J. Ådén, M. Wallgren, P. Storm, C. F. Weise, A. Christiansen et al., Extraordinary µs-ms backbone dynamics in Arabidopsis thaliana peroxiredoxin Q, Biochim. Biophys. Acta BBA -Proteins Proteomics, vol.1814, pp.1880-1890, 2011.

N. J. Adimora, D. P. Jones, and M. L. Kemp, A Model of Redox Kinetics Implicates the Thiol Proteome in Cellular Hydrogen Peroxide Responses, Antioxid. Redox Signal, vol.13, pp.731-743, 2010.

S. Akter, L. Fu, Y. Jung, M. L. Conte, J. R. Lawson et al., Chemical proteomics reveals new targets of cysteine sulfinic acid reductase, Nat. Chem. Biol, vol.14, pp.995-1004, 2018.

C. Appenzeller-herzog, G. Bánhegyi, I. Bogeski, K. J. Davies, A. Delaunay-moisan et al., Transit of H2O2 across the endoplasmic reticulum membrane is not sluggish. Free Radic, Biol. Med, vol.94, pp.157-160, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02393406

W. Augustin, F. Gellerich, I. Wiswedel, Y. Evtodienko, and V. Zinchenko, Inhibition of cation efflux by antioxidants during oscillatory ion transport in mitochondria, FEBS Lett, vol.107, pp.151-154, 1979.

E. V. Bakhmutova-albert, H. Yao, D. E. Denevan, and D. E. Richardson, Kinetics and Mechanism of Peroxymonocarbonate Formation, Inorg. Chem, vol.49, pp.11287-11296, 2010.

M. Bakhtina, S. Lee, Y. Wang, C. Dunlap, B. Lamarche et al., Use of Viscogens, dNTP?S, and Rhodium(III) as Probes in Stopped-Flow Experiments To Obtain New Evidence for the Mechanism of Catalysis by DNA Polymerase ? ?, ? . Biochemistry, vol.44, pp.5177-5187, 2005.

J. S. Beckman and B. A. Freeman, Apparent Hydroxyl Radical Production by Peroxinitrite: Implications for Endothelial Injury from Nitric Oxide and Superoxide, Med. Sci, vol.5, 1990.

B. S. Berlett and E. R. Stadtman, Protein Oxidation in Aging, Disease, and Oxidative Stress, J. Biol. Chem, vol.272, pp.20313-20316, 1997.

A. Bersweiler, B. D'autréaux, H. Mazon, A. Kriznik, G. Belli et al., A scaffold protein that chaperones a cysteine-sulfenic acid in H2O2 signaling, Nat. Chem. Biol, vol.13, pp.909-915, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01652643

G. P. Bienert and F. Chaumont, Aquaporin-facilitated transmembrane diffusion of hydrogen peroxide, Biochim. Biophys. Acta BBA -Gen. Subj, vol.1840, pp.1596-1604, 2014.

B. Biteau, J. Labarre, and M. B. Toledano, ATP-dependent reduction of cysteine-sulphinic acid by S. cerevisiae sulphiredoxin, Nature, vol.425, pp.980-984, 2003.

J. A. Bolduc, K. J. Nelson, A. C. Haynes, J. Lee, J. A. Reisz et al., Novel hyperoxidation resistance motifs in 2-Cys peroxiredoxins, J. Biol. Chem, vol.293, pp.11901-11912, 2018.

J. A. Bolduc, K. J. Nelson, A. C. Haynes, J. Lee, J. A. Reisz et al., Novel hyperoxidation resistance motifs in 2-Cys peroxiredoxins, J. Biol. Chem, vol.293, pp.11901-11912, 2018.

S. Boukhenouna, H. Mazon, G. Branlant, C. Jacob, M. B. Toledano et al., Evidence That Glutathione and the Glutathione System Efficiently Recycle 1-Cys Sulfiredoxin In Vivo, Antioxid. Redox Signal, vol.22, pp.731-743, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01451669

G. Calabrese, E. Peker, P. S. Amponsah, M. N. Hoehne, T. Riemer et al., Hyperoxidation of mitochondrial peroxiredoxin limits H 2 O 2 -induced cell death in yeast, EMBO J, vol.38, 2019.

I. A. Calvo, S. Boronat, A. Domènech, S. García-santamarina, J. Ayté et al., , 2013.

, Dissection of a Redox Relay: H2O2-Dependent Activation of the Transcription Factor Pap1 through the Peroxidatic Tpx1-Thioredoxin Cycle, Cell Rep, vol.5, pp.1413-1424

Z. Cao and J. G. Lindsay, The Peroxiredoxin Family: An Unfolding Story, Macromolecular Protein Complexes, pp.127-147, 2017.

Z. Cao, S. Subramaniam, and N. J. Bulleid, Lack of an Efficient Endoplasmic Reticulumlocalized Recycling System Protects Peroxiredoxin IV from Hyperoxidation, J. Biol. Chem, vol.289, pp.5490-5498, 2014.

