12
Edinburgh Research Explorer Induction of sulfiredoxin expression and reduction of peroxiredoxin hyperoxidation by the neuroprotective Nrf2 activator 3H-1,2-dithiole-3-thione Citation for published version: Soriano, FX, Leveille, F, Papadia, S, Higgins, LG, Varley, J, Baxter, P, Hayes, JD & Hardingham, GE 2008, 'Induction of sulfiredoxin expression and reduction of peroxiredoxin hyperoxidation by the neuroprotective Nrf2 activator 3H-1,2-dithiole-3-thione', Journal of Neurochemistry, vol. 107, no. 2, pp. 533-543. https://doi.org/10.1111/j.1471-4159.2008.05648.x Digital Object Identifier (DOI): 10.1111/j.1471-4159.2008.05648.x Link: Link to publication record in Edinburgh Research Explorer Document Version: Peer reviewed version Published In: Journal of Neurochemistry Publisher Rights Statement: Published in final edited form as: J Neurochem. 2008 October ; 107(2): 533–543. doi:10.1111/j.1471-4159.2008.05648.x. General rights Copyright for the publications made accessible via the Edinburgh Research Explorer is retained by the author(s) and / or other copyright owners and it is a condition of accessing these publications that users recognise and abide by the legal requirements associated with these rights. Take down policy The University of Edinburgh has made every reasonable effort to ensure that Edinburgh Research Explorer content complies with UK legislation. If you believe that the public display of this file breaches copyright please contact [email protected] providing details, and we will remove access to the work immediately and investigate your claim. Download date: 12. Feb. 2021

Edinburgh Research Explorer · Key among these is the thioredoxin-peroxiredoxin system which is an important reducer of oxidative stressors such as peroxides (Winyard et al. 2005)

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Edinburgh Research Explorer · Key among these is the thioredoxin-peroxiredoxin system which is an important reducer of oxidative stressors such as peroxides (Winyard et al. 2005)

Edinburgh Research Explorer

Induction of sulfiredoxin expression and reduction ofperoxiredoxin hyperoxidation by the neuroprotective Nrf2activator 3H-1,2-dithiole-3-thione

Citation for published version:Soriano, FX, Leveille, F, Papadia, S, Higgins, LG, Varley, J, Baxter, P, Hayes, JD & Hardingham, GE 2008,'Induction of sulfiredoxin expression and reduction of peroxiredoxin hyperoxidation by the neuroprotectiveNrf2 activator 3H-1,2-dithiole-3-thione', Journal of Neurochemistry, vol. 107, no. 2, pp. 533-543.https://doi.org/10.1111/j.1471-4159.2008.05648.x

Digital Object Identifier (DOI):10.1111/j.1471-4159.2008.05648.x

Link:Link to publication record in Edinburgh Research Explorer

Document Version:Peer reviewed version

Published In:Journal of Neurochemistry

Publisher Rights Statement:Published in final edited form as:J Neurochem. 2008 October ; 107(2): 533–543. doi:10.1111/j.1471-4159.2008.05648.x.

General rightsCopyright for the publications made accessible via the Edinburgh Research Explorer is retained by the author(s)and / or other copyright owners and it is a condition of accessing these publications that users recognise andabide by the legal requirements associated with these rights.

Take down policyThe University of Edinburgh has made every reasonable effort to ensure that Edinburgh Research Explorercontent complies with UK legislation. If you believe that the public display of this file breaches copyright pleasecontact [email protected] providing details, and we will remove access to the work immediately andinvestigate your claim.

Download date: 12. Feb. 2021

Page 2: Edinburgh Research Explorer · Key among these is the thioredoxin-peroxiredoxin system which is an important reducer of oxidative stressors such as peroxides (Winyard et al. 2005)

*Centre for Neuroscience Research, University of Edinburgh, Edinburgh, UK

�Biomedical Research Centre, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK

Oxidative stress occurs because of an imbalance betweenproduction of reactive oxygen species and the cell’s capacityto neutralize them through its intrinsic antioxidant defences.Key among these is the thioredoxin-peroxiredoxin systemwhich is an important reducer of oxidative stressors such asperoxides (Winyard et al. 2005). The thioredoxin systemprotects against H2O2-induced apoptosis, and its inhibitionpromotes oxidative stress and cell death (Yoshida et al.2005). The thioredoxin-peroxiredoxin system detoxifiesperoxides by transferring reducing equivalents from NADPHto peroxides via thioredoxin reductase, thioredoxin andfinally peroxiredoxins (Prxs). Prxs are a ubiquitous family ofperoxidases with cytoprotective and antioxidative effects(Immenschuh and Baumgart-Vogt 2005). The 2-Cys Prxs isthe predominant Prx subfamily, comprising Prx I-IV (Woodet al. 2003) and are implicated in protecting neuronal cellsfrom Ab toxicity (Yao et al. 2007), excitotoxicity (Hattoriet al. 2003), oxygen-glucose deprivation (Boulos et al.2007), peroxide (Sanchez-Font et al. 2003; Fang et al.2007), and MPP+ toxicity (Qu et al. 2007).

These Prxs contain a peroxidatic cysteine residue, oxidizedby peroxides to cysteine sulfenic acid, which then forms adisulfide bond with the resolving cysteine, which is in turnreduced by thioredoxin (Wood et al. 2003). Sometimes, Prx-sulfenic acid is further oxidized by peroxide to sulfinic(-SO2H) or sulfonic (-SO3H) acid, causing inactivation ofperoxidase activity (Rhee et al. 2007). Prx-SO2/3H is not asubstrate for the resolving cysteine and cannot be reduced by

Received July 23, 2008; revised manuscript received August 8, 2008;accepted August 12, 2008.Address correspondence and reprint requests to Giles E. Hardingham,

Centre for Neuroscience Research, University of Edinburgh, EdinburghEH8 9XD, UK. E-mail: [email protected] used: ARE, antioxidant response element; AP-1, acti-

vator protein-1; D3T, 3H-1,2-dithiole-3-thione; GAPDH, Glycer-aldehyde-3-phosphate dehydrogenase; GFAP, Glial fibrillary acidicprotein; DAPI, 4,6-diamino-2-phenylindole; eGFP, enhanced greenfluorescent protein; NeuN, Neuronal nuclear antigen; Nrf2, Nuclearfactor erythroid 2-related factor; Prx, Peroxiredoxin; tBHQ, tert-butylhydroquinone.

Abstract

Peroxiredoxins are an important family of cysteine-based

antioxidant enzymes that exert a neuroprotective effect in

several models of neurodegeneration. However, under oxi-

dative stress they are vulnerable to inactivation through

hyperoxidation of their active site cysteine residues. We show

that in cortical neurons, the chemopreventive inducer 3H-1,2-

dithiole-3-thione (D3T), that activates the transcription factor

Nuclear factor erythroid 2-related factor (Nrf2), inhibits the

formation of inactivated, hyperoxidized peroxiredoxins fol-

lowing oxidative trauma, and protects neurons against oxi-

dative stress. In both neurons and glia, Nrf2 expression and

treatment with chemopreventive Nrf2 activators, including

D3T and sulforaphane, up-regulates sulfiredoxin, an enzyme

responsible for reducing hyperoxidized peroxiredoxins.

Induction of sulfiredoxin expression is mediated by Nrf2, act-

ing via a cis-acting antioxidant response element (ARE) in its

promoter. The ARE element in Srxn1 contains an embedded

activator protein-1 (AP-1) site which directs induction of Srxn1

by synaptic activity. Thus, raising Nrf2 activity in neurons

prevents peroxiredoxin hyperoxidation and induces a new

member of the ARE-gene family, whose enzymatic function of

reducing hyperoxidized peroxiredoxins may contribute to the

neuroprotective effects of Nrf2 activators.

