Skip to main content
Molecular & Cellular Proteomics : MCP logoLink to Molecular & Cellular Proteomics : MCP
. 2009 Jun;8(6):1206–1218. doi: 10.1074/mcp.M800406-MCP200

The Thioredoxin-like Protein Rod-derived Cone Viability Factor (RdCVFL) Interacts with TAU and Inhibits Its Phosphorylation in the Retina*,S⃞

Ram Fridlich , Francois Delalande §, Céline Jaillard , Jun Lu , Laetitia Poidevin , Thérèse Cronin , Ludivine Perrocheau , Géraldine Millet-Puel , Marie-Laure Niepon , Olivier Poch ‡,‖, Arne Holmgren , Alain Van Dorsselaer §, Jose-Alain Sahel , Thierry Léveillard ‡,**
PMCID: PMC2690495  PMID: 19279044

Abstract

Rod-derived cone viability factor (RdCVF) is produced by the Nxnl1 gene that codes for a second polypeptide, RdCVFL, by alternative splicing. Although the role of RdCVF in promoting cone survival has been described, the implication of RdCVFL, a putative thioredoxin enzyme, in the protection of photoreceptors is presently unknown. Using a proteomics approach we identified 90 proteins interacting with RdCVFL including the microtubule-binding protein TAU. We demonstrate that the level of phosphorylation of TAU is increased in the retina of the Nxnl1−/− mice as it is hyperphosphorylated in the brain of patients suffering from Alzheimer disease, presumably in some cases through oxidative stress. Using a cell-based assay, we show that RdCVFL inhibits TAU phosphorylation. In vitro, RdCVFL protects TAU from oxidative damage. Photooxidative stress is implicated in retinal degeneration, particularly in retinitis pigmentosa, where it is considered to be a contributor to secondary cone death. The functional interaction between RdCVFL and TAU described here is the first characterization of the RdCVFL signaling pathway involved in neuronal cell death mediated by oxidative stress.


Oxidative stress is suspected to play a major role in numerous age-related diseases (1) and neurodegenerative disorders (2). High levels of oxygen result in generation of reactive oxygen species that interact with proteins, lipids, or DNA and lead to cell dysfunction and death. In the eye, several diseases including glaucoma (3) and age-related macular degeneration (4) are associated with oxidative stress. Retinal degenerations are a prevalent cause of severe loss of vision. Retinitis pigmentosa is a monogenic disease in which mutations occur in genes that lead to the death of rod photoreceptors resulting in night blindness. After rods die, cone photoreceptors gradually die, resulting in blindness. Recent evidence has implicated oxidative damage as a contributor to death of cones in retinitis pigmentosa and to death of both photoreceptor cell types in age-related macular degeneration (5). After rods die, oxygen consumption in the outer retina is markedly reduced, and tissue oxygen levels become substantially elevated (6). This results in progressive oxidative damage to cones and gradual cone cell death, which can be slowed by antioxidants (7). The antioxidant defenses include vitamins, minerals, and the antioxidant enzymes glutathione, superoxide dismutase, hydrogen peroxidase, catalase, and thioredoxins (8). The protection of thioredoxin 1 (TRX1)1 against retinal photooxidative damage (9) and its role in neurodegenerative diseases like Alzheimer disease are known (10, 11). TRX1, the substrate of thioredoxin reductase, can protect neuronal cells by scavenging free radicals, binding and inhibiting apoptosis signal-regulating kinase 1 (12), regulating transcription factors, and maintaining redox homeostasis (13). Rod-derived cone viability factor (RdCVF) belongs to the family of thioredoxins (14). RdCVF is a trophic factor secreted by rods that promotes cone viability encoded by the nucleoredoxin-like 1 (Nxnl1) gene, also called thioredoxin-like 6. The loss of rods results in a decrease of RdCVF expression, which presumably leads to cone death due to a lack of trophic support (15, 16). The TRX active site containing two cysteines (CXXC) catalyzes the reduction of disulfide bonds in targeted proteins (17). The Nxnl1 gene encodes two products via alternative splicing that contain an active site: a full-length protein (RdCVFL) that might carry a thioredoxin activity (18, 19) and a C-terminally truncated protein (RdCVF) with trophic activity for cones but without any enzymatic activity. This latter form resembles the cytokine TRX80, a form of human TRX1 that has no thioredoxin activity (20). The trophic effect is carried specifically by the short isoform RdCVF (14). The thiol oxidoreductase activity of RdCVFL has to date only been observed in the arthropod Carcinoscorpius (18), and the participation of RdCVFL in the protection of photoreceptors is currently unknown. To clarify the role of RdCVFL in photoreceptor survival, we identified a specific functional interaction between RdCVFL and TAU using a proteomics approach.