M. D. De-carvalho, J. F. De-mesquita, and E. C. Eleutherio, In Vivo Characterization of I91T Sod2 Polymorphism of Saccharomyces cerevisiae, J. Cell. Biochem, vol.118, pp.1078-1086, 2017.

H. Z. Chae, K. Robison, L. B. Poole, G. Church, G. Storz et al., Cloning and sequencing of thiol-specific antioxidant from mammalian brain: alkyl hydroperoxide reductase and thiol-specific antioxidant define a large family of antioxidant enzymes, Proc. Natl. Acad. Sci, vol.91, pp.7017-7021, 1994.

S. D. Copley, W. R. Novak, and P. C. Babbitt, Divergence of Function in the Thioredoxin Fold Suprafamily: Evidence for Evolution of Peroxiredoxins from a Thioredoxin-like Ancestor ?, Biochemistry, vol.43, pp.13981-13995, 2004.

A. G. Cox, K. K. Brown, E. S. Arner, and M. B. Hampton, The thioredoxin reductase inhibitor auranofin triggers apoptosis through a Bax/Bak-dependent process that involves peroxiredoxin 3 oxidation, Biochem. Pharmacol, vol.76, pp.1097-1109, 2008.

J. Dahl, M. J. Gray, J. , and U. , Protein Quality Control under Oxidative Stress Conditions, J. Mol. Biol, vol.427, pp.1549-1563, 2015.

J. Dalla-rizza, L. M. Randall, J. Santos, G. Ferrer-sueta, and A. Denicola, Differential parameters between cytosolic 2-Cys peroxiredoxins, PRDX1 and PRDX2: PRDX1, PRDX2 differential parameters, Protein Sci, 2018.

J. Dalla-rizza, L. M. Randall, J. Santos, G. Ferrer-sueta, and A. Denicola, Differential parameters between cytosolic 2-Cys peroxiredoxins, PRDX1 and PRDX2: PRDX1, PRDX2 differential parameters, Protein Sci, vol.28, pp.191-201, 2019.

K. D'ambrosio, D. Limauro, E. Pedone, I. Galdi, C. Pedone et al., Insights into the catalytic mechanism of the Bcp family: Functional and structural analysis of Bcp1 from Sulfolobus solfataricus, Proteins Struct. Funct. Bioinforma, vol.76, pp.995-1006, 2009.

E. Damiani, G. Paganga, L. Greci, and C. Rice-evans, Inhibition of copper-mediated low density lipoprotein peroxidation by quinoline and indolinone nitroxide radicals, Biochem. Pharmacol, vol.48, pp.1155-1161, 1994.

A. M. Day, J. D. Brown, S. R. Taylor, J. D. Rand, B. A. Morgan et al., Inactivation of a Peroxiredoxin by Hydrogen Peroxide Is Critical for Thioredoxin-Mediated Repair of Oxidized Proteins and Cell Survival, Mol. Cell, vol.45, pp.398-408, 2012.

A. Delaunay, D. Pflieger, M. B. Barrault, J. Vinh, and M. B. Toledano, A thiol peroxidase is an H2O2 receptor and redox-transducer in gene activation, Cell, vol.111, pp.471-481, 2002.
URL : https://hal.archives-ouvertes.fr/hal-01637040

A. Delaunay, D. Pflieger, M. Barrault, J. Vinh, and M. B. Toledano, A Thiol Peroxidase Is an H2O2 Receptor and Redox-Transducer in Gene Activation, Cell, vol.111, pp.471-481, 2002.
URL : https://hal.archives-ouvertes.fr/hal-01637040

A. Denicola, J. M. Souza, and R. Radi, Diffusion of peroxynitrite across erythrocyte membranes, Proc. Natl. Acad. Sci, vol.95, pp.3566-3571, 1998.

A. Domènech, J. Ayté, F. Antunes, and E. Hidalgo, Using in vivo oxidation status of oneand two-component redox relays to determine H2O2 levels linked to signaling and toxicity, BMC Biol, vol.16, p.61, 2018.

H. R. Ellis and L. B. Poole, Roles for the Two Cysteine Residues of AhpC in Catalysis of Peroxide Reduction by Alkyl Hydroperoxide Reductase from Salmonella typhimurium ?, Biochemistry, vol.36, pp.13349-13356, 1997.

F. Etienne, D. Spector, N. Brot, and H. Weissbach, A methionine sulfoxide reductase in Escherichia coli that reduces the R enantiomer of methionine sulfoxide, Biochem. Biophys. Res. Commun, vol.300, pp.378-382, 2003.

