1
|
Callejo M, Morales-Cano D, Olivencia MA, Mondejar-Parreño G, Barreira B, Tura-Ceide O, Martínez VG, Serrano-Navarro A, Moreno L, Morrell N, Perros F, Vicente A, Cogolludo A, Perez-Vizcaino F. Vitamin D receptor and its antiproliferative effect in human pulmonary arterial hypertension. Sci Rep 2024; 14:27445. [PMID: 39523384 PMCID: PMC11551162 DOI: 10.1038/s41598-024-78380-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Vitamin D (vitD) deficiency is frequently observed in patients with pulmonary arterial hypertension (PAH) and, in these patients, low levels of vitD correlate with worse prognosis. The aim of this study was to examine the expression and the antiproliferative role of vitD receptor (VDR) and its signalling pathway in the human pulmonary vasculature. VDR presence and expression was analyzed in lungs, pulmonary artery smooth muscle cells (PASMC) and endothelial cells (PAEC) from controls and PAH-patients. VDR expression and VDR-target genes were examined in PASMC treated with calcitriol. The antiproliferative effect of 48 h-calcitriol was studied in PASMC by MTT and BrdU assays. VDR is expressed in PASMC. It is downregulated in lungs and in PASMC, but not in PAEC, from PAH-patients compared to non-hypertensive controls. Calcitriol strongly upregulated VDR expression in PASMC and the VDR target genes KCNK3 (encoding TASK1), BIRC5 (encoding survivin) and BMP4. Calcitriol produced an antiproliferative effect which was diminished by silencing or by pharmacological inhibition of survivin or BMPR2, but not of TASK1. In conclusion, the expression of VDR is low in PAH-patients and can be rescued by calcitriol. VDR exerts an antiproliferative effect in PASMC by modulating survivin and the BMP signalling pathway.
Collapse
MESH Headings
- Humans
- Receptors, Calcitriol/metabolism
- Receptors, Calcitriol/genetics
- Cell Proliferation/drug effects
- Calcitriol/pharmacology
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Pulmonary Artery/drug effects
- Survivin/metabolism
- Survivin/genetics
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Female
- Male
- Pulmonary Arterial Hypertension/metabolism
- Pulmonary Arterial Hypertension/drug therapy
- Pulmonary Arterial Hypertension/pathology
- Pulmonary Arterial Hypertension/genetics
- Potassium Channels, Tandem Pore Domain/metabolism
- Potassium Channels, Tandem Pore Domain/genetics
- Signal Transduction/drug effects
- Bone Morphogenetic Protein 4/metabolism
- Bone Morphogenetic Protein 4/genetics
- Middle Aged
- Bone Morphogenetic Protein Receptors, Type II/metabolism
- Bone Morphogenetic Protein Receptors, Type II/genetics
- Endothelial Cells/metabolism
- Endothelial Cells/drug effects
- Lung/metabolism
- Lung/pathology
- Adult
- Cells, Cultured
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/pathology
- Nerve Tissue Proteins
Collapse
Affiliation(s)
- Maria Callejo
- Department of Pharmacology and Toxicology, Facultad de Medicina, School of Medicine, Universidad Complutense de Madrid, Pza Ramón y Cajal s/n., 28040, Madrid, Spain
- CIBER Enfermedades Respiratorias (CibeRes), Madrid, Spain
| | - Daniel Morales-Cano
- Department of Pharmacology and Toxicology, Facultad de Medicina, School of Medicine, Universidad Complutense de Madrid, Pza Ramón y Cajal s/n., 28040, Madrid, Spain
- CIBER Enfermedades Respiratorias (CibeRes), Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
| | - Miguel A Olivencia
- Department of Pharmacology and Toxicology, Facultad de Medicina, School of Medicine, Universidad Complutense de Madrid, Pza Ramón y Cajal s/n., 28040, Madrid, Spain
- CIBER Enfermedades Respiratorias (CibeRes), Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
| | - Gema Mondejar-Parreño
- Department of Medicine, Division of Cardiovascular Medicine, Stanford Cardiovascular Institute, Stanford, USA
| | - Bianca Barreira
- Department of Pharmacology and Toxicology, Facultad de Medicina, School of Medicine, Universidad Complutense de Madrid, Pza Ramón y Cajal s/n., 28040, Madrid, Spain
- CIBER Enfermedades Respiratorias (CibeRes), Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
| | - Olga Tura-Ceide
- CIBER Enfermedades Respiratorias (CibeRes), Madrid, Spain
- Department of Pulmonary Medicine, Servei de Pneumologia, Hospital Clínic-Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, Villarroel, 170, 08036, Barcelona, Spain
- Translational Research Group on Cardiovascular Respiratory Diseases (CAREs), Institut d'Investigació Biomèdica de Girona (IDIBGI-CERCA), Parc Hospitalari Martí i Julià, Edifici M2, 17190, Salt, Spain
| | - Victor G Martínez
- Biomedical Research Institute I + 12, University Hospital, 12 de Octubre, Madrid, Spain
- Molecular Oncology Unit, CIEMAT (Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas), Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | | | - Laura Moreno
- Department of Pharmacology and Toxicology, Facultad de Medicina, School of Medicine, Universidad Complutense de Madrid, Pza Ramón y Cajal s/n., 28040, Madrid, Spain
- CIBER Enfermedades Respiratorias (CibeRes), Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
| | - Nick Morrell
- Department of Medicine, School of Clinical Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Frédéric Perros
- Laboratoire CarMeN, INSERM U.1060, INRAe U.1397, Université Claude Bernard Lyon1, Pierre Bénite, France
| | - Angeles Vicente
- Department of Cell Biology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Angel Cogolludo
- Department of Pharmacology and Toxicology, Facultad de Medicina, School of Medicine, Universidad Complutense de Madrid, Pza Ramón y Cajal s/n., 28040, Madrid, Spain
- CIBER Enfermedades Respiratorias (CibeRes), Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
| | - Francisco Perez-Vizcaino
- Department of Pharmacology and Toxicology, Facultad de Medicina, School of Medicine, Universidad Complutense de Madrid, Pza Ramón y Cajal s/n., 28040, Madrid, Spain.
- CIBER Enfermedades Respiratorias (CibeRes), Madrid, Spain.
