect injury and how their dysregulation can promote PAH pathogenesis. Ion channels are key regulators of vasoconstriction and proliferative/apoptotic phenotypes; however, they are poorly studied at the endothelial level. The current review will describe and categorize different expression, functions, regulation, and remodeling of endothelial ion channels (K+, Ca2+, Na+, and Cl− channels) in PAH. We will focus on the potential pathogenic role of ion channel deregulation in the onset and progression of endothelial dysfunction during the development of PAH and its potential therapeutic role.
Collapse
|
7
|
Bisserier M, Sun XQ, Fazal S, Turnbull IC, Bonnet S, Hadri L. Novel Insights into the Therapeutic Potential of Lung-Targeted Gene Transfer in the Most Common Respiratory Diseases. Cells 2022; 11:984. [PMID: 35326434 PMCID: PMC8947048 DOI: 10.3390/cells11060984] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/08/2022] [Accepted: 03/10/2022] [Indexed: 12/10/2022] Open
Abstract
Over the past decades, a better understanding of the genetic and molecular alterations underlying several respiratory diseases has encouraged the development of new therapeutic strategies. Gene therapy offers new therapeutic alternatives for inherited and acquired diseases by delivering exogenous genetic materials into cells or tissues to restore physiological protein expression and/or activity. In this review, we review (1) different types of viral and non-viral vectors as well as gene-editing techniques; and (2) the application of gene therapy for the treatment of respiratory diseases and disorders, including pulmonary arterial hypertension, idiopathic pulmonary fibrosis, cystic fibrosis, asthma, alpha-1 antitrypsin deficiency, chronic obstructive pulmonary disease, non-small-cell lung cancer, and COVID-19. Further, we also provide specific examples of lung-targeted therapies and discuss the major limitations of gene therapy.
Collapse
Affiliation(s)
- Malik Bisserier
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY 10029, USA; (M.B.); (S.F.); (I.C.T.)
| | - Xiao-Qing Sun
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands;
| | - Shahood Fazal
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY 10029, USA; (M.B.); (S.F.); (I.C.T.)
| | - Irene C. Turnbull
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY 10029, USA; (M.B.); (S.F.); (I.C.T.)
| | - Sébastien Bonnet
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec, QC G1V4G5, Canada;
- Department of Medicine, Laval University, Québec, QC G1V4G5, Canada
| | - Lahouaria Hadri
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY 10029, USA; (M.B.); (S.F.); (I.C.T.)
| |
Collapse
|
8
|
The role of immune cells in pulmonary hypertension: Focusing on macrophages. Hum Immunol 2021; 83:153-163. [PMID: 34844784 DOI: 10.1016/j.humimm.2021.11.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 11/14/2021] [Accepted: 11/15/2021] [Indexed: 01/06/2023]
Abstract
Pulmonary hypertension (PH) is a life-threatening pathological state with elevated pulmonary arterial pressure, resulting in right ventricular failure and heart functional failure. Analyses of human samples and rodent models of pH support the infiltration of various immune cells, including neutrophils, mast cells, dendritic cells, B-cells, T-cells, and natural killer cells, to the lungs and pulmonary perivascular regions and their involvement in the PH development. There is evidence that macrophages are presented in the pulmonary lesions of pH patients as first-line myeloid leucocytes. Macrophage accumulation and presence, both M1 and M2 phenotypes, is a distinctive hallmark of pH which plays a pivotal role in pulmonary artery remodeling through various cellular and molecular interactions and mechanisms, including CCL2 and CX3CL1 chemokines, adventitial fibroblasts, glucocorticoid-regulated kinase 1 (SGK1), crosstalk with other immune cells, leukotriene B4 (LTB4), bone morphogenetic protein receptor 2 (BMPR2), macrophage migration inhibitory factor (MIF), and thrombospondin-1 (TSP-1). In this paper, we reviewed the molecular mechanisms and the role of immune cells and responses are involved in PH development. We also summarized the polarization of macrophages in response to different stimuli and their pathological role and their infiltration in the lung of pH patients and animal models.
Collapse
|
9
|
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: 6.0] [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
|
10
|
Rai N, Shihan M, Seeger W, Schermuly RT, Novoyatleva T. Genetic Delivery and Gene Therapy in Pulmonary Hypertension. Int J Mol Sci 2021; 22:ijms22031179. [PMID: 33503992 PMCID: PMC7865388 DOI: 10.3390/ijms22031179] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 02/06/2023] Open
Abstract
Pulmonary hypertension (PH) is a progressive complex fatal disease of multiple etiologies. Hyperproliferation and resistance to apoptosis of vascular cells of intimal, medial, and adventitial layers of pulmonary vessels trigger excessive pulmonary vascular remodeling and vasoconstriction in the course of pulmonary arterial hypertension (PAH), a subgroup of PH. Multiple gene mutation/s or dysregulated gene expression contribute to the pathogenesis of PAH by endorsing the proliferation and promoting the resistance to apoptosis of pulmonary vascular cells. Given the vital role of these cells in PAH progression, the development of safe and efficient-gene therapeutic approaches that lead to restoration or down-regulation of gene expression, generally involved in the etiology of the disease is the need of the hour. Currently, none of the FDA-approved drugs provides a cure against PH, hence innovative tools may offer a novel treatment paradigm for this progressive and lethal disorder by silencing pathological genes, expressing therapeutic proteins, or through gene-editing applications. Here, we review the effectiveness and limitations of the presently available gene therapy approaches for PH. We provide a brief survey of commonly existing and currently applicable gene transfer methods for pulmonary vascular cells in vitro and describe some more recent developments for gene delivery existing in the field of PH in vivo.
Collapse
Affiliation(s)
- Nabham Rai
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
| | - Mazen Shihan
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
| | - Werner Seeger
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Institute for Lung Health (ILH), 35392 Giessen, Germany
| | - Ralph T. Schermuly
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
| | - Tatyana Novoyatleva
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
- Correspondence:
| |
Collapse
|
11
|
Bortner CD, Cidlowski JA. Ions, the Movement of Water and the Apoptotic Volume Decrease. Front Cell Dev Biol 2020; 8:611211. [PMID: 33324655 PMCID: PMC7723978 DOI: 10.3389/fcell.2020.611211] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 11/04/2020] [Indexed: 12/20/2022] Open
Abstract
The movement of water across the cell membrane is a natural biological process that occurs during growth, cell division, and cell death. Many cells are known to regulate changes in their cell volume through inherent compensatory regulatory mechanisms. Cells can sense an increase or decrease in their cell volume, and compensate through mechanisms known as a regulatory volume increase (RVI) or decrease (RVD) response, respectively. The transport of sodium, potassium along with other ions and osmolytes allows the movement of water in and out of the cell. These compensatory volume regulatory mechanisms maintain a cell at near constant volume. A hallmark of the physiological cell death process known as apoptosis is the loss of cell volume or cell shrinkage. This loss of cell volume is in stark contrast to what occurs during the accidental cell death process known as necrosis. During necrosis, cells swell or gain water, eventually resulting in cell lysis. Thus, whether a cell gains or loses water after injury is a defining feature of the specific mode of cell death. Cell shrinkage or the loss of cell volume during apoptosis has been termed apoptotic volume decrease or AVD. Over the years, this distinguishing feature of apoptosis has been largely ignored and thought to be a passive occurrence or simply a consequence of the cell death process. However, studies on AVD have defined an underlying movement of ions that result in not only the loss of cell volume, but also the activation and execution of the apoptotic process. This review explores the role ions play in controlling not only the movement of water, but the regulation of apoptosis. We will focus on what is known about specific ion channels and transporters identified to be involved in AVD, and how the movement of ions and water change the intracellular environment leading to stages of cell shrinkage and associated apoptotic characteristics. Finally, we will discuss these concepts as they apply to different cell types such as neurons, cardiomyocytes, and corneal epithelial cells.
Collapse
Affiliation(s)
- Carl D. Bortner
- Signal Transduction Laboratory, Department of Health and Human Services, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - John A. Cidlowski
- Signal Transduction Laboratory, Department of Health and Human Services, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| |
Collapse
|
12
|
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: 6.0] [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
|
13
|
Babicheva A, Ayon RJ, Zhao T, Ek Vitorin JF, Pohl NM, Yamamura A, Yamamura H, Quinton BA, Ba M, Wu L, Ravellette KS, Rahimi S, Balistrieri F, Harrington A, Vanderpool RR, Thistlethwaite PA, Makino A, Yuan JXJ. MicroRNA-mediated downregulation of K + channels in pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol 2020; 318:L10-L26. [PMID: 31553627 PMCID: PMC6985878 DOI: 10.1152/ajplung.00010.2019] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 08/19/2019] [Accepted: 09/06/2019] [Indexed: 12/22/2022] Open
Abstract
Downregulated expression of K+ channels and decreased K+ currents in pulmonary artery smooth muscle cells (PASMC) have been implicated in the development of sustained pulmonary vasoconstriction and vascular remodeling in patients with idiopathic pulmonary arterial hypertension (IPAH). However, it is unclear exactly how K+ channels are downregulated in IPAH-PASMC. MicroRNAs (miRNAs) are small non-coding RNAs that are capable of posttranscriptionally regulating gene expression by binding to the 3'-untranslated regions of their targeted mRNAs. Here, we report that specific miRNAs are responsible for the decreased K+ channel expression and function in IPAH-PASMC. We identified 3 miRNAs (miR-29b, miR-138, and miR-222) that were highly expressed in IPAH-PASMC in comparison to normal PASMC (>2.5-fold difference). Selectively upregulated miRNAs are correlated with the decreased expression and attenuated activity of K+ channels. Overexpression of miR-29b, miR-138, or miR-222 in normal PASMC significantly decreased whole cell K+ currents and downregulated voltage-gated K+ channel 1.5 (KV1.5/KCNA5) in normal PASMC. Inhibition of miR-29b in IPAH-PASMC completely recovered K+ channel function and KV1.5 expression, while miR-138 and miR-222 had a partial or no effect. Luciferase assays further revealed that KV1.5 is a direct target of miR-29b. Additionally, overexpression of miR-29b in normal PASMC decreased large-conductance Ca2+-activated K+ (BKCa) channel currents and downregulated BKCa channel β1 subunit (BKCaβ1 or KCNMB1) expression, while inhibition of miR-29b in IPAH-PASMC increased BKCa channel activity and BKCaβ1 levels. These data indicate upregulated miR-29b contributes at least partially to the attenuated function and expression of KV and BKCa channels in PASMC from patients with IPAH.
