1
|
Rahimi Kakavandi N, Asadi T, Hooshangi Shayesteh MR, Baeeri M, Rahimifard M, Baghaei A, Noruzi M, Sharifzadeh M, Abdollahi M. The electrocardiographic, hemodynamic, echocardiographic, and biochemical evaluation of treatment with edaravone on acute cardiac toxicity of aluminum phosphide. Front Pharmacol 2022; 13:1032941. [PMID: 36278198 PMCID: PMC9581139 DOI: 10.3389/fphar.2022.1032941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Aluminum phosphide (AlP) poisoning can be highly fatal due to its severe toxicity to the heart. Based on the evidence, edaravone (EDA) has protective effects on various pathological conditions of the heart. This research aimed to examine the potential protective effects of EDA on AlP-induced cardiotoxicity in rats. The rats were divided into six groups, including almond oil (control), normal saline, AlP (LD50), and AlP + EDA (20, 30, and 45 mg/kg). Thirty minutes following AlP poisoning, the electrocardiographic (ECG), blood pressure (BP), and heart rate (HR) parameters were examined for 180 min. The EDA was injected 60 min following the AlP poisoning intraperitoneally. Also, 24 h after poisoning, echocardiography was carried out to evaluate the ejection fraction (EF), stroke volume (SV), and cardiac output (CO). The biochemical and molecular parameters, such as the activities of the mitochondrial complexes, reactive oxygen species (ROS), apoptosis and necrosis, and troponin I and lactate levels, were also examined after 12 and 24 h in the heart tissue. According to the results, AlP-induced ECG abnormalities, decrease in blood pressure, heart rate, SV, EF%, and CO were significantly improved with EDA at doses of 30 and 45 mg/kg. Likewise, EDA significantly improved complex I and IV activity, apoptosis and necrosis, ROS, troponin I, and lactate levels following AlP-poisoning (p < 0.05). Also, the mean survival time was increased following EDA treatment, which can be attributed to the EDA's protective effects against diverse underlying mechanisms of phosphine-induced cardiac toxicity. These findings suggest that EDA, by ameliorating heart function and modulating mitochondrial activity, might relieve AlP-induced cardiotoxicity. Nonetheless, additional investigations are required to examine any potential clinical advantages of EDA in this toxicity.
Collapse
Affiliation(s)
- Nader Rahimi Kakavandi
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Tayebeh Asadi
- Health and Environment Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | | | - Maryam Baeeri
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mahban Rahimifard
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Amir Baghaei
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Alborz University of Medical Sciences, Karaj, Iran
| | - Marzieh Noruzi
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mohammad Sharifzadeh
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mohammad Abdollahi
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
2
|
Khushbu, Jindal R. Cyclodextrin mediated controlled release of edaravone from pH-responsive sodium alginate and chitosan based nanocomposites. Int J Biol Macromol 2022; 202:11-25. [PMID: 35031316 DOI: 10.1016/j.ijbiomac.2022.01.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 12/09/2021] [Accepted: 01/01/2022] [Indexed: 12/18/2022]
Abstract
The objective of the study is to enhance the aqueous solubility and stability of edaravone, a free radical scavenger drug. Inclusion complexes of edaravone with β-cyclodextrin were prepared by microwave irradiation and physical mixture method and confirmation of inclusion complexes were investigated by different analytical techniques such as FT-IR, ROESY, DSC, and 1H NMR. pH-sensitive nanocomposites based on chitosan (CH), sodium alginate (ALG), and bentonite (BN) were synthesized. To get the maximum percentage swelling different reaction parameters that are responsible for the synthesis of the nanocomposite were optimized and characterized by various techniques such as FESEM, EDS, XRD, and FT-IR. To regulate the drug delivery, inclusion complexes were directly loaded into the CH/ALG hydrogel, and CH/ALG/BN nanocomposite and release studies were evaluated at different pH environments. The solubility of edaravone was investigated by phase solubility and the graph results in a typical AL type behavior, suggesting the formation of a 1:1 stoichiometry inclusion complex. The comparative evaluation of drug release was explored by kinetic models. Controlled release of drug was achieved from CH/ALG/BN nanocomposite in comparison to CH/ALG hydrogel. The exploratory kinetic investigation revealed that β-CD plays a critical role in the drug release process by influencing polymer relaxation, resulting in slow release.