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain.
| |
Collapse
|
2
|
Santos RT, de Sá Freire Onofre ME, de Assis Fernandes Caldeira D, Klein AB, Rocco PRM, Cruz FF, Silva PL. Pharmacological Agents and Potential New Therapies in Pulmonary Arterial Hypertension. Curr Vasc Pharmacol 2024; 22:155-170. [PMID: 38115617 DOI: 10.2174/0115701611266576231211045731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 10/09/2023] [Accepted: 11/15/2023] [Indexed: 12/21/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease characterized by an imbalance between vasoactive mediators, which causes vascular remodeling, increased pulmonary vascular resistance, and right ventricular overload, ultimately leading to heart failure and death. A metabolic theory has been suggested to explain the pathophysiology of PAH whereby abnormalities in mitochondrial biogenesis can trigger a hyperproliferative and apoptosis-resistant phenotype in cardiopulmonary and malignant cells, leading to mitochondrial dysfunction, which in turn causes the Warburg effect. This can culminate in the mitophagy of pulmonary vessels and cardiomyocytes. The present narrative review focuses on the pathophysiology of PAH, the pharmacological agents currently available for its treatment, and promising and challenging areas of therapeutic investigation.
Collapse
Affiliation(s)
- Renata Trabach Santos
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Maria Eduarda de Sá Freire Onofre
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Dayene de Assis Fernandes Caldeira
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Adriane Bello Klein
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Department of Physiology, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Patricia Rieken Macedo Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernanda Ferreira Cruz
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro Leme Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
3
|
Wang E, Zhou S, Zeng D, Wang R. Molecular regulation and therapeutic implications of cell death in pulmonary hypertension. Cell Death Discov 2023; 9:239. [PMID: 37438344 DOI: 10.1038/s41420-023-01535-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/21/2023] [Accepted: 06/28/2023] [Indexed: 07/14/2023] Open
Abstract
Pulmonary hypertension (PH) is a clinical and pathophysiological syndrome caused by changes in pulmonary vascular structure or function that results in increased pulmonary vascular resistance and pulmonary arterial pressure, and it is characterized by pulmonary endothelial dysfunction, pulmonary artery media thickening, pulmonary vascular remodeling, and right ventricular hypertrophy, all of which are driven by an imbalance between the growth and death of pulmonary vascular cells. Programmed cell death (PCD), different from cell necrosis, is an active cellular death mechanism that is activated in response to both internal and external factors and is precisely regulated by cells. More than a dozen PCD modes have been identified, among which apoptosis, autophagy, pyroptosis, ferroptosis, necroptosis, and cuproptosis have been proven to be involved in the pathophysiology of PH to varying degrees. This article provides a summary of the regulatory patterns of different PCD modes and their potential effects on PH. Additionally, it describes the current understanding of this complex and interconnected process and analyzes the therapeutic potential of targeting specific PCD modes as molecular targets.
Collapse
Affiliation(s)
- Enze Wang
- Department of respiratory and critical care medicine, the first affiliated hospital of Anhui medical university, Hefei, 230022, China
| | - Sijing Zhou
- Department of Occupational Disease, Hefei third clinical college of Anhui Medical University, Hefei, 230022, China
| | - Daxiong Zeng
- Department of pulmonary and critical care medicine, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou, 215006, China.
| | - Ran Wang
- Department of respiratory and critical care medicine, the first affiliated hospital of Anhui medical university, Hefei, 230022, China.
| |
Collapse
|
4
|
Blanco I, Marquina M, Tura-Ceide O, Ferrer E, Ramírez AM, Lopez-Meseguer M, Callejo M, Perez-Vizcaino F, Peinado VI, Barberà JA. Survivin inhibition with YM155 ameliorates experimental pulmonary arterial hypertension. Front Pharmacol 2023; 14:1145994. [PMID: 37188265 PMCID: PMC10176173 DOI: 10.3389/fphar.2023.1145994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
Background: Imbalance between cell proliferation and apoptosis underlies the development of pulmonary arterial hypertension (PAH). Current vasodilator treatment of PAH does not target the uncontrolled proliferative process in pulmonary arteries. Proteins involved in the apoptosis pathway may play a role in PAH and their inhibition might represent a potential therapeutic target. Survivin is a member of the apoptosis inhibitor protein family involved in cell proliferation. Objectives: This study aimed to explore the potential role of survivin in the pathogenesis of PAH and the effects of its inhibition. Methods: In SU5416/hypoxia-induced PAH mice we assessed the expression of survivin by immunohistochemistry, western-blot analysis, and RT-PCR; the expression of proliferation-related genes (Bcl2 and Mki67); and the effects of the survivin inhibitor YM155. In explanted lungs from patients with PAH we assessed the expression of survivin, BCL2 and MKI67. Results: SU5416/hypoxia mice showed increased expression of survivin in pulmonary arteries and lung tissue extract, and upregulation of survivin, Bcl2 and Mki67 genes. Treatment with YM155 reduced right ventricle (RV) systolic pressure, RV thickness, pulmonary vascular remodeling, and the expression of survivin, Bcl2, and Mki67 to values similar to those in control animals. Lungs of patients with PAH also showed increased expression of survivin in pulmonary arteries and lung extract, and also that of BCL2 and MKI67 genes, compared with control lungs. Conclusion: We conclude that survivin might be involved in the pathogenesis of PAH and that its inhibition with YM155 might represent a novel therapeutic approach that warrants further evaluation.
