Molecular basis of oxidative stress chemistry mechanisms and disease pathogenesis

448 145 0
Molecular basis of oxidative stress chemistry mechanisms and disease  pathogenesis

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

Thông tin tài liệu

MOLECULAR BASIS OF OXIDATIVE STRESS MOLECULAR BASIS OF OXIDATIVE STRESS Chemistry, Mechanisms, and Disease Pathogenesis Edited by FREDERICK A VILLAMENA Department of Pharmacology and Davis Heart and Lung Institute The Ohio State University Columbus, Ohio Copyright © 2013 by John Wiley & Sons, Inc All rights reserved Published by John Wiley & Sons, Inc., Hoboken, New Jersey Published simultaneously in Canada No part of this publication may be reproduced, stored in a retrieval system, or transmitted in any form or by any means, electronic, mechanical, photocopying, recording, scanning, or otherwise, except as permitted under Section 107 or 108 of the 1976 United States Copyright Act, without either the prior written permission of the Publisher, or authorization through payment of the appropriate per-copy fee to the Copyright Clearance Center, Inc., 222 Rosewood Drive, Danvers, MA 01923, (978) 750-8400, fax (978) 750-4470, or on the web at www.copyright.com Requests to the Publisher for permission should be addressed to the Permissions Department, John Wiley & Sons, Inc., 111 River Street, Hoboken, NJ 07030, (201) 748-6011, fax (201) 748-6008, or online at http://www.wiley.com/go/permissions Limit of Liability/Disclaimer of Warranty: While the publisher and author have used their best efforts in preparing this book, they make no representations or warranties with respect to the accuracy or completeness of the contents of this book and specifically disclaim any implied warranties of merchantability or fitness for a particular purpose No warranty may be created or extended by sales representatives or written sales materials The advice and strategies contained herein may not be suitable for your situation You should consult with a professional where appropriate Neither the publisher nor author shall be liable for any loss of profit or any other commercial damages, including but not limited to special, incidental, consequential, or other damages For general information on our other products and services or for technical support, please contact our Customer Care Department within the United States at (800) 762-2974, outside the United States at (317) 572-3993 or fax (317) 572-4002 Wiley also publishes its books in a variety of electronic formats Some content that appears in print may not be available in electronic formats For more information about Wiley products, visit our web site at www.wiley.com Library of Congress Cataloging-in-Publication Data Molecular basis of oxidative stress : chemistry, mechanisms, and disease pathogenesis / edited by Frederick A Villamena, Department of Pharmacology and Davis Heart and Lung Institute, The Ohio State University, Columbus, Ohio, USA pages cm Includes bibliographical references and index ISBN 978-0-470-57218-4 (cloth) Oxidative stress Oxidation I Villamena, Frederick A QD281.O9M65 2013 571.9'453–dc23 2013002853 Printed in the United States of America 10 CONTENTS Preface xvii About the Contributors xix Contributors xxv Chemistry of Reactive Species Frederick A Villamena 1.1 1.2 1.3 Redox Chemistry, Classification of Reactive Species, 1.2.1 Type of Orbitals, 1.2.2 Stability of Radicals, 1.2.3 ROS, 1.2.3.1 Oxygen Molecule (O2, Triplet Oxygen, Dioxygen), 1.2.3.2 Superoxide Radical Anion (O2•−), 1.2.3.3 Hydroperoxyl Radical (HO2•), 1.2.3.4 Hydrogen Peroxide (H2O2), 10 1.2.3.5 Hydroxyl Radical (HO•), 11 1.2.3.6 Singlet Oxygen (1O21Δg or 1O2*), 13 1.2.4 Reactive Nitrogen Species, 14 1.2.4.1 Nitric Oxide (NO or •NO), 14 1.2.4.2 Nitrogen Dioxide (•NO2), 16 1.2.4.3 Peroxynitrite (ONOO−), 17 1.2.5 Reactive Sulfur and Chlorine Species, 18 1.2.5.1 Thiyl or Sulfhydryl Radical (RS•), 18 1.2.5.2 Disulfide (RSSR), 19 1.2.5.3 Hypochlorous Acid (HOCl), 20 Reactivity, 22 1.3.1 Thermodynamic Considerations, 22 1.3.2 Kinetic Considerations, 24 1.3.2.1 Unimolecular or First-Order Reactions, 25 1.3.2.2 Bimolecular or Second-Order Reactions, 25 1.3.2.3 Transition State Theory, Reaction Coordinates and Activation Energies, 26 v vi CONTENTS 1.4 Origins of Reactive Species, 26 1.4.1 Biological Sources, 26 1.4.1.1 NADPH Oxidase, 26 1.4.1.2 Xanthine Oxidoreductase or Oxidase, 27 1.4.1.3 Mitochondrial Electron Transport Chain (METC), 27 1.4.1.4 Hemoglobin (Hb), 28 1.4.1.5 Nitric Oxide Synthases, 28 1.4.1.6 Cytochrome P450 (CYP), 29 1.4.1.7 Cyclooxygenase (COX) and Lipoxygenase (LPO), 29 1.4.1.8 Endoplasmic Reticulum (ER), 29 1.4.2 Nonbiochemical Sources, 29 1.4.2.1 Photolysis, 29 1.4.2.2 Sonochemical, 30 1.4.2.3 Photochemical, 30 1.4.2.4 Electrochemical, 30 1.4.2.5 Chemical, 30 1.5 Methods of Detection, 31 1.5.1 In Vitro, 32 1.5.1.1 Flourescence and Chemiluminescence, 32 1.5.1.2 UV-Vis Spectrophotometry and HPLC, 33 1.5.1.3 Immunochemical, 34 1.5.1.4 Electron Paramagnetic Resonance (EPR) Spectroscopy, 34 1.5.2 In Vivo, 38 1.5.2.1 Histochemical, 38 1.5.2.2 Immunocytochemical Methods, 38 1.5.2.3 Low Frequency EPR Imaging, 38 1.5.2.4 In Vivo EPR Spin Tapping-Ex Vivo Measurement, 38 References, 38 Lipid Peroxidation and Nitration Sean S Davies and Lilu Guo Overview, 49 2.1 Peroxidation of PUFAs, 49 2.1.1 Hydroperoxy Fatty Acid Isomers (HpETEs and HpODEs), 50 2.1.2 Hydroxy Fatty Acids (HETEs and HODEs), 51 2.1.3 Isoleukotrienes, 51 2.1.4 Epoxy Alcohols, 52 2.2 Cyclic Endoperoxides and Their Products, 52 2.2.1 Isoprostanes, 52 2.2.1.1 Isoprostane Regio- and Stereoisomers, 54 2.2.1.2 F2-Isoprostanes, 54 2.2.1.3 Other Major Isoprostane Products, 54 2.2.1.4 Minor Isoprostane Products, 56 2.2.2 Diepoxide Pathway Products, 57 2.2.2.1 Isofurans and Related Compounds, 57 2.2.3 Serial Cyclic Endoperoxides, 57 2.3 Fragmented Products of Lipid Peroxidation, 58 2.3.1 Short-Chain Alkanes, Aldehydes, and Acids, 58 2.3.2 Oxidatively Fragmented Phospholipids, 58 2.3.3 PAF Acetylhydrolase, 59 2.3.4 Hydroxyalkenals, 59 49 CONTENTS 2.3.5 Malondialdehyde, 61 2.3.6 Acrolein, 61 2.4 Epoxy Fatty Acids, 62 2.5 Lipid Nitrosylation, 62 2.5.1 Formation of Reactive Nitrogen Species, 63 2.5.2 Lipid Nitration Reactions, 63 2.5.3 Detection of Lipid Nitration In Vivo, 64 2.5.4 Bioactivities of Nitrated Lipids, 64 Summary, 65 References, 65 Protein Posttranslational Modification James L Hougland, Joseph Darling, and Susan Flynn 71 Overview, 71 3.1 Oxidative Stress-Related PTMs: Oxidation Reactions, 71 3.1.1 Cysteine, 71 3.1.1.1 Formation of Sulfur–Oxygen Adducts: Sulfenic, Sulfinic, and Sulfonic Acids, 72 3.1.1.2 Formation of Sulfur–Nitrogen Adducts: S-Nitrosothiols and Sulfonamides, 73 3.1.1.3 Formation of Sulfur–Sulfur Adducts: Disulfides and S-Glutathionylation, 74 3.1.1.4 Redoxins: Enzymes Catalyzing Cysteine Reduction, 75 3.1.2 Methionine, 76 3.1.3 Oxidation of Aromatic Amino Acids, 78 3.1.3.1 Tyrosine, 78 3.1.3.2 Tryptophan, 79 3.1.3.3 Histidine, 79 3.1.3.4 Phenylalanine, 79 3.1.4 Oxidation of Aliphatic Amino Acids, 79 3.2 Amino Acid Modification by Oxidation-Produced Electrophiles, 80 3.2.1 Electrophiles Formed by Oxidative Stress, 80 3.2.2 Carbonylation Reactions with Amino Acids, 80 3.3 Detection of Oxidative-Stress Related PTMs, 81 3.3.1 Mass Spectrometry, 81 3.3.2 Chemoselective Functionalization, 82 3.3.3 Cysteine Modifications, 82 3.3.3.1 Sulfenic Acids, 82 3.3.3.2 Cysteine-Nitrosothiols, 82 3.3.3.3 Cysteine-Glutathionylation, 82 3.3.4 Protein Carbonylation, 83 3.4 Role of PTMs in Cellular Redox Signaling, 84 Summary, 85 References, 85 DNA Oxidation Dessalegn B Nemera, Amy R Jones, and Edward J Merino Overview, 93 4.1 The Context of Cellular DNA Oxidation, 93 4.2 Oxidation of Oligonucleotides, 94 4.3 Examination of Specific Oxidative Lesions, 96 4.3.1 8-Oxo-7,8-Dihydro-2′-Deoxyguanosine, 96 93 vii viii CONTENTS 4.3.2 Lesions on Ribose Bases Including Apurinic or Apyrimidinic Sites, 99 4.3.3 Novel Types of Ribose and Guanine Oxidative Lesions and Future Outlook, 101 4.3.3.1 Tandem Lesions, 101 4.3.3.2 Hyperoxidized Guanine, 102 4.3.3.3 Oxidative Cross-Links, 103 Future Outlook of DNA Oxidative Lesions, 103 References, 103 Downregulation of Antioxidants and Phase Proteins Hong Zhu, Jianmin Wang, Arben Santo, and Yunbo Li 113 Overview, 113 5.1 Definitions of Antioxidants and Phase Proteins, 113 5.1.1 Antioxidants, 113 5.1.2 Phase Proteins, 113 5.2 Roles in Oxidative Stress, 114 5.2.1 Superoxide Dismutase, 114 5.2.2 Catalase, 114 5.2.3 GSH and GSH-Related Enzymes, 114 5.2.3.1 GSH, 114 5.2.3.2 Glutathione Peroxidase, 115 5.2.3.3 Glutathione Reductase, 115 5.2.3.4 GST, 115 5.2.4 NAD(P)H:Quinone Oxidoreductase, 116 5.2.5 Heme Oxygenase, 116 5.2.6 Ferritin, 116 5.2.7 UDP-Glucuronosyltransferase, 116 5.3 Molecular Regulation, 116 5.3.1 General Consideration, 116 5.3.2 Nrf2 Signaling, 116 5.3.3 Other Regulators, 117 5.4 Induction in Chemoprevention, 117 5.4.1 Chemical Inducers, 117 5.4.2 Chemoprotection, 117 5.5 Downregulation, 117 5.5.1 Selective Chemical Inhibitors, 117 5.5.1.1 N,N-Diethyldithiocarbamate, 118 5.5.1.2 3-Amino-1,2,4-Triazole, 118 5.5.1.3 BSO, 118 5.5.1.4 Sulfasalazine, 118 5.5.1.5 Dicumarol, 118 5.5.2 Drugs and Environmental Toxic Agents, 118 Conclusions and Perspectives, 119 References, 119 Mitochondrial Dysfunction Yeong-Renn Chen Overview, 123 6.1 Mitochondria and Submitochondrial Particles, 123 6.2 Energy Transduction, 125 6.3 Mitochondrial Stress, 125 123 CONTENTS 6.4 Superoxide Radical Anion Generation as Mediated by ETC and Disease Pathogenesis, 126 6.4.1 Mediation of O2•− Generation by Complex I, 126 6.4.1.1 The Role of FMN Moiety, 126 6.4.1.2 The Role of Ubiquinone-Binding Domain, 126 6.4.1.3 The Role of Iron–Sulfur Clusters, 127 6.4.1.4 The Role of Cysteinyl Redox Domains, 127 6.4.1.5 Complex I, Free Radicals, and Parkinsonism, 129 6.4.2 Mediation of O2•− Generation by Complex II, 129 6.4.2.1 The Role of FAD Moiety, 129 6.4.2.2 The Role of Ubiquinone-Binding Site, 129 6.4.2.3 Mutations of Complex II Are Related with Mitochondrial Diseases, 129 6.4.2.4 Mitochondrial Complex II in Myocardial Infarction, 130 6.4.3 Mediation of O2•− Generation by Complex III, 130 6.4.3.1 The Q-Cycle Mediated by Complex III, 130 6.4.3.2 Role of Q Cycle in O2•− Generation, 131 6.4.3.3 The Role of Cytochrome bL in O2•− Generation, 132 6.4.3.4 Bidirectionality of Superoxide Release as Mediated by Complex III, 132 6.4.4 Complex IV, 132 Summary, 133 References, 134 NADPH Oxidases: Structure and Function Mark