1
|
Bao W, Cheng M, Chen X, Wang T, Xu D, Liao H, Chen L, Wen F, He J, Chen J. Effect of fasudil on clinical outcomes of pulmonary hypertension: a systematic review and meta-analysis. Expert Rev Clin Pharmacol 2024:1-18. [PMID: 39269366 DOI: 10.1080/17512433.2024.2404688] [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: 07/10/2024] [Revised: 09/06/2024] [Accepted: 09/11/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND Pulmonary hypertension (PH) is a life-threatening condition with high mortality, categorized into Group 1-5 by distinct etiologies. Fasudil, a potent vasodilator targeting RhoA/Rho kinase pathway, holds promise for diverse PH pathologies. However, a systematic evaluation of its clinical benefits remains elusive. METHODS We conducted a systematic search in several databases. Meta-analysis using odds ratio and mean difference was performed, with an assessment of studies' quality and pooled evidences. RESULTS Inclusion of 3269 Group-3 PH patients demonstrated that Fasudil increased effective events, forced expiratory volume in one second (FEV1), 6-minute walking distance (6MWD) and arterial partial pressure of oxygen (PaO2), and decreased mean pulmonary artery pressure (mPAP) and pulmonary artery systolic pressure (PASP); Inclusion of 197 Group-2 PH patients suggested that Fasudil increased 6MWD and PaO2, and decreased PASP. Subgroup analysis revealed no significant difference between 30 and 60 mg/day dosages of Fasudil, while administration durations and methods might affect its effectiveness in treating Group-3 PH patients. CONCLUSIONS Our study favors the beneficial effects of Fasudil by enhancing FEV1, 6MWD and PaO2, and reducing mPAP and PASP on Group-3 PH patients, suggesting Fasudil as a viable treatment option and highlighting the need for further studies to inform healthcare policies. PROTOCOL REGISTRATION www.crd.york.ac.uk/prospero identifier is CRD42022308947.
Collapse
Affiliation(s)
- Wanying Bao
- Department of Respiratory and Critical Care Medicine, West China Hospital, West China School of Medicine, and Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Obstetrics and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Mengxin Cheng
- Department of Respiratory and Critical Care Medicine, West China Hospital, West China School of Medicine, and Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Xiaoye Chen
- Department of Respiratory and Critical Care Medicine, West China Hospital, West China School of Medicine, and Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Wang
- Department of Respiratory and Critical Care Medicine, West China Hospital, West China School of Medicine, and Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Dan Xu
- Department of Respiratory and Critical Care Medicine, West China Hospital, West China School of Medicine, and Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Hualin Liao
- Department of Respiratory Medicine, Hospital of Chengdu office of People's Government of Tibetan Autonomous Region of China, Chengdu, Sichuan, China
| | - Lei Chen
- Department of Respiratory and Critical Care Medicine, West China Hospital, West China School of Medicine, and Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Fuqiang Wen
- Department of Respiratory and Critical Care Medicine, West China Hospital, West China School of Medicine, and Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Junyun He
- Department of Respiratory Medicine, Hospital of Chengdu office of People's Government of Tibetan Autonomous Region of China, Chengdu, Sichuan, China
| | - Jun Chen
- Department of Respiratory and Critical Care Medicine, West China Hospital, West China School of Medicine, and Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
2
|
MacLean MR, Fanburg B, Hill N, Lazarus HM, Pack TF, Palacios M, Penumatsa KC, Wring SA. Serotonin and Pulmonary Hypertension; Sex and Drugs and ROCK and Rho. Compr Physiol 2022; 12:4103-4118. [PMID: 36036567 DOI: 10.1002/cphy.c220004] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Serotonin is often referred to as a "happy hormone" as it maintains good mood, well-being, and happiness. It is involved in communication between nerve cells and plays a role in sleeping and digestion. However, too much serotonin can have pathogenic effects and serotonin synthesis is elevated in pulmonary artery endothelial cells from patients with pulmonary arterial hypertension (PAH). PAH is characterized by elevated pulmonary pressures, right ventricular failure, inflammation, and pulmonary vascular remodeling; serotonin has been shown to be associated with these pathologies. The rate-limiting enzyme in the synthesis of serotonin in the periphery of the body is tryptophan hydroxylase 1 (TPH1). TPH1 expression and serotonin synthesis are elevated in pulmonary artery endothelial cells in patients with PAH. The serotonin synthesized in the pulmonary arterial endothelium can act on the adjacent pulmonary arterial smooth muscle cells (PASMCs), adventitial macrophages, and fibroblasts, in a paracrine fashion. In humans, serotonin enters PASMCs cells via the serotonin transporter (SERT) and it can cooperate with the 5-HT1B receptor on the plasma membrane; this activates both contractile and proliferative signaling pathways. The "serotonin hypothesis of pulmonary hypertension" arose when serotonin was associated with PAH induced by diet pills such as fenfluramine, aminorex, and chlorphentermine; these act as indirect serotonergic agonists causing the release of serotonin from platelets and cells through the SERT. Here the role of serotonin in PAH is reviewed. Targeting serotonin synthesis or signaling is a promising novel alternative approach which may lead to novel therapies for PAH. © 2022 American Physiological Society. Compr Physiol 12: 1-16, 2022.
Collapse
Affiliation(s)
- Margaret R MacLean
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland
| | - Barry Fanburg
- Pulmonary, Critical Care and Sleep Division, Department of Medicine, Tufts Medical Center, Boston, Massachusetts, USA
| | - Nicolas Hill
- Pulmonary, Critical Care and Sleep Division, Department of Medicine, Tufts Medical Center, Boston, Massachusetts, USA
| | | | | | | | - Krishna C Penumatsa
- Pulmonary, Critical Care and Sleep Division, Department of Medicine, Tufts Medical Center, Boston, Massachusetts, USA
| | | |
Collapse
|
3
|
Dieffenbach PB, Aravamudhan A, Fredenburgh LE, Tschumperlin DJ. The Mechanobiology of Vascular Remodeling in the Aging Lung. Physiology (Bethesda) 2022; 37:28-38. [PMID: 34514871 PMCID: PMC8742727 DOI: 10.1152/physiol.00019.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Aging is accompanied by declining lung function and increasing susceptibility to lung diseases. The role of endothelial dysfunction and vascular remodeling in these changes is supported by growing evidence, but underlying mechanisms remain elusive. In this review we summarize functional, structural, and molecular changes in the aging pulmonary vasculature and explore how interacting aging and mechanobiological cues may drive progressive vascular remodeling in the lungs.
Collapse
Affiliation(s)
- Paul B. Dieffenbach
- 1Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Aja Aravamudhan
- 2Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| | - Laura E. Fredenburgh
- 1Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Daniel J. Tschumperlin
- 2Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| |
Collapse
|
4
|
Shoji H, Yoshida Y, Sanada TJ, Naito A, Maruyama J, Zhang E, Sumi K, Sakao S, Maruyama K, Hidaka H, Tatsumi K. The Isoquinoline-Sulfonamide Compound H-1337 Attenuates SU5416/Hypoxia-Induced Pulmonary Arterial Hypertension in Rats. Cells 2021; 11:66. [PMID: 35011628 PMCID: PMC8750965 DOI: 10.3390/cells11010066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 12/17/2021] [Accepted: 12/23/2021] [Indexed: 12/11/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterized by elevated pulmonary arterial pressure and right heart failure. Selective pulmonary vasodilators have improved the prognosis of PAH; however, they are not able to reverse pulmonary vascular remodeling. Therefore, a search for new treatment agents is required. H-1337 is an isoquinoline-sulfonamide compound that inhibits multiple serine/threonine kinases, including Rho-associated protein kinase (ROCK) and mammalian target of rapamycin (mTOR). Here, we investigated the effects of H-1337 on pulmonary hypertension and remodeling in the pulmonary vasculature and right ventricle in experimental PAH induced by SU5416 and hypoxia exposure. H-1337 and H-1337M1 exerted inhibitory effects on ROCK and Akt. H-1337 inhibited the phosphorylation of myosin light chain and mTOR and suppressed the proliferation of smooth muscle cells in vitro. H-1337 treatment also suppressed the phosphorylation of myosin light chain and mTOR in the pulmonary vasculature and decreased right ventricular systolic pressure and the extent of occlusive pulmonary vascular lesions. Furthermore, H-1337 suppressed aggravation of right ventricle hypertrophy. In conclusion, our data demonstrated that inhibition of ROCK and mTOR pathways with H-1337 suppressed the progression of pulmonary vascular remodeling, pulmonary hypertension, and right ventricular remodeling.
Collapse
Affiliation(s)
- Hiroki Shoji
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; (H.S.); (A.N.); (S.S.); (K.T.)
- Department of Respiratory Medicine, Tokyo Rosai Hospital, Tokyo 143-0013, Japan
| | - Yoko Yoshida
- D. Western Therapeutics Institute, Inc., Nagoya 460-0003, Japan; (Y.Y.); (K.S.); (H.H.)
- Human Research Promotion and Drug Development, Mie University, Mie 514-8507, Japan
| | - Takayuki Jujo Sanada
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; (H.S.); (A.N.); (S.S.); (K.T.)
| | - Akira Naito
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; (H.S.); (A.N.); (S.S.); (K.T.)
| | - Junko Maruyama
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Mie University, Mie 514-8507, Japan; (J.M.); (E.Z.); (K.M.)
- Faculty of Medical Engineering, Suzuka University of Medical Science, Mie 510-0293, Japan
| | - Erquan Zhang
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Mie University, Mie 514-8507, Japan; (J.M.); (E.Z.); (K.M.)
| | - Kengo Sumi
- D. Western Therapeutics Institute, Inc., Nagoya 460-0003, Japan; (Y.Y.); (K.S.); (H.H.)
- Human Research Promotion and Drug Development, Mie University, Mie 514-8507, Japan
| | - Seiichiro Sakao
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; (H.S.); (A.N.); (S.S.); (K.T.)
| | - Kazuo Maruyama
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Mie University, Mie 514-8507, Japan; (J.M.); (E.Z.); (K.M.)
| | - Hiroyoshi Hidaka
- D. Western Therapeutics Institute, Inc., Nagoya 460-0003, Japan; (Y.Y.); (K.S.); (H.H.)
- Human Research Promotion and Drug Development, Mie University, Mie 514-8507, Japan
| | - Koichiro Tatsumi
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; (H.S.); (A.N.); (S.S.); (K.T.)
| |
Collapse
|
5
|
Ali MK, Ichimura K, Spiekerkoetter E. Promising therapeutic approaches in pulmonary arterial hypertension. Curr Opin Pharmacol 2021; 59:127-139. [PMID: 34217109 DOI: 10.1016/j.coph.2021.05.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 04/12/2021] [Accepted: 05/11/2021] [Indexed: 12/19/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a debilitating multifactorial disease characterized by progressive pulmonary vascular remodeling, elevated pulmonary arterial pressure, and pulmonary vascular resistance, resulting in right ventricular failure and subsequent death. Current available therapies do not reverse the disease, resulting in a persistent high morbidity and mortality. Thus, there is an urgent unmet medical need for novel effective therapies to better treat patients with PAH. Over the past few years, enthusiastic attempts have been made to identify novel effective therapies that address the essential roots of PAH with targeting key signaling pathways in both preclinical models and patients with PAH. This review aims to discuss the most emerging and promising therapeutic interventions in PAH pathogenesis.
Collapse
Affiliation(s)
- Md Khadem Ali
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford Medical School, USA; Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, USA
| | - Kenzo Ichimura
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford Medical School, USA; Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, USA
| | - Edda Spiekerkoetter
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford Medical School, USA; Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, USA.
| |
Collapse
|
6
|
Shimoda LA. Cellular Pathways Promoting Pulmonary Vascular Remodeling by Hypoxia. Physiology (Bethesda) 2021; 35:222-233. [PMID: 32490752 DOI: 10.1152/physiol.00039.2019] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Exposure to hypoxia increases pulmonary vascular resistance, leading to elevated pulmonary arterial pressure and, potentially, right heart failure. Vascular remodeling is an important contributor to the increased pulmonary vascular resistance. Hyperproliferation of smooth muscle, endothelial cells, and fibroblasts, and deposition of extracellular matrix lead to increased wall thickness, extension of muscle into normally non-muscular arterioles, and vascular stiffening. This review highlights intrinsic and extrinsic modulators contributing to the remodeling process.