TAU protein is a microtubule-associated protein that has a role in assembly and stabilization of microtubules (21). In the brains of patients with Alzheimer disease (AD) TAU was found to be hyperphosphorylated (22), leading to aggregation of the protein and to a decrease in TAU binding to microtubules (23) resulting in cell death. Phosphorylated TAU is also toxic to neuronal cells (24). The interaction between RdCVFL and TAU is the first description of the participation of RdCVFL in the protection of photoreceptors and of a novel redox signaling pathway involved in regulating neuronal cell death.

EXPERIMENTAL PROCEDURES

Animal Models and Retina Extraction—

Care and handling of mice in these studies conformed to the rules set by the Association for Research on Vision and Ophthalmology Resolution. Chicken retinal extracts are described in Fintz et al. (25). The Nxnl1−/− mice were generated on a pure BALB/c background. Retinas were solubilized in Nonidet P-40 lysis buffer (25 mm Hepes, pH 7.8, 1 mm EDTA, 1 mm DTT, 0.05 m KCl, 1% Nonidet P-40, 1 mm PMSF, protease inhibitors) followed by sonication and centrifugation. The soluble fraction was subjected to further analysis. Unless otherwise specified, all chemicals were obtained from Sigma.

GST-RdCVFL Affinity Chromatography—

RdCVFL was cloned into pGEX-2TK plasmid (GH Healthcare, catalogue number 27-4587-01), overexpressed, and purified as described previously (14). GST and GST-RdCVFL were bound to Sepharose 4B beads (GE Healthcare), which were washed and packed in a column. An amount of GST and GST-RdCVFL equivalent in molarity was used. The columns were equilibrated with 25 mm Hepes, pH 7.8, 1 mm EDTA, 1 mm DTT, 0.05 m KCl. Chick retinas (postnatal day 17) were lysed in Nonidet P-40 lysis buffer followed by Polytron disruption and centrifugation. The soluble fraction was diluted to a concentration of 0.5 mg/ml and loaded on the equilibrated columns at a flow of 50 μl/min. After washing, elution was carried out with a KCl gradient (25 mm Hepes, pH 7.8, 1 mm EDTA, 1 mm DTT, 0.05–0.5 m KCl). Four fractions were collected from each column, desalted with Centricon (Amicon), and loaded on a gel followed by silver staining (ProteosilverPlus, Sigma). Lanes (except lane C) were cut out and subjected to MS analysis.

In-gel Digestion—

In-gel digestion was performed with an automated protein digestion system, MassPREP Station (Waters). The gel slices were washed three times in a mixture containing 25 mm NH4HCO3, CH3CN (1:1, v/v). The cysteine residues were reduced by 50 μl of 10 mm DTT at 57 °C and alkylated by 50 μl of 55 mm iodoacetamide. After dehydration with acetonitrile, the proteins were cleaved in gel with 40 μl of 12.5 ng/μl modified porcine trypsin (Promega) in 25 mm NH4HCO3 at 37 °C for 4 h. The tryptic peptides were extracted with 60% acetonitrile in 5% formic acid followed by a second extraction with 100% (v/v) acetonitrile.

Mass Spectrometry Analysis—

The peptides extracted from the gel were directly analyzed by nano-LC-MS/MS. Nano-LC-MS/MS was performed on an Agilent 1100 Series HPLC-Chip/MS system (Agilent Technologies) coupled to an Ultra High-Capacity Trap mass spectrometer system (Bruker Daltonics). The voltage applied to the capillary cap was optimized to −1850 V. For tandem MS experiments, the system was operated with automatic switching between MS and MS/MS modes. The three most abundant peptides, preferring doubly charged ions, were selected on each MS spectrum for further isolation and fragmentation. The MS/MS scanning was performed in the ultrascan resolution mode at a scan rate of 26,000 m/z per second. A total of six scans were averaged to obtain a MS/MS spectrum. The complete system was fully controlled by ChemStation (Agilent Technologies) and EsquireControl (Bruker Daltonics) software. The mass data recorded during nano-LC-MS/MS analyses were processed and converted into .mgf peak list format.