G. Ferrer-sueta, B. Manta, H. Botti, R. Radi, M. Trujillo et al., Factors Affecting Protein Thiol Reactivity and Specificity in Peroxide Reduction, Chem. Res. Toxicol, vol.24, pp.434-450, 2011.
URL : https://hal.archives-ouvertes.fr/pasteur-00685049

L. K. Folkes, L. P. Candeias, and P. Wardman, Kinetics and Mechanisms of Hypochlorous Acid Reactions, Arch. Biochem. Biophys, vol.323, pp.120-126, 1995.

H. J. Forman and I. Fridovich, Superoxide dismutase: A comparison of rate constants, Arch. Biochem. Biophys, vol.158, pp.396-400, 1973.

S. Fourquet, M. Huang, B. D'autreaux, and M. B. Toledano, The Dual Functions of Thiol-Based Peroxidases in H 2 O 2 Scavenging and Signaling, Antioxid. Redox Signal, vol.10, pp.1565-1576, 2008.

M. M. Gaschler and B. R. Stockwell, Lipid peroxidation in cell death, Biochem. Biophys. Res. Commun, vol.482, pp.419-425, 2017.

M. Giorgio, M. Trinei, E. Migliaccio, and P. G. Pelicci, Hydrogen peroxide: a metabolic by-product or a common mediator of ageing signals? 7, 2007.

A. Gruez and G. Branlant, Structural diversity in the recognition between reduced thioredoxin and its oxidized enzyme partners, Biomol. Concepts, vol.3, 2012.

A. Gruez, M. Libiad, S. Boschi-muller, and G. Branlant, Structural and Biochemical Characterization of Free Methionine-R -sulfoxide Reductase from Neisseria meningitidis, J. Biol. Chem, vol.285, pp.25033-25043, 2010.
URL : https://hal.archives-ouvertes.fr/hal-01690430

A. Hall, P. A. Karplus, and L. B. Poole, Typical 2-Cys peroxiredoxins--structures, mechanisms and functions, Febs J, vol.276, pp.2469-2477, 2009.

A. Hall, D. Parsonage, L. B. Poole, and P. A. Karplus, Structural Evidence that Peroxiredoxin Catalytic Power Is Based on Transition-State Stabilization, J. Mol. Biol, vol.402, pp.194-209, 2010.

A. Hall, K. Nelson, L. B. Poole, and P. A. Karplus, Structure-based insights into the catalytic power and conformational dexterity of peroxiredoxins, Antioxid. Redox Signal, vol.15, pp.795-815, 2011.

S. Hanzén, K. Vielfort, J. Yang, F. Roger, V. Andersson et al., Lifespan Control by Redox-Dependent Recruitment of Chaperones to Misfolded Proteins, Cell, vol.166, pp.140-151, 2016.

A. C. Haynes, J. Qian, J. A. Reisz, C. M. Furdui, and W. T. Lowther, Molecular Basis for the Resistance of Human Mitochondrial 2-Cys Peroxiredoxin 3 to Hyperoxidation, J. Biol. Chem, vol.288, pp.29714-29723, 2013.

A. C. Haynes, J. Qian, J. A. Reisz, C. M. Furdui, and W. T. Lowther, Molecular basis for the resistance of human mitochondrial 2-Cys peroxiredoxin 3 to hyperoxidation, J. Biol. Chem, vol.288, pp.29714-29723, 2013.

A. Higdon, A. R. Diers, J. Y. Oh, A. Landar, and V. M. Darley-usmar, Cell signalling by reactive lipid species: new concepts and molecular mechanisms, Biochem. J, vol.442, pp.453-464, 2012.

B. K. Huang and H. D. Sikes, Quantifying intracellular hydrogen peroxide perturbations in terms of concentration, Redox Biol, vol.2, pp.955-962, 2014.

I. Ingold, C. , and M. , Selenium and iron, two elemental rivals in the ferroptotic death process, Oncotarget, vol.9, 2018.

Y. Inoue, T. Matsuda, K. Sugiyama, S. Izawa, and A. Kimura, Genetic Analysis of Glutathione Peroxidase in Oxidative Stress Response of Saccharomyces cerevisiae, J. Biol. Chem, vol.274, pp.27002-27009, 1999.

D. J. Jamieson, Oxidative stress responses of the yeast Saccharomyces cerevisiae, vol.17, 1998.

H. H. Jang, K. O. Lee, Y. H. Chi, B. G. Jung, S. K. Park et al., Two enzymes in one; two yeast peroxiredoxins display oxidative stressdependent switching from a peroxidase to a molecular chaperone function, Cell, vol.117, pp.625-635, 2004.

R. M. Jarvis, S. M. Hughes, and E. C. Ledgerwood, Peroxiredoxin 1 functions as a signal peroxidase to receive, transduce, and transmit peroxide signals in mammalian cells. Free Radic, Biol. Med, vol.53, pp.1522-1530, 2012.