Collapse
Affiliation(s)
- Khushbu
- Polymer and Nanomaterial Lab, Department of Chemistry, Dr BR Ambedkar National Institute of Technology, Jalandhar 144011, Punjab, India.
| | - Rajeev Jindal
- Polymer and Nanomaterial Lab, Department of Chemistry, Dr BR Ambedkar National Institute of Technology, Jalandhar 144011, Punjab, India.
| |
Collapse
|
3
|
Wang S, Dong J, Li L, Wu R, Xu L, Ren Y, Hu X. Exosomes derived from miR-129-5p modified bone marrow mesenchymal stem cells represses ventricular remolding of mice with myocardial infarction. J Tissue Eng Regen Med 2021; 16:177-187. [PMID: 34814233 DOI: 10.1002/term.3268] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 11/14/2021] [Accepted: 11/15/2021] [Indexed: 12/23/2022]
Abstract
Myocardial infraction (MI) is a severe disease with great mortality. Mesenchymal stem cells-derived exosomes display protection against MI. MicroRNA-129-5p was reported to exert anti-inflammation activity by targeting high mobility group box 1 (HMGB1). In the present study, the effects of MSCs derived exosomes overexpressing miR-129-5p on MI were evaluated. Bone marrow mesenchymal stem cells (BMSCs) were transfected with miR-129-5p for exosomes isolation. Myocardial infraction mice model was established and administrated exosomes overexpressing miR-129-5p. The cardiac function, expression of HMGB1, inflammatory cytokines, apoptosis and fibrosis in heart tissues were measured. miR-129-5p inhibited HMGB1 expression in BMSCs. Myocardial infraction mice treated with exosomes overexpressing miR-129-5p had enhanced cardiac function and decreased expression of HMGB1 and production of inflammatory cytokines. Exosomes overexpressing miR-129-5p further prevented apoptosis and fibrosis. Exosome-mediated transfer of miR-129-5p suppressed inflammation in MI mice by targeting HMGB1.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Cardiology, Shijiazhuang People's Hospital, Shijiazhuang, Hebei, China
| | - Jingjie Dong
- Department of Cardiology, Shijiazhuang People's Hospital, Shijiazhuang, Hebei, China
| | - Liu Li
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Rubing Wu
- Department of Cardiology, Shijiazhuang People's Hospital, Shijiazhuang, Hebei, China
| | - Lei Xu
- Department of Cardiology, Shijiazhuang People's Hospital, Shijiazhuang, Hebei, China
| | - Yanchun Ren
- Department of Cardiology, Shijiazhuang People's Hospital, Shijiazhuang, Hebei, China
| | - Xitian Hu
- Department of Cardiology, Shijiazhuang People's Hospital, Shijiazhuang, Hebei, China
| |
Collapse
|
4
|
Gianazza E, Brioschi M, Martinez Fernandez A, Casalnuovo F, Altomare A, Aldini G, Banfi C. Lipid Peroxidation in Atherosclerotic Cardiovascular Diseases. Antioxid Redox Signal 2021; 34:49-98. [PMID: 32640910 DOI: 10.1089/ars.2019.7955] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Significance: Atherosclerotic cardiovascular diseases (ACVDs) continue to be a primary cause of mortality worldwide in adults aged 35-70 years, occurring more often in countries with lower economic development, and they constitute an ever-growing global burden that has a considerable socioeconomic impact on society. The ACVDs encompass diverse pathologies such as coronary artery disease and heart failure (HF), among others. Recent Advances: It is known that oxidative stress plays a relevant role in ACVDs and some of its effects are mediated by lipid oxidation. In particular, lipid peroxidation (LPO) is a process under which oxidants such as reactive oxygen species attack unsaturated lipids, generating a wide array of oxidation products. These molecules can interact with circulating lipoproteins, to diffuse inside the cell and even to cross biological membranes, modifying target nucleophilic sites within biomolecules such as DNA, lipids, and proteins, and resulting in a plethora of biological effects. Critical Issues: This review summarizes the evidence of the effect of LPO in the development and progression of atherosclerosis-based diseases, HF, and other cardiovascular diseases, highlighting the role of protein adduct formation. Moreover, potential therapeutic strategies targeted at lipoxidation in ACVDs are also discussed. Future Directions: The identification of valid biomarkers for the detection of lipoxidation products and adducts may provide insights into the improvement of the cardiovascular risk stratification of patients and the development of therapeutic strategies against the oxidative effects that can then be applied within a clinical setting.