Protein Identification—

The MS and the MS/MS data were searched using a local Mascot server (version, Mascot 2.2.0; Matrix Science). The MS/MS data were analyzed against a composite target-decoy database including the National Center for Biotechnology Information (NCBI) protein sequences of human, Rattus norvegicus, Mus musculus, and Aves downloaded in February 2008 and reversed versions of these sequences (total 1,693,251 entries; see supplemental methods). Searches were performed with a mass tolerance of 250 ppm for MS mode and 0.4 Da in MS/MS mode for nano-LC-MS/MS analysis. One missed cleavage per peptide was allowed, and variable modifications were taken into account such as carbamidomethylation of cysteine, oxidation of methionine, and N-acetyl protein. Searches were performed without constraining protein molecular weight or isoelectric point and without any taxonomic restriction. To minimize false positive identifications, the results were subjected to very stringent filtering criteria. For the identification of a protein with two peptides or more, at least two unique peptides had to have a Mascot ion score above 25. In the case of single peptide hits, the score of the unique peptide must be greater (minimal “difference score” of 12) than the 95% significance Mascot threshold. The spectra of these single peptide hits are provided (supplemental data). For the estimation of the false positive rate, a target-decoy database search was performed (26, 27). In this approach peptides are matched against a database consisting of the native protein sequences found in the database (target) and of the sequence-reversed entries (decoy). The evaluations were performed using the peptide validation software Scaffold (Proteome Software). This strategy was used to obtain a final catalogue of proteins with an estimated false positive rate below 1%.

Bioinformatics—

Protein-protein interactions were obtained from the STRING database (28) containing known and predicted physical and functional protein-protein interactions. STRING in protein mode was used, and only interactions with high confidence levels (>0.7) were kept. Network visualization was done with the Cytoscape software (29). We used the DAVID (Database for Annotation, Visualization and Integrated Discovery) functional annotation tool (30) to identify the enriched gene ontology (GO) terms within the gene lists. We used p values after Benjamini correction and considered only those where the corrected p value was inferior to 10−7.

RNA Purification, Reverse Transcription-PCR, Plasmid Construction—

Cone-enriched cultures were made as described previously (14, 25). Total RNA from chicken cone-enriched cultures was purified by cesium gradient (14) and subjected to RT analysis. PCR was performed with 100 ng of cDNA. Cycling conditions were as follow: 94 °C for 5 min followed by 35 cycles of amplification (94 °C denaturation for 30 s, annealing for 30 s at a temperature range of 52–58 °C, and 72 °C elongation for 1 min) with a final incubation at 72 °C for 3 min. Details of primers are provided in supplemental Table II. The PCR products of human TRX1 and chicken TAU were cloned into the pCMV-HA/DEST vector (a gift from R. Roepman) by Gateway system cloning (BP and LR reactions). pCMV4-GSK-3β and the cDNA of human TAU (383 aa) were kindly provided by C. Gespach and I. Ginzburg, respectively. Chicken genomic DNA was purified using standard methods.

Cell Culture and Transfection—

COS-1 and HEK 293 cells were grown in Dulbecco's modified Eagle's medium with 10% fetal bovine serum. The cells were transfected using the calcium phosphate co-precipitation technique (14). 48 h after transfection, cells were resuspended in lysis buffer (50 mm Tris, pH 7.5, 1 mm EDTA, 1 mm DTT, 50 μg/ml Nα-tosyl-l-lysinyl-chloromethyl ketone, 1% Triton X-100, protease inhibitors), sonicated, and centrifuged, and the supernatant was kept at −80 °C.

Co-immunoprecipitation—

Diluted transfected cell extracts were incubated overnight with different antibodies. Immunocomplexes were immobilized on protein A-Sepharose beads. Beads were washed, and protein complexes were eluted by adding Laemmli buffer.

Western Blotting—

Western blotting is described in Leveillard et al. (14). The antibodies used were as follows: anti-HA (Covance, catalogue number MMS-101R-0500; 1:1000), anti-RdCVF (RdCVF-N, 1:500), anti-Tau5 (Calbiochem, catalogue number 577801; 1:500), anti-AT8 (Autogen Bioclear, catalogue number 90206; 1:100), anti-actin (Chemicon, catalogue number MAB1501; 1:7500), and anti-human TRX1 (IMCO Corp., catalogue number ATRX-03; 1:1000).