M. Jeng, A. P. Campbell, T. Begley, A. Holmgren, D. A. Case et al., High-resolution solution structures of oxidized and reduced Escherichia coli thioredoxin, Structure, vol.2, pp.853-868, 1994.

T. J. Jonsson, M. S. Murray, L. C. Johnson, and W. T. Lowther, Reduction of cysteine sulfinic acid in peroxiredoxin by sulfiredoxin proceeds directly through a sulfinic phosphoryl ester intermediate, J Biol Chem, vol.283, pp.23846-23851, 2008.

T. J. Jönsson, L. C. Johnson, and W. T. Lowther, Structure of the sulphiredoxin-peroxiredoxin complex reveals an essential repair embrace, Nature, vol.451, pp.98-101, 2008.

T. J. Jonsson, L. C. Johnson, and W. T. Lowther, Protein engineering of the quaternary sulfiredoxin.peroxiredoxin enzyme.substrate complex reveals the molecular basis for cysteine sulfinic acid phosphorylation, J Biol Chem, vol.284, pp.33305-33310, 2009.

P. A. Karplus, A primer on peroxiredoxin biochemistry. Free Radic, Biol. Med, vol.80, pp.183-190, 2015.

Y. Kato, The Formation of Lipid Hydroperoxide-Derived Amide-Type Lysine Adducts on Proteins: A Review of Current Knowledge, Lipid Hydroperoxide-Derived Modification of Biomolecules, pp.21-39, 2014.

I. S. Kil, S. K. Lee, K. W. Ryu, H. A. Woo, M. Hu et al., Feedback Control of Adrenal Steroidogenesis via H2O2-Dependent, Reversible Inactivation of Peroxiredoxin III in Mitochondria, Mol. Cell, vol.46, pp.584-594, 2012.

K. Kim, H. Kim, and E. R. Stadtmann, The Isolation and Purification of a Specific "Protector"Protein Which Inhibits Enzyme Inactivation by a Thiol/Fe(III)/02Mixed-function Oxidation System, vol.8, 1988.

K. H. Kim, W. Lee, and E. E. Kim, Crystal structures of human peroxiredoxin 6 in different oxidation states, Biochem. Biophys. Res. Commun, vol.477, pp.717-722, 2016.

S. Kwon, S. K. Kim, D. Lee, and J. F. Kim, Comparative genomics and experimental evolution of, Escherichia coli, vol.21, p.3, 2015.

J. D. Lambeth, NOX enzymes and the biology of reactive oxygen, Nat. Rev. Immunol, vol.4, pp.181-189, 2004.

J. Lee, D. Spector, C. Godon, J. Labarre, and M. B. Toledano, A New Antioxidant with Alkyl Hydroperoxide Defense Properties in Yeast, J. Biol. Chem, vol.274, pp.4537-4544, 1999.

S. Li, N. A. Peterson, M. Kim, C. Kim, L. Hung et al., Crystal Structure of AhpE from Mycobacterium tuberculosis, a 1-Cys Peroxiredoxin, J. Mol. Biol, vol.346, pp.1035-1046, 2005.

T. W. Lyons, C. T. Reinhard, and N. J. Planavsky, The rise of oxygen in Earth's early ocean and atmosphere, Nature, vol.506, pp.307-315, 2014.

S. S. Marla, J. Lee, and J. T. Groves, Peroxynitrite rapidly permeates phospholipid membranes, Proc. Natl. Acad. Sci, vol.94, pp.14243-14248, 1997.

J. L. Martin, Thioredoxin -a fold for all reasons, Structure, vol.3, pp.245-250, 1995.

D. B. Medinas, J. C. Toledo, G. Cerchiaro, A. T. Do-amaral, L. De-rezende et al., Peroxymonocarbonate and Carbonate Radical Displace the Hydroxyl-like Oxidant in the Sod1 Peroxidase Activity under Physiological Conditions, Chem. Res. Toxicol, vol.22, pp.639-648, 2009.

J. L. Meitzler, S. Antony, Y. Wu, A. Juhasz, H. Liu et al., NADPH Oxidases: A Perspective on Reactive Oxygen Species Production in Tumor Biology, Antioxid. Redox Signal, vol.20, pp.2873-2889, 2014.

N. B. Milev, S. Rhee, and A. B. Reddy, Cellular Timekeeping: It's Redox o'Clock, Cold Spring Harb. Perspect. Biol, vol.10, 2018.

B. Miroux and J. E. Walker, Over-production of Proteins inEscherichia coli: Mutant Hosts that Allow Synthesis of some Membrane Proteins and Globular Proteins at High Levels, J. Mol. Biol, vol.260, pp.289-298, 1996.

P. Nagy, A. Karton, A. Betz, A. V. Peskin, P. Pace et al., Model for the Exceptional Reactivity of Peroxiredoxins 2 and 3 with Hydrogen Peroxide: A KINETIC AND COMPUTATIONAL STUDY, J. Biol. Chem, vol.286, pp.18048-18055, 2011.