Collapse
Affiliation(s)
- Aleksandra Babicheva
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
- Departments of Medicine and Physiology, The University of Arizona, Tucson, Arizona
| | - Ramon J Ayon
- Departments of Medicine and Physiology, The University of Arizona, Tucson, Arizona
| | - Tengteng Zhao
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
- Departments of Medicine and Physiology, The University of Arizona, Tucson, Arizona
| | - Jose F Ek Vitorin
- Departments of Medicine and Physiology, The University of Arizona, Tucson, Arizona
| | - Nicole M Pohl
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Aya Yamamura
- Kinjo Gakuin University School of Pharmacy, Nagoya, Japan
| | - Hisao Yamamura
- Nagoya City University Graduate School of Pharmaceutical Sciences, Nagoya, Japan
| | - Brooke A Quinton
- Departments of Medicine and Physiology, The University of Arizona, Tucson, Arizona
| | - Manqing Ba
- Departments of Medicine and Physiology, The University of Arizona, Tucson, Arizona
| | - Linda Wu
- Departments of Medicine and Physiology, The University of Arizona, Tucson, Arizona
| | - Keeley S Ravellette
- Departments of Medicine and Physiology, The University of Arizona, Tucson, Arizona
| | - 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
| | - Rebecca R Vanderpool
- Departments of Medicine and Physiology, The University of Arizona, Tucson, Arizona
| | | | - Ayako Makino
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, California
- Departments of Medicine and Physiology, The University of Arizona, Tucson, Arizona
| | - Jason X-J Yuan
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
- Departments of Medicine and Physiology, The University of Arizona, Tucson, Arizona
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
14
|
McClenaghan C, Woo KV, Nichols CG. Pulmonary Hypertension and ATP-Sensitive Potassium Channels. Hypertension 2019; 74:14-22. [PMID: 31132951 DOI: 10.1161/hypertensionaha.119.12992] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Conor McClenaghan
- From the Department of Cell Biology and Physiology, and Center for the Investigation of Membrane Excitability Diseases (CIMED), Washington University, St Louis, MO (C.M., C.G.N.)
| | - Kel Vin Woo
- Department of Pediatrics, Division of Cardiology, Washington University School of Medicine, St Louis, MO (K.V.W.)
| | - Colin G Nichols
- From the Department of Cell Biology and Physiology, and Center for the Investigation of Membrane Excitability Diseases (CIMED), Washington University, St Louis, MO (C.M., C.G.N.)
| |
Collapse
|
15
|
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: 3.7] [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
|
16
|
Chen J, Sysol JR, Singla S, Zhao S, Yamamura A, Valdez-Jasso D, Abbasi T, Shioura KM, Sahni S, Reddy V, Sridhar A, Gao H, Torres J, Camp SM, Tang H, Ye SQ, Comhair S, Dweik R, Hassoun P, Yuan JXJ, Garcia JGN, Machado RF. Nicotinamide Phosphoribosyltransferase Promotes Pulmonary Vascular Remodeling and Is a Therapeutic Target in Pulmonary Arterial Hypertension. Circulation 2017; 135:1532-1546. [PMID: 28202489 DOI: 10.1161/circulationaha.116.024557] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 02/06/2017] [Indexed: 11/16/2022]
Abstract
BACKGROUND Pulmonary arterial hypertension is a severe and progressive disease, a hallmark of which is pulmonary vascular remodeling. Nicotinamide phosphoribosyltransferase (NAMPT) is a cytozyme that regulates intracellular nicotinamide adenine dinucleotide levels and cellular redox state, regulates histone deacetylases, promotes cell proliferation, and inhibits apoptosis. We hypothesized that NAMPT promotes pulmonary vascular remodeling and that inhibition of NAMPT could attenuate pulmonary hypertension. METHODS Plasma, mRNA, and protein levels of NAMPT were measured in the lungs and isolated pulmonary artery endothelial cells from patients with pulmonary arterial hypertension and in the lungs of rodent models of pulmonary hypertension. Nampt+/- mice were exposed to 10% hypoxia and room air for 4 weeks, and the preventive and therapeutic effects of NAMPT inhibition were tested in the monocrotaline and Sugen hypoxia models of pulmonary hypertension. The effects of NAMPT activity on proliferation, migration, apoptosis, and calcium signaling were tested in human pulmonary artery smooth muscle cells. RESULTS Plasma and mRNA and protein levels of NAMPT were increased in the lungs and isolated pulmonary artery endothelial cells from patients with pulmonary arterial hypertension, as well as in lungs of rodent models of pulmonary hypertension. Nampt+/- mice were protected from hypoxia-mediated pulmonary hypertension. NAMPT activity promoted human pulmonary artery smooth muscle cell proliferation via a paracrine effect. In addition, recombinant NAMPT stimulated human pulmonary artery smooth muscle cell proliferation via enhancement of store-operated calcium entry by enhancing expression of Orai2 and STIM2. Last, inhibition of NAMPT activity attenuated monocrotaline and Sugen hypoxia-induced pulmonary hypertension in rats. CONCLUSIONS Our data provide evidence that NAMPT plays a role in pulmonary vascular remodeling and that its inhibition could be a potential therapeutic target for pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Jiwang Chen
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Justin R Sysol
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Sunit Singla
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Shuangping Zhao
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Aya Yamamura
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Daniela Valdez-Jasso
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Taimur Abbasi
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Krystyna M Shioura
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Sakshi Sahni
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Vamsi Reddy
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Arvind Sridhar
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Hui Gao
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Jaime Torres
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Sara M Camp
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Haiyang Tang
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Shui Q Ye
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Suzy Comhair
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Raed Dweik
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Paul Hassoun
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Jason X-J Yuan
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.)
| | - Joe G N Garcia
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.).
| | - Roberto F Machado
- From Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine (J.C., J.R.S., S.S., S.Z., A.Y., T.A., K.M.S., S.S., V.R., A.S., H.G., J.T., R.F.M.), Department of Pharmacology (J.R.S., R.F.M.), and Department of Bioengineering (A.V.-J., T.A.), University of Illinois at Chicago; Institute of Precision Medicine, Jining Medical University, China (J.C.); Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan (A.Y.); Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL (T.A.); Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.G.); Department of Medicine, University of Arizona, Tucson (S.M.C., H.T., J.X.-J.Y., J.G.N.G.); Department of Biomedical and Health Informatics and Department of Pediatrics, Children's Mercy Hospital and University of Missouri-Kansas City School of Medicine (S.Q.Y.); Department of Pathobiology, Lerner Research Institute, Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, OH (S.C., R.D.); and Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (P.H.).
| |
Collapse
|
17
|
Evans AM, Mahmoud AD, Moral-Sanz J, Hartmann S. The emerging role of AMPK in the regulation of breathing and oxygen supply. Biochem J 2016; 473:2561-72. [PMID: 27574022 PMCID: PMC5003690 DOI: 10.1042/bcj20160002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 04/20/2016] [Accepted: 05/03/2016] [Indexed: 01/25/2023]
Abstract
Regulation of breathing is critical to our capacity to accommodate deficits in oxygen availability and demand during, for example, sleep and ascent to altitude. It is generally accepted that a fall in arterial oxygen increases afferent discharge from the carotid bodies to the brainstem and thus delivers increased ventilatory drive, which restores oxygen supply and protects against hypoventilation and apnoea. However, the precise molecular mechanisms involved remain unclear. We recently identified as critical to this process the AMP-activated protein kinase (AMPK), which is key to the cell-autonomous regulation of metabolic homoeostasis. This observation is significant for many reasons, not least because recent studies suggest that the gene for the AMPK-α1 catalytic subunit has been subjected to natural selection in high-altitude populations. It would appear, therefore, that evolutionary pressures have led to AMPK being utilized to regulate oxygen delivery and thus energy supply to the body in the short, medium and longer term. Contrary to current consensus, however, our findings suggest that AMPK regulates ventilation at the level of the caudal brainstem, even when afferent input responses from the carotid body are normal. We therefore hypothesize that AMPK integrates local hypoxic stress at defined loci within the brainstem respiratory network with an index of peripheral hypoxic status, namely afferent chemosensory inputs. Allied to this, AMPK is critical to the control of hypoxic pulmonary vasoconstriction and thus ventilation-perfusion matching at the lungs and may also determine oxygen supply to the foetus by, for example, modulating utero-placental blood flow.