Collapse
Affiliation(s)
- Erica Gianazza
- Proteomics Unit, Monzino Cardiology Center IRCCS, Milan, Italy
| | - Maura Brioschi
- Proteomics Unit, Monzino Cardiology Center IRCCS, Milan, Italy
| | | | | | | | - Giancarlo Aldini
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Cristina Banfi
- Proteomics Unit, Monzino Cardiology Center IRCCS, Milan, Italy
| |
Collapse
|
5
|
Zhao X, Zhang E, Ren X, Bai X, Wang D, Bai L, Luo D, Guo Z, Wang Q, Yang J. Edaravone alleviates cell apoptosis and mitochondrial injury in ischemia-reperfusion-induced kidney injury via the JAK/STAT pathway. Biol Res 2020; 53:28. [PMID: 32620154 PMCID: PMC7333427 DOI: 10.1186/s40659-020-00297-0] [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: 02/27/2020] [Accepted: 06/26/2020] [Indexed: 01/31/2023] Open
Abstract
Background Kidney ischemia–reperfusion injury is a common pathophysiological phenomenon in the clinic. A large number of studies have found that the tyrosine protein kinase/signal transducer and activator of transcription (JAK/STAT) pathway is involved in the development of a variety of kidney diseases and renal protection associated with multiple drugs. Edaravone (EDA) is an effective free radical scavenger that has been used clinically for the treatment of postischemic neuronal injury. This study aimed to identify whether EDA improved kidney function in rats with ischemia–reperfusion injury by regulating the JAK/STAT pathway and clarify the underlying mechanism. Methods Histomorphological analysis was used to assess pathological kidney injury, and mitochondrial damage was observed by transmission electron microscopy. Terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling (TUNEL) staining was performed to detect tubular epithelial cell apoptosis. The expression of JAK2, P-JAK2, STAT3, P-STAT3, STAT1, P-STAT1, BAX and Bcl-2 was assessed by western blotting. Mitochondrial function in the kidney was assessed by mitochondrial membrane potential (ΔΨm) measurement. Results The results showed that EDA inhibited the expression of p-JAK2, p-STAT3 and p-STAT1, accompanied by downregulation of the expression of Bax and caspase-3, and significantly ameliorated kidney damage caused by ischemia–reperfusion injury (IRI). Furthermore, the JC-1 dye assay showed that edaravone attenuated ischemia–reperfusion-induced loss of kidney ΔΨm. Conclusion Our findings indicate that EDA protects against kidney damage caused by ischemia–reperfusion through JAK/STAT signaling, inhibiting apoptosis and improving mitochondrial injury.
Collapse
Affiliation(s)
- Xiaoying Zhao
- Department of Anesthesiology, Second Hospital of Shanxi Medical University, Taiyuan, China.
