1
|
Mladenov M, Lubomirov L, Grisk O, Avtanski D, Mitrokhin V, Sazdova I, Keremidarska-Markova M, Danailova Y, Nikolaev G, Konakchieva R, Gagov H. Oxidative Stress, Reductive Stress and Antioxidants in Vascular Pathogenesis and Aging. Antioxidants (Basel) 2023; 12:antiox12051126. [PMID: 37237992 DOI: 10.3390/antiox12051126] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/22/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
This review is focused on the mechanisms that regulate health, disease and aging redox status, the signal pathways that counteract oxidative and reductive stress, the role of food components and additives with antioxidant properties (curcumin, polyphenols, vitamins, carotenoids, flavonoids, etc.), and the role of the hormones irisin and melatonin in the redox homeostasis of animal and human cells. The correlations between the deviation from optimal redox conditions and inflammation, allergic, aging and autoimmune responses are discussed. Special attention is given to the vascular system, kidney, liver and brain oxidative stress processes. The role of hydrogen peroxide as an intracellular and paracrine signal molecule is also reviewed. The cyanotoxins β-N-methylamino-l-alanine (BMAA), cylindrospermopsin, microcystins and nodularins are introduced as potentially dangerous food and environment pro-oxidants.
Collapse
Affiliation(s)
- Mitko Mladenov
- Faculty of Natural Sciences and Mathematics, Institute of Biology, "Ss. Cyril and Methodius" University, P.O. Box 162, 1000 Skopje, North Macedonia
| | - Lubomir Lubomirov
- Institute of Physiology, Brandenburg Medical School Theodor Fontane, 16816 Neuruppin, Germany
| | - Olaf Grisk
- Institute of Physiology, Brandenburg Medical School Theodor Fontane, 16816 Neuruppin, Germany
| | - Dimiter Avtanski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, 110 E 59th Street, New York, NY 10003, USA
| | - Vadim Mitrokhin
- Department of Physiology, Pirogov Russian National Research Medical University, 1 Ostrovityanova Street, 117997 Moscow, Russia
| | - Iliyana Sazdova
- Department of Animal and Human Physiology, Faculty of Biology, Sofia University "St. Kliment Ohridski", 8 Dragan Tzankov Blvd., 1164 Sofia, Bulgaria
| | - Milena Keremidarska-Markova
- Department of Animal and Human Physiology, Faculty of Biology, Sofia University "St. Kliment Ohridski", 8 Dragan Tzankov Blvd., 1164 Sofia, Bulgaria
| | - Yana Danailova
- Department of Animal and Human Physiology, Faculty of Biology, Sofia University "St. Kliment Ohridski", 8 Dragan Tzankov Blvd., 1164 Sofia, Bulgaria
| | - Georgi Nikolaev
- Department of Cell and Developmental Biology, Faculty of Biology, Sofia University "St. Kliment Ohridski", 8 Dragan Tsankov Blvd., 1164 Sofia, Bulgaria
| | - Rossitza Konakchieva
- Department of Cell and Developmental Biology, Faculty of Biology, Sofia University "St. Kliment Ohridski", 8 Dragan Tsankov Blvd., 1164 Sofia, Bulgaria
| | - Hristo Gagov
- Department of Animal and Human Physiology, Faculty of Biology, Sofia University "St. Kliment Ohridski", 8 Dragan Tzankov Blvd., 1164 Sofia, Bulgaria
| |
Collapse
|
2
|
The dynamics of thrombin formation in patients with pulmonary arterial hypertension. Thromb Res 2021; 208:230-232. [PMID: 34404541 DOI: 10.1016/j.thromres.2021.07.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 06/29/2021] [Accepted: 07/26/2021] [Indexed: 11/20/2022]
|
3
|
Kurahara LH, Hiraishi K, Yamamura A, Zhang Y, Abe K, Yahiro E, Aoki M, Koga K, Yokomise H, Go T, Ishikawa K, Bo Z, Kishi H, Kobayashi S, Aoki-Shoi N, Toru S, Inoue R, Hirano K. Eicosapentaenoic acid ameliorates pulmonary hypertension via inhibition of tyrosine kinase Fyn. J Mol Cell Cardiol 2020; 148:50-62. [PMID: 32889002 DOI: 10.1016/j.yjmcc.2020.08.