Keywords: chemoprevention, neurodegeneration, neuropro-

tection, oxidative stress, phase II enzymes, thioredoxin.

J. Neurochem. (2008) 107, 533–543.

JOURNAL OF NEUROCHEMISTRY | 2008 | 107 | 533–543 doi: 10.1111/j.1471-4159.2008.05648.x

� 2008 The AuthorsJournal Compilation � 2008 International Society for Neurochemistry, J. Neurochem. (2008) 107, 533–543 533

Page 3: Edinburgh Research Explorer · Key among these is the thioredoxin-peroxiredoxin system which is an important reducer of oxidative stressors such as peroxides (Winyard et al. 2005)

thioredoxin. Hyperoxidation takes place when there is notenough reduced Prx to deal with peroxides present, and isassociated with oxidative neuronal death in vitro, and alsoischemic brain damage in vivo (Papadia et al. 2008).Previously, hyperoxidation of Prx was thought to beirreversible. However, more recently it has been found thatPrx-SO2H can be reduced back to the catalytically activethiol form in eukaryotic cells by the ATP-dependentreductase, sulfiredoxin (Biteau et al. 2003; Rhee et al.2007; Jonsson et al. 2008). The activity of sulfiredoxinrestores inactive Prxs back to the thioredoxin cycle andprevents permanent oxidative inactivation of Prxs by strongoxidative insults. Over-expression of sulfiredoxin has beenshown to prevent Prx hyperoxidation in response to anoxidative insult (Woo et al. 2005). Conversely, knockdownof sulfiredoxin prevents reduction of Prx-SO2H following atransient oxidative insult (Chang et al. 2004).

One known defence against oxidative insults is theinduction of a group of genes encoding antioxidative anddrug-metabolizing enzymes (also known as Phase IIenzymes). These genes are induced by a variety of smallthiol-active molecules including the potent chemopreventiveagent 3H-1,2-dithiole-3-thione (D3T), as well as dietaryphytochemicals such as Sulforaphane (Nguyen et al. 2004).Mild oxidative stress also induces these genes (Giudice andMontella 2006). Transcriptional regulation of this group ofgenes is mediated by a cis-acting promoter element termedthe antioxidant response element (ARE), which recruits thetranscription factor Nuclear factor erythroid 2-related factor(Nrf2) as a heterodimer with small Maf proteins (Zhang2006). Nrf2 levels are constitutively low because of beingtargeted for degradation by Keap1. Under conditions ofoxidative stress, Nrf2 degradation is slowed and Nrf2accumulates in the nucleus and activates ARE-containinggenes, with a net antioxidative effect (Nguyen et al. 2004).Small molecule activators of Nrf2 also act by interferingwith Keap1-mediated degradation. Activation of Nrf2 andinduction of ARE-driven defences is implicated in protect-ing against a variety of diseases in many organs and tissues,including autoimmune diseases, cancer, cardiovasculardisease, neurodegenerative disease and ischemia (Lee et al.2005; Shih et al. 2005; Giudice and Montella 2006; Zhang2006). The ability of Nrf2 to exert a cytoprotective effect inneural cells has mainly been studied in the context of glia.In a mixed culture of neurons and glial cells, Nrf2expression in glial cells can confer protection on a largenumber of normal non-overexpressing neurons (Shih et al.2003; Kraft et al. 2004).

Here we find that oxidant-induced neuronal apoptosis canbe prevented by expression of Nrf2 specifically in neurons,and also by the Nrf2 activator D3T. Furthermore, D3Tstrongly impairs oxidant-induced peroxiredoxin hyperoxida-tion in neurons. D3T induces expression of the enzymeresponsible for reducing hyperoxidized peroxiredoxins, sul-

firedoxin. We find that this induction is mediated at thetranscriptional level and that sulfiredoxin is a new member ofthe Nrf2/ARE regulated gene family. Given its function, itmay form part of the cytoprotective gene battery, thetranscription of which is promoted by chemopreventiveagents.

Materials and methods

Tissue culture and the induction of oxidative stressCortical rat neurons were cultured as described (Hardingham et al.2002) from E21 rats except that growth medium contained B27

(Invitrogen, Carlsbad, CA, USA). A single dose of anti-mitotic

agent (AraC, 4.8 lM) was added to the cultures at days in vitro(DIV) 4 to minimize glial numbers. Experiments were carried out

after being cultured for 8–10 days during which cortical neurons

develop a network of processes, express functional NMDA-type and

a-amino-3-hydroxy-5-methylisoxazole-4-propionate/kainate-type

glutamate receptors, and form synaptic contacts. Experiments were

performed after transferring neurons at DIV8 into defined medium

lacking trophic support ‘TMo’ (Papadia et al. 2005): this is

composed of 10% MEM (Invitrogen) and 90% Salt-Glucose-

Glycine medium (SGG: 114 mM NaCl, 0.219% NaHCO3,

5.292 mM KCl, 1 mM MgCl2, 2 mM CaCl2, 10 mM HEPES,

1 mM Glycine, 30 mM Glucose, 0.5 mM sodium pyruvate, 0.1%

Phenol Red; osmolarity 325 mosm/L, hereafter TMo). D3T (10–

25 lM) was applied to neurons 16 h prior to the application of an

oxidative insult in the form of H2O2 (100 lM, stabilized solution:

Sigma, St Louis, MO, USA). Neurons were fixed after a further 24 h

and subjected to 4,6-diamino-2-phenylindole (DAPI) staining and

cell death quantified by counting (blind) the number of apoptotic

nuclei as a percentage of the total. Approximately 1500 cells were

counted per treatment, across four independent experiments.

Morphologically, peroxide-treated neurons show typical signs of

apoptotic-like cell death (shrunken cell body and large round

chromatin clumps).

Cultures of cortical glial cells were obtained by plating mixed

neuronal/glial cultures at low density in 10% fetal bovine serum/

Dulbecco’s modified Eagle’s medium (no anti-mitotic). After 6 days

the small number of neurons that remained were killed by 1 mM

NMDA overnight. This procedure leaves > 99% Glial fibrillary

acidic protein (GFAP) positive glial cells. HEK293 cells were

maintained in 10% FBS/Dulbecco’s modified Eagle’s medium and

passaged 1 : 5 every 4 days.

Transfection and following the fate of transfected cellsAll transfections on all cell types were performed using Lipofec-

tamine 2000 (Invitrogen). Neurons and glial cells were transfected at

DIV8 in TMo (see above). Transfection efficiency was approxi-

mately 5% for neuronal cultures. It was found that more than 99% of

enhanced green fluorescent protein (eGFP)-expressing transfected

neurons were neuronal nuclear antigen (NeuN)-positive, and < 1%

were GFAP positive (see results) confirming their neuronal identity.

For monitoring the fate of Nrf2-expressing neurons, peGFP was

used to track the fate of transfected neurons expressing the plasmid

of interest (Nrf2 vs. globin control). To ensure that green fluorescent

protein (GFP)-positive neurons were also expressing the plasmid of

Journal Compilation � 2008 International Society for Neurochemistry, J. Neurochem. (2008) 107, 533–543� 2008 The Authors

534 | F. X. Soriano et al.

Page 4: Edinburgh Research Explorer · Key among these is the thioredoxin-peroxiredoxin system which is an important reducer of oxidative stressors such as peroxides (Winyard et al. 2005)

interest, a favorable ratio was used (peGFP: plasmid of interest,

1 : 2). Coexpression at this ratio was confirmed in the case of a

plasmid encoding red fluorescent protein (Papadia et al. 2008).