| | - Erfei Zhang
- Department of Anesthesiology, The Affiliated Hospital of Yan'an University, Yan'an, China
| | - Xiaofen Ren
- Department of Anesthesiology, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaoli Bai
- Department of Anesthesiology, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Dongming Wang
- Department of Anesthesiology, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Ling Bai
- Department of Anesthesiology, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Danlei Luo
- Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Zheng Guo
- Department of Anesthesiology, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Qiang Wang
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jianxin Yang
- Department of Anesthesiology, Second Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
6
|
Liu D, Cao Y, Zhang X, Peng C, Tian X, Yan C, Liu Y, Liu M, Han Y. Chemokine CC-motif ligand 2 participates in platelet function and arterial thrombosis by regulating PKCα-P38MAPK-HSP27 pathway. Biochim Biophys Acta Mol Basis Dis 2018; 1864:2901-2912. [PMID: 29864522 DOI: 10.1016/j.bbadis.2018.05.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 05/10/2018] [Accepted: 05/30/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND Studies indicate that chemokine CC-motif ligand 2 (CCL2) is involved in inflammation and atherosclerosis. However, the roles and mechanisms of CCL2 on platelet function and arterial thrombosis are unknown. METHODS The expressions of CCL2 or CCR2 in the plasma, platelets and coronary thrombus of ST-elevated myocardial infarction (STEMI) patients were examined by ELISA, Western blot, immunohistochemistry and immunofluorescence. The roles of CCL2 on platelet aggregation, activation and secretion were examined by light transmission aggregometry, flow cytometry and ELISA. RESULTS The expressions of CCL2 or CCR2 in the plasma or platelets of STEMI patients with platelet high response were higher than those with platelet normal response; In vitro, exogenous recombinant human CCL2 markedly increased platelet aggregation, activation and granule secretion, which were abolished by CCL2 neutralizing antibody or CCR2 inhibiter. CCL2 increased the phosphorylation levels of PKCα (Thr638), P38MAPK (Thr180/Tyr182) and HSP27 (S78/S82) in human platelets, which were abrogated by PKCα inhibitor (RO 318220) or P38MAPK inhibitor (SB 203580). RO 318220 or SB 203580 diminished CCL2-induced platelet function. In CCL2-/- mice, platelet aggregation and secretion were attenuated; the phosphorylation of PKCα, P38MAPK and HSP27 were decreased. In a carotid arterial thrombus mouse model, CCL2-/- mice displayed a significantly extended carotid artery occlusion time compared with wild type. CONCLUSIONS CCL2 played important roles in regulating platelet function and arterial thrombosis through the PKCα-P38MAPK-HSP27 pathway, which might provide theoretical basis for searching new antiplatelet drugs and the treatment for cardiovascular diseases.
Collapse
Affiliation(s)
- Dan Liu
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Shenyang Military Region, Shenyang, China
| | - Yu Cao
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Shenyang Military Region, Shenyang, China
| | - Xiaolin Zhang
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Shenyang Military Region, Shenyang, China
| | - Chengfei Peng
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Shenyang Military Region, Shenyang, China
| | - Xiaoxiang Tian
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Shenyang Military Region, Shenyang, China
| | - Chenghui Yan
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Shenyang Military Region, Shenyang, China
| | - Yanxia Liu
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Shenyang Military Region, Shenyang, China
| | - Meili Liu
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Shenyang Military Region, Shenyang, China
| | - Yaling Han
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Shenyang Military Region, Shenyang, China.
| |
Collapse
|
7
|
Abstract
Each month, subscribers to The Formulary Monograph Service receive 5 to 6 well-documented monographs on drugs that are newly released or are in late phase 3 trials. The monographs are targeted to Pharmacy & Therapeutics Committees. Subscribers also receive monthly 1-page summary monographs on agents that are useful for agendas and pharmacy/nursing in-services. A comprehensive target drug utilization evaluation/medication use evaluation (DUE/MUE) is also provided each month. With a subscription, the monographs are available online to subscribers. Monographs can be customized to meet the needs of a facility. Through the cooperation of The Formulary, Hospital Pharmacy publishes selected reviews in this column. For more information about The Formulary Monograph Service, contact Wolters Kluwer customer service at 866-397-3433.
Collapse
|
8
|
Warren JS, Oka SI, Zablocki D, Sadoshima J. Metabolic reprogramming via PPARα signaling in cardiac hypertrophy and failure: From metabolomics to epigenetics. Am J Physiol Heart Circ Physiol 2017. [PMID: 28646024 DOI: 10.1152/ajpheart.00103.2017] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Studies using omics-based approaches have advanced our knowledge of metabolic remodeling in cardiac hypertrophy and failure. Metabolomic analysis of the failing heart has revealed global changes in mitochondrial substrate metabolism. Peroxisome proliferator-activated receptor-α (PPARα) plays a critical role in synergistic regulation of cardiac metabolism through transcriptional control. Metabolic reprogramming via PPARα signaling in heart failure ultimately propagates into myocardial energetics. However, emerging evidence suggests that the expression level of PPARα per se does not always explain the energetic state in the heart. The transcriptional activities of PPARα are dynamic, yet highly coordinated. An additional level of complexity in the PPARα regulatory mechanism arises from its ability to interact with various partners, which ultimately determines the metabolic phenotype of the diseased heart. This review summarizes our current knowledge of the PPARα regulatory mechanisms in cardiac metabolism and the possible role of PPARα in epigenetic modifications in the diseased heart. In addition, we discuss how metabolomics can contribute to a better understanding of the role of PPARα in the progression of cardiac hypertrophy and failure.