T Quinn Overview, 137 7.1 Introduction, 137 7.2 Phagocyte NADPH Oxidase Structure, 137 7.2.1 Flavocytochrome b, 138 7.2.2 p47phox, 139 7.2.3 p67phox, 140 7.2.4 p40 phox, 141 7.2.5 Rac1/2, 141 7.2.6 Rap1A, 142 7.3 Phagocyte ROS Production, 142 7.3.1 Superoxide Anion (O2•−), 142 7.3.2 Hydrogen Peroxide (H2O2), 143 7.3.3 Hypochlorous Acid (HOCl), 143 7.3.4 Hydroxyl Radical (HO•), 143 7.3.5 Singlet Oxygen (1O2*), 144 7.3.6 Nitric Oxide (•NO) and Peroxynitrite (OONO−), 144 7.4 Phagocyte NADPH Oxidase Function, 145 7.5 Nonphagocyte NADPH Oxidase Structure, 146 7.5.1 NOX1, 147 7.5.2 NOX3, 149 7.5.3 NOX4, 149 7.5.4 NOX5, 150 7.5.5 DUOX1 and DUOX2, 150 7.5.6 NOXO1, 150 7.5.7 NOXA1, 151 137 ix x CONTENTS 7.6 7.7 Nonphagocyte ROS Production, 151 Functions of Nonphagocyte NADPH Oxidases, 152 7.7.1 Cardiovascular System, 152 7.7.2 Renal System, 154 7.7.3 Pulmonary System, 155 7.7.4 Central Nervous System, 156 7.7.5 Gastrointestinal System, 157 7.7.6 Hepatic System, 158 7.7.7 Thyroid Gland, 159 Summary, 159 Acknowledgments, 159 References, 160 Cell Signaling and Transcription Imran Rehmani, Fange Liu, and Aimin Liu 179 Overview, 179 8.1 Common Mechanisms of Redox Signaling, 179 8.2 Redox and Oxygen-Sensitive Transcription Factors in Prokaryotes, 181 8.2.1 Fe–S Cluster Proteins, 181 8.2.2 Prokaryotic Hydrogen Peroxide Sensors: Proteins Utilizing Reactive Thiols, 182 8.2.3 PerR: A Unique Metalloprotein Sensor of Hydrogen Peroxide, 182 8.2.4 Summary, 184 8.3 Redox Signaling in Metazoans, 185 8.3.1 Primary Sources of ROS in Eukaryotic Redox Signaling, 185 8.3.2 The Floodgate Hypothesis, 186 8.3.3 Redox Regulation of Kinase and Phosphatase Activity, 187 8.3.4 Communication between ROS and Calcium Signaling, 188 8.3.5 Redox Modulation of Transcription Factors, 188 8.3.6 Summary, 189 8.4 Oxygen Sensing in Metazoans, 190 8.4.1 HIF, 190 8.4.2 PHD Enzymes, 190 8.4.3 FIH, 191 8.4.4 Factors Influencing Fe(II)/α-KG Dependent Enzymes, 192 8.4.5 ROS and Oxygen Sensing, 193 8.4.6 Summary, 193 8.5 Medical Significance of Redox and Oxygen-Sensing Pathways, 194 8.5.1 Cancer, 194 8.5.2 Vascular Pathophysiology, 194 Concluding Remarks, 195 References, 195 Oxidative Stress and Redox Signaling in Carcinogenesis Rodrigo Franco, Aracely Garcia-Garcia, Thomas B Kryston, Alexandros G Georgakilas, Mihalis I Panayiotidis, and Aglaia Pappa Overview, 203 9.1 Redox Environment and Cancer, 203 9.1.1 Pro-Oxidant Environment and Endogenous Sources of RS in Cancer, 203 9.1.1.1 Reactive Oxygen Species (ROS)-Generating NADPH Oxidases and Cancer, 203 203 396 SYNTHETIC ANTIOXIDANTS equal in the two groups, and no evidence of efficacy for any secondary end points, including scores on neurologic and activities of daily living scales, was observed A pooled analysis of the SAINT I and II trials further concluded that NXY-059 was not effective in the combined trials.215 Although both preclinical and clinical trials were supposed to be developed in accordance with the Stroke Therapy Academic Industry Roundtable (STAIR), the very disappointing outcome of the SAINT II trial led to the termination of NXY-059 as therapeutics for acute ischemic stroke Critiques on how the development of NXY-059 was conducted have been raised Savitz pointed out the need for more rigorous and strenuous testing at the preclinical stage.216 Among Savitz’s critiques about the preclinical studies, one can cite the lack of sufficient independent studies in different laboratories in order to confirm preliminary findings as well as the absence of information on NXY-059 mechanisms of action and whether it enters the brain parenchyma after embolic stroke Clinical trial also appeared to be inadequately designed because of inappropriate treatment windows and inclusion of diverse stroke patients 15.4.5 The Controversial Mode of Action of Nitrones Considering the very broad activity of nitrones as discussed above and reviewed by others, there had been controversy on the mechanism of action of nitrone in biological systems Since they were initially employed as probes for spin-trapping experiments, their biological activity had been first explained based on their radical trapping activity However, several experimental evidences have strongly put aside the trapping activity and other mechanisms have been suggested For instance, the rate of reaction of PBN with radical is quite slow,135 that is, 105–107 M−1.s−1 and therefore, during spin trapping experiments in chemical milieu, PBN must be present at high concentration (10–50 mM) to trap a significant fraction of free radicals These concentrations used are roughly 1000 times higher than those commonly employed in biological studies of protection (10–50 μM) Moreover, the concentration of nitrones in the target tissues is usually inferior at 0.5 mM, which is not sufficient to quench all the radical species As previously discussed above, in experimental stroke model, it had been observed that PBN led to protection even if administered up to hours after the start of reperfusion.198 Preclinical and clinical studies with PBN and NXY-059 confirmed without any doubt the ability of nitrones to prevent stroke induced-damages even when administered of few hours after the onset of stroke These findings further support the invalidation of the spin-trapping mechanism as the primary mode of action of nitrones Another demonstration by Floyd and Carney was provided with aging gerbils that were chronically administered with PBN for 14 days.217 Although the treatment ceased several days before the stroke, treated animals were more resistant than untreated ones Considering the half-life of PBN ∼2 hours, it is very likely that none of the nitrone is still present days after cessation of the treatment, thus demonstrating that the classical mass action of spin traps is not solely responsible for the pharmacological activities of nitrones 15.4.5.1 Antioxidant Property of PBN against Lipid Peroxydation Its very poor antioxidant activity have been observed in simple models of lipid peroxidation demonstrating that nitrones not act as classical chainbreaking antioxidants.218,219 While PBN inhibited lipid peroxidation at mM, antioxidants such as γ-tocopherol or BHT were a thousand-fold more efficient, exhibiting antioxidant protection at only μM.218 15.4.5.2 Anti-Inflammatory and Anti-Apopotic Properties of Nitrones There is strong evidence that PBN acts to quell signal transduction processes and therefore provides potent anti-inflammatory and antiapoptotic properties.161,162,164,191 PBN has been shown to inhibit the activity of cytokines and transcription factors such as NF-κB, which can rapidly activate the expression of genes involved in inflammation.220–222 The inhibition of the COX2 catalytic activity and the decrease of the steady state COX2 mRNA level was also demonstrated Other rationales for nitrone protection in animal models showing that PBN can inhibit gene induction of heat shock proteins include c-fos and inducible nitric oxide synthase as well One of the first and yet convincing study that illustrated the importance of neuro-inflammation in brain injury and the potency of PBN to preserve brain function by quelling exacerbated signal transduction processes is that by Floyd et al in 2000.223 Kainate administration to rats led to injury to the hippocampus providing a good experimental model of epilepsy Administration of PBN prevented apoptotic neuron loss and mortality, and immunohistochemical examination showed that the Kainate-induced activation of p38 MAP kinase and NF-κB was suppressed by PBN Since then, numerous reports have shown the ability of nitrone to suppress the proinflammatory cytokine and stressors mediated induction of genes in a wide range of biological systems In the review by Green et al are listed some important biochemical consequences of nitrone administration in vitro, ex vivo, and in vivo, which may be associated with neuroprotection (Table 15.2).204 NITRONES TABLE 15.2 et al.204) 397 Some Biochemical Consequences of PBN Administration In Vitro, In Vivo, and Ex Vivo (Adapted from Green In vitro In vivo or ex vivo 10 11 12 13 14 Inhibition of iNOS induction in HIV-1 envelope Potentiation of H2O2-induced Erk and Src kinase in human Stimulation of H2O2-induced activation of the prosurvival Erk signal transduction pathway Protection of primary cerebellar neurones from glutamate toxicity Facilitation of postischemic reperfusion following transient MCAO Inhibition of apoptosis-associated gene expression in endotoxin-treated rats Suppression of caspase-3 activation following global ischemia Prevention of the decrease in stimulated (+ADP), nonstimulated (–ADP) and uncoupled rats following transient MCAO Inhibition of endotoxin-induced induction of nitric oxide synthase Improvement of the rate of metabolic recovery, acidosis rebound, and ATP renewal in rat brain following transient focal ischemia Suppresion of c-fos expression in postischemic gerbil brain Attenuation of the secondary mitochondrial dysfunction after transient focal ischemia Reduction of the number of positive τ-oligodendrocytes after focal ischemia Prevention of cytotoxic ischemia following malonate 15.4.5.3 Action on Membrane Enzymes PBN was shown to act as a reversible calcium channel blocker at concentrations far lower than those required for free radical detection by EPR, and it was concluded that removal of free radicals may not contribute to the nitrone-induced vasorelaxation.224 The potency of PBN to inhibit acetylcholinesterase activity, an enzyme that converts acetylcholine into the inactive metabolites choline and acetate, with a Ki of 0.58 mM, was also reported.225 15.4.5.4 Interaction with the Mitochondrial Metabolism Hensley et al demonstrated that PBN interacts with the mitochondrial complex I, inhibiting complex-I stimulated H2O2 flux and nitro blue tetrazolium reduction.226 This site-specific interaction of PBN with mitochondrial flavin deshydrogenases, which leads to alteration of the electron transit within the enzymes, is believed to participate to the broad antioxidant and anti-inflammatory action of nitrones Interaction of PBN with mitochondrial complex I was also demonstrated in the prevention of the doxorubicin-induced apoptosis in bovine aortic endothelial cells.171 Indeed, it was shown that doxorubicin inactivated complex-I by a superoxide-dependant mechanism and that complex-I activity was restored by PBN, confirming the first observations by Hensley et al Pretreatment of cells with