F. Neiers, A. Kriznik, S. Boschi-muller, and G. Branlant, Evidence for a New Sub-class of, 2004.
URL : https://hal.archives-ouvertes.fr/hal-01636385

, Methionine Sulfoxide Reductases B with an Alternative Thioredoxin Recognition Signature, J. Biol. Chem, vol.279, pp.42462-42468

K. J. Nelson, S. T. Knutson, L. Soito, C. Klomsiri, L. B. Poole et al., Analysis of the peroxiredoxin family: Using active-site structure and sequence information for global classification and residue analysis, Proteins Struct. Funct. Bioinforma, vol.79, pp.947-964, 2011.

K. J. Nelson, D. Parsonage, P. A. Karplus, and L. B. Poole, Evaluating peroxiredoxin sensitivity towards inactivation by peroxide substrates, Methods Enzymol, vol.527, 2013.

M. H. Nielsen, R. T. Kidmose, and L. B. Jenner, Structure of TSA2 reveals novel features of the active-site loop of peroxiredoxins, Acta Crystallogr. Sect. Struct. Biol, vol.72, pp.158-167, 2016.

R. Ogusucu, D. Rettori, D. C. Munhoz, L. E. Soares-netto, A. et al., Reactions of yeast thioredoxin peroxidases I and II with hydrogen peroxide and peroxynitrite: Rate constants by competitive kinetics. Free Radic, Biol. Med, vol.42, pp.326-334, 2007.

M. Del-olmo, A. Kramer, and H. Herzel, A Robust Model for Circadian Redox Oscillations, Int. J. Mol. Sci, vol.20, p.2368, 2019.

A. Olry, S. Boschi-muller, M. Marraud, S. Sanglier-cianferani, A. Van-dorsselear et al., Characterization of the methionine sulfoxide reductase activities of PILB, a probable virulence factor from Neisseria meningitidis, J Biol Chem, vol.277, pp.12016-12022, 2002.
URL : https://hal.archives-ouvertes.fr/hal-01690847

J. S. O'neill and A. B. Reddy, Circadian clocks in human red blood cells, Nature, vol.469, pp.498-503, 2011.

J. S. O'neill, G. Van-ooijen, L. E. Dixon, C. Troein, F. Corellou et al., Circadian rhythms persist without transcription in a eukaryote, Nature, vol.469, pp.554-558

M. A. Packer and M. P. Murphy, Peroxynitrite Formed by Simultaneous Nitric Oxide and Superoxide Generation Causes Cyclosporin-A-Sensitive Mitochondrial Calcium Efflux and Depolarisation, Eur. J. Biochem, vol.234, pp.231-239, 1995.

P. B. Palde and K. S. Carroll, A universal entropy-driven mechanism for thioredoxin-target recognition, Proc. Natl. Acad. Sci, vol.112, pp.7960-7965, 2015.

S. G. Park, M. Cha, W. Jeong, and I. Kim, Distinct Physiological Functions of Thiol Peroxidase Isoenzymes in Saccharomyces cerevisiae, J. Biol. Chem, vol.275, pp.5723-5732, 2000.

D. Parsonage, K. J. Nelson, G. Ferrer-sueta, S. Alley, P. A. Karplus et al., Dissecting Peroxiredoxin Catalysis: Separating Binding, Peroxidation, and Resolution for a Bacterial AhpC, Biochemistry, vol.54, pp.1567-1575, 2015.

C. E. Paulsen and K. S. Carroll, Orchestrating Redox Signaling Networks through Regulatory Cysteine Switches, ACS Chem. Biol, vol.5, pp.47-62, 2010.

A. Perkins, M. C. Gretes, K. J. Nelson, L. B. Poole, and P. A. Karplus, Mapping the Active Site Helix-to-Strand Conversion of CxxxxC Peroxiredoxin Q Enzymes, Biochemistry, vol.51, pp.7638-7650, 2012.

A. Perkins, K. J. Nelson, J. R. Williams, D. Parsonage, L. B. Poole et al., The Sensitive Balance between the Fully Folded and Locally Unfolded Conformations of a Model Peroxiredoxin, Biochemistry, vol.52, pp.8708-8721, 2013.

A. Perkins, K. J. Nelson, D. Parsonage, L. B. Poole, and P. A. Karplus, Peroxiredoxins: guardians against oxidative stress and modulators of peroxide signaling, Trends Biochem. Sci, vol.40, pp.435-445, 2015.

A. Perkins, D. Parsonage, K. J. Nelson, O. M. Ogba, P. H. Cheong et al., Peroxiredoxin Catalysis at Atomic Resolution. Structure, vol.24, pp.1668-1678, 2016.

