An evolving understanding of the S-glutathionylation cycle in pathways of redox regulation

Free Radical Biology and Medicine - Tập 120 - Trang 204-216 - 2018
Jie Zhang1, Zhiwei Ye1, Shweta Singh1, Danyelle M. Townsend2, Kenneth D. Tew1
1Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, 70 President Street, DDB410, Charleston, SC 29425, United States
2Department of Pharmaceutical and Biomedical Sciences, Medical University of South Carolina, 274 Calhoun Street, MSC141, Charleston, SC 29425, United States

Tóm tắt

Từ khóa


Tài liệu tham khảo

Grek, 2013, Causes and consequences of cysteine S-glutathionylation, J. Biol. Chem., 288, 26497, 10.1074/jbc.R113.461368

Bechtel, 2017, From structure to redox: the diverse functional roles of disulfides and implications in disease, Proteomics, 17, 10.1002/pmic.201600391

Hatahet, 2009, Protein disulfide isomerase: a critical evaluation of its function in disulfide bond formation, Antioxid. Redox Signal., 11, 2807, 10.1089/ars.2009.2466

Mailloux, 2014, S-glutathionylation reactions in mitochondrial function and disease, Front. Cell Dev. Biol., 2, 68, 10.3389/fcell.2014.00068

Gallogly, 2007, Mechanisms of reversible protein glutathionylation in redox signaling and oxidative stress, Curr. Opin. Pharmacol., 7, 381, 10.1016/j.coph.2007.06.003

Gilbert, 1995, Thiol/disulfide exchange equilibria and disulfide bond stability, Methods Enzymol., 251, 8, 10.1016/0076-6879(95)51107-5

Gilbert, 1990, Molecular and cellular aspects of thiol-disulfide exchange, Adv. Enzymol. Relat. Areas Mol. Biol., 63, 69

Klatt, 1999, Redox regulation of c-Jun DNA binding by reversible S-glutathiolation, FASEB J., 13, 1481, 10.1096/fasebj.13.12.1481

Arbault, 1997, Activation of the NADPH oxidase in human fibroblasts by mechanical intrusion of a single cell with an ultramicroelectrode, Carcinogenesis, 18, 569, 10.1093/carcin/18.3.569

Starke, 2003, Glutathione-thiyl radical scavenging and transferase properties of human glutaredoxin (thioltransferase). Potential role in redox signal transduction, J. Biol. Chem., 278, 14607, 10.1074/jbc.M210434200

Gallogly, 2008, Kinetic and mechanistic characterization and versatile catalytic properties of mammalian glutaredoxin 2: implications for intracellular roles, Biochemistry, 47, 11144, 10.1021/bi800966v

Giustarini, 2005, S-nitrosation versus S-glutathionylation of protein sulfhydryl groups by S-nitrosoglutathione, Antioxid. Redox Signal., 7, 930, 10.1089/ars.2005.7.930

Townsend, 2006, A glutathione S-transferase pi-activated prodrug causes kinase activation concurrent with S-glutathionylation of proteins, Mol. Pharmacol., 69, 501, 10.1124/mol.105.018523

Townsend, 2009, Nitrosative stress-induced s-glutathionylation of protein disulfide isomerase leads to activation of the unfolded protein response, Cancer Res., 69, 7626, 10.1158/0008-5472.CAN-09-0493

Konorev, 2000, Modification of creatine kinase by S-nitrosothiols: s-nitrosation vs. S-thiolation, Free Radic. Biol. Med., 28, 1671, 10.1016/S0891-5849(00)00281-1

Manevich, 2004, Activation of the antioxidant enzyme 1-CYS peroxiredoxin requires glutathionylation mediated by heterodimerization with pi GST, Proc. Natl. Acad. Sci. USA, 101, 3780, 10.1073/pnas.0400181101

Ralat, 2006, Direct evidence for the formation of a complex between 1-cysteine peroxiredoxin and glutathione S-transferase pi with activity changes in both enzymes, Biochemistry, 45, 360, 10.1021/bi0520737