Collapse
Affiliation(s)
- Isabel Blanco
- Department of Pulmonary Medicine, Hospital Clínic-University of Barcelona, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
- *Correspondence: Isabel Blanco,
| | - Maribel Marquina
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
| | - Olga Tura-Ceide
- Department of Pulmonary Medicine, Hospital Clínic-University of Barcelona, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
- Biomedical Research Institute-IDIBGI, Girona, Spain
| | - Elisabet Ferrer
- Department of Pulmonary Medicine, Hospital Clínic-University of Barcelona, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Department of Medicine, Addenbrooke’s Hospital, University of Cambridge, Cambridge, United Kingdom
| | - Ana M. Ramírez
- Department of Pulmonary Medicine, Hospital Clínic-University of Barcelona, Barcelona, Spain
| | | | - Maria Callejo
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
- Departament of Pharmacology and Toxicology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Universidad Complutense de Madrid, Madrid, Spain
| | - Francisco Perez-Vizcaino
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
- Departament of Pharmacology and Toxicology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Universidad Complutense de Madrid, Madrid, Spain
| | - Victor Ivo Peinado
- Department of Pulmonary Medicine, Hospital Clínic-University of Barcelona, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
- Department of Experimental Pathology, Institut d’Investigacions Biomèdiques de Barcelona (IIBB-CSIC), Barcelona, Spain
| | - Joan Albert Barberà
- Department of Pulmonary Medicine, Hospital Clínic-University of Barcelona, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
| |
Collapse
|
5
|
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: 24] [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
|
6
|
Liu G, Fu D, Tian H, Dai A. The mechanism of ions in pulmonary hypertension. Pulm Circ 2021; 11:2045894020987948. [PMID: 33614016 PMCID: PMC7869166 DOI: 10.1177/2045894020987948] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 12/23/2020] [Indexed: 12/15/2022] Open
Abstract
Pulmonary hypertension(PH)is a kind of hemodynamic and pathophysiological state, in which the pulmonary artery pressure (PAP) rises above a certain threshold. The main pathological manifestation is pulmonary vasoconstriction and remodelling progressively. More and more studies have found that ions play a major role in the pathogenesis of PH. Many vasoactive substances, inflammatory mediators, transcription-inducing factors, apoptosis mediators, redox substances and translation modifiers can control the concentration of ions inside and outside the cell by regulating the activity of ion channels, which can regulate vascular contraction, cell proliferation, migration, apoptosis, inflammation and other functions. We all know that there are no effective drugs to treat PH. Ions are involved in the occurrence and development of PH, so it is necessary to clarify the mechanism of ions in PH as a therapeutic target for PH. The main ions involved in PH are calcium ion (Ca2+), potassium ion (K+), sodium ion (Na+) and chloride ion (Cl-). Here, we mainly discuss the distribution of these ions and their channels in pulmonary arteries and their role in the pathogenesis of PH.
Collapse
Affiliation(s)
- Guogu Liu
- Department of Graduate School, University of South China,
Hengyang, China
- Department of Respiratory Medicine, Hunan Provincial People’s
Hospital, Changsha, China
| | - Daiyan Fu
- Department of Respiratory Medicine, Hunan Provincial People’s
Hospital, Changsha, China
| | - Heshen Tian
- Department of Graduate School, University of South China,
Hengyang, China
- Department of Respiratory Medicine, Hunan Provincial People’s
Hospital, Changsha, China
| | - Aiguo Dai
- Department of Respiratory Diseases, Hunan University of Chinese
Medicine, Changsha, China
| |
Collapse
|
7
|
Shang L, Wang K, Liu D, Qin S, Huang J, Zhao Y, Pang Y. TMEM16A regulates the cell cycle of pulmonary artery smooth muscle cells in high-flow-induced pulmonary arterial hypertension rat model. Exp Ther Med 2020; 19:3275-3281. [PMID: 32266023 PMCID: PMC7132240 DOI: 10.3892/etm.2020.8589] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 08/20/2019] [Indexed: 12/11/2022] Open
Abstract
High-flow-induced pulmonary arterial hypertension (PAH) has attained global notoriety, the mechanism of which remains elusive. The present study investigated the regulation of Anoctamin-1, also known as transmembrane member 16A (TMEM16A), in the cell cycle progression of pulmonary artery smooth muscle cells (PASMCs) from a PAH rat model induced by high pulmonary blood flow. A total of 30 Sprague-Dawley rats were randomly assigned into control, sham and shunt groups. A rat model of high pulmonary blood flow-induced PAH was established by surgery using abdominal aorta-inferior vena cava fistula. Right ventricular pressure, right ventricular hypertrophy index and pulmonary arteriole structural remodeling were assessed 11 weeks following operation. The cell cycle statuses of PASMCs was assessed via flow cytometry, whereas western blot analysis was performed to measure the expression of cyclin D1, CDK2, p27KIP and cyclin E in primary PASMCs isolated from rats. The expression of cyclin E and cyclin D1 was revealed to be increased in the shunt group compared with the control group, which was accompanied with an increased expression of TMEM16A in the shunt group. Changes in the ratio of PASMCs in the G0/G1, S and G2/M phases of cycle induced by PAH were reversed by TMEM16A knockdown. The expression of cyclin E and cyclin D1 in the shunt group was significantly higher compared with the control group in vitro, which was reversed by TMEM16A-siRNA transfection. In conclusion, TMEM16A may be involved in high pulmonary blood flow-induced PAH by regulating PASMC cell cycle progression.
Collapse
Affiliation(s)
- Lifeng Shang
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guanxi 530021, P.R. China
| | - Kai Wang
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Dongli Liu
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guanxi 530021, P.R. China
| | - Suyuan Qin
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guanxi 530021, P.R. China
| | - Jinglin Huang
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guanxi 530021, P.R. China
| | - Yijue Zhao
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guanxi 530021, P.R. China
| | - Yusheng Pang
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guanxi 530021, P.R. China
| |
Collapse
|
8
|
Li G, Zhang H, Zhao L, Zhang Y, Yan D, Liu Y, Su J, Fan X. The expression of survivin in irreversible pulmonary arterial hypertension rats and its value in evaluating the reversibility of pulmonary arterial hypertension secondary to congenital heart disease. Pulm Circ 2019; 9:2045894019859480. [PMID: 31428312 PMCID: PMC6681502 DOI: 10.1177/2045894019859480] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 05/24/2019] [Indexed: 01/07/2023] Open
Abstract
The reversibility of pulmonary arterial hypertension (PAH) determines the
operability of congenital heart disease (CHD) complicating with PAH, but it
lacks a method for evaluating the reversibility. The current study aims to
investigate the serum survivin level in irreversible PAH rats and to explore its
potential as a biomarker for evaluating the reversibility of PAH in CHD
patients. Irreversible PAH rats were characterized by prominent obstructive
lesions resulting from the intimal formation, which was associated with
decreased apoptosis and increased survivin expression, while reversible PAH rats
were featured by medial hypertrophy resulting in mild occlusion, with increased
apoptosis and unchanged survivin expression. In addition, the serum survivin was
significantly increased in irreversible PAH rats when compared to both
reversible PAH and control rats, and a positive correlation of serum survivin
with survivin expression in the lung was confirmed. Third, the preoperative
serum survivin was significantly higher in patients with irreversible CHD-PAH
than in these with reversible CHD-PAH, and significant correlations between the
serum survivin and BNP, preoperative pulmonary vascular resistance index, and
postoperative mean pulmonary arterial pressure were also identified. In
conclusion, the increased survivin level is a feature of irreversible PAH and
the serum survivin represents a candidate biomarker reflecting the operability
of CHD-PAH patients.