A. V. Peskin, F. M. Low, L. N. Paton, G. J. Maghzal, M. B. Hampton et al., , 2007.

, The High Reactivity of Peroxiredoxin 2 with H 2 O 2 Is Not Reflected in Its Reaction with Other Oxidants and Thiol Reagents, J. Biol. Chem, vol.282, pp.11885-11892

A. V. Peskin, N. Dickerhof, R. A. Poynton, L. N. Paton, P. E. Pace et al., Hyperoxidation of Peroxiredoxins 2 and 3: RATE CONSTANTS FOR THE REACTIONS OF THE SULFENIC ACID OF THE PEROXIDATIC CYSTEINE, J. Biol. Chem, vol.288, pp.14170-14177, 2013.

C. S. Pillay, J. S. Hofmeyr, B. G. Olivier, J. L. Snoep, and J. M. Rohwer, Enzymes or redox couples? The kinetics of thioredoxin and glutaredoxin reactions in a systems biology context, Biochem. J, vol.417, pp.269-277, 2009.

S. Portillo-ledesma, L. M. Randall, D. Parsonage, J. Dalla-rizza, P. A. Karplus et al., Differential Kinetics of Two-Cysteine Peroxiredoxin Disulfide Formation Reveal a Novel Model for Peroxide Sensing, Biochemistry, vol.57, pp.3416-3424, 2018.

C. Prolo, M. N. Álvarez, and R. Radi, Peroxynitrite, a potent macrophage-derived oxidizing cytotoxin to combat invading pathogens: Peroxynitrite in Pathogen Invasion, BioFactors, vol.40, pp.215-225, 2014.

T. Rabilloud, M. Heller, F. Gasnier, S. Luche, C. Rey et al., Proteomics Analysis of Cellular Response to Oxidative Stress: EVIDENCE FOR IN VIVO OVEROXIDATION OF PEROXIREDOXINS AT THEIR ACTIVE SITE, J. Biol, 2002.

, Chem, vol.277, pp.19396-19401

S. Rahuel-clermont, R. Bchini, S. Barbe, S. Boutserin, I. André et al., Enzyme Active Site Loop Revealed as a Gatekeeper for Cofactor Flip by Targeted Molecular Dynamics Simulations and FRET-Based Kinetics, ACS Catal, vol.9, pp.1337-1346, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02022828

L. M. Randall, J. Dalla-rizza, D. Parsonage, J. Santos, R. A. Mehl et al., Unraveling the effects of peroxiredoxin 2 nitration; role of C-terminal tyrosine 193. Free Radic, Biol. Med, vol.141, pp.492-501, 2019.

K. G. Reddie and K. S. Carroll, Expanding the functional diversity of proteins through cysteine oxidation, Curr. Opin. Chem. Biol, vol.12, pp.746-754, 2008.

A. M. Reyes, M. Hugo, A. Trostchansky, L. Capece, R. Radi et al., Oxidizing substrate specificity of Mycobacterium tuberculosis alkyl hydroperoxide reductase E: kinetics and mechanisms of oxidation and overoxidation. Free Radic, Biol. Med, vol.51, pp.464-473, 2011.

S. G. Rhee, S. W. Kang, W. Jeong, T. S. Chang, K. S. Yang et al., Intracellular messenger function of hydrogen peroxide and its regulation by peroxiredoxins, Curr Opin Cell Biol, vol.17, pp.183-189, 2005.

N. Rouhier, E. Gelhaye, C. Corbier, and J. P. Jacquot, Active site mutagenesis and phospholipid hydroperoxide reductase activity of poplar type II peroxiredoxin, Physiol. Plant, vol.120, pp.57-62, 2004.
URL : https://hal.archives-ouvertes.fr/hal-02672433

X. Roussel, G. Béchade, A. Kriznik, A. Van-dorsselaer, S. Sanglier-cianferani et al., Evidence for the Formation of a Covalent Thiosulfinate Intermediate with Peroxiredoxin in the Catalytic Mechanism of Sulfiredoxin, J. Biol. Chem, vol.283, pp.22371-22382, 2008.
URL : https://hal.archives-ouvertes.fr/hal-01652689

X. Roussel, A. Kriznik, C. Richard, S. Rahuel-clermont, and G. Branlant, Catalytic Mechanism of Sulfiredoxin from Saccharomyces cerevisiae Passes through an Oxidized Disulfide Sulfiredoxin Intermediate That Is Reduced by Thioredoxin, J. Biol. Chem, vol.284, pp.33048-33055, 2009.
URL : https://hal.archives-ouvertes.fr/hal-01682337

F. Saccoccia, P. Di-micco, G. Boumis, M. Brunori, I. Koutris et al., Moonlighting by Different Stressors: Crystal Structure of the Chaperone Species of a 2-Cys Peroxiredoxin, Structure, vol.20, pp.429-439, 2012.
URL : https://hal.archives-ouvertes.fr/pasteur-00952065

T. Sakamoto, K. Maebayashi, Y. Nakagawa, and H. Imai, Deletion of the four phospholipid hydroperoxide glutathione peroxidase genes accelerates aging in Caenorhabditis elegans, Genes Cells, vol.19, pp.778-792, 2014.