Pictures of GFP-expressing neurons were taken using a Leica

AF6000 LX imaging system (Leica Microsystems, Wetzlar,

Germany), with a DFC350 FX digital camera. Neurons were then

treated with 100 lM H2O2 and images of the same cells were taken

24 h after H2O2 exposure. Cell death was assessed by counting the

number of surviving GFP-positive neurons pre- and post-exposure

to H2O2. In the vast majority of cases, death was easily spotted as an

absence of a healthy GFP-expressing cell where one once was. In

place of the cell, there was in most cases (> 90%) evidence of death

in the form of fragmented neurites, fluorescent cell debris, and an

apoptotic nucleus. This confirmed that the cells were genuinely

dying as opposed to more unlikely scenario such as peroxide-

induced quenching of eGFP fluorescence in a sub-population of

neurons. This is also underlined by the fact that death measured by

this technique is blocked by caspase inhibitors (Papadia et al. 2008).For each plasmid/condition, the fate of approximately 150 neurons

was monitored over three independent experiments.

Plasmids and reporter assaysSrxn1-Luc (Papadia et al. 2008) was subjected to site-directed

mutagenesis of its ARE with the QuikChange II XL site-

directed mutagenesis kit (Stratagene), using the oligonucleotide 5¢-ACCTGCAAACTCACCCTGGGCCCGCGACCCGACGCGTC-3¢and its reverse-complementary sequence. The mutation was verified

by sequencing. ARE-Luc (Numazawa et al. 2003), pcDNA3.1/

V5HisBmNrf2 and pcDNA3.1/V5HisCmKeap1 (McMahon et al.2003) and pEF-Nrf2 (Kotkow and Orkin 1995) have been described

previously. Firefly luciferase-based reporter gene constructs (Srxn1-Luc and its ARE mutated variant were transfected along with a

renilla expression vector (pTK-RL), and also, where relevant, other

expression vectors. Luciferase assays were performed 30 h post-

transfection using the Dual Glo assay kit (Promega, Madison, WI,

USA) with Firefly luciferase-based reporter gene activity normalized

to the Renilla control (pTK-RL plasmid) in all cases.

Immunocytochemistry, western blotting and antibodiesImmunofluorescence was performed as described (Mckenzie et al.2005). Antibodies (Sulfiredoxin-S17, 1 : 500, Santa Cruz Bio-

technology, Santa Cruz, CA, USA; NeuN, 1 : 15 Chemicon,

Temecula, CA, USA; GFAP, 1 : 1000, Sigma) were incubated

with fixed cells overnight and visualized using biotinylated

secondary antibody/cy3-conjugated streptavidin. Nuclei were

counter-stained with DAPI. Pictures were taken on a Leica

AF6000 LX imaging system, with a DFC350 FX digital camera.

For western blotting, total cell lysates were boiled at 100�C for

5 min in 1.5· sample buffer (1.5 M Tris pH 6.8; Glycerol 15%;

sodium dodecyl sulfate 3%; b-mercaptoethanol 7.5%; bromophe-

nol blue 0.0375%). Gel electrophoresis and western blotting were

performed using Xcell Surelock system (Invitrogen) using pre-cast

gradient gels (4–20%) according to the manufacturer’s instruc-

tions. The gels were blotted onto polyvinylidene difluoride

membranes, which were then blocked for 1 h at 20�C with 5%

(w/v) non-fat dried milk in Tris-buffered saline with 0.1% Tween

20. The membranes were then incubated at 4�C overnight with

the primary antibodies diluted in blocking solution: 2-Cys Prx

(1 : 500, Abcam, Cambridge, UK), Prx-SO2/3H (1 : 1000, Ab-

cam), Sulfiredoxin (P16, 1 : 250, Santa Cruz), b-tubulin isotype

III (1 : 125 000, Sigma). For visualisation of western blots,

horseradish peroxidase-based secondary antibodies were used

followed by chemiluminescent detection on Kodak X-Omat film.

Western blots were analysed by digitally scanning the blots,

followed by densitometric analysis (ImageJ, National Institutes of

Health, Bethesda, MD, USA). All analyses involved normalizing

to a loading control (b-tubulin or Prx).

RNA isolation and qPCRRNA was isolated using the Qiagen RNeasy kit (Qiagen, Valencia,

CA, USA) (including DNAse treatment) following disruption of cells

(QiaShredder column). cDNA was synthesized from 1–3 lg RNA

using the Stratascript QPCR cDNA Synthesis kit. Dilutions of this

cDNA were used for real-time PCR [cDNA equivalent to 6 ng of

initial RNA per reaction for Srxn1; 3 ng equivalent for Glyceralde-

hyde-3-phosphate dehydrogenase (GAPDH)]. qPCR was performed

in anMx3000P QPCR System (Stratagene, La Jolla, CA, USA) using

Brilliant SYBR Green QPCR Master Mix. No-template and no-RT

negative controls were included. Primers used: Srxn1–F: 5¢-GAC-GTCCTCTGGATCAAAG-3¢ 200 nM, -R: 5¢-GCAGGAATGGTCT-CTCTCTG-3¢ 200 nM; GAPDH-F: 5¢-GGGTGTGAACCACGAGAAAT-3¢ 200 nM, -R: 5¢-CCTTCCACAATGCCAAAGTT-3¢100 nM. 18s rRNA-F: 5¢-GTGGAGCGATTTGTCTGGTT-3¢, -R:5¢-CAAGCTTATGACCCGCACTT-3¢. Sesn2 F: 5¢-GGATTATACC-TGGGAAGACC-3¢ 200 nM, -R: 5¢-CGCAGTGGATGTAGTTCC-3¢ 200 nM. Hmox1 F: 5¢-AGCACAGGGTGACAGAAGAG-3¢200 nM, -R: 5¢-GGAGCGGTGTCTGGGATG-3¢. The data were

analysed using theMxProQPCR software (Stratagene). Expression of

the Srxn1 was normalized to GAPDH.

Results

Neuronal Nrf2 expression, and the Nrf2 activator D3T,prevent oxidative neuronal deathNuclear factor erythroid 2-related factor (Nrf2) over-expres-sion in glial cells can confer protection on a large number ofnormal non-overexpressing neurons (Shih et al. 2003; Kraftet al. 2004). It is not clear whether Nrf2 can act solely inneurons to confer neuroprotection against oxidative insults.We analyzed the neuroprotective effect of boosting Nrf2activity specifically in neurons, using a neuron-specifictransfection protocol previously employed (Papadia et al.2008) and briefly described below. Cortical ‘neuronal’cultures naturally contain a small percentage of glial cells(5–10%). Transfection of these cultures results in tinynumbers of transfected glial cells, fewer than the 5–10%expected if both cell types were equally amenable totransfection (Papadia et al. 2008). To quantify this, mixedcultures were transfected with eGFP and subjected to NeuNimmunofluorescence. Of 189 eGFP-expressing cells, 186were NeuN-positive. We then transfected cultures again witheGFP but this time stained for GFAP. Of 271 eGFP-

� 2008 The AuthorsJournal Compilation � 2008 International Society for Neurochemistry, J. Neurochem. (2008) 107, 533–543

Nrf2 prevents peroxiredoxin hyperoxidation | 535

Page 5: Edinburgh Research Explorer · Key among these is the thioredoxin-peroxiredoxin system which is an important reducer of oxidative stressors such as peroxides (Winyard et al. 2005)

Journal Compilation � 2008 International Society for Neurochemistry, J. Neurochem. (2008) 107, 533–543� 2008 The Authors

536 | F. X. Soriano et al.

