1
|
Agrawal V, Hemnes AR. Integrin Targeting Therapies in Pulmonary Arterial Hypertension: A Roadmap for Traversing the Translational Valley of Death? Circulation 2025; 151:1184-1186. [PMID: 40258053 PMCID: PMC12014143 DOI: 10.1161/circulationaha.125.074059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/23/2025]
Affiliation(s)
- Vineet Agrawal
- Pulmonary Circulation Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Veterans Affairs, Nashville, TN, USA
| | - Anna R. Hemnes
- Pulmonary Circulation Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Division of Allergy, Pulmonary, and Critical Care Medicine, Nashville, TN, USA
| |
Collapse
|
2
|
Chu Z, Li Y, Ge J. Identification of nitric oxide-related key genes in pulmonary hypertension via bioinformatics and in vitro validation for therapeutic target discovery. Comput Methods Biomech Biomed Engin 2025:1-12. [PMID: 40099537 DOI: 10.1080/10255842.2025.2468322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 01/17/2025] [Accepted: 02/10/2025] [Indexed: 03/20/2025]
Abstract
This study aims to uncover key genes and associated pathways related to nitric oxide (NO) in pulmonary hypertension (PH). By analyzing datasets GSE131793 and GSE703 from the Gene Expression Omnibus (GEO), differentially expressed genes (DEGs) associated with PH were identified. NO-related genes were selected from the GeneCards database and intersected with the DEGs. Subsequently, Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) analyses were conducted to evaluate pathway enrichment, and key genes were selected using the random forest and least absolute shrinkage and selection operator (LASSO) regression algorithms. Immune cell infiltration was analyzed using the CIBERSORT algorithm, and Gene Set Enrichment Analysis (GSEA) was performed to explore potential mechanisms. The transcriptional regulatory networks of key genes were constructed using Cytoscape software. The expression levels of the key genes were validated in peripheral blood samples from PH patients using quantitative real-time PCR (RT-qPCR). A total of 97 DEGs were identified, of which 20 were NO-related genes. Three key genes, HBG2, PRKAB1, and THBD, were further selected. RT-qPCR results revealed significant upregulation of HBG2 and THBD, and downregulation of PRKAB1 in PH patients. CIBERSORT analysis indicated the significant role of immune cells in the pathology of PH. GSEA and transcriptional network analyses further suggested that key genes may participate in the pathogenesis of PH through immune regulation and metabolic pathways. Through bioinformatics analysis and clinical sample validation, this study systematically elucidates the potential mechanisms of NO-related key genes in PH, providing new molecular targets for early diagnosis and targeted therapy of PH.
Collapse
Affiliation(s)
- Zhuyang Chu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
| | - Yiming Li
- Department of Cardiac Surgery, The First Affiliated Hospital of USTC, Hefei, China
| | - Jianjun Ge
- Department of Cardiovascular Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
| |
Collapse
|
3
|
Murugesan P, Zhang Y, Huang Y, Chenggong Zong N, Youn JY, Chen W, Wang C, Loscalzo J, Cai H. Reversal of Pulmonary Hypertension in a Human-Like Model: Therapeutic Targeting of Endothelial DHFR. Circ Res 2024; 134:351-370. [PMID: 38299369 PMCID: PMC10880947 DOI: 10.1161/circresaha.123.323090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 01/06/2024] [Accepted: 01/15/2024] [Indexed: 02/02/2024]
Abstract
BACKGROUND Pulmonary hypertension (PH) is a progressive disorder characterized by remodeling of the pulmonary vasculature and elevated mean pulmonary arterial pressure, resulting in right heart failure. METHODS Here, we show that direct targeting of the endothelium to uncouple eNOS (endothelial nitric oxide synthase) with DAHP (2,4-diamino 6-hydroxypyrimidine; an inhibitor of GTP cyclohydrolase 1, the rate-limiting synthetic enzyme for the critical eNOS cofactor tetrahydrobiopterin) induces human-like, time-dependent progression of PH phenotypes in mice. RESULTS Critical phenotypic features include progressive elevation in mean pulmonary arterial pressure, right ventricular systolic blood pressure, and right ventricle (RV)/left ventricle plus septum (LV+S) weight ratio; extensive vascular remodeling of pulmonary arterioles with increased medial thickness/perivascular collagen deposition and increased expression of PCNA (proliferative cell nuclear antigen) and alpha-actin; markedly increased total and mitochondrial superoxide production, substantially reduced tetrahydrobiopterin and nitric oxide bioavailabilities; and formation of an array of human-like vascular lesions. Intriguingly, novel in-house generated endothelial-specific dihydrofolate reductase (DHFR) transgenic mice (tg-EC-DHFR) were completely protected from the pathophysiological and molecular features of PH upon DAHP treatment or hypoxia exposure. Furthermore, DHFR overexpression with a pCMV-DHFR plasmid transfection in mice after initiation of DAHP treatment completely reversed PH phenotypes. DHFR knockout mice spontaneously developed PH at baseline and had no additional deterioration in response to hypoxia, indicating an intrinsic role of DHFR deficiency in causing PH. RNA-sequencing experiments indicated great similarity in gene regulation profiles between the DAHP model and human patients with PH. CONCLUSIONS Taken together, these results establish a novel human-like murine model of PH that has long been lacking in the field, which can be broadly used for future mechanistic and translational studies. These data also indicate that targeting endothelial DHFR deficiency represents a novel and robust therapeutic strategy for the treatment of PH.
Collapse
Affiliation(s)
- Priya Murugesan
- Division of Molecular Medicine, Department of Anesthesiology, Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (P.M., Y.Z., Y.H., N.C.Z., J.Y.Y., H.C.)
| | - Yixuan Zhang
- Division of Molecular Medicine, Department of Anesthesiology, Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (P.M., Y.Z., Y.H., N.C.Z., J.Y.Y., H.C.)
| | - Yuanli Huang
- Division of Molecular Medicine, Department of Anesthesiology, Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (P.M., Y.Z., Y.H., N.C.Z., J.Y.Y., H.C.)
| | - Nobel Chenggong Zong
- Division of Molecular Medicine, Department of Anesthesiology, Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (P.M., Y.Z., Y.H., N.C.Z., J.Y.Y., H.C.)
| | - Ji Youn Youn
- Division of Molecular Medicine, Department of Anesthesiology, Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (P.M., Y.Z., Y.H., N.C.Z., J.Y.Y., H.C.)
| | - Wenhui Chen
- Peking Union Medical College and Chinese Academy of Medical Sciences, Department of Respiratory Medicine, China-Japan Friendship Hospital, Beijing (W.C., C.W.)
| | - Chen Wang
- Peking Union Medical College and Chinese Academy of Medical Sciences, Department of Respiratory Medicine, China-Japan Friendship Hospital, Beijing (W.C., C.W.)
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA (J.L.)
| | - Hua Cai
- Division of Molecular Medicine, Department of Anesthesiology, Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (P.M., Y.Z., Y.H., N.C.Z., J.Y.Y., H.C.)
| |
Collapse
|
4
|
Hye T, Hossain MR, Saha D, Foyez T, Ahsan F. Emerging biologics for the treatment of pulmonary arterial hypertension. J Drug Target 2023; 31:1-15. [PMID: 37026714 PMCID: PMC10228297 DOI: 10.1080/1061186x.2023.2199351] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 01/11/2023] [Accepted: 01/16/2023] [Indexed: 04/08/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a rare pulmonary vascular disorder, wherein mean systemic arterial pressure (mPAP) becomes abnormally high because of aberrant changes in various proliferative and inflammatory signalling pathways of pulmonary arterial cells. Currently used anti-PAH drugs chiefly target the vasodilatory and vasoconstrictive pathways. However, an imbalance between bone morphogenetic protein receptor type II (BMPRII) and transforming growth factor beta (TGF-β) pathways is also implicated in PAH predisposition and pathogenesis. Compared to currently used PAH drugs, various biologics have shown promise as PAH therapeutics that elicit their therapeutic actions akin to endogenous proteins. Biologics that have thus far been explored as PAH therapeutics include monoclonal antibodies, recombinant proteins, engineered cells, and nucleic acids. Because of their similarity with naturally occurring proteins and high binding affinity, biologics are more potent and effective and produce fewer side effects when compared with small molecule drugs. However, biologics also suffer from the limitations of producing immunogenic adverse effects. This review describes various emerging and promising biologics targeting the proliferative/apoptotic and vasodilatory pathways involved in PAH pathogenesis. Here, we have discussed sotatercept, a TGF-β ligand trap, which is reported to reverse vascular remodelling and reduce PVR with an improved 6-minute walk distance (6-MWDT). We also elaborated on other biologics including BMP9 ligand and anti-gremlin1 antibody, anti-OPG antibody, and getagozumab monoclonal antibody and cell-based therapies. Overall, recent literature suggests that biologics hold excellent promise as a safe and effective alternative to currently used PAH therapeutics.
Collapse
Affiliation(s)
- Tanvirul Hye
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, Michigan
| | - Md Riajul Hossain
- Department of Biological Sciences, University of Arkansas, Fayetteville, Arkansas
| | - Dipongkor Saha
- Department of Pharmaceutical and Biomedical Sciences, California Northstate College of Pharmacy, Elk Grove, California
| | - Tahmina Foyez
- Department of Hematology Blood Research Center School of Medicine, The University of North Carolina at Chapel Hill, North Carolina
| | - Fakhrul Ahsan
- Department of Pharmaceutical and Biomedical Sciences, California Northstate College of Pharmacy, Elk Grove, California
- MedLuidics LLC, Elk Grove, California, USA
| |
Collapse
|
5
|
Bender AM, Valentine MS, Bauer JA, Days E, Lindsley CW, Merryman WD. Identification of Potent, Selective, and Peripherally Restricted Serotonin Receptor 2B Antagonists from a High-Throughput Screen. Assay Drug Dev Technol 2023; 21:89-96. [PMID: 36930852 PMCID: PMC10122230 DOI: 10.1089/adt.2022.116] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2023] Open
Abstract
Antagonists of the serotonin receptor 2B (5-HT2B) have shown great promise as therapeutics for the treatment of pulmonary arterial hypertension, valvular heart disease, and related cardiopathies. Herein, we describe a high-throughput screen campaign that led to the identification of highly potent and selective 5-HT2B antagonists. Furthermore, selected compounds were profiled for their predicted ability to cross the blood-brain barrier. Two exemplary compounds, VU0530244 and VU0631019, were predicted to have very limited potential for brain penetration in human subjects, a critical profile for the development of 5-HT2B antagonists devoid of centrally-mediated adverse effects.
Collapse
Affiliation(s)
- Aaron M. Bender
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Franklin, Tennessee, USA
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
| | - Michael S. Valentine
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Joshua A. Bauer
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA
| | - Emily Days
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Craig W. Lindsley
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Franklin, Tennessee, USA
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA
| | - W. David Merryman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
6
|
He YF, Liu Y, Yu JH, Cheng H, Odilov A, Yang FP, Tian GH, Yao XM, Duan HQ, Yu CY, Yu C, Liu YM, Liu GY, Shen JS, Wang Z, Diao XX. Pharmacokinetics, mass balance, and metabolism of [ 14C]TPN171, a novel PDE5 inhibitor, in humans for the treatment of pulmonary arterial hypertension. Acta Pharmacol Sin 2023; 44:221-233. [PMID: 35676531 DOI: 10.1038/s41401-022-00922-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 05/11/2022] [Indexed: 01/18/2023]
Abstract
TPN171 is a novel phosphodiesterase-5 (PDE5) inhibitor used to treat pulmonary arterial hypertension (PAH) and erectile dysfunction (ED), which currently is undergoing phase II clinical trials in China. In this single-center, single-dose, nonrandomized, and open design study, radiolabeled [14C]TPN171 was used to investigate the metabolic mechanism, pharmacokinetic characteristics, and clearance pathways of TPN171 in 6 healthy Chinese male volunteers. Each volunteer was administered a single oral suspension of 10 mg (100 μCi) of [14C]TPN171. We found that TPN171 was absorbed rapidly in humans with a peak time (Tmax) of 0.667 h and a half-life (t1/2) of approximately 9.89 h in plasma. Excretion of radiopharmaceutical-related components was collected 216 h after administration, accounting for 95.21% of the dose (46.61% in urine and 48.60% in feces). TPN171 underwent extensive metabolism in humans. Twenty-two metabolites were detected in human plasma, urine, and feces using a radioactive detector combined with a high-resolution mass spectrometer. According to radiochromatograms, a glucuronide metabolite of O-dealkylated TPN171 exceeded 10% of the total drug-related components in human plasma. However, according to the Food and Drug Administration (FDA) guidelines, no further tests are needed to evaluate the safety of this metabolite because it is a phase II metabolite, but the compound is still worthy of attention. The main metabolic biotransformation of TPN171 was mono-oxidation (hydroxylation and N-oxidation), dehydrogenation, N-dealkylation, O-dealkylation, amide hydrolysis, glucuronidation, and acetylation. Cytochrome P450 3A4 (CYP3A4) mainly catalyzed the formation of metabolites, and CYP2E1 and CYP2D6 were involved in the oxidative metabolism of TPN171 to a lesser extent. According to the incubation data, M1 was mainly metabolized to M1G by UDP-glucuronosyltransferase 1A9 (UGT1A9), followed by UGT1A7 and UGT1A10.
Collapse
Affiliation(s)
- Yi-Fei He
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.,University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Yin Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.,University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Jing-Hua Yu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Huan Cheng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.,School of Pharmaceutical Sciences, Shandong University of Traditional Chinese Medicine, Ji-nan, 250355, China
| | - Abdullajon Odilov
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.,University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Fei-Pu Yang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | | | - Xiu-Mei Yao
- Vigonvita Life Sciences Co., Ltd, Suzhou, 215000, China
| | - Hua-Qing Duan
- Vigonvita Life Sciences Co., Ltd, Suzhou, 215000, China
| | - Cheng-Yin Yu
- Shanghai Xuhui Central Hospital, Shanghai, 200030, China
| | - Chen Yu
- Shanghai Xuhui Central Hospital, Shanghai, 200030, China
| | - Yan-Mei Liu
- Shanghai Xuhui Central Hospital, Shanghai, 200030, China
| | - Gang-Yi Liu
- Shanghai Xuhui Central Hospital, Shanghai, 200030, China
| | - Jing-Shan Shen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.,University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhen Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China. .,Lingang Laboratory, Shanghai, 201602, China.
| | - Xing-Xing Diao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China. .,University of the Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
7
|
Hu Y, Wang X, Xiao S, Wu H, Huan C, Xu T, Guo M, Liu A, Jiang X, Wang J, Zhu H, Pan D. Development and validation of a risk nomogram model for predicting pulmonary hypertension in patients with stage 3-5 chronic kidney disease. Int Urol Nephrol 2022; 55:1353-1363. [PMID: 36562902 PMCID: PMC10105676 DOI: 10.1007/s11255-022-03431-x] [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: 04/02/2022] [Accepted: 11/18/2022] [Indexed: 12/24/2022]
Abstract
OBJECTIVES The occurrence of pulmonary arterial hypertension (PAH) can greatly affect the prognosis of patients with chronic kidney disease (CKD). We aimed to construct a nomogram to predict the probability of PAH development in patients with stage 3-5 CKD to guide early intervention and to improve prognosis. METHODS From August 2018 to December 2021, we collected the data of 1258 patients with stage 3-5 CKD hospitalized at the Affiliated Hospital of Xuzhou Medical University as a training set and 389 patients hospitalized at Zhongda Hospital as a validation set. These patients were divided into PAH and N-PAH groups with pulmonary arterial systolic pressure ≥ 35 mmHg as the cutoff. The results of univariate and multivariate logistic regression analyses were used to establish the nomogram. Then, areas under the receiver operating characteristic curve (AUC-ROCs), a calibration plot, and decision curve analysis (DCA) were used to validate the nomogram. RESULTS The nomogram included nine variables: age, diabetes mellitus, hemoglobin, platelet count, serum creatinine, left ventricular end-diastolic diameter, left atrial diameter, main pulmonary artery diameter and left ventricular ejection fraction. The AUC-ROCs of the training set and validation set were 0.801 (95% confidence interval (CI) 0.771-0.830) and 0.760 (95% CI 0.699-0.818), respectively, which showed good discriminative ability of the nomogram. The calibration diagram showed good agreement between the predicted and observed results. DCA also demonstrated that the nomogram could be clinically useful. CONCLUSION The evaluation of the nomogram model for predicting PAH in patients with CKD based on risk factors showed its ideal efficacy. Thus, the nomogram can be used to screen for patients at high risk for PAH and has guiding value for the subsequent formulation of prevention strategies and clinical treatment.