Townsend, 2009, Novel role for glutathione S-transferase pi. Regulator of protein S-Glutathionylation following oxidative and nitrosative stress, J. Biol. Chem., 284, 436, 10.1074/jbc.M805586200

Wetzelberger, 2010, Postischemic deactivation of cardiac aldose reductase: role of glutathione S-transferase P and glutaredoxin in regeneration of reduced thiols from sulfenic acids, J. Biol. Chem., 285, 26135, 10.1074/jbc.M110.146423

de Luca, 2011, Treatment of doxorubicin-resistant MCF7/Dx cells with nitric oxide causes histone glutathionylation and reversal of drug resistance, Biochem. J., 440, 175, 10.1042/BJ20111333

Klaus, 2013, Glutathione S-transferases interact with AMP-activated protein kinase: evidence for S-glutathionylation and activation in vitro, PLoS One, 8, e62497, 10.1371/journal.pone.0062497

Ye, 2017, Glutathione S-transferase P-mediated protein S-glutathionylation of resident endoplasmic reticulum proteins influences sensitivity to drug-induced unfolded protein response, Antioxid. Redox Signal., 26, 247, 10.1089/ars.2015.6486

Zhang, 2014, Glutathione S-transferase P influences redox and migration pathways in bone marrow, PLoS One, 9, e107478, 10.1371/journal.pone.0107478

Kamada, 2004, Nuclear glutathione S-transferase pi prevents apoptosis by reducing the oxidative stress-induced formation of exocyclic DNA products, Free Radic. Biol. Med., 37, 1875, 10.1016/j.freeradbiomed.2004.09.002

Goto, 2009, Glutathione S-transferase pi localizes in mitochondria and protects against oxidative stress, Free Radic. Biol. Med., 46, 1392, 10.1016/j.freeradbiomed.2009.02.025

Tajc, 2004, Direct determination of thiol pKa by isothermal titration microcalorimetry, J. Am. Chem. Soc., 126, 10508, 10.1021/ja047929u

Dirr, 1994, X-ray crystal structures of cytosolic glutathione S-transferases. Implications for protein architecture, substrate recognition and catalytic function, Eur. J. Biochem., 220, 645, 10.1111/j.1432-1033.1994.tb18666.x

Tew, 2011, The role of glutathione S-transferase P in signaling pathways and S-glutathionylation in cancer, Free Radic. Biol. Med., 51, 299, 10.1016/j.freeradbiomed.2011.04.013

Xiong, 2011, S-glutathionylation: from molecular mechanisms to health outcomes, Antioxid. Redox Signal., 15, 233, 10.1089/ars.2010.3540

Mannervik, 1980, Role of cytoplasmic thioltransferase in cellular regulation by thiol-disulphide interchange, Biochem. J., 190, 125, 10.1042/bj1900125

Gladyshev, 2001, Identification and characterization of a new mammalian glutaredoxin (thioltransferase), Grx2, J. Biol. Chem., 276, 30374, 10.1074/jbc.M100020200

Lundberg, 2001, Cloning and expression of a novel human glutaredoxin (Grx2) with mitochondrial and nuclear isoforms, J. Biol. Chem., 276, 26269, 10.1074/jbc.M011605200

Pai, 2007, What is the functional significance of the unique location of glutaredoxin 1 (GRx1) in the intermembrane space of mitochondria?, Antioxid. Redox Signal., 9, 2027, 10.1089/ars.2007.1642

Gallogly, 2009, Mechanistic and kinetic details of catalysis of thiol-disulfide exchange by glutaredoxins and potential mechanisms of regulation, Antioxid. Redox Signal., 11, 1059, 10.1089/ars.2008.2291

Martin, 1995, Thioredoxin–a fold for all reasons, Structure, 3, 245, 10.1016/S0969-2126(01)00154-X

Gravina, 1993, Thioltransferase is a specific glutathionyl mixed disulfide oxidoreductase, Biochemistry, 32, 3368, 10.1021/bi00064a021

Peltoniemi, 2006, Insights into deglutathionylation reactions. Different intermediates in the glutaredoxin and protein disulfide isomerase catalyzed reactions are defined by the gamma-linkage present in glutathione, J. Biol. Chem., 281, 33107, 10.1074/jbc.M605602200