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 08/03/2020] [Accepted: 08/18/2020] [Indexed: 12/26/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a multifactorial disease characterized by pulmonary arterial vasoconstriction and remodeling. Src family tyrosine kinases, including Fyn, play critical roles in vascular remodeling via the inhibition of STAT3 signaling. EPA is known to inhibit Fyn kinase activity. This study investigated the therapeutic potential and underlying mechanisms of EPA and its metabolite, resolvin E1 (RvE1), to treat PAH using monocrotaline-induced PAH model rats (MCT-PAH), human pulmonary artery endothelial cells (HPAECs), and human pulmonary artery smooth muscle cells (HPASMCs). Administration of EPA 1 and 2 weeks after MCT injection both ameliorated right ventricular hypertrophy, remodeling and dysfunction, and medial wall thickening of the pulmonary arteries and prolonged survival in MCT-PAH rats. EPA attenuated the enhanced contractile response to 5-hydroxytryptamine in isolated pulmonary arteries of MCT-PAH rats. Mechanistically, the treatment with EPA and RvE1 or the introduction of dominant-negative Fyn prevented TGF-β2-induced endothelial-to-mesenchymal transition and IL-6-induced phosphorylation of STAT3 in cultured HPAECs. EPA and RvE1 suppressed Src family kinases' activity as evaluated by their phosphorylation status in cultured HPAECs and HPASMCs. EPA and RvE1 suppressed vasocontraction of rat and human PA. Furthermore, EPA and RvE1 inhibited the enhanced proliferation and activity of Src family kinases in HPASMCs derived from patients with idiopathic PAH. EPA ameliorated PAH's pathophysiology by mitigating vascular remodeling and vasoconstriction, probably inhibiting Src family kinases, especially Fyn. Thus, EPA is considered a potent therapeutic agent for the treatment of PAH.
Collapse
Affiliation(s)
- Lin Hai Kurahara
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University, Kita-gun, Miki-cho, Kagawa, Japan; Department of Physiology, Fukuoka University School of Medicine, Fukuoka, Japan.
| | - Keizo Hiraishi
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University, Kita-gun, Miki-cho, Kagawa, Japan; Department of Physiology, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Aya Yamamura
- Department of Physiology, Aichi Medical University, Nagakute, Japan
| | - Ying Zhang
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Yamaguchi University, Minami-Kogushi, Ube, Yamaguchi, Japan
| | - Kohtaro Abe
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Eiji Yahiro
- Fukuoka University Medical Education Center, Fukuoka University School of Medicine, Fukuoka University, Fukuoka, Japan
| | - Mikiko Aoki
- Department of Pathology, School of Medicine, Fukuoka University, Fukuoka, Japan
| | - Kaori Koga
- Department of Pathology, School of Medicine, Fukuoka University, Fukuoka, Japan
| | - Hiroyasu Yokomise
- Department of General Thoracic Surgery, Faculty of Medicine, Kagawa University, Kita-gun, Miki-cho, Kagawa, Japan
| | - Tetsuhiko Go
- Department of General Thoracic Surgery, Faculty of Medicine, Kagawa University, Kita-gun, Miki-cho, Kagawa, Japan
| | - Kaori Ishikawa
- Department of General Medicine, Faculty of Medicine, Kagawa University, Kita-gun, Miki-cho, Kagawa, Japan
| | - Zhang Bo
- Department of Biochemistry, Fukuoka University School of Medicine, Fukuoka University, Fukuoka, Japan
| | - Hiroko Kishi
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Yamaguchi University, Minami-Kogushi, Ube, Yamaguchi, Japan
| | - Sei Kobayashi
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Yamaguchi University, Minami-Kogushi, Ube, Yamaguchi, Japan
| | - Narumi Aoki-Shoi
- Department of Chemistry, Faculty of Science, Fukuoka University, Fukuoka, Japan
| | - Satoh Toru
- Division of Cardiology, Department of Medicine, Kyorin University School of Medicine, Tokyo, Japan
| | - Ryuji Inoue
- Department of Physiology, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Katsuya Hirano
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University, Kita-gun, Miki-cho, Kagawa, Japan
| |
Collapse
|
4
|
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: 2.8] [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
|
5
|