Collapse
Affiliation(s)
- Yue Hu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Intensive Care Unit Department, No. 23, Mei Shu Guan Hou Jie, Beijing, 100010, Dongcheng, China
| | - Xiaotong Wang
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Shengjue Xiao
- Department of Cardiology, School of Medicine, Zhongda Hospital, Southeast University, 87 Dingjiaqiao, Nanjing, 210009, Jiangsu, China
| | - Huimin Wu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Intensive Care Unit Department, No. 23, Mei Shu Guan Hou Jie, Beijing, 100010, Dongcheng, China
| | - Chunyan Huan
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Tao Xu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Minjia Guo
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Ailin Liu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Xiaoyao Jiang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Intensive Care Unit Department, No. 23, Mei Shu Guan Hou Jie, Beijing, 100010, Dongcheng, China
| | - Jia Wang
- Department of Nephrology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Hong Zhu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Defeng Pan
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
8
|
Liu B, Peng Y, Yi D, Machireddy N, Dong D, Ramirez K, Dai J, Vanderpool R, Zhu MM, Dai Z, Zhao YY. Endothelial PHD2 deficiency induces nitrative stress via suppression of caveolin-1 in pulmonary hypertension. Eur Respir J 2022; 60:2102643. [PMID: 35798360 PMCID: PMC9791795 DOI: 10.1183/13993003.02643-2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 06/24/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND Nitrative stress is a characteristic feature of the pathology of human pulmonary arterial hypertension. However, the role of nitrative stress in the pathogenesis of obliterative vascular remodelling and severe pulmonary arterial hypertension remains largely unclear. METHOD Our recently identified novel mouse model (Egln1Tie2Cre, Egln1 encoding prolyl hydroxylase 2 (PHD2)) has obliterative vascular remodelling and right heart failure, making it an excellent model to use in this study to examine the role of nitrative stress in obliterative vascular remodelling. RESULTS Nitrative stress was markedly elevated whereas endothelial caveolin-1 (Cav1) expression was suppressed in the lungs of Egln1Tie2Cre mice. Treatment with a superoxide dismutase mimetic, manganese (III) tetrakis (1-methyl-4-pyridyl) porphyrin pentachloride or endothelial Nos3 knockdown using endothelial cell-targeted nanoparticle delivery of CRISPR-Cas9/guide RNA plasmid DNA inhibited obliterative pulmonary vascular remodelling and attenuated severe pulmonary hypertension in Egln1Tie2Cre mice. Genetic restoration of Cav1 expression in Egln1Tie2Cre mice normalised nitrative stress, reduced pulmonary hypertension and improved right heart function. CONCLUSION These data suggest that suppression of Cav1 expression secondary to PHD2 deficiency augments nitrative stress through endothelial nitric oxide synthase activation, which contributes to obliterative vascular remodelling and severe pulmonary hypertension. Thus, a reactive oxygen/nitrogen species scavenger might have therapeutic potential for the inhibition of obliterative vascular remodelling and severe pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Bin Liu
- Division of Pulmonary, Critical Care and Sleep, Dept of Internal Medicine, University of Arizona, Phoenix, AZ, USA
- Translational Cardiovascular Research Center, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Yi Peng
- Program for Lung and Vascular Biology, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Section for Injury Repair and Regeneration Research, Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Division of Critical Care, Dept of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Dan Yi
- Division of Pulmonary, Critical Care and Sleep, Dept of Internal Medicine, University of Arizona, Phoenix, AZ, USA
- Translational Cardiovascular Research Center, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Narsa Machireddy
- Program for Lung and Vascular Biology, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Section for Injury Repair and Regeneration Research, Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Division of Critical Care, Dept of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Daoyin Dong
- Program for Lung and Vascular Biology, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Section for Injury Repair and Regeneration Research, Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Division of Critical Care, Dept of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Karina Ramirez
- Division of Pulmonary, Critical Care and Sleep, Dept of Internal Medicine, University of Arizona, Phoenix, AZ, USA
- Translational Cardiovascular Research Center, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Jingbo Dai
- Program for Lung and Vascular Biology, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Section for Injury Repair and Regeneration Research, Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Division of Critical Care, Dept of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Rebecca Vanderpool
- College of Medicine Division of Cardiovascular Medicine, The Ohio State University, Columbus, OH, USA
| | - Maggie M Zhu
- Program for Lung and Vascular Biology, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Section for Injury Repair and Regeneration Research, Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Division of Critical Care, Dept of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Zhiyu Dai
- Division of Pulmonary, Critical Care and Sleep, Dept of Internal Medicine, University of Arizona, Phoenix, AZ, USA
- Translational Cardiovascular Research Center, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
- Zhiyu Dai and You-Yang Zhao contributed equally to this article as lead authors and supervised the work
| | - You-Yang Zhao
- Program for Lung and Vascular Biology, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Section for Injury Repair and Regeneration Research, Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Division of Critical Care, Dept of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Dept of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Dept of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Zhiyu Dai and You-Yang Zhao contributed equally to this article as lead authors and supervised the work
| |
Collapse
|
9
|
Bikou O, Tharakan S, Yamada KP, Kariya T, Aguero J, Gordon A, Mazurek R, Aikawa T, Kohlbrenner E, Fish KM, Hajjar RJ, Ishikawa K. Endobronchial Aerosolized AAV1.SERCA2a Gene Therapy in a Pulmonary Hypertension Pig Model: Addressing the Lung Delivery Bottleneck. Hum Gene Ther 2022; 33:550-559. [PMID: 35293228 PMCID: PMC9142769 DOI: 10.1089/hum.2021.274] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/20/2022] [Indexed: 11/12/2022] Open
Abstract
A disappointing number of new therapies for pulmonary hypertension (PH) have been successfully translated to the clinic. Adeno-associated viral (AAV) gene therapy has the potential to treat the underlying pathology of PH, but the challenge remains in efficient and safe delivery. The aims of this study were (1) to test the efficacy of endobronchial aerosolization delivery for AAV1-mediated sarcoplasmic/endoplasmic reticulum Ca2+ ATPase 2a (SERCA2a) gene therapy in a PH pig model and (2) to identify the most efficient airway administration modality for in-lung gene therapy in PH. We hypothesized that delivery to the distal bronchi increases lung viral uptake and avoids virus loss in off-target compartments. In part 1 of the study, PH was induced in pigs by surgically banding the pulmonary veins. Two months postsurgery, 1 × 1013 viral genomes (vg) of AAV1.SERCA2a or saline was endobronchially aerosolized using a bronchoscope. Two months after aerosolization, high vg copies (vgc) were detected in the lungs, accompanied by functional and morphometrical amelioration of PH. In part 2 of the study, we directly compared the endobronchial aerosolization gene delivery to the intratracheal aerosolization in PH pigs. Endobronchial delivery demonstrated higher viral expression (6,719 ± 927 vs. 1,444 ± 402 vgc/100 ng DNA, p = 0.0017), suggesting this delivery modality is a promising method for clinical AAV gene therapy for PH.
Collapse
Affiliation(s)
- Olympia Bikou
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Serena Tharakan
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kelly P. Yamada
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Taro Kariya
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jaume Aguero
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Alexandra Gordon
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Renata Mazurek
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Phospholamban Foundation, Amsterdam, Netherlands
| | - Tadao Aikawa
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Erik Kohlbrenner
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Phospholamban Foundation, Amsterdam, Netherlands
| | - Kenneth M. Fish
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | - Kiyotake Ishikawa
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
10
|
Endobronchial Gene Delivery for Pulmonary Hypertension in a Large Animal Model. Methods Mol Biol 2022; 2573:279-289. [PMID: 36040602 DOI: 10.1007/978-1-0716-2707-5_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Pulmonary hypertension (PH) is a devastating disease with high morbidity and mortality. Despite significant progress in the pharmacotherapy, current treatments only ameliorate the symptoms and cannot heal PH. Gene therapy may target the roots of the disease and holds evident promise. The current bottleneck for lung gene therapy is the delivery method. The requirements for the delivery mode are efficiency, safety, and the ability to target the anatomical site of interest, while avoiding off-target effects. Aerosolized gene delivery has been used in several studies and proven to be an efficient mode of administration for lung gene therapy. In this chapter, we describe a protocol of endobronchial aerosolization for PH gene therapy in a large animal model. Testing of a gene therapy in large animals is essential before clinical testing, since the lung anatomy and (patho)physiology differ immensely between humans and rodents, where most of the proof-of-concept studies are tested. The gene delivery vector is being aerosolized in the peripheral bronchi using a sprayer inserted through a flexible bronchoscope. This delivery mode results in efficient lung uptake and less off-target distribution relative to other airway delivery methods.
Collapse
|
11
|
Zhang Y, Zervopoulos SD, Boukouris AE, Lorenzana-Carrillo MA, Saleme B, Webster L, Liu Y, Haromy A, Tabatabaei Dakhili SA, Ussher JR, Sutendra G, Michelakis ED. SNPs for Genes Encoding the Mitochondrial Proteins Sirtuin3 and Uncoupling Protein 2 Are Associated With Disease Severity, Type 2 Diabetes, and Outcomes in Patients With Pulmonary Arterial Hypertension and This Is Recapitulated in a New Mouse Model Lacking Both Genes. J Am Heart Assoc 2021; 10:e020451. [PMID: 34719264 PMCID: PMC9075406 DOI: 10.1161/jaha.120.020451] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Isolated loss‐of‐function single nucleotide polymorphisms (SNPs) for SIRT3 (a mitochondrial deacetylase) and UCP2 (an atypical uncoupling protein enabling mitochondrial calcium entry) have been associated with both pulmonary arterial hypertension (PAH) and insulin resistance, but their collective role in animal models and patients is unknown. Methods and Results In a prospective cohort of patients with PAH (n=60), we measured SNPs for both SIRT3 and UCP2, along with several clinical features (including invasive hemodynamic data) and outcomes. We found SIRT3 and UCP2 SNPs often both in the same patient in a homozygous or heterozygous manner, correlating positively with PAH severity and associated with the presence of type 2 diabetes and 10‐year outcomes (death and transplantation). To explore this mechanistically, we generated double knockout mice for Sirt3 and Ucp2 and found increasing severity of PAH (mean pulmonary artery pressure, right ventricular hypertrophy/dilatation and extensive vascular remodeling, including inflammatory plexogenic lesions, in a gene dose‐dependent manner), along with insulin resistance, compared with wild‐type mice. The suppressed mitochondrial function (decreased respiration, increased mitochondrial membrane potential) in the double knockout pulmonary artery smooth muscle cells was associated with apoptosis resistance and increased proliferation, compared with wild‐type mice. Conclusions Our work supports the metabolic theory of PAH and shows that these mice exhibit spontaneous severe PAH (without environmental or chemical triggers) that mimics human PAH and may explain the findings in our patient cohort. Our study offers a new mouse model of PAH, with several features of human disease that are typically absent in other PAH mouse models.
Collapse
Affiliation(s)
- Yongneng Zhang
- Department of Medicine (Cardiology), Faculty of Medicine and Dentistry University of Alberta Edmonton Alberta Canada
| | - Sotirios D Zervopoulos
- Department of Medicine (Cardiology), Faculty of Medicine and Dentistry University of Alberta Edmonton Alberta Canada
| | - Aristeidis E Boukouris
- Department of Medicine (Cardiology), Faculty of Medicine and Dentistry University of Alberta Edmonton Alberta Canada
| | | | - Bruno Saleme
- Department of Medicine (Cardiology), Faculty of Medicine and Dentistry University of Alberta Edmonton Alberta Canada
| | - Linda Webster
- Department of Medicine (Cardiology), Faculty of Medicine and Dentistry University of Alberta Edmonton Alberta Canada
| | - Yongsheng Liu
- Department of Medicine (Cardiology), Faculty of Medicine and Dentistry University of Alberta Edmonton Alberta Canada
| | - Alois Haromy
- Department of Medicine (Cardiology), Faculty of Medicine and Dentistry University of Alberta Edmonton Alberta Canada
| | | | - John R Ussher
- Faculty of Pharmacy and Pharmaceutical Sciences University of Alberta Edmonton Alberta Canada
| | - Gopinath Sutendra
- Department of Medicine (Cardiology), Faculty of Medicine and Dentistry University of Alberta Edmonton Alberta Canada
| | - Evangelos D Michelakis
- Department of Medicine (Cardiology), Faculty of Medicine and Dentistry University of Alberta Edmonton Alberta Canada
| |
Collapse
|
12
|
Evans CE, Cober ND, Dai Z, Stewart DJ, Zhao YY. Endothelial cells in the pathogenesis of pulmonary arterial hypertension. Eur Respir J 2021; 58:13993003.03957-2020. [PMID: 33509961 DOI: 10.1183/13993003.03957-2020] [Citation(s) in RCA: 159] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 01/13/2021] [Indexed: 12/11/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a devastating disease that involves pulmonary vasoconstriction, small vessel obliteration, large vessel thickening and obstruction, and development of plexiform lesions. PAH vasculopathy leads to progressive increases in pulmonary vascular resistance, right heart failure and, ultimately, premature death. Besides other cell types that are known to be involved in PAH pathogenesis (e.g. smooth muscle cells, fibroblasts and leukocytes), recent studies have demonstrated that endothelial cells (ECs) have a crucial role in the initiation and progression of PAH. The EC-specific role in PAH is multi-faceted and affects numerous pathophysiological processes, including vasoconstriction, inflammation, coagulation, metabolism and oxidative/nitrative stress, as well as cell viability, growth and differentiation. In this review, we describe how EC dysfunction and cell signalling regulate the pathogenesis of PAH. We also highlight areas of research that warrant attention in future studies, and discuss potential molecular signalling pathways in ECs that could be targeted therapeutically in the prevention and treatment of PAH.
Collapse
Affiliation(s)
- Colin E Evans
- Program for Lung and Vascular Biology, Section of Injury Repair and Regeneration, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA.,Dept of Pediatrics, Division of Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Nicholas D Cober
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Dept of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Zhiyu Dai
- Program for Lung and Vascular Biology, Section of Injury Repair and Regeneration, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA.,Dept of Pediatrics, Division of Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Dept of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Duncan J Stewart
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Dept of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - You-Yang Zhao
- Program for Lung and Vascular Biology, Section of Injury Repair and Regeneration, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA .,Dept of Pediatrics, Division of Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Dept of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Dept of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
13
|
Al-Hilal TA, Keshavarz A, Kadry H, Lahooti B, Al-Obaida A, Ding Z, Li W, Kamm R, McMurtry IF, Lahm T, Nozik-Grayck E, Stenmark KR, Ahsan F. Pulmonary-arterial-hypertension (PAH)-on-a-chip: fabrication, validation and application. LAB ON A CHIP 2020; 20:3334-3345. [PMID: 32749432 PMCID: PMC7592346 DOI: 10.1039/d0lc00605j] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Currently used animal and cellular models for pulmonary arterial hypertension (PAH) only partially recapitulate its pathophysiology in humans and are thus inadequate in reproducing the hallmarks of the disease, inconsistent in portraying the sex-disparity, and unyielding to combinatorial study designs. Here we sought to deploy the ingenuity of microengineering in developing and validating a tissue chip model for human PAH. We designed and fabricated a microfluidic device to emulate the luminal, intimal, medial, adventitial, and perivascular layers of a pulmonary artery. By growing three types of pulmonary arterial cells (PACs)-endothelial, smooth muscle, and adventitial cells, we recreated the PAH pathophysiology on the device. Diseased (PAH) PACs, when grown on the chips, moved of out their designated layers and created phenomena similar to the major pathologies of human PAH: intimal thickening, muscularization, and arterial remodeling and show an endothelial to mesenchymal transition. Flow-induced stress caused control cells, grown on the chips, to undergo morphological changes and elicit arterial remodeling. Our data also suggest that the newly developed chips can be used to elucidate the sex disparity in PAH and to study the therapeutic efficacy of existing and investigational anti-PAH drugs. We believe this miniaturized device can be deployed for testing various prevailing and new hypotheses regarding the pathobiology and drug therapy in human PAH.
Collapse
Affiliation(s)
- Taslim A Al-Hilal
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Jerry H. Hodge School of Pharmacy, 1300 Coulter Dr., Amarillo, 79119 Texas, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Tian L, Xiong PY, Alizadeh E, Lima PDA, Potus F, Mewburn J, Martin A, Chen K, Archer SL. Supra-coronary aortic banding improves right ventricular function in experimental pulmonary arterial hypertension in rats by increasing systolic right coronary artery perfusion. Acta Physiol (Oxf) 2020; 229:e13483. [PMID: 32339403 DOI: 10.1111/apha.13483] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 12/13/2022]
Abstract
AIM Pulmonary arterial hypertension (PAH) results in right ventricular (RV) dysfunction owing, in part, to RV ischemia. The relative contribution of RV microvascular rarefaction vs reduced right coronary artery perfusion pressure (RCA-PP) to RV ischemia remains unknown. We hypothesize that increasing RCA-PP improves RV function in PAH by increasing RV systolic perfusion. METHODS Supra-coronary aortic banding (SAB) or sham surgery was performed on male Sprague-Dawley rats. Seven to ten days later, rats received either monocrotaline (MCT; 60 mg/kg) or saline. After 1 month, echocardiography, cardiac catheterization, 99m Tc-sestamibi single-photon emission computed tomography (SPECT) and microsphere infusion studies were performed. The RV was harvested for measurement of hypertrophy (RVH), fibrosis and immunoblotting, and the lung was harvested for pulmonary artery (PA) histology. RESULTS Supra-coronary aortic banding increased systolic pressures in proximal aorta and systolic RCA-PP in SAB + MCT vs MCT rats (114 ± 12 vs 5 ± 9 mm Hg), without altering diastolic RCA-PP. SAB + MCT rats had improved RV function vs MCT rats, evident from their significantly increased cardiac output (CO), RV free wall (RVFW) thickening, tricuspid annular plane systolic excursion (TAPSE) and RV-PA coupling indices. RV-PA coupling indices and CO correlated directly with systolic RCA-PP. RV perfusion was increased in SAB + MCT vs MCT rats and correlated well with CO; whereas microvascular rarefaction was unaltered. SAB + MCT rats had less RVH and fibrosis and lower PA pressures vs MCT rats. SAB + MCT rats had significantly lower RV pyruvate kinase muscle isoform 2/1 ratios than MCT rats, consistent with restoration of oxidative metabolism. CONCLUSION A SAB-induced increase in systolic RCA-PP improves RV perfusion and function in MCT rats. Maintaining systolic RCA perfusion can preserve RV function in PAH.