Collapse
Affiliation(s)
- Larissa A Shimoda
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
7
|
Pulmonary Hypertension in Acute and Chronic High Altitude Maladaptation Disorders. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18041692. [PMID: 33578749 PMCID: PMC7916528 DOI: 10.3390/ijerph18041692] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/05/2021] [Accepted: 02/07/2021] [Indexed: 12/13/2022]
Abstract
Alveolar hypoxia is the most prominent feature of high altitude environment with well-known consequences for the cardio-pulmonary system, including development of pulmonary hypertension. Pulmonary hypertension due to an exaggerated hypoxic pulmonary vasoconstriction contributes to high altitude pulmonary edema (HAPE), a life-threatening disorder, occurring at high altitudes in non-acclimatized healthy individuals. Despite a strong physiologic rationale for using vasodilators for prevention and treatment of HAPE, no systematic studies of their efficacy have been conducted to date. Calcium-channel blockers are currently recommended for drug prophylaxis in high-risk individuals with a clear history of recurrent HAPE based on the extensive clinical experience with nifedipine in HAPE prevention in susceptible individuals. Chronic exposure to hypoxia induces pulmonary vascular remodeling and development of pulmonary hypertension, which places an increased pressure load on the right ventricle leading to right heart failure. Further, pulmonary hypertension along with excessive erythrocytosis may complicate chronic mountain sickness, another high altitude maladaptation disorder. Importantly, other causes than hypoxia may potentially underlie and/or contribute to pulmonary hypertension at high altitude, such as chronic heart and lung diseases, thrombotic or embolic diseases. Extensive clinical experience with drugs in patients with pulmonary arterial hypertension suggests their potential for treatment of high altitude pulmonary hypertension. Small studies have demonstrated their efficacy in reducing pulmonary artery pressure in high altitude residents. However, no drugs have been approved to date for the therapy of chronic high altitude pulmonary hypertension. This work provides a literature review on the role of pulmonary hypertension in the pathogenesis of acute and chronic high altitude maladaptation disorders and summarizes current knowledge regarding potential treatment options.
Collapse
|
8
|
Yan S, Resta TC, Jernigan NL. Vasoconstrictor Mechanisms in Chronic Hypoxia-Induced Pulmonary Hypertension: Role of Oxidant Signaling. Antioxidants (Basel) 2020; 9:E999. [PMID: 33076504 PMCID: PMC7602539 DOI: 10.3390/antiox9100999] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 10/06/2020] [Accepted: 10/06/2020] [Indexed: 02/06/2023] Open
Abstract
Elevated resistance of pulmonary circulation after chronic hypoxia exposure leads to pulmonary hypertension. Contributing to this pathological process is enhanced pulmonary vasoconstriction through both calcium-dependent and calcium sensitization mechanisms. Reactive oxygen species (ROS), as a result of increased enzymatic production and/or decreased scavenging, participate in augmentation of pulmonary arterial constriction by potentiating calcium influx as well as activation of myofilament sensitization, therefore mediating the development of pulmonary hypertension. Here, we review the effects of chronic hypoxia on sources of ROS within the pulmonary vasculature including NADPH oxidases, mitochondria, uncoupled endothelial nitric oxide synthase, xanthine oxidase, monoamine oxidases and dysfunctional superoxide dismutases. We also summarize the ROS-induced functional alterations of various Ca2+ and K+ channels involved in regulating Ca2+ influx, and of Rho kinase that is responsible for myofilament Ca2+ sensitivity. A variety of antioxidants have been shown to have beneficial therapeutic effects in animal models of pulmonary hypertension, supporting the role of ROS in the development of pulmonary hypertension. A better understanding of the mechanisms by which ROS enhance vasoconstriction will be useful in evaluating the efficacy of antioxidants for the treatment of pulmonary hypertension.
Collapse
Affiliation(s)
| | | | - Nikki L. Jernigan
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (S.Y.); (T.C.R.)
| |
Collapse
|
9
|
Kuwabara Y, Tanaka-Ishikawa M, Abe K, Hirano M, Hirooka Y, Tsutsui H, Sunagawa K, Hirano K. Proteinase-activated receptor 1 antagonism ameliorates experimental pulmonary hypertension. Cardiovasc Res 2020; 115:1357-1368. [PMID: 30423156 DOI: 10.1093/cvr/cvy284] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 10/28/2018] [Accepted: 11/12/2018] [Indexed: 12/12/2022] Open
Abstract
AIMS Pulmonary hypertension (PH) is characterized by progressive increases in pulmonary vascular resistance (PVR). Thrombotic lesions are common pathological findings. The pulmonary artery has a unique property regarding the vasoconstrictive response to thrombin, which is mediated by proteinase-activated receptor 1 (PAR1). We aim to elucidate the role of PAR1 in the development and progression of PH. METHODS AND RESULTS A rat model of monocrotaline-induced PH and a mouse model of hypoxia (Hx)-induced PH were used to investigate the effects of atopaxar (a PAR1 antagonist) and PAR1 knockout on haemodynamic parameters, right ventricular hypertrophy (RVH), vascular remodelling and survival. In perfused lung preparations, the pressor response to PAR1 agonist was significantly augmented in monocrotaline-induced PH. Both the preventive and therapeutic administration of atopaxar significantly inhibited the increase in PVR and the development of RVH and prolonged survival. A real-time PCR revealed that the level of PAR1 mRNA in the pulmonary artery was significantly higher than that in any of the systemic arteries examined in control rats, and the level was significantly up-regulated in monocrotaline-induced PH. PAR1 gene knockout significantly attenuated the haemodynamic and histological findings in the mouse model of Hx-induced PH. CONCLUSION The specific expression of PAR1 in the pulmonary artery and its up-regulation were suggested to play a critical role in the development and progression of experimental PH in murine models. PAR1 is a potential therapeutic target for the treatment of PH.
Collapse
Affiliation(s)
- Yukimitsu Kuwabara
- Department of Molecular Cardiology, Research Institute of Angiocardiology, Graduate School of Medicine, Kyushu University, Fukuoka, Japan.,Department of Cardiovascular Medicine, Research Institute of Angiocardiology, Graduate School of Medicine, Kyushu University, Fukuoka, Japan
| | - Mariko Tanaka-Ishikawa
- Department of Cardiovascular Medicine, Research Institute of Angiocardiology, Graduate School of Medicine, Kyushu University, Fukuoka, Japan.,Department of Anesthesiology and Critical Care, Graduate School of Medicine, Kyushu University, Fukuoka, Japan
| | - Kohtaro Abe
- Department of Cardiovascular Medicine, Research Institute of Angiocardiology, Graduate School of Medicine, Kyushu University, Fukuoka, Japan
| | - Mayumi Hirano
- Department of Molecular Cardiology, Research Institute of Angiocardiology, Graduate School of Medicine, Kyushu University, Fukuoka, Japan
| | - Yoshitaka Hirooka
- Department of Advanced Cardiovascular Regulation and Therapeutics, Graduate School of Medicine, Kyushu University, Fukuoka, Japan
| | - Hiroyuki Tsutsui
- Department of Cardiovascular Medicine, Research Institute of Angiocardiology, Graduate School of Medicine, Kyushu University, Fukuoka, Japan
| | - Kenji Sunagawa
- Department of Cardiovascular Medicine, Research Institute of Angiocardiology, Graduate School of Medicine, Kyushu University, Fukuoka, Japan
| | - Katsuya Hirano
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, Japan
| |
Collapse
|
10
|
Weiss A, Boehm M, Egemnazarov B, Grimminger F, Savai Pullamsetti S, Kwapiszewska G, Schermuly RT. Kinases as potential targets for treatment of pulmonary hypertension and right ventricular dysfunction. Br J Pharmacol 2020; 178:31-53. [PMID: 31709514 DOI: 10.1111/bph.14919] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 10/07/2019] [Accepted: 10/21/2019] [Indexed: 12/11/2022] Open
Abstract
Pulmonary hypertension (PH) is a progressive pulmonary vasculopathy that causes chronic right ventricular pressure overload and often leads to right ventricular failure. Various kinase inhibitors have been studied in the setting of PH and either improved or worsened the disease, highlighting the importance of understanding the specific role of the respective kinases in a spatiotemporal cellular context. In this review, we will summarize the knowledge on the role of kinases in PH and focus on druggable targets for which certain criteria are met: (a) deregulation of the kinase in PH; (b) small-molecule inhibitors are available (e.g. from the oncology field); (c) preclinical studies have shown their efficacy in PH models; and (d) when available, therapeutic exploitation in human PH has been initiated. Along this line, clinical considerations such as personalized medicine approaches to predict therapy response and adverse side events such as cardiotoxicity together with their clinical management are discussed. LINKED ARTICLES: This article is part of a themed issue on Risk factors, comorbidities, and comedications in cardioprotection. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.1/issuetoc.
Collapse
Affiliation(s)
- Astrid Weiss
- Department of Internal Medicine, Justus-Liebig University Giessen, Giessen, Germany.,German Center for Lung Research (DZL), Giessen, Germany
| | - Mario Boehm
- Department of Internal Medicine, Justus-Liebig University Giessen, Giessen, Germany.,German Center for Lung Research (DZL), Giessen, Germany
| | | | - Friedrich Grimminger
- Department of Internal Medicine, Justus-Liebig University Giessen, Giessen, Germany.,German Center for Lung Research (DZL), Giessen, Germany
| | | | - Grazyna Kwapiszewska
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria.,Otto Loewi Center, Physiology, Medical University of Graz, Graz, Austria
| | - Ralph T Schermuly
- Department of Internal Medicine, Justus-Liebig University Giessen, Giessen, Germany
| |
Collapse
|
11
|
Liberale L, Bonaventura A, Montecucco F, Dallegri F, Carbone F. Impact of Red Wine Consumption on Cardiovascular Health. Curr Med Chem 2019; 26:3542-3566. [PMID: 28521683 DOI: 10.2174/0929867324666170518100606] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 03/05/2017] [Accepted: 03/05/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND The devastating effects of heavy alcohol drinking have been long time recognized. In the last decades, potential benefits of modest red wine drinking were suggested. In European countries in which red wide intake is not negligible (such as France), the association between cholesterol and cardiovascular (CV) risk was less evident, suggesting the action of some protective molecules in red wine or other foods and drinks. METHODS This narrative review is based on the material searched for and obtained via PubMed up to May 2016. The search terms we used were: "red wine, cardiovascular, alcohol" in combination with "polyphenols, heart failure, infarction". RESULTS Epidemiological and mechanistic evidence of a J-shaped relationship between red wine intake and CV risk further supported the "French paradox". Specific components of red wine both in vitro and in animal models were discovered. Polyphenols and especially resveratrol largely contribute to CV prevention mainly through antioxidant properties. They exert beneficial effects on endothelial dysfunction and hypertension, dyslipidemia, metabolic diseases, thus reducing the risk of adverse CV events such as myocardial infarction ischemic stroke and heart failure. Of interest, recent studies pointed out the role of ethanol itself as a potential cardioprotective agent, but a clear epidemiological evidence is still missing. The aim of this narrative review is to update current knowledge on the intracellular mechanism underlying the cardioprotective effects of polyphenols and ethanol. Furthermore, we summarized the results of epidemiological studies, emphasizing their methodological criticisms and the need for randomized clinical trials able to clarify the potential role of red wine consumption in reducing CV risk. CONCLUSION Caution in avowing underestimation of the global burden of alcohol-related diseases was particularly used.