Immunohistochemistry—

Nxnl1+/+ and Nxnl1−/− mice aged 10 months were used. Retinas were removed from eyes and fixed in 4% paraformaldehyde in 0.1 m phosphate buffer, pH 7.2, for 2 h. Immunohistochemistry was performed on 5-μm sections obtained from paraffin-embedded tissue blocks. TAU was visualized using anti-AT8 and -Tau5 antibodies. Sections were subjected to microwave irradiation for antigen retrieval. Nonspecific binding sites were blocked by incubation of sections for 1 h at room temperature with normal goat serum in PBS. Sections were incubated overnight at 4 °C with AT8 antibody (1:250) in PBS. Immunostaining was visualized using the avidin-biotin system (Vectastain, Vector Laboratories) and nitro blue tetrazolium/5-bromo-4-chloro-3-indolyl phosphate as the chromogen. After washing, sections were mounted in Vecstatin and imaged with a microscope (Leica).

In Vitro Thiol Oxidoreductase Assay—

GST and GST-RdCVFL were eluted from beads using a buffer containing 10 mm glutathione. GST, GST-RdCVFL (in PBS), and the recombinant human TAU protein samples (Sigma, catalogue number T9392) in 50 mm MOPS, pH 6.8, 100 mm NaCl, 1 mm EDTA, 5 mm DTT, 1 mm PMSF were dialyzed against 50 mm Tris-HCl, pH 7.5, 1 mm EDTA overnight with Dispo-Biodialyzer devices (molecular mass cutoff, 10 kDa) (Sigma). 1 μm TAU protein was mixed with 2 μm GST or GST-RdCVFL. The mixture or TAU protein alone was treated with 200 μm H2O2 for 2 h at room temperature. The oxidized TAU was incubated with the thioredoxin system (500 μm NADPH (Sigma), 100 nm recombinant thioredoxin reductase, 20 μm thioredoxin1 (IMCO Corp., catalogue number TRX-01)) for 1 h. The free cysteines of the proteins were labeled by 100 μm fluorescent 5-(iodoacetamido)fluorescein for 20 min. This labeling was stopped by addition of 50 mm iodoacetamide. The sample loading buffer with or without 50 mm DTT was incubated with the samples at 56 °C for 20 min, and the proteins were separated on a gel. The fluorescent bands were detected under a UV light, and subsequently the protein bands were detected by Coomassie Blue staining.

On-line Supplemental Material—

Supplemental Table I displays the 44 and 121 proteins that were identified as GST- and GST-RdCVFL-interacting proteins, respectively. Supplemental Table II displays the sequence primers used in this study. Supplemental data display the spectra of all the proteins identified with one peptide. Recombinant protein sequences used in the study are provided in supplemental Table III.

RESULTS

RdCVFL Is Expressed in Cone-enriched Cultures—

RdCVF was originally identified using cone-enriched cultures from chicken embryos. Bird retinas are cone-dominated, and in culture, cones represent 70% of the cell population (25). In the mouse retina RdCVF and RdCVFL are encoded by two alternative mRNA transcripts corresponding to exon 1 and to the spliced exons 1 and 2, respectively. In the chicken genome there is no in-frame stop codon immediately 3′ to exon 1 (31). We analyzed the expression of NXNL1 mRNAs by RT-PCR in the cone-enriched culture (Fig. 1A). In these cultures, visinin is selectively expressed by cone cells (25). We detected an amplification product of RdCVFL mRNA but not RdCVF (Fig. 1B). The efficiency of RdCVF primers was validated on genomic DNA. The data demonstrate that RdCVFL is expressed by the chick cone culture.

Fig. 1.

Fig. 1.

RdCVFL is expressed in chicken cone-enriched cultures. A, schematic presentation of the chicken Nxnl1 gene and its products. RdCVFL is composed of two exons (CDS, 624 bp), and RdCVF (predicted) is composed of the first exon and a part of the first intron (CDS, 450 bp). The arrows with numbers correspond to the primers used for RT-PCR. B, RT-PCR analysis on chicken cone cultures. Specific primers were used for the detection of RdCVF (predicted) (lanes 3–5) and RdCVFL (lanes 6–7). For controls visinin primers were used (lanes 1–2) as well as genomic DNA for primers 3 and 4 (lane 5).

Identification of RdCVFL-interacting Proteins—

To identify RdCVFL-interacting proteins, we prepared soluble protein extract from chicken retina at postnatal day 17, a developmental stage rich in mature cones (result not shown). This retinal extract was loaded on GST-RdCVFL or GST columns. After washing, the elution was carried out with a 50–500 mm KCl gradient. The eluted proteins collected in four fractions were loaded on a gel and silver-stained (Fig. 2). For both columns the amount of protein eluted decreased with increasing KCl concentration. Some differences in the eluted profile are indicated by an asterisk in Fig. 2. Each lane was cut into 2-mm slices corresponding to different molecular weights and subjected to proteomics analysis.