Collapse
Affiliation(s)
- Lian Tian
- Department of Medicine Queen's University Kingston ON Canada
| | - Ping Yu Xiong
- Department of Medicine Queen's University Kingston ON Canada
- Department of Biomedical and Molecular Sciences Queen's University Kingston ON Canada
| | | | | | - François Potus
- Department of Medicine Queen's University Kingston ON Canada
| | - Jeffrey Mewburn
- Department of Medicine Queen's University Kingston ON Canada
| | - Ashley Martin
- 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) Kingston ON Canada
| |
Collapse
|
15
|
Wang HL, Tang FQ, Jiang YH, Zhu Y, Jian Z, Xiao YB. AMPKα2 deficiency exacerbates hypoxia-induced pulmonary hypertension by promoting pulmonary arterial smooth muscle cell proliferation. J Physiol Biochem 2020; 76:445-456. [PMID: 32592088 DOI: 10.1007/s13105-020-00742-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 04/23/2020] [Indexed: 12/13/2022]
Abstract
Increased evidence indicates that adenosine monophosphate-activated protein kinase (AMPK) plays a vital role in vascular homeostasis, especially under hypoxia, and protects against the progression of pulmonary hypertension (PH). However, the role of AMPK in the pathogenesis of PH remains to be clarified. In the present study, we confirmed that a loss of AMPKα2 exacerbated the development of PH by using hypoxia-induced PH model in AMPKα2 -/- mice. After a 4-week period of hypoxic exposure, AMPKα2 -/- mice exhibited more severe pulmonary vascular remodeling and pulmonary vascular smooth muscle cell (SMC) proliferation when compared with wild type (WT) mice. In vitro, AMPKα2 knockdown promoted the proliferation of pulmonary arterial smooth muscle cells (PASMCs) under hypoxia. This phenomenon was accompanied by upregulated Skp2 and downregulated p27kip1 expression and was abolished by rapamycin, an inhibitor of mTOR. These results indicate that AMPKα2 deficiency exacerbates hypoxia-induced PH by promoting PASMC proliferation via the mTOR/Skp2/p27kip1 signaling axis. Therefore, enhanced AMPKα2 activity might underlie a novel therapeutic strategy for the management of PH.
Collapse
Affiliation(s)
- Hai-Long Wang
- Department of Cardiovascular Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, People's Republic of China
| | - Fu-Qin Tang
- Department of Cardiovascular Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, People's Republic of China
| | - Yun-Han Jiang
- Department of Cardiovascular Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, People's Republic of China
| | - Yu Zhu
- Department of Cardiovascular Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, People's Republic of China
| | - Zhao Jian
- Department of Cardiovascular Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, People's Republic of China.
| | - Ying-Bin Xiao
- Department of Cardiovascular Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, People's Republic of China.
| |
Collapse
|
16
|
Daniell H, Mangu V, Yakubov B, Park J, Habibi P, Shi Y, Gonnella PA, Fisher A, Cook T, Zeng L, Kawut SM, Lahm T. Investigational new drug enabling angiotensin oral-delivery studies to attenuate pulmonary hypertension. Biomaterials 2020; 233:119750. [PMID: 31931441 PMCID: PMC7045910 DOI: 10.1016/j.biomaterials.2019.119750] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 12/16/2019] [Accepted: 12/28/2019] [Indexed: 01/21/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a deadly and uncurable disease characterized by remodeling of the pulmonary vasculature and increased pulmonary artery pressure. Angiotensin Converting Enzyme 2 (ACE2) and its product, angiotensin-(1-7) [ANG-(1-7)] were expressed in lettuce chloroplasts to facilitate affordable oral drug delivery. Lyophilized lettuce cells were stable up to 28 months at ambient temperature with proper folding, assembly of CTB-ACE2/ANG-(1-7) and functionality. When the antibiotic resistance gene was removed, Ang1-7 expression was stable in subsequent generations in marker-free transplastomic lines. Oral gavage of monocrotaline-induced PAH rats resulted in dose-dependent delivery of ANG-(1-7) and ACE2 in plasma/tissues and PAH development was attenuated with decreases in right ventricular (RV) hypertrophy, RV systolic pressure, total pulmonary resistance and pulmonary artery remodeling. Such attenuation correlated well with alterations in the transcription of Ang-(1-7) receptor MAS and angiotensin II receptor AGTRI as well as IL-1β and TGF-β1. Toxicology studies showed that both male and female rats tolerated ~10-fold ACE2/ANG-(1-7) higher than efficacy dose. Plant cell wall degrading enzymes enhanced plasma levels of orally delivered protein drug bioencapsulated within plant cells. Efficient attenuation of PAH with no toxicity augurs well for clinical advancement of the first oral protein therapy to prevent/treat underlying pathology for this disease.
Collapse
Affiliation(s)
- Henry Daniell
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Venkata Mangu
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bakhtiyor Yakubov
- Department of Medicine, Division of Pulmonary, Critical Care and Occupational Medicine, Indianapolis, IN, USA
| | - Jiyoung Park
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Peyman Habibi
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yao Shi
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Patricia A Gonnella
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amanda Fisher
- Department of Medicine, Division of Pulmonary, Critical Care and Occupational Medicine, Indianapolis, IN, USA
| | - Todd Cook
- Department of Medicine, Division of Pulmonary, Critical Care and Occupational Medicine, Indianapolis, IN, USA
| | - Lily Zeng
- Department of Medicine, Division of Pulmonary, Critical Care and Occupational Medicine, Indianapolis, IN, USA
| | - Steven M Kawut
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Tim Lahm
- Department of Medicine, Division of Pulmonary, Critical Care and Occupational Medicine, Indianapolis, IN, USA; Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN, USA; Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA
| |
Collapse
|
17
|
Valuparampil Varghese M, James J, Eccles CA, Niihori M, Rafikova O, Rafikov R. Inhibition of Anaplerosis Attenuated Vascular Proliferation in Pulmonary Arterial Hypertension. J Clin Med 2020; 9:jcm9020443. [PMID: 32041182 PMCID: PMC7074087 DOI: 10.3390/jcm9020443] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/20/2020] [Accepted: 02/04/2020] [Indexed: 01/09/2023] Open
Abstract
Vascular remodeling is considered a key event in the pathogenesis of pulmonary arterial hypertension (PAH). However, mechanisms of gaining the proliferative phenotype by pulmonary vascular cells are still unresolved. Due to well-established pyruvate dehydrogenase (PDH) deficiency in PAH pathogenesis, we hypothesized that the activation of another branch of pyruvate metabolism, anaplerosis, via pyruvate carboxylase (PC) could be a key contributor to the metabolic reprogramming of the vasculature. In sugen/hypoxic PAH rats, vascular proliferation was found to be accompanied by increased activation of Akt signaling, which upregulated membrane Glut4 translocation and caused upregulation of hexokinase and pyruvate kinase-2, and an overall increase in the glycolytic flux. Decreased PDH activity and upregulation of PC shuttled more pyruvate to oxaloacetate. This results in the anaplerotic reprogramming of lung vascular cells and their subsequent proliferation. Treatment of sugen/hypoxia rats with the PC inhibitor, phenylacetic acid 20 mg/kg, starting after one week from disease induction, significantly attenuated right ventricular systolic pressure, Fulton index, and pulmonary vascular cell proliferation. PC inhibition reduced the glycolytic shift by attenuating Akt-signaling, glycolysis, and restored mitochondrial pyruvate oxidation. Our findings suggest that targeting PC mediated anaplerosis is a potential therapeutic intervention for the resolution of vascular remodeling in PAH.
Collapse
Affiliation(s)
| | | | | | | | - Olga Rafikova
- Correspondence: (O.R.); (R.R.); Tel.: +1-520-626-1303 (O.R.); +1-520-626-6092 (R.R.)
| | - Ruslan Rafikov
- Correspondence: (O.R.); (R.R.); Tel.: +1-520-626-1303 (O.R.); +1-520-626-6092 (R.R.)
| |
Collapse
|
18
|
Suliman HB, Nozik-Grayck E. Mitochondrial Dysfunction: Metabolic Drivers of Pulmonary Hypertension. Antioxid Redox Signal 2019; 31:843-857. [PMID: 30604624 PMCID: PMC6751393 DOI: 10.1089/ars.2018.7705] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Pulmonary hypertension (PH) is a progressive disease characterized by pulmonary vascular remodeling and lung vasculopathy. The disease displays progressive dyspnea, pulmonary artery uncoupling and right ventricular (RV) dysfunction. The overall survival rate is ranging from 28-72%. Recent Advances: The molecular events that promote the development of PH are complex and incompletely understood. Metabolic impairment has been proposed to contribute to the pathophysiology of PH with evidence for mitochondrial dysfunction involving the electron transport chain proteins, antioxidant enzymes, apoptosis regulators, and mitochondrial quality control. Critical Issues: It is vital to characterize the mechanisms by which mitochondrial dysfunction contribute to PH pathogenesis. This review focuses on the currently available publications that supports mitochondrial mechanisms in PH pathophysiology. Future Directions: Further studies of these metabolic mitochondrial alterations in PH could be viable targets of diagnostic and therapeutic intervention.
Collapse
Affiliation(s)
- Hagir B Suliman
- Department of Anesthesiology, Duke University Medical Centers, Durham, North Carolina
| | - Eva Nozik-Grayck
- Department of Pediatrics, Cardiovascular Pulmonary Research Labs and Pediatric Critical Care Medicine, University of Colorado Denver, Aurora, Colorado
| |
Collapse
|
19
|
Bloodworth NC, Clark CR, West JD, Snider JC, Gaskill C, Shay S, Scott C, Bastarache J, Gladson S, Moore C, D'Amico R, Brittain EL, Tanjore H, Blackwell TS, Majka SM, Merryman WD. Bone Marrow-Derived Proangiogenic Cells Mediate Pulmonary Arteriole Stiffening via Serotonin 2B Receptor Dependent Mechanism. Circ Res 2019; 123:e51-e64. [PMID: 30566041 DOI: 10.1161/circresaha.118.313397] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
RATIONALE Pulmonary arterial hypertension is a deadly disease of the pulmonary vasculature for which no disease-modifying therapies exist. Small-vessel stiffening and remodeling are fundamental pathological features of pulmonary arterial hypertension that occur early and drive further endovascular cell dysfunction. Bone marrow (BM)-derived proangiogenic cells (PACs), a specialized heterogeneous subpopulation of myeloid lineage cells, are thought to play an important role in pathogenesis. OBJECTIVE To determine whether BM-derived PACs directly contributed to experimental pulmonary hypertension (PH) by promoting small-vessel stiffening through 5-HT2B (serotonin 2B receptor)-mediated signaling. METHODS AND RESULTS We performed BM transplants using transgenic donor animals expressing diphtheria toxin secondary to activation of an endothelial-specific tamoxifen-inducible Cre and induced experimental PH using hypoxia with SU5416 to enhance endovascular injury and ablated BM-derived PACs, after which we measured right ventricular systolic pressures in a closed-chest procedure. BM-derived PAC lineage tracing was accomplished by transplanting BM from transgenic donor animals with fluorescently labeled hematopoietic cells and treating mice with a 5-HT2B antagonist. BM-derived PAC ablation both prevented and reversed experimental PH with SU5416-enhanced endovascular injury, reducing the number of muscularized pulmonary arterioles and normalizing arteriole stiffness as measured by atomic force microscopy. Similarly, treatment with a pharmacological antagonist of 5-HT2B also prevented experimental PH, reducing the number and stiffness of muscularized pulmonary arterioles. PACs accelerated pulmonary microvascular endothelial cell injury response in vitro, and the presence of BM-derived PACs significantly correlated with stiffer pulmonary arterioles in pulmonary arterial hypertension patients and mice with experimental PH. RNA sequencing of BM-derived PACs showed that 5-HT2B antagonism significantly altered biologic pathways regulating cell proliferation, locomotion and migration, and cytokine production and response to cytokine stimulus. CONCLUSIONS Together, our findings illustrate that BM-derived PACs directly contribute to experimental PH with SU5416-enhanced endovascular injury by mediating small-vessel stiffening and remodeling in a 5-HT2B signaling-dependent manner.
Collapse
Affiliation(s)
- Nathaniel C Bloodworth
- From the Department of Biomedical Engineering (N.C.B., C.R.C., J.C.S., C.S., R.D., W.D.M.), Vanderbilt University Medical Center, Nashville, TN
| | - Cynthia R Clark
- From the Department of Biomedical Engineering (N.C.B., C.R.C., J.C.S., C.S., R.D., W.D.M.), Vanderbilt University Medical Center, Nashville, TN
| | - James D West
- Division of Allergy, Pulmonary, and Critical Care, Department of Medicine (J.D.W., C.G., S.S., J.B., S.G., C.M., H.T., T.S.B., S.M.M.), Vanderbilt University Medical Center, Nashville, TN
| | - J Caleb Snider
- From the Department of Biomedical Engineering (N.C.B., C.R.C., J.C.S., C.S., R.D., W.D.M.), Vanderbilt University Medical Center, Nashville, TN
| | - Christa Gaskill
- Division of Allergy, Pulmonary, and Critical Care, Department of Medicine (J.D.W., C.G., S.S., J.B., S.G., C.M., H.T., T.S.B., S.M.M.), Vanderbilt University Medical Center, Nashville, TN
| | - Sheila Shay
- Division of Allergy, Pulmonary, and Critical Care, Department of Medicine (J.D.W., C.G., S.S., J.B., S.G., C.M., H.T., T.S.B., S.M.M.), Vanderbilt University Medical Center, Nashville, TN
| | - Christine Scott
- From the Department of Biomedical Engineering (N.C.B., C.R.C., J.C.S., C.S., R.D., W.D.M.), Vanderbilt University Medical Center, Nashville, TN
| | - Julie Bastarache
- Division of Allergy, Pulmonary, and Critical Care, Department of Medicine (J.D.W., C.G., S.S., J.B., S.G., C.M., H.T., T.S.B., S.M.M.), Vanderbilt University Medical Center, Nashville, TN
| | - Santhi Gladson
- Division of Allergy, Pulmonary, and Critical Care, Department of Medicine (J.D.W., C.G., S.S., J.B., S.G., C.M., H.T., T.S.B., S.M.M.), Vanderbilt University Medical Center, Nashville, TN
| | - Christy Moore
- Division of Allergy, Pulmonary, and Critical Care, Department of Medicine (J.D.W., C.G., S.S., J.B., S.G., C.M., H.T., T.S.B., S.M.M.), Vanderbilt University Medical Center, Nashville, TN
| | - Reid D'Amico
- From the Department of Biomedical Engineering (N.C.B., C.R.C., J.C.S., C.S., R.D., W.D.M.), Vanderbilt University Medical Center, Nashville, TN
| | - Evan L Brittain
- Division of Cardiovascular Medicine, Department of Medicine (E.L.B.), Vanderbilt University Medical Center, Nashville, TN
| | - Harikrishna Tanjore
- Division of Allergy, Pulmonary, and Critical Care, Department of Medicine (J.D.W., C.G., S.S., J.B., S.G., C.M., H.T., T.S.B., S.M.M.), Vanderbilt University Medical Center, Nashville, TN
| | - Timothy S Blackwell
- Division of Allergy, Pulmonary, and Critical Care, Department of Medicine (J.D.W., C.G., S.S., J.B., S.G., C.M., H.T., T.S.B., S.M.M.), Vanderbilt University Medical Center, Nashville, TN.,Department of Veterans Affairs Medical Center, Nashville, TN (T.S.B.)
| | - Susan M Majka
- Division of Allergy, Pulmonary, and Critical Care, Department of Medicine (J.D.W., C.G., S.S., J.B., S.G., C.M., H.T., T.S.B., S.M.M.), Vanderbilt University Medical Center, Nashville, TN
| | - W David Merryman
- From the Department of Biomedical Engineering (N.C.B., C.R.C., J.C.S., C.S., R.D., W.D.M.), Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
20
|
Dai Z, Zhu MM, Peng Y, Jin H, Machireddy N, Qian Z, Zhang X, Zhao YY. Endothelial and Smooth Muscle Cell Interaction via FoxM1 Signaling Mediates Vascular Remodeling and Pulmonary Hypertension. Am J Respir Crit Care Med 2019; 198:788-802. [PMID: 29664678 DOI: 10.1164/rccm.201709-1835oc] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
RATIONALE Angioproliferative vasculopathy is a hallmark of pulmonary arterial hypertension (PAH). However, little is known about how endothelial cell (EC) and smooth muscle cell (SMC) crosstalk regulates the angioproliferative vascular remodeling. OBJECTIVES To investigate the role of EC and SMC interaction and underlying signaling pathways in pulmonary hypertension (PH) development. METHODS SMC-specific Foxm1 (forkhead box M1) or Cxcr4 knockout mice, EC-specific Foxm1 or Egln1 knockout mice, and EC-specific Egln1/Cxcl12 double knockout mice were used to assess the role of FoxM1 on SMC proliferation and PH. Lung tissues and cells from patients with PAH were used to validate clinical relevance. FoxM1 inhibitor thiostrepton was used in Sugen 5416/hypoxia- and monocrotaline-challenged rats. MEASUREMENTS AND MAIN RESULTS FoxM1 expression was markedly upregulated in lungs and pulmonary arterial SMCs of patients with idiopathic PAH and four discrete PH rodent models. Mice with SMC- (but not EC-) specific deletion of Foxm1 were protected from hypoxia- or Sugen 5416/hypoxia-induced PH. The upregulation of FoxM1 in SMCs induced by multiple EC-derived factors (PDGF-B, CXCL12, ET-1, and MIF) mediated SMC proliferation. Genetic deletion of endothelial Cxcl12 in Egln1Tie2Cre mice or loss of its cognate receptor Cxcr4 in SMCs in hypoxia-treated mice inhibited FoxM1 expression, SMC proliferation, and PH. Accordingly, pharmacologic inhibition of FoxM1 inhibited severe PH in both Sugen 5416/hypoxia and monocrotaline-challenged rats. CONCLUSIONS Multiple factors derived from dysfunctional ECs induced FoxM1 expression in SMCs and activated FoxM1-dependent SMC proliferation, which contributes to pulmonary vascular remodeling and PH. Thus, targeting FoxM1 signaling represents a novel strategy for treatment of idiopathic PAH.