Collapse
Affiliation(s)
- Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Aldo Bonaventura
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy.,IRCCS AOU San Martino - IST, Genova, 10 Largo Benzi, 16132 Genoa, Italy.,Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 9 viale Benedetto XV, 16132 Genoa, Italy
| | - Franco Dallegri
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy.,IRCCS AOU San Martino - IST, Genova, 10 Largo Benzi, 16132 Genoa, Italy
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| |
Collapse
|
12
|
Abstract
Pulmonary hypertension (PH) and its severe subtype pulmonary arterial hypertension (PAH) encompass a set of multifactorial diseases defined by sustained elevation of pulmonary arterial pressure and pulmonary vascular resistance leading to right ventricular failure and subsequent death. Pulmonary hypertension is characterized by vascular remodeling in association with smooth muscle cell proliferation of the arterioles, medial thickening, and plexiform lesion formation. Despite our recent advances in understanding its pathogenesis and related therapeutic discoveries, PH still remains a progressive disease without a cure. Nevertheless, development of drugs that specifically target molecular pathways involved in disease pathogenesis has led to improvement in life quality and clinical outcomes in patients with PAH. There are presently more than 12 Food and Drug Administration-approved vasodilator drugs in the United States for the treatment of PAH; however, mortality with contemporary therapies remains high. More recently, there have been exuberant efforts to develop new pharmacologic therapies that target the fundamental origins of PH and thus could represent disease-modifying opportunities. This review aims to summarize recent developments on key signaling pathways and molecular targets that drive PH disease progression, with emphasis on new therapeutic options under development.
Collapse
Affiliation(s)
- Chen-Shan Chen Woodcock
- Division of Cardiology, Department of Medicine, Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Stephen Y. Chan
- Division of Cardiology, Department of Medicine, Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|
13
|
Dieffenbach PB, Maracle M, Tschumperlin DJ, Fredenburgh LE. Mechanobiological Feedback in Pulmonary Vascular Disease. Front Physiol 2018; 9:951. [PMID: 30090065 PMCID: PMC6068271 DOI: 10.3389/fphys.2018.00951] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 06/28/2018] [Indexed: 01/06/2023] Open
Abstract
Vascular stiffening in the pulmonary arterial bed is increasingly recognized as an early disease marker and contributor to right ventricular workload in pulmonary hypertension. Changes in pulmonary artery stiffness throughout the pulmonary vascular tree lead to physiologic alterations in pressure and flow characteristics that may contribute to disease progression. These findings have led to a greater focus on the potential contributions of extracellular matrix remodeling and mechanical signaling to pulmonary hypertension pathogenesis. Several recent studies have demonstrated that the cellular response to vascular stiffness includes upregulation of signaling pathways that precipitate further vascular remodeling, a process known as mechanobiological feedback. The extracellular matrix modifiers, mechanosensors, and mechanotransducers responsible for this process have become increasingly well-recognized. In this review, we discuss the impact of vascular stiffening on pulmonary hypertension morbidity and mortality, evidence in favor of mechanobiological feedback in pulmonary hypertension pathogenesis, and the major contributors to mechanical signaling in the pulmonary vasculature.
Collapse
Affiliation(s)
- Paul B Dieffenbach
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States
| | - Marcy Maracle
- Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Daniel J Tschumperlin
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Laura E Fredenburgh
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States
| |
Collapse
|
14
|
Karoor V, Fini MA, Loomis Z, Sullivan T, Hersh LB, Gerasimovskaya E, Irwin D, Dempsey EC. Sustained Activation of Rho GTPases Promotes a Synthetic Pulmonary Artery Smooth Muscle Cell Phenotype in Neprilysin Null Mice. Arterioscler Thromb Vasc Biol 2018; 38:154-163. [PMID: 29191928 PMCID: PMC5746466 DOI: 10.1161/atvbaha.117.310207] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 11/15/2017] [Indexed: 01/16/2023]
Abstract
OBJECTIVE Pulmonary artery smooth muscle cells (PASMCs) from neprilysin (NEP) null mice exhibit a synthetic phenotype and increased activation of Rho GTPases compared with their wild-type counterparts. Although Rho GTPases are known to promote a contractile SMC phenotype, we hypothesize that their sustained activity decreases SM-protein expression in these cells. APPROACH AND RESULTS PASMCs isolated from wild-type and NEP-/- mice were used to assess levels of SM-proteins (SM-actin, SM-myosin, SM22, and calponin) by Western blotting, and were lower in NEP-/- PASMCs compared with wild-type. Rac and Rho (ras homology family member) levels and activity were higher in NEP-/- PASMCs, and ShRNA to Rac and Rho restored SM-protein, and attenuated the enhanced migration and proliferation of NEP-/- PASMCs. SM-gene repressors, p-Elk-1, and Klf4 (Kruppel lung factor 4), were higher in NEP-/- PASMCs and decreased by shRNA to Rac and Rho. Costimulation of wild-type PASMCs with PDGF (platelet-derived growth factor) and the NEP substrate, ET-1 (endothelin-1), increased Rac and Rho activity, and decreased SM-protein levels mimicking the NEP knock-out phenotype. Activation of Rac and Rho and downstream effectors was observed in lung tissue from NEP-/- mice and humans with chronic obstructive pulmonary disease. CONCLUSIONS Sustained Rho activation in NEP-/- PASMCs is associated with a decrease in SM-protein levels and increased migration and proliferation. Inactivation of RhoGDI (Rho guanine dissociation inhibitor) and RhoGAP (Rho GTPase activating protein) by phosphorylation may contribute to prolonged activation of Rho in NEP-/- PASMCs. Rho GTPases may thus have a role in integration of signals between vasopeptides and growth factor receptors and could influence pathways that suppress SM-proteins to promote a synthetic phenotype.
Collapse
MESH Headings
- Actins/biosynthesis
- Animals
- Becaplermin/pharmacology
- Calcium-Binding Proteins/biosynthesis
- Cell Movement
- Cell Proliferation
- Cells, Cultured
- Endothelin-1/pharmacology
- Enzyme Activation
- Genotype
- Humans
- Kruppel-Like Factor 4
- Kruppel-Like Transcription Factors/genetics
- Kruppel-Like Transcription Factors/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Microfilament Proteins/biosynthesis
- Muscle Proteins/biosynthesis
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- Neprilysin/deficiency
- Neprilysin/genetics
- Phenotype
- Pulmonary Artery/drug effects
- Pulmonary Artery/enzymology
- Pulmonary Artery/pathology
- Pulmonary Disease, Chronic Obstructive/enzymology
- Pulmonary Disease, Chronic Obstructive/pathology
- Signal Transduction
- Smooth Muscle Myosins/biosynthesis
- ets-Domain Protein Elk-1/genetics
- ets-Domain Protein Elk-1/metabolism
- rho GTP-Binding Proteins/genetics
- rho GTP-Binding Proteins/metabolism
- Calponins
Collapse
Affiliation(s)
- Vijaya Karoor
- From the Cardiovascular Pulmonary Research Laboratory (V.K., M.A.F., Z.L., T.S., E.G., D.I., E.C.D.) and Division of Pulmonary Sciences and Critical Care Medicine (V.K., M.A.F., E.C.D.), University of Colorado Anschutz Medical Campus, Aurora; Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington (L.B.H.); and Pulmonary and Critical Care, Denver VA Medical Center, CO (E.C.D.).
| | - Mehdi A Fini
- From the Cardiovascular Pulmonary Research Laboratory (V.K., M.A.F., Z.L., T.S., E.G., D.I., E.C.D.) and Division of Pulmonary Sciences and Critical Care Medicine (V.K., M.A.F., E.C.D.), University of Colorado Anschutz Medical Campus, Aurora; Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington (L.B.H.); and Pulmonary and Critical Care, Denver VA Medical Center, CO (E.C.D.)
| | - Zoe Loomis
- From the Cardiovascular Pulmonary Research Laboratory (V.K., M.A.F., Z.L., T.S., E.G., D.I., E.C.D.) and Division of Pulmonary Sciences and Critical Care Medicine (V.K., M.A.F., E.C.D.), University of Colorado Anschutz Medical Campus, Aurora; Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington (L.B.H.); and Pulmonary and Critical Care, Denver VA Medical Center, CO (E.C.D.)
| | - Timothy Sullivan
- From the Cardiovascular Pulmonary Research Laboratory (V.K., M.A.F., Z.L., T.S., E.G., D.I., E.C.D.) and Division of Pulmonary Sciences and Critical Care Medicine (V.K., M.A.F., E.C.D.), University of Colorado Anschutz Medical Campus, Aurora; Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington (L.B.H.); and Pulmonary and Critical Care, Denver VA Medical Center, CO (E.C.D.)
| | - Louis B Hersh
- From the Cardiovascular Pulmonary Research Laboratory (V.K., M.A.F., Z.L., T.S., E.G., D.I., E.C.D.) and Division of Pulmonary Sciences and Critical Care Medicine (V.K., M.A.F., E.C.D.), University of Colorado Anschutz Medical Campus, Aurora; Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington (L.B.H.); and Pulmonary and Critical Care, Denver VA Medical Center, CO (E.C.D.)
| | - Evgenia Gerasimovskaya
- From the Cardiovascular Pulmonary Research Laboratory (V.K., M.A.F., Z.L., T.S., E.G., D.I., E.C.D.) and Division of Pulmonary Sciences and Critical Care Medicine (V.K., M.A.F., E.C.D.), University of Colorado Anschutz Medical Campus, Aurora; Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington (L.B.H.); and Pulmonary and Critical Care, Denver VA Medical Center, CO (E.C.D.)
| | - David Irwin
- From the Cardiovascular Pulmonary Research Laboratory (V.K., M.A.F., Z.L., T.S., E.G., D.I., E.C.D.) and Division of Pulmonary Sciences and Critical Care Medicine (V.K., M.A.F., E.C.D.), University of Colorado Anschutz Medical Campus, Aurora; Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington (L.B.H.); and Pulmonary and Critical Care, Denver VA Medical Center, CO (E.C.D.)
| | - Edward C Dempsey
- From the Cardiovascular Pulmonary Research Laboratory (V.K., M.A.F., Z.L., T.S., E.G., D.I., E.C.D.) and Division of Pulmonary Sciences and Critical Care Medicine (V.K., M.A.F., E.C.D.), University of Colorado Anschutz Medical Campus, Aurora; Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington (L.B.H.); and Pulmonary and Critical Care, Denver VA Medical Center, CO (E.C.D.)
| |
Collapse
|
15
|
Zhang Y, Wu S. Effects of fasudil on pulmonary hypertension in clinical practice. Pulm Pharmacol Ther 2017; 46:54-63. [PMID: 28782712 DOI: 10.1016/j.pupt.2017.08.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 06/02/2017] [Accepted: 08/03/2017] [Indexed: 01/09/2023]
Abstract
Pulmonary hypertension (PH) is a pathophysiologic disorder that may involve multiple clinical conditions and can complicate the majority of cardiovascular and respiratory diseases. The presence of PH is associated with worse outcomes, but the efficacy of current therapy is still unsatisfactory. Because Rho-kinase (ROCK) plays an important role in the pathogenesis of PH, the ROCK inhibitor fasudil is expected to contribute to PH treatment. In animal models of PH, fasudil reduced pulmonary artery pressure (PAP) and improved survival. Furthermore, the short-term efficacy and safety of fasudil in the treatment of PH are demonstrated in clinical trials. Both PAP and pulmonary vascular resistance in patients with PH are significantly decreased by intravenous or inhaled fasudil without apparent side effect. However, no clinical trial has assessed the long-term efficacy of fasudil in the treatment of PH. Limited data suggest that the mid-term use of fasudil could improve exercise capacity and reduce in-hospital mortality. We also discuss the combined use of fasudil and other drugs for PH treatment. However, these combinations have not yet been evaluated in a clinical trial. According to animal studies, the combination of fasudil with beraprost or sildenafil shows synergistic effects, whereas the combination of fasudil with bosentan has no additional ameliorating effects on PH development.