Jensen, 2014, The pKa value and accessibility of cysteine residues are key determinants for protein substrate discrimination by glutaredoxin, Biochemistry, 53, 2533, 10.1021/bi4016633

Menon, 2013, A role for glutathione transferase Omega 1 (GSTO1-1) in the glutathionylation cycle, J. Biol. Chem., 288, 25769, 10.1074/jbc.M113.487785

Findlay, 2006, A novel role for human sulfiredoxin in the reversal of glutathionylation, Cancer Res., 66, 6800, 10.1158/0008-5472.CAN-06-0484

Park, 2009, Deglutathionylation of 2-Cys peroxiredoxin is specifically catalyzed by sulfiredoxin, J. Biol. Chem., 284, 23364, 10.1074/jbc.M109.021394

Board, 2000, Identification, characterization, and crystal structure of the Omega class glutathione transferases, J. Biol. Chem., 275, 24798, 10.1074/jbc.M001706200

Board, 2016, Structure, function and disease relevance of Omega-class glutathione transferases, Arch. Toxicol., 90, 1049, 10.1007/s00204-016-1691-1

Kim, 2012, Glutathione s-transferase omega 1 activity is sufficient to suppress neurodegeneration in a Drosophila model of Parkinson disease, J. Biol. Chem., 287, 6628, 10.1074/jbc.M111.291179

Chen, 2017, Glycosylation of KEAP1 links nutrient sensing to redox stress signaling, EMBO J., 36, 2233, 10.15252/embj.201696113

Uys, 2011, Nitrosative stress-induced S-glutathionylation of protein disulfide isomerase, Methods Enzymol., 490, 321, 10.1016/B978-0-12-385114-7.00018-0

Ghezzi, 2005, Regulation of protein function by glutathionylation, Free Radic. Res., 39, 573, 10.1080/10715760500072172

Watanabe, 2014, Methods of measuring protein disulfide isomerase activity: a critical overview, Front. Chem., 2, 73, 10.3389/fchem.2014.00073

Xiong, 2012, S-glutathionylation of protein disulfide isomerase regulates estrogen receptor alpha stability and function, Int. J. Cell Biol., 2012, 273549, 10.1155/2012/273549

Hofmann, 2002, Peroxiredoxins, Biol. Chem., 383, 347

Ralat, 2008, Characterization of the complex of glutathione S-transferase pi and 1-cysteine peroxiredoxin, Arch. Biochem. Biophys., 474, 109, 10.1016/j.abb.2008.02.043

Mieyal, 1991, Thioltransferase in human red blood cells: purification and properties, Biochemistry, 30, 6088, 10.1021/bi00239a002

Manevich, 2013, Allelic variants of glutathione S-transferase P1-1 differentially mediate the peroxidase function of peroxiredoxin VI and alter membrane lipid peroxidation, Free Radic. Biol. Med., 54, 62, 10.1016/j.freeradbiomed.2012.10.556

Choi, 1998, Crystal structure of a novel human peroxidase enzyme at 2.0 A resolution, Nat. Struct. Biol., 5, 400, 10.1038/nsb0598-400

Manevich, 2014, Peroxiredoxin VI oxidation in cerebrospinal fluid correlates with traumatic brain injury outcome, Free Radic. Biol. Med., 72, 210, 10.1016/j.freeradbiomed.2014.04.002

Zhang, 2014, Pleiotropic functions of glutathione S-transferase P, Adv. Cancer Res., 122, 143, 10.1016/B978-0-12-420117-0.00004-9

Murphy, 2012, Mitochondrial thiols in antioxidant protection and redox signaling: distinct roles for glutathionylation and other thiol modifications, Antioxid. Redox Signal., 16, 476, 10.1089/ars.2011.4289

O'Brien, 2017, Protein S-glutathionylation alters superoxide/hydrogen peroxide emission from pyruvate dehydrogenase complex, Free Radic. Biol. Med., 106, 302, 10.1016/j.freeradbiomed.2017.02.046