Collapse
Affiliation(s)
- Zhiyu Dai
- 1 Program for Lung and Vascular Biology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.,2 Division of Critical Care, Department of Pediatrics.,3 Department of Pharmacology and.,4 The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois; and
| | - Maggie M Zhu
- 1 Program for Lung and Vascular Biology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.,2 Division of Critical Care, Department of Pediatrics.,3 Department of Pharmacology and.,4 The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois; and
| | - Yi Peng
- 1 Program for Lung and Vascular Biology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.,2 Division of Critical Care, Department of Pediatrics.,3 Department of Pharmacology and.,4 The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois; and
| | - Hua Jin
- 1 Program for Lung and Vascular Biology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.,2 Division of Critical Care, Department of Pediatrics.,3 Department of Pharmacology and.,4 The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois; and
| | - Narsa Machireddy
- 1 Program for Lung and Vascular Biology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.,2 Division of Critical Care, Department of Pediatrics.,3 Department of Pharmacology and.,4 The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois; and
| | - Zhijian Qian
- 5 Department of Medicine, Division of Hematology/Oncology, University of Illinois at Chicago, Chicago, Illinois
| | - Xianming Zhang
- 1 Program for Lung and Vascular Biology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.,2 Division of Critical Care, Department of Pediatrics.,3 Department of Pharmacology and.,4 The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois; and
| | - You-Yang Zhao
- 1 Program for Lung and Vascular Biology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.,2 Division of Critical Care, Department of Pediatrics.,6 Department of Pharmacology, and.,7 Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,3 Department of Pharmacology and.,4 The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois; and
| |
Collapse
|
21
|
Stenmark KR, Frid MG, Graham BB, Tuder RM. Dynamic and diverse changes in the functional properties of vascular smooth muscle cells in pulmonary hypertension. Cardiovasc Res 2019; 114:551-564. [PMID: 29385432 DOI: 10.1093/cvr/cvy004] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 01/26/2018] [Indexed: 12/21/2022] Open
Abstract
Pulmonary hypertension (PH) is the end result of interaction between pulmonary vascular tone and a complex series of cellular and molecular events termed 'vascular remodelling'. The remodelling process, which can involve the entirety of pulmonary arterial vasculature, almost universally involves medial thickening, driven by increased numbers and hypertrophy of its principal cellular constituent, smooth muscle cells (SMCs). It is noted, however that SMCs comprise heterogeneous populations of cells, which can exhibit markedly different proliferative, inflammatory, and extracellular matrix production changes during remodelling. We further consider that these functional changes in SMCs of different phenotype and their role in PH are dynamic and may undergo significant changes over time (which we will refer to as cellular plasticity); no single property can account for the complexity of the contribution of SMC to pulmonary vascular remodelling. Thus, the approaches used to pharmacologically manipulate PH by targeting the SMC phenotype(s) must take into account processes that underlie dominant phenotypes that drive the disease. We present evidence for time- and location-specific changes in SMC proliferation in various animal models of PH; we highlight the transient nature (rather than continuous) of SMC proliferation, emphasizing that the heterogenic SMC populations that reside in different locations along the pulmonary vascular tree exhibit distinct responses to the stresses associated with the development of PH. We also consider that cells that have often been termed 'SMCs' may arise from many origins, including endothelial cells, fibroblasts and resident or circulating progenitors, and thus may contribute via distinct signalling pathways to the remodelling process. Ultimately, PH is characterized by long-lived, apoptosis-resistant SMC. In line with this key pathogenic characteristic, we address the acquisition of a pro-inflammatory phenotype by SMC that is essential to the development of PH. We present evidence that metabolic alterations akin to those observed in cancer cells (cytoplasmic and mitochondrial) directly contribute to the phenotype of the SM and SM-like cells involved in PH. Finally, we raise the possibility that SMCs transition from a proliferative to a senescent, pro-inflammatory and metabolically active phenotype over time.
Collapse
Affiliation(s)
- Kurt R Stenmark
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, 12700 E. 19th Avenue, RC2, B131, Aurora, CO 80045, USA
| | - Maria G Frid
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, 12700 E. 19th Avenue, RC2, B131, Aurora, CO 80045, USA
| | - Brian B Graham
- Pulmonary and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, 12700 E. 19th Avenue, RC2, B131, Aurora, CO 80045, USA
| | - Rubin M Tuder
- Pulmonary and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, 12700 E. 19th Avenue, RC2, B131, Aurora, CO 80045, USA
| |
Collapse
|
22
|
Gao H, Chen J, Chen T, Wang Y, Song Y, Dong Y, Zhao S, Machado RF. MicroRNA410 Inhibits Pulmonary Vascular Remodeling via Regulation of Nicotinamide Phosphoribosyltransferase. Sci Rep 2019; 9:9949. [PMID: 31289307 PMCID: PMC6616369 DOI: 10.1038/s41598-019-46352-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 06/20/2019] [Indexed: 11/21/2022] Open
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) upregulation in human pulmonary artery endothelial cells (hPAECs) is associated with pulmonary arterial hypertension (PAH) progression and pulmonary vascular remodeling. The underlying mechanisms regulating NAMPT expression are still not clear. In this study, we aimed to study the regulation of NAMPT expression by microRNA410 (miR410) in hPAECs and explore the role of miR410 in the pathogenesis of experimental pulmonary hypertension. We show that miR410 targets the 3' UTR of NAMPT and that, concomitant with NAMPT upregulation, miR410 is downregulated in lungs of mice exposed to hypoxia-induced pulmonary hypertension (HPH). Our results also demonstrate that miR410 directly inhibits NAMPT expression. Overexpression of miR410 in hPAECs inhibits basal and VEGF-induced proliferation, migration and promotes apoptosis of hPAECs, while miR410 inhibition via antagomirs has the opposite effect. Finally, administration of miR410 mimics in vivo attenuated induction of NAMPT in PAECs and prevented the development of HPH in mice. Our results highlight the role of miR410 in the regulation of NAMPT expression in hPAECs and show that miR410 plays a potential role in PAH pathobiology by targeting a modulator of pulmonary vascular remodeling.
Collapse
Affiliation(s)
- Hui Gao
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jiwang Chen
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Tianji Chen
- Department of Pediatrics, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Yifang Wang
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Yang Song
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Yangbasai Dong
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Shuangping Zhao
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Roberto F Machado
- Division of Pulmonary, Critical Care, Sleep, and Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, IN, 46202, USA.
| |
Collapse
|
23
|
Kurosawa R, Satoh K, Kikuchi N, Kikuchi H, Saigusa D, Al-Mamun ME, Siddique MAH, Omura J, Satoh T, Sunamura S, Nogi M, Numano K, Miyata S, Uruno A, Kano K, Matsumoto Y, Doi T, Aoki J, Oshima Y, Yamamoto M, Shimokawa H. Identification of Celastramycin as a Novel Therapeutic Agent for Pulmonary Arterial Hypertension. Circ Res 2019; 125:309-327. [PMID: 31195886 DOI: 10.1161/circresaha.119.315229] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
RATIONALE Pulmonary arterial hypertension (PAH) is characterized by enhanced proliferation of pulmonary artery smooth muscle cells (PASMCs) accompanying increased production of inflammatory factors and adaptation of the mitochondrial metabolism to a hyperproliferative state. However, all the drugs in clinical use target pulmonary vascular dilatation, which may not be effective for patients with advanced PAH. OBJECTIVE We aimed to discover a novel drug for PAH that inhibits PASMC proliferation. METHODS AND RESULTS We screened 5562 compounds from original library using high-throughput screening system to discover compounds which inhibit proliferation of PASMCs from patients with PAH (PAH-PASMCs). We found that celastramycin, a benzoyl pyrrole-type compound originally found in a bacteria extract, inhibited the proliferation of PAH-PASMCs in a dose-dependent manner with relatively small effects on PASMCs from healthy donors. Then, we made 25 analogs of celastramycin and selected the lead compound, which significantly inhibited cell proliferation of PAH-PASMCs and reduced cytosolic reactive oxygen species levels. Mechanistic analysis demonstrated that celastramycin reduced the protein levels of HIF-1α (hypoxia-inducible factor 1α), which impairs aerobic metabolism, and κB (nuclear factor-κB), which induces proinflammatory signals, in PAH-PASMCs, leading to reduced secretion of inflammatory cytokine. Importantly, celastramycin treatment reduced reactive oxygen species levels in PAH-PASMCs with increased protein levels of Nrf2 (nuclear factor erythroid 2-related factor 2), a master regulator of cellular response against oxidative stress. Furthermore, celastramycin treatment improved mitochondrial energy metabolism with recovered mitochondrial network formation in PAH-PASMCs. Moreover, these celastramycin-mediated effects were regulated by ZFC3H1 (zinc finger C3H1 domain-containing protein), a binding partner of celastramycin. Finally, celastramycin treatment ameliorated pulmonary hypertension in 3 experimental animal models, accompanied by reduced inflammatory changes in the lungs. CONCLUSIONS These results indicate that celastramycin ameliorates pulmonary hypertension, reducing excessive proliferation of PAH-PASMCs with less inflammation and reactive oxygen species levels, and recovered mitochondrial energy metabolism. Thus, celastramycin is a novel drug for PAH that targets antiproliferative effects on PAH-PASMCs.
Collapse
Affiliation(s)
- Ryo Kurosawa
- From the Department of Cardiovascular Medicine (R.K., K.S., N.K., E.A.M., M.A.H.S., J.O., T.S., S.S., M.N., K.N., S.M., H.S.), Sendai, Japan.,Japan Society for the Promotion of Science, Tokyo, Japan (R.K.)
| | - Kimio Satoh
- From the Department of Cardiovascular Medicine (R.K., K.S., N.K., E.A.M., M.A.H.S., J.O., T.S., S.S., M.N., K.N., S.M., H.S.), Sendai, Japan
| | - Nobuhiro Kikuchi
- From the Department of Cardiovascular Medicine (R.K., K.S., N.K., E.A.M., M.A.H.S., J.O., T.S., S.S., M.N., K.N., S.M., H.S.), Sendai, Japan
| | - Haruhisa Kikuchi
- Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan (H.K., K.K., Y.M., T.D., J.A., Y.O.)
| | - Daisuke Saigusa
- Department of Integrative Genomics, Tohoku University Tohoku Medical Megabank Organizaition (D.S., A.U., M.Y.), Sendai, Japan.,Department of Medical Biochemistry, Tohoku University Graduate School of Medicine (D.S., A.U., M.Y.), Sendai, Japan
| | - Md Elias Al-Mamun
- From the Department of Cardiovascular Medicine (R.K., K.S., N.K., E.A.M., M.A.H.S., J.O., T.S., S.S., M.N., K.N., S.M., H.S.), Sendai, Japan
| | - Mohammad A H Siddique
- From the Department of Cardiovascular Medicine (R.K., K.S., N.K., E.A.M., M.A.H.S., J.O., T.S., S.S., M.N., K.N., S.M., H.S.), Sendai, Japan
| | - Junichi Omura
- From the Department of Cardiovascular Medicine (R.K., K.S., N.K., E.A.M., M.A.H.S., J.O., T.S., S.S., M.N., K.N., S.M., H.S.), Sendai, Japan
| | - Taijyu Satoh
- From the Department of Cardiovascular Medicine (R.K., K.S., N.K., E.A.M., M.A.H.S., J.O., T.S., S.S., M.N., K.N., S.M., H.S.), Sendai, Japan
| | - Shinichiro Sunamura
- From the Department of Cardiovascular Medicine (R.K., K.S., N.K., E.A.M., M.A.H.S., J.O., T.S., S.S., M.N., K.N., S.M., H.S.), Sendai, Japan
| | - Masamichi Nogi
- From the Department of Cardiovascular Medicine (R.K., K.S., N.K., E.A.M., M.A.H.S., J.O., T.S., S.S., M.N., K.N., S.M., H.S.), Sendai, Japan
| | - Kazuhiko Numano
- From the Department of Cardiovascular Medicine (R.K., K.S., N.K., E.A.M., M.A.H.S., J.O., T.S., S.S., M.N., K.N., S.M., H.S.), Sendai, Japan
| | - Satoshi Miyata
- From the Department of Cardiovascular Medicine (R.K., K.S., N.K., E.A.M., M.A.H.S., J.O., T.S., S.S., M.N., K.N., S.M., H.S.), Sendai, Japan
| | - Akira Uruno
- Department of Integrative Genomics, Tohoku University Tohoku Medical Megabank Organizaition (D.S., A.U., M.Y.), Sendai, Japan.,Department of Medical Biochemistry, Tohoku University Graduate School of Medicine (D.S., A.U., M.Y.), Sendai, Japan
| | - Kuniyuki Kano
- Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan (H.K., K.K., Y.M., T.D., J.A., Y.O.)
| | - Yotaro Matsumoto
- Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan (H.K., K.K., Y.M., T.D., J.A., Y.O.)
| | - Takayuki Doi
- Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan (H.K., K.K., Y.M., T.D., J.A., Y.O.)
| | - Junken Aoki
- Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan (H.K., K.K., Y.M., T.D., J.A., Y.O.)
| | - Yoshiteru Oshima
- Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan (H.K., K.K., Y.M., T.D., J.A., Y.O.)
| | - Masayuki Yamamoto
- Department of Integrative Genomics, Tohoku University Tohoku Medical Megabank Organizaition (D.S., A.U., M.Y.), Sendai, Japan.,Department of Medical Biochemistry, Tohoku University Graduate School of Medicine (D.S., A.U., M.Y.), Sendai, Japan
| | - Hiroaki Shimokawa
- From the Department of Cardiovascular Medicine (R.K., K.S., N.K., E.A.M., M.A.H.S., J.O., T.S., S.S., M.N., K.N., S.M., H.S.), Sendai, Japan
| |
Collapse
|
24
|
Wang Z, Jiang X, Zhang X, Tian G, Yang R, Wu J, Zou X, Liu Z, Yang X, Wu C, Shi J, Li J, Suo J, Wang Y, Zhang R, Xu Z, Gong X, He Y, Zhu W, Aisa HA, Jiang H, Xu Y, Shen J. Pharmacokinetics-Driven Optimization of 4(3 H)-Pyrimidinones as Phosphodiesterase Type 5 Inhibitors Leading to TPN171, a Clinical Candidate for the Treatment of Pulmonary Arterial Hypertension. J Med Chem 2019; 62:4979-4990. [PMID: 31021628 DOI: 10.1021/acs.jmedchem.9b00123] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Phosphodiesterase type 5 (PDE5) inhibitors are first-line therapy for pulmonary arterial hypertension (PAH) and erectile dysfunction. As a continuing work to improve the terminal half-lives and oral bioavailabilities of our previously reported 4(3 H)-pyrimidones, a pharmacokinetics-driven optimization focusing on the terminal substituent is described. Two major congeneric series of 4(3 H)-pyrimidones, the aminosulfonylphenylpyrimidones and acylaminophenylpyrimidones, were designed, synthesized, and pharmacologically assessed in vitro and in vivo. Among them, compound 15 (TPN171) with subnanomolar potency for PDE5 and good selectivity over PDE6 was finally recognized as a potential drug candidate, and its pharmacokinetic profiles in rats and dogs are significantly improved compared to the starting compound (3). Moreover, TPN171 was proven to exert a longer lasting effect than sildenafil in animal models, providing a foundation for a once-daily oral administration for its clinical use. TPN171 is currently being investigated in a phase II clinical trial for the treatment of PAH.