Collapse
Affiliation(s)
- Yiqing Zhang
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Research Unit of Respiratory Disease, Central South University, Changsha, Hunan 410011, China; Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan 410011, China.
| | - Shangjie Wu
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Research Unit of Respiratory Disease, Central South University, Changsha, Hunan 410011, China; Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|
16
|
Nakamura K, Matsubara H, Akagi S, Sarashina T, Ejiri K, Kawakita N, Yoshida M, Miyoshi T, Watanabe A, Nishii N, Ito H. Nanoparticle-Mediated Drug Delivery System for Pulmonary Arterial Hypertension. J Clin Med 2017; 6:jcm6050048. [PMID: 28468233 PMCID: PMC5447939 DOI: 10.3390/jcm6050048] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 04/10/2017] [Accepted: 04/22/2017] [Indexed: 11/16/2022] Open
Abstract
Nanoparticles have been used as a novel drug delivery system. Drug-incorporated nanoparticles for local delivery might optimize the efficacy and minimize the side effects of drugs. The efficacy and safety of intratracheal administration of prostacyclin analog (beraprost) -incorporated nanoparticles and imatinib (a PDGF-receptor tyrosine kinase inhibitor) -incorporated nanoparticles in Sugen-hypoxia-normoxia or monocrotaline rat models of pulmonary arterial hypertension (PAH) and in human PAH-pulmonary arterial smooth muscle cells have been reported. The use of inhaled drug-incorporated nanoparticles might be a novel approach for the treatment of PAH.
Collapse
Affiliation(s)
- Kazufumi Nakamura
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| | - Hiromi Matsubara
- Division of Cardiology, National Hospital Organization Okayama Medical Center, Okayama 701-1192, Japan.
| | - Satoshi Akagi
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| | - Toshihiro Sarashina
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| | - Kentaro Ejiri
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| | - Norifumi Kawakita
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| | - Masashi Yoshida
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| | - Toru Miyoshi
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| | - Atsuyuki Watanabe
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| | - Nobuhiro Nishii
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| | - Hiroshi Ito
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| |
Collapse
|
17
|
Jernigan NL, Resta TC, Gonzalez Bosc LV. Altered Redox Balance in the Development of Chronic Hypoxia-induced Pulmonary Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 967:83-103. [PMID: 29047083 DOI: 10.1007/978-3-319-63245-2_7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Normally, the pulmonary circulation is maintained in a low-pressure, low-resistance state with little resting tone. Pulmonary arteries are thin-walled and rely heavily on pulmonary arterial distension and recruitment for reducing pulmonary vascular resistance when cardiac output is elevated. Under pathophysiological conditions, however, active vasoconstriction and vascular remodeling lead to enhanced pulmonary vascular resistance and subsequent pulmonary hypertension (PH). Chronic hypoxia is a critical pathological factor associated with the development of PH resulting from airway obstruction (COPD, sleep apnea), diffusion impairment (interstitial lung disease), developmental lung abnormalities, or high altitude exposure (World Health Organization [WHO]; Group III). The rise in pulmonary vascular resistance increases right heart afterload causing right ventricular hypertrophy that can ultimately lead to right heart failure in patients with chronic lung disease. PH is typically characterized by diminished paracrine release of vasodilators, antimitogenic factors, and antithrombotic factors (e.g., nitric oxide and protacyclin) and enhanced production of vasoconstrictors and mitogenic factors (e.g., reactive oxygen species and endothelin-1) from the endothelium and lung parenchyma. In addition, phenotypic changes to pulmonary arterial smooth muscle cells (PASMC), including alterations in Ca2+ homeostasis, Ca2+ sensitivity, and activation of transcription factors are thought to play prominent roles in the development of both vasoconstrictor and arterial remodeling components of hypoxia-associated PH. These changes in PASMC function are briefly reviewed in Sect. 1 and the influence of altered reactive oxygen species homeostasis on PASMC function discussed in Sects. 2-4.
Collapse
Affiliation(s)
- Nikki L Jernigan
- Department Cell Biology and Physiology, Vascular Physiology Group, University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Thomas C Resta
- Department Cell Biology and Physiology, Vascular Physiology Group, University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Laura V Gonzalez Bosc
- Department Cell Biology and Physiology, Vascular Physiology Group, University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, NM, 87131, USA.
| |
Collapse
|
18
|
Maron BA, Machado RF, Shimoda L. Pulmonary vascular and ventricular dysfunction in the susceptible patient (2015 Grover Conference series). Pulm Circ 2016; 6:426-438. [PMID: 28090285 PMCID: PMC5210067 DOI: 10.1086/688315] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 06/21/2016] [Indexed: 12/14/2022] Open
Abstract
Pulmonary blood vessel structure and tone are maintained by a complex interplay between endogenous vasoactive factors and oxygen-sensing intermediaries. Under physiological conditions, these signaling networks function as an adaptive interface between the pulmonary circulation and environmental or acquired perturbations to preserve oxygenation and maintain systemic delivery of oxygen-rich hemoglobin. Chronic exposure to hypoxia, however, triggers a range of pathogenetic mechanisms that include hypoxia-inducible factor 1α (HIF-1α)-dependent upregulation of the vasoconstrictor peptide endothelin 1 in pulmonary endothelial cells. In pulmonary arterial smooth muscle cells, chronic hypoxia induces HIF-1α-mediated upregulation of canonical transient receptor potential proteins, as well as increased Rho kinase-Ca2+ signaling and pulmonary arteriole synthesis of the profibrotic hormone aldosterone. Collectively, these mechanisms contribute to a contractile or hypertrophic pulmonary vascular phenotype. Genetically inherited disorders in hemoglobin structure are also an important etiology of abnormal pulmonary vasoreactivity. In sickle cell anemia, for example, consumption of the vasodilator and antimitogenic molecule nitric oxide by cell-free hemoglobin is an important mechanism underpinning pulmonary hypertension. Contemporary genomic and transcriptomic analytic methods have also allowed for the discovery of novel risk factors relevant to sickle cell disease, including GALNT13 gene variants. In this report, we review cutting-edge observations characterizing these and other pathobiological mechanisms that contribute to pulmonary vascular and right ventricular vulnerability.
Collapse
Affiliation(s)
- Bradley A. Maron
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA; and Department of Cardiology, Boston Veterans Affairs Healthcare System, Boston, Massachusetts, USA
| | - Roberto F. Machado
- Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Larissa Shimoda
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Asthma and Allergy Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
19
|
MURC deficiency in smooth muscle attenuates pulmonary hypertension. Nat Commun 2016; 7:12417. [PMID: 27546070 PMCID: PMC4996946 DOI: 10.1038/ncomms12417] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 06/30/2016] [Indexed: 02/06/2023] Open
Abstract
Emerging evidence suggests that caveolin-1 (Cav1) is associated with pulmonary arterial hypertension. MURC (also called Cavin-4) is a member of the cavin family, which regulates caveolar formation and functions together with caveolins. Here, we show that hypoxia increased Murc mRNA expression in the mouse lung, and that Murc-null mice exhibited attenuation of hypoxia-induced pulmonary hypertension (PH) accompanied by reduced ROCK activity in the lung. Conditional knockout mice lacking Murc in smooth muscle also resist hypoxia-induced PH. MURC regulates the proliferation and migration of pulmonary artery smooth muscle cells (PASMCs) through Rho/ROCK signalling. Cav1 suppresses RhoA activity in PASMCs, which is reversed by MURC. MURC binds to Cav1 and inhibits the association of Cav1 with the active form of Gα13, resulting in the facilitated association of the active form of Gα13 with p115RhoGEF. These results reveal that MURC has a function in the development of PH through modulating Rho/ROCK signalling. MURC protein regulates the function of caveolae, the small invaginations of the plasma membrane in muscle cells. Here the authors show that by interacting with caveolin proteins, MURC affects RhoA/ROCK signalling and regulates proliferation and migration of pulmonary artery smooth muscle cells, suggesting a new target in therapy of pulmonary hypertension.
Collapse
|
20
|
Jasińska-Stroschein M, Owczarek J, Sołtysiak U, Orszulak-Michalak D. Rosuvastatin intensifies the beneficial effects of rho-kinase inhibitor in reversal of monocrotaline-induced pulmonary hypertension. Arch Med Sci 2016; 12:898-905. [PMID: 27478473 PMCID: PMC4947607 DOI: 10.5114/aoms.2015.49740] [Citation(s) in RCA: 8] [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] [Received: 09/22/2014] [Accepted: 11/24/2014] [Indexed: 11/23/2022] Open
Abstract
INTRODUCTION It remains controversial whether statins have a beneficial effect on pulmonary arterial hypertension (PAH). This study is intended to evaluate whether statin, co-administered with Rho-kinase inhibitor, could enhance its efficacy. Although Rho-kinase inhibitors, including fasudil, have been reported to improve pulmonary hypertension in experimental and clinical studies, the combination of these agents has not been tested in the treatment of pulmonary hypertension (PH). MATERIAL AND METHODS The effects of such a regimen on hemodynamics, right ventricle hypertrophy, and Rho-associated protein kinase (ROCK) activity in experimental monocrotaline (MCT)-induced pulmonary hypertension were examined. Fourteen days after monocrotaline injection (60 mg/kg), male rats were treated orally for another 14 days with fasudil (15 mg/kg per day), or with a combination of fasudil + rosuvastatin (10 mg/kg per day). RESULTS The drug combination reversed the MCT-induced increase in right ventricle pressure (RVP) and reduced right ventricular hypertrophy (RV/LV + S ratio) more than Rho kinase inhibitor alone. The simultaneous administration of fasudil and rosuvastatin caused a further decrease of RhoA kinase activity in isolated lung tissues as compared to fasudil alone. CONCLUSIONS The results indicate that rosuvastatin intensifies the beneficial effects of Rho-kinase inhibitor on the Rho/Rho-kinase pathway and such a combination may represent an option for the treatment of pulmonary arterial hypertension.
Collapse
Affiliation(s)
| | - Jacek Owczarek
- Department of Biopharmacy, Medical University of Lodz, Lodz, Poland
| | | | | |
Collapse
|
21
|
Abstract
Twenty years ago, Rho-kinase was identified as an important downstream effector of the small GTP-binding protein, RhoA. Thereafter, a series of studies demonstrated the important roles of Rho-kinase in the cardiovascular system. The RhoA/Rho-kinase pathway is now widely known to play important roles in many cellular functions, including contraction, motility, proliferation, and apoptosis, and its excessive activity induces oxidative stress and promotes the development of cardiovascular diseases. Furthermore, the important role of Rho-kinase has been demonstrated in the pathogenesis of vasospasm, arteriosclerosis, ischemia/reperfusion injury, hypertension, pulmonary hypertension, and heart failure. Cyclophilin A is secreted by vascular smooth muscle cells and inflammatory cells and activated platelets in a Rho-kinase-dependent manner, playing important roles in a wide range of cardiovascular diseases. Thus, the RhoA/Rho-kinase pathway plays crucial roles under both physiological and pathological conditions and is an important therapeutic target in cardiovascular medicine. Recently, functional differences between ROCK1 and ROCK2 have been reported in vitro. ROCK1 is specifically cleaved by caspase-3, whereas granzyme B cleaves ROCK2. However, limited information is available on the functional differences and interactions between ROCK1 and ROCK2 in the cardiovascular system in vivo. Herein, we will review the recent advances about the importance of RhoA/Rho-kinase in the cardiovascular system.
Collapse
Affiliation(s)
- Hiroaki Shimokawa
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Shinichiro Sunamura
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kimio Satoh
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
22
|
Zychowski KE, Lucas SN, Sanchez B, Herbert G, Campen MJ. Hypoxia-induced pulmonary arterial hypertension augments lung injury and airway reactivity caused by ozone exposure. Toxicol Appl Pharmacol 2016; 305:40-45. [PMID: 27286659 DOI: 10.1016/j.taap.2016.06.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 05/10/2016] [Accepted: 06/03/2016] [Indexed: 11/29/2022]
Abstract
Ozone (O3)-related cardiorespiratory effects are a growing public health concern. Ground level O3 can exacerbate pre-existing respiratory conditions; however, research regarding therapeutic interventions to reduce O3-induced lung injury is limited. In patients with chronic obstructive pulmonary disease, hypoxia-associated pulmonary hypertension (HPH) is a frequent comorbidity that is difficult to treat clinically, yet associated with increased mortality and frequency of exacerbations. In this study, we hypothesized that established HPH would confer vulnerability to acute O3 pulmonary toxicity. Additionally, we tested whether improvement of pulmonary endothelial barrier integrity via rho-kinase inhibition could mitigate pulmonary inflammation and injury. To determine if O3 exacerbated HPH, male C57BL/6 mice were subject to either 3 weeks continuous normoxia (20.9% O2) or hypoxia (10.0% O2), followed by a 4-h exposure to either 1ppm O3 or filtered air (FA). As an additional experimental intervention fasudil (20mg/kg) was administered intraperitoneally prior to and after O3 exposures. As expected, hypoxia significantly increased right ventricular pressure and hypertrophy. O3 exposure in normoxic mice caused lung inflammation but not injury, as indicated by increased cellularity and edema in the lung. However, in hypoxic mice, O3 exposure led to increased inflammation and edema, along with a profound increase in airway hyperresponsiveness to methacholine. Fasudil administration resulted in reduced O3-induced lung injury via the enhancement of pulmonary endothelial barrier integrity. These results indicate that increased pulmonary vascular pressure may enhance lung injury, inflammation and edema when exposed to pollutants, and that enhancement of pulmonary endothelial barrier integrity may alleviate such vulnerability.