Collapse
Affiliation(s)
- A Mark Evans
- Centre for Integrative Physiology, College of Medicine and Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, U.K.
| | - Amira D Mahmoud
- Centre for Integrative Physiology, College of Medicine and Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, U.K
| | - Javier Moral-Sanz
- Centre for Integrative Physiology, College of Medicine and Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, U.K
| | - Sandy Hartmann
- Centre for Integrative Physiology, College of Medicine and Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, U.K
| |
Collapse
|
18
|
Moral-Sanz J, Mahmoud AD, Ross FA, Eldstrom J, Fedida D, Hardie DG, Evans AM. AMP-activated protein kinase inhibits Kv 1.5 channel currents of pulmonary arterial myocytes in response to hypoxia and inhibition of mitochondrial oxidative phosphorylation. J Physiol 2016; 594:4901-15. [PMID: 27062501 PMCID: PMC5009768 DOI: 10.1113/jp272032] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 03/26/2016] [Indexed: 12/29/2022] Open
Abstract
KEY POINTS Progression of hypoxic pulmonary hypertension is thought to be due, in part, to suppression of voltage-gated potassium channels (Kv ) in pulmonary arterial smooth muscle by hypoxia, although the precise molecular mechanisms have been unclear. AMP-activated protein kinase (AMPK) has been proposed to couple inhibition of mitochondrial metabolism by hypoxia to acute hypoxic pulmonary vasoconstriction and progression of pulmonary hypertension. Inhibition of complex I of the mitochondrial electron transport chain activated AMPK and inhibited Kv 1.5 channels in pulmonary arterial myocytes. AMPK activation by 5-aminoimidazole-4-carboxamide riboside, A769662 or C13 attenuated Kv 1.5 currents in pulmonary arterial myocytes, and this effect was non-additive with respect to Kv 1.5 inhibition by hypoxia and mitochondrial poisons. Recombinant AMPK phosphorylated recombinant human Kv 1.5 channels in cell-free assays, and inhibited K(+) currents when introduced into HEK 293 cells stably expressing Kv 1.5. These results suggest that AMPK is the primary mediator of reductions in Kv 1.5 channels following inhibition of mitochondrial oxidative phosphorylation during hypoxia and by mitochondrial poisons. ABSTRACT Progression of hypoxic pulmonary hypertension is thought to be due, in part, to suppression of voltage-gated potassium channels (Kv ) in pulmonary arterial smooth muscle cells that is mediated by the inhibition of mitochondrial oxidative phosphorylation. We sought to determine the role in this process of the AMP-activated protein kinase (AMPK), which is intimately coupled to mitochondrial function due to its activation by LKB1-dependent phosphorylation in response to increases in the cellular AMP:ATP and/or ADP:ATP ratios. Inhibition of complex I of the mitochondrial electron transport chain using phenformin activated AMPK and inhibited Kv currents in pulmonary arterial myocytes, consistent with previously reported effects of mitochondrial inhibitors. Myocyte Kv currents were also markedly inhibited upon AMPK activation by A769662, 5-aminoimidazole-4-carboxamide riboside and C13 and by intracellular dialysis from a patch-pipette of activated (thiophosphorylated) recombinant AMPK heterotrimers (α2β2γ1 or α1β1γ1). Hypoxia and inhibitors of mitochondrial oxidative phosphorylation reduced AMPK-sensitive K(+) currents, which were also blocked by the selective Kv 1.5 channel inhibitor diphenyl phosphine oxide-1 but unaffected by the presence of the BKCa channel blocker paxilline. Moreover, recombinant human Kv 1.5 channels were phosphorylated by AMPK in cell-free assays, and K(+) currents carried by Kv 1.5 stably expressed in HEK 293 cells were inhibited by intracellular dialysis of AMPK heterotrimers and by A769662, the effects of which were blocked by compound C. We conclude that AMPK mediates Kv channel inhibition by hypoxia in pulmonary arterial myocytes, at least in part, through phosphorylation of Kv 1.5 and/or an associated protein.
Collapse
Affiliation(s)
- Javier Moral-Sanz
- Centre for Integrative Physiology, College of Medicine and Veterinary Medicine, Hugh Robson Building, George Square, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Amira D Mahmoud
- Centre for Integrative Physiology, College of Medicine and Veterinary Medicine, Hugh Robson Building, George Square, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Fiona A Ross
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Dow Street, Dundee, DD1 5EH, UK
| | - Jodene Eldstrom
- Department of Anaesthesiology. Pharmacology and Therapeutics, University of British Columbia, 2350 Health Science Mall, Vancouver, Canada, V6T 1Z3
| | - David Fedida
- Department of Anaesthesiology. Pharmacology and Therapeutics, University of British Columbia, 2350 Health Science Mall, Vancouver, Canada, V6T 1Z3
| | - D Grahame Hardie
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Dow Street, Dundee, DD1 5EH, UK
| | - A Mark Evans
- Centre for Integrative Physiology, College of Medicine and Veterinary Medicine, Hugh Robson Building, George Square, University of Edinburgh, Edinburgh, EH8 9XD, UK
| |
Collapse
|
19
|
Song S, Zhang M, Yi Z, Zhang H, Shen T, Yu X, Zhang C, Zheng X, Yu L, Ma C, Liu Y, Zhu D. The role of PDGF-B/TGF-β1/neprilysin network in regulating endothelial-to-mesenchymal transition in pulmonary artery remodeling. Cell Signal 2016; 28:1489-501. [PMID: 27373199 DOI: 10.1016/j.cellsig.2016.06.022] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 05/07/2016] [Accepted: 06/29/2016] [Indexed: 12/13/2022]
Abstract
Endothelial-to-mesenchymal transition (EndoMT) has been recognized as a major reason for the pulmonary artery remodeling (PAR) in pulmonary artery hypertension (PAH). However, the molecular mechanisms and regulatory pathways involved in the EndoMT remain undefined. In the present study, we have confirmed that EndoMT was occurred in pulmonary arteries of rats induced by hypoxia and monocrotaline and in hypoxic pulmonary artery endothelial cells (PAECs). Moreover, hypoxia increased the expression of platelet-derived growth factor (PDGF) and transforming growth factor-β1 (TGF-β1) and decreased the expression of neprilysin (NEP), which contributed to the hypoxia-induced EndoMT of PAECs. Furthermore, a reciprocal regulation of PDGF-B and TGF-β1 induced by decreasing NEP promoted the EndoMT of PAECs under hypoxia, which was a novel molecular mechanism to reveal the EndoMT participating in PAR. More importantly, imatinib, a PDGF receptor antagonist, relieved PAR and EndoMT in PAH rats. Thus, our results identify a novel mechanism to reveal the formation of EndoMT in PAH, and imply that imatinib may serve as a new therapeutic approach for treatment of the third cardiovascular disease.
Collapse
Affiliation(s)
- Shasha Song
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University (Daqing), Daqing 163319, China
| | - Min Zhang
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University (Daqing), Daqing 163319, China
| | - Zhi Yi
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University (Daqing), Daqing 163319, China
| | - Hongyue Zhang
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University (Daqing), Daqing 163319, China
| | - Tingting Shen
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University (Daqing), Daqing 163319, China
| | - Xiufeng Yu
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University (Daqing), Daqing 163319, China
| | - Chen Zhang
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University (Daqing), Daqing 163319, China
| | - Xiaodong Zheng
- Department of Pathophysiology, College of Basic Medicine, Harbin Medical University (Daqing), Daqing 163319, China
| | - Lei Yu
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University (Daqing), Daqing 163319, China
| | - Cui Ma
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University (Daqing), Daqing 163319, China; Department of Immunology, College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing 163319, China
| | - Yang Liu
- Department of clinical nursing, College of nursing, Harbin Medical University (Daqing), Daqing 163319, China
| | - Daling Zhu
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University (Daqing), Daqing 163319, China; Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin 150081, China.
| |
Collapse
|
20
|
Zhang Q, Fan K, Wang P, Yu J, Liu R, Qi H, Sun H, Cao Y. Carvacrol induces the apoptosis of pulmonary artery smooth muscle cells under hypoxia. Eur J Pharmacol 2016; 770:134-46. [DOI: 10.1016/j.ejphar.2015.11.037] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 11/17/2015] [Accepted: 11/18/2015] [Indexed: 12/11/2022]
|
21
|
Boucherat O, Chabot S, Antigny F, Perros F, Provencher S, Bonnet S. Potassium channels in pulmonary arterial hypertension. Eur Respir J 2015; 46:1167-77. [PMID: 26341985 DOI: 10.1183/13993003.00798-2015] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 07/09/2015] [Indexed: 12/15/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a devastating cardiopulmonary disorder with various origins. All forms of PAH share a common pulmonary arteriopathy characterised by vasoconstriction, remodelling of the pre-capillary pulmonary vessel wall, and in situ thrombosis. Although the pathogenesis of PAH is recognised as a complex and multifactorial process, there is growing evidence that potassium channels dysfunction in pulmonary artery smooth muscle cells is a hallmark of PAH. Besides regulating many physiological functions, reduced potassium channels expression and/or activity have significant effects on PAH establishment and progression. This review describes the molecular mechanisms and physiological consequences of potassium channel modulation. Special emphasis is placed on KCNA5 (Kv1.5) and KCNK3 (TASK1), which are considered to play a central role in determining pulmonary vascular tone and may represent attractive therapeutic targets in the treatment of PAH.