Collapse
Affiliation(s)
- Zhen Wang
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203 , China
| | - Xiangrui Jiang
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203 , China
| | - Xianglei Zhang
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203 , China.,School of Pharmacy , University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Guanghui Tian
- Vigonvita Life Science Co., Ltd. , Suzhou 215123 , China
| | - Rulei Yang
- Vigonvita Life Science Co., Ltd. , Suzhou 215123 , China
| | - Jianzhong Wu
- Vigonvita Life Science Co., Ltd. , Suzhou 215123 , China
| | - Xiaoli Zou
- Vigonvita Life Science Co., Ltd. , Suzhou 215123 , China
| | - Zheng Liu
- Topharman Shanghai Co., Ltd. , Shanghai 201203 , China
| | - Xiaojun Yang
- Topharman Shanghai Co., Ltd. , Shanghai 201203 , China
| | - Chunhui Wu
- Topharman Shanghai Co., Ltd. , Shanghai 201203 , China
| | - Jing Shi
- Topharman Shanghai Co., Ltd. , Shanghai 201203 , China
| | - Jianfeng Li
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203 , China
| | - Jin Suo
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203 , China
| | - Yu Wang
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203 , China
| | - Rongxia Zhang
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203 , China
| | - Zhijian Xu
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203 , China
| | - Xudong Gong
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203 , China.,Key Laboratory of Plant Resources and Chemistry in Arid Regions , Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences , Urumqi 830011 , China
| | - Yang He
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203 , China
| | - Weiliang Zhu
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203 , China.,School of Pharmacy , University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Haji Akber Aisa
- Key Laboratory of Plant Resources and Chemistry in Arid Regions , Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences , Urumqi 830011 , China
| | - Hualiang Jiang
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203 , China.,School of Pharmacy , University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Yechun Xu
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203 , China.,School of Pharmacy , University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Jingshan Shen
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203 , China.,School of Pharmacy , University of Chinese Academy of Sciences , Beijing 100049 , China
| |
Collapse
|
25
|
Grinnan D, Trankle C, Andruska A, Bloom B, Spiekerkoetter E. Drug repositioning in pulmonary arterial hypertension: challenges and opportunities. Pulm Circ 2019; 9:2045894019832226. [PMID: 30729869 PMCID: PMC6852366 DOI: 10.1177/2045894019832226] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Despite many advances in medical therapy for pulmonary arterial hypertension (PAH) over the past 20 years, long-term survival is still poor. Novel therapies which target the underlying pathology of PAH and which could be added to current vasodilatory therapies to halt disease progression and potentially reverse pulmonary vascular remodeling are highly sought after. Given the high attrition rates, substantial costs, and slow pace of new drug development, repositioning of “old” drugs is increasingly becoming an attractive path to identify novel treatment options, especially for a rare disease such as PAH. We here summarize the limitations of current PAH therapy, the general concept of repurposing and repositioning, success stories of approved repositioned drugs in PAH as well as novel repositioned drugs that show promise in preclinical models of pulmonary hypertension (PH) and are currently tested in clinical trials. We furthermore discuss various data-driven as well as experimental approaches currently used to identify repurposed drug candidates and review challenges for the “repositioning community” with regards to funding and patent and regulatory considerations, and to illustrate opportunities for collaborative solutions for drug repositioning relevant to PAH.
Collapse
Affiliation(s)
- Daniel Grinnan
- 1 Department of Medicine, Division of Pulmonary and Critical Care Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Cory Trankle
- 2 Department of Medicine, Division of Cardiology, Virginia Commonwealth University, Richmond, VA, USA
| | - Adam Andruska
- 3 Department of Medicine, Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, CA, USA.,4 Wall Center for Pulmonary Vascular Disease, Stanford, CA, USA
| | | | - Edda Spiekerkoetter
- 3 Department of Medicine, Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, CA, USA.,4 Wall Center for Pulmonary Vascular Disease, Stanford, CA, USA
| |
Collapse
|
26
|
Kitagawa MG, Reynolds JO, Durgan D, Rodney G, Karmouty‐Quintana H, Bryan R, Pandit LM. Twik-2 -/- mouse demonstrates pulmonary vascular heterogeneity in intracellular pathways for vasocontractility. Physiol Rep 2019; 7:e13950. [PMID: 30632293 PMCID: PMC6328926 DOI: 10.14814/phy2.13950] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/06/2018] [Accepted: 11/15/2018] [Indexed: 11/24/2022] Open
Abstract
We have previously shown Twik-2-/- mice develop pulmonary hypertension and vascular remodeling. We hypothesized that distal pulmonary arteries (D-PAs) of the Twik-2-/- mice are hypercontractile under physiological venous conditions due to altered electrophysiologic properties between the conduit and resistance vessels in the pulmonary vascular bed. We measured resting membrane potential and intracellular calcium through Fura-2 in freshly digested pulmonary artery smooth muscles (PASMCs) from both the right main (RM-PA) and D-PA (distal) regions of pulmonary artery from WT and Twik-2-/- mice. Whole segments of RM-PAs and D-PAs from 20 to 24-week-old wildtype (WT) and Twik-2-/- mice were also pressurized between two glass micropipettes and bathed in buffer with either arterial or venous conditions. Abluminally-applied phenylephrine (PE) and U46619 were added to the buffer at log increments and vessel diameter was measured. All values were expressed as averages with ±SEM. Vasoconstrictor responses did not differ between WT and Twik-2-/- RM-PAs under arterial conditions. Under venous conditions, Twik-2-/- RM-PAs showed an increased sensitivity to PE with a lower EC50 (P = 0.02). Under venous conditions, Twik-2-/- D-PAs showed an increase maximal vasoconstrictor response to both phenylephrine and U46619 compared to the WT mice (P < 0.05). Isolated PASMCs from Twik-2 -/- D-PA were depolarized and had higher intracellular calcium levels compared to PASMCs from RM-PA of both WT and Twik-2-/- mice. These studies suggest that hypercontractile responses and electrophysiologic properties unique to the anatomic location of the D-PAs may contribute to pulmonary hypertensive vasculopathy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Lavannya M. Pandit
- Baylor College of MedicineHoustonTexas
- Michael E.DeBakey Veterans Affairs Medical CenterHoustonTexas
| |
Collapse
|
27
|
Jansen JM. Evangelos Michelakis. Circ Res 2018; 123:938-941. [PMID: 30355034 DOI: 10.1161/circresaha.118.314005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
28
|
Gurtu V, Michelakis ED. A Paradigm Shift Is Needed in the Field of Pulmonary Arterial Hypertension for Its Entrance Into the Precision Medicine Era. Circ Res 2018; 119:1276-1279. [PMID: 27932471 DOI: 10.1161/circresaha.116.309689] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Vikram Gurtu
- From the Division of Cardiology, Department of Medicine, University of Alberta, 2C2 Walter Mackenzie Centre, Edmonton, Canada
| | - Evangelos D Michelakis
- From the Division of Cardiology, Department of Medicine, University of Alberta, 2C2 Walter Mackenzie Centre, Edmonton, Canada.
| |
Collapse
|
29
|
Kikuchi N, Satoh K, Kurosawa R, Yaoita N, Elias-Al-Mamun M, Siddique MAH, Omura J, Satoh T, Nogi M, Sunamura S, Miyata S, Saito Y, Hoshikawa Y, Okada Y, Shimokawa H. Selenoprotein P Promotes the Development of Pulmonary Arterial Hypertension: Possible Novel Therapeutic Target. Circulation 2018; 138:600-623. [PMID: 29636330 DOI: 10.1161/circulationaha.117.033113] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 03/28/2018] [Indexed: 02/05/2023]
Abstract
BACKGROUND Excessive proliferation and apoptosis resistance of pulmonary artery smooth muscle cells (PASMCs) are key mechanisms of pulmonary arterial hypertension (PAH). Despite the multiple combination therapy, a considerable number of patients develop severe pulmonary hypertension (PH) because of the lack of diagnostic biomarker and antiproliferative therapies for PASMCs. METHODS Microarray analyses were used to identify a novel therapeutic target for PAH. In vitro experiments, including lung and serum samples from patients with PAH, cultured PAH-PASMCs, and high-throughput screening of 3336 low-molecular-weight compounds, were used for mechanistic study and exploring a novel therapeutic agent. Five genetically modified mouse strains, including PASMC-specific selenoprotein P (SeP) knockout mice and PH model rats, were used to study the role of SeP and therapeutic capacity of the compounds for the development of PH in vivo. RESULTS Microarray analysis revealed a 32-fold increase in SeP in PAH-PASMCs compared with control PASMCs. SeP is a widely expressed extracellular protein maintaining cellular metabolism. Immunoreactivity of SeP was enhanced in the thickened media of pulmonary arteries in PAH. Serum SeP levels were also elevated in patients with PH compared with controls, and high serum SeP predicted poor outcome. SeP-knockout mice ( SeP-/-) exposed to chronic hypoxia showed significantly reduced right ventricular systolic pressure, right ventricular hypertrophy, and pulmonary artery remodeling compared with controls. In contrast, systemic SeP-overexpressing mice showed exacerbation of hypoxia-induced PH. Furthermore, PASMC-specific SeP-/- mice showed reduced hypoxia-induced PH compared with controls, whereas neither liver-specific SeP knockout nor liver-specific SeP-overexpressing mice showed significant differences with controls. Altogether, protein levels of SeP in the lungs were associated with the development of PH. Mechanistic experiments demonstrated that SeP promotes PASMC proliferation and resistance to apoptosis through increased oxidative stress and mitochondrial dysfunction, which were associated with activated hypoxia-inducible factor-1α and dysregulated glutathione metabolism. It is important to note that the high-throughput screening of 3336 compounds identified that sanguinarine, a plant alkaloid with antiproliferative effects, reduced SeP expression and proliferation in PASMCs and ameliorated PH in mice and rats. CONCLUSIONS These results indicate that SeP promotes the development of PH, suggesting that it is a novel biomarker and therapeutic target of the disorder.
Collapse
MESH Headings
- Animals
- Antihypertensive Agents/pharmacology
- Apoptosis
- Arterial Pressure/drug effects
- Benzophenanthridines/pharmacology
- Cell Proliferation
- Cells, Cultured
- Disease Models, Animal
- Humans
- Hypertension, Pulmonary/etiology
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/physiopathology
- Hypertension, Pulmonary/prevention & control
- Hypoxia/complications
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Isoquinolines/pharmacology
- Male
- Mice, Knockout
- Mitochondria, Muscle/metabolism
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/physiopathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Oxidative Stress
- Pulmonary Artery/metabolism
- Pulmonary Artery/physiopathology
- Rats, Sprague-Dawley
- Selenoprotein P/metabolism
- Signal Transduction
- Vascular Remodeling/drug effects
Collapse
Affiliation(s)
- Nobuhiro Kikuchi
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (N.K., K.S., R.K., S.M., N.Y., M.E.-A.-M., M.A.H.S., J.O., T.S., M.N., S.S., H.S.)
- Research Fellow of Japan Society for the Promotion of Science, Tokyo (N.K., R.K.)
| | - Kimio Satoh
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (N.K., K.S., R.K., S.M., N.Y., M.E.-A.-M., M.A.H.S., J.O., T.S., M.N., S.S., H.S.)
| | - Ryo Kurosawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (N.K., K.S., R.K., S.M., N.Y., M.E.-A.-M., M.A.H.S., J.O., T.S., M.N., S.S., H.S.)
- Research Fellow of Japan Society for the Promotion of Science, Tokyo (N.K., R.K.)
| | - Nobuhiro Yaoita
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (N.K., K.S., R.K., S.M., N.Y., M.E.-A.-M., M.A.H.S., J.O., T.S., M.N., S.S., H.S.)
| | - Md Elias-Al-Mamun
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (N.K., K.S., R.K., S.M., N.Y., M.E.-A.-M., M.A.H.S., J.O., T.S., M.N., S.S., H.S.)
| | - Mohammad Abdul Hai Siddique
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (N.K., K.S., R.K., S.M., N.Y., M.E.-A.-M., M.A.H.S., J.O., T.S., M.N., S.S., H.S.)
| | - Junichi Omura
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (N.K., K.S., R.K., S.M., N.Y., M.E.-A.-M., M.A.H.S., J.O., T.S., M.N., S.S., H.S.)
| | - Taijyu Satoh
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (N.K., K.S., R.K., S.M., N.Y., M.E.-A.-M., M.A.H.S., J.O., T.S., M.N., S.S., H.S.)
| | - Masamichi Nogi
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (N.K., K.S., R.K., S.M., N.Y., M.E.-A.-M., M.A.H.S., J.O., T.S., M.N., S.S., H.S.)
| | - Shinichiro Sunamura
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (N.K., K.S., R.K., S.M., N.Y., M.E.-A.-M., M.A.H.S., J.O., T.S., M.N., S.S., H.S.)
| | - Satoshi Miyata
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (N.K., K.S., R.K., S.M., N.Y., M.E.-A.-M., M.A.H.S., J.O., T.S., M.N., S.S., H.S.)
| | - Yoshiro Saito
- Department of Medical Life Systems, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe, Japan (Y.S.)
| | - Yasushi Hoshikawa
- Department of Thoracic Surgery, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan (Y.H., Y.O.)
| | - Yoshinori Okada
- Department of Thoracic Surgery, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan (Y.H., Y.O.)
| | - Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (N.K., K.S., R.K., S.M., N.Y., M.E.-A.-M., M.A.H.S., J.O., T.S., M.N., S.S., H.S.)
| |
Collapse
|
30
|
Michelakis ED, Gurtu V, Webster L, Barnes G, Watson G, Howard L, Cupitt J, Paterson I, Thompson RB, Chow K, O'Regan DP, Zhao L, Wharton J, Kiely DG, Kinnaird A, Boukouris AE, White C, Nagendran J, Freed DH, Wort SJ, Gibbs JSR, Wilkins MR. Inhibition of pyruvate dehydrogenase kinase improves pulmonary arterial hypertension in genetically susceptible patients. Sci Transl Med 2018; 9:9/413/eaao4583. [PMID: 29070699 DOI: 10.1126/scitranslmed.aao4583] [Citation(s) in RCA: 204] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 09/22/2017] [Indexed: 12/17/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a progressive vascular disease with a high mortality rate. It is characterized by an occlusive vascular remodeling due to a pro-proliferative and antiapoptotic environment in the wall of resistance pulmonary arteries (PAs). Proliferating cells exhibit a cancer-like metabolic switch where mitochondrial glucose oxidation is suppressed, whereas glycolysis is up-regulated as the major source of adenosine triphosphate production. This multifactorial mitochondrial suppression leads to inhibition of apoptosis and downstream signaling promoting proliferation. We report an increase in pyruvate dehydrogenase kinase (PDK), an inhibitor of the mitochondrial enzyme pyruvate dehydrogenase (PDH, the gatekeeping enzyme of glucose oxidation) in the PAs of human PAH compared to healthy lungs. Treatment of explanted human PAH lungs with the PDK inhibitor dichloroacetate (DCA) ex vivo activated PDH and increased mitochondrial respiration. In a 4-month, open-label study, DCA (3 to 6.25 mg/kg b.i.d.) administered to patients with idiopathic PAH (iPAH) already on approved iPAH therapies led to reduction in mean PA pressure and pulmonary vascular resistance and improvement in functional capacity, but with a range of individual responses. Lack of ex vivo and clinical response was associated with the presence of functional variants of SIRT3 and UCP2 that predict reduced protein function. Impaired function of these proteins causes PDK-independent mitochondrial suppression and pulmonary hypertension in mice. This first-in-human trial of a mitochondria-targeting drug in iPAH demonstrates that PDK is a druggable target and offers hemodynamic improvement in genetically susceptible patients, paving the way for novel precision medicine approaches in this disease.