Collapse
Affiliation(s)
- Junco Shibayama Warren
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah; .,Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah; and
| | - Shin-Ichi Oka
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Daniela Zablocki
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, New Jersey
| |
Collapse
|
9
|
Nelson MAM, Baba SP, Anderson EJ. Biogenic Aldehydes as Therapeutic Targets for Cardiovascular Disease. Curr Opin Pharmacol 2017; 33:56-63. [PMID: 28528297 DOI: 10.1016/j.coph.2017.04.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 03/28/2017] [Accepted: 04/19/2017] [Indexed: 12/21/2022]
Abstract
Aldehydes are continuously formed in biological systems through enzyme-dependent and spontaneous oxidation of lipids, glucose, and primary amines. These highly reactive, biogenic electrophiles can become toxic via covalent modification of proteins, lipids and DNA. Thus, agents that scavenge aldehydes through conjugation have therapeutic value for a number of major cardiovascular diseases. Several commonly-prescribed drugs (e.g., hydralazine) have been shown to have potent aldehyde-conjugating properties which may contribute to their beneficial effects. Herein, we briefly describe the major sources and toxicities of biogenic aldehydes in cardiovascular system, and provide an overview of drugs that are known to have aldehyde-conjugating effects. Some compounds of phytochemical origin, and histidyl-dipeptides with emerging therapeutic value in this area are also discussed.
Collapse
Affiliation(s)
- Margaret-Ann M Nelson
- Department of Pharmacology & Toxicology, East Carolina University, Greenville, NC, USA
| | - Shahid P Baba
- Department of Medicine, Diabetes and Obesity Center, University of Louisville, Louisville, KY, USA
| | - Ethan J Anderson
- Department of Pharmaceutical Sciences & Experimental Therapeutics, College of Pharmacy, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
10
|
Reduced Oxidative Stress in STEMI Patients Treated by Primary Percutaneous Coronary Intervention and with Antioxidant Therapy: A Systematic Review. Cardiovasc Drugs Ther 2014; 28:173-81. [DOI: 10.1007/s10557-014-6511-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
11
|
Onodera H, Arito M, Sato T, Ito H, Hashimoto T, Tanaka Y, Kurokawa MS, Okamoto K, Suematsu N, Kato T. Novel effects of edaravone on human brain microvascular endothelial cells revealed by a proteomic approach. Brain Res 2013; 1534:87-94. [PMID: 23958343 DOI: 10.1016/j.brainres.2013.08.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 08/09/2013] [Indexed: 12/20/2022]
Abstract
Edaravone (3-methyl-1-phenyl-2-pyrazolin-5-one) is a free radical scavenger used for acute ischemic stroke. However, it is not known whether edaravone works only as a free radical scavenger or possess other pharmacological actions. Therefore, we elucidated the effects of edaravone on human brain microvascular endothelial cells (HBMECs) by 2 dimensional fluorescence difference gel electrophoresis (2D-DIGE). We found 38 protein spots the intensity of which was significantly altered 1.3 fold on average (p< 0.05) by the edaravone treatment and successfully identified 17 proteins of those. Four of those 17 proteins were cytoskeleton proteins or cytoskeleton-regulating proteins. Therefore, we subsequently investigated the change of size and shape of the cells, the actin network, and the tight junction of HBMEC by immunocytochemistry. As a result, most edaravone-treated HBMECs became larger and rounder compared with those that were not treated. Furthermore, edaravone-treated HBMECs formed gathering zona occludens (ZO)-1, a tight junction protein, along the junction of the cells. In addition, we found that edaravone suppressed interleukin (IL)-1β-induced secretion of monocyte chemoattractant protein-1 (MCP-1), which was reported to increase cell permeability. We found a novel function of edaravone is the promotion of tight junction formations of vascular endothelial cells partly via the down-regulation of MCP-1 secretion. These data provide fundamental and useful information in the clinical use of edaravone in patients with cerebral vascular diseases.