Collapse
Affiliation(s)
- Olivier Boucherat
- Pulmonary Hypertension Research Group, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, QC, Canada
| | - Sophie Chabot
- Pulmonary Hypertension Research Group, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, QC, Canada
| | - Fabrice Antigny
- Pulmonary Hypertension Research Group, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, QC, Canada UMRS 999, INSERM and Univ. Paris-Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France
| | - Frédéric Perros
- Pulmonary Hypertension Research Group, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, QC, Canada UMRS 999, INSERM and Univ. Paris-Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France
| | - Steeve Provencher
- Pulmonary Hypertension Research Group, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, QC, Canada
| | - Sébastien Bonnet
- Pulmonary Hypertension Research Group, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, QC, Canada
| |
Collapse
|
22
|
Liu Y, Zheng D, Liu M, Bai J, Zhou X, Gong B, Lü J, Zhang Y, Huang H, Luo W, Huang G. Downregulation of glypican-3 expression increases migration, invasion, and tumorigenicity of human ovarian cancer cells. Tumour Biol 2015; 36:7997-8006. [PMID: 25967456 DOI: 10.1007/s13277-015-3528-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 05/04/2015] [Indexed: 01/03/2023] Open
Abstract
Glypican-3 (GPC3) is a membrane of heparan sulfate proteoglycan family involved in cell proliferation, adhesion, migration, invasion, and differentiation during the development of the majority of mesodermal tissues and organs. GPC3 is explored as a potential biomarker for hepatocellular carcinoma screening. However, as a tumor-associated antigen, its role in ovarian cancer remains elusive. In this report, the expression levels of GPC3 in the various ovarian cancer cells were determined with quantitative reverse transcription-polymerase chain reaction (qRT-PCR), and GPC3 expression in ovarian cancer UCI 101 and A2780 cells was knocked down by siRNA transfection, and the effects of GPC3 knockdown on in vitro cell proliferation, migration, and invasion were respectively analyzed by 3-[4, 5-dimethylthiazol-2-yl]-2, 5-diphenyltetrazolium bromide (MTT) assay and Transwell migration assay. Additionally, the effect of GPC3 knockdown on in vivo tumorigenesis were investigated in athymic nude mice. The results indicated that GPC3 knockdown significantly promoted cell proliferation and increased cell migration and invasion by upregulation of matrix metalloproteinase (MMP)-2 and MMP-9 expression and downregulation of tissue inhibitor of metalloproteinase-1 expression. Additionally, GPC3 knockdown also increased in vivo tumorigenicity of UCI 101 and A2780 cells and final tumor weights and volumes after subcutaneous cell injection in the nude mice. The results of immunohistochemical staining and Western blotting both demonstrated a lower expression of GPC3 antigen in the tumors of GPC3 knockdown groups than that of negative control groups. Moreover, transforming growth factor-β2 protein expression in the tumors of GPC3 knockdown groups was significantly increased, which at least contributed to tumor growth in the nude mice. Taken together, these findings suggest that GPC3 knockdown promotes the progression of human ovarian cancer cells by increasing their migration, invasion, and tumorigenicity, and suggest that GPC3 is a potential therapeutic target for ovarian cancer patients.
Collapse
Affiliation(s)
- Ying Liu
- Department of Obstetrics and Gynecology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei Province, China
| | - Dongping Zheng
- Ultrasonic Imaging Division, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei Province, China
| | - Mingming Liu
- Department of Obstetrics and Gynecology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei Province, China
| | - Jiao Bai
- Ultrasonic Imaging Division, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei Province, China
| | - Xi Zhou
- Department of Obstetrics and Gynecology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei Province, China
| | - Baolan Gong
- Department of Obstetrics and Gynecology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei Province, China
| | - Jieyu Lü
- Department of Obstetrics and Gynecology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei Province, China
| | - Yi Zhang
- Department of Obstetrics and Gynecology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei Province, China
| | - Hui Huang
- Department of Obstetrics and Gynecology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei Province, China
| | - Wenying Luo
- Department of Clinical Laboratory, Affiliated Hospital of Guangdong Medical College, Zhanjiang, 524001, China
| | - Guangrong Huang
- Department of Obstetrics and Gynecology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei Province, China.
| |
Collapse
|
23
|
Zhang P, Li J, Tang X, Zhang J, Liang J, Zeng G. Dracorhodin perchlorate induces apoptosis in primary fibroblasts from human skin hypertrophic scars via participation of caspase-3. Eur J Pharmacol 2014; 728:82-92. [DOI: 10.1016/j.ejphar.2014.01.068] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 01/21/2014] [Accepted: 01/24/2014] [Indexed: 10/25/2022]
|
24
|
Makino A, Firth AL, Yuan JXJ. Endothelial and smooth muscle cell ion channels in pulmonary vasoconstriction and vascular remodeling. Compr Physiol 2013; 1:1555-602. [PMID: 23733654 DOI: 10.1002/cphy.c100023] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The pulmonary circulation is a low resistance and low pressure system. Sustained pulmonary vasoconstriction and excessive vascular remodeling often occur under pathophysiological conditions such as in patients with pulmonary hypertension. Pulmonary vasoconstriction is a consequence of smooth muscle contraction. Many factors released from the endothelium contribute to regulating pulmonary vascular tone, while the extracellular matrix in the adventitia is the major determinant of vascular wall compliance. Pulmonary vascular remodeling is characterized by adventitial and medial hypertrophy due to fibroblast and smooth muscle cell proliferation, neointimal proliferation, intimal, and plexiform lesions that obliterate the lumen, muscularization of precapillary arterioles, and in situ thrombosis. A rise in cytosolic free Ca(2+) concentration ([Ca(2+)]cyt) in pulmonary artery smooth muscle cells (PASMC) is a major trigger for pulmonary vasoconstriction, while increased release of mitogenic factors, upregulation (or downregulation) of ion channels and transporters, and abnormalities in intracellular signaling cascades are key to the remodeling of the pulmonary vasculature. Changes in the expression, function, and regulation of ion channels in PASMC and pulmonary arterial endothelial cells play an important role in the regulation of vascular tone and development of vascular remodeling. This article will focus on describing the ion channels and transporters that are involved in the regulation of pulmonary vascular function and structure and illustrating the potential pathogenic role of ion channels and transporters in the development of pulmonary vascular disease.
Collapse
Affiliation(s)
- Ayako Makino
- Department of Medicine, The University of Illinois at Chicago, Chicago, Illinois, USA
| | | | | |
Collapse
|
25
|
Song S, Wang S, Ma J, Yao L, Xing H, Zhang L, Liao L, Zhu D. Biliverdin reductase/bilirubin mediates the anti-apoptotic effect of hypoxia in pulmonary arterial smooth muscle cells through ERK1/2 pathway. Exp Cell Res 2013; 319:1973-1987. [PMID: 23722043 DOI: 10.1016/j.yexcr.2013.05.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Revised: 05/13/2013] [Accepted: 05/14/2013] [Indexed: 10/26/2022]
Abstract
Inhibition of pulmonary arterial smooth muscle cell (PASMC) apoptosis induced by hypoxia plays an important role in pulmonary arterial remodeling leading to aggravate hypoxic pulmonary arterial hypertension. However, the mechanisms of hypoxia acting on PASMC apoptosis remain exclusive. Biliverdin reductase (BVR) has many essential biologic roles in physiological and pathological processes. Nevertheless, it is unclear whether the hypoxia-induced inhibition on PASMC apoptosis is mediated by BVR. In the present work, we found BVR majorly localized in PASMCs and was up-regulated in levels of protein and mRNA by hypoxia. Then we studied the contribution of BVR to anti-apoptotic response of hypoxia in PASMCs. Our results showed that siBVR, blocking generation of bilirubin, reversed the effect of hypoxia on enhancing cell survival and apoptotic protein (Bcl-2, procasepase-9, procasepase-3) expression, preventing nuclear shrinkage, DNA fragmentation and mitochondrial depolarization in starved PASMCs, which were recovered by exogenous bilirubin. Moreover, the inhibitory effect of bilirubin on PASMC apoptosis under hypoxic condition was blocked by the inhibitor of ERK1/2 pathway. Taken together, our data indicate that BVR contributes to the inhibitory process of hypoxia on PASMC apoptosis, which is mediated by bilirubin through ERK1/2 pathway.
Collapse
Affiliation(s)
- Shasha Song
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical, University (Daqing), Daqing 163319, China
| | - Shuang Wang
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical, University (Daqing), Daqing 163319, China; Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin 150081, China
| | - Jun Ma
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical, University (Daqing), Daqing 163319, China
| | - Lan Yao
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical, University (Daqing), Daqing 163319, China
| | - Hao Xing
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical, University (Daqing), Daqing 163319, China
| | - Lei Zhang
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical, University (Daqing), Daqing 163319, China
| | - Lin Liao
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical, University (Daqing), Daqing 163319, China
| | - Daling Zhu
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical, University (Daqing), Daqing 163319, China; Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin 150081, China.
| |
Collapse
|
26
|
Incretin-stimulated interaction between β-cell Kv1.5 and Kvβ2 channel proteins involves acetylation/deacetylation by CBP/SirT1. Biochem J 2013; 451:227-34. [DOI: 10.1042/bj20121669] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The incretins, GIP (glucose-dependent insulinotropic polypeptide) and GLP-1 (glucagon-like peptide-1) are gastrointestinal hormones conferring a number of beneficial effects on β-cell secretion, survival and proliferation. In a previous study, it was demonstrated that delayed rectifier channel protein Kv2.1 contributes to β-cell apoptosis and that the prosurvival effects of incretins involve Kv2.1 PTMs (post-translational modifications), including phosphorylation and acetylation. Since Kv1.5 overexpression was also shown to stimulate β-cell death, the present study was initiated in order to determine whether incretins modulate Kv1.5α–Kvβ2 interaction via PTM and the mechanisms involved. GIP and GLP-1 reduced apoptosis in INS-1 β-cells (clone 832/13) overexpressing Kv1.5, and RNAi (RNA interference)-mediated knockdown of endogenous Kv1.5 attenuated apoptotic β-cell death. Both GIP and GLP-1 increased phosphorylation and acetylation of Kv1.5 and its Kvβ2 protein subunit, leading to their enhanced interaction. Further studies demonstrated that CBP [CREB (cAMP-response-element-binding protein)-binding protein]/SirT1 mediated acetylation/deacetylation and interaction between Kvβ2 and Kv1.5 in response to GIP or GLP-1. Incretin regulation of β-cell function therefore involves the acetylation of multiple Kvα and Kvβ subunits.