Wang R, Wang M, He S, Sun G, Sun X. Targeting Calcium Homeostasis in Myocardial Ischemia/Reperfusion Injury: An Overview of Regulatory Mechanisms and Therapeutic Reagents. Front Pharmacol 2020; 11:872. [PMID: 32581817 PMCID: PMC7296066 DOI: 10.3389/fphar.2020.00872] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 05/27/2020] [Indexed: 12/17/2022] Open
Abstract
Calcium homeostasis plays an essential role in maintaining excitation–contraction coupling (ECC) in cardiomyocytes, including calcium release, recapture, and storage. Disruption of calcium homeostasis may affect heart function, leading to the development of various heart diseases. Myocardial ischemia/reperfusion (MI/R) injury may occur after revascularization, which is a treatment used in coronary heart disease. MI/R injury is a complex pathological process, and the main cause of increased mortality and disability after treatment of coronary heart disease. However, current methods and drugs for treating MI/R injury are very scarce, not ideal, and have limitations. Studies have shown that MI/R injury can cause calcium overload that can further aggravate MI/R injury. Therefore, we reviewed the effects of critical calcium pathway regulators on MI/R injury and drew an intuitive diagram of the calcium homeostasis pathway. We also summarized and analyzed calcium pathway-related or MI/R drugs under research or marketing by searching Therapeutic Target and PubMed Databases. The data analysis showed that six drugs and corresponding targets are used to treat MI/R injury and involved in calcium signaling pathways. We emphasize the relevance of further detailed investigation of MI/R injury and calcium homeostasis and the therapeutic role of calcium homeostasis in MI/R injury, which bridges basic research and clinical applications of MI/R injury.
Collapse
Affiliation(s)
- Ruiying Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, China
| | - Min Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, China
| | - Shuaibing He
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, China
| | - Guibo Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
6
|
Gorr MW, Sriram K, Muthusamy A, Insel PA. Transcriptomic analysis of pulmonary artery smooth muscle cells identifies new potential therapeutic targets for idiopathic pulmonary arterial hypertension. Br J Pharmacol 2020; 177:3505-3518. [PMID: 32337710 DOI: 10.1111/bph.15074] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/25/2020] [Accepted: 04/17/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND PURPOSE Pulmonary arterial hypertension (PAH, type 1 pulmonary hypertension) has a 3-year survival of ~50% and is in need of new, effective therapies. In PAH, remodelling of the pulmonary artery (PA) increases pulmonary vascular resistance and can result in right heart dysfunction and failure. Genetic mutations can cause PAH but it can also be idiopathic (IPAH). Enhanced contractility and proliferation of PA smooth muscle cells (PASMCs) are key contributors to the pathophysiology of PAH, but the underlying mechanisms are not well understood. EXPERIMENTAL APPROACH We utilized RNA-sequencing (RNA-seq) of IPAH and control patient-derived PASMCs as an unbiased approach to define differentially expressed (DE) genes that may identify new biology and potential therapeutic targets. KEY RESULTS Analysis of DE genes for shared gene pathways revealed increases in genes involved in cell proliferation and mitosis and decreases in a variety of gene sets, including response to cytokine signalling. ADGRG6/GPR126, an adhesion G protein-coupled receptor (GPCR), was increased in IPAH-PASMCs compared to control-PASMCs. Increased expression of this GPCR in control-PASMCs decreased their proliferation; siRNA knockdown of ADGRG6/GPR126 in IPAH-PASMCs tended to increase proliferation. CONCLUSION AND IMPLICATIONS These data provide insights regarding the expression of current and experimental PAH drug targets, GPCRs and GPCR-related genes as potentially new therapeutic targets in PAH-PASMCs. Overall, the findings identify genes and pathways that may contribute to IPAH-PASMC function and suggest that ADGRG6/GPR126 is a novel therapeutic target for IPAH.