PBN also resulted in a full inhibition of DOX-induced cytochrome c release and a complete restoration of GSH levels Other mitochondrial-specific interactions of nitrones were next reported An increase of endogenous mitochondrial superoxide production, following intra- thecal injection of complex III inhibitor antimycin A, has been demonstrated to induce hyperalgesia in mice.227 PBN significantly reduced hyperalgesia 30 and 60 minutes following the treatment providing transient antinociception irrespective to the administration mode of PBN, that is, intraperitoneal or intrathecal Mitochondrial impairment has been associated with cardiac dysfunction in Chagas’ disease, a parasitic disease whose about 40% of seropositive patients develop cardiomyopathy In infected mice, oxidative stress and alteration of mitochondrial functions in the myocardium are observed as well as similar cardiac dysfunction to what is observed in human chagasic patients Treatment of infected mice with PBN prevented mitochondrial oxidative damage and significantly improved respiratory complex activities.228,229 The impairment of a specific site of complex III was identified as the main target in infected myocardium, leading to an increased electron leakage and O2 production Treatment with PBN improved the respiratory chain function by preserving electron transport chain (ETC) activity thereby limiting electron leakage and mitochondrial ROS production The two amphiphilic amide nitrones, the LPBNAH and its reverse analogue the LPBNH15 (Fig 15.15), have recently shown very promising results.230 These two compounds exhibited hydroxyl radical scavenging activity and radical reducing potency in ABTS assays Experimental and theoretical data showed that substitution of the PBN by hydrophilic and lipophilic groups alters its redox properties, with the amphiphilic amide nitrones being easier to oxidize and reduce than the parent PBN Moreover, very high protective effects 398 SYNTHETIC ANTIOXIDANTS were demonstrated both in in vitro and in vivo experiments With regard to the mitochondrial interaction, these two nitrones showed interesting properties They both decreased electron and proton leakage as well as hydrogen peroxide formation in isolated rat brain mitochondria at nanomolar concentration They also significantly enhanced mitochondrial membrane potential, and the dopamine-induced inhibition of complex I activity was antagonized by pretreatment with these agents These findings strongly suggest that new nitrone analogues are more than just radical scavenging antioxidants but may act as a new class of bioenergetic agents directly interacting with mitochondrial electron and proton transport 13 14 15 16 REFERENCES Halliwell, B., Gutteridge, J.M.C Free Radicals in Biology and Medicine, 4th ed Oxford University Press, Oxford, 2007 The Alpha-Tocopherol, Beta Carotene Cancer Prevention Study Group The effect of vitamin E and beta carotene on the incidence of lung cancer and other cancers in male smokers N Engl J Med 1994, 330, 1029–1035 Ascherio, A., Rimm, E.B., Hernan, M.A., Giovannucci, E., Kawachi, I., Stampfer, M.J., Willet, W.C Relation of consumption of vitamin E, vitamin C, and carotenoids to risk for stroke among men in the United States Ann Intern Med 1999, 130, 963–970 Krirtharides, L., Stocker, R The use of antioxidant supplements in coronary heart disease Atherosclerosis 2002, 164, 211–219 Meyers, D.G., Maloley, P.A., Weeks, D Safety of antioxidant vitamins Arch Intern Med 1996, 156, 925–935 Riley, D.P Functional mimics of superoxide dismutase enzymes as therapeutic agents Chem Rev 1999, 99, 2573–2587 Day, B.J Catalytic antioxidants: A radical approach to new therapeutics Drug Discov Today 2004, 9, 557–566 Day, B.J Catalase and glutathione peroxidase mimics Biochem Pharmacol 2009, 77, 285–296 Patel, M., Day, B.J Metalloporphyrin class of therapeutic catalytic antioxidants Trends Pharmacol Sci 1999, 20, 359–364 10 Baudry, M., Etienne, S., Bruce, A., Palucki, M., Jacobsen, E., Malfroy, B Salen-manganese complexes are superoxide dismutase-mimics Biochem Biophys Res Commun 1993, 192, 964 11 Doctrow, S.R., Huffman, K., Marcus, C.B., Malfroy, B Salen-manganese complexes: Combined superoxide dismutase/catalase mimics with broad pharmacological efficacy Adv Pharmacol 1997, 38, 247–269 12 Sharpe, M.A., Ollosson, R., Stewart, V.C., Clark, J.B Oxidation of nitric oxide by oxomanganese-salen complexes: 17 18 19 20 21 22 23 24 25 A new mechanism for cellular protection by superoxide dismutase/catalase mimetics Biochem J 2002, 366, 97– 107 Boucher, L.J Maganese Schiff’s base complexes II: Synthesis and spectroscopy of chloro-complexes of some derivatives of (salicylaldehydeethylenediimato) maganese(III) J Inorg Nucl Chem 1974, 36, 531–536 Bruce, A.J., Malfroy, B., Baudry, M beta-Amyloid toxicity in organotypic hippocampal cultures: Protection by EUK-8, a synthetic catalytic free radical scavenger Proc Natl Acad Sci U.S.A 1996, 93, 2312–2316 Musleh, W., Bruce, A., Malfroy, B., Baudry, M Effects of EUK-8, a synthetic catalytic superoxide scavenger, on hypoxia- and acidosis-induced damage in hippocampal slices Neuropharmacology 1994, 33, 929–934 Baker, K., Bucay-Marcus, C., Huffman, K., Kruk, H., Malfroy, B., Doctrow, S.R Synthetic combined superoxide dismutase/catalase mimics are protective as a delayed treatment in a rat stroke model: A key role for reactive oxygen species in ischemic brain injury J Pharmacol Exp Ther 1998, 284, 215–221 Doctrow, S.R., Huffman, K., Marcus, C.B., Mulesh, W., Bruce, A., Baudry, M., Malfroy, B Salen-manganese complexes: Combined superoxide dismutase/catalase mimics with broad pharmacological efficacy In: Antioxidants in Disease Mechanisms and Therapeutic Strategies, ed Helmut Sies Academic Press, New York, 1997, pp 247–270 Doctrow, S.R., Huffman, K., Marcus, C.B., Tocco, G., Malfroy, E., Adinolfi, C.A., Kruk, H., Baker, K., Lazarowych, N., Mascarenhas, J., Malfroy, B Salen-manganese complexes as catalytic scavengers of hydrogen peroxide and cytoprotective agents: Structure-activity relationship studies J Med Chem 2002, 45, 4549–4558 Rosenthal, R.A., Huffman, K., Fisette, L.W., Damphouse, C.A., Callaway, W.B., Malfroy, B., Doctrow, S.R Orally available Mn porphyrins with superoxide dismutase and catalse activity J Biol Inorg Chem 2009, 14, 979–991 Pasternack, R.F., Banth, A., Pasternack, J.M., Johnson, C.S Catalysis of the disproportionation of superoxide by metalloporphyrins III J Inorg Biochem 1981, 15, 261– 267 Pasternack, R.F., Skowronek, W.R Catalysis of the disproportionation of superoxide by metalloporphyrins J Inorg Biochem 1979, 11, 261–267 Day, B.J., Fridovich, I., Crapo, J.D Maganic porphyrins possess catalase activity and protect endothelial cells against hydrogen peroxide-mediated injury Arch Biochem Biophys 1997, 347, 256–262 Groves, J.T., Marla, S.S Peroxynitrite-induced DNA strand scission mediated by a manganese porphyrin J Am Chem Soc 1995, 117, 9578–9579 Marla, S.S., Lee, J., Groves, J.T Peroxynitrite rapidly permeates phospholipid membranes Proc Natl Acad Sci U.S.A 1997, 94, 14243–14248 Groves, J.T., Lee, J., Marla, S.S Detection and characterization of an oxomanganese(V) porphyrin complex by REFERENCES rapid-mixing stopped-flow spectrophotometry J Am Chem Soc 1997, 119, 6269–6273 26 Lee, J., Hunt, J.A., Groves, J.T Manganese porphyrins as redox-coupled peroxynitrite reductases J Am Chem Soc 1998, 120, 6053–6061 27 Day, B.J., Batini-Haberle, I., Crapo, J.D Metalloporphyrins are potent inhibitors of lipid peroxidation Free Radic Biol Med 1999, 26, 730–736 28 Bloodsworth, A., O’Donnell, V.B., Batinic-Haberle, I., Chumley, P.H., Hurt, J.B., Day, B.J., Crow, J.P., Freeman, B.A Manganese-porphyrin reactions with lipids and lipoproteins Free Radic Biol Med 2000, 28, 1017–1029 29 Batinic-Haberle, I., Benov, L., Spasojevic, I., Fridovich, I The ortho effect makes manganese(III)meso-tetrakis(Nmethylpyridinium-2-yl)porphyrin a powerful and potentially useful superoxide dismutase mimic J Biol Chem 1998, 273, 24521–24528 30 Spasojevic, I., Batinic-Haberle, I., Rebouỗas, J.S., Idemori, Y.M., Fridovich, I Electrostatic contribution in the catalysis of O2− dismutation by superoxide dismutase mimics J Biol Chem 2003, 278, 6831–6837 31 Spasojevic, I., Chen, Y., Noel, T.J., Fan, P., Zhang, L., Rebouỗas, J.S., St Clair, D.K., Batinic-Haberle, I Pharmacokinetics of the potent redox-modulating manganese porphyrin, MnTE-2-PyP5+, in plasma and major organs of B6C3F1 mice Free Radic Biol Med 2008, 45, 943– 949 32 Batinic-Haberle, I., Ndengele, M.M., Cuzzocrea, S., Rebouỗas, J.S., Spasojevic, I., Salvemini, D Lipophilicity is a critical parameter that dominates the efficacy of metalloporphyrins in blocking the development of morphine antinociceptive tolerance through peroxynitritemediated pathways Free Radic Biol Med 2009, 46, 212–219 33 O’Neill, H.C., White, C.W., Veress, L.A., Hendry-Hofer, T.B., Loader, J.E., Min, E., Huang, J., Rancourt, R.C., Day, B.J Treatment with the catalytic metalloporphyrin AEOL 10150 reduces inflammation and oxidative stress due to inhalation of the sulfur mustard analog 2-chloroethyl ethyl sulfide Free Radic Biol Med 2010, 48, 1188–1196 34 Riley, D.P., Weiss, R.H Manganese macrocyclic ligand complexes as mimics of superoxide dismutase J Am Chem Soc 1994, 116, 387–388 35 Weiss, R.H., Fretland, D.J., Baron, D.A., Ryan, U.S., Riley, D.P Manganese-based superoxide dismutase mimetics inhibit neutrophil infiltration in vivo J Biol Chem 1996, 271, 26149–26156 36 Black, S.C., Schasteen, C.S., Weiss, R.H., Riley, D.P., Driscoll, E.M., Lucchesi, B.R Inhibition of in vivo myocardial ischemic and reperfusion injury by a synthetic manganese-based superoxide dismutase mimetic J Pharmacol Exp Ther 1994, 270, 1208–1215 37 Riley, D.P., Henke, S.L., Lennon, P.J., Aston, K Computeraided design (CAD) of synzymes: Use of molecular mechanics (MM) for the rational design of superoxide dismutase mimics Inorg Chem 1999, 38, 1908–1917 399 38 Riley, D.P., Lennon, P.J., Neumann, W.L., Weiss, R.H Toward the rational design of superoxide dismutase mimics: Mechanistic studies for the elucidation of substituent effects on the catalytic activity of macrocyclic manganese(II) complexes J Am Chem Soc 1997, 119, 6522–6528 39 Rauen, U., Li, T., Sustmann, R., de Groot, H Protection against iron- and hydrogen peroxide-dependent cell injuries by a novel synthetic iron catalase mimic and its precursor, the iron-free ligand Free Radic Biol Med 2004, 37, 1369–1383 40 Stern, M.K., Jensen, M.P., Kramer, K Peroxynitrite decomposition catalysts J Am Chem Soc 1996, 118, 8735–8736 41 Lee, J., Hunt, J.A., Groves, J.T Mechanisms of iron porphyrin reactions with peroxynitrite J Am Chem Soc 1998, 120, 7493–7501 42 Salvemini, D., Wang, Z.