Kil, 2005, Regulation of mitochondrial NADP+-dependent isocitrate dehydrogenase activity by glutathionylation, J. Biol. Chem., 280, 10846, 10.1074/jbc.M411306200

Applegate, 2008, Reversible inhibition of alpha-ketoglutarate dehydrogenase by hydrogen peroxide: glutathionylation and protection of lipoic acid, Biochemistry, 47, 473, 10.1021/bi7017464

Eaton, 2002, Purification of proteins susceptible to oxidation at cysteine residues: identification of malate dehydrogenase as a target for S-glutathiolation, Ann. N. Y. Acad. Sci., 973, 529, 10.1111/j.1749-6632.2002.tb04694.x

Garcia, 2010, Regulation of mitochondrial glutathione redox status and protein glutathionylation by respiratory substrates, J. Biol. Chem., 285, 39646, 10.1074/jbc.M110.164160

Han, 2005, Sites and mechanisms of aconitase inactivation by peroxynitrite: modulation by citrate and glutathione, Biochemistry, 44, 11986, 10.1021/bi0509393

Baradaran, 2013, Crystal structure of the entire respiratory complex I, Nature, 494, 443, 10.1038/nature11871

Passarelli, 2010, GSSG-mediated Complex I defect in isolated cardiac mitochondria, Int. J. Mol. Med., 26, 95

Hurd, 2008, Complex I within oxidatively stressed bovine heart mitochondria is glutathionylated on Cys-531 and Cys-704 of the 75-kDa subunit: potential role of CYS residues in decreasing oxidative damage, J. Biol. Chem., 283, 24801, 10.1074/jbc.M803432200

Kumar, 2013, Redox proteomics of thiol proteins in mouse heart during ischemia/reperfusion using ICAT reagents and mass spectrometry, Free Radic. Biol. Med., 58, 109, 10.1016/j.freeradbiomed.2013.01.021

Mailloux, 2014, Glutaredoxin-2 is required to control oxidative phosphorylation in cardiac muscle by mediating deglutathionylation reactions, J. Biol. Chem., 289, 14812, 10.1074/jbc.M114.550574

Grivennikova, 2001, Catalytic activity of NADH-ubiquinone oxidoreductase (complex I) in intact mitochondria. evidence for the slow active/inactive transition, J. Biol. Chem., 276, 9038, 10.1074/jbc.M009661200

Drose, 2014, Mitochondrial respiratory chain complexes as sources and targets of thiol-based redox-regulation, Biochim. Biophys. Acta, 1844, 1344, 10.1016/j.bbapap.2014.02.006

Galkin, 2008, Identification of the mitochondrial ND3 subunit as a structural component involved in the active/deactive enzyme transition of respiratory complex I, J. Biol. Chem., 283, 20907, 10.1074/jbc.M803190200

Chen, 2007, Mitochondrial complex II in the post-ischemic heart: oxidative injury and the role of protein S-glutathionylation, J. Biol. Chem., 282, 32640, 10.1074/jbc.M702294200

Wang, 2011, Redox regulation of mitochondrial ATP synthase: implications for cardiac resynchronization therapy, Circ. Res., 109, 750, 10.1161/CIRCRESAHA.111.246124

Mailloux, 2011, Uncoupling proteins and the control of mitochondrial reactive oxygen species production, Free Radic. Biol. Med., 51, 1106, 10.1016/j.freeradbiomed.2011.06.022

Mailloux, 2011, Glutathionylation acts as a control switch for uncoupling proteins UCP2 and UCP3, J. Biol. Chem., 286, 21865, 10.1074/jbc.M111.240242

Queiroga, 2010, Glutathionylation of adenine nucleotide translocase induced by carbon monoxide prevents mitochondrial membrane permeabilization and apoptosis, J. Biol. Chem., 285, 17077, 10.1074/jbc.M109.065052

Kramer, 2015, The measurement of reversible redox dependent post-translational modifications and their regulation of mitochondrial and skeletal muscle function, Front. Physiol., 6, 347, 10.3389/fphys.2015.00347

Zhang, 2018, S-Glutathionylation of estrogen receptor alpha affects dendritic cell function, J. Biol. Chem.