Page 6: Edinburgh Research Explorer · Key among these is the thioredoxin-peroxiredoxin system which is an important reducer of oxidative stressors such as peroxides (Winyard et al. 2005)

expressing cells, 0 (zero) were GFAP-positive (see examplepictures in Fig. 1a). Thus, nearly all transfected cells inneuronal cultures are neurons, potentially due the fact thatquiescent (AraC-treated) glial cells are not amenable totransfection (NB. Non-quiescent glial cells are amenable totransfection-see below).

We transfected the Nrf2-expressing constructs into neu-rons, along with an eGFP co-transfection marker to enable usto monitor the fate of the neurons [a technique previouslydescribed (Papadia et al. 2008)]. Pictures of eGFP-express-ing neurons were taken 24 h post-transfection, after whichneurons were treated with an oxidative insult (100 lMH2O2). After a further 24 h pictures of the same cells weretaken, before the cultures were fixed and nuclear integrityassessed by DAPI staining. By monitoring the neuronsbefore and after H2O2 treatment, we found that neuronsexpressing Nrf2 were completely resistant to cell deathfollowing H2O2 treatment (100 lM, Fig. 1b, see examplepictures in Fig. 1d). We also studied the fate of neuronswithin a 150 lm radius of Nrf2-transfected neurons andfound that susceptibility of neurons to death by H2O2

treatment was not changed by being in close proximity to aNrf2-expressing neuron (Fig. 1c), in contrast to the reportednon-autonomous effects of Nrf2-expressing glial cells (Shihet al. 2003).

We next investigated the influence of known activators ofNrf2 activity on cortical neuronal vulnerability to anoxidative insult (H2O2) in vitro. We tested D3T, sulforaphaneand tert-butylhydroquinone (tBHQ). The drugs were appliedat a wide range of concentrations for 16 h prior to theapplication of H2O2 (for further 24 h). The protection thatwas achieved by these inducing agents varied: D3T conferredsignificant and strong neuroprotection (Fig. 1e), whereastBHQ and sulforaphane were significantly less potent at therange of doses tested (data not shown). Further studies usingD3T revealed that increased neuroprotection at a lower dose

(10 lM) could be achieved by applying a second identicaldose of the compound immediately prior to the oxidativeinsult (Fig. 1e).

D3T prevents the thioredoxin-peroxiredoxin system frombecoming overwhelmed by oxidative stressThe existence of hyperoxidized Prx-SO2/3H in neuronsindicates an overwhelmed thioredoxin-peroxiredoxin sys-tem, and its formation is associated with oxidative neuronaldeath (Papadia et al. 2008). We therefore investigatedwhether the neuroprotective effects of D3T were associatedwith changes in Prx-SO2/3H levels. Western analysis using aPrx-SO2/3H-specific antibody revealed that H2O2 caused Prxhyperoxidation in vehicle (control)-treated neurons, whileD3T-treated neurons displayed far less hyperoxidation(Fig. 1f). The strong Prx-SO2/3H band represents hyperox-idized PrxII (Papadia et al. 2008), although a higherexposure of the blot reveals hyperoxidation of the upperband identified as Prx III (Papadia et al. 2008) which alsoappears to be weaker in D3T-treated neurons (Fig. 1f).Therefore, D3T pre-treatment renders neurons resistant tooxidative stress and inhibits the appearance of hyperoxi-dized Prx protein.

Sulfiredoxin expression is induced in glia and neurons byactivators of Nrf2Because D3T-treated neurons displayed lower levels of Prx-SO2/3H in response to H2O2 treatment (Fig. 1f), we studiedthe transcriptional regulation of the two genes whoseproducts mediate reduction of Prx-SO2/3H: sulfiredoxin[Srxn1, (Rhee et al. 2007)] and sestrin2 [Sesn2 (Budanovet al. 2004)]. We found that D3T induced transcription ofSrxn1, producing an increase at both the mRNA and proteinlevel in cortical neurons (Fig. 2a and b). In contrast, D3T didnot induce expression of Sesn2. Other Nrf2 activators (tBHQand sulforaphane) also promoted expression of Srxn1 but not

Fig. 1 Neuronal Nrf2 expression and the Nrf2 activator D3T protects

neurons against oxidative stress; D3T inhibits the formation of

inactivated, hyperoxidized peroxiredoxins following oxidative trauma.

(a) Upper: example of an e-GFP expressing cell immuno-positive for

NeuN. Lower: example of an e-GFP expressing cell immuno-negative

for GFAP. Scale bar = 30 lm. (b) Effect of expressing Nrf2 in cortical

neurons on H2O2-induced neuronal death. The neurons were

transfected with control vector or pNrf2, plus an eGFP expression

vector to monitor cell fate (see methods). Twenty four hours post-

transfection neurons were treated where indicated with H2O2 (100 lM

here and throughout the study) and cell fate monitored after a further

24 h. For each vector/treatment, approx. One hundred and fifty cells

were studied across three independent experiments.*p < 0.05 Bon-

feronni two-tailed paired t-test. (c) Effect of expressing Nrf2 in cortical

neurons on survival of nearby cells in response to H2O2-treatment.

Survival/death of neurons within a 150 lm radius of Nrf2-expressing

neurons was analyzed by assessing nuclear morphology of DAPI

stained fixed cells. As can be seen in (d), healthy neurons have large

nuclei with diffuse DAPI staining. Apoptotic nuclei are characterised by

pyknotic brightly stained clumps of condensed chromatin. Survival/

death of over 1000 cells was scored across three independent

experiments. *p < 0.05 Bonferonni two-tailed paired t-test (n = 3). (d)

Example pictures relating to the data shown in (b). Arrows point to the

transfected cells identified by co-expression of eGFP. Pictures before

and after H2O2 treatment are taken of the cell, and DAPI stained

images are also taken post-treatment. Scale bar = 30 lm (e) Cell

death because of 24 h H2O2 insult in the face of the indicated

treatments. (Lower) Examples pictures, scale bar = 25 lm. *p < 0.05

Bonferonni two-tailed paired t-test (compared to control H2O2 treated,

n = 4), mean ± SEM shown in this and all cases. (f) Western analysis

of Prx hyperoxidation using an anti-PrxSO2/3H specific antibody. Two

exposures are shown for optimal visibility of hyperoxidized Prx II and

Prx III respectively. Analysis of PrxSO2/3H levels is restricted to Prx II,

and is normalized to total Prx II expression. *p < 0.05 Bonferonni

two-tailed paired t-test (compared to control, H2O2-treated neurons,

n = 7).

� 2008 The AuthorsJournal Compilation � 2008 International Society for Neurochemistry, J. Neurochem. (2008) 107, 533–543

Nrf2 prevents peroxiredoxin hyperoxidation | 537

Page 7: Edinburgh Research Explorer · Key among these is the thioredoxin-peroxiredoxin system which is an important reducer of oxidative stressors such as peroxides (Winyard et al. 2005)

Sesn2 (Fig. 2a) strongly suggesting that Srxn1 is a Nrf2target gene.

Since our cortical neuronal cultures contain 5–10% of glialcells (Papadia et al. 2008) we wanted to know whether theseNrf2 activators are promoting Srxn1 expression in neurons orglia or both. We performed experiments comparing Srxn1expression in pure glial cultures (> 98% GFAP-positive,Fig. 2c) with that of our mixed cultures. Expression of Srxn1within the neuronal cultures was not significantly different tothat in glial cultures (normalizing to 18S rRNA levels, data notshown). Therefore, in our neuronal cultures, the 5–10%of glialcells by number represent a similar percentage of total Srxn1mRNA. We found that D3T induced Srxn1 expression in pure

glial cultures at 4 h by around 6-fold (Fig. 2d), similar to thelevel of induction of Hmox1 (a known Nrf2-regulated gene,Fig. 2d). Assuming that the glial cells within the neuronalcultures respond similarly, induction of Srxn1 in glial cellsalone cannot account for the 3.5-fold induction of Srxn1 byD3T in the neuronal cultures. The 3.5-fold induction observedcan only be explained if D3T can induce Srxn1 in the neuronsas well as glial cells (possibly by around 3.2-fold).