-Q., Stern, M.K., Currie, M.G., Misko, T.P Peroxynitrite decomposition catalysts: Therapeutics for peroxynitrite-mediated pathology Proc Natl Acad Sci U.S.A 1998, 95, 2659–2663 43 Filipovska, A., Kelso, G.F., Brown, S.E., Beer, S.M., Smith, R.A.J., Murphy, M.P Synthesis and characterization of a triphenylphosphonium-conjugated peroxidase mimetic J Biol Chem 2005, 280, 24113–24126 44 Imai, H., Masayasu, H., Dewar, D., Graham, D.I., Macrae, I.M Ebselen protects both gray and white matter in a rodent model of focal cerebral ischemia Stroke 2001, 32, 2149–2154 45 Ullrich, V., Weber, P., Meisch, F., von Appen, F Ebselenbinding equilibria between plasma and target proteins Biochem Pharmacol 1996, 52, 15–19 46 Lesser, R., Weiss, R Über selenhaltige aromatische Verbindungen (VI) Ber Dtsch Chem Ges 1924, 57, 1077 47 Weber, R., Renson, M Bull Soc Chim Fr 1976, 1024 48 Engman, L., Hallberg, A Expedient synthesis of ebselen and related compounds J Org Chem 1989, 54, 2964– 2966 49 Morgenstern, R., Cotgreave, I.A., Engman, L Determination of the relative contributions of the diselenide and selenol forms of ebselen in the mechanism of its glutathione peroxidase-like activity Chem Biol Interact 1992, 84, 77–84 50 Sies, H Ebselen, a selenoorganic compound as glutathione peroxidase mimic Free Radic Biol Med 1993, 14, 313–323 51 Sies, H., Lester, P [47] Ebselen: A glutathione peroxidase mimic In: Methods in Enzymology, Vol 234, ed Lester Packer Academic Press, San Diego, CA, 1994, pp 476–482 52 Schewe, T Molecular actions of Ebselen—An antiinflammatory antioxidant Gen Pharmacol 1995, 26, 1153– 1169 53 Haenen, G.R., De Rooij, B.M., Vermeulen, N.P., Bast, A Mechanism of the reaction of ebselen with endogenous thiols: Dihydrolipoate is a better cofactor than glutathione 400 54 55 56 57 58 59 60 61 62 63 64 65 66 SYNTHETIC ANTIOXIDANTS in the peroxidase activity of ebselen Mol Pharmacol 1990, 37, 412–422 Maiorino, M., Roveri, A., Coassin, M., Ursini, F Kinetic mechanism and substrate specificity of glutathione peroxidase activity of ebselen (PZ51) Biochem Pharmacol 1988, 37, 2267–2271 Schöneich, C., Narayanaswami, V., Asmus, K.-D., Sies, H Reactivity of ebselen and related selenoorganic compounds with 1,2-dichloroethane radical cations and halogenated peroxyl radicals Arch Biochem Biophys 1990, 282, 18–25 Maiorino, M., Roveri, A., Ursini, F Antioxidant effect of ebselen (PZ 51): Peroxidase mimetic activity on phospholipid and cholesterol hydroperoxides vs free radical scavenger activity Arch Biochem Biophys 1992, 295, 404–409 Noguchi, N., Yoshida, Y., Kaneda, H., Yamamoto, Y., Niki, E Action of ebselen as an antioxidant against lipid peroxidation Biochem Pharmacol 1992, 44, 39–44 Scurlock, R., Rougee, M., Bensasson, R.V., Evers, M., Dereu, N Deactivation of singlet molecular oxygen by organo-selenium compounds exhibiting glutathione peroxidase activity and by sulfur-containing homologs Photochem Photobiol 1991, 54, 733–736 Hiroshi, M., Kissner, R., Koppenol, W.H., Sies, H Kinetic study of the reaction of ebselen with peroxynitrite FEBS Lett 1996, 398(2–3), 179–182 Masumoto, H., Sies, H The reaction of ebselen with peroxynitrite Chem Res Toxicol 1996, 9, 262–267 Daiber, A., Zou, M.-H., Bachschmid, M., Ullrich, V Ebselen as a peroxynitrite scavenger in vitro and ex vivo Biochem Pharmacol 2000, 59, 153–160 Schewe, C., Schewe, T., Wendel, A Strong inhibition of mammalian lipoxygenases by the antiinflammatory seleno-organic compound ebselen in the absence of glutathione Biochem Pharmacol 1994, 48, 65–74 Cotgreave, I.A., Duddy, S.K., Kass, G.E.N., Thompson, D., Moldéus, P Studies on the anti-inflammatory activity of ebselen: Ebselen interferes with granulocyte oxidative burst by dual inhibition of NADPH oxidase and protein kinase C? Biochem Pharmacol 1989, 38, 649–656 Hattori, R., Inoue, R., Sase, K., Eizawa, H., Kosuga, K., Aoyama, T., Masayasu, H., Kawai, C., Sasayama, S., Yui, Y Preferential inhibition of inducible nitric oxide synthase by ebselen Eur J Pharmacol 1994, 267, R1–R2 Porciúncula, L.O., Rocha, J.B.T., Cimarosti, H., Vinadé, L., Ghisleni, G., Salbego, C.G., Souza, D.O Neuroprotective effect of ebselen on rat hippocampal slices submitted to oxygen-glucose deprivation: Correlation with immunocontent of inducible nitric oxide synthase Neurosci Lett 2003, 346, 101–104 Rossato, J.I., Zeni, G., Mello, C.F., Rubin, M.A., Rocha, J.B.T Ebselen blocks the quinolinic acid-induced production of thiobarbituric acid reactive species but does not prevent the behavioral alterations produced by intrastriatal quinolinic acid administration in the rat Neurosci Lett 2002, 318, 137–140 67 Yamaguchi, T., Sano, K., Takakura, K., Saito, I., Shinohara, Y., Asano, T., Yasuhara, H Ebselen in acute ischemic stroke: A placebo-controlled, double-blind clinical trial Stroke 1998, 29, 12–17 68 Saito, I., Asano, T., Sano, K., Takakura, K., Abe, H., Yoshimoto, T., Kikuchi, H., Ohta, T., Ishibashi, S Neuroprotective effect of an antioxidant, ebselen, in patients with delayed neurological deficits after aneurysmal subarachnoid hemorrhage Neurosurgery 1998, 42, 269–277 69 Zhao, R., Holmgren, A A novel antioxidant mechanism of ebselen involving ebselen diselenide, a substrate of mammalian thioredoxin and thioredoxin reductase J Biol Chem 2002, 277(42), 39456–39462 70 Zhao, R., Masayasu, H., Holmgren, A Ebselen: A substrate for human thioredoxin reductase strongly stimulating its hydroperoxide reductase activity and a superfast thioredoxin oxidant Proc Natl Acad Sci U.S.A 2002, 99, 8579–8584 71 Moutet, M., D’Alessio, P., Malette, P., Devaux, V., Chaudière, J Glutathione peroxidase Mimmics prevent TNFα- and neutrophil-induced endothelial alterations Free Radic Biol Med 1998, 25, 270–281 72 D’Alessio, P., Moutet, M., Marsac, C., Chaudière, J Pharmacological Inhibition of Endothelial Cytoskeleton Alterations Induced by Hydrogen Peroxide and TNF-α Marcel Dekker Inc., New York, 1997 73 D’Alessio, P., Moutet, M., Coudrier, E., Darquenne, S., Chaudiere, J ICAM-1 and VCAM-1 expression induced by TNF-α are inhibited by a glutathione peroxidase mimic Free Radic Biol Med 1998, 24, 979–987 74 Nogueira, C.W., Zeni, G., Rocha, J.B.T Organoselenium and organotellurium compounds: Toxicology and pharmacology Chem Rev 2004, 104, 6255–6286 75 Watanabe, T., Tahara, M., Todo, S The novel antioxidant edaravone: From bench to bedside Cardiovasc Ther 2008, 26, 101–114 76 Higashi, Y., Jitsuiki, D., Chayama, K., Yoshizumi, M Edaravone (3-methyl-1-phenyl-2-pyrazolin-5-one), a novel free radical scavenger, for treatment of cardiovascular diseases Recent Pat Cardiovas Drug Discov 2006, 1, 85–93 77 Chegaev, K., Cena, C., Giorgis, M., Rolando, B., Tosco, P., Bertinaria, M., Fruttero, R., Carrupt, P.-A., Gasco, A Edaravone derivatives containing NO-donor functions J Med Chem 2009, 52, 574–578 78 Nishinaka, Y., Mori, H., Endo, N., Miyoshi, T., Yamashita, K., Adachi, S., Arai, T Edaravone directly reacts with singlet oxygen and protects cells from attack Life Sci 2010, 86, 808–813 79 Komatsu, T., Nakai, H., Takamastu, Y., Morinaka, Y., Watanabe, K., Shinoda, M., Iida, S Pharmacokinetic studies of 3-methyl-1-phenyl-2-pyrazolin-5-one-(MCI-186): Metabolism in rats, dogs and human Drug Metab Pharmacokinet 1996, 11, 451–462 80 Abe, S., Kirima, K., Tsuchiya, K., Okamoto, M., Hasegawa, T., Houchi, H., Yoshizumi, M., Tamaki, T The reaction REFERENCES 81 82 83 84 85 86 87 88 89 90 91 92 93 rate of edaravone (3-methyl-1-phenyl-2-pyrazolin-5-one (MCI-186)) with hydroxyl radical Chem Pharm Bull 2004, 52(2), 186–191 Ono, S., Okazaki, K., Sakurai, M., Inoue, Y Density functional study of the radical reactions of 3-methyl-1-phenyl2-pyrazolin-5-one (MCI-186): Implication for the biological function of MCI-186 as a highly potent antioxidative radical scavenger J Phys Chem A 1997, 101, 3769–3775 Lin, M., Katsumura, Y., Hata, K., Muroya, Y., Nakagawa, K Pulse radiolysis study on free radical scavenger edaravone (3-methyl-1-phenyl-2-pyrazolin-5-one) J Photochem Photobiol B 2007, 89(1), 36–43 Yamamoto, Y., Kuwahara, T., Watanabe, K., Watanabe, K Antioxidant activity of 3-methyl-1-phenyl-2-pyrazoline5-one Redox Rep 1996, 2, 333–338 Wang, L.-F., Zhang, H.-Y A theoretical investigation on DPPH radical-scavenging mechanism of edaravone Bioorg Med Chem Lett 2003, 13, 3789–3792 Nakagawa, H., Ohyama, R., Kimata, A., Suzuki, T., Miyata, N Hydroxyl radical scavenging by edaravone derivatives: Efficient scavenging by 3-methyl-1-(pyridin2-yl)-5-pyrazolone with an intramolecular base Bioorg Med Chem Lett 2006, 16, 5939–5942 Satoh, K., Ikeda, Y., Shioda, S., Tobe, T., Yoshikawa, T Edaravone scavenges nitric oxide Redox Rep 2002, 7, 219–222 Anzai, K., Furuse, M., Yoshida, A., Matsumaya, A., Moritake, T., Tsuboi, K., Ikota, N In vivo radioprotection of mice by 3-methyl-1-phenyl-2-pyrazolin-5-one (edaravone; Radicut), a clinical drug J Radiat Res 2004, 45, 319–323 Murota, S., Morita, I., Suda, N The control of vascular endothelial cell injury Ann N Y Acad Sci 1990, 598, 182–187 Watanabe, T., Morita, I., Nishi, H., Murota, S Prenventive effect of MC-181 on 15-HPETE induced vascular endothelial cell injury in vivo Prostaglandins Leukot Essent Fatty Acids 1988, 33, 81–87 Yoshida, H., Sasaki, K., Namiki, Y., Sato, N., Tada, N Edaravone, a novel radical scavenger, inhibits oxidative modification of low-density lipoprotein (LDL) and reverses oxidized LDL-mediated reduction in the expression of endothelial nitric oxide synthase Atherosclerosis 2005, 179, 97–102 Zhang, X.