A. Smeets, C. Marchand, D. Linard, B. Knoops, and J. Declercq, The crystal structures of oxidized forms of human peroxiredoxin 5 with an intramolecular disulfide bond confirm the proposed enzymatic mechanism for atypical 2-Cys peroxiredoxins, Arch. Biochem. Biophys, vol.477, pp.98-104, 2008.

M. C. Sobotta, W. Liou, S. Stöcker, D. Talwar, M. Oehler et al., Peroxiredoxin-2 and STAT3 form a redox relay for H2O2 signaling, Nat. Chem. Biol, vol.11, pp.64-70, 2015.

L. Soito, C. Williamson, S. T. Knutson, J. S. Fetrow, L. B. Poole et al., PREX: PeroxiRedoxin classification indEX, a database of subfamily assignments across the diverse peroxiredoxin family, Nucleic Acids Res, vol.39, pp.332-337, 2011.

S. Stöcker, K. Van-laer, A. Mijuskovic, and T. P. Dick, The Conundrum of Hydrogen Peroxide Signaling and the Emerging Role of Peroxiredoxins as Redox Relay Hubs, Antioxid. Redox Signal, vol.28, pp.558-573, 2018.

F. W. Studier, Protein production by auto-induction in high-density shaking cultures, Protein Expr. Purif, vol.41, pp.207-234, 2005.

C. A. Tairum, M. A. De-oliveira, B. B. Horta, F. J. Zara, and L. E. Netto, Disulfide Biochemistry in 2-Cys Peroxiredoxin: Requirement of Glu50 and Arg146 for the Reduction of Yeast Tsa1 by Thioredoxin, J. Mol. Biol, vol.424, pp.28-41, 2012.

G. Takebe, J. Yarimizu, Y. Saito, T. Hayashi, H. Nakamura et al., A Comparative Study on the Hydroperoxide and Thiol Specificity of the Glutathione Peroxidase Family and Selenoprotein P, J. Biol. Chem, vol.277, pp.41254-41258, 2002.

T. Tanaka, S. Izawa, and Y. Inoue, GPX2 , Encoding a Phospholipid Hydroperoxide Glutathione Peroxidase Homologue, Codes for an Atypical 2-Cys Peroxiredoxin in Saccharomyces cerevisiae, J. Biol. Chem, vol.280, pp.42078-42087, 2005.

J. J. Tanner, Z. D. Parsons, A. H. Cummings, H. Zhou, and K. S. Gates, Redox Regulation of Protein Tyrosine Phosphatases: Structural and Chemical Aspects, Antioxid. Redox Signal, vol.15, pp.77-97, 2011.

L. Tarrago, E. Laugier, M. Zaffagnini, C. Marchand, P. Le-maréchal et al., Regeneration Mechanisms of Arabidopsis thaliana Methionine Sulfoxide Reductases B by Glutaredoxins and Thioredoxins, J. Biol. Chem, vol.284, pp.18963-18971, 2009.
URL : https://hal.archives-ouvertes.fr/hal-02665795

F. Teixeira, E. Tse, H. Castro, K. A. Makepeace, B. A. Meinen et al., Chaperone activation and client binding of a 2-cysteine peroxiredoxin, Nat. Commun, vol.10, 2019.

D. M. Tobin and L. Ramakrishnan, TB: the Yin and Yang of lipid mediators, Curr. Opin. Pharmacol, vol.13, pp.641-645, 2013.

M. B. Toledano, A. Delaunay, L. Monceau, and F. Tacnet, Microbial H2O2 sensors as archetypical redox signaling modules, Trends Biochem Sci, vol.29, pp.351-357, 2004.

D. F. Trindade, G. Cerchiaro, A. , and O. , A Role for Peroxymonocarbonate in the Stimulation of Biothiol Peroxidation by the Bicarbonate/Carbon Dioxide Pair, Chem. Res. Toxicol, vol.19, pp.1475-1482, 2006.

D. R. Truzzi, F. R. Coelho, V. Paviani, S. V. Alves, L. E. Netto et al., The bicarbonate/carbon dioxide pair increases hydrogen peroxide-mediated hyperoxidation of human peroxiredoxin 1, J. Biol. Chem. jbc, 2019.

B. Turner-ivey, Y. Manevich, J. Schulte, E. Kistner-griffin, A. Jezierska-drutel et al., Role for Prdx1 as a specific sensor in redox-regulated senescence in breast cancer, Oncogene, vol.32, pp.5302-5314, 2013.

M. Valko, C. J. Rhodes, J. Moncol, M. Izakovic, and M. Mazur, Free radicals, metals and antioxidants in oxidative stress-induced cancer, Chem. Biol. Interact, vol.160, pp.1-40, 2006.