Collapse
Affiliation(s)
- Matthew W Gorr
- Department of Pharmacology, University of California, San Diego, La Jolla, California, USA.,Colleges of Nursing and Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Krishna Sriram
- Department of Pharmacology, University of California, San Diego, La Jolla, California, USA
| | - Abinaya Muthusamy
- Department of Pharmacology, University of California, San Diego, La Jolla, California, USA
| | - Paul A Insel
- Department of Pharmacology, University of California, San Diego, La Jolla, California, USA.,Department of Medicine, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
7
|
Chikuda M, Sato K. Effects of dexmedetomidine on porcine pulmonary artery vascular smooth muscle. BMC Anesthesiol 2019; 19:176. [PMID: 31510933 PMCID: PMC6740015 DOI: 10.1186/s12871-019-0843-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 08/26/2019] [Indexed: 11/23/2022] Open
Abstract
Background The α2-receptor agonist dexmedetomidine (Dex) has been shown to produce sedative and analgesic effects not only with systemic administration but also when administered in the extradural space and around peripheral nerves. The effects and mechanism of action of Dex on pulmonary arteries, however, have not been determined. This study therefore aimed to investigate the effect of Dex on pulmonary arterial vascular smooth muscle by evaluating changes in isometric contraction tension. We then attempted to determine the effects of Dex on depolarization stimulation and receptor stimulation. Methods Endothelium-denuded porcine pulmonary arteries were sliced into 2- to 3-mm rings. We then exposed them to certain substances at various concentrations under different conditions of baseline stimulation (with KCl, adrenaline, caffeine, or histamine) and to α2-receptor stimulants or antagonists, or α1-receptor antagonists (imidazoline, yohimbine, rauwolscine, prazosin), and different conditions of Ca2+ depletion of the intracellular reservoir or extracellular stores. We measured the changes in isometric contraction tension with each addition or change in conditions. Results Dex enhanced the contraction induced by high-concentration KCl stimulation. Dex-induced enhancement of contraction induced by high-concentration KCl was completely suppressed by yohimbine and rauwolscine, which are α2-receptor antagonists, but not by prazosin. Dex, imidazoline, yohimbine, and rauwolscine reduced the increases in contraction tension induced by the receptor stimulant adrenaline. Dex suppressed the adrenaline-induced increases in contraction tension after depletion of the Ca2+ reservoir. In the absence of extracellular Ca2+, Dex suppressed the adrenaline- and histamine-induced increases in contraction tension but did not affect caffeine-induced increases. Conclusions Dex-enhanced, high-concentration KCl-induced contraction was mediated by α2-receptors. Adrenaline-induced contraction was suppressed by the α2-receptor stimulant Dex and α2-receptor antagonists yohimbine and rauwolscine, suggesting that the effect of Dex on adrenaline-induced contraction is attributable to its α2-receptor-blocking action. Dex inhibited receptor-activated Ca2+ channels and phosphatidylinositol-1,4,5-triphosphate-induced Ca2+ release but not Ca2+-induced Ca2+ release.