Collapse
|
27
|
Bossini-Castillo L, Simeon CP, Beretta L, Broen J, Vonk MC, Callejas JL, Carreira P, Rodríguez-Rodríguez L, García-Portales R, González-Gay MA, Castellví I, Camps MT, Tolosa C, Vicente-Rabaneda E, Egurbide MV, Schuerwegh AJ, Hesselstrand R, Lunardi C, van Laar JM, Shiels P, Herrick A, Worthington J, Denton C, Radstake TRDJ, Fonseca C, Martin J. KCNA5 gene is not confirmed as a systemic sclerosis-related pulmonary arterial hypertension genetic susceptibility factor. Arthritis Res Ther 2012; 14:R273. [PMID: 23270786 PMCID: PMC3674598 DOI: 10.1186/ar4124] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Accepted: 12/20/2012] [Indexed: 01/22/2023] Open
Abstract
INTRODUCTION Potassium voltage-gated channel shaker-related subfamily member 5 (KCNA5) is implicated in vascular tone regulation, and its inhibition during hypoxia produces pulmonary vasoconstriction. Recently, a protective association of the KCNA5 locus with systemic sclerosis (SSc) patients with pulmonary arterial hypertension (PAH) was reported. Hence, the aim of this study was to replicate these findings in an independent multicenter Caucasian SSc cohort. METHODS The 2,343 SSc cases (179 PAH positive, confirmed by right-heart catheterization) and 2,690 matched healthy controls from five European countries were included in this study. Rs10744676 single-nucleotide polymorphism (SNP) was genotyped by using a TaqMan SNP genotyping assay. RESULTS Individual population analyses of the selected KCNA5 genetic variant did not show significant association with SSc or any of the defined subsets (for example, limited cutaneous SSc, diffuse cutaneous SSc, anti-centromere autoantibody positive and anti-topoisomerase autoantibody positive). Furthermore, pooled analyses revealed no significant evidence of association with the disease or any of the subsets, not even the PAH-positive group. The comparison of PAH-positive patients with PAH-negative patients showed no significant differences among patients. CONCLUSIONS Our data do not support an important role of KCNA5 as an SSc-susceptibility factor or as a PAH-development genetic marker for SSc patients.
Collapse
|
28
|
Abstract
Endothelial injury related to oxidative stress is a key event in cardiovascular diseases, such as hypertension and atherosclerosis. The activation of the redox-sensitive Kv1.5 potassium channel mediates mitochondrial reactive oxygen species (ROS)-induced apoptosis in vascular smooth muscle cells and some cancer cells. Kv1.5 channel is therefore taken as a new potential therapeutic target for pulmonary hypertension and cancers. Although Kv1.5 is abundantly expressed in vascular endothelium, there is little knowledge of its role in endothelial injury related to oxidative stress. We found that DPO-1, a specific inhibitor of Kv1.5, attenuated H2O2-evoked endothelial cell apoptosis in an in vivo rat carotid arterial model. In human umbilical vein endothelial cells (HUVECs) and human pulmonary arterial endothelial cells (HPAECs), angiotensin II and oxLDL time- or concentration-dependently enhanced Kv1.5 protein expression in parallel with the production of intracellular ROS and endothelial cell injury. Moreover, siRNA-mediated knockdown of Kv1.5 attenuated, whereas adenovirus-mediated Kv1.5 cDNA overexpression enhanced oxLDL–induced cellular damage, NADPH oxidase and mitochondria-derived ROS production and restored the decrease in protein expression of mitochondria uncoupling protein 2 (UCP2). Collectively, these data suggest that Kv1.5 may play an important role in oxidative vascular endothelial injury.
Collapse
|
29
|
Sikka P, Kapoor S, Bindra VK, Saini M, Saxena KK. Iptakalim: A novel multi-utility potassium channel opener. J Pharmacol Pharmacother 2012; 3:12-4. [PMID: 22368410 PMCID: PMC3284029 DOI: 10.4103/0976-500x.92495] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Pranav Sikka
- Department of Pharmacology, LLRM Medical College, Meerut, Uttar Pradesh, India
| | | | | | | | | |
Collapse
|
30
|
Kuhr FK, Smith KA, Song MY, Levitan I, Yuan JXJ. New mechanisms of pulmonary arterial hypertension: role of Ca²⁺ signaling. Am J Physiol Heart Circ Physiol 2012; 302:H1546-62. [PMID: 22245772 DOI: 10.1152/ajpheart.00944.2011] [Citation(s) in RCA: 145] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a severe and progressive disease that usually culminates in right heart failure and death if left untreated. Although there have been substantial improvements in our understanding and significant advances in the management of this disease, there is a grim prognosis for patients in the advanced stages of PAH. A major cause of PAH is increased pulmonary vascular resistance, which results from sustained vasoconstriction, excessive pulmonary vascular remodeling, in situ thrombosis, and increased pulmonary vascular stiffness. In addition to other signal transduction pathways, Ca(2+) signaling in pulmonary artery smooth muscle cells (PASMCs) plays a central role in the development and progression of PAH because of its involvement in both vasoconstriction, through its pivotal effect of PASMC contraction, and vascular remodeling, through its stimulatory effect on PASMC proliferation. Altered expression, function, and regulation of ion channels and transporters in PASMCs contribute to an increased cytosolic Ca(2+) concentration and enhanced Ca(2+) signaling in patients with PAH. This review will focus on the potential pathogenic role of Ca(2+) mobilization, regulation, and signaling in the development and progression of PAH.
Collapse
Affiliation(s)
- Frank K Kuhr
- Section of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612, USA
| | | | | | | | | |
Collapse
|
31
|
Zhuang JL, Wang CY, Zhou MH, Duan KZ, Mei YA. TGF-β1 enhances Kv2.1 potassium channel protein expression and promotes maturation of cerebellar granule neurons. J Cell Physiol 2011; 227:297-307. [PMID: 21412780 DOI: 10.1002/jcp.22735] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Members of the transforming growth factor-β (TGF-β) family of cytokines are involved in diverse physiological processes. Although TGF-β is known to play multiple roles in the mammalian central nervous system (CNS), its role in neuronal development has not been explored. We have studied the effects of TGF-β1 on the electrophysiological properties and maturation of rat primary cerebellar granule neurons (CGNs). We report that incubation with TGF-β1 increased delayed rectifier potassium current (I(K) ) amplitudes in a dose- and time-dependent manner, but did not affect the kinetic properties of the channel. Exposure to TGF-β1 (20 ng/ml) for 36 h led to a 37.2% increase in I(K) amplitudes. There was no significant change in mRNA levels for the key Kv2.1 channel protein, but translation blockade abolished the increase in protein levels and channel activity, arguing that TGF-β1 increases I(K) amplitudes by upregulating translation of the Kv2.1 channel protein. Although TGF-β1 treatment did not affect the activity of protein kinase A (PKA), and constitutive activation of PKA with forskolin failed to increase I(K) amplitudes, inhibition of PKA prevented channel upregulation, demonstrating that basal PKA activity is required for TGF-β1 stimulation of I(K) channel activity. TGF-β1 also promoted the expression of the γ-aminobutyric acid (GABA(A) ) receptor α6 subunit, a marker of mature CGNs, and calcium influx during depolarizing stimuli was reduced by TGF-β1. The effects of TGF-β1 were only observed during a narrow developmental time-window, and were lost as CGNs matured. These findings suggest that TGF-β1 upregulates K(+) channel expression and I(K) currents and thereby promotes CGN maturation.
Collapse
Affiliation(s)
- Jia-Li Zhuang
- Institutes of Brain Science, School of Life Sciences and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | | | | | | | | |
Collapse
|
32
|
Inhibition of TASK-3 (KCNK9) channel biosynthesis changes cell morphology and decreases both DNA content and mitochondrial function of melanoma cells maintained in cell culture. Melanoma Res 2011; 21:308-22. [DOI: 10.1097/cmr.0b013e3283462713] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
33
|
Mo J, Xia Y, Wade TJ, DeMarini DM, Davidson M, Mumford J. Altered gene expression by low-dose arsenic exposure in humans and cultured cardiomyocytes: assessment by real-time PCR arrays. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2011; 8:2090-108. [PMID: 21776218 PMCID: PMC3138013 DOI: 10.3390/ijerph8062090] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Revised: 05/21/2011] [Accepted: 05/27/2011] [Indexed: 11/16/2022]
Abstract
Chronic arsenic exposure results in higher risk of skin, lung, and bladder cancer, as well as cardiovascular disease and diabetes. The purpose of this study was to investigate the effects on expression of selected genes in the blood lymphocytes from 159 people exposed chronically to arsenic in their drinking water using a novel RT-PCR TaqMan low-density array (TLDA). We found that expression of tumor necrosis factor-α (TNF-α), which activates both inflammation and NF-κB-dependent survival pathways, was strongly associated with water and urinary arsenic levels. Expression of KCNA5, which encodes a potassium ion channel protein, was positively associated with water and toe nail arsenic levels. Expression of 2 and 11 genes were positively associated with nail and urinary arsenic, respectively. Because arsenic exposure has been reported to be associated with long QT intervals and vascular disease in humans, we also used this TLDA for analysis of gene expression in human cardiomyocytes exposed to arsenic in vitro. Expression of the ion-channel genes CACNA1, KCNH2, KCNQ1 and KCNE1 were down-regulated by 1-μM arsenic. Alteration of some common pathways, including those involved in oxidative stress, inflammatory signaling, and ion-channel function, may underlay the seemingly disparate array of arsenic-associated diseases, such as cancer, cardiovascular disease, and diabetes.