Fig. 2.

Fig. 2.

Silver-stained gel of GST- and GST-RdCVFL interacting proteins. Soluble chicken retinal extracts were loaded on GST and GST-RdCVFL columns. Columns were washed, and four fractions were eluted from each column using a gradient of increasing KCl concentration. The first fractions (1 and A) contain the lowest KCl concentration (50 mm), and the last fractions (4 and D) contain the highest KCl concentration (500 mm). Proteins present in the GST-RdCVFL fractions and missing in the GST fractions are indicated by asterisks on the left side of the lane.

To identify the proteins, the trypsin-digested peptides in the individual slices were analyzed by nano-LC-MS/MS. Mass data collected during an LC-MS/MS analysis were processed and converted into a .pkl file to be submitted to the search software Mascot. A composite target-decoy database strategy was used to generate score criteria that yielded an estimated false positive rate of 1% (26). 44 and 121 proteins were identified as GST- and GST-RdCVFL-interacting proteins, respectively (supplemental Table I). After subtraction of the redundant proteins and the proteins eluted from the GST column, 90 proteins were found to interact specifically with RdCVFL (Table I). Among the proteins identified in this study, we found proteins related to apoptosis (apoptosis inhibitor 5 (API5) and poly(ADP-ribose) polymerase 1 (PARP1)) and linked to neurodegenerative diseases (TAU/MAPT (microtubule-associated protein Tau)). TAU was identified in several gel slices corresponding to molecular mass of 30–60 kDa by three different peptides (supplemental Table I).

Table I.

RdCVFL-interacting proteins identified by nano-LC-MS/MS

The first column lists the gene name for each protein. Four eluted fractions were collected. A, B, and D in the fifth column represent the KCl concentration at which the protein was eluted: A, 50–162.5 mm; B, 162.5–275 mm; and D, 387.5–500 mm. snRNP, small nuclear ribonucleoprotein; SH3, Src homology 3; Map, mitogen-activated protein; CNS, central nervous system; SNRPF, small nuclear ribonucleoprotein polypeptide F.