-H., Matsuda, N., Jesmin, S., Sakuraya, F., Gando, S., Kemmotsu, O., Hattori, Y Normalization by edaravone, a free radical scavenger, of irradiationreduced endothelial nitric oxide synthase expression Eur J Pharmacol 2003, 476, 131–137 Takahashi, G., Sakurai, M., Abe, K., Itoyama, Y., Tabayashi, K MCI-186 prevents spinal cord damage and affects enzyme levels of nitric oxide synthase and Cu/Zn superoxide dismutase after transient ischemia in rabbits J Thorac Cardiovasc Surg 2003, 126, 1461–1466 Yanagisawa, A., Miyagawa, M., Ishikawa, K., Murota, S Cardioprotective effect of MC-186 (3-methyl-1-phenyl- 401 2-pyrazolin-5-one) during acute ischemia-reperfusion injury in rats Int J Angiol 1994, 3, 12–15 94 Minhaz, U., Tanaka, M., Tsukamoto, H., Watanabe, K., Koide, S., Shohtsu, A., Nakazawa, H Effect of MCI-186 on postischemic reperfusion injury in isolated rat heart Free Radic Res 1996, 24, 361–367 95 Tsujimoto, I., Hikoso, S., Yamaguchi, O., Kashiwase, K., Nakai, A., Takeda, T., Watanabe, T., Taniike, M., Matsumura, Y., Nishida, K., Hori, M., Kogo, M., Otsu, K The antioxidant edaravone attenuates pressure overloadinduced left ventricular hypertrophy Hypertension 2005, 45, 921–926 96 Tsujita, K., Shimomura, H., Kawano, H., Hokamaki, J., Fukuda, M., Yamashita, T., Hida, S., Nakamura, Y., Nagayoshi, Y., Sakamoto, T., Yoshimura, M., Arai, H., Ogawa, H Effects of edaravone on reperfusion injury in patients with acute myocardial infarction Am J Cardiol 2004, 94, 481–484 97 Yoshida, H., Yanai, H., Namiki, Y., Fukatsu-Sasaki, K., Furutani, N., Tada, N Neuroprotective effects of edaravone: A novel free radical scavenger in cerebrovascular injury CNS Drug Rev 2006, 12, 9–20 98 Otani, H., Togashi, H., Jesmin, S., Sakuma, I., Yamaguchi, T., Matsumoto, M., Kakehata, H., Yoshioka, M Temporal effects of edaravone, a free radical scavenger, on transient ischemia-induced neuronal dysfunction in the rat hippocampus Eur J Pharmacol 2005, 512, 129–137 99 The Edaravone Acute Brain Infarction Study Group Effect of a novel free radical scavenger, edaravone (MCI-186), on acute brain infarction Cerebrovasc Dis 2003, 15, 222–229 100 Takahashi, R Edaravone in ALS Exp Neurol 2009, 217, 235–236 101 Yoshino, H., Kimura, A Investigation of the therapeutic effects of edaravone, a free radical scavenger, on amyotrophic lateral sclerosis (Phase II study) Amyotroph Lateral Scler 2006, 7, 247–251 102 Ito, H., Wate, R., Zhang, J., Ohnishi, S., Kaneko, S., Ito, H., Nakano, S., Kusaka, H Treatment with edaravone, initiated at symptom onset, slows motor decline and decreases SOD1 deposition in ALS mice Exp Neurol 2008, 213, 448–455 103 Inokuchi, Y., Imai, S., Nakajima, Y., Shimazawa, M., Aihara, M., Araie, M., Hara, H Edaravone, a free radical scavenger, protects against retinal damage in vitro and in vivo J Pharmacol Exp Ther 2009, 329, 687–698 104 Imai, S., Inokuchi, Y., Nakamura, S., Tsuruma, K., Shimazawa, M., Hara, H Systemic administration of a free radical scavenger, edaravone, protects against lightinduced photoreceptor degeneration in the mouse retina Eur J Pharmacol 2010, 642, 77–85 105 Tanaka, K., Takemoto, T., Sugahara, K., Okuda, T., Mikuriya, T., Takeno, K., Hashimoto, M., Shimogori, H., Yamashita, H Post-exposure administration of edaravone attenuates noise-induced hearing loss Eur J Pharmacol 2005, 522, 116–121 402 SYNTHETIC ANTIOXIDANTS 106 Sueishi, K., Mishima, K., Makino, K., Itoh, Y., Tsuruya, K., Hirakata, H., Oishi, R Protection by a radical scavenger edaravone against cisplatin-induced nephrotoxicity in rats Eur J Pharmacol 2002, 451, 203–208 107 Fukudome, D., Matsuda, M., Kawasaki, T., Ago, Y., Matsuda, T The radical scavenger edaravone counteracts diabetes in multiple low-dose streptozotocin-treated mice Eur J Pharmacol 2008, 583, 164–169 108 Spapen, H., Zhang, H., Vincent, J.-L Potential therapeutic value of lazaroids in endotoxemia and other forms of sepsis Shock 1997, 8, 321–327 109 Braughler, J.M., Pregenzer, J.F., Chase, R.L., Duncan, L.A., Jacobsen, E.J., McCall, J.M Novel 21-amino steroids as potent inhibitors of iron-dependent lipid peroxidation J Biol Chem 1987, 262, 10438–10440 110 Braughler, J.M., Pregenzer, J.F The 21-aminosteroid inhibitors of lipid peroxidation: Reactions with lipid peroxyl and phenoxy radicals Free Radic Biol Med 1989, 7, 125–130 111 Hinzmann, J.S., McKenna, R.L., Pierson, T.S., Han, F., Kézdy, F.J., Epps, D.E Interaction of antioxidants with depth-dependent fluorescence quenchers and energy transfer probes in lipid bilayers Chem Phys Lipids 1992, 62, 123–138 112 Noguchi, N., Takahashi, M., Tsuchiya, J., Yamashita, H., Komuro, E., Niki, E Action of 21-aminosteroid U74006F as an antioxidant against lipid peroxidation Biochem Pharmacol 1998, 55, 785–791 113 Audus, K.L., Guillot, F.L., Mark Braughler, J Evidence for 21-aminosteroid association with the hydrophobic domains of brain microvessel endothelial cells Free Radic Biol Med 1991, 11, 361–371 114 Braughler, J.M., Chase, R.L., Neff, G.L., Yonkers, P.A., Day, J.S., Hall, E.D., Sethy, V.H., Lahti, R.A A new 21-aminosteroid antioxidant lacking glucocorticoid activity stimulates adrenocorticotropin secretion and blocks arachidonic acid release from mouse pituitary tumor (AtT-20) cells J Pharmacol Exp Ther 1988, 244, 423– 427 115 Munns, P.L., Leach, K.L Two novel antioxidants, U74006F and U78517F, inhibit oxidant-stimulated calcium influx Free Radic Biol Med 1995, 18, 467–478 116 Lesiuk, H., Sutherland, G., Peeling, J., Butler, K., Saunders, J Effect of U74006F on forebrain ischemia in rats Stroke 1991, 22, 896–901 117 Perkins, W.J., Milde, L.N., Milde, J.H., Michenfelder, J.D Pretreatment with U74006F improves neurologic outcome following complete cerebral ischemia in dogs Stroke 1991, 22, 902–909 118 Hall, E.D., Vaishnav, R.A., Mustafa, A.G Antioxidant therapies for traumatic brain injury Neurotherapeutics 2010, 7, 51–61 119 Holzgrefe, H.H., Buchanan, L.V., Gibson, J.K Effects of U74006F, a novel inhibitor of lipid peroxidation, in stunned reperfused canine myocardium J Cardiovasc Pharmacol 1990, 15, 239–248 120 Ovize, M., de Lorgeril, M., Ovize, A., Ciavatti, M., Delaye, J., Renaud, S U74006F, a novel 21-aminosteroid, inhibits in vivo lipid peroxidation but fails to limit infarct size in a canine model of myocardial ischemia reperfusion Am Heart J 1991, 122, 681–689 121 Levitt, M.A., Sievers, R.E., Wolfe, C.L Reduction of infarct size during myocardial ischemia and reperfusion by lazaroid U-74500A, a nonglucocorticoid 21-aminosteroid J Cardiovasc Pharmacol 1994, 23, 136–140 122 Liu, P., Vonderfecht, S.L., McGuire, G.M., Fisher, M.A., Farhood, A., Jaeschke, H The 21-aminosteroid tirilazad mesylate protects against endotoxin shock and acute liver failure in rats J Pharmacol Exp Ther 1994, 271, 438–445 123 McGuire, G.M., Liu, P., Jaeschke, H Neutrophil-induced lung damage after hepatic ischemia and endotoxemia Free Radic Biol Med 1996, 20, 189–197 124 Wang, Y., Mathews, W.R., Guido, D.M., Jaeschke, H The 21-aminosteroid tirilazad mesylate protects against liver injury via membrane stabilization not inhibition of lipid peroxidation J Pharmacol Exp Ther 1996, 277, 714–720 125 Zhang, H., Spapen, H., Manikis, P., Rogiers, P., Metz, G., Buurman, W.A., Vincent, J.L Tirilazad mesylate (U-74006F) inhibits effects of endotoxin in dogs Am J Physiol Heart Circ Physiol 1995, 268, 1847–1855 126 Bath, P.M.W., Blecic, S., Bogousslavsky, J., Boysen, G., Davis, S., Diez-Tejedor, E., Ferro, J.M., Gommans, J., Hacke, W., Indredavik, B., Norrving, B., Orgogozo, J.M., Ringelstein, E.B., Sacchetti, M.L., Iddenden, R., Bath, F.J., Musch, B.C., Brosse, D.M., Naberhuis-Stehouwer, S.A Tirilazad mesylate in acute ischemic stroke: Asystematic review Stroke 2000, 31, 2257–2265 127 Hall, E.D., Pazara, K.E., Braughler, J.M 21-Aminosteroid lipid peroxidation inhibitor U74006F protects against cerebral ischemia in gerbils Stroke 1988, 19, 997–1002 128 Beck, T., Bielenberg, G.W Failure of the lipid peroxidation inhibitor U74006F to improve neurological outcome after transient forebrain ischemia in the rat Brain Res 1990, 532, 336–338 129 Reith, J., Jorgensen, H.S., Pedersen, P.M., Nakamaya, H., Jeppesen, L.L., Olsen, T.S., Raaschou, H.O Body temperature in acute stroke: Relation to stroke severity, infarct size, mortality, and outcome Lancet 1996, 347, 422–425 130 Marshall, L.F., Maas, A.I.R., Marshall, S.B., Bricolo, A., Fearnside, M., Iannotti, F., Klauber, M.R., Lagarrigue, J., Lobato, R., Persson, L., Pickard, J.D., Piek, J., Servadei, F., Wellis, G.N., Morris, G.F., Means, E.D., Musch, B A multicenter trial on the efficacy of using tirilazad mesylate in cases of head injury J Neurosurg 1998, 89, 519–525 131 Haley, E.C., Kassell, N.F., Apperson-Hansen, C., Maile, M.H., Alves, W.M A randomized, double-blind, vehiclecontrolled trial of tirilazad mesylate in patients with aneurysmal subarachnoid hemorrhage: A cooperative study in North America J Neurosurg 1997, 86, 467–474 132 Kassell, N.F., Haley, E.C., Apperson-Hansen, C., Alves, W.M Randomized, double-blind, vehicle-controlled trial REFERENCES 133 134 135 136 137 138 139 140 141 142 143 144 of tirilazad mesylate in patients with aneurysmal subarachnoid hemorrhage: A cooperative study in Europe, Australia, and New Zealand J Neurosurg 1996, 84, 221–228 Lanzino, G., Kassell, N.F., Dorsch, N.W.C., Pasqualin, A., Brandt, L., Schmiedek, P., Truskowski, L.L., Alves, W.M Double-blind, randomized, vehicle-controlled study of high-dose tirilazad mesylate in women with aneurysmal subarachnoid hemorrhage Part I A cooperative study in Europe, Australia, New Zealand, and South Africa J Neurosurg 1999, 90, 1011–1017 Lanzino, G., Kassell, N.F Double-blind, randomized, vehicle-controlled study of high-dose tirilazad mesylate in women with aneurysmal subarachnoid hemorrhage Part II A cooperative study in North America J Neurosurg 1999, 90, 1018–1024 Janzen, E.G Spin trapping Acc Chem Res 1971, 4, 31–40 Villamena, F.A., Zweier, J.L Detection of reactive oxygen and nitrogen species by EPR spin trapping Antioxid Redox Signal 2004, 6, 619–629 Fréjaville, C., Karoui, H., Tuccio, B., Moigne, F.L., Culcasi, M., Pietri, S., Lauricella, R., Tordo, P 5(Diethoxyphosphoryl)-5-methyl-1-pyrroline N-oxide: A new efficient phosphorylated nitrone for the in vitro and in vivo spin trapping of oxygen-centered radicals J Med Chem 1995, 38, 258–265 Olive, G., Mercier, A., Le Moigne, F., Rockenbauer, A., Tordo, P 2-ethoxycarbonyl-2-methyl-3,4-dihydro-2Hpyrrole-1-oxide: Evaluation of the spin trapping properties Free Radic Biol Med 2000, 28, 403–408 Zhao, H., Joseph, J., Zhang, H., Karoui, H., Kalyanaraman, B Synthesis and biochemical applications of a solid cyclic nitrone spin trap: A relatively superior trap for detecting superoxide anions and glutathiyl radicals Free Radic Biol Med 2001, 31, 599–606 Villamena, F.A., Rockenbauer, A., Gallucci, J., Velayutham, M., Hadad, C.M., Zweier, J.L Spin trapping by 5-carbamoyl-5-methyl-1-pyrroline N-oxide (AMPO): Theoretical and experimental studies J Org Chem 2004, 69, 7994–8004 Villamena, F.A., Xia, S., Merle, J.K., Lauricella, R., Tuccio, B., Hadad, C.M., Zweier, J.L Reactivity of superoxide radical anion with cyclic nitrones: Role of intramolecular H-Bond and electrostatic effects J Am Chem Soc 2007, 129, 8177–8191 Zeghdaoui, A., Tuccio, B., Finet, J.