J. M. Van-raamsdonk and S. Hekimi, Superoxide dismutase is dispensable for normal animal lifespan, Proc. Natl. Acad. Sci, vol.109, pp.5785-5790, 2012.

D. S. Vazquez, A. Zeida, W. A. Agudelo, M. R. Montes, G. Ferrer-sueta et al., , 2020.

, Exploring the conformational transition between the fully folded and locally unfolded substates of Escherichia coli thiol peroxidase, Phys. Chem. Chem. Phys

E. Veal, T. Jackson, and H. Latimer, Role/s of 'Antioxidant' Enzymes in Ageing, Biochemistry and Cell Biology of Ageing: Part I Biomedical Science, pp.425-450, 2018.

E. A. Veal, Z. E. Underwood, L. E. Tomalin, B. A. Morgan, and C. S. Pillay, Hyperoxidation of Peroxiredoxins: Gain or Loss of Function?, Antioxid. Redox Signal, 2017.

A. P. Vivancos, E. A. Castillo, B. Biteau, C. Nicot, J. Ayté et al., , 2005.

, A cysteine-sulfinic acid in peroxiredoxin regulates H2O2-sensing by the antioxidant Pap1 pathway, Proc. Natl. Acad. Sci. U. S. A, vol.102, pp.8875-8880

Y. Wang, R. Branicky, A. Noë, and S. Hekimi, Superoxide dismutases: Dual roles in controlling ROS damage and regulating ROS signaling, J. Cell Biol, vol.217, pp.1915-1928, 2018.

T. Watanabe and T. Nakamura, The Effects of Linoleate Hydroperoxide on Respiration and Oxidative Phosphorylation of Rat Liver Mitochondria, J. Biochem, vol.86, pp.1041-1047, 1979.

J. West, T. Roston, J. David, K. Allan, and M. Loberg, Piecing Together How Peroxiredoxins Maintain Genomic Stability, Antioxidants, vol.7, p.177, 2018.

C. C. Winterbourn, Reconciling the chemistry and biology of reactive oxygen species, Nat. Chem. Biol, vol.4, pp.278-286, 2008.

C. C. Winterbourn, The Biological Chemistry of Hydrogen Peroxide, In Methods in Enzymology, pp.3-25, 2013.

C. C. Winterbourn and M. B. Hampton, Thiol chemistry and specificity in redox signaling. Free Radic, Biol. Med, vol.45, pp.549-561, 2008.

C. C. Winterbourn and D. Metodiewa, Reactivity of biologically important thiol compounds with superoxide and hydrogen peroxide. Free Radic, Biol. Med, vol.27, pp.322-328, 1999.

J. Wong-ekkabut, Z. Xu, W. Triampo, I. Tang, D. P. Tieleman et al., Effect of Lipid Peroxidation on the Properties of Lipid Bilayers: A Molecular Dynamics Study, Biophys. J, 2017.

H. A. Woo, S. Kang, H. K. Kim, K. Yang, H. Z. Chae et al., Reversible Oxidation of the Active Site Cysteine of Peroxiredoxins to Cysteine Sulfinic Acid: IMMUNOBLOT DETECTION WITH ANTIBODIES SPECIFIC FOR THE HYPEROXIDIZED CYSTEINE-CONTAINING SEQUENCE, J. Biol. Chem, vol.278, pp.47361-47364, 2003.

Z. A. Wood, E. Schröder, J. R. Harris, and L. B. Poole, Structure, mechanism and regulation of peroxiredoxins, Trends Biochem. Sci, vol.28, pp.32-40, 2003.

Z. A. Wood, L. B. Poole, and P. A. Karplus, Peroxiredoxin evolution and the regulation of hydrogen peroxide signaling, Science, vol.300, pp.650-653, 2003.

N. A. Yewdall, H. Venugopal, A. Desfosses, V. Abrishami, Y. Yosaatmadja et al., Structures of Human Peroxiredoxin 3 Suggest Self-Chaperoning Assembly that Maintains Catalytic State, Structure, vol.24, pp.1120-1129, 2016.

A. Zeida, M. Trujillo, G. Ferrer-sueta, A. Denicola, D. A. Estrin et al., Catalysis of Peroxide Reduction by Fast Reacting Protein Thiols: Focus Review, Chem. Rev. acs.chemrev.9b00371, 2019.

, Marouane Libiad 3 , Hélène Le Cordier 1 , Samia Boukhenouna 1 , Michel B Toledano 3 & Sophie Rahuel, Alexandre Kriznik, vol.1

U. Imopa, . De-lorraine, and B. Cnrs, Campus Biologie Santé, F-54000 Nancy, France 2 UMS2008 IBSLor, Biophysics and Structural Biology core facility, vol.2

*. ,