Collapse
Affiliation(s)
| | - Vikram Gurtu
- Department of Medicine, University of Alberta, Edmonton, Alberta T6G2B7, Canada
| | - Linda Webster
- Department of Medicine, University of Alberta, Edmonton, Alberta T6G2B7, Canada
| | - Gareth Barnes
- Department of Medicine, Imperial College London, London W12 0NN, UK
| | - Geoffrey Watson
- Department of Medicine, Imperial College London, London W12 0NN, UK
| | - Luke Howard
- National Pulmonary Hypertension Service, Imperial College Healthcare National Health Service Trust, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - John Cupitt
- Department of Medicine, Imperial College London, London W12 0NN, UK
| | - Ian Paterson
- Department of Medicine, University of Alberta, Edmonton, Alberta T6G2B7, Canada
| | - Richard B Thompson
- Department of Biomedical Engineering, University of Alberta, Edmonton, Alberta T6G2B7, Canada
| | - Kelvin Chow
- Department of Biomedical Engineering, University of Alberta, Edmonton, Alberta T6G2B7, Canada
| | - Declan P O'Regan
- Medical Research Council, London Institute of Medical Sciences, Hammersmith Hospital Campus, Imperial College London, London W12 0NN, UK
| | - Lan Zhao
- Department of Medicine, Imperial College London, London W12 0NN, UK
| | - John Wharton
- Department of Medicine, Imperial College London, London W12 0NN, UK
| | - David G Kiely
- Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, Sheffield S10 2JF, UK
| | - Adam Kinnaird
- Department of Medicine, University of Alberta, Edmonton, Alberta T6G2B7, Canada
| | | | - Chris White
- Department of Surgery, University of Alberta, Edmonton, Alberta T6G2B7, Canada
| | - Jayan Nagendran
- Department of Surgery, University of Alberta, Edmonton, Alberta T6G2B7, Canada
| | - Darren H Freed
- Department of Surgery, University of Alberta, Edmonton, Alberta T6G2B7, Canada
| | - Stephen J Wort
- National Heart and Lung Institute, Imperial College London, Dovehouse Street, London SW3 6LY, UK
| | - J Simon R Gibbs
- National Pulmonary Hypertension Service, Imperial College Healthcare National Health Service Trust, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - Martin R Wilkins
- Department of Medicine, Imperial College London, London W12 0NN, UK.
| |
Collapse
|
31
|
Dumas SJ, Bru-Mercier G, Courboulin A, Quatredeniers M, Rücker-Martin C, Antigny F, Nakhleh MK, Ranchoux B, Gouadon E, Vinhas MC, Vocelle M, Raymond N, Dorfmüller P, Fadel E, Perros F, Humbert M, Cohen-Kaminsky S. NMDA-Type Glutamate Receptor Activation Promotes Vascular Remodeling and Pulmonary Arterial Hypertension. Circulation 2018; 137:2371-2389. [PMID: 29444988 DOI: 10.1161/circulationaha.117.029930] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 12/22/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND Excessive proliferation and apoptosis resistance in pulmonary vascular cells underlie vascular remodeling in pulmonary arterial hypertension (PAH). Specific treatments for PAH exist, mostly targeting endothelial dysfunction, but high pulmonary arterial pressure still causes heart failure and death. Pulmonary vascular remodeling may be driven by metabolic reprogramming of vascular cells to increase glutaminolysis and glutamate production. The N-methyl-d-aspartate receptor (NMDAR), a major neuronal glutamate receptor, is also expressed on vascular cells, but its role in PAH is unknown. METHODS We assessed the status of the glutamate-NMDAR axis in the pulmonary arteries of patients with PAH and controls through mass spectrometry imaging, Western blotting, and immunohistochemistry. We measured the glutamate release from cultured pulmonary vascular cells using enzymatic assays and analyzed NMDAR regulation/phosphorylation through Western blot experiments. The effect of NMDAR blockade on human pulmonary arterial smooth muscle cell proliferation was determined using a BrdU incorporation assay. We assessed the role of NMDARs in vascular remodeling associated to pulmonary hypertension, in both smooth muscle-specific NMDAR knockout mice exposed to chronic hypoxia and the monocrotaline rat model of pulmonary hypertension using NMDAR blockers. RESULTS We report glutamate accumulation, upregulation of the NMDAR, and NMDAR engagement reflected by increases in GluN1-subunit phosphorylation in the pulmonary arteries of human patients with PAH. Kv channel inhibition and type A-selective endothelin receptor activation amplified calcium-dependent glutamate release from human pulmonary arterial smooth muscle cell, and type A-selective endothelin receptor and platelet-derived growth factor receptor activation led to NMDAR engagement, highlighting crosstalk between the glutamate-NMDAR axis and major PAH-associated pathways. The platelet-derived growth factor-BB-induced proliferation of human pulmonary arterial smooth muscle cells involved NMDAR activation and phosphorylated GluN1 subunit localization to cell-cell contacts, consistent with glutamatergic communication between proliferating human pulmonary arterial smooth muscle cells via NMDARs. Smooth-muscle NMDAR deficiency in mice attenuated the vascular remodeling triggered by chronic hypoxia, highlighting the role of vascular NMDARs in pulmonary hypertension. Pharmacological NMDAR blockade in the monocrotaline rat model of pulmonary hypertension had beneficial effects on cardiac and vascular remodeling, decreasing endothelial dysfunction, cell proliferation, and apoptosis resistance while disrupting the glutamate-NMDAR pathway in pulmonary arteries. CONCLUSIONS These results reveal a dysregulation of the glutamate-NMDAR axis in the pulmonary arteries of patients with PAH and identify vascular NMDARs as targets for antiremodeling treatments in PAH.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Calcium/pharmacology
- Cell Proliferation/drug effects
- Disease Models, Animal
- Dizocilpine Maleate/pharmacology
- Endothelin-1/pharmacology
- Glutamic Acid/metabolism
- Humans
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/pathology
- Lung/metabolism
- Lung/pathology
- Mice
- Mice, Knockout
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Potassium Channels, Voltage-Gated/metabolism
- Rats
- Receptors, Endothelin/chemistry
- Receptors, Endothelin/metabolism
- Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors
- Receptors, N-Methyl-D-Aspartate/genetics
- Receptors, N-Methyl-D-Aspartate/metabolism
- Signal Transduction/drug effects
- Vascular Remodeling/drug effects
Collapse
Affiliation(s)
- Sébastien J Dumas
- INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France (S.J.D., G.B.-M., A.C., M.Q., C.R.-M, F.A., M.K.N., B.R., E.G., M.-C.V., M.V., N.R., P.D., E.F., F.P., M.H., S.C.-K.)
- University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France (S.J.D., G.B.-M., A.C., M.Q., C.R.-M, F.A., M.K.N., B.R., E.G., M.-C.V., M.V., N.R., P.D., E.F., F.P., M.H., S.C.-K.)
| | - Gilles Bru-Mercier
- INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France (S.J.D., G.B.-M., A.C., M.Q., C.R.-M, F.A., M.K.N., B.R., E.G., M.-C.V., M.V., N.R., P.D., E.F., F.P., M.H., S.C.-K.)
- University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France (S.J.D., G.B.-M., A.C., M.Q., C.R.-M, F.A., M.K.N., B.R., E.G., M.-C.V., M.V., N.R., P.D., E.F., F.P., M.H., S.C.-K.)
| | - Audrey Courboulin
- INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France (S.J.D., G.B.-M., A.C., M.Q., C.R.-M, F.A., M.K.N., B.R., E.G., M.-C.V., M.V., N.R., P.D., E.F., F.P., M.H., S.C.-K.)
- University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France (S.J.D., G.B.-M., A.C., M.Q., C.R.-M, F.A., M.K.N., B.R., E.G., M.-C.V., M.V., N.R., P.D., E.F., F.P., M.H., S.C.-K.)
| | - Marceau Quatredeniers
- INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France (S.J.D., G.B.-M., A.C., M.Q., C.R.-M, F.A., M.K.N., B.R., E.G., M.-C.V., M.V., N.R., P.D., E.F., F.P., M.H., S.C.-K.)
- University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France (S.J.D., G.B.-M., A.C., M.Q., C.R.-M, F.A., M.K.N., B.R., E.G., M.-C.V., M.V., N.R., P.D., E.F., F.P., M.H., S.C.-K.)
| | - Catherine Rücker-Martin
- INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France (S.J.D., G.B.-M., A.C., M.Q., C.R.-M, F.A., M.K.N., B.R., E.G., M.-C.V., M.V., N.R., P.D., E.F., F.P., M.H., S.C.-K.)
- University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France (S.J.D., G.B.-M., A.C., M.Q., C.R.-M, F.A., M.K.N., B.R., E.G., M.-C.V., M.V., N.R., P.D., E.F., F.P., M.H., S.C.-K.)
| | - Fabrice Antigny
- INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France (S.J.D., G.B.-M., A.C., M.Q., C.R.-M, F.A., M.K.N., B.R., E.G., M.-C.V., M.V., N.R., P.D., E.F., F.P., M.H., S.C.-K.)
- University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France (S.J.D., G.B.-M., A.C., M.Q., C.R.-M, F.A., M.K.N., B.R., E.G., M.-C.V., M.V., N.R., P.D., E.F., F.P., M.H., S.C.-K.)
| | - Morad K Nakhleh
- INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France (S.J.D., G.B.-M., A.C., M.Q., C.R.-M, F.A., M.K.N., B.R., E.G., M.-C.V., M.V., N.R., P.D., E.F., F.P., M.H., S.C.-K.)
- University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France (S.J.D., G.B.-M., A.C., M.Q., C.R.-M, F.A., M.K.N., B.R., E.G., M.-C.V., M.V., N.R., P.D., E.F., F.P., M.H., S.C.-K.)
| | - Benoit Ranchoux
- INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France (S.J.D., G.B.-M., A.C., M.Q., C.R.-M, F.A., M.K.N., B.R., E.G., M.-C.V., M.V., N.R., P.D., E.F., F.P., M.H., S.C.-K.)
- University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France (S.J.D., G.B.-M., A.C., M.Q., C.R.-M, F.A., M.K.N., B.R., E.G., M.-C.V., M.V., N.R., P.D., E.F., F.P., M.H., S.C.-K.)
| | - Elodie Gouadon
- INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France (S.J.D., G.B.-M., A.C., M.Q., C.R.-M, F.A., M.K.N., B.R., E.G., M.-C.V., M.V., N.R., P.D., E.F., F.P., M.H., S.C.-K.)
- University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France (S.J.D., G.B.-M., A.C., M.Q., C.R.-M, F.A., M.K.N., B.R., E.G., M.-C.V., M.V., N.R., P.D., E.F., F.P., M.H., S.C.-K.)
| | - Maria-Candida Vinhas
- INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France (S.J.D., G.B.-M., A.C., M.Q., C.R.-M, F.A., M.K.N., B.R., E.G., M.-C.V., M.V., N.R., P.D., E.F., F.P., M.H., S.C.-K.)
- University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France (S.J.D., G.B.-M., A.C., M.Q., C.R.-M, F.A., M.K.N., B.R., E.G., M.-C.V., M.V., N.R., P.D., E.F., F.P., M.H., S.C.-K.)
| | - Matthieu Vocelle
- INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France (S.J.D., G.B.-M., A.C., M.Q., C.R.-M, F.A., M.K.N., B.R., E.G., M.-C.V., M.V., N.R., P.D., E.F., F.P., M.H., S.C.-K.)
- University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France (S.J.D., G.B.-M., A.C., M.Q., C.R.-M, F.A., M.K.N., B.R., E.G., M.-C.V., M.V., N.R., P.D., E.F., F.P., M.H., S.C.-K.)
| | - Nicolas Raymond
- INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France (S.J.D., G.B.-M., A.C., M.Q., C.R.-M, F.A., M.K.N., B.R., E.G., M.-C.V., M.V., N.R., P.D., E.F., F.P., M.H., S.C.-K.)
- University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France (S.J.D., G.B.-M., A.C., M.Q., C.R.-M, F.A., M.K.N., B.R., E.G., M.-C.V., M.V., N.R., P.D., E.F., F.P., M.H., S.C.-K.)
| | - Peter Dorfmüller
- INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France (S.J.D., G.B.-M., A.C., M.Q., C.R.-M, F.A., M.K.N., B.R., E.G., M.-C.V., M.V., N.R., P.D., E.F., F.P., M.H., S.C.-K.)
- University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France (S.J.D., G.B.-M., A.C., M.Q., C.R.-M, F.A., M.K.N., B.R., E.G., M.-C.V., M.V., N.R., P.D., E.F., F.P., M.H., S.C.-K.)
| | - Elie Fadel
- INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France (S.J.D., G.B.-M., A.C., M.Q., C.R.-M, F.A., M.K.N., B.R., E.G., M.-C.V., M.V., N.R., P.D., E.F., F.P., M.H., S.C.-K.)
- University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France (S.J.D., G.B.-M., A.C., M.Q., C.R.-M, F.A., M.K.N., B.R., E.G., M.-C.V., M.V., N.R., P.D., E.F., F.P., M.H., S.C.-K.)
| | - Frédéric Perros
- INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France (S.J.D., G.B.-M., A.C., M.Q., C.R.-M, F.A., M.K.N., B.R., E.G., M.-C.V., M.V., N.R., P.D., E.F., F.P., M.H., S.C.-K.)
- University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France (S.J.D., G.B.-M., A.C., M.Q., C.R.-M, F.A., M.K.N., B.R., E.G., M.-C.V., M.V., N.R., P.D., E.F., F.P., M.H., S.C.-K.)
| | - Marc Humbert
- INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France (S.J.D., G.B.-M., A.C., M.Q., C.R.-M, F.A., M.K.N., B.R., E.G., M.-C.V., M.V., N.R., P.D., E.F., F.P., M.H., S.C.-K.)
- University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France (S.J.D., G.B.-M., A.C., M.Q., C.R.-M, F.A., M.K.N., B.R., E.G., M.-C.V., M.V., N.R., P.D., E.F., F.P., M.H., S.C.-K.)
- AP-HP Assistance Publique-Hôpitaux de Paris, Service de Pneumologie, Hôpital Bicêtre, Le Kremlin-Bicêtre, France (M.H.)
| | - Sylvia Cohen-Kaminsky
- INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France (S.J.D., G.B.-M., A.C., M.Q., C.R.-M, F.A., M.K.N., B.R., E.G., M.-C.V., M.V., N.R., P.D., E.F., F.P., M.H., S.C.-K.).
- University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France (S.J.D., G.B.-M., A.C., M.Q., C.R.-M, F.A., M.K.N., B.R., E.G., M.-C.V., M.V., N.R., P.D., E.F., F.P., M.H., S.C.-K.)
| |
Collapse
|
32
|
Abstract
Chronic pulmonary hypertension (PH) is associated with right ventricular failure and high mortality regardless of the underlying disease. Currently, therapies can improve clinical outcomes in specific subsets of patients, but have little impact on the progression of pulmonary vascular remodeling. Upon new advances in vector development and delivery techniques, gene therapy is a novel strategy in this field with the potential of overcoming the main limitations of approved drug therapies: modulation of novel anti-remodeling targets and selective pulmonary vasculature targeting with minimal systemic effects. In the recent years, several reports have shown that gene transfer to the pulmonary vascular system is feasible in rodent models of PH. Our group has focused on the translation of airway delivery of viral vectors in small and large animals. Here, we describe a procedure to achieve vector transduction at the distal vasculature in animal models of PH and the methods to evaluate the outcomes of this intervention as a promising new approach in pulmonary vascular diseases.
Collapse
|
33
|
Image-based computational assessment of vascular wall mechanics and hemodynamics in pulmonary arterial hypertension patients. J Biomech 2017; 68:84-92. [PMID: 29310945 DOI: 10.1016/j.jbiomech.2017.12.022] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 11/30/2017] [Accepted: 12/17/2017] [Indexed: 11/20/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a disease characterized by an elevated pulmonary arterial (PA) pressure. While several computational hemodynamic models of the pulmonary vasculature have been developed to understand PAH, they are lacking in some aspects, such as the vessel wall deformation and its lack of calibration against measurements in humans. Here, we describe a computational modeling framework that addresses these limitations. Specifically, computational models describing the coupling of hemodynamics and vessel wall mechanics in the pulmonary vasculature of a PAH patient and a normal subject were developed. Model parameters, consisting of linearized stiffness E of the large vessels and Windkessel parameters for each outflow branch, were calibrated against in vivo measurements of pressure, flow and vessel wall deformation obtained, respectively, from right-heart catheterization, phase-contrast and cine magnetic resonance images. Calibrated stiffness E of the proximal PA was 2.0 and 0.5 MPa for the PAH and normal models, respectively. Calibrated total compliance CT and resistance RT of the distal vessels were, respectively, 0.32 ml/mmHg and 11.3 mmHg∗min/l for the PAH model, and 2.93 ml/mmHg and 2.6 mmHg∗min/l for the normal model. These results were consistent with previous findings that the pulmonary vasculature is stiffer with more constricted distal vessels in PAH patients. Individual effects on PA pressure due to remodeling of the distal and proximal compartments of the pulmonary vasculature were also investigated in a sensitivity analysis. The analysis suggests that the remodeling of distal vasculature contributes more to the increase in PA pressure than the remodeling of proximal vasculature.
Collapse
|
34
|
Schäfer M, Barker AJ, Kheyfets V, Stenmark KR, Crapo J, Yeager ME, Truong U, Buckner JK, Fenster BE, Hunter KS. Helicity and Vorticity of Pulmonary Arterial Flow in Patients With Pulmonary Hypertension: Quantitative Analysis of Flow Formations. J Am Heart Assoc 2017; 6:JAHA.117.007010. [PMID: 29263034 PMCID: PMC5779020 DOI: 10.1161/jaha.117.007010] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Background Qualitative and quantitative flow hemodynamic indexes have been shown to reflect right ventricular (RV) afterload and function in pulmonary hypertension (PH). We aimed to quantify flow hemodynamic formations in pulmonary arteries using 4‐dimensional flow cardiac magnetic resonance imaging and the spatial velocity derivatives helicity and vorticity in a heterogeneous PH population. Methods and Results Patients with PH (n=35) and controls (n=10) underwent 4‐dimensional flow magnetic resonance imaging study for computation of helicity and vorticity in the main pulmonary artery (MPA), the right pulmonary artery, and the RV outflow tract. Helicity and vorticity were correlated with standard RV volumetric and functional indexes along with MPA stiffness assessed by measuring relative area change. Patients with PH had a significantly decreased helicity in the MPA (8 versus 32 m/s2; P<0.001), the right pulmonary artery (24 versus 50 m/s2; P<0.001), and the RV outflow tract–MPA unit (15 versus 42 m/s2; P<0.001). Vorticity was significantly decreased in patients with PH only in the right pulmonary artery (26 versus 45 1/s; P<0.001). Total helicity computed correlated with the cardiac magnetic resonance imaging–derived ventricular‐vascular coupling (−0.927; P<0.000), the RV ejection fraction (0.865; P<0.0001), cardiac output (0.581; P<0.0001), mean pulmonary arterial pressure (−0.581; P=0.0008), and relative area change measured at the MPA (0.789; P<0.0001). Conclusions The flow hemodynamic character in patients with PH assessed via quantitative analysis is considerably different when compared with healthy and normotensive controls. A strong association between helicity in pulmonary arteries and ventricular‐vascular coupling suggests a relationship between the mechanical and flow hemodynamic domains.