-P., Cerri, V., Tordo, P β-Phosphorylated α-phenyl-N-tert-butylnitrone (PBN) analogs: A new series of spin traps for oxyl radicals J Chem Soc Perkin Trans 1995, 12, 2087–2089 Roubaud, V., Lauricella, R., Tuccio, B., Bouteiller, J.-C., Tordo, P Decay of superoxide spin adducts of new PBNtype phosphorylated nitrones Res Chem Intermed 1996, 22, 405–416 Hinton, R.D., Janzen, E.G Synthesis and characterization of phenyl-substituted C-phenyl-N-tert-butylnitrones and some of their radical adducts J Org Chem 1992, 57, 2646–2651 403 145 Rosen, G.M., Britigan, B.E., Halpern, H.J., Pou, S Free Radicals: Biology and Detection by Spin Trapping Oxford Univeristy Press, New York, 1999 146 Durand, G., Poeggeler, B., Böker, J., Raynal, S., Polidori, A., Pappolla, M.A., Hardeland, R., Pucci, B Fine-tuning the amphiphilicity: A crucial parameter in the design of potent alpha-phenyl-N-tert-butylnitrone analogues J Med Chem 2007, 50, 3976–3979 147 Durand, G., Polidori, A., Ouari, O., Tordo, P., Geromel, V., Rustin, P., Pucci, B Synthesis and preliminary biological evaluations of ionic and nonionic amphiphilic alphaphenyl-N-tert-butylnitrone derivatives J Med Chem 2003, 46, 5230–5237 148 Durand, G., Polidori, A., Salles, J.-P., Pucci, B Synthesis of a new family of glycolipidic nitrones as potential antioxidant drugs for neurodegenerative disorders Bioorg Med Chem Lett 2003, 13, 859–862 149 Poeggeler, B., Durand, G., Polidori, A., Pappolla, M.A., Vega-Naredo, I., Coto-Montes, A., Boeker, J., Hardeland, R., Pucci, B Mitochondrial medicine: Neuroprotection and life extension by the new amphiphilic nitrone LPBNAH acting as a highly potent antioxidant agent J Neurochem 2005, 95, 962–973 150 Bernotas, R.C., Thomas, C.E., Carr, A.A., Nieduzak, T.R., Adams, G., Ohlweiler, D.F., Hay, D.A Synthesis and radical scavenging activity of 3,3-dialkyl-3,4-dihydroisoquinoline 2-oxides Bioorg Med Chem Lett 1996, 6, 1105–1110 151 Janzen, E.G., Poyer, J.L., Schaefer, C.F., Downs, P.E., BuBose, C.M Biological spin trapping II Toxicity of nitrones spin traps: Dose-ranging in the rat J Biochem Biophys Methods 1995, 30, 239–247 152 Schaefer, C.F., Janzen, E.G., West, M.S., Poyer, J.L., Kosanke, S.D Blood chemistry changes in the rat induced by high doses of nitronyl free radicals spin traps Free Radic Biol Med 1996, 21, 427–436 153 Haseloff, R.F., Mertsch, K., Rohde, E., Baeger, I., Grigor’ev, I.A., Blasig, I.E Cytotoxicity of spin trapping compounds FEBS Lett 1997, 418, 73–75 154 Chen, G., Bray, T.M., Janzen, E.G., McCay, P.B Excretion, metabolism and tissue distribution of a spin trapping agent, α-phenyl-N-tert-butyl-nitrone (PBN) in rats Free Radic Res Commun 1990, 9(3–6), 317–323 155 Cheng, H.-Y., Liu, T., Feurerstein, G., Barone, F.C Distribution of spin-trapping compounds in rat blood and brain: In vivo microdialysis determination Free Radic Biol Med 1993, 14, 243–250 156 Liu, K.J., Kotake, Y., Lee, M., Miyake, M., Sugden, K., Yu, Z., Swartz, H.M High-performance liquid chromatography study of the pharmacokinetics of various spin traps for application to in vivo spin trapping Free Radic Biol Med 1999, 27, 82–89 157 Trudeau-Lame, M.E., Kalgutkar, A.S., LaFontaine, M Pharmakokinetics and metabolism of the reactive oxygen scavenger α-phenyl-N-tert-butylnitrone in the male sprague-dawley Drug Metab Dispos 2003, 31, 147–152 404 SYNTHETIC ANTIOXIDANTS 158 Cova, D., De Angelis, L., Monti, E., Piccinini, F Subcellular distribution of two spin trapping agents in rat heart: Possible explanation for their different protective effects against doxorubicin-induced cardiotoxicity Free Radic Res Commun 1992, 15, 353–360 159 Reinke, L.A., Moore, D.R., Sang, H., Janzen, E.G., Kotake, Y Aromatic hydroxylation in PBN spin trapping by hydroxyl radicals and cytochrome P-450 Free Radic Biol Med 2000, 28, 345–350 160 Novelli, G.P., Angiolini, P., Tani, R., Consales, G., Bordi, L Phenyl-tert-butylnitrone is active against traumatic shock in rats Free Radic Res Commun 1986, 1, 321–327 161 Floyd, R.A., Hensley, K., Forster, M.J., KelleherAndersson, J.A., Wood, P.L Nitrones, their value as therapeutics and probes to understand aging Mech Ageing Dev 2002, 123, 1021–1031 162 Kotake, Y Pharmacologic properties of phenyl N-tertbutylnitrone Antioxid Redox Signal 1999, 1, 481–499 163 Floyd, R.A Nitrones as therapeutics in age-related diseases Aging Cell 2006, 5, 51–57 164 Floyd, R.A., Kopke, R.D., Choi, C.-H., Foster, S.B., Doblas, S., Towner, R.A Nitrones as therapeutics Free Radic Biol Med 2008, 45, 1361–1374 165 Hamburger, S.A., McCay, P.B Endotoxin-induced mortality in rats is reduced by nitrones Circ Shock 1989, 29, 329–334 166 McKechnie, K., Furman, B.L., Parrat, J.R Modification by oxygen free radical scavengers of the metabolic and cardiovascular effects of endotoxin infusion in conscious rats Circ Shock 1986, 19, 429–439 167 Progrebniak, H.W., Merino, M.J., Hahn, S.M., Mitchell, J.B., Pass, H.I Spin trap salvage from endotoxemia: The role of cytokine down-regulation Surgery 1992, 112, 130–139 168 Iovino, G., Kubow, S., Marliss, E.B Effect of α-phenyl -N-tert-butylnitrone on diabetes and lipid peroxidation in BB rats Can J Physiol Pharmacol 1999, 77, 166–172 169 Ho, E., Chen, G., Bray, T.M Alpha-phenyl-tertbutylnitrone (PBN) inhibits NFκB activation offering protection against chemically induced diabetes Free Radic Biol Med 2000, 28, 604–614 170 Jotti, A., Paracchini, L., Perletti, G., Piccinini, F Cardiotoxicity induced by doxorubicin in vivo: Protective activity of the spin trap alpha-phenyl-tert-butyl nitrone Pharmacol Res 1992, 26, 143–150 171 Kotamraju, S., Konorev, E., Joseph, J., Kalyanaraman, B Doxorubicin-induced apoptosis in endothelial cells and cardiomycetes is ameliorated by nitrone spin traps and ebselen J Biol Chem 2000, 275, 33585–33592 172 Parman, T., Wiley, M.J., Wells, P.G Free radical-medical oxidative DNA damage in the mechanism of thalidomide teratogenicity Nat Med 1999, 5, 582–585 173 Rao, D.B., Fechter, L.D Protective effects of phenyl N-tert-butylnitrone on the potentiation of noise-induced hearing loss by carbone monoxide Toxicol Appl Pharmacol 2000, 167, 125–131 174 Pouyatos, B., Gearhart, C.A., Fechter, L.D Acrylonitrile potentiates hearing loss and cochlear damage induced by moderate noise exposure Toxicol Appl Pharmacol 2005, 204, 46–56 175 Fechter, L.D., Liu, Y., Pearce, T.A Cochlear protection from carbon monoxide exposure by free radical blockers in the guinea pig Toxicol Appl Pharmacol 1997, 142, 47–55 176 Choi, C.-H., Chen, K., Vasquez-Weldon, A., Jackson, R.L., Floyd, R.A., Kopke, R.D Effectiveness of 4-hydroxy phenyl N-tert-butylnitrone (4-OHPBN) alone and in combination with other antioxidant drugs in the treatment of acute acoustic trauma in chinchilla Free Radic Biol Med 2008, 44, 1772–1784 177 Ranchon, I., Chen, S., Alvarez, K., Anderson, R.E Systemic administration of phenyl-N-tert-butylnitrone protects the retina from light damage Invest Ophthalmol Vis Sci 2001, 46, 427–434 178 Tomita, H., Kotake, Y., Anderson, R.E Mechanism of protection from light-induced retinal degeneration by the synthetic antioxidant phenyl-N-tert-butylnitrone Invest Ophthalmol Vis Sci 2005, 46, 427–434 179 Choteau, F., Durand, G., Ranchon-Cole, I., Cercy, C., Pucci, B Cholesterol-based α-phenyl-N-tert-butyl nitrone derivatives as antioxidants against light-induced retinal degeneration Bioorg Med Chem Lett 2010, 20, 7405– 7409 180 Ranchon, I., LaVail, M.M., Kotake, Y., Anderson, D.E Free radical trap phenyl-N-tert-butylnitrone protects against light damage but does not rescue P23H and S334ter rhodopsin transgenic rats from inherited retinal degeneration J Neurosci 2003, 23, 6050–6057 181 Yamashita, T., Ohshima, H., Asanuma, T., Inukai, N., Miyoshi, I., Kasai, N., Kon, Y., Watanabe, T., Sato, F., Kuwabara, M The effects of α-phenyl-N-tert-butyl nitrone (PBN) on copper induced rat fulminant hepatitis with jaundice Free Radic Biol Med 1996, 26, 755–761 182 Asanuma, T., Yasui, H., Inanami, O., Waki, K., Takahashi, M., Iizuka, D., Uemura, T., Durand, G., Polidori, A., Kon, Y., Pucci, B., Kuwabara, M A new amphiphilic derivative, N -{[4-(lactobionamido)methyl]benzylidene}-1,1dimethyl-2-(octylsulfanyl)ethylamine N-oxide, has a protective effect against copper-induced fulminant hepatitis in Long-Evans cinnamon rats at an extremely low concentration compared with its original form α-phenyl-N(tert-butyl) nitrone Chem Biodivers 2007, 4, 2253– 2267 183 Tosaki, A., Blasig, I.E., Pali, T., Ebert, B Heart protection and radical trapping by DMPO during reperfusion in isolated working rat hearts Free Radic Biol Med 1990, 8, 363–372 184 Zhuo, L., Chen, Y.R., Reyes, L.A., Lee, H.L., Chen, C.L., Villamena, F.A., Zweier, J.L The radical spin-trap 5,5-dimethyl-1-pyrroline N-oxide exerts dose-dependant protection against myocardial ischemia-reperfusion injury trough preservation of mitochondrial electron transport J Pharmacol Exp Ther 2009, 329, 515–523 REFERENCES 185 Tanguy, S., Durand, G., Reboul, C., Polidori, A., Pucci, B., Dauzat, M., Obert, P Protection against reactive oxygen species injuries in rat isolated perfused hearts: Effect of LPBNAH, a new amphiphilic spin-trap derived from PBN Cardiovasc Drugs Ther 2006, 20, 147–149 186 Edamatsu, R., Mori, A., Packer, L The spin-trap N-tertα-phenyl-butylnitrone prolongs the life span of the senescence accelerated mouse Biochem Biophys Res Commun 1995, 221, 847–849 187 Saito, K., Yoshioka, H., Cutler, R.G A spin trap, N-tertbutyl-α-phenylnitrone extends the life span of mice Biosci Biotechnol Biochem 1998, 62, 792–794 188 Sack, C.A., Socci, D.J., Crandall, B.M., Arendash, G.W Antioxidant treatment with phenyl-α-tert-butyl nitrone (PBN) improves the cognitive performance Neurosci Lett 1996, 205, 181–184 189 Atamna, H., Paler-Martinez, A., Ames, B.N N-t-butylhydroxylamine, a hydrolysis product of a-phenyl-N-t-butyl nitrone, is more potent in delaying senescence in human lung fibroblasts J Biol Chem 2000, 275, 6741–6748 190 von Zglinicki, T., Pilger, R., Sitte, N Accumulation of single-strand breaks is the major cause of telomere shortening in human fibroblasts Free Radic Biol Med 2000, 28, 64–74 191 Floyd, R.A., Hensley, K., Forster, M.J., Kelleher-Andersson, J.A., Wood, P.L Nitrones as neuroprotectants and antiaging drugs Ann N Y Acad Sci 2002, 959, 321–329 192 Floyd, R.A Role of oxygen free radicals in carcinogenesis and brain ischemia FASEB J 1990, 4, 2587–2597 193 Carney, J.M., Floyd, R.A Phenyl butyl nitrone compositions and methods for treatment of oxidative tissue damage, US Patent 5025032 Issued 18 June 1991 194 Phillis, J.W., Clough-Helfman, C Protection from cerebral ischemic injury in gerbils with the spin trap agent N-tert-butyl-α-phenylnitrone (PBN) Neurosci Lett 1990, 116, 315–319 195 Cao, X., Phillis, J.W α-Phenyl-tert-butyl nitrone reduces cortical infarct and edema in rats subjected to focal ischemia Brain Res 1994, 644, 267–272 196 Mori, H., Arai, T., Ishii, H., Adachi, T., Endo, N., Makino, K., Mori, K Neuroprotective effects of pterin-6-aldehyde in gerbil global brain ischemia: Comparison with those of [alpha]-phenyl-N-tert-butyl nitrone Neurosci Lett 1998, 241(2–3), 99–102 197 Yue, T.