Collapse
Affiliation(s)
- Jinyao Mo
- Center for Environmental Medicine, Asthma and Lung Biology, University of North Carolina, Chapel Hill, NC 27599, USA; E-Mail:
| | - Yajuan Xia
- Inner Mongolia Center for Endemic Disease Control and Research, Huhhot 010031, Inner Mongolia, China; E-Mail:
| | - Timothy J. Wade
- National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA; E-Mails: (D.M.D.); (J.M.)
| | - David M. DeMarini
- National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA; E-Mails: (D.M.D.); (J.M.)
| | - Mercy Davidson
- Department of Radiation Oncology, Columbia University, New York, NY 10032, USA; E-Mail:
| | - Judy Mumford
- National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA; E-Mails: (D.M.D.); (J.M.)
| |
Collapse
|
34
|
Song MY, Makino A, Yuan JXJ. STIM2 Contributes to Enhanced Store-operated Ca Entry in Pulmonary Artery Smooth Muscle Cells from Patients with Idiopathic Pulmonary Arterial Hypertension. Pulm Circ 2011; 1:84-94. [PMID: 21709766 PMCID: PMC3121304 DOI: 10.4103/2045-8932.78106] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Pulmonary vasoconstriction and vascular remodeling are two major causes for elevated pulmonary vascular resistance and pulmonary arterial pressure in patients with idiopathic pulmonary arterial hypertension (IPAH). An increase in cytosolic free Ca2+concentration ([Ca2+]cyt) in pulmonary artery smooth muscle cells (PASMC) is a major trigger for pulmonary vasoconstriction and an important stimulus for PASMC proliferation, which causes pulmonary vascular remodeling. Store-operated Ca2+ entry (SOCE), induced by depletion of stored Ca2+ in the sarcoplasmic reticulum (SR), can increase [Ca2+]cyt in PASMC, independent of other means of Ca2+ entry. Stromal interaction molecule (STIM) proteins, STIM1 and STIM2, were both recently identified as sensors for store depletion and also signaling molecules to open store-operated Ca2+ channels. We previously reported that SOCE was significantly enhanced in PASMC from IPAH patients compared to PASMC from normotensive control subjects. Enhanced SOCE plays an important role in the pathophysiological changes in PASMC associated with pulmonary arterial hypertension. In this study, we examine whether the expression levels of STIM1 and STIM2 are altered in IPAH-PASMC compared to control PASMC, and whether these putative changes in the STIM1 and STIM2 expression levels are responsible for enhanced SOCE and proliferation in IPAH-PASMC. Compared to control PASMC, the protein expression level of STIM2 was significantly increased in IPAH-PASMC, whereas STIM1 protein expression was not significantly changed. In IPAH-PASMC, the small interfering RNA (siRNA)-mediated knockdown of STIM2 decreased SOCE and proliferation, while knockdown of STIM2 in control PASMC had no effect on either SOCE or proliferation. Overexpression of STIM2 in the control PASMC failed to enhance SOCE or proliferation. These data indicate that enhanced protein expression of STIM2 is necessary, but not sufficient, for enhanced SOCE and proliferation of IPAH-PASMC.
Collapse
Affiliation(s)
- Michael Y Song
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093-0725
| | | | | |
Collapse
|
35
|
Bilirubin oxidation end products directly alter K+ channels important in the regulation of vascular tone. J Cereb Blood Flow Metab 2011; 31:102-12. [PMID: 20424637 PMCID: PMC2970662 DOI: 10.1038/jcbfm.2010.54] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The exact etiology of delayed cerebral vasospasm following cerebral hemorrhage is not clear, but a family of compounds termed 'bilirubin oxidation end products (BOXes)' derived from heme has been implicated. As proper regulation of vascular smooth muscle tone involves large-conductance Ca(2+)- and voltage-dependent Slo1 K(+) (BK, maxiK, K(Ca)1.1) channels, we examined whether BOXes altered functional properties of the channel. Electrophysiological measurements of Slo1 channels heterologously expressed in a human cell line and of native mouse BK channels in isolated cerebral myocytes showed that BOXes markedly diminished open probability. Biophysically, BOXes specifically stabilized the conformations of the channel with its ion conduction gate closed. The results of chemical amino-acid modifications and molecular mutagenesis together suggest that two specific lysine residues in the structural element linking the transmembrane ion-permeation domain to the carboxyl cytosolic domain of the Slo1 channel are critical in determining the sensitivity of the channel to BOXes. Inhibition of Slo1 BK channels by BOXes may contribute to the development of delayed cerebral vasospasm following brain hemorrhage.
Collapse
|
36
|
Ko EA, Park WS, Firth AL, Kim N, Yuan JXJ, Han J. Pathophysiology of voltage-gated K+ channels in vascular smooth muscle cells: Modulation by protein kinases. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2010; 103:95-101. [DOI: 10.1016/j.pbiomolbio.2009.10.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2009] [Accepted: 10/07/2009] [Indexed: 10/20/2022]
|
37
|
Zhang L, Ma J, Li Y, Guo L, Ran Y, Liu S, Jiang C, Zhu D. 15-Hydroxyeicosatetraenoic acid (15-HETE) protects pulmonary artery smooth muscle cells against apoptosis via HSP90. Life Sci 2010; 87:223-31. [DOI: 10.1016/j.lfs.2010.06.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2009] [Revised: 05/20/2010] [Accepted: 06/14/2010] [Indexed: 01/11/2023]
|
38
|
Chimote AA, Adragna NC, Lauf PK. Ion transport in a human lens epithelial cell line exposed to hyposmotic and apoptotic stress. J Cell Physiol 2010; 223:110-22. [PMID: 20049853 DOI: 10.1002/jcp.22015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Membrane transport changes in human lens epithelial (HLE-B3) cells under hyposmotic and apoptotic stress were compared. Cell potassium content, K(i), uptake of the K congener rubidium, Rb(i), and water content were measured after hyposmotic stress induced by hypotonicity, and apoptotic stress by the protein-kinase inhibitor staurosporine (STP). Cell water increased in hyposmotic (150 mOsm) as compared to isosmotic (300 mOsm) balanced salt solution (BSS) by >2-fold at 5 min and decreased within 15 min to baseline values accompanied by a 40% K(i) loss commensurate with cell swelling and subsequent cell shrinkage likely due to regulatory volume decrease (RVD). Loss of K(i), and accompanying water, and Rb(i) uptake in hyposmotic BSS were prevented by clotrimazole (CTZ) suggesting water shifts associated with K and Rb flux via intermediate conductance K (IK) channels, also detected at the mRNA and protein level. In contrast, 2 h after 2 microM STP exposure, the cells lost approximately 40% water and approximately 60% K(i), respectively, consistent with apoptotic volume decrease (AVD). Indeed, water and K(i) loss was at least fivefold greater after hyposmotic than after apoptotic stress. High extracellular K and 2 mM 4-aminopyridine (4-AP) but not CTZ significantly reduced apoptosis. Annexin labeling phosphatidylserine (PS) at 15 min suggested loss of lipid asymmetry. Quantitative PCR revealed significant IK channel expression during prolonged hyposmotic stress. Results suggest in HLE-B3 cells, IK channels likely partook in and were down regulated after RVD, whereas pro-apoptotic STP-activation of 4-AP-sensitive voltage-gated K channels preceded or accompanied PS externalization before subsequent apoptosis.
Collapse
Affiliation(s)
- Ameet A Chimote
- Cell Biophysics Group, Boonshoft School of Medicine, Wright State University, Dayton, Ohio 45435, USA
| | | | | |
Collapse
|
39
|
Contribution of voltage-gated potassium channels to the regulation of apoptosis. FEBS Lett 2010; 584:2049-56. [DOI: 10.1016/j.febslet.2010.01.038] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2009] [Revised: 01/18/2010] [Accepted: 01/19/2010] [Indexed: 01/25/2023]
|
40
|
Burg ED, Platoshyn O, Tsigelny IF, Lozano-Ruiz B, Rana BK, Yuan JXJ. Tetramerization domain mutations in KCNA5 affect channel kinetics and cause abnormal trafficking patterns. Am J Physiol Cell Physiol 2009; 298:C496-509. [PMID: 20018952 DOI: 10.1152/ajpcell.00464.2009] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The activity of voltage-gated K(+) (K(V)) channels plays an important role in regulating pulmonary artery smooth muscle cell (PASMC) contraction, proliferation, and apoptosis. The highly conserved NH(2)-terminal tetramerization domain (T1) of K(V) channels is important for proper channel assembly, association with regulatory K(V) beta-subunits, and localization of the channel to the plasma membrane. We recently reported two nonsynonymous mutations (G182R and E211D) in the KCNA5 gene of patients with idiopathic pulmonary arterial hypertension, which localize to the T1 domain of KCNA5. To study the electrophysiological properties and expression patterns of the mutants compared with the wild-type (WT) channel in vitro, we transfected HEK-293 cells with WT KCNA5, G182R, E211D, or the double mutant G182R/E211D channel. The mutants form functional channels; however, whole cell current kinetic differences between WT and mutant channels exist. Steady-state inactivation curves of the G182R and G182R/E211D channels reveal accelerated inactivation; the mutant channels inactivated at more hyperpolarized potentials compared with the WT channel. Channel protein expression was also decreased by the mutations. Compared with the WT channel, which was present in its mature glycosylated form, the mutant channels are present in greater proportion in their immature form in HEK-293 cells. Furthermore, G182R protein level is greatly reduced in COS-1 cells compared with WT. Immunostaining data support the hypothesis that, while WT protein localizes to the plasma membrane, mutant protein is mainly retained in intracellular packets. Overall, these data support a role for the T1 domain in channel kinetics as well as in KCNA5 channel subcellular localization.