Collapse
Affiliation(s)
- Michal Schäfer
- Division of Cardiology, National Jewish Health, Denver, CO .,Division of Cardiology, Children's Hospital Colorado, Aurora, CO.,Department of Bioengineering, University of Colorado Denver
- Anschutz Medical Campus, Denver, CO
| | - Alex J Barker
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Vitaly Kheyfets
- Department of Bioengineering, University of Colorado Denver
- Anschutz Medical Campus, Denver, CO
| | - Kurt R Stenmark
- Department of Bioengineering, University of Colorado Denver
- Anschutz Medical Campus, Denver, CO.,Pediatric Division, Department of Critical Care and Pulmonary Medicine, University of Colorado Denver
- Anschutz Medical Campus, Denver, CO
| | - James Crapo
- Division of Pulmonary Medicine, National Jewish Health, Denver, CO
| | - Michael E Yeager
- Department of Bioengineering, University of Colorado Denver
- Anschutz Medical Campus, Denver, CO
| | - Uyen Truong
- Division of Cardiology, National Jewish Health, Denver, CO.,Department of Bioengineering, University of Colorado Denver
- Anschutz Medical Campus, Denver, CO
| | - J Kern Buckner
- Division of Cardiology, National Jewish Health, Denver, CO
| | | | - Kendall S Hunter
- Division of Cardiology, National Jewish Health, Denver, CO.,Department of Bioengineering, University of Colorado Denver
- Anschutz Medical Campus, Denver, CO
| |
Collapse
|
35
|
Yildirim E, Celik M, Yuksel UC, Gungor M, Bugan B, Dogan D, Gokoglan Y, Kabul HK, Gormel S, Yasar S, Koklu M, Barcin C. Relationship between Pulmonary Artery Stiffness and Functional Capacity in Patients with Heart Failure with Reduced Ejection Fraction. Korean Circ J 2017; 47:929-938. [PMID: 29171209 PMCID: PMC5711685 DOI: 10.4070/kcj.2017.0081] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Revised: 06/07/2017] [Accepted: 06/21/2017] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Functional capacity varies significantly among patients with heart failure with reduced ejection fraction (HFrEF), and it remains unclear why functional capacity is severely compromised in some patients with HFrEF while it is preserved in others. In this study, we aimed to evaluate the role of pulmonary artery stiffness (PAS) in the functional status of patients with HFrEF. METHODS A total of 46 heart failure (HF) patients without overt pulmonary hypertension or right HF and 52 controls were enrolled in the study. PAS was assessed on parasternal short-axis view using pulsed-wave Doppler recording of pulmonary flow one centimeter distal to the pulmonic valve annulus at a speed of 100 mm/sec. PAS was calculated according to the following formula: the ratio of maximum flow velocity shift of pulmonary flow to pulmonary acceleration time. RESULTS PAS was significantly increased in the HFrEF group compared to the control group (10.53±2.40 vs. 7.41±1.32, p<0.001). In sub-group analysis of patients with HFrEF, PAS was significantly associated with the functional class of the patients. HFrEF patients with poor New York Heart Association (NYHA) functional capacity had higher PAS compared those with good functional capacity. In multivariate regression analysis, NYHA class was independently correlated with PAS. CONCLUSION PAS is associated with functional status and should be taken into consideration as an underlying pathophysiological mechanism of dyspnea in patients with HFrEF.
Collapse
Affiliation(s)
- Erkan Yildirim
- Department of Cardiology, Gulhane Training and Research Hospital, Ankara, Turkey.
| | - Murat Celik
- Department of Cardiology, Gulhane Training and Research Hospital, Ankara, Turkey
| | - Uygar Cagdas Yuksel
- Department of Cardiology, Gulhane Training and Research Hospital, Ankara, Turkey
| | - Mutlu Gungor
- Department of Cardiology, Memorial Sisli Hospital, Istanbul, Turkey
| | - Baris Bugan
- Department of Cardiology, Corlu State Hospital, Tekirdag, Turkey
| | - Deniz Dogan
- Department of Thoracic Medicine, Gulhane Training and Research Hospital, Ankara, Turkey
| | - Yalcin Gokoglan
- Department of Cardiology, Gulhane Training and Research Hospital, Ankara, Turkey
| | - Hasan Kutsi Kabul
- Department of Cardiology, Gulhane Training and Research Hospital, Ankara, Turkey
| | - Suat Gormel
- Department of Cardiology, Gulhane Training and Research Hospital, Ankara, Turkey
| | - Salim Yasar
- Department of Cardiology, Gulhane Training and Research Hospital, Ankara, Turkey
| | - Mustafa Koklu
- Department of Cardiology, Gulhane Training and Research Hospital, Ankara, Turkey
| | - Cem Barcin
- Department of Cardiology, Gulhane Training and Research Hospital, Ankara, Turkey
| |
Collapse
|
36
|
Frump AL, Bonnet S, de Jesus Perez VA, Lahm T. Emerging role of angiogenesis in adaptive and maladaptive right ventricular remodeling in pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2017; 314:L443-L460. [PMID: 29097426 DOI: 10.1152/ajplung.00374.2017] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Right ventricular (RV) function is the primary prognostic factor for both morbidity and mortality in pulmonary hypertension (PH). RV hypertrophy is initially an adaptive physiological response to increased overload; however, with persistent and/or progressive afterload increase, this response frequently transitions to more pathological maladaptive remodeling. The mechanisms and disease processes underlying this transition are mostly unknown. Angiogenesis has recently emerged as a major modifier of RV adaptation in the setting of pressure overload. A novel paradigm has emerged that suggests that angiogenesis and angiogenic signaling are required for RV adaptation to afterload increases and that impaired and/or insufficient angiogenesis is a major driver of RV decompensation. Here, we summarize our current understanding of the concepts of maladaptive and adaptive RV remodeling, discuss the current literature on angiogenesis in the adapted and failing RV, and identify potential therapeutic approaches targeting angiogenesis in RV failure.
Collapse
Affiliation(s)
- Andrea L Frump
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University School of Medicine , Indianapolis, Indiana
| | - Sébastien Bonnet
- Pulmonary Hypertension Research Group, Institut Universitaire de Cardiologie et de Pneumologie de Québec Research Center, Laval University , Quebec City, Quebec , Canada
| | - Vinicio A de Jesus Perez
- Division of Pulmonary/Critical Care, Stanford University School of Medicine , Stanford, California.,Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University School of Medicine , Stanford, California
| | - Tim Lahm
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University School of Medicine , Indianapolis, Indiana.,Richard L. Roudebush Veterans Affairs Medical Center , Indianapolis, Indiana.,Department of Cellular and Integrative Physiology, Indiana University School of Medicine , Indianapolis, Indiana
| |
Collapse
|
37
|
Michelakis ED. PVDOMICS Drive the Pulmonary Hypertension Field Into the Precision Medicine Era. Circ Res 2017; 121:1106-1108. [PMID: 29074523 DOI: 10.1161/circresaha.117.312022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
38
|
Kitagawa MG, Reynolds JO, Wehrens XHT, Bryan RM, Pandit LM. Hemodynamic and Pathologic Characterization of the TASK-1 -/- Mouse Does Not Demonstrate Pulmonary Hypertension. Front Med (Lausanne) 2017; 4:177. [PMID: 29109948 PMCID: PMC5660113 DOI: 10.3389/fmed.2017.00177] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 10/02/2017] [Indexed: 01/22/2023] Open
Abstract
Introduction Pulmonary hypertension (PH) carries significant associated morbidity and mortality and the underlying molecular mechanisms of PH are not well understood. Loss-of-function mutations in TASK-1 potassium channels are associated with PH in humans. Although TASK-1 has been considered in the development of PH for over a decade, characterization of TASK-1 knockout mice has been limited to in vitro studies or in vivo studies in room air at isolated time points. The purpose of this study was twofold. First, we sought to determine if TASK-/- male and female mice developed PH over the span of one year. Second, we sought to determine the effect of chronic hypoxia, a stimulus for PH, and its recovery on PH in TASK-1-/- mice. Methods We measured right ventricular systolic pressure (RVSP) and vascular remodeling in male and female C57BL/6 WT and TASK-1-/- mice at separate time points: 20-24 weeks and 1 year of age. Additionally, we measured RVSP and vascular remodeling in TASK-1-/- and wild-type mice between 13 and 16 weeks of age exposed to 10% hypoxia for 3 weeks followed by recovery to room air conditions for an additional 6 weeks. Results RVSP was similar between WT and TASK-/- mice. Male and female WT and TASK-1-/- mice all demonstrated age-related increases in RVSP, which correlated to age-related vascular remodeling in male mice but not in female mice. Male TASK-1-/- and WT mice exposed to chronic hypoxia demonstrated increased RVSP, which decreased following room air recovery. WT and TASK-1-/- male mice demonstrated vascular remodeling upon exposure to hypoxia that persisted in room air recovery. Conclusion Female and male TASK-1-/- mice do not develop hemodynamic or vascular evidence for PH, but RVSP rises in an age-dependent manner independent of genotype. TASK-1-/- and WT male mice develop hypoxia-induced elevations in RVSP that decrease to baseline after recovery in room air. TASK-1-/- and WT male mice demonstrate vascular remodeling after exposure to hypoxia that persists despite recovery to room air conditions and does not correlate with RVSP normalization.
Collapse
Affiliation(s)
- Melanie G Kitagawa
- Baylor College of Medicine, Texas Children's Hospital, Houston, TX, United States
| | - Julia O Reynolds
- Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, United States.,Baylor College of Medicine, Houston, TX, United States
| | | | | | - Lavannya M Pandit
- Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, United States.,Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
39
|
Ghataorhe P, Rhodes CJ, Harbaum L, Attard M, Wharton J, Wilkins MR. Pulmonary arterial hypertension - progress in understanding the disease and prioritizing strategies for drug development. J Intern Med 2017; 282:129-141. [PMID: 28524624 DOI: 10.1111/joim.12623] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Pulmonary arterial hypertension (PAH), at one time a largely overlooked disease, is now the subject of intense study in many academic and biotech groups. The availability of new treatments has increased awareness of the condition. This in turn has driven a change in the demographics of PAH, with an increase in the mean age at diagnosis. The diagnosis of PAH in more elderly patients has highlighted the need for careful phenotyping of patients and for further studies to understand how best to manage pulmonary hypertension associated with, for example, left heart disease. The breadth and depth of expertise focused on unravelling the molecular pathology of PAH has yielded novel insights, including the role of growth factors, inflammation and metabolic remodelling. The description of the genetic architecture of PAH is accelerating in parallel, with novel variants, such as those reported in potassium two-pore domain channel subfamily K member 3 (KCNK3), adding to the list of more established mutations in genes associated with bone morphogenetic protein receptor type 2 (BMPR2) signalling. These insights have supported a paradigm shift in treatment strategies away from simply addressing the imbalance of vasoactive mediators observed in PAH towards tackling more directly the structural remodelling of the pulmonary vasculature. Here, we summarize the changing clinical and molecular landscape of PAH. We highlight novel drug therapies that are in various stages of clinical development, targeting for example cell proliferation, metabolic, inflammatory/immune and BMPR2 dysfunction, and the challenges around developing these treatments. We argue that advances in the treatment of PAH will come through deep molecular phenotyping with the integration of clinical, genomic, transcriptomic, proteomic and metabolomic information in large populations of patients through international collaboration. This approach provides the best opportunity for identifying key signalling pathways, both as potential drug targets and as biomarkers for patient selection. The expectation is that together these will enable the prioritization of potential therapies in development and the evolution of personalized medicine for PAH.
Collapse
Affiliation(s)
- P Ghataorhe
- Department of Medicine, Imperial College London, London, UK
| | - C J Rhodes
- Department of Medicine, Imperial College London, London, UK
| | - L Harbaum
- Department of Medicine, Imperial College London, London, UK
| | - M Attard
- Department of Medicine, Imperial College London, London, UK
| | - J Wharton
- Department of Medicine, Imperial College London, London, UK
| | - M R Wilkins
- Department of Medicine, Imperial College London, London, UK
| |
Collapse
|
40
|
Maarman GJ. Editorial commentary: "Discovery of a Murine Model of clinical pulmonary arterial hypertension: Mission impossible?" by Dai and Zhao. Trends Cardiovasc Med 2017; 27:237-238. [PMID: 28189476 DOI: 10.1016/j.tcm.2017.01.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Accepted: 01/08/2017] [Indexed: 01/07/2023]
Affiliation(s)
- Gerald J Maarman
- Hatter Institute for Cardiovascular Disease in Africa (HICRA), Department of Medicine, Faculty of Health Sciences, University of Cape Town, Chris Barnard Building, Anzio Road, Observatory 7925, South Africa; Division of Exercise Science & Sports Medicine (ESSM), Department of Medicine, Department of Human Biology, Boundary Road, Newlands, University of Cape Town, South Africa.
| |
Collapse
|
41
|
Role of dynorphin in hypoxic pulmonary hypertension. Eur J Pharmacol 2016; 791:78-84. [DOI: 10.1016/j.ejphar.2016.08.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 08/23/2016] [Accepted: 08/24/2016] [Indexed: 11/21/2022]
|
42
|
Pulmonary Arterial Stiffness: Toward a New Paradigm in Pulmonary Arterial Hypertension Pathophysiology and Assessment. Curr Hypertens Rep 2016; 18:4. [PMID: 26733189 DOI: 10.1007/s11906-015-0609-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Stiffening of the pulmonary arterial bed with the subsequent increased load on the right ventricle is a paramount feature of pulmonary hypertension (PH). The pathophysiology of vascular stiffening is a complex and self-reinforcing function of extracellular matrix remodeling, driven by recruitment of circulating inflammatory cells and their interactions with resident vascular cells, and mechanotransduction of altered hemodynamic forces throughout the ventricular-vascular axis. New approaches to understanding the cell and molecular determinants of the pathophysiology combine novel biopolymer substrates, controlled flow conditions, and defined cell types to recapitulate the biomechanical environment in vitro. Simultaneously, advances are occurring to assess novel parameters of stiffness in vivo. In this comprehensive state-of-art review, we describe clinical hemodynamic markers, together with the newest translational echocardiographic and cardiac magnetic resonance imaging methods, to assess vascular stiffness and ventricular-vascular coupling. Finally, fluid-tissue interactions appear to offer a novel route of investigating the mechanotransduction processes and disease progression.
Collapse
|
43
|
Aguero J, Ishikawa K, Hadri L, Santos-Gallego CG, Fish KM, Kohlbrenner E, Hammoudi N, Kho C, Lee A, Ibáñez B, García-Alvarez A, Zsebo K, Maron BA, Plataki M, Fuster V, Leopold JA, Hajjar RJ. Intratracheal Gene Delivery of SERCA2a Ameliorates Chronic Post-Capillary Pulmonary Hypertension: A Large Animal Model. J Am Coll Cardiol 2016; 67:2032-46. [PMID: 27126531 PMCID: PMC6019268 DOI: 10.1016/j.jacc.2016.02.049] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 02/16/2016] [Accepted: 02/17/2016] [Indexed: 01/16/2023]
Abstract
BACKGROUND Pulmonary hypertension (PH) is characterized by pulmonary arterial remodeling that results in increased pulmonary vascular resistance, right ventricular (RV) failure, and premature death. Down-regulation of sarcoplasmic reticulum Ca(2+)-ATPase 2a (SERCA2a) in the pulmonary vasculature leads to perturbations in calcium ion (Ca(2+)) homeostasis and transition of pulmonary artery smooth muscle cells to a proliferative phenotype. OBJECTIVES We assessed the feasibility of sustained pulmonary vascular SERCA2a gene expression using aerosolized delivery of adeno-associated virus type 1 (AAV1) in a large animal model of chronic PH and evaluated the efficacy of gene transfer regarding progression of pulmonary vascular and RV remodeling. METHODS A model of chronic post-capillary PH was created in Yorkshire swine by partial pulmonary vein banding. Development of chronic PH was confirmed hemodynamically, and animals were randomized to intratracheal administration of aerosolized AAV1 carrying the human SERCA2a gene (n = 10, AAV1.SERCA2a group) or saline (n = 10). Therapeutic efficacy was evaluated 2 months after gene delivery. RESULTS Transduction efficacy after intratracheal delivery of AAV1 was confirmed by β-galactosidase detection in the distal pulmonary vasculature. Treatment with aerosolized AAV1.SERCA2a prevented disease progression as evaluated by mean pulmonary artery pressure, vascular resistance, and limited vascular remodeling quantified by histology. Therapeutic efficacy was supported further by the preservation of RV ejection fraction (p = 0.014) and improvement of the RV end-diastolic pressure-volume relationship in PH pigs treated with aerosolized AAV1.SERCA2a. CONCLUSIONS Airway-based delivery of AAV vectors to the pulmonary arteries was feasible, efficient, and safe in a clinically relevant chronic PH model. Vascular SERCA2a overexpression resulted in beneficial effects on pulmonary arterial remodeling, with attendant improvements in pulmonary hemodynamics and RV performance, and might offer therapeutic benefit by modifying fundamental pathophysiology in pulmonary vascular diseases.