Nrf2 directly regulates Srxn1 expression via a cis-actingAREWe next investigated whether Srxn1 is directly regulated byNrf2. Nrf2 acts by binding the cis-acting antioxidant responseelement (ARE) in the promoters of target genes.Analysis of therat Srxn1 promoter revealed a sequence at –188 relative to thetranscription start site (TCACCCTGAGTCAGCG) whichresembled an ARE (Nioi et al. 2003), and was conserved inthe gene from various mammalian species (Fig. 3a). To testwhether this putative element is functional we created aluciferase reporter of the Srxn1 promoter containing 585 nt of5¢ promoter sequence (from –577 to +8). We then analysed theeffect of Nrf2 expression on activity of this reporter. We firstused primary cortical neuronal cultures and found that drivenNrf2 expression induced Srxn1-Luc reporter gene activity(Fig. 3b). Furthermore, mutation of the core of the putativeARE (to TCACCCTGGGCCCGCG, Srxn1(mut)-Luc) abol-ished Nrf2-responsiveness (Fig. 3b). The capacity of Nrf2 toinduce Srxn1-Luc was similar to its capacity to induce anartificial ARE-Luc reporter (Numazawa et al. 2003), gener-ated by placing AREs from the Heme oxygenase-1 (Hmox1)promoter upstream of the luciferase open reading frame(Fig. 3c). This indicates that the Srxn1 putative ARE is a bonafide Nrf2-responsive element, and is similarly responsive toNrf2 as the Hmox1 AREs.

To test whether endogenous Srxn1 was also induced byNrf2, we over-expressed Nrf2 in neurons and performedimmunocytochemistry with an appropriate antibody. Thetransfected neurons exhibited elevated levels of Srxn1(Fig. 3d), confirming that the endogenous gene is alsoNrf2-responsive. We next investigated whether the Srxn1ARE was responsive to Nrf2 in other cell types, since Srxn1expression is induced in glial cells by D3T as well as neurons(Fig. 2). Nrf2 induced Srxn1 promoter activity in glial cells(Fig. 3e) to a similar degree as it activated ARE-Luc(Fig. 3f). We also observed Nrf2-responsiveness of theSrxn1 ARE in HEK293 cells (Fig. 3g) confirming the ARE’sfunctionality in several cell types.

Nrf2 controls the basal expression of Srxn1 in glia but notin neuronsTo determine whether Nrf2 controls basal levels of Srxn1expression in neurons and glia, we first looked at the relativebasal activity of Srxn1-Luc and Srxn1(mut)-Luc in bothneurons and glia (Fig. 3b and f). Mutation of the ARE

Fig. 2 Chemopreventive inducers of endogenous Nrf2 induce Srxn1

expression in neurons and glia. (a) Neuronal cultures were treated with

D3T (25 lM), tBHQ (10 lM, Aldrich, Milwaukee, WI, USA) or sulfor-

aphane (5 lM, Merck Biosciences, Darmstadt, Germany) for 4 h

followed by RNA extraction and q-RT-PCR analysis of Srxn1 and

Sesn2 (normalized to GAPDH). *p < 0.05 (Bonferonni two-tailed t-test,

n = 3–5). (b) Western analysis of Srxn1 protein expression in extracts

taken from neuronal cultures treated for 24 h with D3T (25 lM).

*p < 0.05 (n = 6). (c) GFAP immunofluorescence of glial cultures.

(d) Glial cultures were treated with D3T (25 lM) for 4 h followed by

RNA extraction and q-RT-PCR analysis of Srxn1, Sesn2 and Hmox1

(normalized to GAPDH, *p < 0.05, two-tailed t-test in this and sub-

sequent experiments unless otherwise stated, n = 6).

Journal Compilation � 2008 International Society for Neurochemistry, J. Neurochem. (2008) 107, 533–543� 2008 The Authors

538 | F. X. Soriano et al.

Page 8: Edinburgh Research Explorer · Key among these is the thioredoxin-peroxiredoxin system which is an important reducer of oxidative stressors such as peroxides (Winyard et al. 2005)

sequence within the construct had only a modest effect onbasal Srxn1 promoter activity in neurons (Fig. 3b) butreduced basal promoter activity in glia by around 80%(Fig. 3f), suggesting that glial Srxn1 mRNA levels rely onNrf2 activity. However, the Srxn1 ARE contains an embed-ded AP-1 like sequence, so basal activity could be because ofAP-1 activity rather than Nrf2. To investigate this further, weexamined the effect of suppressing basal Nrf2 activity byexpressing Keap1, an endogenous repressor of Nrf2 whichtargets Nrf2 for proteosomal degradation (McMahon et al.2003; Zhang 2006). Keap1 over-expression suppressed basalARE-Luc expression in glial cells but not in neurons (Fig. 4a

and b). In both glia and neurons, Keap1 impaired AREactivity induced by Nrf2 expression (Fig. 4a and b),confirming that Keap1 was functioning properly in neurons.These experiments indicate that basal Nrf2 activity isrelatively low in neurons compared to glia consistent withother studies (Shih et al. 2003). Analysis of Srxn1 reporteractivity revealed that Keap1 effectively suppressed basalglial Srxn1 expression but failed to affect neuronal basalSrxn1 reporter activity (Fig. 4c and d), consistent withnormal constitutive levels of Nrf2 activity in neurons beinglow. This indicates that factors other than Nrf2 are respon-sible for maintaining basal levels of sulfiredoxin in neurons.

Fig. 3 The Srxn1 promoter contains an ARE which is induced by Nrf2

expression in cortical neurons, as is the endogenous gene. (a)

Schematic showing the putative ARE within mammalian Srxn1

promoters. (b) Effect of Nrf2 expression on wild-type and mutant

Srxn1-Luc reporters (normalized to Renilla control-see methods).

Neurons were transfected with Srxn1 reporter plus pcDNA3.1-Nrf2 or

pGlobin control plasmid. NB. In this and all experiments ‘con’ denotes

pGlobin control vector. *p < 0.05 (2-tailed paired t-test, n = 3–5). (c)

Effect of Nrf2 expression on ARE-Luc activity. *p < 0.05 (n = 5). (d)

Effect of transfecting pEF-Nrf2 on endogenous Srxn1 expression.

Neurons were transfected with the indicated vectors and after 24 h

subjected to immunocytochemical analysis of sulfiredoxin expression.

Immunofluorescence performed as described (McKenzie et al. 2006).

Scale bar = 40 lm. (e) Effect of Nrf2 expression on WT and mutant

Srxn1-Luc reporters in glial cells *p < 0.05 (n = 7). (f) Effect of Nrf2

expression on ARE-Luc activity in glial cells. *p < 0.05 (n = 4).

(g) Effect of Nrf2 expression on WT and mutant Srxn1-Luc reporters in

HEK293cells. *p < 0.05 (n = 3).