Collapse
Affiliation(s)
- Mami Chikuda
- Division of Dental Anesthesiology, Department of Reconstructive Oral and Maxillofacial Surgery, School of Dentistry, Iwate Medical University, 1-3-27 Chuo-dori, Morioka, Iwate, 020-8505, Japan
| | - Kenichi Sato
- Division of Dental Anesthesiology, Department of Reconstructive Oral and Maxillofacial Surgery, School of Dentistry, Iwate Medical University, 1-3-27 Chuo-dori, Morioka, Iwate, 020-8505, Japan.
| |
Collapse
|
8
|
Satoh K, Kamada S, Kumagai M, Sato M, Kuji A, Joh S. Effect of lidocaine on swine lingual and pulmonary arteries. J Anesth 2015; 29:529-34. [PMID: 25596945 DOI: 10.1007/s00540-014-1965-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 12/14/2014] [Indexed: 10/24/2022]
Abstract
PURPOSE Lidocaine has a biphasic action on smooth muscle of peripheral blood vessels, with vasoconstriction at low concentrations and vasodilation at higher concentrations. Many in vivo studies have demonstrated the effects of lidocaine on aortic or coronary arteries in several animals, but there are few reports about the effect on peripheral vessels. This study was designed to investigate the direct effects of lidocaine on peripheral vessels, namely swine lingual and pulmonary arterial rings. METHODS Swine lingual artery and pulmonary artery segments, about 2-3 mm in diameter, were cut into 3-mm-long rings, and the lumen surface was gently rubbed to remove the endothelium. Isometric tension was measured using a displacement transducer and recorded. After a stable constriction was developed with 5 µM noradrenaline, 5 µM noradrenaline containing lidocaine (0.5, 1.0, 10, 20, 50 or 100 µg/ml) was perfused for 5 min, and then all drug perfusion was stopped. The strength of any isometric tension during an experiment was normalized to the strength of the isometric tension immediately before lidocaine perfusion, and expressed as a percentage. RESULTS Lidocaine elicited a concentration-dependent biphasic response of lingual and pulmonary arterial rings. The lidocaine concentration at 1 µg/ml caused mild contraction. Dilation occurred at 10 µg/ml and increased with increasing dose. CONCLUSIONS Lidocaine-induced vasoconstriction of swine lingual and pulmonary smooth muscle may occur at low concentration when lidocaine is infiltrated into the oral submucosa or administered intravenously for the treatment of ventricular arrhythmia.
Collapse
Affiliation(s)
- Kenichi Satoh
- Division of Dental Anesthesiology, Department of Reconstructive Oral Maxillofacial Surgery, School of Dentistry, Iwate Medical University, 1-3-27 Chuo-dori, Morioka, Iwate, 020-8505, Japan,
| | | | | | | | | | | |
Collapse
|
9
|
Antoniak S, Sparkenbaugh E, Pawlinski R. Tissue factor, protease activated receptors and pathologic heart remodelling. Thromb Haemost 2014; 112:893-900. [PMID: 25104210 DOI: 10.1160/th14-03-0243] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 05/30/2014] [Indexed: 12/13/2022]
Abstract
Tissue factor is the primary initiator of coagulation cascade and plays an essential role in haemostasis and thrombosis. In addition, tissue factor and coagulation proteases contribute to many cellular responses via activation of protease activated receptors. The heart is an organ with high levels of constitutive tissue factor expression. This review focuses on the role of tissue factor, coagulation proteases and protease activated receptors in heart haemostasis and the pathological heart remodelling associated with myocardial infarction, viral myocarditis and hypertension.