Collapse
Affiliation(s)
- Hidetaka Onodera
- Clinical Proteomics and Molecular Medicine, St. Marianna University Graduate School of Medicine, 2-16-1 Sugao, Miyamae, Kawasaki, Kanagawa 216-8512, Japan; Department of Neurosurgery, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae, Kawasaki, Kanagawa 216-8512, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Kikuchi K, Tancharoen S, Takeshige N, Yoshitomi M, Morioka M, Murai Y, Tanaka E. The efficacy of edaravone (radicut), a free radical scavenger, for cardiovascular disease. Int J Mol Sci 2013; 14:13909-30. [PMID: 23880849 PMCID: PMC3742225 DOI: 10.3390/ijms140713909] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 06/19/2013] [Accepted: 06/21/2013] [Indexed: 02/07/2023] Open
Abstract
Edaravone was originally developed as a potent free radical scavenger, and has been widely used to treat acute ischemic stroke in Japan since 2001. Free radicals play an important role in the pathogenesis of a variety of diseases, such as cardiovascular diseases and stroke. Therefore, free radicals may be targets for therapeutic intervention in these diseases. Edaravone shows protective effects on ischemic insults and inflammation in the heart, vessel, and brain in experimental studies. As well as scavenging free radicals, edaravone has anti-apoptotic, anti-necrotic, and anti-cytokine effects in cardiovascular diseases and stroke. Edaravone has preventive effects on myocardial injury following ischemia and reperfusion in patients with acute myocardial infarction. Edaravone may represent a new therapeutic intervention for endothelial dysfunction in the setting of atherosclerosis, heart failure, diabetes, or hypertension, because these diseases result from oxidative stress and/or cytokine-induced apoptosis. This review evaluates the potential of edaravone for treatment of cardiovascular disease, and covers clinical and experimental studies conducted between 1984 and 2013. We propose that edaravone, which scavenges free radicals, may offer a novel option for treatment of cardiovascular diseases. However, additional clinical studies are necessary to verify the efficacy of edaravone.
Collapse
Affiliation(s)
- Kiyoshi Kikuchi
- Department of Pharmacology, Faculty of Dentistry, Mahidol University, 6 Yothe Road, Rajthevee, Bangkok 10400, Thailand; E-Mails: (K.K.); (S.T.)
- Division of Brain Science, Department of Physiology, Kurume University School of Medicine, 67 Asahi-machi, Kurume 830-0011, Japan; E-Mail:
- Department of Neurosurgery, Kurume University School of Medicine, 67 Asahi-machi, Kurume 830-0011, Japan; E-Mails: (N.T.); (M.Y.); (M.M.)
| | - Salunya Tancharoen
- Department of Pharmacology, Faculty of Dentistry, Mahidol University, 6 Yothe Road, Rajthevee, Bangkok 10400, Thailand; E-Mails: (K.K.); (S.T.)
| | - Nobuyuki Takeshige
- Department of Neurosurgery, Kurume University School of Medicine, 67 Asahi-machi, Kurume 830-0011, Japan; E-Mails: (N.T.); (M.Y.); (M.M.)
| | - Munetake Yoshitomi
- Department of Neurosurgery, Kurume University School of Medicine, 67 Asahi-machi, Kurume 830-0011, Japan; E-Mails: (N.T.); (M.Y.); (M.M.)
| | - Motohiro Morioka
- Department of Neurosurgery, Kurume University School of Medicine, 67 Asahi-machi, Kurume 830-0011, Japan; E-Mails: (N.T.); (M.Y.); (M.M.)