Collapse
Affiliation(s)
- Elyssa D Burg
- Dept. of Medicine, Univ. of California, San Diego, 9500 Gilman Dr., MC 0725, La Jolla, CA 92093-0725, USA
| | | | | | | | | | | |
Collapse
|
41
|
Nakamura T, Bhatt S, Sapru HN. Cardiovascular responses to hypothalamic arcuate nucleus stimulation in the rat: role of sympathetic and vagal efferents. Hypertension 2009; 54:1369-75. [PMID: 19884562 DOI: 10.1161/hypertensionaha.109.140715] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Experiments were carried out in urethane-anesthetized, artificially ventilated, adult male Wistar rats. Microinjections (50 nL) of N-methyl-d-aspartic acid (1, 5, and 10 mmol/L), but not artificial cerebrospinal fluid, into the hypothalamic arcuate nucleus (ARCN) elicited increases in mean arterial pressure (5.7+/-0.5, 13.2+/-1.4, and 17.3+/-1.1 mm Hg, respectively) and heart rate (24.3+/-4.3, 49.3+/-5.2, and 75.2+/-8.0 bpm, respectively). ARCN stimulation was accomplished by microinjections of a maximally effective concentration of N-methyl-d-aspartic acid (10 mmol/L). The tachycardic responses to the ARCN stimulation were significantly attenuated after bilateral vagotomy. Intrathecal injections of ionotropic glutamate receptor (iGLUR) antagonists completely blocked pressor responses to the ARCN stimulation, whereas the tachycardic responses were significantly attenuated but not abolished. Intrathecal injections of iGLUR antagonists at T9 to T10, combined with bilateral vagotomy, completely blocked the tachycardic responses to ARCN stimulation. ARCN stimulation with N-methyl-d-aspartic acid elicited increased activities of the greater splanchnic nerve (91.7+/-14.8%) and the renal nerve (109.3+/-13%). Intrathecal injections of iGLURs at T9 to T10 blocked the increase in the greater splanchnic nerve activity in response to ARCN stimulation. These results indicate the following: (1) the chemical stimulation of the ARCN elicits increases in mean arterial pressure, greater splanchnic nerve and renal nerve activity, and heart rate; (2) the increases in mean arterial pressure and sympathetic nerve activity are mediated via the activation of spinal cord iGLURs; and (3) the increases in heart rate are mediated via the activation of spinal cord iGLURs and decreases in vagal input to the heart.
Collapse
Affiliation(s)
- Takeshi Nakamura
- Department of Neurological Surgery, MSB H-586, UMDNJ-New Jersey Medical School, 185 South Orange Ave, Newark, NJ 07103, USA
| | | | | |
Collapse
|
42
|
Merkel OM, Beyerle A, Librizzi D, Pfestroff A, Behr TM, Sproat B, Barth PJ, Kissel T. Nonviral siRNA delivery to the lung: investigation of PEG-PEI polyplexes and their in vivo performance. Mol Pharm 2009; 6:1246-60. [PMID: 19606864 DOI: 10.1021/mp900107v] [Citation(s) in RCA: 143] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
This study describes the physicobiological characterization of PEI- and PEG-PEI polyplexes containing partially 2'-OMe modified 25/27mer dicer substrate siRNAs (DsiRNAs) and their in vivo behavior regarding biodistribution and systemic bioavailability after pulmonary application as well as their ability to knock down gene expression in the lung. Biophysical characterization included circular dichroism of siRNA in polyplexes, condensation efficiency of polymers and in vitro stability. After in vivo application, biodistribution and kinetics of radiolabeled polyplexes were quantified and recorded over time in three-dimensional SPECT images and by end point scintillation counting. The influence on lung tissue and on the humoral and cellular immunosystem was investigated, and finally knockdown of endogenous gene expression in the lung was determined qualitatively. While all of the polymers used in our study were proven to effectively condense siRNA, stability of the complexes depended on the PEG grafting degree. Interestingly, PEI 25 kDa, which showed the least interaction with mucin or surfactant in vitro, performed poorly in vivo. Our nuclear imaging approach enabled us to follow biodistribution of the instilled nanocarriers over time and indicated that PEGylated nanocarriers are more suitable for lung application. While moderate proinflammatory effects were attributed to PEI25k-PEG(2k)(10) nanocarriers, none of the treatments caused histological abnormalities. Our preliminary in vivo knockdown experiment suggests that PEG-PEI/siRNA complexes are promising nanomedicines for pulmonary siRNA delivery. These results encouraged us to further investigate possible adverse effects and to quantify in vivo gene silencing in the lung after intratracheal instillation of PEG-PEI/siRNA complexes.
Collapse
Affiliation(s)
- Olivia M Merkel
- Department of Pharmaceutics and Biopharmacy, Philipps Universitat Marburg, Ketzerbach 63, 35032 Marburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Bonnet S, Paulin R, Sutendra G, Dromparis P, Roy M, Watson KO, Nagendran J, Haromy A, Dyck JRB, Michelakis ED. Dehydroepiandrosterone reverses systemic vascular remodeling through the inhibition of the Akt/GSK3-{beta}/NFAT axis. Circulation 2009; 120:1231-40. [PMID: 19752325 DOI: 10.1161/circulationaha.109.848911] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND The remodeled vessel wall in many vascular diseases such as restenosis after injury is characterized by proliferative and apoptosis-resistant vascular smooth muscle cells. There is evidence that proproliferative and antiapoptotic states are characterized by a metabolic (glycolytic phenotype and hyperpolarized mitochondria) and electric (downregulation and inhibition of plasmalemmal K(+) channels) remodeling that involves activation of the Akt pathway. Dehydroepiandrosterone (DHEA) is a naturally occurring and clinically used steroid known to inhibit the Akt axis in cancer. We hypothesized that DHEA will prevent and reverse the remodeling that follows vascular injury. METHODS AND RESULTS We used cultured human carotid vascular smooth muscle cell and saphenous vein grafts in tissue culture, stimulated by platelet-derived growth factor to induce proliferation in vitro and the rat carotid injury model in vivo. DHEA decreased proliferation and increased vascular smooth muscle cell apoptosis in vitro and in vivo, reducing vascular remodeling while sparing healthy tissues after oral intake. Using pharmacological (agonists and antagonists of Akt and its downstream target glycogen-synthase-kinase-3beta [GSK-3beta]) and molecular (forced expression of constitutively active Akt1) approaches, we showed that the effects of DHEA were mediated by inhibition of Akt and subsequent activation of GSK-3beta, leading to mitochondrial depolarization, increased reactive oxygen species, activation of redox-sensitive plasmalemmal voltage-gated K(+) channels, and decreased [Ca(2+)](i). These functional changes were accompanied by sustained molecular effects toward the same direction; by decreasing [Ca(2+)](i) and inhibiting GSK-3beta, DHEA inhibited the nuclear factor of activated T cells transcription factor, thus increasing expression of Kv channels (Kv1.5) and contributing to sustained mitochondrial depolarization. These results were independent of any steroid-related effects because they were not altered by androgen and estrogen inhibitors but involved a membrane G protein-coupled receptor. CONCLUSIONS We suggest that the orally available DHEA might be an attractive candidate for the treatment of systemic vascular remodeling, including restenosis, and we propose a novel mechanism of action for this important hormone and drug.
Collapse
Affiliation(s)
- Sébastien Bonnet
- Centre de Recherche de L'Hôtel-Dieu de Québec, 9 Rue McMahon, Québec, Qc, G1R 2J6, Canada.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Lefebvre T, Gonzalez BJ, Vaudry D, Desrues L, Falluel-Morel A, Aubert N, Fournier A, Tonon MC, Vaudry H, Castel H. Paradoxical effect of ethanol on potassium channel currents and cell survival in cerebellar granule neurons. J Neurochem 2009; 110:976-89. [PMID: 19493160 DOI: 10.1111/j.1471-4159.2009.06197.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Transient exposure to ethanol (EtOH) results in a massive neurodegeneration in the developing brain leading to behavioral and cognitive deficits observed in fetal alcohol syndrome. There is now compelling evidence that K+ channels play an important role in the control of programmed cell death. The aim of the present work was to investigate the involvement of K+ channels in the EtOH-induced cerebellar granule cell death and/or survival. At low and high concentrations, EtOH evoked membrane depolarization and hyperpolarization, respectively. Bath perfusion of EtOH (10 mM) depressed the I(A) (transient K+ current) potassium current whereas EtOH (400 mM) provoked a marked potentiation of the specific I(K) (delayed rectifier K+ current) current. Pipette dialysis with GTPgammaS or GDPbetaS did not modify the effects of EtOH (400 mM) on both membrane potential and I(K) current. In contrast, the reversible depolarization and slowly recovering inhibition of I(A) induced by EtOH (10 mM) became irreversible in the presence of GTPgammaS. EtOH (400 mM) induced prodeath responses whereas EtOH (10 mM) and K+ channel blockers promoted cell survival. Altogether, these results indicate that in cerebellar granule cells, EtOH mediates a dual effect on K+ currents partly involved in the control of granule cell death.
Collapse
Affiliation(s)
- Thomas Lefebvre
- Laboratory of Neuroendocrine and Neuronal Cell Differentiation and Communication, European Institute for Peptide Research, University of Rouen, Mont-Saint-Aignan, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Zhai FG, Zhang XH, Wang HL. Fluoxetine protects against monocrotaline-induced pulmonary arterial hypertension: potential roles of induction of apoptosis and upregulation of Kv1.5 channels in rats. Clin Exp Pharmacol Physiol 2009; 36:850-6. [PMID: 19298536 DOI: 10.1111/j.1440-1681.2009.05168.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
1. Suppressing apoptosis and downregulating K(+) channels in pulmonary artery smooth muscle cells (PASMC) have been implicated in the development of pulmonary vascular medial hypertrophy and pulmonary arterial hypertension (PAH). Previous studies have shown that selective serotonin re-uptake inhibitors (SSRIs) protected against PAH. The aim of the present study was to investigate the involvement of Kv1.5 channels and apoptosis in the protective effect of the SSRI fluoxetine against PAH. 2. Monocrotaline (MCT) was used to establish PAH in Wistar rats. Fluoxetine (2 and 10 mg/kg per day) was administered by gavage once a day for 3 weeks. Three weeks after the induction of PAH by MCT, pulmonary haemodynamic measurements and pulmonary artery morphological assessments were undertaken, along with detection of apoptosis and Kv1.5. 3. Fluoxetine (2 and 10 mg/kg per day) decreased pulmonary artery pressure, reduced the right ventricular index and inhibited the increase in medial wall thickness of pulmonary arteries in established PAH. Fluoxetine (10 mg/kg per day) reduced the expression of Bcl-2 and Bcl-xL protein, increased the expression of cleaved caspase 3 protein and enhanced the expression of Kv1.5 protein and mRNA in pulmonary arteries. Furthermore, fluoxetine (10 mg/kg per day) significantly suppressed proliferation and enhanced apoptosis of PASMC in MCT-induced PAH. 4. In conclusion, fluoxetine protects against MCT-induced PAH by suppressing PASMC proliferation, inducing PASMC apoptosis and upregulating Kv1.5 channels.