� 2008 The AuthorsJournal Compilation � 2008 International Society for Neurochemistry, J. Neurochem. (2008) 107, 533–543

Nrf2 prevents peroxiredoxin hyperoxidation | 539

Page 9: Edinburgh Research Explorer · Key among these is the thioredoxin-peroxiredoxin system which is an important reducer of oxidative stressors such as peroxides (Winyard et al. 2005)

Activity-dependent induction of Srxn1 is largelyindependent of Nrf2We reported previously that synaptic activity stronglyinduces Srxn1 via the two AP-1 sites at –188 and –239(Papadia et al. 2008). The proximal AP-1 site identified inthis earlier study is in fact contained within the core of theARE identified here. This raises the possibility that some ofthe activity-dependent induction of Srxn1 is actually due toNrf2 activation. We used an established method of networkdisinhibition to enhance synaptic activity, applying theGABAA receptor antagonist bicuculline to induce bursting,and the K+ channel antagonist 4-aminopyridine [whichenhances burst frequency, hereafter BiC/4-AP (Hardinghamet al. 2001)]. We analyzed the effect of Keap1 on activity-dependent induction of the Srxn1 promoter and found a smallbut significant reduction after 8 h stimulation (Fig. 5a), butnot at 24 h stimulation [the time-point used in our previousstudy (Papadia et al. 2008)]. By comparison we found thatinduction of a reporter of Sesn2, which is also induced by

synaptic activity, was not inhibited by Keap1 at either time-point (Fig. 5a). To test this further, we investigated thedegree to which activity-dependent induction of the com-posite ARE/AP-1 site is dependent on AP-1. We utilizedexpression of TAM67, a dominant negative form of c-junwhich interferes with AP-1 mediated gene expression.Activity-dependent induction of a reporter containing fivetandem AP-1 sites is inhibited by 65% by TAM67 (Fig. 5b).Induction of the mutant Srxn1 reporter dependent on thedistal AP-1 site (i.e. with the composite ARE/AP-1 sitemutated) was inhibited by 55% (Fig. 5b), similar to inhibi-tion of a pure AP-1 reporter. In contrast, induction of themutant Srxn1 reporter dependent on the composite ARE/AP-1 site (i.e. with the distal AP-1 site mutated) wasinhibited by only 30% (Fig. 5b). Thus, our data indicate thatat early time-points the activity-dependent induction of thecomposite ARE/AP-1 site is partly dependent on AP-1, andis also partly dependent on Nrf2.

Fig. 4 Basal levels of Srxn1 promoter activity are sensitive to Keap1

expression in glia but not neurons. (a,b) Effect of Keap1 expression on

basal and Nrf2-driven ARE-Luc reporter activity in neurons (a) and glia

(b). Cells were transfected with ARE-Luc plus pcDNA3.1-Nrf2 (or

pGlobin), plus pcDNA3.1-Keap1 (or pGlobin). *p < 0.05 (n = 4–6).

(c,d) Effect of Keap1 expression on basal Srxn1-Luc reporter activity in

neurons (c) and glia (d). *p < 0.05 (n = 4).

Fig. 5 Activity-dependent induction of Srxn1 via the ARE/AP-1 site is

primarily mediated by AP-1. (a) Effect of Keap1 expression on activity-

dependent induction of reporters of Srxn1 and Sesn2. Neurons were

transfected with the reporter plus TK-driven Renilla control vector.

Twenty four hours post-transfection, bursts of action potential firing

were induced by treatment of neurons with 50 lM bicuculline, and

burst frequency was enhanced by addition of 250 lM 4-amino pyridine

(Hardingham et al. 2001). Reporter activity was assayed after the

indicated time periods. *p < 0.05 Bonferonni two-tailed paired t-test

(compared to control, BiC/4-AP-stimulated level, n = 6). (b) Effect of

an interfering mutant of AP-1 (TAM67) on activity-dependent induction

of the putative ARE/AP-1 composite site, compared to an upstream

AP-1 site. Neurons were transfected with the indicated reporter con-

structs, plus pTK-RL and either control vector or expression vector for

TAM67. Twenty four hours post-transfection neurons were stimulated

with BiC/4-AP overnight prior to assay of reporter activity (n = 5).

Journal Compilation � 2008 International Society for Neurochemistry, J. Neurochem. (2008) 107, 533–543� 2008 The Authors

540 | F. X. Soriano et al.

Page 10: Edinburgh Research Explorer · Key among these is the thioredoxin-peroxiredoxin system which is an important reducer of oxidative stressors such as peroxides (Winyard et al. 2005)

Discussion

This study provides evidence that activation of the Nrf2pathway specifically within neurons can antagonize H2O2-stimulated apoptosis. We also show that activation of thispathway can prevent hyperoxidation of peroxiredoxins thatoccurs in neurons upon exposure to an oxidative insult. Alsowe have found that expression of sulfiredoxin, an enzymethat reduces hyperoxidized peroxiredoxins, is regulated byNrf2 in neurons and other cell types. Thus, expression ofsulfiredoxin may contribute to the antioxidative effects ofchemopreventive agents that activate Nrf2.

Reactivation of peroxiredoxins by sulfiredoxinPeroxiredoxins are a family of thiol-based antioxidants thatprotect against damage caused by oxidative stressors. PrxIIprotects cortical neurons against Ab toxicity (Yao et al.2007) and oxygen-glucose deprivation (Boulos et al. 2007).Interfering with PrxII expression renders neuroblastomacells vulnerable to oxidative stress (Sanchez-Font et al.2003), and renders cortical neurons vulnerable to MPP+

(Qu et al. 2007). PrxIII protects hippocampal neuronsagainst excitotoxicity (Hattori et al. 2003). Thus, molecularevents that render Prxs inactive in vivo in pathologicalscenarios may contribute to disease progression. We havepreviously observed Prx-inactivating Prx-SO2/3H formationfollowing cerebral ischemia in vivo (Papadia et al. 2008). Itremains to be seen whether Prx hyperoxidation occurs inmore chronic neurodegenerative diseases associated withoxidative stress.

Sulfiredoxin was initially characterized in yeast (Biteauet al. 2003) and then in mammalian cells (Rhee et al. 2007).It acts by catalysing the ATP-dependent formation of asulfinic acid phosphoric ester on Prx (Rhee et al. 2007)which is then reduced by thiol equivalents such as thiore-doxin. Interfering with Srxn1 expression impairs the abilityof cells to reduce hyperoxidized Prxs, while over-expressionof Srxn1 enhances it (Chang et al. 2004; Woo et al. 2005).Our finding that Srxn1 expression can be induced by smallthiol-active molecules that activate Nrf2 suggests thatexposure to such inducers may increase the level ofperoxides that a cell can tolerate before Prx is inactivatedby hyperoxidation. Given the antioxidant and cytoprotectiveeffects of Prxs, induction of sulfiredoxin expression may be asignificant contributor to the net effects of Nrf2 activation. Itwill be of considerable interest to see whether Nrf2activators are able to reduce Prx-SO2/3H formation followingcerebral ischemia in vivo. This seems possible, since pre-administration of the Nrf2 activators tBHQ and sulforaphanecan protect the brain from cerebral ischemia in vivo (Shihet al. 2005; Zhao et al. 2006). According to our observa-tions, D3T is a superior neuroprotectant, so it will be ofinterest to know whether it is an efficient protector againstischemic injury.