Collapse
Affiliation(s)
- Gang Li
- Pediatric Cardiac Center, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Han Zhang
- Pediatric Cardiac Center, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Lei Zhao
- Department of Molecular Physiology and Biophysics, Holden Comprehensive Cancer Center, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Yaozhong Zhang
- Pediatric Cardiac Center, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Daole Yan
- Pediatric Cardiac Center, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Yinglong Liu
- Pediatric Cardiac Center, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Junwu Su
- Pediatric Cardiac Center, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Xiangming Fan
- Pediatric Cardiac Center, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| |
Collapse
|
9
|
Tian H, Fan F, Geng J, Deng J, Tian H. Beraprost Upregulates KV Channel Expression and Function via EP4 Receptor in Pulmonary Artery Smooth Muscle Cells Obtained from Rats with Hypoxia-Induced Pulmonary Hypertension. J Vasc Res 2019; 56:204-214. [PMID: 31189158 DOI: 10.1159/000500424] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 04/15/2019] [Indexed: 11/19/2022] Open
Abstract
The reduced expression and function of voltage-dependent potassium (KV) channels have been involved in the pathogenesis of hypoxia-induced pulmonary hypertension (HPH), leading to pulmonary vasoconstriction and vascular remodeling, while the upregulation of KV channels is of therapeutic significance for pulmonary hypertension. Beraprost sodium (BPS) has been shown to be effective in patients with pulmonary hypertension. However, the effect of BPS on O2-sensitive KV channels in pulmonary artery smooth muscle cells (PASMCs) remains unclear. In the present study, the effect of BPS on rats with HPH was observed, and the influence of BPS on the expression and function of O2-sensitive KV channels in PASMCs was investigated. The results revealed that BPS reduced mean pulmonary artery pressure, suppressed right ventricular hypertrophy, and attenuated the remodeling of pulmonary arteries in rats exposed to discontinuous hypoxia for 4 weeks (8 h/day). This was accompanied with the significantly upregulated expression of KV channel α-subunits (KV1.2, KV1.5 and KV2.1) and O2-sensitive voltage-gated K+ (KV) channel current (IK(V)) in small pulmonary arteries in HPH model rats, as well as in hypoxia-induced PASMCs. Furthermore, in vitrostudies have revealed that the upregulation of BPS on O2-sensitive KV channels was significantly inhibited after treatment with prostaglandin E2 receptor subtype EP4 antagonist GW627368X. Taken together, these results suggest that BPS attenuates the development of HPH through the upregulation of O2-sensitive KV channels, which was probably via the EP4 receptor-related pathway.
Collapse
Affiliation(s)
- Hua Tian
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Fenling Fan
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jie Geng
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jizhao Deng
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Hongyan Tian
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China,
| |
Collapse
|
10
|
Dogan MF, Yildiz O, Arslan SO, Ulusoy KG. Potassium channels in vascular smooth muscle: a pathophysiological and pharmacological perspective. Fundam Clin Pharmacol 2019; 33:504-523. [PMID: 30851197 DOI: 10.1111/fcp.12461] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 02/28/2019] [Accepted: 03/07/2019] [Indexed: 12/23/2022]
Abstract
Potassium (K+ ) ion channel activity is an important determinant of vascular tone by regulating cell membrane potential (MP). Activation of K+ channels leads to membrane hyperpolarization and subsequently vasodilatation, while inhibition of the channels causes membrane depolarization and then vasoconstriction. So far five distinct types of K+ channels have been identified in vascular smooth muscle cells (VSMCs): Ca+2 -activated K+ channels (BKC a ), voltage-dependent K+ channels (KV ), ATP-sensitive K+ channels (KATP ), inward rectifier K+ channels (Kir ), and tandem two-pore K+ channels (K2 P). The activity and expression of vascular K+ channels are changed during major vascular diseases such as hypertension, pulmonary hypertension, hypercholesterolemia, atherosclerosis, and diabetes mellitus. The defective function of K+ channels is commonly associated with impaired vascular responses and is likely to become as a result of changes in K+ channels during vascular diseases. Increased K+ channel function and expression may also help to compensate for increased abnormal vascular tone. There are many pharmacological and genotypic studies which were carried out on the subtypes of K+ channels expressed in variable amounts in different vascular beds. Modulation of K+ channel activity by molecular approaches and selective drug development may be a novel treatment modality for vascular dysfunction in the future. This review presents the basic properties, physiological functions, pathophysiological, and pharmacological roles of the five major classes of K+ channels that have been determined in VSMCs.
Collapse
Affiliation(s)
- Muhammed Fatih Dogan
- Department of Pharmacology, Ankara Yildirim Beyazit University, Bilkent, Ankara, 06010, Turkey
| | - Oguzhan Yildiz
- Department of Pharmacology, Gulhane Faculty of Medicine, University of Health Sciences, Etlik, Ankara, 06170, Turkey
| | - Seyfullah Oktay Arslan
- Department of Pharmacology, Ankara Yildirim Beyazit University, Bilkent, Ankara, 06010, Turkey
| | - Kemal Gokhan Ulusoy
- Department of Pharmacology, Gulhane Faculty of Medicine, University of Health Sciences, Etlik, Ankara, 06170, Turkey
| |
Collapse
|
11
|
Kosanovic D, Platzek SM, Petrovic A, Sydykov A, Maripov A, Mamazhakypov A, Sartmyrzaeva M, Muratali Uulu K, Cholponbaeva M, Toktosunova A, Omurzakova N, Duishobaev M, Vroom C, Pak O, Weissmann N, Ghofrani HA, Sarybaev A, Schermuly RT. Circulating Apoptotic Signals During Acute and Chronic Exposure to High Altitude in Kyrgyz Population. Front Physiol 2019; 10:54. [PMID: 30804801 PMCID: PMC6370645 DOI: 10.3389/fphys.2019.00054] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 01/17/2019] [Indexed: 12/21/2022] Open
Abstract
Background: Circulating apoptotic signals (CASs) have been described in the pathologies associated with dysregulated apoptosis, such as cancer, heart diseases, and pulmonary hypertension (PH). However, nothing is known about the expression profiles of these markers in the circulation of humans exposed to acute and chronic effects of high altitude (HA). Methods: Gene expression levels of different apoptotic signals (ASs) were analyzed in human pulmonary artery smooth muscle cells (PASMCs) upon hypoxia incubation. In addition, we measured the plasma values of relevant CAS in Kyrgyz volunteers during acute and chronic exposure to HA. Finally, we analyzed the effects of pro-apoptotic mediator Fas ligand (FasL) on apoptosis and proliferation of human PASMCs. Results: Several cellular AS were increased in PASMCs exposed to hypoxia, in comparison to normoxia condition. Among analyzed CAS, there was a prominent reduction of FasL in lowlanders exposed to HA environment. Furthermore, decreased circulatory levels of FasL were found in highlanders with HA-induced PH (HAPH), as compared to the lowland controls. Furthermore, FasL concentration in plasma negatively correlated with tricuspid regurgitant gradient values. Finally, FasL exerted pro-apoptotic and anti-proliferative effects on PASMCs. Conclusion: Our data demonstrated that circulating levels of FasL are reduced during acute and chronic exposure to HA environment. In addition, dysregulated FasL may play a role in the context of HAPH due to its relevant functions on apoptosis and proliferation of PASMCs.