Collapse
Affiliation(s)
- Feng-Guo Zhai
- Department of Clinical Pharmacology, China Medical University, Shenyang, China
| | | | | |
Collapse
|
46
|
Yang MX, Liu ZX, Zhang S, Jing Y, Zhang SJ, Xie WP, Ma L, Zhu CL, Wang H. Proteomic analysis of the effect of iptakalim on human pulmonary arterial smooth muscle cell proliferation. Acta Pharmacol Sin 2009; 30:175-83. [PMID: 19169269 DOI: 10.1038/aps.2008.30] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
AIM To investigate the anti-proliferative effect of iptakalim (Ipt), a newly selective K(ATP) channel opener, in endothelin-1 (ET-1)-induced human pulmonary arterial smooth muscle cells (PASMCs) using proteomic analysis. METHODS Human PASMCs were incubated with ET-1 (10(-8) mol/L) and ET-1 (10(-8) mol/L) plus iptaklim (10(-5) mol/L) for 24 h. Analysis via 2-DE gel electrophoresis and MALDI-TOF-MS was employed to display the different protein profiles of whole-cell protein from cultures of control, ET-1 treatment alone, and treatment with ET-1 and iptaklim combined. Real time RT-PCR and Western blot analysis were used to confirm the proteomic analysis. RESULTS When iptakalim inhibited the proliferative effect of ET-1 in human PASMCs by opening the K(ATP) channels, the expression of different groups of cellular proteins was changed, including cytoskeleton-associated proteins, plasma membrane proteins and receptors, chaperone proteins, ion transport-associated proteins, and glycolytic and metabolism-associated proteins. We found that iptakalim could inhibit the proliferation of human PASMCs partly by affecting the expression of Hsp60, vimentin, nucleoporin P54 (NUP54) and Bcl-X(L) by opening the K(ATP) channel. CONCLUSION The data suggest that a wide range of signaling pathways may be involved in abolishing ET-1-induced proliferation of human PASMCs following iptakalim treatment.
Collapse
|
47
|
Li Y, Li Q, Wang Z, Liang D, Liang S, Tang X, Guo L, Zhang R, Zhu D. 15-HETE suppresses K+ channel activity and inhibits apoptosis in pulmonary artery smooth muscle cells. Apoptosis 2008; 14:42-51. [DOI: 10.1007/s10495-008-0286-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
48
|
Pritchard DM, Berry D, Przemeck SMC, Campbell F, Edwards SW, Varro A. Gastrin increases mcl-1 expression in type I gastric carcinoid tumors and a gastric epithelial cell line that expresses the CCK-2 receptor. Am J Physiol Gastrointest Liver Physiol 2008; 295:G798-805. [PMID: 18719002 PMCID: PMC2575912 DOI: 10.1152/ajpgi.00015.2008] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Elevated serum concentrations of the hormone gastrin are associated with the development of gastric carcinoid tumors, but the mechanisms of tumor development are not fully understood. We hypothesized that the antiapoptotic effects of gastrin may be implicated and have therefore investigated the role of antiapoptotic members of the bcl-2 family of proteins. AGS-G(R) human gastric carcinoma cells stably transfected with the CCK-2 receptor were used to assess changes in the expression of bcl-2 family members following gastrin treatment and the function of mcl-1 during apoptosis was investigated by use of small-interfering RNA (siRNA). Treatment of AGS-G(R) cells with 10 nM gastrin for 6 h caused maximally increased mcl-1 protein abundance. Gastrin-induced mcl-1 expression was inhibited by the transcription inhibitor actinomycin D and by the protein synthesis inhibitor cycloheximide. Downstream signaling of mcl-1 expression occurred via the CCK-2 receptor, protein kinase C, and MAP kinase pathways, but not via PI 3-kinase. Transfection with mcl-1 siRNA significantly suppressed mcl-1 protein expression and abolished the antiapoptotic effects of gastrin on serum starvation-induced apoptosis. Mcl-1 protein expression was also specifically increased in the type I enterochromaffin-like cell carcinoid tumors of 10 patients with autoimmune atrophic gastritis and hypergastrinemia. Gastrin therefore signals via the CCK-2 receptor, protein kinase C, and MAP kinase to induce expression of antiapoptotic mcl-1 in AGS-G(R) cells, and mcl-1 expression is also increased in human hypergastrinemia-associated type I gastric carcinoid tumors. Gastrin-induced mcl-1 expression may therefore be an important mechanism contributing toward type I gastric carcinoid development.
Collapse
Affiliation(s)
- D. M. Pritchard
- Division of Gastroenterology, School of Clinical Sciences, University of Liverpool; Department of Pathology, Royal Liverpool and Broadgreen University Hospitals National Health Service Trust, Liverpool; and School of Biological Sciences and Division of Physiology, School of Biomedical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - D. Berry
- Division of Gastroenterology, School of Clinical Sciences, University of Liverpool; Department of Pathology, Royal Liverpool and Broadgreen University Hospitals National Health Service Trust, Liverpool; and School of Biological Sciences and Division of Physiology, School of Biomedical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - S. M. C. Przemeck
- Division of Gastroenterology, School of Clinical Sciences, University of Liverpool; Department of Pathology, Royal Liverpool and Broadgreen University Hospitals National Health Service Trust, Liverpool; and School of Biological Sciences and Division of Physiology, School of Biomedical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - F. Campbell
- Division of Gastroenterology, School of Clinical Sciences, University of Liverpool; Department of Pathology, Royal Liverpool and Broadgreen University Hospitals National Health Service Trust, Liverpool; and School of Biological Sciences and Division of Physiology, School of Biomedical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - S. W. Edwards
- Division of Gastroenterology, School of Clinical Sciences, University of Liverpool; Department of Pathology, Royal Liverpool and Broadgreen University Hospitals National Health Service Trust, Liverpool; and School of Biological Sciences and Division of Physiology, School of Biomedical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - A. Varro
- Division of Gastroenterology, School of Clinical Sciences, University of Liverpool; Department of Pathology, Royal Liverpool and Broadgreen University Hospitals National Health Service Trust, Liverpool; and School of Biological Sciences and Division of Physiology, School of Biomedical Sciences, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
49
|
Weir EK, Obreztchikova M, Vargese A, Cabrera JA, Peterson DA, Hong Z. Mechanisms of oxygen sensing: a key to therapy of pulmonary hypertension and patent ductus arteriosus. Br J Pharmacol 2008; 155:300-7. [PMID: 18641675 PMCID: PMC2567893 DOI: 10.1038/bjp.2008.291] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2008] [Accepted: 06/23/2008] [Indexed: 11/08/2022] Open
Abstract
Specialized tissues that sense acute changes in the local oxygen tension include type 1 cells of the carotid body, neuroepithelial bodies in the lungs, and smooth muscle cells of the resistance pulmonary arteries and the ductus arteriosus (DA). Hypoxia inhibits outward potassium current in carotid body type 1 cells, leading to depolarization and calcium entry through L-type calcium channels. Increased intracellular calcium concentration ([Ca+ +]i) leads to exocytosis of neurotransmitters, thus stimulating the carotid sinus nerve and respiration. The same K+ channel inhibition occurs with hypoxia in pulmonary artery smooth muscle cells (PASMCs), causing contraction and providing part of the mechanism of hypoxic pulmonary vasoconstriction (HPV). In the SMCs of the DA, the mechanism works in reverse. It is the shift from hypoxia to normoxia that inhibits K+ channels and causes normoxic ductal contraction. In both PA and DA, the contraction is augmented by release of Ca+ + from the sarcoplasmic reticulum, entry of Ca+ + through store-operated channels (SOC) and by Ca+ + sensitization. The same three 'executive' mechanisms are partly responsible for idiopathic pulmonary arterial hypertension (IPAH). While vasoconstrictor mediators constrict both PA and DA and vasodilators dilate both vessels, only redox changes mimic oxygen by having directly opposite effects on the K+ channels, membrane potential, [Ca(++)]i and tone in the PA and DA. There are several different hypotheses as to how redox might alter tone, which remain to be resolved. However, understanding the mechanism will facilitate drug development for pulmonary hypertension and patent DA.
Collapse
Affiliation(s)
- E K Weir
- Department of Medicine, VA Medical Center and University of Minnesota, Minneapolis, MN 55417, USA.
| | | | | | | | | | | |
Collapse
|
50
|
Rai PR, Cool CD, King JAC, Stevens T, Burns N, Winn RA, Kasper M, Voelkel NF. The cancer paradigm of severe pulmonary arterial hypertension. Am J Respir Crit Care Med 2008; 178:558-64. [PMID: 18556624 DOI: 10.1164/rccm.200709-1369pp] [Citation(s) in RCA: 192] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The plexiform lesions of severe pulmonary arterial hypertension (PAH) are similar in histologic appearance, whether the disease is idiopathic or secondary. Both forms of the disease show actively proliferating endothelial cells without evidence of apoptosis. Here, we discuss the pathobiology of the atypical, angioproliferative endothelial cells in severe PAH. The concept of the endothelial cell as a "quasi-malignant" cell provides a new framework for antiproliferative, antiangiogenic therapy in severe PAH.
Collapse
Affiliation(s)
- Pradeep R Rai
- Department of Pathology, University of Colorado at Denver and Health Sciences Center, Denver, Colorado, USA
| | | | | | | | | | | | | | | |
Collapse
|