Collapse
Affiliation(s)
- Katherine E Zychowski
- Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, NM 87131, United States
| | - Selita N Lucas
- Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, NM 87131, United States
| | - Bethany Sanchez
- Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, NM 87131, United States
| | - Guy Herbert
- Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, NM 87131, United States
| | - Matthew J Campen
- Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, NM 87131, United States.
| |
Collapse
|
23
|
Challenges, priorities and novel therapies for hypoxemic respiratory failure and pulmonary hypertension in the neonate. J Perinatol 2016; 36 Suppl 2:S32-6. [PMID: 27225964 DOI: 10.1038/jp.2016.47] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 02/10/2016] [Accepted: 02/16/2016] [Indexed: 01/31/2023]
Abstract
Future priorities for the management of hypoxemic respiratory failure (HRF) and pulmonary hypertension include primary prevention of neonatal lung diseases, 'precision medicine' and translating promising clinical and preclinical research into novel therapies. Promising areas of investigation include noninvasive ventilation strategies, emerging pulmonary vasodilators (for example, cinaciguat, intravenous bosentan, rho-kinase inhibitors, peroxisome proliferator-activated receptor-γ agonists) and hemodynamic support (arginine vasopressin). Research challenges include the optimal timing for primary prevention interventions and development of validated biomarkers that predict later disease or serve as surrogates for long-term respiratory outcomes. Differentiating respiratory disease endotypes using biomarkers and experimental therapies tailored to the underlying pathobiology are central to the concept of 'precision medicine' (that is, prevention and treatment strategies that take individual variability into account). The ideal biomarker should be expressed early in the neonatal course to offer an opportunity for effective and targeted interventions to modify outcomes. The feasibility of this approach will depend on the identification and validation of accurate, rapid and affordable point-of-care biomarker tests. Trials targeting patient-specific pathobiology may involve less risk than traditional randomized controlled trials that enroll all at-risk neonates. Such approaches would reduce trial costs, potentially with fewer negative trials and improved health outcomes. Initiatives such as the Prematurity and Respiratory Outcomes Program, supported by the National Heart, Lung, and Blood Institute, provide a framework to develop refined outcome measures and early biomarkers that will enhance our understanding of novel, mechanistic therapeutic targets that can be tested in clinical trials in neonates with HRF.
Collapse
|
24
|
Qiao F, Zou Z, Liu C, Zhu X, Wang X, Yang C, Jiang T, Chen Y. ROCK2 mediates the proliferation of pulmonary arterial endothelial cells induced by hypoxia in the development of pulmonary arterial hypertension. Exp Ther Med 2016; 11:2567-2572. [PMID: 27284349 DOI: 10.3892/etm.2016.3214] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 03/10/2016] [Indexed: 11/06/2022] Open
Abstract
It has been reported that RhoA activation and Rho-kinase (ROCK) expression are increased in chronic hypoxic lungs, and the long-term inhibition of ROCK markedly improves the survival of patients with pulmonary arterial hypertension (PAH). However, whether Rho-kinase α (ROCK2) participates in regulation of the growth of pulmonary arterial endothelial cells (PAECs) remains unknown. The aim of the present study was to investigate the effect of hypoxia on the proliferation of PAECs and the role of ROCK2 in the underlying mechanism. The results of western blotting and reverse transcription-quantitative polymerase chain reaction analysis showed that hypoxia increased the activity and expression of ROCK2 in PAECs, and the stimulating effects of hypoxia on the proliferation of PAECs were attenuated by either the ROCK inhibitor Y27632 or transfection with ROCK2 small interfering RNA. Moreover, analysis of cyclin A and cyclin D1 mRNA expression indicated that ROCK2 mediates the cell cycle progression promoted by hypoxia. These results indicate that hypoxia promotes the proliferation of pulmonary arterial endothelial cells via activation of the ROCK2 signaling pathway.
Collapse
Affiliation(s)
- Feng Qiao
- Department of Thoracic Surgery, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154002, P.R. China
| | - Zhitian Zou
- Department of Thoracic Surgery, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154002, P.R. China
| | - Chunhui Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154002, P.R. China
| | - Xiaofeng Zhu
- Department of Thoracic Surgery, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154002, P.R. China
| | - Xiaoqiang Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154002, P.R. China
| | - Chengpeng Yang
- Department of Thoracic Surgery, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154002, P.R. China
| | - Tengjiao Jiang
- Department of Thoracic Surgery, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154002, P.R. China
| | - Ying Chen
- Department of Intensive Care Unit, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154002, P.R. China
| |
Collapse
|
25
|
Light and Dark of Reactive Oxygen Species for Vascular Function: 2014 ASVB (Asian Society of Vascular Biology). J Cardiovasc Pharmacol 2016; 65:412-8. [PMID: 25162437 DOI: 10.1097/fjc.0000000000000159] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Vascular-derived hydrogen peroxide (H2O2) serves as an important signaling molecule in the cardiovascular system and contributes to vascular homeostasis. H2O2 is a second messenger, transducing the oxidative signal into biological responses through posttranslational protein modification. The balance between oxidant and antioxidant systems regulates intracellular redox status, and their imbalance causes oxidative or reductive stress, leading to cellular damage in cardiovascular systems. Excessive H2O2 deteriorates vascular functions and promotes vascular disease through multiple pathways. The RhoA/Rho-kinase pathway plays an important role in various fundamental cellular functions, including production of excessive reactive oxygen species, leading to the development of cardiovascular diseases. Rho-kinase (ROCK1 and ROCK2) belongs to the family of serine/threonine kinases and is an important downstream effector of the small GTP-binding protein RhoA. Rho-kinase plays a crucial role in the pathogenesis of vasospasm, arteriosclerosis, ischemia/reperfusion injury, hypertension, pulmonary hypertension, stroke, and heart failure. Thus, Rho-kinase inhibitors may be useful for the treatment of cardiovascular diseases in humans. In this review, we will briefly discuss the roles of vascular-derived H2O2 and review the recent progress in the translational research on the therapeutic importance of the Rho-kinase pathway in cardiovascular medicine.
Collapse
|
26
|
Abstract
Vascular homeostasis is regulated by complex interactions between many vascular cell components, including endothelial cells, vascular smooth muscle cells (VSMCs), adventitial inflammatory cells, and autonomic nervous system. The balance between oxidant and antioxidant systems determines intracellular redox status, and their imbalance can cause oxidative stress. Excessive oxidative stress is one of the important stimuli that induce cellular damage and dysregulation of vascular cell components, leading to vascular diseases through multiple pathways. Cyclophilin A (CyPA) is one of the causative proteins that mediate oxidative stress-induced cardiovascular dysfunction. CyPA was initially discovered as the intracellular receptor of the immunosuppressive drug cyclosporine 30 years ago. However, recent studies have established that CyPA is secreted from vascular cell components, such as endothelial cells and VSMCs. Extracellular CyPA augments the development of cardiovascular diseases. CyPA secretion is regulated by Rho-kinase, which contributes to the pathogenesis of vasospasm, arteriosclerosis, ischemia/reperfusion injury, hypertension, pulmonary hypertension, and heart failure. We recently reported that plasma CyPA levels are significantly higher in patients with coronary artery disease, which is associated with increased numbers of stenotic coronary arteries and the need for coronary intervention in such patients. Furthermore, we showed that the vascular erythropoietin (Epo)/Epo receptor system plays an important role in production of nitric oxide and maintenance of vascular redox state and homeostasis, with a potential mechanistic link to the Rho-kinase-CyPA pathway. In this article, I review the data on the protective role of the vascular Epo/Epo receptor system and discuss the roles of the CyPA/Rho-kinase system in cardiovascular diseases.
Collapse
Affiliation(s)
- Kimio Satoh
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine
| |
Collapse
|
27
|
Hu J, Xu Q, McTiernan C, Lai YC, Osei-Hwedieh D, Gladwin M. Novel Targets of Drug Treatment for Pulmonary Hypertension. Am J Cardiovasc Drugs 2015; 15:225-34. [PMID: 26016608 DOI: 10.1007/s40256-015-0125-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Biomedical advances over the last decade have identified the central role of proliferative pulmonary arterial smooth muscle cells (PASMCs) in the development of pulmonary hypertension (PH). Furthermore, promoters of proliferation and apoptosis resistance in PASMCs and endothelial cells, such as aberrant signal pathways involving growth factors, G protein-coupled receptors, kinases, and microRNAs, have also been described. As a result of these discoveries, PH is currently divided into subgroups based on the underlying pathology, which allows focused and targeted treatment of the condition. The defining features of PH, which subsequently lead to vascular wall remodeling, are dysregulated proliferation of PASMCs, local inflammation, and apoptosis-resistant endothelial cells. Efforts to assess the relative contributions of these factors have generated several promising targets. This review discusses recent novel targets of therapies for PH that have been developed as a result of these advances, which are now in pre-clinical and clinical trials (e.g., imatinib [phase III]; nilotinib, AT-877ER, rituximab, tacrolimus, paroxetine, sertraline, fluoxetine, bardoxolone methyl [phase II]; and sorafenib, FK506, aviptadil, endothelial progenitor cells (EPCs) [phase I]). While substantial progress has been made in recent years in targeting key molecular pathways, PH still remains without a cure, and these novel therapies provide an important conceptual framework of categorizing patients on the basis of molecular phenotype(s) for effective treatment of the disease.
Collapse
|
28
|
Shimokawa H, Satoh K. 2015 ATVB Plenary Lecture: translational research on rho-kinase in cardiovascular medicine. Arterioscler Thromb Vasc Biol 2015; 35:1756-69. [PMID: 26069233 DOI: 10.1161/atvbaha.115.305353] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 05/27/2015] [Indexed: 02/07/2023]
Abstract
Rho-kinase (ROCKs) is an important downstream effector of the small GTP-binding protein Ras homolog gene family member A. There are 2 isoforms of ROCK, ROCK1 and ROCK2, and they have different functions in several vascular components. The Ras homolog gene family member A/ROCK pathway plays an important role in various fundamental cellular functions, including contraction, motility, proliferation, and apoptosis, whereas its excessive activity is involved in the pathogenesis of cardiovascular diseases. For the past 20 years, a series of translational research studies have demonstrated the important roles of ROCK in the pathogenesis of cardiovascular diseases. At the molecular and cellular levels, ROCK upregulates several molecules related to inflammation, thrombosis, and fibrosis. In animal experiments, ROCK plays an important role in the pathogenesis of vasospasm, arteriosclerosis, hypertension, pulmonary hypertension, and heart failure. Finally, at the human level, ROCK is substantially involved in the pathogenesis of coronary vasospasm, angina pectoris, hypertension, pulmonary hypertension, and heart failure. Furthermore, ROCK activity in circulating leukocytes is a useful biomarker for the assessment of disease severity and therapeutic responses in vasospastic angina, heart failure, and pulmonary hypertension. In addition to fasudil, many other ROCK inhibitors are currently under development for various indications. Thus, the ROCK pathway is an important novel therapeutic target in cardiovascular medicine.