Hwang, 1992, Oxidized redox state of glutathione in the endoplasmic reticulum, Science, 257, 1496, 10.1126/science.1523409

Coe, 2009, Calcium binding chaperones of the endoplasmic reticulum, Gen. Physiol. Biophys. 28 Spec. No Focus, F96

Carreras-Sureda, 2017, Calcium signaling at the endoplasmic reticulum: fine-tuning stress responses, Cell Calcium

Aracena-Parks, 2006, Identification of cysteines involved in S-nitrosylation, S-glutathionylation, and oxidation to disulfides in ryanodine receptor type 1, J. Biol. Chem., 281, 40354, 10.1074/jbc.M600876200

Aracena, 2005, Effects of S-glutathionylation and S-nitrosylation on calmodulin binding to triads and FKBP12 binding to type 1 calcium release channels, Antioxid. Redox Signal., 7, 870, 10.1089/ars.2005.7.870

Zima, 2016, Functional impact of ryanodine receptor oxidation on intracellular calcium regulation in the heart, Rev. Physiol. Biochem. Pharmacol., 171, 39, 10.1007/112_2016_2

Sanchez, 2005, Tachycardia increases NADPH oxidase activity and RyR2 S-glutathionylation in ventricular muscle, J. Mol. Cell Cardiol., 39, 982, 10.1016/j.yjmcc.2005.08.010

Lock, 2012, Protein S-glutathionylation enhances Ca2+-induced Ca2+ release via the IP3 receptor in cultured aortic endothelial cells, J. Physiol., 590, 3431, 10.1113/jphysiol.2012.230656

Viner, 1996, The oxidative inactivation of sarcoplasmic reticulum Ca(2+)-ATPase by peroxynitrite, Free Radic. Res., 24, 243, 10.3109/10715769609088022

Adachi, 2004, S-Glutathiolation by peroxynitrite activates SERCA during arterial relaxation by nitric oxide, Nat. Med., 10, 1200, 10.1038/nm1119

Vangheluwe, 2005, Modulating sarco(endo)plasmic reticulum Ca2+ ATPase 2 (SERCA2) activity: cell biological implications, Cell Calcium, 38, 291, 10.1016/j.ceca.2005.06.033

Wang, 2016, Formation and reversibility of BiP Protein cysteine oxidation facilitate cell survival during and post oxidative stress, J. Biol. Chem., 291, 7541, 10.1074/jbc.M115.694810

Ali-Osman, 1997, Molecular cloning, characterization, and expression in Escherichia coli of full-length cDNAs of three human glutathione S-transferase Pi gene variants. Evidence for differential catalytic activity of the encoded proteins, J. Biol. Chem., 272, 10004, 10.1074/jbc.272.15.10004

Harries, 1997, Identification of genetic polymorphisms at the glutathione S-transferase Pi locus and association with susceptibility to bladder, testicular and prostate cancer, Carcinogenesis, 18, 641, 10.1093/carcin/18.4.641

Harris, 1998, Polymorphism of the Pi class glutathione S-transferase in normal populations and cancer patients, Pharmacogenetics, 8, 27, 10.1097/00008571-199802000-00004

Grek, 2014, S-glutathionylation of buccal cell proteins as biomarkers of exposure to hydrogen peroxide, BBA Clin., 2, 31, 10.1016/j.bbacli.2014.08.003

Karihtala, 2003, Peroxiredoxins in breast carcinoma, Clin. Cancer Res., 9, 3418

Chang, 2007, Identification of the functional role of peroxiredoxin 6 in the progression of breast cancer, Breast Cancer Res., 9, R76, 10.1186/bcr1789

Manevich, 2004, Activation of the antioxidant enzyme 1-CYS peroxiredoxin requires glutathionylation mediated by heterodimerization with pi GST, Proc. Natl. Acad. Sci. USA, 101, 3780, 10.1073/pnas.0400181101

Manevich, 2005, Peroxiredoxin 6, a 1-Cys peroxiredoxin, functions in antioxidant defense and lung phospholipid metabolism, Free Radic. Biol. Med., 38, 1422, 10.1016/j.freeradbiomed.2005.02.011