Symbol Protein name GI kDa Eluted fraction
ACTB β-Actin 211055 9 Da
AMFR Autocrine motility factor receptor 118096216 88 BD
API5 API5 57529974 59 AB
ATP6V1E1 Vacuolar H+-ATPase E1 57525423 26 ABD
ATP6V1G1 ATPase, H+-transporting 118099214 14 A
CAPZA1 F-actin-capping protein subunit α-1 (CapZ 36/32) 115592 33 Ba
CAPZA2 F-actin-capping protein subunit α-2 (CapZ 36/32) 115579 33 Da
CAPZB F-actin-capping protein subunit β isoforms 1 and 2 (CapZ 36/32) 115597 31 Da
CCT3 Chaperonin containing TCP1 (Homo sapiens) 119573352 61 Da
CPSF6 Cleavage and polyadenylation-specific factor 6 82233874 59 BD
DBN1 Drebrin 211726 72 D
DCAMKL1 KIAA0369 118084966 115 BD
DCD Dermcidin (H. sapiens) 119617200 11 D
DDX3X DEA(D/H) (Asp-Glu-Ala-(Asp/His)) box polypeptide 3 71895253 72 BD
DDX48 DEAD (Asp-Glu-Ala-Asp) box polypeptide 48 71897163 47 ADa
DDX5 DEAD (Asp-Glu-Ala-Asp) box polypeptide 5 45382259 67 BD
DDX6 DEAD (Asp-Glu-Ala-Asp) box polypeptide 6 118102030 54 B
DLAT Dihydrolipoamide S-acetyltransferase 118102025 72 Aa
DSTN Destrin 53128989 19 Da
EEF1B2 Peptide elongation factor 1-β 4324407 25 Da
EEF1G Eukaryotic translation elongation factor 1 118126430 6 Aa
EFTUD2 U5 snRNP-specific protein 71895651 109 Ba
EIF1AY EIF1AY 71895237 16 A
EIF3S1 Eukaryotic translation initiation factor 3 61098232 29 ABa
EIF4A2 Translational eukaryotic initiation factor 4AII 21435808 46 ABa
ELAVL1 RNA-binding protein HuA 5738249 36 Da
ERH Enhancer of rudimentary homologue 57529979 12 D
FKBP3 Putative FK506-binding protein 13992475 25 A
FUS FUS/TLS 47420845 52 ABDa
FXR1 Fragile X mental retardation protein 50746068 64 B
G3BP Ras-GTPase-activating protein SH3 domain-binding protein 57525015 52 B
GNAO1 Guanine nucleotide-binding protein 118096242 40 ABD
H1FX Histone H1x 118096909 23 ABDa
HIST1H1E Histone H1.01 121895 23 BD
HIST2H2AC Histone H2A 71896919 14 ABDa
HIST2H2BE Histone H2B 71895559 14 ABDa
HMGB3 HMG-1 391636 23 ABD
HNRPA0 Heterogeneous nuclear ribonucleoprotein A0 118097545 31 Da
HNRPH3 Heterogeneous nuclear ribonucleoprotein H3 60302824 17 D
HNRPK Heterogeneous nuclear ribonucleoprotein K 50762370 47 ABDa
HNRPR Heterogeneous nuclear ribonucleoprotein R 132626770 71 ABDa
HSD17B4 17-β-Hydroxysteroid dehydrogenase type IV 2315981 80 D
HSPA8 Heat shock cognate 70 2996407 71 ADa
HSPE1 Heat shock protein 10 2623879 11 A
KIAA1232 mKIAA1232 118089402 90 B
LASP1 LIM and SH3 protein 1 (H. sapiens) 119580947 30 Da
MAGOH2 MAGO protein 25990444 17 BDa
MAPK1 Chain A, structure of penta mutant human Erk2 Map kinase complexed with a specific inhibitor of human p38 Map kinase (H. sapiens) 157833528 43 Aa
MAPT CNS-specific microtubule-associated protein Tau 118102862 44 ABa
NPM1 Nucleophosmin 1 isoform 1 45383996 33 Aa
NUDT16L1 Nudix 45382147 33 D
PAFAH1B1 Platelet-activating factor acetylhydrolase (PAF) 45383504 47 B
PARP1 PARP1 3220000 114 ABa
PKM2 Pyruvate kinase muscle isozyme 125608 58 ABa
PPP2R2A α isoform of regulatory subunit B55, protein phosphatase 2 71894705 52 AD
PRDX1 Natural killer cell-enhancing factor 118094464 23 AD
PRP19 PRP19/PSO4 pre-mRNA processing factor 19 homolog (H. sapiens) 119594310 55 D
PRSS3 hCG22067 (H. sapiens) 119572363 30 ABD
RBBP4 Retinoblastoma-binding protein 4 45382339 48 B
RBMX RNA binding motif protein, X-linked 119331082 41 Da
RPL23A Rpl23a protein 118100216 18 Da
RPL30 Ribosomal protein L30 56118966 13 BDa
RPL5 RPL5 132989 34 Da
RPS10 Ribosomal protein S10 50760453 19 Da
RPS15A Ribosomal protein S15 118098058 15 Da
RPS19 Ribosomal protein S19 (M. musculus) 12963511 16 BDa
RPS20 Ribosomal protein S20 118086993 17 Aa
SDHA Chain A, avian mitochondrial respiratory complex 110590574 68 ABDa
SDHB Chain B, avian mitochondrial respiratory complex 110590575 29 D
SF3A1 Splicing factor 3a 71895509 88 Ba
SFRS1 Splicing factor, arginine/serine-rich 1 166091440 28 Da
SMD1 Small nuclear riboprotein Sm-D 118086133 14 AD
SND1 Transcriptional coactivator p100 16660153 80 AB
SNRPA1 SNRPA1 50753184 28 ABDa
SNRPD3 Small nuclear ribonucleoprotein D3 56119060 14 BDa
SNRPF SNRPF protein 118082523 10 Ba
SNRPN snRNP-B 10720263 25 ABDa
SPTBN1 Spectrin 118087525 274 AB
STAU Staufen 61098218 78 BD
SUB1 Activated RNA polymerase II transcription cofactor 4 56605972 14 A
TAGLN3 Transgelin 3 50729726 22 A
THOC4 REF1-I 109492372 28 ABDa
TNC Tenascin C 118099212 199 A
TNR Tenascin R 45384054 148 AB
UQCRC2 Ubiquinol-cytochrome c reductase 118098350 49 Da
VIM Vimentin 114326309 54 Aa
WDR48 WD repeat domain 48 82231191 76 ABDa
XLRS1 X-linked juvenile retinoschisis protein (M. musculus) 5114035 26 ABD
YWHAG YWHAG 71895543 28 B
ZNF207 ZNF207 118099726 53 A
a

The proteins that figure in the interactome scheme (Fig. 3).