Collapse
Affiliation(s)
| | | | - Rafal Pawlinski
- Rafal Pawlinski, PhD, Division of Hematology/Oncology, Department of Medicine, McAllister Heart Institute, University of North Carolina, 320A Mary Ellen Jones Bldg, 98 Manning Drive, Chapel Hill, NC 27599, USA, Tel: 919 843 8387, Fax: 919 843 4896, E-mail:
| |
Collapse
|
10
|
Mukherjee S, Trice J, Shinde P, Willis RE, Pressley TA, Perez-Zoghbi JF. Ca2+ oscillations, Ca2+ sensitization, and contraction activated by protein kinase C in small airway smooth muscle. ACTA ACUST UNITED AC 2013; 141:165-78. [PMID: 23359281 PMCID: PMC3557311 DOI: 10.1085/jgp.201210876] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Protein kinase C (PKC) has been implicated in the regulation of smooth muscle cell (SMC) contraction and may contribute to airway hyperresponsiveness. Here, we combined optical and biochemical analyses of mouse lung slices to determine the effects of PKC activation on Ca2+ signaling, Ca2+ sensitivity, protein phosphorylation, and contraction in SMCs of small intrapulmonary airways. We found that 10 µM phorbol-12-myristate-13-acetate or 1 µM phorbol 12,13-dibutyrate induced repetitive, unsynchronized, and transient contractions of the SMCs lining the airway lumen. These contractions were associated with low frequency Ca2+ oscillations in airway SMCs that resulted from Ca2+ influx through L-type voltage-gated Ca2+ channels and the subsequent release of Ca2+ from intracellular stores through ryanodine receptors. Phorbol ester stimulation of lung slices in which SMC intracellular Ca2+ concentration ([Ca2+]i) was “clamped” at a high concentration induced strong airway contraction, indicating that PKC mediated sensitization of the contractile response to [Ca2+]i. This Ca2+ sensitization was accompanied by phosphorylation of both the PKC-potentiated PP1 inhibitory protein of 17 kD (CPI-17) and the regulatory myosin light chain. Thrombin, like the phorbol esters, induced a strong Ca2+ sensitization that was inhibited by the PKC inhibitor GF-109203X and also potentiated airway contraction to membrane depolarization with KCl. In conclusion, we suggest that PKC activation in small airways leads to both the generation of Ca2+ oscillations and strong Ca2+ sensitization; agents associated with airway inflammation, such as thrombin, may activate this pathway to sensitize airway smooth muscle to agonists that cause membrane depolarization and Ca2+ entry and induce airway hyperresponsiveness.
Collapse
Affiliation(s)
- Seema Mukherjee
- Department of Cell Physiology and Molecular Biophysics, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | | | | | | | | | | |
Collapse
|
11
|
Nickel KF, Laux V, Heumann R, von Degenfeld G. Thrombin has biphasic effects on the nitric oxide-cGMP pathway in endothelial cells and contributes to experimental pulmonary hypertension. PLoS One 2013; 8:e63504. [PMID: 23785394 PMCID: PMC3681801 DOI: 10.1371/journal.pone.0063504] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Accepted: 04/06/2013] [Indexed: 01/29/2023] Open
Abstract
Background A potential role for coagulation factors in pulmonary arterial hypertension has been recently described, but the mechanism of action is currently not known. Here, we investigated the interactions between thrombin and the nitric oxide-cGMP pathway in pulmonary endothelial cells and experimental pulmonary hypertension. Principal Findings Chronic treatment with the selective thrombin inhibitor melagatran (0.9 mg/kg daily via implanted minipumps) reduced right ventricular hypertrophy in the rat monocrotaline model of experimental pulmonary hypertension. In vitro, thrombin was found to have biphasic effects on key regulators of the nitric oxide-cGMP pathway in endothelial cells (HUVECs). Acute thrombin stimulation led to increased expression of the cGMP-elevating factors endothelial nitric oxide synthase (eNOS) and soluble guanylate cyclase (sGC) subunits, leading to increased cGMP levels. By contrast, prolonged exposition of pulmonary endothelial cells to thrombin revealed a characteristic pattern of differential expression of the key regulators of the nitric oxide-cGMP pathway, in which specifically the factors contributing to cGMP elevation (eNOS and sGC) were reduced and the cGMP-hydrolyzing PDE5 was elevated (qPCR and Western blot). In line with the differential expression of key regulators of the nitric oxide-cGMP pathway, a reduction of cGMP by prolonged thrombin stimulation was found. The effects of prolonged thrombin exposure were confirmed in endothelial cells of pulmonary origin (HPAECs and HPMECs). Similar effects could be induced by activation of protease-activated receptor-1 (PAR-1). Conclusion These findings suggest a link between thrombin generation and cGMP depletion in lung endothelial cells through negative regulation of the nitric oxide-cGMP pathway, possibly mediated via PAR-1, which could be of relevance in pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Katrin F. Nickel