Townsend, 2007, S-glutathionylation: indicator of cell stress and regulator of the unfolded protein response, Mol. Interv., 7, 313, 10.1124/mi.7.6.7

Morgan, 1996, Isozyme-specific glutathione S-transferase inhibitors potentiate drug sensitivity in cultured human tumor cell lines, Cancer Chemother. Pharmacol., 37, 363, 10.1007/s002800050398

Raza, 2012, A phase 2 randomized multicenter study of 2 extended dosing schedules of oral ezatiostat in low to intermediate-1 risk myelodysplastic syndrome, Cancer, 118, 2138, 10.1002/cncr.26469

Mahadevan, 2015, Ezatiostat hydrochloride for the treatment of myelodysplastic syndromes, Expert Opin. Investig. Drugs, 24, 725, 10.1517/13543784.2015.1021003

Quddus, 2010, Oral Ezatiostat HCl (TLK199) and Myelodysplastic syndrome: a case report of sustained hematologic response following an abbreviated exposure, J. Hematol. Oncol., 3, 16, 10.1186/1756-8722-3-16

Lyons, 2011, Oral ezatiostat HCl (Telintra(R), TLK199) and idiopathic chronic neutropenia (ICN): a case report of complete response of a patient with G-CSF resistant ICN following treatment with ezatiostat, a glutathione S-transferase P1-1 (GSTP1-1) inhibitor, J. Hematol. Oncol., 4, 43, 10.1186/1756-8722-4-43

Flatgaard, 1993, Isozyme specificity of novel glutathione-S-transferase inhibitors, Cancer Chemother. Pharmacol., 33, 63, 10.1007/BF00686025

Lyttle, 1994, Isozyme-specific glutathione-S-transferase inhibitors: design and synthesis, J. Med. Chem., 37, 189, 10.1021/jm00027a024

Gate, 2004, Increased myeloproliferation in glutathione S-transferase pi-deficient mice is associated with a deregulation of JNK and Janus kinase/STAT pathways, J. Biol. Chem., 279, 8608, 10.1074/jbc.M308613200

McMillan, 2016, Attenuation of lung fibrosis in mice with a clinically relevant inhibitor of glutathione-S-transferase pi, JCI Insight, 1, 10.1172/jci.insight.85717

Zhang, 2017, Hydrogen polysulfide biosignal-responsive polymersomes as a nanoplatform for distinguishing intracellular reactive sulfur species (RSS), Small

Cortese-Krott, 2017, The reactive species interactome: evolutionary emergence, biological significance, and opportunities for redox metabolomics and personalized medicine, Antioxid. Redox Signal., 27, 684, 10.1089/ars.2017.7083

Paulsen, 2013, Cysteine-mediated redox signaling: chemistry, biology, and tools for discovery, Chem. Rev., 113, 4633, 10.1021/cr300163e

Casas, 2015, Reactive oxygen-related diseases: therapeutic targets and emerging clinical indications, Antioxid. Redox Signal., 23, 1171, 10.1089/ars.2015.6433

Watson, 2014, Type 2 diabetes as a redox disease, Lancet, 383, 841, 10.1016/S0140-6736(13)62365-X

Sparaco, 2009, Friedreich's ataxia: oxidative stress and cytoskeletal abnormalities, J. Neurol. Sci., 287, 111, 10.1016/j.jns.2009.08.052

Mileo, 2009, Human papillomavirus-16 E7 interacts with glutathione S-transferase P1 and enhances its role in cell survival, PLoS One, 4, e7254, 10.1371/journal.pone.0007254

Trujillo, 2014, The cellular redox environment alters antigen presentation, J. Biol. Chem., 289, 27979, 10.1074/jbc.M114.573402

Butturini, 2018, S-glutathionylation exerts opposing roles in the regulation of STAT1 and STAT3 signaling in reactive microglia, Free Radic. Biol. Med., 117, 191, 10.1016/j.freeradbiomed.2018.02.005