RdCVFL Interactome—

We analyzed the relation between the 90 identified proteins by bioinformatics. We searched in the STRING database for interactomic connections using the human orthologue of the chicken proteins. The STRING database contains predicted interactions that are based on several methods, including gene co-occurrence in genomes (i.e. phylogeny), gene co-expression, gene fusion events, genomic neighborhood (i.e. synteny), text mining, and experimental data such as co-immunoprecipitation and yeast two hybrid (28). To minimize the rate of false positives, we eliminated 43 of the 90 RdCVF partners using stringent criteria. Three major clusters were found: actin-interacting proteins and cytoskeleton proteins, ribosomal proteins, and proteins involved in translation and spliceosome proteins (Fig. 3). We noticed the presence of proteins of these three clusters eluted at highest salt concentration (fraction D in Table I). These 47 proteins represent the first comprehensive analysis of the RdCVFL network. To confirm our results, we tested for enrichment of gene ontology terms for the initial list of 90 proteins and the 49 negative control proteins (GST) (30). We observed a statistically significant enrichment of spliceosome proteins (GO RNA splicing, RNA binding, and ribonucleoprotein complex) (Table II). For the GST-interacting proteins all corrected p values were superior to 10−5, representing only very weak enrichment (data not shown).

Fig. 3.

Fig. 3.

RdCVFL interactome network. A graph of the RdCVFL network deduced from the identified RdCVFL-interacting proteins (Table I) is shown. Three clusters are displayed: actin-binding proteins, ribosomal proteins, and spliceosome proteins. The analysis of the proteins binding to GST, the negative control (supplemental Table I), as well as the high level of expression of their corresponding mRNA probed by Affymetrix gene chip indicate that the ribosomal proteins are more likely nonspecific binding proteins (data not shown).

Table II.

Enrichment of gene ontology terms among RdCVFL-interacting proteins

Ontology categories and terms No. of genes p values
Biological process
    RNA splicing (GO:0008380) 17 2.6 × 10−10
    mRNA metabolic process (GO:0016071) 19 3.5 × 10−10
    mRNA processing (GO:0006397) 17 1.4 × 10−9
    RNA processing (GO:0006396) 18 5.4 × 10−7
Molecular function
    RNA binding (GO:0003723) 31 2.1 × 10−15
    Protein binding (GO:0005515) 74 8.6 × 10−11
    Nucleic acid binding (GO:0003676) 47 7.9 × 10−7
Cellular component
    Ribonucleoprotein complex (GO:0030529) 28 2.1 × 10−16
    Macromolecular complex (GO:0032991) 48 4.1 × 10−15
    Spliceosome (GO:0005681) 16 3.2 × 10−14
    Organelle part (GO:0044422) 52 2.5 × 10−13
    Intracellular organelle part (GO:0044446) 52 2.7 × 10−13
    Nuclear part (GO:0044428) 27 7.3 × 10−10
    Intracellular part (GO:0044424) 74 4.3 × 10−7
TAU, API5, and Ras-GTPase Are Expressed in Chicken Cone-enriched Cultures—

The 90 proteins identified are expressed in chicken retina, but we focused on proteins co-expressed with RdCVFL in cones for their potential implication in neurodegenerative diseases. TAU is a microtubule-associated protein that has been identified in aberrant protein aggregates found in Alzheimer disease. API5 was initially isolated as a gene whose expression promoted cell survival following serum deprivation (32). The Ras-GTPase-activating protein SH3 domain-binding protein (G3BP) plays an important role in the trafficking of macromolecules between the cytoplasm and nucleus (33). Finally histone H2A is potentially involved in transcription regulation at the level of the chromatin.

We tested the mRNA expression of these RdCVFL-interacting proteins in the cone-enriched cultures. We detected the products of amplification of the mRNA encoding TAU, API5, and Ras-GTPase but not that of histone H2A (Fig. 4). This shows that TAU, API5, and Ras-GTPase are expressed in cones, whereas histone H2A, whose mRNA expression is cell cycle-dependent, is not present in cultured cone cells. We focused our analysis on the protein TAU because of its implication in neurodegenerative diseases and potentially in retinal degenerations.