Collapse
Affiliation(s)
- Jaume Aguero
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York; Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Kiyotake Ishikawa
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Lahouaria Hadri
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Carlos G Santos-Gallego
- Atherothrombosis Research Unit, Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Kenneth M Fish
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Erik Kohlbrenner
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Nadjib Hammoudi
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Changwon Kho
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ahyoung Lee
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Borja Ibáñez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; IIS Fundacion Jimenez-Diaz Hospital, Madrid, Spain
| | - Ana García-Alvarez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | | | - Bradley A Maron
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Maria Plataki
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Valentin Fuster
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Zena and Michael A. Wiener Cardiovascular Institute, Mount Sinai School of Medicine, New York, New York
| | - Jane A Leopold
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Roger J Hajjar
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
44
|
Dai Z, Li M, Wharton J, Zhu MM, Zhao YY. Prolyl-4 Hydroxylase 2 (PHD2) Deficiency in Endothelial Cells and Hematopoietic Cells Induces Obliterative Vascular Remodeling and Severe Pulmonary Arterial Hypertension in Mice and Humans Through Hypoxia-Inducible Factor-2α. Circulation 2016; 133:2447-58. [PMID: 27143681 DOI: 10.1161/circulationaha.116.021494] [Citation(s) in RCA: 185] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 04/19/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND Vascular occlusion and complex plexiform lesions are hallmarks of the pathology of severe pulmonary arterial hypertension (PAH) in patients. However, the mechanisms of obliterative vascular remodeling remain elusive; hence, current therapies have not targeted the fundamental disease-modifying mechanisms and result in only modest improvement in morbidity and mortality. METHODS AND RESULTS Mice with Tie2Cre-mediated disruption of Egln1 (encoding prolyl-4 hydroxylase 2 [PHD2]; Egln1(Tie2)) in endothelial cells and hematopoietic cells exhibited spontaneous severe PAH with extensive pulmonary vascular remodeling, including vascular occlusion and plexiform-like lesions, resembling the hallmarks of the pathology of clinical PAH. As seen in patients with idiopathic PAH, Egln1(Tie2) mice exhibited unprecedented right ventricular hypertrophy and failure and progressive mortality. Consistently, PHD2 expression was diminished in lung endothelial cells of obliterated pulmonary vessels in patients with idiopathic PAH. Genetic deletions of both Egln1 and Hif1a or Egln1 and Hif2a identified hypoxia-inducible factor-2α as the critical mediator of the severe PAH seen in Egln1(Tie2) mice. We also observed altered expression of many pulmonary hypertension-causing genes in Egln1(Tie2) lungs, which was normalized in Egln1(Tie2)/Hif2a(Tie2) lungs. PHD2-deficient endothelial cells promoted smooth muscle cell proliferation in part through hypoxia-inducible factor-2α-activated CXCL12 expression. Genetic deletion of Cxcl12 attenuated PAH in Egln1(Tie2) mice. CONCLUSIONS These studies defined an unexpected role of PHD2 deficiency in the mechanisms of severe PAH and identified the first genetically modified mouse model with obliterative vascular remodeling and pathophysiology recapitulating clinical PAH. Thus, targeting PHD2/hypoxia-inducible factor-2α signaling is a promising strategy to reverse vascular remodeling for treatment of severe PAH.
Collapse
Affiliation(s)
- Zhiyu Dai
- From Department of Pharmacology (Z.D., M.L., M.M.Z., Y.-Y.Z.) and Center for Lung and Vascular Biology (Z.D., M.L., M.M.Z., Y.-Y.Z.), University of Illinois College of Medicine, Chicago; and Centre for Pharmacology and Therapeutics, Department of Medicine, Imperial College of London, Hammersmith Hospital, UK (J.W.)
| | - Ming Li
- From Department of Pharmacology (Z.D., M.L., M.M.Z., Y.-Y.Z.) and Center for Lung and Vascular Biology (Z.D., M.L., M.M.Z., Y.-Y.Z.), University of Illinois College of Medicine, Chicago; and Centre for Pharmacology and Therapeutics, Department of Medicine, Imperial College of London, Hammersmith Hospital, UK (J.W.)
| | - John Wharton
- From Department of Pharmacology (Z.D., M.L., M.M.Z., Y.-Y.Z.) and Center for Lung and Vascular Biology (Z.D., M.L., M.M.Z., Y.-Y.Z.), University of Illinois College of Medicine, Chicago; and Centre for Pharmacology and Therapeutics, Department of Medicine, Imperial College of London, Hammersmith Hospital, UK (J.W.)
| | - Maggie M Zhu
- From Department of Pharmacology (Z.D., M.L., M.M.Z., Y.-Y.Z.) and Center for Lung and Vascular Biology (Z.D., M.L., M.M.Z., Y.-Y.Z.), University of Illinois College of Medicine, Chicago; and Centre for Pharmacology and Therapeutics, Department of Medicine, Imperial College of London, Hammersmith Hospital, UK (J.W.)
| | - You-Yang Zhao
- From Department of Pharmacology (Z.D., M.L., M.M.Z., Y.-Y.Z.) and Center for Lung and Vascular Biology (Z.D., M.L., M.M.Z., Y.-Y.Z.), University of Illinois College of Medicine, Chicago; and Centre for Pharmacology and Therapeutics, Department of Medicine, Imperial College of London, Hammersmith Hospital, UK (J.W.).
| |
Collapse
|
45
|
Abe K, Shinoda M, Tanaka M, Kuwabara Y, Yoshida K, Hirooka Y, McMurtry IF, Oka M, Sunagawa K. Haemodynamic unloading reverses occlusive vascular lesions in severe pulmonary hypertension. Cardiovasc Res 2016; 111:16-25. [PMID: 27037259 DOI: 10.1093/cvr/cvw070] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 03/17/2016] [Indexed: 11/13/2022] Open
Abstract
AIMS An important pathogenic mechanism in the development of idiopathic pulmonary arterial hypertension is hypothesized to be a cancer-like cellular proliferation independent of haemodynamics. However, because the vascular lesions are inseparably coupled with haemodynamic stress, the fate of the lesions is unknown when haemodynamic stress is eliminated. METHODS AND RESULTS We applied left pulmonary artery banding to a rat model with advanced pulmonary hypertension to investigate the effects of decreased haemodynamic stress on occlusive vascular lesions. Rats were given an injection of the VEGF blocker Sugen5416 and exposed to 3 weeks of hypoxia plus an additional 7 weeks of normoxia (total 10 weeks) (SU/Hx/Nx rats). The banding surgery to reduce haemodynamic stress to the left lung was done at 1 week prior to (preventive) or 5 weeks after (reversal) the SU5416 injection. All SU/Hx/Nx-exposed rats developed severe pulmonary hypertension and right ventricular hypertrophy. Histological analyses showed that the non-banded right lungs developed occlusive lesions including plexiform lesions with marked perivascular cell accumulation. In contrast, banding the left pulmonary artery not only prevented the development of but also reversed the established occlusive lesions as well as perivascular inflammation in the left lungs. CONCLUSION Our results indicate that haemodynamic stress is prerequisite to the development and progression of occlusive neointimal lesions in this rat model of severe pulmonary hypertension. We conclude that perivascular inflammation and occlusive neointimal arteriopathy are driven by haemodynamic stress.
Collapse
Affiliation(s)
- Kohtaro Abe
- Department of Advanced Cardiovascular Regulation and Therapeutics, Center for Disruptive Cardiovascular Medicine, Kyushu University, Fukuoka 811-1347, Japan
| | - Masako Shinoda
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medicine, Fukuoka 812-8582, Japan
| | - Mariko Tanaka
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medicine, Fukuoka 812-8582, Japan Division of Anesthesiology and Critical Care Medicine, Kyushu University Graduate School of Medicine, Fukuoka 812-8582, Japan
| | - Yukimitsu Kuwabara
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medicine, Fukuoka 812-8582, Japan
| | - Keimei Yoshida
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medicine, Fukuoka 812-8582, Japan
| | - Yoshitaka Hirooka
- Department of Advanced Cardiovascular Regulation and Therapeutics, Center for Disruptive Cardiovascular Medicine, Kyushu University, Fukuoka 811-1347, Japan
| | - Ivan F McMurtry
- Department of Pharmacology, University of South Alabama Mobile, 307 N University Blvd #130, Mobile, AL 36608, USA Department of Internal Medicine, University of South Alabama Mobile, 307 N University Blvd #130, Mobile, AL 36608, USA Center for Lung Biology, University of South Alabama Mobile, 307 N University Blvd #130, Mobile, AL 36608, USA
| | - Masahiko Oka
- Department of Pharmacology, University of South Alabama Mobile, 307 N University Blvd #130, Mobile, AL 36608, USA Department of Internal Medicine, University of South Alabama Mobile, 307 N University Blvd #130, Mobile, AL 36608, USA Center for Lung Biology, University of South Alabama Mobile, 307 N University Blvd #130, Mobile, AL 36608, USA
| | - Kenji Sunagawa
- Therapeutic Regulation of Cardiovascular Homeostasis, Center for Disruptive Cardiovascular Medicine, Kyushu University, Fukuoka 811-1347, Japan
| |
Collapse
|
46
|
Shi L, Kojonazarov B, Elgheznawy A, Popp R, Dahal BK, Böhm M, Pullamsetti SS, Ghofrani HA, Gödecke A, Jungmann A, Katus HA, Müller OJ, Schermuly RT, Fisslthaler B, Seeger W, Fleming I. miR-223-IGF-IR signalling in hypoxia- and load-induced right-ventricular failure: a novel therapeutic approach. Cardiovasc Res 2016; 111:184-93. [PMID: 27013635 DOI: 10.1093/cvr/cvw065] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 03/18/2016] [Indexed: 12/11/2022] Open
Abstract
AIMS Pulmonary hypertension is a progressive disease with poor prognosis, characterized by pathological inward remodelling and loss of patency of the lung vasculature. The right ventricle is co-affected by pulmonary hypertension, which triggers events such as hypoxia and/or increased mechanical load. Initially the right ventricle responds with 'adaptive' hypertrophy, which is often rapidly followed by 'maladaptive' changes leading to right heart decompensation and failure, which is the ultimate cause of death. METHODS AND RESULTS We report here that miR-223 is expressed in the murine lung and right ventricle at higher levels than in the left ventricle. Moreover, lung and right-ventricular miR-223 levels were markedly down-regulated by hypoxia. Correspondingly, increasing right-ventricular load by pulmonary artery banding, induced right-ventricular ischaemia, and the down-regulation of miR-223. Lung and right ventricle miR-223 down-regulation were linked with increased expression of the miR-223 target; insulin-like growth factor-I receptor (IGF-IR) and IGF-I downstream signalling. Similarly, miR-223 was decreased and IGF-IR increased in human pulmonary hypertension. Notably in young mice, miR-223 overexpression, the genetic inactivation or pharmacological inhibition of IGF-IR, all attenuated right-ventricular hypertrophy and improved right heart function under conditions of hypoxia or increased afterload. CONCLUSION These findings highlight the early role of pulmonary and right-ventricular miR-223 and the IGF-IR in the right heart failure programme initiated by pulmonary hypoxia and increased mechanical load and may lead to the development of novel therapeutic strategies that target the development of PH and right heart failure.
Collapse
Affiliation(s)
- Lei Shi
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe-University, Frankfurt am Main, and German Center of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt am Main, Germany
| | - Baktybek Kojonazarov
- University of Giessen and Marburg Lung Center, Justus-Liebig-University Giessen and German Center for Lung Research (DZL), Germany
| | - Amro Elgheznawy
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe-University, Frankfurt am Main, and German Center of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt am Main, Germany
| | - Rüdiger Popp
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe-University, Frankfurt am Main, and German Center of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt am Main, Germany
| | - Bhola Kumar Dahal
- University of Giessen and Marburg Lung Center, Justus-Liebig-University Giessen and German Center for Lung Research (DZL), Germany
| | - Mario Böhm
- University of Giessen and Marburg Lung Center, Justus-Liebig-University Giessen and German Center for Lung Research (DZL), Germany
| | - Soni Savai Pullamsetti
- University of Giessen and Marburg Lung Center, Justus-Liebig-University Giessen and German Center for Lung Research (DZL), Germany Max-Planck-Institute for Heart and Lung Research, Department of Lung Development and Remodeling, Bad Nauheim, Germany
| | - Hossein-Ardeschir Ghofrani
- University of Giessen and Marburg Lung Center, Justus-Liebig-University Giessen and German Center for Lung Research (DZL), Germany
| | - Axel Gödecke
- Institut für Herz- und Kreislaufphysiologie, Universitätsklinikum, Heinrich-Heine-Universität, Dusseldorf, Germany
| | - Andreas Jungmann
- Department of Internal Medicine III, University of Heidelberg, Im Neuenheimer Feld 410, Heidelberg 69120, Germany German Center for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim, Germany
| | - Hugo A Katus
- Department of Internal Medicine III, University of Heidelberg, Im Neuenheimer Feld 410, Heidelberg 69120, Germany German Center for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim, Germany
| | - Oliver J Müller
- Department of Internal Medicine III, University of Heidelberg, Im Neuenheimer Feld 410, Heidelberg 69120, Germany German Center for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim, Germany
| | - Ralph T Schermuly
- University of Giessen and Marburg Lung Center, Justus-Liebig-University Giessen and German Center for Lung Research (DZL), Germany
| | - Beate Fisslthaler
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe-University, Frankfurt am Main, and German Center of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt am Main, Germany
| | - Werner Seeger
- University of Giessen and Marburg Lung Center, Justus-Liebig-University Giessen and German Center for Lung Research (DZL), Germany Max-Planck-Institute for Heart and Lung Research, Department of Lung Development and Remodeling, Bad Nauheim, Germany
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe-University, Frankfurt am Main, and German Center of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt am Main, Germany
| |
Collapse
|
47
|
Sung YK, Yuan K, de Jesus Perez VA. Novel approaches to pulmonary arterial hypertension drug discovery. Expert Opin Drug Discov 2016; 11:407-14. [PMID: 26901465 PMCID: PMC4933595 DOI: 10.1517/17460441.2016.1153625] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Pulmonary arterial hypertension (PAH) is a rare disorder associated with abnormally elevated pulmonary pressures that, if untreated, leads to right heart failure and premature death. The goal of drug development for PAH is to develop effective therapies that halt, or ideally, reverse the obliterative vasculopathy that results in vessel loss and obstruction of blood flow to the lungs. AREAS COVERED This review summarizes the current approach to candidate discovery in PAH and discusses the currently available drug discovery methods that should be implemented to prioritize targets and obtain a comprehensive pharmacological profile of promising compounds with well-defined mechanisms. EXPERT OPINION To improve the successful identification of leading drug candidates, it is necessary that traditional pre-clinical studies are combined with drug screening strategies that maximize the characterization of biological activity and identify relevant off-target effects that could hinder the clinical efficacy of the compound when tested in human subjects. A successful drug discovery strategy in PAH will require collaboration of clinician scientists with medicinal chemists and pharmacologists who can identify compounds with an adequate safety profile and biological activity against relevant disease mechanisms.
Collapse
Affiliation(s)
- Yon K. Sung
- Division of Pulmonary and Critical Care Medicine, The Vera Moulton Wall Center for Pulmonary Vascular Medicine, Stanford Cardiovascular Institute, Stanford, California
| | - Ke Yuan
- Division of Pulmonary and Critical Care Medicine, The Vera Moulton Wall Center for Pulmonary Vascular Medicine, Stanford Cardiovascular Institute, Stanford, California
| | - Vinicio A. de Jesus Perez
- Division of Pulmonary and Critical Care Medicine, The Vera Moulton Wall Center for Pulmonary Vascular Medicine, Stanford Cardiovascular Institute, Stanford, California
| |
Collapse
|
48
|
Calvier L, Legchenko E, Grimm L, Sallmon H, Hatch A, Plouffe BD, Schroeder C, Bauersachs J, Murthy SK, Hansmann G. Galectin-3 and aldosterone as potential tandem biomarkers in pulmonary arterial hypertension. Heart 2016; 102:390-6. [DOI: 10.1136/heartjnl-2015-308365] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
49
|
(1)H NMR-Based Analysis of Serum Metabolites in Monocrotaline-Induced Pulmonary Arterial Hypertensive Rats. DISEASE MARKERS 2016; 2016:5803031. [PMID: 27057080 PMCID: PMC4745193 DOI: 10.1155/2016/5803031] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 12/07/2015] [Indexed: 01/18/2023]
Abstract
AIMS To study the changes of the metabolic profile during the pathogenesis in monocrotaline (MCT) induced pulmonary arterial hypertension (PAH). METHODS Forty male Sprague-Dawley (SD) rats were randomly divided into 5 groups (n = 8, each). PAH rats were induced by a single dose intraperitoneal injection of 60 mg/kg MCT, while 8 rats given intraperitoneal injection of 1 ml normal saline and scarified in the same day (W0) served as control. Mean pulmonary arterial pressure (mPAP) was measured through catherization. The degree of right ventricular hypertrophy and pulmonary hyperplasia were determined at the end of first to fourth weeks; nuclear magnetic resonance (NMR) spectra of sera were then acquired for the analysis of metabolites. Principal component analysis (PCA) and orthogonal partial least-squares discriminant analysis (OPLS-DA) were used to discriminate different metabolic profiles. RESULTS The prominent changes of metabolic profiles were seen during these four weeks. Twenty specific metabolites were identified, which were mainly involved in lipid metabolism, glycolysis, energy metabolism, ketogenesis, and methionine metabolism. Profiles of correlation between these metabolites in each stage changed markedly, especially in the fourth week. Highly activated methionine and betaine metabolism pathways were selected by the pathway enrichment analysis. CONCLUSIONS Metabolic dysfunction is involved in the development and progression of PAH.
Collapse
|
50
|
Wang AP, Li XH, Gong SX, Li WQ, Hu CP, Zhang Z, Li YJ. miR-100 suppresses mTOR signaling in hypoxia-induced pulmonary hypertension in rats. Eur J Pharmacol 2015; 765:565-73. [DOI: 10.1016/j.ejphar.2015.09.031] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Revised: 09/15/2015] [Accepted: 09/21/2015] [Indexed: 12/31/2022]
|