Collapse
Affiliation(s)
- Djuro Kosanovic
- Chair for Pulmonary Pharmacotherapy, Member of the German Center for Lung Research, Universities of Giessen and Marburg Lung Center, Giessen, Germany.,Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Simon Maximilian Platzek
- Chair for Pulmonary Pharmacotherapy, Member of the German Center for Lung Research, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - Aleksandar Petrovic
- Chair for Pulmonary Pharmacotherapy, Member of the German Center for Lung Research, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - Akylbek Sydykov
- Chair for Pulmonary Pharmacotherapy, Member of the German Center for Lung Research, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - Abdirashit Maripov
- Kyrgyz National Centre for Cardiology and Internal Medicine, named after Academician Mirsaid Mirrakhimov, Bishkek, Kyrgyzstan
| | - Argen Mamazhakypov
- Chair for Pulmonary Pharmacotherapy, Member of the German Center for Lung Research, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - Meerim Sartmyrzaeva
- Kyrgyz National Centre for Cardiology and Internal Medicine, named after Academician Mirsaid Mirrakhimov, Bishkek, Kyrgyzstan
| | - Kubatbek Muratali Uulu
- Kyrgyz National Centre for Cardiology and Internal Medicine, named after Academician Mirsaid Mirrakhimov, Bishkek, Kyrgyzstan
| | - Meerim Cholponbaeva
- Kyrgyz National Centre for Cardiology and Internal Medicine, named after Academician Mirsaid Mirrakhimov, Bishkek, Kyrgyzstan
| | - Aidana Toktosunova
- Kyrgyz National Centre for Cardiology and Internal Medicine, named after Academician Mirsaid Mirrakhimov, Bishkek, Kyrgyzstan
| | - Nazgul Omurzakova
- Kyrgyz National Centre for Cardiology and Internal Medicine, named after Academician Mirsaid Mirrakhimov, Bishkek, Kyrgyzstan
| | - Melis Duishobaev
- Kyrgyz National Centre for Cardiology and Internal Medicine, named after Academician Mirsaid Mirrakhimov, Bishkek, Kyrgyzstan
| | - Christina Vroom
- Chair for Pulmonary Pharmacotherapy, Member of the German Center for Lung Research, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - Oleg Pak
- Chair for Pulmonary Pharmacotherapy, Member of the German Center for Lung Research, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - Norbert Weissmann
- Chair for Pulmonary Pharmacotherapy, Member of the German Center for Lung Research, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - Hossein Ardeschir Ghofrani
- Chair for Pulmonary Pharmacotherapy, Member of the German Center for Lung Research, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - Akpay Sarybaev
- Kyrgyz National Centre for Cardiology and Internal Medicine, named after Academician Mirsaid Mirrakhimov, Bishkek, Kyrgyzstan
| | - Ralph Theo Schermuly
- Chair for Pulmonary Pharmacotherapy, Member of the German Center for Lung Research, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| |
Collapse
|
12
|
Mazzio EA, Lewis CA, Elhag R, Soliman KF. Effects of Sepantronium Bromide (YM-155) on the Whole Transcriptome of MDA-MB-231 Cells: Highlight on Impaired ATR/ATM Fanconi Anemia DNA Damage Response. Cancer Genomics Proteomics 2018; 15:249-264. [PMID: 29976630 PMCID: PMC6070710 DOI: 10.21873/cgp.20083] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 05/16/2018] [Accepted: 05/25/2018] [Indexed: 12/18/2022] Open
Abstract
Sepantronium bromide (YM-155) is believed to elicit apoptosis and mitotic arrest in tumor cells by reducing (BIRC5, survivin) mRNA. In this study, we monitored changes in survivin mRNA and protein after treating MDA-MB-231 cells with YM-155 concurrent with evaluation of whole transcriptomic (WT) mRNA and long intergenic non-coding RNA at 2 time points: 8 h sub-lethal (83 ng/mL) and 20 h at the LC50 (14.6 ng/mL). The data show a tight association between cell death and the precipitating loss of survivin protein and mRNA (-2.67 fold-change (FC), p<0.001) at 20 h, questioning if the decline in survivin is attributed to cell death or drug impact. The meager loss of survivin mRNA was overshadowed by enormous differential change to the WT in both magnitude and significance for over 2000 differentially up/down-regulated transcripts: (+22 FC to -12 FC, p<0.001). The data show YM-155 to up-regulate transcripts in control of circadian rhythm (NOCT, PER, BHLHe40, NFIL3), tumor suppression (SIK1, FOSB), histone methylation (KDM6B) and negative feedback of NF-kappa B signaling (TNFAIP3). Down-regulated transcripts by YM-155 include glucuronidase (GUSBP3), numerous micro-RNAs, DNA damage repair elements (CENPI, POLQ, RAD54B) and the most affected system was the ataxia-telangiectasia mutated (ATM)/Fanconi anemia E3 monoubiquitin ligase core complexes (FANC transcripts - A/B/E/F/G/M), FANC2, FANCI, BRCA1, BRCA2, RAD51, PALB2 gene and ATR (ATM- and Rad3-Related) pathway. In conclusion, these findings suggest that a primary target of YM-155 is the loss of replicative DNA repair systems.