| | - Yoshinaka Murai
- Division of Brain Science, Department of Physiology, Kurume University School of Medicine, 67 Asahi-machi, Kurume 830-0011, Japan; E-Mail:
| | - Eiichiro Tanaka
- Division of Brain Science, Department of Physiology, Kurume University School of Medicine, 67 Asahi-machi, Kurume 830-0011, Japan; E-Mail:
- Author to whom correspondence should be addressed. E-Mail: ; Tel.: +81-942-31-7542; Fax: +81-942-31-7695
| |
Collapse
|
13
|
Gupta M, Chaturvedi R, Jain A. Role of monocyte chemoattractant protein-1 (MCP-1) as an immune-diagnostic biomarker in the pathogenesis of chronic periodontal disease. Cytokine 2013; 61:892-7. [PMID: 23375122 DOI: 10.1016/j.cyto.2012.12.012] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 12/06/2012] [Accepted: 12/19/2012] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Monocyte chemoattractant protein-1 (MCP-1) is an important chemokine responsible for the initiation, regulation and mobilization of monocytes to the active sites of severe periodontal inflammation. The present study aims at evaluating the levels of MCP-1 in GCF, saliva and serum and to analyze the changes following phase I periodontal therapy. Assessment of possible correlations between levels of MCP-1 in the three biological fluids was also done. METHODS Fifteen healthy and 30 patients of severe chronic periodontitis (diseased) participated in the study. Patients of the diseased group underwent scaling/root planing. Evaluation of PI, GI, PD, CAL and collection of samples of GCF, serum and saliva was done at baseline and 6 weeks following periodontal therapy. MCP-1 levels were quantified in all samples using ELISA. RESULTS Compared to healthy controls, MCP-1 levels were statistically significantly higher in GCF (p<0.001), saliva (p=0.002) and serum (p<0.001) in subjects with chronic periodontitis. Levels of MCP-1 in all the three fluids decreased significantly in patients after periodontal therapy (p<0.001). There was a significant positive correlation between MCP-1 levels in GCF, saliva and serum in patients of chronic periodontitis both pre (r>0.9) and post-treatment (r>0.6). CONCLUSIONS The results suggest that levels of MCP-1 in GCF and saliva can be reliable indicators of severity of periodontal destruction and their serum levels reflect the systemic impact of this local inflammatory disease thereby strengthening the reciprocal oro-systemic association.
Collapse
Affiliation(s)
- Mili Gupta
- Department of Biochemistry, Dr. Harvansh Singh Judge, Institute of Dental Sciences & Hospital, Panjab University, Chandigarh, India.
| | | | | |
Collapse
|
14
|
Somekawa-Kondo T, Yamaguchi K, Ishitsuka Y, Ito S, Tanaka K, Irikura M, Moriuchi H, Takahama K, Ando Y, Yamazaki T, Irie T. Aminophylline, administered at usual doses for rodents in pharmacological studies, induces hippocampal neuronal cell injury under low tidal volume hypoxic conditions in guinea-pigs. J Pharm Pharmacol 2012; 65:102-14. [PMID: 23215693 DOI: 10.1111/j.2042-7158.2012.01566.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES To establish whether aminophylline, administered at usual doses for rodents in pharmacological studies, induces brain injury in systemic hypoxaemia in guinea-pigs. METHODS A hypoxaemia (partial oxygen tension of arterial blood (PaO₂) = 40-60 mmHg) model was developed by low tidal volume mechanical ventilation in guinea-pigs. KEY FINDINGS Under hypoxic conditions, aminophylline significantly increased the concentration of brain-specific creatine kinase in the serum in a dose- and time-dependent manner. A reduced number of hippocampal neuronal cells in the CA1 region, an increase in the concentration of neuron-specific enolase (NSE) in cerebrospinal fluid (CSF), an increase in lipid hydroperoxides and a decrease in the ratio of glutathione to glutathione disulfide in the brain tissues were also observed. These effects were not observed when aminophylline at the same doses was administered under normoxic conditions (PaO₂ = 80-100 mmHg). There was no difference in either serum or CSF concentrations of theophylline between normoxic and hypoxic conditions. Another methylxanthine, caffeine, did not increase the concentration of NSE in CSF. CONCLUSIONS Aminophylline potentially induces brain damage under hypoxic conditions. We suggest that aminophylline treatment has adverse effects in patients with hypoxaemia subsequent to respiratory disorders such as asthma.