Nrf2 is a therapeutic target for many disordersUp-regulation of Nrf2 activity in Drosphila increases lifespanand resistance to oxidative stress (Sykiotis and Bohmann2008). In mammals, gene expression programs induced byNrf2 can protect many different organs against a variety oftraumas. For example, Nrf2 protects lung against hyperoxicinjury (Cho et al. 2002), and the liver against paracetamol-induced hepatotoxicity (Enomoto et al. 2001). Nrf2 is theprimary molecular target of cancer chemopreventive block-ing agents (Giudice and Montella 2006) which in general actby preventing carcinogens from forming adducts with DNAthat lead to mutations. The boosting of intrinsic antioxidantdefences of the cell by activation of Nrf2 is believed to be animportant mediator of these protective effects, as is the co-ordinated induction of detoxification enzymes such asglutathione S-transferases and NAD(P)H:quinone oxidore-ductase 1. The program of gene expression induced by Nrf2is well-placed to combat the actions and the production of avariety of free radicals. For example, the Nrf2 target geneFerritin sequesters Fe(II) and can thus restrict hydroxylradical-generating Fenton chemistry. Also, the Nrf2 targetgene Hmox1 degrades the pro-oxidant heme molecule,generating bilirubin as a breakdown product. Bilirubin canreact directly with, and neutralize, superoxide, hydroxyl andperoxynitrite radicals (Stocker 2004). The actions of these,and other, gene products, in concert with those involved inthe thiol-based antioxidant systems result in a powerful andgeneral up-regulation of antioxidant defenses (Lee et al.2005).

In the CNS, Nrf2 has been suggested to be a therapeutictarget in excitotoxic disorders such as stroke and seizure, aswell as neurodegenerative diseases (Lee et al. 2005; Shihet al. 2005). While Nrf2 over-expressing glial cells stronglyprotect surrounding untransfected neurons (Shih et al. 2003;Kraft et al. 2004; Jakel et al. 2007), it was not clear whetherNrf2 can act solely in neurons to confer neuroprotectionagainst oxidative insults. The electrophile NEPP11 inducesNrf2 preferentially but not exclusively in neurons (Satohet al. 2006) and protects against models of NMDA receptor-dependent excitotoxicity. This suggested that Nrf2 can act inneurons, but contributions from glial cells could not be ruledout. Our results show that Nrf2 is strongly neuroprotective inneurons and that neuroprotection need not rely on glial Nrf2expression.

The neuroprotective effect of Nrf2 places the many knownactivators of Nrf2 as potential lead compounds. We havefound that the ability of different agonists of Nrf2 to induceSrxn1 and Hmox1 expression in neurons is not matched bytheir neuroprotective effects. In our hands, D3T is afar superior neuroprotectant than sulforaphane or tBHQ(G. E. Hardingham, unpublished data). Oltipraz, a struc-tural relative of D3T, has also been found to be protective(G. E. Hardingham, unpublished data). We hypothesize thatin our particular system, tBHQ and sulforaphane induce Nrf2

� 2008 The AuthorsJournal Compilation � 2008 International Society for Neurochemistry, J. Neurochem. (2008) 107, 533–543

Nrf2 prevents peroxiredoxin hyperoxidation | 541

Page 11: Edinburgh Research Explorer · Key among these is the thioredoxin-peroxiredoxin system which is an important reducer of oxidative stressors such as peroxides (Winyard et al. 2005)

target genes only at a dose that may exert significant toxicity,hence their relative lack of efficacy. This toxicity is likelybecause of unspecified off-target effects. Thus, while activa-tors of Nrf2 remain attractive candidates for acute andchronic neurodegenerative disorders associated with oxida-tive stress, the therapeutic window of each molecule is ofparamount importance with regard to its direct efficacy, evenbefore pharmacodynamic and pharmacokinetic factors areconsidered.

The AP-1/ARE composite site in the Srxn1 promoter canintegrate multiple signalsOur observations here and elsewhere (Papadia et al. 2008)identify Srxn1 as a gene regulated by both AP-1 (via twophorbol 12-O-tetradecanoate 13-acetate-response elementsites) and Nrf2 (one ARE site). Moreover, the proximalAP-1 site is contained within the ARE sequence. SeveralAREs such as those in the promoters of human NQO1 andHMOX1 contain AP-1 like sequences and can respond to AP-1-activating stimuli, as well as Nrf2 (Nguyen et al. 2004).The composite ARE/AP-1 site on the Srxn1 promoterenables it to respond both to small molecule Nrf2 inducerssuch as D3T and tBHQ, as well as to synaptic activity(mainly via AP-1). Induction of Srxn1 by synaptic activitycontributes to the protection of cortical neurons by synapticactivity in the face of an oxidative insult (Papadia et al.2008). It is an intriguing possibility that the protective effectsof synaptic activity and Nrf2 inducers may have a partiallyoverlapping mechanism: induction of certain genes contai-ning ARE/AP-1 composite sites.

Acknowledgements

This work was supported by the Royal Society, the Wellcome Trust

and the Biotechnology and Biological Sciences Research Council

(BBSRC). We sincerely thank Drs Mike McMahon, Satoshi

Numazawa and Jawed Alam for providing plasmids.

References

Biteau B., Labarre J. and Toledano M. B. (2003) ATP-dependentreduction of cysteine-sulphinic acid by S. cerevisiae sulphiredoxin.Nature 425, 980–984.

Boulos S., Meloni B. P., Arthur P. G., Bojarski C. and Knuckey N. W.(2007) Peroxiredoxin 2 overexpression protects cortical neuronalcultures from ischemic and oxidative injury but not glutamateexcitotoxicity, whereas Cu/Zn superoxide dismutase 1 overex-pression protects only against oxidative injury. J. Neurosci. Res.85, 3089–3097.

Budanov A. V., Sablina A. A., Feinstein E., Koonin E. V. and ChumakovP. M. (2004) Regeneration of peroxiredoxins by p53-regulatedsestrins, homologs of bacterial AhpD. Science 304, 596–600.

Chang T. S., Jeong W., Woo H. A., Lee S. M., Park S. and Rhee S. G.(2004) Characterization of mammalian sulfiredoxin and its reacti-vation of hyperoxidized peroxiredoxin through reduction of cys-teine sulfinic acid in the active site to cysteine. J. Biol. Chem. 279,50994–51001.

Cho H. Y., Jedlicka A. E., Reddy S. P., Kensler T. W., Yamamoto M.,Zhang L. Y. and Kleeberger S. R. (2002) Role of NRF2 in pro-tection against hyperoxic lung injury in mice. Am. J. Respir. CellMol. Biol. 26, 175–182.

Enomoto A., Itoh K., Nagayoshi E., Haruta J., Kimura T., O’Connor T.,Harada T. and Yamamoto M. (2001) High sensitivity of Nrf2knockout mice to acetaminophen hepatotoxicity associated withdecreased expression of ARE-regulated drug metabolizingenzymes and antioxidant genes. Toxicol. Sci. 59, 169–177.

Fang J., Nakamura T., Cho D. H., Gu Z. and Lipton S. A. (2007)S-nitrosylation of peroxiredoxin 2 promotes oxidative stress-induced neuronal cell death in Parkinson’s disease. Proc. NatlAcad. Sci. USA 104, 18742–18747.

Giudice A. and Montella M. (2006) Activation of the Nrf2-ARE sig-naling pathway: a promising strategy in cancer prevention.Bioessays 28, 169–181.

Hardingham G. E., Arnold F. J. and Bading H. (2001) Nuclear calciumsignaling controls CREB-mediated gene expression triggered bysynaptic activity. Nat. Neurosci. 4, 261–267.

Hardingham G. E., Fukunaga Y. and Bading H. (2002) ExtrasynapticNMDARs oppose synaptic NMDARs by triggering CREB shut-offand cell death pathways. Nat. Neurosci. 5, 405–414.

Hattori F., Murayama N., Noshita T. and Oikawa S. (2003) Mitochon-drial peroxiredoxin-3 protects hippocampal neurons from excito-toxic injury in vivo. J. Neurochem. 86, 860–868.