Collapse
Affiliation(s)
- Elizabeth A Mazzio
- College of Pharmacy & Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, U.S.A
| | - Charles A Lewis
- College of Pharmacy & Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, U.S.A
| | - Rashid Elhag
- College of Pharmacy & Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, U.S.A
| | - Karam F Soliman
- College of Pharmacy & Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, U.S.A.
| |
Collapse
|
13
|
Jia Y, Hu R, Li P, Zheng Y, Wang Y, Ma X. DEC1 is required for anti-apoptotic activity of gastric cancer cells under hypoxia by promoting Survivin expression. Gastric Cancer 2018; 21:632-642. [PMID: 29204860 DOI: 10.1007/s10120-017-0780-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 11/15/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Human differentiated embryonic chondrocyte-expressed gene 1 (DEC1), which has been reported to be overexpressed in several types of cancer, is associated with tumorigenesis through participation in several biological processes. However, the complex mechanisms underlying DEC1 during carcinogenesis are controversial, and its roles in the development and malignancy of gastric cancer (GC) remain unclear. METHODS We measured DEC1 expression in human GC cell lines. DEC1 levels in GC cells were downregulated by shRNA lentivirus infection. We also evaluated the effect of DEC1 downregulation on xenograft growth in vivo. The viability and apoptosis of the cells were assayed using the CCK8 assay and flow cytometry. The levels of DEC1, Survivin, and Bcl-2 were evaluated by Western blotting. Luciferase reporter was used to verify the downstream target of DEC1. The association of DEC1 and Survivin expression with prognosis was investigated by immunohistochemistry. RESULTS Downregulation of DEC1 inhibits GC cell proliferation in vitro and tumorigenicity in vivo. We observed that hypoxia-induced expression of DEC1 protects GC cells from apoptosis via transcriptional upregulation of Survivin. Furthermore, positive correlations between DEC1 with Survivin expression were observed in tissue sections from GC patients. Notably, GC patients with high expression levels of DEC1 and Survivin showed poor prognosis. CONCLUSIONS DEC1 acts as an anti-apoptotic regulator in GC cells under hypoxia by promoting Survivin expression. Our study demonstrates the critical role of the DEC1 in oncogenesis and highlights a novel role for DEC1 in the regulation of cell apoptosis in GC.
Collapse
Affiliation(s)
- Yanfei Jia
- Central Laboratory, Jinan Central Hospital Affiliated to Shandong University, Jinan, 250013, People's Republic of China
| | - Rui Hu
- Department of Reproduction, Jinan Central Hospital Affiliated to Shandong University, Jinan, 250013, People's Republic of China
| | - Ping Li
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, 250012, People's Republic of China
| | - Yan Zheng
- Central Laboratory, Jinan Central Hospital Affiliated to Shandong University, Jinan, 250013, People's Republic of China
| | - Yunshan Wang
- Central Laboratory, Jinan Central Hospital Affiliated to Shandong University, Jinan, 250013, People's Republic of China. .,Shandong Province Key Lab of Tumor Target Molecule, Jinan Central Hospital Affiliated to Shandong University, Jinan, 250013, People's Republic of China.
| | - Xiaoli Ma
- Central Laboratory, Jinan Central Hospital Affiliated to Shandong University, Jinan, 250013, People's Republic of China.
| |
Collapse
|
14
|
Al-Owais MM, Hettiarachchi NT, Boyle JP, Scragg JL, Elies J, Dallas ML, Lippiat JD, Steele DS, Peers C. Multiple mechanisms mediating carbon monoxide inhibition of the voltage-gated K + channel Kv1.5. Cell Death Dis 2017; 8:e3163. [PMID: 29095440 PMCID: PMC5775415 DOI: 10.1038/cddis.2017.568] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 09/19/2017] [Accepted: 09/21/2017] [Indexed: 12/13/2022]
Abstract
The voltage-gated K+ channel has key roles in the vasculature and in atrial excitability and contributes to apoptosis in various tissues. In this study, we have explored its regulation by carbon monoxide (CO), a product of the cytoprotective heme oxygenase enzymes, and a recognized toxin. CO inhibited recombinant Kv1.5 expressed in HEK293 cells in a concentration-dependent manner that involved multiple signalling pathways. CO inhibition was partially reversed by superoxide dismutase mimetics and by suppression of mitochondrial reactive oxygen species. CO also elevated intracellular nitric oxide (NO) levels. Prevention of NO formation also partially reversed CO inhibition of Kv1.5, as did inhibition of soluble guanylyl cyclase. CO also elevated intracellular peroxynitrite levels, and a peroxynitrite scavenger markedly attenuated the ability of CO to inhibit Kv1.5. CO caused nitrosylation of Kv1.5, an effect that was also observed in C331A and C346A mutant forms of the channel, which had previously been suggested as nitrosylation sites within Kv1.5. Augmentation of Kv1.5 via exposure to hydrogen peroxide was fully reversed by CO. Native Kv1.5 recorded in HL-1 murine atrial cells was also inhibited by CO. Action potentials recorded in HL-1 cells were increased in amplitude and duration by CO, an effect mimicked and occluded by pharmacological inhibition of Kv1.5. Our data indicate that Kv1.5 is a target for modulation by CO via multiple mechanisms. This regulation has important implications for diverse cellular functions, including excitability, contractility and apoptosis.
Collapse
Affiliation(s)
- Moza M Al-Owais
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Nishani T Hettiarachchi
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - John P Boyle
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Jason L Scragg
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Jacobo Elies
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Mark L Dallas
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Jon D Lippiat
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Derek S Steele
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Chris Peers
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
15
|
Sugimoto K, Nakazato K, Sato A, Suzuki S, Yoshihisa A, Machida T, Saitoh SI, Sekine H, Takeishi Y. Autoimmune disease mouse model exhibits pulmonary arterial hypertension. PLoS One 2017; 12:e0184990. [PMID: 28926602 PMCID: PMC5605000 DOI: 10.1371/journal.pone.0184990] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 09/05/2017] [Indexed: 11/30/2022] Open
Abstract
Background Pulmonary arterial hypertension is often associated with connective tissue disease. Although there are some animal models of pulmonary hypertension, an autoimmune disease-based model has not yet been reported. MRL/lpr mice, which have hypergammaglobulinemia, produce various autoimmune antibodies, and develop vasculitis and nephritis spontaneously. However, little is known about pulmonary circulation in these mice. In the present study, we examined the pulmonary arterial pressure in MRL/lpr mice. Methods and results We used female MRL/lpr mice aged between 12 and 14 weeks. Fluorescent immunostaining showed that there was no deposition of immunoglobulin or C3 in the lung tissue of the MRL/lpr mice. Elevation of interferon-γ and interleukin-6 was recognized in the lung tissue of the MRL/lpr mice. Right ventricular systolic pressure, Fulton index and the ratio of right ventricular weight to body weight in the MRL/lpr mice were significantly higher than those in wild type mice with same background (C57BL/6). The medial smooth muscle area and the proportion of muscularized vessels in the lung tissue of the MRL/lpr mice were larger than those of the C57BL/6 mice. Western blot analysis demonstrated markedly elevated levels of prepro-endothelin-1 and survivin as well as decreased endothelial nitric oxide synthase phosphorylation in the lung tissue of the MRL/lpr mice. Terminal deoxynucleotidyl-transferase-mediated dUTP nick end-labeling assay showed the resistance against apoptosis of pulmonary arterial smooth muscle cells in the MRL/lpr mice. Conclusion We showed that MRL/lpr mice were complicated with pulmonary hypertension. MRL/lpr mice appeared to be a useful model for studying the mechanism of pulmonary hypertension associated with connective tissue diseases.