Collapse
MESH Headings
- Animals
- Cardiovascular Agents/therapeutic use
- Cardiovascular Diseases/drug therapy
- Cardiovascular Diseases/enzymology
- Cardiovascular Diseases/pathology
- Cardiovascular Diseases/physiopathology
- Disease Models, Animal
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/enzymology
- Endothelium, Vascular/pathology
- Endothelium, Vascular/physiopathology
- Humans
- Molecular Targeted Therapy
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/physiopathology
- Protein Kinase Inhibitors/therapeutic use
- Signal Transduction
- Translational Research, Biomedical
- rho-Associated Kinases/antagonists & inhibitors
- rho-Associated Kinases/metabolism
Collapse
Affiliation(s)
- Hiroaki Shimokawa
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Kimio Satoh
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
29
|
Abstract
Pulmonary arterial hypertension (PAH) includes a heterogeneous group of diseases characterized by pulmonary vasoconstriction and remodeling of the lung circulation. Although PAH is a disease of the lungs, patients with PAH frequently die of right heart failure. Indeed, survival of patients with PAH depends on the adaptive response of the right ventricle (RV) to the changes in the lung circulation. PAH-specific drugs affect the function of the RV through afterload reduction and perhaps also through direct effects on the myocardium. Prostacyclins, type 5 phosphodiesterase inhibitors, and guanylyl cyclase stimulators may directly enhance myocardial contractility through increased cyclic adenosine and guanosine monophosphate availability. Although this may initially improve cardiac performance, the long-term effects on myocardial oxygen consumption and function are unclear. Cardiac effects of endothelin receptor antagonists may be opposite, as endothelin-1 is known to suppress cardiac contractility. Because PAH is increasingly considered as a disease with quasimalignant growth of cells in the pulmonary vascular wall, therapies are being developed that inhibit hypertrophy and angiogenesis, and promote apoptosis. The inherent danger of these therapies is a further compromise to the already ischemic, fibrotic, and dysfunctional RV. More recently, the right heart has been identified as a direct treatment target in PAH. The effects of well established therapies for left heart failure, such as β-adrenergic receptor blockers, inhibitors of the renin-angiotensin system, exercise training, and assist devices, are currently being investigated in PAH. Future treatment of patients with PAH will likely consist of a multifaceted approaches aiming to reduce the pressure in the lung circulation and improving right heart adaptation simultaneously.
Collapse
|
30
|
Vaidya B, Gupta V. Novel therapeutic approaches for pulmonary arterial hypertension: Unique molecular targets to site-specific drug delivery. J Control Release 2015; 211:118-33. [PMID: 26036906 DOI: 10.1016/j.jconrel.2015.05.287] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 05/26/2015] [Accepted: 05/28/2015] [Indexed: 01/07/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a cardiopulmonary disorder characterized by increased blood pressure in the small arterioles supplying blood to lungs for oxygenation. Advances in understanding of molecular and cellular biology techniques have led to the findings that PAH is indeed a cascade of diseases exploiting multi-faceted complex pathophysiology, with cellular proliferation and vascular remodeling being the key pathogenic events along with several cellular pathways involved. While current therapies for PAH do provide for amelioration of disease symptoms and acute survival benefits, their full therapeutic potential is hindered by patient incompliance and off-target side effects. To overcome the issues related with current therapy and to devise a more selective therapy, various novel pathways are being investigated for PAH treatment. In addition, inability to deliver anti-PAH drugs to the disease site i.e., distal pulmonary arterioles has been one of the major challenges in achieving improved patient outcomes and improved therapeutic efficacy. Several novel carriers have been explored to increase the selectivity of currently approved anti-PAH drugs and to act as suitable carriers for the delivery of investigational drugs. In the present review, we have discussed potential of various novel molecular pathways/targets including RhoA/Rho kinase, tyrosine kinase, endothelial progenitor cells, vasoactive intestinal peptide, and miRNA in PAH therapeutics. We have also discussed various techniques for site-specific drug delivery of anti-PAH therapeutics so as to improve the efficacy of approved and investigational drugs. This review will provide gainful insights into current advances in PAH therapeutics with an emphasis on site-specific drug payload delivery.
Collapse
Affiliation(s)
- Bhuvaneshwar Vaidya
- School of Pharmacy, Keck Graduate Institute, 535 Watson Drive, Claremont, CA 91711, United States
| | - Vivek Gupta
- School of Pharmacy, Keck Graduate Institute, 535 Watson Drive, Claremont, CA 91711, United States.
| |
Collapse
|
31
|
Lu A, Zuo C, He Y, Chen G, Piao L, Zhang J, Xiao B, Shen Y, Tang J, Kong D, Alberti S, Chen D, Zuo S, Zhang Q, Yan S, Fei X, Yuan F, Zhou B, Duan S, Yu Y, Lazarus M, Su Y, Breyer RM, Funk CD, Yu Y. EP3 receptor deficiency attenuates pulmonary hypertension through suppression of Rho/TGF-β1 signaling. J Clin Invest 2015; 125:1228-42. [PMID: 25664856 DOI: 10.1172/jci77656] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 01/05/2015] [Indexed: 01/27/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is commonly associated with chronic hypoxemia in disorders such as chronic obstructive pulmonary disease (COPD). Prostacyclin analogs are widely used in the management of PAH patients; however, clinical efficacy and long-term tolerability of some prostacyclin analogs may be compromised by concomitant activation of the E-prostanoid 3 (EP3) receptor. Here, we found that EP3 expression is upregulated in pulmonary arterial smooth muscle cells (PASMCs) and human distal pulmonary arteries (PAs) in response to hypoxia. Either pharmacological inhibition of EP3 or Ep3 deletion attenuated both hypoxia and monocrotaline-induced pulmonary hypertension and restrained extracellular matrix accumulation in PAs in rodent models. In a murine PAH model, Ep3 deletion in SMCs, but not endothelial cells, retarded PA medial thickness. Knockdown of EP3α and EP3β, but not EP3γ, isoforms diminished hypoxia-induced TGF-β1 activation. Expression of either EP3α or EP3β in EP3-deficient PASMCs restored TGF-β1 activation in response to hypoxia. EP3α/β activation in PASMCs increased RhoA-dependent membrane type 1 extracellular matrix metalloproteinase (MMP) translocation to the cell surface, subsequently activating pro-MMP-2 and promoting TGF-β1 signaling. Activation or disruption of EP3 did not influence PASMC proliferation. Together, our results indicate that EP3 activation facilitates hypoxia-induced vascular remodeling and pulmonary hypertension in mice and suggest EP3 inhibition as a potential therapeutic strategy for pulmonary hypertension.
Collapse
MESH Headings
- Animals
- Cell Hypoxia
- Cells, Cultured
- Extracellular Matrix/metabolism
- Extracellular Matrix Proteins/metabolism
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/physiopathology
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Pulmonary Artery/metabolism
- Rats, Sprague-Dawley
- Receptors, Prostaglandin E, EP3 Subtype/antagonists & inhibitors
- Receptors, Prostaglandin E, EP3 Subtype/genetics
- Receptors, Prostaglandin E, EP3 Subtype/metabolism
- Signal Transduction
- Sulfonamides/pharmacology
- Transforming Growth Factor beta1/physiology
- Vascular Remodeling
- rho GTP-Binding Proteins/metabolism
- rhoA GTP-Binding Protein
Collapse
|
32
|
Dong Z, Gong K, Huang D, Zhu W, Sun W, Zhang Y, Xin P, Shen Y, Wu P, Li J, Lu Z, Zhang X, Wei M. Myocardial infarction accelerates glomerular injury and microalbuminuria in diabetic rats via local hemodynamics and immunity. Int J Cardiol 2014; 179:397-408. [PMID: 25464495 DOI: 10.1016/j.ijcard.2014.11.033] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Revised: 11/02/2014] [Accepted: 11/04/2014] [Indexed: 02/08/2023]
Abstract
BACKGROUND Clinically, approximately one-third of patients with chronic heart failure (CHF) exhibit some degree of renal dysfunction. This renal dysfunction is referred to as cardiorenal syndrome (CRS) and plays an important role in the poor prognosis of CHF. Mounting evidence suggests that diabetes is the most common underlying risk factor for CRS. However, the underlying pathophysiological mechanisms are poorly understood. METHODS We performed the following comparisons in two separate protocols: 1) surgically induced myocardial infarction rats (MI, n=10), sham operation rats (Ctr, n=10) and MI rats treated with Fasudil, a Rho-kinase inhibitor (MI+Fas, n=9); and 2) STZ-induced type 1 diabetic rats (DB, n=10), DB+MI rats (n=10) and DB+MI rats treated with Fasudil (DB+MI+Fas, n=9). Renal hemodynamics and vasoconstrictor reactivity were evaluated using the DMT myograph system. Renal immunity was evaluated by flow cytometry, electron microscopy, immunofluorescence, etc. RESULTS Twelve weeks after the operation, compared with DB or MI rats, DB+MI rats exhibited the following characteristics: 1) significantly increased glomerular enlargement, fibrosis, glomerulosclerosis, podocyte injury and microalbuminuria; 2) significantly increased vasoconstrictor reactivity of the renal interlobular arteries and renal venous pressure; 3) significantly increased infiltration of CD₃+ and CD₄+ T cells and decreased Treg/Th17 ratios; and 4) significantly increased glomerular deposition of IgG and C₄. In contrast, rats with MI only showed mildly accelerated glomerular remodeling and microalbuminuria, with little change in renal hemodynamics and immunity. Fasudil treatment significantly improved the renal lesions in DB+MI rats but not MI rats. CONCLUSIONS Post-MI cardiac dysfunction significantly accelerated glomerular remodeling, podocyte injury and microalbuminuria in STZ-induced diabetic rats. These changes were accompanied by altered local hemodynamics and immunity.
Collapse
Affiliation(s)
- Zhifeng Dong
- Department of Cardiology, Shanghai Sixth Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200233, China
| | - Kaizheng Gong
- Department of Cardiology, The Second Clinical Medical College, Yangzhou University, Yangzhou 225001, China
| | - Dong Huang
- Department of Cardiology, Shanghai Sixth Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200233, China
| | - Wei Zhu
- Department of Cardiology, Shanghai Sixth Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200233, China
| | - Wanfeng Sun
- Affiliated Yancheng Hospital of Medical School, Southeast University, Yancheng 224001, China
| | - Ying Zhang
- Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ping Xin
- Department of Cardiology, Shanghai Sixth Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200233, China
| | - Yuan Shen
- Affiliated Yancheng Hospital of Medical School, Southeast University, Yancheng 224001, China
| | - Penglong Wu
- Department of Cardiology, Shanghai Sixth Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200233, China
| | - Jingbo Li
- Department of Cardiology, Shanghai Sixth Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200233, China
| | - Zhigang Lu
- Department of Cardiology, Shanghai Sixth Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200233, China
| | - Xiaoming Zhang
- Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Meng Wei
- Department of Cardiology, Shanghai Sixth Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200233, China.
| |
Collapse
|
33
|
Pandit LM, Lloyd EE, Reynolds JO, Lawrence WS, Reynolds C, Wehrens XHT, Bryan RM. TWIK-2 channel deficiency leads to pulmonary hypertension through a rho-kinase-mediated process. Hypertension 2014; 64:1260-5. [PMID: 25245387 DOI: 10.1161/hypertensionaha.114.03406] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
TWIK-2 (KCNK6) is a member of the 2-pore domain (K2P) family of potassium channels, which are highly expressed in the vascular system. We tested the hypothesis that TWIK-2 deficiency leads to pulmonary hypertension. TWIK-2 knockout mice and their wildtype littermates at 8 weeks of age had similar mean right ventricular systolic pressures (24±3 and 21±3 mm Hg, respectively.) Significantly, by 20 weeks of age, the mean right ventricular systolic pressures in TWIK-2 knockout mice increased to 35±3 mm Hg (P≤0.036), whereas mean right ventricular systolic pressures in wildtype littermates remained at 22±3 mm Hg. Elevated mean right ventricular systolic pressures in the TWIK-2 knockout mice was accompanied by pulmonary vascular remodeling as determined by a 25% increase in the cross-sectional area of the vessels occupied by the vessel wall. Additionally, secondary branches of the pulmonary artery from 20-week-old TWIK-2 knockout mice showed an enhanced contractile response to U46619 (10(-6) moles/L), a thromboxane A2 mimetic, which was completely abolished with the Rho-kinase inhibitor, Y27632 (10(-6) and 10(-5) moles/L). Treatment of TWIK-2 knockout mice with the Rho-kinase inhibitor, fasudil, in the drinking water for 12 weeks, abolished the development of pulmonary hypertension and attenuated the vessel remodeling. We concluded that mice deficient in the TWIK-2 channel develop pulmonary hypertension between 8 and 20 weeks of age through a mechanism involving Rho-kinase. Our results suggest that downregulation of TWIK-2 in the pulmonary vasculature may be an underlying mechanism in the development of pulmonary hypertension.