- Cardiology Research, Bayer HealthCare AG, Wuppertal, Germany
- Department of Molecular Medicine and Surgery, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Volker Laux
- Cardiology Research, Bayer HealthCare AG, Wuppertal, Germany
| | - Rolf Heumann
- Biochemistry II – Molecular Neurobiochemistry, Ruhr-University Bochum, Bochum, Germany
| | - Georges von Degenfeld
- Common Mechanism Research, Bayer HealthCare AG, Wuppertal, Germany, and Institute for Research in Operative Medicine, University of Witten/Herdecke, Cologne, Germany
- * E-mail:
| |
Collapse
|
12
|
Abstract
Currently available antiplatelet agents have shown improved short- and long-term clinical outcomes but are associated with increased bleeding risk, and the rates of recurrent ischemic events still remain high. Selective inhibition of protease-activated receptor-1 for thrombin represents a potential novel strategy to reduce ischemic events without increasing the risk of bleeding. Two protease-activated receptor-1 antagonists are currently being evaluated in clinical trials: SCH 530348 and E5555. Results of phase II trials have shown that SCH 530348, when added to standard antiplatelet therapy, was well tolerated and not associated with increased bleeding risk. Two large-scale phase III trials assessing the efficacy of SCH 530348 in addition to the standard of care are currently ongoing. This review provides an outline of the current status of understanding on platelet thrombin-receptor antagonist SCH 530348, focusing on its pharmacologic properties and clinical development.
Collapse
|
13
|
Maki J, Hirano M, Hoka S, Kanaide H, Hirano K. Involvement of reactive oxygen species in thrombin-induced pulmonary vasoconstriction. Am J Respir Crit Care Med 2010; 182:1435-44. [PMID: 20639439 DOI: 10.1164/rccm.201002-0255oc] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
RATIONALE Pulmonary vascular thrombosis and thrombotic arteriopathy are common pathological findings in pulmonary arterial hypertension. Thrombin may thus play an important role in the pathogenesis and pathophysiology of pulmonary arterial hypertension. OBJECTIVES The present study aimed to elucidate the contractile effect of thrombin in the pulmonary artery and clarify its underlying mechanisms. METHODS The changes in cytosolic Ca²(+) concentrations ([Ca²(+)](i)), 20-kD myosin light chain (MLC20) phosphorylation, and contraction were monitored in the isolated porcine pulmonary artery. The production of reactive oxygen species (ROS) was evaluated by fluorescence imaging. MEASUREMENTS AND MAIN RESULTS In the presence of extracellular Ca²(+), thrombin induced a sustained contraction accompanied by an increase in [Ca²(+)](i) and the phosphorylation of MLC20. In the absence of extracellular Ca²(+), thrombin induced a contraction without either [Ca²(+)](i) elevation or MLC20 phosphorylation. This Ca²(+)- and MLC20 phosphorylation-independent contraction was mimicked by hydrogen peroxide and inhibited by N-acetyl cysteine. Fluorescence imaging revealed thrombin to induce the production of ROS. A Rho-kinase inhibitor, Y27632, inhibited not only the thrombin-induced Ca²(+)- and MLC20 phosphorylation-dependent contraction, but also the Ca²(+)- and MLC20 phosphorylation-independent contraction and the ROS production. These effects of thrombin were mimicked by a proteinase-activated receptor 1 (PAR₁)-activating peptide. CONCLUSIONS This study elucidated the Ca²(+)- and MLC20 phosphorylation-independent ROS-mediated noncanonical mechanism as well as Ca²(+)- and MLC20 phosphorylation-dependent canonical mechanism that are involved in the thrombin-induced PAR₁-mediated pulmonary vasoconstriction. Rho-kinase was suggested to play multiple roles in the development of thrombin-induced pulmonary vasoconstriction.
Collapse
Affiliation(s)
- Jun Maki
- Research Institute of Angiocardiology, Kyushu University, Higashi-ku, Fukuoka, Japan
| | | | | | | | | |
Collapse
|