Collapse
Affiliation(s)
- Koichi Sugimoto
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
- Department of Pulmonary Hypertension, Fukushima Medical University, Fukushima, Japan
- * E-mail:
| | - Kazuhiko Nakazato
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| | - Akihiko Sato
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| | - Satoshi Suzuki
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| | - Akiomi Yoshihisa
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| | - Takeshi Machida
- Department of Immunology, Fukushima Medical University, Fukushima, Japan
| | - Shu-ichi Saitoh
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| | - Hideharu Sekine
- Department of Immunology, Fukushima Medical University, Fukushima, Japan
| | - Yasuchika Takeishi
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
- Department of Pulmonary Hypertension, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
16
|
Gravina G, Wasén C, Garcia-Bonete MJ, Turkkila M, Erlandsson MC, Töyrä Silfverswärd S, Brisslert M, Pullerits R, Andersson KM, Katona G, Bokarewa MI. Survivin in autoimmune diseases. Autoimmun Rev 2017; 16:845-855. [PMID: 28564620 DOI: 10.1016/j.autrev.2017.05.016] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Accepted: 05/18/2017] [Indexed: 12/11/2022]
Abstract
Survivin is a protein functionally important for cell division, apoptosis, and possibly, for micro-RNA biogenesis. It is an established marker of malignant cell transformation. In non-malignant conditions, the unique properties of survivin make it indispensable for homeostasis of the immune system. Indeed, it is required for the innate and adaptive immune responses, controlling differentiation and maintenance of CD4+ and CD8+ memory T-cells, and in B cell maturation. Recently, survivin has emerged as an important player in the pathogenesis of autoimmune diseases. Under the conditions of unreserved inflammation, survivin enhances antigen presentation, maintains persistence of autoreactive cells, and supports production of autoantibodies. In this context, survivin takes its place as a diagnostic and prognostic marker in rheumatoid arthritis, psoriasis, systemic sclerosis and pulmonary arterial hypertension, neuropathology and multiple sclerosis, inflammatory bowel diseases and oral lichen planus. In this review, we summarise the knowledge about non-malignant properties of survivin and focus on its engagement in cellular and molecular pathology of autoimmune diseases. The review highlights utility of survivin measures for clinical applications. It provides rational for the survivin inhibiting strategies and presents results of recent reports on survivin inhibition in modern therapies of cancers and autoimmune diseases.
Collapse
Affiliation(s)
- G Gravina
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - C Wasén
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - M J Garcia-Bonete
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden.
| | - M Turkkila
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - M C Erlandsson
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Rheumatology Clinic, Sahlgrenska University Hospital, Gothenburg, Sweden.
| | - S Töyrä Silfverswärd
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - M Brisslert
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - R Pullerits
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden.
| | - K M Andersson
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - G Katona
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden.
| | - M I Bokarewa
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Rheumatology Clinic, Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
17
|
Taglieri L, De Iuliis F, Giuffrida A, Giantulli S, Silvestri I, Scarpa S. Resistance to the mTOR inhibitor everolimus is reversed by the downregulation of survivin in breast cancer cells. Oncol Lett 2017; 14:3832-3838. [PMID: 28927154 PMCID: PMC5587981 DOI: 10.3892/ol.2017.6597] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 04/07/2017] [Indexed: 12/17/2022] Open
Abstract
Everolimus (RAD001) is an inhibitor of mammalian target of rapamycin used in combination with exemestane to treat hormone receptor-positive advanced breast cancer. However, not all patients are equally sensitive to RAD001 and certain patients develop resistance. Therefore, the present study analyzed the mechanisms involved in the resistance of breast cancer cells to RAD001 in order to identify a potential tool to overcome it. The effects of RAD001 on the inhibition of cell viability, on the induction of apoptosis and autophagy and on the regulation of survivin, an anti-apoptotic protein, were evaluated in two breast cancer cell lines: BT474 (luminal B) and MCF7 (luminal A). RAD001 was demonstrated to induce autophagy in the two cell lines at following a short period of treatment (4 h) and to induce apoptosis exclusively in BT474 cells following longer periods of treatment (48 h). RAD001 induced the downregulation of survivin in BT474 cells and its upregulation in MCF7 cells. Consequently, inhibiting survivin with YM155 resulted in the acquired resistance of MCF7 cells to RAD001 being reverted, restoring RAD001-induced apoptosis. These data demonstrated that RAD001 exerted anti-proliferative and pro-apoptotic effects on breast cancer cells, but that these effects were repressed by the simultaneous up-regulation of survivin. Finally, the results demonstrated that inhibiting the expression of survivin resulted in the restoration of the anti-neoplastic activity of RAD001.