Immenschuh S. and Baumgart-Vogt E. (2005) Peroxiredoxins, oxida-tive stress, and cell proliferation. Antioxid. Redox Signal. 7, 768–777.

Jakel R. J., Townsend J. A., Kraft A. D. and Johnson J. A. (2007) Nrf2-mediated protection against 6-hydroxydopamine. Brain Res. 1144,192–201.

Jonsson T. J., Johnson L. C. and Lowther W. T. (2008) Structure of thesulphiredoxin-peroxiredoxin complex reveals an essential repairembrace. Nature 451, 98–101.

Kotkow K. J. and Orkin S. H. (1995) Dependence of globin geneexpression in mouse erythroleukemia cells on the NF-E2 hetero-dimer. Mol. Cell. Biol. 15, 4640–4647.

Kraft A. D., Johnson D. A. and Johnson J. A. (2004) Nuclear factor E2-related factor 2-dependent antioxidant response element activationby tert-butylhydroquinone and sulforaphane occurring preferen-tially in astrocytes conditions neurons against oxidative insult.J. Neurosci. 24, 1101–1112.

Lee J. M., Li J., Johnson D. A., Stein T. D., Kraft A. D., Calkins M. J.,Jakel R. J. and Johnson J. A. (2005) Nrf2, a multi-organ protector?FASEB J. 19, 1061–1066.

Mckenzie G. J., Stephenson P., Ward G., Papadia S., Bading H., ChawlaS., Privalsky M. and Hardingham G. E. (2005) Nuclear Ca2+ andCaM kinase IV specify hormonal- and Notch-responsiveness.J. Neurochem. 93, 171–185.

McKenzie G., Ward G., Stallwood Y., Briend E., Papadia S., Lennard A.,Turner M., Champion B. and Hardingham G. E. (2006) CellularNotch responsiveness is defined by phosphoinositide 3-kinase-dependent signals. BMC Cell Biol. 7, 10.

McMahon M., Itoh K., Yamamoto M. and Hayes J. D. (2003) Keap1-dependent proteasomal degradation of transcription factor Nrf2contributes to the negative regulation of antioxidant responseelement-driven gene expression. J. Biol. Chem. 278, 21592–21600.

Nguyen T., Yang C. S. and Pickett C. B. (2004) The pathways andmolecular mechanisms regulating Nrf2 activation in response tochemical stress. Free Radic. Biol. Med. 37, 433–441.

Nioi P., McMahon M., Itoh K., Yamamoto M. and Hayes J. D. (2003)Identification of a novel Nrf2-regulated antioxidant response ele-ment (ARE) in the mouse NAD(P)H:quinone oxidoreductase 1

Journal Compilation � 2008 International Society for Neurochemistry, J. Neurochem. (2008) 107, 533–543� 2008 The Authors

542 | F. X. Soriano et al.

Page 12: Edinburgh Research Explorer · Key among these is the thioredoxin-peroxiredoxin system which is an important reducer of oxidative stressors such as peroxides (Winyard et al. 2005)

gene: reassessment of the ARE consensus sequence. Biochem. J.374, 337–348.

Numazawa S., Ishikawa M., Yoshida A., Tanaka S. and Yoshida T.(2003) Atypical protein kinase C mediates activation of NF-E2-related factor 2 in response to oxidative stress. Am. J. Physiol. CellPhysiol. 285, C334–C342.

Papadia S., Stevenson P., Hardingham N. R., Bading H. and HardinghamG. E. (2005) Nuclear Ca2+ and the cAMP response element-binding protein family mediate a late phase of activity-dependentneuroprotection. J. Neurosci. 25, 4279–4287.

Papadia S., Soriano F. X., Leveille F. et al. (2008) Synaptic NMDAreceptor activity boosts intrinsic antioxidant defenses. Nat. Neu-rosci. 11, 476–487.

Qu D., Rashidian J., Mount M. P. et al. (2007) Role of Cdk5-mediatedphosphorylation of Prx2 in MPTP toxicity and Parkinson’s disease.Neuron 55, 37–52.

Rhee S. G., Jeong W., Chang T. S. and Woo H. A. (2007) Sulfiredoxin,the cysteine sulfinic acid reductase specific to 2-Cys peroxiredoxin:its discovery, mechanism of action, and biological significance.Kidney Int. Suppl., 106, S3–S8.

Sanchez-Font M. F., Sebastia J., Sanfeliu C., Cristofol R., Marfany G.and Gonzalez-Duarte R. (2003) Peroxiredoxin 2 (PRDX2), anantioxidant enzyme, is under-expressed in Down syndrome fetalbrains. Cell. Mol. Life Sci. 60, 1513–1523.

Satoh T., Okamoto S. I., Cui J., Watanabe Y., Furuta K., Suzuki M.,Tohyama K. and Lipton S. A. (2006) Activation of the Keap1/Nrf2pathway for neuroprotection by electrophilic [correction of electro-phillic] phase II inducers. Proc. Natl Acad. Sci. USA 103, 768–773.

ShihA. Y., JohnsonD. A.,WongG., Kraft A. D., Jiang L., ErbH., JohnsonJ. A. and Murphy T. H. (2003) Coordinate regulation of glutathionebiosynthesis and release by Nrf2-expressing glia potently protectsneurons from oxidative stress. J. Neurosci. 23, 3394–3406.

Shih A. Y., Li P. and Murphy T. H. (2005) A small-molecule-inducibleNrf2-mediated antioxidant response provides effective prophylaxisagainst cerebral ischemia in vivo. J. Neurosci. 25, 10321–10335.

Stocker R. (2004) Antioxidant activities of bile pigments. Antioxid.Redox Signal. 6, 841–849.

Sykiotis G. P. and Bohmann D. (2008) Keap1/Nrf2 signaling regulatesoxidative stress tolerance and lifespan in Drosophila. Dev. Cell 14,76–85.

Winyard P. G., Moody C. J. and Jacob C. (2005) Oxidative activation ofantioxidant defence. Trends Biochem. Sci. 30, 453–461.

Woo H. A., Jeong W., Chang T. S., Park K. J., Park S. J., Yang J. S. andRhee S. G. (2005) Reduction of cysteine sulfinic acid by sulfire-doxin is specific to 2-cys peroxiredoxins. J. Biol. Chem. 280,3125–3128.

Wood Z. A., Schroder E., Robin Harris J. and Poole L. B. (2003)Structure, mechanism and regulation of peroxiredoxins. TrendsBiochem. Sci. 28, 32–40.

Yao J., Taylor M., Davey F., Ren Y., Aiton J., Coote P., Fang F., ChenJ. X., Yan S. D. and Gunn-Moore F. J. (2007) Interaction ofamyloid binding alcohol dehydrogenase/Abeta mediates up-regu-lation of peroxiredoxin II in the brains of Alzheimer’s diseasepatients and a transgenic Alzheimer’s disease mouse model. Mol.Cell. Neurosci. 35, 377–382.

Yoshida T., Nakamura H., Masutani H. and Yodoi J. (2005) Theinvolvement of thioredoxin and thioredoxin binding protein-2 oncellular proliferation and aging process. Ann. NY Acad. Sci. 1055,1–12.

Zhang D. D. (2006) Mechanistic studies of the Nrf2-Keap1 signalingpathway. Drug Metab. Rev. 38, 769–789.

Zhao J., Kobori N., Aronowski J. and Dash P. K. (2006) Sulforaphanereduces infarct volume following focal cerebral ischemia inrodents. Neurosci. Lett. 393, 108–112.

� 2008 The AuthorsJournal Compilation � 2008 International Society for Neurochemistry, J. Neurochem. (2008) 107, 533–543

Nrf2 prevents peroxiredoxin hyperoxidation | 543