-L., Gu, J.-L., Lysko, P.G., Cheng, H.-Y., Barone, F.C., Feuerstein, G Neuroprotective effects of phenyl-tbutyl-nitrone in gerbil global brain ischemia and in cultured rat cerebellar neurons Brain Res 1992, 574, 193–197 198 Zhao, Q., Pahlmark, K., Smith, M.-I., Siesjo, B.K Delayed treatment with the spin trap α-phenyl-N-tert-butyl nitrone (PBN) reduces infarct size following transient middle cerebral artery occlusion in rats Acta Physiol Scand 1994, 152, 349–350 199 Nakao, N., Grabson-Frodl, E.M., Widner, H., Brundin, P Antioxidant treatment protects striatal neurons against excitotoxic insults Neuroscience 1996, 73, 185–200 405 200 Nakao, N., Brundin, P Effects of α-phenyl-tert-butylnitrone on neuronal survival and motor function following intrastriatal injections of quinolinic or 3nitropropoionic acid Neuroscience 1997, 76, 749–761 201 Schulz, J.B., Henshaw, D.R., Siwek, D., Jenkins, B.G., Ferrante, R.J., Cipolloni, P.B., Kowall, N.W., Rosen, B.R., Beal, M.F Involvement of free radicals in excitotoxicity in vivo J Neurochem 1995, 64, 2239–2247 202 Marklund, N., Clausen, F., McIntosh, T.K., Hillered, L Free radical scavenger post-treatment improves functional and morphological outcome after fluid percussion injury in the rat J Neurotrauma 2001, 19, 821–832 203 Marklund, N., Lewander, T., Clausen, F., Hillered, L Effects of the nitrone radical scavengers PBN and S-PBN on in vivo trapping of reactive oxygen species after traumatic brain injury in rats J Cereb Blood Flow Metab 2001, 21, 1259–1267 204 Green, A.R., Ashwood, T., Odergren, T., Jackson, D.M Nitrones as neuroprotective agents in cerebral ischemia, with particular reference to NXY-059 Pharmacol Ther 2003, 100, 195–214 205 Kuroda, S., Tsuchidate, R., Smith, M.-L., Maples, K.R., Siesjo, B.K Neuroprotective effects of a novel nitrone, NXY-059, after transient focal cerebral ischemia in the rat J Cereb Blood Flow Metab 1999, 19, 778–787 206 Sydserff, S.G., Borelli, A.R., Green, A.R., Cross, A.J Effect of NXY-059 on infarct volume after transient or permanent middle cerebral artery occlusion in the rat; studies on dose, plasma concentration and therapeutic time window Br J Pharmacol 2002, 135, 103–112 207 Marshall, J.W.B., Duffin, K.J., Green, A.R., Ridley, R.M., Finklestein, S.P NXY-059, a free radical-trapping agent, substantially lessens the functional disability resulting from cerebral ischemia in a primate species Stroke 2001, 32, 190–198 208 Marshall, J.W.B., Cummings, R.M., Bowes, L.J., Ridley, R.M., Green, A.R Functional and Histological evidence for the protective effect of NXY-059 in a primate model of stroke when given hours after occlusion Stroke 2003, 34, 2228–2233 209 Edenius, C., Strid, S., Borgå, O., Breitholtz-Emanuelsson, A., Vallén, K.L., Fransson, B Pharmacokinetics of NXY059, a nitrone-based free radical trapping agent, in healthy young and elderly subjects J Stroke Cerebrovasc Dis 2002, 11, 34–43 210 Lees, K.R., Sharma, A.K., Barer, D., Ford, G.A., Kostulas, V., Cheng, Y.F., Odergren, T Tolerability and pharmacokinetics of the nitrone NXY-059 in patients with acute stroke Stroke 2001, 32, 675–680 211 Lees, K.R., Barer, D., Ford, G.A., Hacke, W., Kostulas, V., Sharma, A.K., Odergren, T Tolerability of NXY-059 at Higher target concentrations in patients with acute stroke Stroke 2003, 34, 482–487 212 Lees, K.R., Zivin, J.A., Ashwood, T., Davalos, A., Davis, S.M., Diener, H.-C., Grotta, J., Lyden, P., Shuaib, A., Hårdemark, H.-G., Wasiewski, W.W NXY-059 for acute ischemic stroke N Engl J Med 2006, 354, 588–600 406 SYNTHETIC ANTIOXIDANTS 213 Shuaib, A., Lees, K.R., Lyden, P., Grotta, J., Davalos, A., Davis, S.M., Diener, H.-C., Ashwood, T., Wasiewski, W.W., Emeribe, U NXY-059 for the treatment of acute ischemic stroke N Engl J Med 2007, 357, 562–571 214 Lees, K.R., Davalos, A., Davis, S.M., Diener, H.-C., Grotta, J., Lyden, P., Shuaib, A., Ashwood, T., Hårdemark, H.-G., Wasiewski, W., Emeribe, U., Zivin, J.A Additional outcomes and subgroup analyses of NXY-059 for acute ischemic stroke in the SAINT I trial Stroke 2006, 37, 2970–2978 215 Diener, H.-C., Lees, K.R., Lyden, P., Grotta, J., Davalos, A., Davis, S.M., Shuaib, A., Ashwood, T., Wasiewski, W., Alderfer, V., Hårdemark, H.-G., Rodichok, L NXY-059 for the treatment of acute stroke: Pooled analysis of the SAINT I and II trials Stroke 2008, 39, 1751–1758 216 Savitz, S.I A critical appraisal of the NXY-059 neuroprotection studies for acute stroke: A need for more rigorous testing of neuroprotective agents in animal models of stroke Exp Neurol 2007, 205, 20–25 217 Floyd, R.A., Carney, J.M Nitrone Radical Traps Protect in Experimental Neurodegenerative Diseases Academin Press, London, 1996 218 Barclay, L.R.C., Vinqvist, M.R Do spin traps also act as classical chain-breaking antioxidants? A quantitative kinetic study of phenyl-tert-butylnitrone (PBN) in solution and in liposomes Free Radic Biol Med 2000, 28, 1079–1090 219 Janzen, E.G., West, M.S., Poyer, J.L Comparison of Antioxidant Activity of PBN with Hindered Phenols in Initiated Rat Liver Microsomal Lipid Peroxidation Elsevier, San Diego, CA, 1994 220 Kotake, Y., Sang, H., Miyajima, T., Wallis, G.L Inhibition of NF-kB, iNOS mRNA, COX2 mRNA, and COX catalytic activity by phenyl-N-tert-butylnitrone (PBN) Biochim Biophys Acta 1998, 1448, 77–84 221 Sang, H., Wallis, G.L., Stewart, C.A., Kotake, Y Expression of cytokines and activation of transcription factors in lipopolysaccharide-administered rats and their inhibition by phenyl N-tert-butylnitrone (PBN) Arch Biochem Biophys 1999, 363, 341–348 222 Floyd, R.A., Hensley, K., Jaffrey, F., Maidt, L., Robinson, K., Pye, Q., Stewart, C Increased oxidative stress brought on by pro-inflammatory cytokines in neurodegenerative 223 224 225 226 227 228 229 230 processes and the protective role of nitrone-based free radicals traps Life Sci 1999, 65, 1893–1899 Floyd, R.A., Hensley, K., Bing, G Evidence for enhanced neuro-inflammatory processes in neurodegenerative diseases and the action of nitrones as potential therapeutics J Neural Transm 2000, 60, 337–364 Anderson, D.E., Yuan, X.J., Tseng, C.M., Rubin, L.J., Rosen, G.M., Tod, M.L Nitrone spin-traps block calcium channels and induce pulmonary artery relaxation independant of free radicals Biochem Biophys Res Commun 1993, 193, 878–885 Milatovic, D., Radic, Z., Zivin, M., Dettbarn, W.D Atypical effect of some spin trapping agents: Reversible inhibition of acetylcholinesterase Free Radic Res Commun 2000, 28, 597–603 Hensley, K., Pye, Q.N., Maidt, M.L., Stewart, C.A., Robinson, K.A., Jaffrey, F., Floyd, R.A Interaction of α-phenyl-N-tert-butyl nitrone and alternative electron acceptors with complex I indicates a substrate reduction site upstream from the rotenone binding site J Neurochem 1998, 71, 2549–2557 Kim, H.Y., Chung, J.M., Chung, K Increased production of mitochondrial superoxide in the spinal cord induces pain behaviors in mice: The effect of mitochondrial electron transport complex inhibitors Neurosci Lett 2008, 447, 87–91 Wen, J.-J., Bhatia, V., Popov, V.L., Garg, N.J Phenyl{alpha}-tert-butyl nitrone reverses mitochondrial decay in acute Chagas’ disease Am J Pathol 2006, 169, 1953–1964 Wen, J.-J., Garg, N Mitochondrial generation of reactive oxygen species is enhanced at the Q0 site of the complex III in the myocardium of Trypanosoma cruzi-infected mice: Beneficial effects of an antioxidant J Bioenerg Biomembr 2008, 40, 587–598 Durand, G., Poeggeler, B., Ortial, S., Polidori, A., Villamena, F.A., Böker, J., Hardeland, R., Pappolla, M.A., Pucci, B Amphiphilic amide nitrones: A new class of protective agents acting as modifiers of mitochondrial metabolism J Med Chem 2010, 53, 4849–4861 INDEX ABCC7, 345 Acetylcholine, 194 Acetylsalicylic acid (ASA, aspirin), 248–249, 252–553, 262 Acidosis, intracellular, 317 Acrolein, 58, 61–62 Actin, 74, 78 Activator protein 1, 346 Adderall, 256 Adenine, 96 Adenosine, 316, 321 Adenosine diphosphate (ADP), 27, 114, 123–125, 316, 318 Adenosine monophosphate (AMP), 27, 316, 318 cyclic, 243, 245, 352 Adenosine triphosphate (ATP), 27, 73, 114, 123–125, 130, 132, 190, 220, 238, 240–241, 316, 318–319, 352 synthase, 362–364, 397, Adenylation, 78 ADMA, see Asymmetric dimethylarginine Advanced lipidation end products (ALE), 80 Aflatoxin B1, 250 Aging, 237, 252 and DNA repair, 268–269 and METH, 262 Airway inflammation, 347 obstructions, 348 Alanine, 318 Alcohol dehydrogenase class III (ADH), 74 Aldehyde oxidase (AO), 314, 316, 318 Aldehydes in atherosclerosis, 329–335 in I-R, 314 Alkoxyl radical, 51–52, 58, 62 Allopurinol, 318, 320 See also Inhibitors Allysine, 79 Alzheimer’s disease, 80, 194, 238–241, 250, 253–254, 263, 266, 277–278, 360 Amadori rearrangements, 80 Amino acids, free, 318–319 Aminoglycoside, 351 2-amino-3-keto butyric acid, 80 3-amino-1,2,4-triazole, 117 Amphetamine(s), 252, 255–263 affinities to uptake transporters, 260 bioactivation by PHSs, 252–253 history, 255–256 metabolism by CYPs, 257–259 neurotoxicity, 261–263 pharmacokinetics, 256–257 SOD protection against toxicity, 266 uses, 255–256 AMPO, 390 See also Nitrones Amyotrophic lateral sclerosis (ALS), 239, 241, 253–4, 277–278, 360 Angiotensin II, 150, 152–156, 158 Antibiotics, 181 Antibody, 127–128 Antidepressants, 240 Antiinflammatory cytokines, 346 Antimicrobial, 346–350 Antioxidant response element (ARE), 269–270, 274–275 Antioxidants in cell signaling, 179, 180, 182, 185–186, 188, 190, 193–195 enzymes, 94, 114–116, 263–268, 312–313, 318, 321 synthetic, 377–398 Antipsychotics, 240 AP-1, see Activator protein Apical transporter, 348 Molecular Basis of Oxidative Stress: Chemistry, Mechanisms, and Disease Pathogenesis, First Edition Edited by Frederick A Villamena © 2013 John Wiley & Sons, Inc Published 2013 by John Wiley & Sons, Inc 407 408 INDEX Apolipoprotein A1 (apo A1), 334 B (apo B), 331 Apoptosis, 75, 216–219 Bcl-2, 218 Bcl-xl, 218 BH3-only proteins, 218 cytochrome c (Cyt c), 217–218 death receptors, 217–218 endoplasmic reticulum (ER) stress, 218–219 extrinsic pathway, see Death receptors glutathione, 217–219 in I-R, 313, 315, 319, 320 intrinsic pathway, see Mitochondrial pathway mitochondrial pathway, 218 multidrug-resistance proteins (MRP1), 220 from NADPH oxidases, 145, 146, 148, 155–156, 158 p53, 219 and PTM, 75 TNF-related apoptosis-inducing ligand (TRAIL), 217–218 Apurinic/apyrimidinic (AP) sites, 99, 100–101 Aquaporins, 351 Arachidonic acid (AA), 50–52, 54, 57–65, 240, 242, 246–249, 252 ARE, see Antioxidant response element Arsenic, 94–95 Ascorbic acid, 262, 268, 347 Asparaginyl hydroxylase factor inhibiting HIF (FIH), 191–192 Aspartate, 318 Astrocytes, 239–241, 250, 254, 263, 266–268, 276–279 Asymmetric dimethylarginine (ADMA), 318 Ataluren, 351 Ataxia telangiectasia mutated (ATM), 238–239, 268 Atherosclerosis, 329–333, 384 Attention deficit–hyperactivity disorder (ADHD), 256, 260 Autophagy, 219, 319–320 See also Cell death Azelaic acid (AZA), 334 Bacteria colonization, 346–347 BALF, see Broncho alveolar lavage fluid Benzo[a]pyrene-7,8-dihydrodiol, 250 Beta-amyloid (AB), 241, 250 β-carotene, 347 Beta-scission reactions formation of fragmented lipid products, 51–52, 58–62 role of iron and copper, 50–51, 58, 61–62 Biomarker for COPD, 349 for neurodegenerative disorders, 360 Biotin, 82 Blood–brain barrier (BBB), 263 and METH, 256 Brad 1, 189 Bradykinin, 321 Brain infarction, 385, 387 Breast cancer (Brca1), 238–239, 269 Broncho alveolar lavage fluid, 352 Bronchodilation, 349 Buthionine sulfoximine, 117–118 Calcification, 331 Calcium in cell signaling, 186–188 in dendritic spines, 250 in excitotoxicity, 241 homeostasis, 185, 188, 195 in mitochondria, 238 Calcium-dependent proteases, 241 Calicheamicin, 100 CAMP, see Adenosine monophosphate, cyclic Cancer, 203–235 apoptosis, 216–219 autophagy, 219 catalase, 206 cell signaling, 193–194 cycloxygenases, 205 DNA methylation, 216 DNA methyl-transferases (DMTs), 216 drug resistance, 219–220 epigenetics, 213, 216 glucose-6-phosphate dehydrogenase, 206 glutathione, 205–206, 208, 217 glutathione peroxidases (GPx), 206 glutathione-SNO (S-nitrosoglutathione or GSNO), 208 glutathione-S-transferases, 216, 220 protein glutathionylation, 208 γ -glutamylcysteine synthetase, 205 Carbohydrate oxidation biomarkers in neurodegenerative disorders, 366 Carcinogenesis, 203–235 See also Cancer Cardiac, 311–321 See also Heart ischemia, 311–314, 318 reperfusion, 311, 314, 318 transplantation, 321 Cardiolipin, 315 Cardiomyocyte(s), 311, 314–315, 318–320 Cardioplegic, arrest, 312, 318 buffer, 318 ischemia, 312 solution, 312 Cardiovascular disease (CVD), 330, 334–335 Catalase, 114, 143–146, 159, 238–239, 251–252, 377–378 in brain, 262, 267 in cancer, 206 in I-R, 313 and NADPH, 264 mimetics, 380 in neurodegenerative disease, 278 Celecoxib (Celebrex), 249, 254 Cell death, 311, 315, 319–321 apoptosis, 319–320 autophagy, 319–320 necrosis, 319–320 programmed, 315, 319 Cell signaling, 179, 180, 184–186, 193–194, 317–318 INDEX Cellular enzymes, 313 organelles, 312–313 thiolstat, 71, 84 CF, see cystic fibrosis CFTR, see Cystic fibrosis transmembrane regulator CGMP-dependent protein kinase (PKG)I-α, 85 Chemiluminescence, 32 Chemoprevention, 117 Chemoselective functionalization, 82 Chloride channel, 345, 348 conductance, 351–352 transport, 348 transporter, 348, 351 ChPBNL, 393 See also Nitrones Chromium, 94–95, 98 Chronic granulomatous disease (CGD), 138–141, 145–146, 154, infections, 346 lung infections, 345 obstructive pulmonary disease COPD, 80 Citric acid cycle, 180, 192 Clorgyline, 240 Cockayne syndrome B (CSB), 238–239, 269 Cofactors, 316 BH4, 315, 317–318, 320 Colon epithelial cells, 186 Complex I, 123, 125–129, 133, 312, 314, 316 Complex II, 126, 129, 130, 133 Complex III, 125–126, 129–133, 312, 315 Complex IV, 125–126, 132–133, 315 Conserved sequence block, 96 Copper-zinc superoxide dismutase (CuZnSOD, SOD1), see Superoxide dismutase Coronary artery disease, 311, 320 Coronary heart disease (CHD), 333 COX, see Cyclooxygenase CPI-1429, 393–394 See also Nitrones C-Rel, see Proto-oncogene cRel Cross-links, see Lesions Cu,Zn superoxide dismutase (CuZnSOD, SOD1), 79, 253, 313 in neurodegenerative diseases, 278 Cyanuric acid, 102 Cyclooxygenase-1 (COX-1), 242 See also PHS-1; prostaglandin H synthases Cyclooxygenase-1b (COX-1b) (COX-3), 244 Cyclooygenase, 29 See also COX-1; COX-2 Cyclooxygenase-2 (COX-2), 242 See also PHS-2; Prostaglandin H synthases CYP1A2, 258 See also Cytochromes P450 CYP2D6, 240, 257–259 See also Cytochromes P450 expression in brain, 259 CYP2E1, 240 See also Cytochromes P450 Cysteinyl redox domains, 127 Cystic fibrosis (CF), 80, 345 diagnostic tests for, 348 gene, 345 lung disease, 345–347, 351 Cystic fibrosis transmembrane regulator (CFTR) KO mice, 347–349, 352 mutations, 349, 351 potentiators, 352 Cytochrome bH or heme bH, 130–131 Cytochrome bL or heme bL, 130–133 Cytochrome c, 131–132 in I-R, 314–316, 318–319 See also Nitrate/Nitrite reductase Cytochromes P450 (CYPs), 240 metabolism of amphetamines, 257–259 Cytokine, 187, 193–194 and microglia, 241 and NOS, 240 as PHS activators, 245 Cytosolic phospholipase A2 (cPLA2), 240 Death receptors, 217, 219 Deoxymyoglobin, 316 See also Nitrate/Nitrite reductase 2-deoxypentos-4-ulose abasic site, 96 2′-deoxyribonolactone, 96,100 Deoxyribonucleic acid (DNA), 94–103 and histone proteins, 94 backbone reactivity, 96 damage, 349 See also Oxidative DNA damage duplex, kinetic changes to, 99 holes, 96 oxidation, 252–253 repair, 268–269 2′-deoxyribose (dR), 99–102 and hydrogen abstraction, 100 lesions, 101 oxidation, 100 radical, 100 DEPMPO, 390 See also Nitrones Detection of reactive species, 31–38 in vitro, 32 in vivo, 38 Dexedrine, 256 Diabetes, 194 Dicumarol, 118 Dihydroxyphenylacetic acid (DOPAC), 251–252, 255 Dilated cardiomyopathy, 313 Dimedone, 82 Dimethyl fumarate (DMF), 276–277, 279 Dioxetane, 52, 59, 61 1,2-dioxilane ring, 52 1,2-dioxolanylcarbinyl, 52, 57 Diradical, 190 Dismutation, see Superoxide radical Disulfide, 19, 180 formation, 19 with nucleophiles, 19 oxidation, 19 reaction with thiols, 19 reduction, 20 3,3′-dityrosine, 78 DJ-1, 269–270, 272, 277–278 409 410 INDEX DMPO, 36 as antioxidant, 389–394 DNA, see Deoxyribonucleic acid DNA-mediated charge transport, 96 Dopamine, 154, 156, 240, 251–252, 255, 259 and METH neurotoxicity, 261–262 quinones, 251, 254 Double strand breaks (DSBs), 100 DPI, 126–127 Drug resistance in cancer, 219 glutathione, 219 Dual oxidases, 347 See also DUOX, DUOX1, DUOX2 DUOX, 147–148, 150, 152, 155–159 DUOX1, 147–148, 150, 152, 155, 159 DUOX2, 147–148, 150, 152, 155, 157, 159 Ebselen, 382–384 anti-inflammatory effect of, 382, 384 BXT-51072, 385 BXT-51077, 385 clinical trial (ischemic stroke), 384 ebselen diselenide, 383–385 ebselen selenol, 383–384 ebselen Se-oxide, 384 glutathione peroxydase activity of, 382 inhibiting activity of lipoxygenases, 384 inhibition of iNOS by, 384 inhibition of lipid peroxidation by, 384 peroxynitrite scavenging by, 384 reaction with thiols, 383–384 reduction and scavenging properties of, 383 substrate for mammalian thioredoxin reductase, 384–385 synthesis of, 382 Edaravone (also radicut, MC-186) , 385–388 enhancement of eNOS expression, 385, 387 inhibition of iNOS, 385, 387 neuroprotective effect of, 387 pharmacokinetics of, 385 protection against amyotrophic lateral sclerosis (ALS), 387 protection against brain infraction, 385, 387 protection against noise-induced hearing loss, 388 protection against retinal degeneration, 387 reduction of myocardial injuries, 385–387 scavenging properties of, 385–386 Eicosanoids, 241–242 Elastase, 347 Electron leakage, 28, 312 Electron paramagnetic resonance (EPR) imaging, 38 oximetry, 38 spectroscopy, 34, 126–130, 317, 385–386, 389–390, 397 Electron transfer, Electron transport chain (ETC), see Mitochondrial electron transport chain (METC) Electronegative atoms, Electronegativity, Electrophile response element (EpRE), 269 See also Antioxidant response element Electrospray ionization ( ESI) mass spectrometry, 81 EMPO, 390 See also Nitrones Endonuclease III ( NTH), 102 Endoperoxides, 52–54, 57, 61–62 Endoplasmic reticulum (ER), 29, 312 stress, 313 Endothelial, 311, 315–316, 318–320 cell(s), 311, 316, 318–319 dysfunction, 320 Endothelial nitric oxide synthase (eNOS, NOS3), see Nitric oxide synthase Endothelin, 314, 321 Energy transduction, 125 ENOS/NOS3, see Nitric oxide synthase Epithelial lining fluid, 346, 349 See also ELF Epoxide hydrolase, 313 Epoxy alcohols and peroxyls , 51–53, 57 aldehydes, 61 fatty acids, 59, 62 isoprostanes, 57 Erythorose abasic site, 96 Excision repair cross complementing (ERCC1), 101 Excitotoxicity, 241, 253 source of ROS, 241 4-exo-cyclization, 52 5-exo-cyclization, 52, 55, 57 ΔF508 mutation, 345 FADH•− or FADH• semiquinone, 129 Fat soluble antioxidant vitamins, 347 Fe(IV)=O intermediate, see Iron-oxo intermediate Febuxostat, 320 See also Inhibitors 2Fe-2S, 3Fe-4S, 4Fe-4S, see Iron-sulfur cluster Fe-DETC, see Iron diethyldithiocarbamate Fe-MGD, see Iron methyldithiocarbamate Fenton-type reaction, 125, 183–184, 378, 382 Ferritin, 116, 347 FIH, see Asparaginyl hydroxylase factor inhibiting HIF Flavin adenine dinucleotide (FAD), 129–130, 138–140, 147, 149, 152, 186 Flavin mononucleotide (FMN), 126–128 Flavocytochrome b, 138 See also Flavocytochrome b558, gp91phox, NOX2 Flavocytochrome b558, 139 See also Flavocytochrome b, gp91phox, NOX2 Fluorescence, 32 Fluoroquinolone, 181 FMN, see Flavin mononucleotide FMNH•− or FMNH• semiquinone, 126 Forced vital capacity, 352 3-formyl phosphate, 96 Forskolin, 352 Fp subcomplex, 126–127 Fragmentation ... MOLECULAR BASIS OF OXIDATIVE STRESS Chemistry, Mechanisms, and Disease Pathogenesis Edited by FREDERICK A VILLAMENA Department of Pharmacology and Davis Heart and Lung Institute... Cataloging-in-Publication Data Molecular basis of oxidative stress : chemistry, mechanisms, and disease pathogenesis / edited by Frederick A Villamena, Department of Pharmacology and Davis Heart and Lung Institute,... encompasses the understanding of the mechanisms of oxidative stress and autophagy in experimental Parkinson’s disease models Alexandros G Georgakilas is an Associate Professor of Biology at East

Ngày đăng: 13/03/2018, 15:16

Từ khóa liên quan

Tài liệu cùng người dùng

  • Đang cập nhật ...

Tài liệu liên quan