Collapse
Affiliation(s)
- Lavannya M Pandit
- From the Departments of Internal Medicine (L.M.P., X.H.T.W., R.M.B.), Anesthesiology (E.E.L., R.M.B.), Cardiovascular Research Institute, Department of Molecular Physiology and Biophysics (J.O.R., C.R., X.H.T.W., R.M.B.), Baylor College of Medicine, and Department of Microbiology and Immunology (W.S.L.), The University of Texas Medical Branch
| | - Eric E Lloyd
- From the Departments of Internal Medicine (L.M.P., X.H.T.W., R.M.B.), Anesthesiology (E.E.L., R.M.B.), Cardiovascular Research Institute, Department of Molecular Physiology and Biophysics (J.O.R., C.R., X.H.T.W., R.M.B.), Baylor College of Medicine, and Department of Microbiology and Immunology (W.S.L.), The University of Texas Medical Branch
| | - Julia O Reynolds
- From the Departments of Internal Medicine (L.M.P., X.H.T.W., R.M.B.), Anesthesiology (E.E.L., R.M.B.), Cardiovascular Research Institute, Department of Molecular Physiology and Biophysics (J.O.R., C.R., X.H.T.W., R.M.B.), Baylor College of Medicine, and Department of Microbiology and Immunology (W.S.L.), The University of Texas Medical Branch
| | - William S Lawrence
- From the Departments of Internal Medicine (L.M.P., X.H.T.W., R.M.B.), Anesthesiology (E.E.L., R.M.B.), Cardiovascular Research Institute, Department of Molecular Physiology and Biophysics (J.O.R., C.R., X.H.T.W., R.M.B.), Baylor College of Medicine, and Department of Microbiology and Immunology (W.S.L.), The University of Texas Medical Branch
| | - Corey Reynolds
- From the Departments of Internal Medicine (L.M.P., X.H.T.W., R.M.B.), Anesthesiology (E.E.L., R.M.B.), Cardiovascular Research Institute, Department of Molecular Physiology and Biophysics (J.O.R., C.R., X.H.T.W., R.M.B.), Baylor College of Medicine, and Department of Microbiology and Immunology (W.S.L.), The University of Texas Medical Branch
| | - Xander H T Wehrens
- From the Departments of Internal Medicine (L.M.P., X.H.T.W., R.M.B.), Anesthesiology (E.E.L., R.M.B.), Cardiovascular Research Institute, Department of Molecular Physiology and Biophysics (J.O.R., C.R., X.H.T.W., R.M.B.), Baylor College of Medicine, and Department of Microbiology and Immunology (W.S.L.), The University of Texas Medical Branch
| | - Robert M Bryan
- From the Departments of Internal Medicine (L.M.P., X.H.T.W., R.M.B.), Anesthesiology (E.E.L., R.M.B.), Cardiovascular Research Institute, Department of Molecular Physiology and Biophysics (J.O.R., C.R., X.H.T.W., R.M.B.), Baylor College of Medicine, and Department of Microbiology and Immunology (W.S.L.), The University of Texas Medical Branch
| |
Collapse
|
34
|
Kolluru GK, Majumder S, Chatterjee S. Rho-kinase as a therapeutic target in vascular diseases: striking nitric oxide signaling. Nitric Oxide 2014; 43:45-54. [PMID: 25196952 DOI: 10.1016/j.niox.2014.09.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 09/03/2014] [Accepted: 09/03/2014] [Indexed: 11/27/2022]
Abstract
Rho GTPases are a globular, monomeric group of small signaling G-protein molecules. Rho-associated protein kinase/Rho-kinase (ROCK) is a downstream effector protein of the Rho GTPase. Rho-kinases are the potential therapeutic targets in the treatment of cardiovascular diseases. Here, we have primarily discussed the intriguing roles of ROCK in cardiovascular health in relation to nitric oxide signaling. Further, we highlighted the biphasic effects of Y-27632, a ROCK inhibitor under shear stress, which acts as an agonist of nitric oxide production in endothelial cells. The biphasic effects of this inhibitor raised the question of safety of the drug usage in treating cardiovascular diseases.
Collapse
Affiliation(s)
| | - Syamantak Majumder
- Aab Cardiovascular Research Institute, School of Medicine and Dentistry, University of Rochester, Rochester, NY, USA
| | - Suvro Chatterjee
- Department of Biotechnology, Anna University, Chennai, India; Vascular Biology Lab, AU-KBC Research Centre, Anna University, Chennai, India.
| |
Collapse
|
35
|
Liu M, Wang Y, Zheng L, Zheng W, Dong K, Chen S, Zhang B, Li Z. Fasudil reversed MCT-induced and chronic hypoxia-induced pulmonary hypertension by attenuating oxidative stress and inhibiting the expression of Trx1 and HIF-1α. Respir Physiol Neurobiol 2014; 201:38-46. [PMID: 24973470 DOI: 10.1016/j.resp.2014.06.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 05/30/2014] [Accepted: 06/02/2014] [Indexed: 01/07/2023]
Abstract
Antioxidant therapy attenuates pulmonary hypertension (PH). In the present study, we tested the antioxidant effects of fasudil against PH in rats. Monocrotaline (MCT)-induced and chronic hypoxia-induced PH models of rats were established, and the haemodynamic and pathomorphologic results of three different doses of fasudil (10 mg/kg, 30 mg/kg, and 75 mg/kg per day) were subsequently compared with those of bosentan (30 mg/kg per day). Additionally, the protein expressions of thioredoxin-1 (Trx1) and hypoxia inducible factor-1α (HIF-1α), the content of superoxide dismutase (SOD), and the levels of hydrogen peroxide (H2O2), malonyldialdehyde (MDA), and hydroxy radical (·OH) were investigated. Fasudil effectively reduced the right ventricular systolic pressure (RVSP) and alleviated right ventricle (RV) hypertrophy, as well as the histological changes in the pulmonary arterioles. Moreover, fasudil markedly lessened the expression of Trx1 and HIF-1α, up-regulated the concentration of SOD, and lowered the levels of H2O2, MDA, and ·OH. In conclusion, fasudil is a notably attractive potential therapy for PH.
Collapse
Affiliation(s)
- Manling Liu
- Department of Pathology and Pathophysiology, Fourth Military Medical University, Xi'an 710032, PR China
| | - Yanxia Wang
- Department of Pathology and Pathophysiology, Fourth Military Medical University, Xi'an 710032, PR China
| | - Lianhe Zheng
- Centre of Orthopedic Surgery, Orthopedics Oncology Institute of Chinese PLA, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, PR China
| | - Wansong Zheng
- Department of Information, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, PR China
| | - Kai Dong
- Tianjin Chase Sun Pharmaceutical Co., Ltd., Tianjin 301700, PR China
| | - Shuai Chen
- Tianjin Chase Sun Pharmaceutical Co., Ltd., Tianjin 301700, PR China
| | - Bo Zhang
- Department of Pathology and Pathophysiology, Fourth Military Medical University, Xi'an 710032, PR China
| | - Zhichao Li
- Department of Pathology and Pathophysiology, Fourth Military Medical University, Xi'an 710032, PR China.
| |
Collapse
|
36
|
Fukumoto Y. [Role of the Rho-kinase pathway in pulmonary arterial hypertension]. Nihon Yakurigaku Zasshi 2014; 143:178-81. [PMID: 24717605 DOI: 10.1254/fpj.143.178] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
37
|
Liu CP, Dai ZK, Huang CH, Yeh JL, Wu BN, Wu JR, Chen IJ. Endothelial nitric oxide synthase-enhancing G-protein coupled receptor antagonist inhibits pulmonary artery hypertension by endothelin-1-dependent and endothelin-1-independent pathways in a monocrotaline model. Kaohsiung J Med Sci 2014; 30:267-78. [DOI: 10.1016/j.kjms.2014.02.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 12/25/2013] [Accepted: 12/31/2013] [Indexed: 11/15/2022] Open
|
38
|
Nergui S, Fukumoto Y, Do E Z, Nakajima S, Shimizu T, Ikeda S, Elias-Al-Mamun M, Shimokawa H. Role of endothelial nitric oxide synthase and collagen metabolism in right ventricular remodeling due to pulmonary hypertension. Circ J 2014; 78:1465-74. [PMID: 24705390 DOI: 10.1253/circj.cj-13-1586] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Pulmonary hypertension (PH) causes elevated right ventricular (RV) systolic pressure, RV remodeling and finally RV failure to death. However, the mechanisms of RV remodeling in PH remain to be fully elucidated. METHODS AND RESULTS RV autopsy samples from 6 PH patients with RV failure against 3 age- and sex-matched controls were first examined. Next, RV remodeling in 2 mouse models of chronic hypoxia-induced PH with endothelial nitric oxide synthase-deficient (eNOS(-/-)) and collagenase-resistant knock-in (Col(R/R)) mice were examined. In humans, RV failure was associated with RV hypertrophy, interstitial and perivascular fibrosis, decreased RV capillary density and increased macrophage recruitment. Furthermore, immunostaining showed that perivascular matrix metalloproteinase-2 was increased in PH patients with RV failure. In animals, both hypoxic eNOS(-/-) and Col(R/R) mice developed a greater extent of RV hypertrophy, perivascular remodeling and macrophage infiltration compared with wild-type mice. Capillary rarefaction was developed in hypoxic eNOS(-/-) mice, while Col(R/R) mice were able to increase their capillary density in the RV in response to chronic hypoxia. Both mouse models showed increased autophagy even under normoxic condition. CONCLUSIONS These results indicate that RV remodeling occurs early during PH development through fibrosis, perivascular remodeling, capillary rarefaction and autophagy, in which the eNOS pathway and collagen metabolism might be involved.
Collapse
Affiliation(s)
- Suvd Nergui
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Jasińska-Stroschein M, Owczarek J, Plichta P, Orszulak-Michalak D. Concurrent rho-kinase and tyrosine kinase platelet-derived growth factor inhibition in experimental pulmonary hypertension. Pharmacology 2014; 93:145-50. [PMID: 24662671 DOI: 10.1159/000360182] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 01/31/2014] [Indexed: 11/19/2022]
Abstract
BACKGROUND We hypothesized that inhibition of Rho-kinase by fasudil, together with tyrosine kinase platelet-derived growth factor (PDGF) receptor inhibition by imatinib, results in greater pulmonary arterial hypertension (PAH) improvement. METHODS The effects of such regimens were investigated on hemodynamics, right ventricle hypertrophy, PDGF and ROCK in experimental monocrotaline (MCT)-induced pulmonary hypertension. Fourteen days after MCT injection, male rats were treated orally for another 14 days with imatinib, fasudil or their combination. RESULTS Concurrent imatinib and fasudil administration reversed an MCT-induced increase in right ventricular pressure more than either drug alone and decreased right ventricle hypertrophy (right ventricle weight to left ventricle plus septum weight ratio) significantly. The simultaneous administration of fasudil and imatinib caused a further decrease in plasma PDGF-BB levels compared to either drug alone. CONCLUSIONS Inhibition of Rho-kinase by fasudil in addition to tyrosine kinase PDGF inhibition by imatinib can result in further PAH improvement. Such outcome may result from additional impact of the Rho-kinase inhibitor on the decrease in PDGF-induced effects.