Collapse
Affiliation(s)
- Tomoko Somekawa-Kondo
- Department of Clinical Chemistry and Informatics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Kohno T, Anzai T, Kaneko H, Sugano Y, Shimizu H, Shimoda M, Miyasho T, Okamoto M, Yokota H, Yamada S, Yoshikawa T, Okada Y, Yozu R, Ogawa S, Fukuda K. High-mobility group box 1 protein blockade suppresses development of abdominal aortic aneurysm. J Cardiol 2012; 59:299-306. [DOI: 10.1016/j.jjcc.2012.01.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Revised: 12/09/2011] [Accepted: 01/09/2012] [Indexed: 01/22/2023]
|
16
|
Kikuchi K, Takeshige N, Miura N, Morimoto Y, Ito T, Tancharoen S, Miyata K, Kikuchi C, Iida N, Uchikado H, Miyagi N, Shiomi N, Kuramoto T, Maruyama I, Morioka M, Kawahara KI. Beyond free radical scavenging: Beneficial effects of edaravone (Radicut) in various diseases (Review). Exp Ther Med 2011; 3:3-8. [PMID: 22969835 DOI: 10.3892/etm.2011.352] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Accepted: 09/13/2011] [Indexed: 12/19/2022] Open
Abstract
Free radicals play an important role in the pathogenesis of a variety of diseases; thus, they are an attractive target for therapeutic intervention in these diseases. Compounds capable of scavenging free radicals have been developed for this purpose and some, developed for the treatment of cerebral ischemic stroke, have progressed to clinical trials. One such scavenger, edaravone, is used to treat patients within 24 h of stroke. Edaravone, which can diffuse into many disease-affected organs, also shows protective effects in the heart, lung, intestine, liver, pancreas, kidney, bladder and testis. As well as scavenging free radicals, edaravone has anti-apoptotic, anti-necrotic and anti-cytokine effects in various diseases. Here, we critically review the literature on its clinical efficacy and examine whether edaravone should be considered a candidate for worldwide development, focusing on its effects on diseases other than cerebral infarction. Edaravone has been safely used as a free radical scavenger for more than 10 years; we propose that edaravone may offer a novel treatment option for several diseases.
Collapse
Affiliation(s)
- Kiyoshi Kikuchi
- Department of Neurosurgery, Yame Public General Hospital, Yame 834-0034
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Miyazaki Y, Kaikita K, Endo M, Horio E, Miura M, Tsujita K, Hokimoto S, Yamamuro M, Iwawaki T, Gotoh T, Ogawa H, Oike Y. C/EBP homologous protein deficiency attenuates myocardial reperfusion injury by inhibiting myocardial apoptosis and inflammation. Arterioscler Thromb Vasc Biol 2011; 31:1124-32. [PMID: 21330607 DOI: 10.1161/atvbaha.111.224519] [Citation(s) in RCA: 118] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVE To investigate whether and how the endoplasmic reticulum (ER) stress-induced, CCAAT/enhancer-binding protein-homologous protein (CHOP)-mediated pathway regulates myocardial ischemia/reperfusion injury. METHODS AND RESULTS Wild-type and chop-deficient mice underwent 50 minutes of left coronary artery occlusion followed by reperfusion. Expression of chop and spliced x-box binding protein-1 (sxbp1) mRNA was rapidly and significantly increased in reperfused myocardium of wild-type mice. chop-deficient mice exhibited markedly reduced injury size after reperfusion compared with wild-type mice, accompanied by a decreasing number of terminal deoxynucleotidyl transferase dUTP nick-end labeling-positive cardiomyocytes. Interestingly, myocardial inflammation, as assessed by expression of inflammatory cytokines and chemokines and numbers of infiltrated inflammatory cells, was also attenuated in chop-deficient mice. Moreover, expression of interleukin-6 mRNA in response to lipopolysaccharide was enhanced by simultaneous stimulation with thapsigargin, a potent ER stressor, in wild-type cardiomyocytes but not in chop-deficient cardiomyocytes. Finally, we found that superoxide was produced in reperfused myocardium and that intravenous administration of edaravone, a free radical scavenger, immediately before reperfusion significantly suppressed the superoxide overproduction and subsequent expression of sxbp1 and chop mRNA, followed by reduced injury size in wild-type mice. CONCLUSIONS The ER stress-induced, CHOP-mediated pathway, which is activated in part by superoxide overproduction after reperfusion, exacerbates myocardial ischemia/reperfusion injury by inducing cardiomyocyte apoptosis and myocardial inflammation.
Collapse
Affiliation(s)
- Yuji Miyazaki
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University, 2-2-1 Honjo, Kumamoto 860-0811, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|