Collapse
Affiliation(s)
- Ludovica Taglieri
- Department of Experimental Medicine, Sapienza University, I-00161 Rome, Italy
| | - Francesca De Iuliis
- Department of Experimental Medicine, Sapienza University, I-00161 Rome, Italy
| | - Anna Giuffrida
- Department of Experimental Medicine, Sapienza University, I-00161 Rome, Italy
| | - Sabrina Giantulli
- Department of Molecular Medicine, Sapienza University, I-00161 Rome, Italy
| | - Ida Silvestri
- Department of Molecular Medicine, Sapienza University, I-00161 Rome, Italy
| | - Susanna Scarpa
- Department of Experimental Medicine, Sapienza University, I-00161 Rome, Italy
| |
Collapse
|
18
|
Li KL, Wang YF, Qin JR, Wang F, Yang YT, Zheng LW, Li MH, Kong J, Zhang W, Yang HY. Rapamycin enhances the anti-angiogenesis and anti-proliferation ability of YM155 in oral squamous cell carcinoma. Tumour Biol 2017; 39:1010428317706213. [PMID: 28618939 DOI: 10.1177/1010428317706213] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
YM155, a small molecule inhibitor of survivin, has been studied in many tumors. It has been shown that YM155 inhibited oral squamous cell carcinoma through promoting apoptosis and autophagy and inhibiting proliferation. It was found that YM155 also inhibited the oral squamous cell carcinoma-mediated angiogenesis through the inactivation of the mammalian target of rapamycin pathway. Rapamycin, a mammalian target of rapamycin inhibitor, played an important role in the proliferation and angiogenesis of oral squamous cell carcinoma cell lines. In our study, cell proliferation assay, transwell assay, tube formation assay, and western blot assay were used to investigate the synergistic effect of rapamycin on YM155 in oral squamous cell carcinoma. Either in vitro or in vivo, rapamycin and YM155 exerted a synergistic effect on the inhibition of survivin and vascular endothelial growth factor through mammalian target of rapamycin pathway. Overall, our results revealed that low-dose rapamycin strongly promoted the sensitivity of oral squamous cell carcinoma cell lines to YM155.
Collapse
Affiliation(s)
- Kong-Liang Li
- 1 Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yu-Fan Wang
- 1 Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jia-Ruo Qin
- 2 Department of Stomatology, Tenth People's Hospital of Tongji University, Shanghai, China
| | - Feng Wang
- 1 Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yong-Tao Yang
- 1 Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Li-Wu Zheng
- 3 Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Ming-Hua Li
- 4 Central Laboratory, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jie Kong
- 5 Department of Stomatology, Shenzhen Children's Hospital, Shenzhen, China
| | - Wei Zhang
- 6 Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Hong-Yu Yang
- 1 Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
19
|
Lu Y, Huang J, Geng S, Chen H, Song C, Zhu S, Zhao S, Yuan M, Li X, Hu H. MitoKATP regulating HIF/miR210/ISCU signaling axis and formation of a positive feedback loop in chronic hypoxia-induced PAH rat model. Exp Ther Med 2017; 13:1697-1701. [PMID: 28565755 PMCID: PMC5443288 DOI: 10.3892/etm.2017.4161] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 09/29/2016] [Indexed: 02/06/2023] Open
Abstract
In the present study, we studied the mechanism of mitochondrial ATP-sensitive potassium (mitoKATP) channels regulating hypoxia-inducible factor (HIF)-1α/microRNA (miR)-210/mitochondrial iron-sulfur protein integrin (ISCU) signaling axis and forming a positive feedback loop in chronic hypoxia-induced pulmonary arterial hypertension (PAH) by using in vivo animal model. Two hundred healthy adult SPF Sprague-Dawley rats were randomly divided into five groups: Control, a mimic miR-210 agent (mimic-210) intervention, a miR-210 inhibitor (anti-210) intervention, a chronic PAH and an anti-210 intervention PAH groups, with 40 rats in each group. After the chronic PAH rat model was successfully established, the rats were intervened with mimic-210 and anti-210. The pulmonary artery smooth muscle cells (PASMCs) of rats in each group were acutely isolated and the activity of mitoKATP and mitochondria-derived oxygen free radicals reactive oxygen species (ROS) was detected. RT-qPCR was used to detect the gene of HIF-1α/miR-210/ISCU and western blot analysis was used to detect the protein of HIF-1α and ISCU. The gene and protein expression were detected again after mitoKATP-specific opener diazoxide and blocker 5-HD was given via tail vein and took effect on each group of rats, respectively. Additionally, the indicators were detected again after ISCU recombinant protein was given via tail vein and ISCU small interfering RNA (siRNA) via nasal feeding and took effect on each group of rats, respectively. It was found that the activity of mitoKATP and ROS and the gene and protein levels of HIF-1α/miR-210/ISCU of the mimic-210 group were significantly higher than those of the control group while that of the anti-210 group was significantly reduced (P<0.05). The indicators in the chronic PAH group were significantly higher than those of the control group while those of the anti-210 intervention PAH group were significantly reduced (P<0.05). The indicators of all the groups were increased after being given mitoKATP specific opener diazoxide. The indicators of all the groups were significantly reduced after receiving blocker 5-HD (P<0.05). The indicators of all the groups were significantly reduced after given ISCU recombinant protein. The indicators of all the groups increased following ISCU siRNA, and there was a statistically significant difference (P<0.05). In conclusion, the mechanism of mitoKATP regulating the HIF-1α/miR-210/ISCU signaling axis and formation of a positive feedback loop exists in the PAH rat model.
Collapse
Affiliation(s)
- Yang Lu
- Department of Respiratory Medicine, Key Laboratory for Molecular Diagnosis of Hubei Province, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430017, P.R. China
| | - Jing Huang
- Department of Respiratory Medicine, Key Laboratory for Molecular Diagnosis of Hubei Province, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430017, P.R. China
| | - Shuang Geng
- Department of Respiratory Medicine, Key Laboratory for Molecular Diagnosis of Hubei Province, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430017, P.R. China
| | - Hao Chen
- Department of Respiratory Medicine, Key Laboratory for Molecular Diagnosis of Hubei Province, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430017, P.R. China
| | - Cheng Song
- Department of Respiratory Medicine, Key Laboratory for Molecular Diagnosis of Hubei Province, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430017, P.R. China
| | - Shan Zhu
- Department of Respiratory Medicine, Key Laboratory for Molecular Diagnosis of Hubei Province, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430017, P.R. China
| | - Su Zhao
- Department of Respiratory Medicine, Key Laboratory for Molecular Diagnosis of Hubei Province, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430017, P.R. China
| | - Mingli Yuan
- Department of Respiratory Medicine, Key Laboratory for Molecular Diagnosis of Hubei Province, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430017, P.R. China
| | - Xueying Li
- Department of Respiratory Medicine, Key Laboratory for Molecular Diagnosis of Hubei Province, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430017, P.R. China
| | - Hongling Hu
- Department of Respiratory Medicine, Key Laboratory for Molecular Diagnosis of Hubei Province, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430017, P.R. China
| |
Collapse
|