Collapse
|
40
|
Targeted therapies in pulmonary arterial hypertension. Pharmacol Ther 2014; 141:172-91. [DOI: 10.1016/j.pharmthera.2013.10.002] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 08/21/2013] [Indexed: 12/21/2022]
|
41
|
Elias-Al-Mamun M, Satoh K, Tanaka SI, Shimizu T, Nergui S, Miyata S, Fukumoto Y, Shimokawa H. Combination Therapy With Fasudil and Sildenafil Ameliorates Monocrotaline-Induced Pulmonary Hypertension and Survival in Rats. Circ J 2014; 78:967-76. [DOI: 10.1253/circj.cj-13-1174] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Md. Elias-Al-Mamun
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine
| | - Kimio Satoh
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine
| | - Shin-ichi Tanaka
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine
| | - Toru Shimizu
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine
| | - Suvd Nergui
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine
| | - Satoshi Miyata
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine
| | - Yoshihiro Fukumoto
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine
| | - Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine
| |
Collapse
|
42
|
Gien J, Tseng N, Seedorf G, Roe G, Abman SH. Peroxisome proliferator activated receptor-γ-Rho-kinase interactions contribute to vascular remodeling after chronic intrauterine pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2013; 306:L299-308. [PMID: 24375792 DOI: 10.1152/ajplung.00271.2013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Peroxisome proliferator-activated receptor-γ (PPARγ) and Rho-kinase (ROCK) regulate smooth muscle cell (SMC) proliferation and contribute to vascular remodeling in adult pulmonary hypertension. Whether these pathways interact to contribute to the development of vascular remodeling in persistent pulmonary hypertension of the newborn (PPHN) remains unknown. We hypothesized that ROCK-PPARγ interactions increase SMC proliferation resulting in vascular remodeling in experimental PPHN. Pulmonary artery SMCs (PASMCs) were harvested from fetal sheep after partial ligation of the ductus arteriosus in utero (PPHN) and controls. Cell counts were performed daily for 5 days with or without PPARγ agonists and ROCK inhibition. PPARγ and ROCK protein expression/activity were measured by Western blot in normal and PPHN PASMCs. We assessed PPARγ-ROCK interactions by studying the effect of ROCK activation on PPARγ activity and PPARγ inhibition (siRNA) on ROCK activity and PASMC proliferation. At baseline, PPHN PASMC cell number was increased by 38% above controls on day 5. ROCK protein expression/activity were increased by 25 and 34% and PPARγ protein/activity decreased by 40 and 50% in PPHN PASMC. ROCK inhibition and PPARγ activation restored PPHN PASMC growth to normal values. ROCK inhibition increased PPARγ activity by 50% in PPHN PASMC, restoring PPARγ activity to normal. In normal PASMCs, ROCK activation decreased PPARγ activity and PPARγ inhibition increased ROCK activity and cell proliferation, resulting in a PPHN hyperproliferative PASMC phenotype. PPARγ-ROCK interactions regulate SMC proliferation and contribute to increased PPHN PASMC proliferation and vascular remodeling in PPHN. Restoring normal PPARγ-ROCK signaling may prevent vascular remodeling and improve outcomes in PPHN.
Collapse
Affiliation(s)
- Jason Gien
- Perinatal Research Facility, 13243 E. 23rd Ave., Mail Stop F441, Aurora, CO 80045.
| | | | | | | | | |
Collapse
|
43
|
Long-term treatment with fasudil improves bleomycin-induced pulmonary fibrosis and pulmonary hypertension via inhibition of Smad2/3 phosphorylation. Pulm Pharmacol Ther 2013; 26:635-43. [DOI: 10.1016/j.pupt.2013.07.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Revised: 07/26/2013] [Accepted: 07/27/2013] [Indexed: 11/18/2022]
|
44
|
Shimizu T, Fukumoto Y, Tanaka SI, Satoh K, Ikeda S, Shimokawa H. Crucial role of ROCK2 in vascular smooth muscle cells for hypoxia-induced pulmonary hypertension in mice. Arterioscler Thromb Vasc Biol 2013; 33:2780-91. [PMID: 24135024 DOI: 10.1161/atvbaha.113.301357] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Rho/Rho-kinase (ROCK) pathway in vascular smooth muscle cells (VSMCs) plays an important role in the pathogenesis of cardiovascular diseases, including pulmonary arterial hypertension (PAH). Rho-kinase has 2 isoforms, ROCK1 and ROCK2, with different functions in different cells; ROCK1 for circulating inflammatory cells and ROCK2 for the vasculature. In the present study, we aimed to examine whether ROCK2 in VSMC is involved in the pathogenesis of PAH. APPROACH AND RESULTS In patients with PAH, the expression of ROCK2 was increased in pulmonary arterial media and primary pulmonary arterial smooth muscle cells when compared with controls. To investigate the role of ROCK2 in VSMC, we generated VSMC-specific heterozygous ROCK2-deficient (ROCK2(+/-)) mice and VSMC-specific ROCK2-overexpressing transgenic (ROCK2-Tg) mice. The extent of hypoxia-induced pulmonary hypertension was reduced in ROCK2(+/-) mice and was enhanced in ROCK2-Tg mice compared with respective littermates. The protein expression of ROCK activity and phosphorylated extracellular signal-regulated kinase and the number of Ki67-positive proliferating cells in the lung were reduced in ROCK2(+/-) mice and were increased in ROCK2-Tg mice compared with respective littermates. In cultured mouse aortic VSMC, migration and proliferation activities were reduced in ROCK2(+/-) mice, and migration activity was increased in ROCK2-Tg mice compared with respective littermates. In addition, in primary pulmonary arterial smooth muscle cells from a patient with PAH, ROCK2 was required for migration and proliferation through ROCK and extracellular signal-regulated kinase activation. CONCLUSIONS ROCK2 in VSMC contributes to the pathogenesis of PAH.
Collapse
Affiliation(s)
- Toru Shimizu
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | | | | | | |
Collapse
|
45
|
Role of Rho-kinase and its inhibitors in pulmonary hypertension. Pharmacol Ther 2013; 137:352-64. [DOI: 10.1016/j.pharmthera.2012.12.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Accepted: 10/27/2012] [Indexed: 11/20/2022]
|
46
|
Firth AL, Choi IW, Park WS. Animal models of pulmonary hypertension: Rho kinase inhibition. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2012; 109:67-75. [PMID: 22713173 DOI: 10.1016/j.pbiomolbio.2012.05.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Revised: 05/17/2012] [Accepted: 05/21/2012] [Indexed: 10/28/2022]
Abstract
Pulmonary Hypertension is a terminology encompassing a range of etiologically different pulmonary vascular diseases. The most common is that termed pulmonary arterial hypertension or PAH; a rare but often fatal disease characterized by a mean pulmonary arterial pressure of >25 mmHg. PAH is associated with a complex etiology highlighted by core characteristics of increased pulmonary vascular resistance and elevation of mean pulmonary artery pressure. When sustained, pulmonary vascular remodeling occurs and eventually patients pass away due to right heart failure. Hypoxic pulmonary vasoconstriction is an early event occurring in pulmonary hypertension due to chronic exposure to hypoxia. While the underlying mechanisms of hypoxic pulmonary vasoconstriction may be controversial, a role for RhoA/Rho kinase mediated regulation of intracellular Ca(2+) has been recently identified. Further study suggests that RhoA may have an integral role in other pathophysiological processes such as cell proliferation and migration occurring in all forms of PH. Indeed Rho proteins are known to play essential roles in actin cytoskeleton organization in all eukaryotic cells and thus Rho and Rho-GTPases are implicated in fundamental cellular processes such as cellular proliferation, migration, adhesion, apoptosis and gene expression. This review focuses on providing an overview of the role of RhoA/Rho kinase in currently available animal models of pulmonary hypertension.
Collapse
Affiliation(s)
- Amy L Firth
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA, USA
| | | | | |
Collapse
|
47
|
Seto M. [Development of Rho kinase inhibitors for pulmonary arterial hypertension]. Nihon Yakurigaku Zasshi 2012; 139:251-255. [PMID: 22728987 DOI: 10.1254/fpj.139.251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
|
48
|
Wojciak-Stothard B, Zhao L, Oliver E, Dubois O, Wu Y, Kardassis D, Vasilaki E, Huang M, Mitchell JA, Harrington LS, Louise H, Prendergast GC, Wilkins MR. Role of RhoB in the regulation of pulmonary endothelial and smooth muscle cell responses to hypoxia. Circ Res 2012; 110:1423-34. [PMID: 22539766 DOI: 10.1161/circresaha.112.264473] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
RATIONALE RhoA and Rho kinase contribute to pulmonary vasoconstriction and vascular remodeling in pulmonary hypertension. RhoB, a protein homologous to RhoA and activated by hypoxia, regulates neoplastic growth and vasoconstriction but its role in the regulation of pulmonary vascular function is not known. OBJECTIVE To determine the role of RhoB in pulmonary endothelial and smooth muscle cell responses to hypoxia and in pulmonary vascular remodeling in chronic hypoxia-induced pulmonary hypertension. METHODS AND RESULTS Hypoxia increased expression and activity of RhoB in human pulmonary artery endothelial and smooth muscle cells, coincidental with activation of RhoA. Hypoxia or adenoviral overexpression of constitutively activated RhoB increased actomyosin contractility, induced endothelial permeability, and promoted cell growth; dominant negative RhoB or manumycin, a farnesyltransferase inhibitor that targets the vascular function of RhoB, inhibited the effects of hypoxia. Coordinated activation of RhoA and RhoB maximized the hypoxia-induced stress fiber formation caused by RhoB/mammalian homolog of Drosophila diaphanous-induced actin polymerization and RhoA/Rho kinase-induced phosphorylation of myosin light chain on Ser19. Notably, RhoB was specifically required for hypoxia-induced factor-1α stabilization and for hypoxia- and platelet-derived growth factor-induced cell proliferation and migration. RhoB deficiency in mice markedly attenuated development of chronic hypoxia-induced pulmonary hypertension, despite compensatory expression of RhoA in the lung. CONCLUSIONS RhoB mediates adaptational changes to acute hypoxia in the vasculature, but its continual activation by chronic hypoxia can accentuate vascular remodeling to promote development of pulmonary hypertension. RhoB is a potential target for novel approaches (eg, farnesyltransferase inhibitors) aimed at regulating pulmonary vascular tone and structure.
Collapse
Affiliation(s)
- Beata Wojciak-Stothard
- Centre for Pharmacology and Therapeutics, Experimental Medicine, Imperial College London, London, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Cahill E, Rowan SC, Sands M, Banahan M, Ryan D, Howell K, McLoughlin P. The pathophysiological basis of chronic hypoxic pulmonary hypertension in the mouse: vasoconstrictor and structural mechanisms contribute equally. Exp Physiol 2012; 97:796-806. [DOI: 10.1113/expphysiol.2012.065474] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
50
|
Abstract
INTRODUCTION Pulmonary arterial hypertension (PAH) is a rare disease with a complex pathogenesis. It is often associated with an increased vascular resistance, whilst in the more advanced stages there is a remodelling of the vascular walls. PAH has an intricate involvement of various signaling pathways, including the ras homolog family member A (RhoA)-Rho kinase (ROCK) axis. Currently, available therapies are not always able to significantly slow PAH progression. Therefore, newer approaches are needed. AREAS COVERED In this review, areas covered include the role of the RhoA/ROCK in PAH pathogenesis and the plausibility of its therapeutic targeting. Furthermore, various inhibitory compounds are discussed, including Fasudil and SB-772077-B. EXPERT OPINION Currently, specific RhoA/ROCK inhibition is the most promising therapeutic approach for PAH. Research has shown that it suppresses both the components of this axis and the upstream upregulating mediators. An inhaled RhoA/ROCK inhibitor may be a successful future therapy; however, further clinical trials are needed to support this approach.
Collapse
Affiliation(s)
- Sabina Antonela Antoniu
- 'Gr T Popa' University of Medicine and Pharmacy Iaşi, Pulmonary Disease University Hospital, Department of Medicine II -Pulmonary Disease, Romania.
| |
Collapse
|