Hoffman, 2016, Ablation of the thiol transferase Glutaredoxin-1 augments protein S-glutathionylation and modulates type 2 inflammatory responses and IL-17 in a house dust mite model of allergic airway disease in mice, Ann. Am. Thorac. Soc., 13, S97, 10.1513/AnnalsATS.201510-656MG

Kovats, 2012, Estrogen receptors regulate an inflammatory pathway of dendritic cell differentiation: mechanisms and implications for immunity, Horm. Behav., 62, 254, 10.1016/j.yhbeh.2012.04.011

Michalek, 2011, Estrogen-related receptor-alpha is a metabolic regulator of effector T-cell activation and differentiation, Proc. Natl. Acad. Sci. USA, 108, 18348, 10.1073/pnas.1108856108

Mitchell, 2017, Glutathionylation of Yersinia pestis LcrV and its effects on plague pathogenesis, MBio, 8, 10.1128/mBio.00646-17

Gu, 2016, Proteomic approaches to quantify cysteine reversible modifications in aging and neurodegenerative diseases, Proteom. Clin. Appl., 10, 1159, 10.1002/prca.201600015

Yang, 2016, The expanding landscape of the thiol redox proteome, Mol. Cell. Proteom., 15, 1, 10.1074/mcp.O115.056051

Su, 2014, Proteomic identification and quantification of S-glutathionylation in mouse macrophages using resin-assisted enrichment and isobaric labeling, Free Radic. Biol. Med., 67, 460, 10.1016/j.freeradbiomed.2013.12.004

Guo, 2014, Resin-assisted enrichment of thiols as a general strategy for proteomic profiling of cysteine-based reversible modifications, Nat. Protoc., 9, 64, 10.1038/nprot.2013.161

Lind, 2002, Identification of S-glutathionylated cellular proteins during oxidative stress and constitutive metabolism by affinity purification and proteomic analysis, Arch. Biochem. Biophys., 406, 229, 10.1016/S0003-9861(02)00468-X

Reynaert, 2006, In situ detection of S-glutathionylated proteins following glutaredoxin-1 catalyzed cysteine derivatization, Biochim. Biophys. Acta, 1760, 380, 10.1016/j.bbagen.2006.01.006

Abo, 2015, Electrophilic probe for global analysis of cysteine reactivity in living cells, J. Am. Chem. Soc., 137, 7087, 10.1021/jacs.5b04350

Hurd, 2007, Detection of reactive oxygen species-sensitive thiol proteins by redox difference gel electrophoresis: implications for mitochondrial redox signaling, J. Biol. Chem., 282, 22040, 10.1074/jbc.M703591200

Comini, 2016, Measurement and meaning of cellular thiol: disufhide redox status, Free Radic. Res., 50, 246, 10.3109/10715762.2015.1110241

Pan, 2014, Mass spectrometry-based quantitative proteomics for dissecting multiplexed redox cysteine modifications in nitric oxide-protected cardiomyocyte under hypoxia, Antioxid. Redox Signal., 20, 1365, 10.1089/ars.2013.5326

Forrester, 2009, Proteomic analysis of S-nitrosylation and denitrosylation by resin-assisted capture, Nat. Biotechnol., 27, 557, 10.1038/nbt.1545

Michelet, 2008, In vivo targets of S-thiolation in Chlamydomonas reinhardtii, J. Biol. Chem., 283, 21571, 10.1074/jbc.M802331200

Brennan, 2006, The utility of N,N-biotinyl glutathione disulfide in the study of protein S-glutathiolation, Mol. Cell. Proteom., 5, 215, 10.1074/mcp.M500212-MCP200

Yang, 2010, Oxidative stress inhibits vascular K(ATP) channels by S-glutathionylation, J. Biol. Chem., 285, 38641, 10.1074/jbc.M110.162578

Samarasinghe, 2014, Metabolic synthesis of clickable glutathione for chemoselective detection of glutathionylation, J. Am. Chem. Soc., 136, 11566, 10.1021/ja503946q

Samarasinghe, 2016, A clickable glutathione approach for identification of protein glutathionylation in response to glucose metabolism, Mol. Biosyst., 12, 2471, 10.1039/C6MB00175K