Fig. 4.

Fig. 4.

TAU, API5, and Ras-GTPase are expressed in cone-enriched culture from chicken embryos. RT-PCR analysis was performed. Specific primers were used for the detection of TAU, API5, Ras-GTPase, and histone H2A. TAU cDNA was detected as a smear in the range of 1000–1550 bp. The visinin primers were used as a positive control. Amplification product lengths are as follows: TAU, 1266 bp; API5, 1572 bp; and Ras-GTPase, 1419 bp.

Identification of Two Isoforms of TAU—

In human brain, six TAU isoforms arising from alternative mRNA splicing of exons 2, 3, and 10 of the TAU gene have been identified (34). By RT-PCR, the primers referring to TAU detected several amplification products that form a smear in a range of 1000–1550 bp (Fig. 4). This is in agreement with the proteomics analysis (various molecular masses, range of 30–60 kDa) and may reflect the expression of several isoforms of TAU in the cones. To sequence the complete TAU coding DNA sequence (CDS), we amplified the chicken TAU products (detected as a smear) and cloned the products into the pDEST/HA vector. We isolated two clones (Fig. 5A) and submitted the sequences of the isoforms 1 and 2 to NCBI under GenBank™ accession numbers EU380277 and EU380278 that correspond to the human isoforms 0N3R (352 aa) and 0N4R (383 aa), respectively. Both isoforms lack exons 2 and 3 present in the chicken TAU protein (GenBank accession number XM_424354). The N-terminal domain of isoform 1 is longer by 34 amino acids than the N-terminal of isoform 2, but the presence of an additional repeat domain (tubulin binding domain) in isomer 2 (exon 10) results in two proteins with similar length (Fig. 5A). We transfected COS-1 cells with plasmids encoding TAU isoforms 1 and 2 tagged with HA. By Western blotting analysis (anti-HA) we detected two bands of 68 and 66 kDa corresponding to isomers 1 and 2, respectively (Fig. 5B).

Fig. 5.

Fig. 5.

Identification of two novel isoforms of the TAU protein in the chicken retina. A, schematic presentation of the predicted form and the two isoforms of chicken TAU. The predicted TAU, GenBank accession number XM_424354, contains in its N terminus exons 2 and 3 and in its C terminus exon 10. Isoforms 1 and 2 lack exons 2 and 3. Isoform 2 carries exon 10 resulting from alternative splicing. This supplementary exon in isoform 2 results in one more “tubulin binding domain” in the C terminus of the sequence. Between exons 3 and 10, the shaded regions differ between the two isoforms. B, transfection of isoforms 1 and 2 in COS-1 cells. The isoforms were tagged with the epitope HA, and Western blotting was carried out using anti-HA antibody. The two bands corresponding to the two isoforms, 68 and 66 kDa, respectively, were detected.

Mapping of the Interaction between RdCVFL and TAU—

To validate the interaction between RdCVFL and TAU, we performed several co-transfections followed by co-immunoprecipitations followed by Western blot. First we transfected RdCVFL and isoform 1 of chicken TAU in COS-1 cells. The input lane shows that RdCVFL and TAU were expressed after transfection (Fig. 6A). When precipitated with anti-RdCVF-N antibodies (14) the Western blot using anti-HA (TAU was tagged with HA) revealed the presence of co-immunoprecipitated TAU protein (lane 2). Conversely precipitation of TAU with anti-HA led to the presence of RdCVFL in association with TAU (lane 3). We then assessed the specificity of this interaction by testing another member of the thioredoxin superfamily, TRX1. COS-1 cells were transfected with HA-TRX1 and HA-TAU (Fig. 6B). Because both proteins are tagged with HA, the immunoprecipitation with anti-HA revealed the presence of both proteins (lane 3). When precipitation was carried out with anti-TRX1, we only detected TRX1 and not TAU demonstrating the absence of an interaction between TRX1 and TAU (lane 2). We also tested RdCVF, the trophic factor (Fig. 6C). The immunoprecipitation of TAU did not co-precipitate RdCVF (lane 3). Altogether these results indicate that TAU interacts specifically with RdCVFL.

Fig. 6.

Fig. 6.

Mapping of the interaction between RdCVFL and TAU. COS-1 transfections followed by co-immunoprecipitation and Western blotting were carried out to validate and to characterize the interaction between RdCVFL and TAU. As a negative control no antibody (Ab) was used for the immunoprecipitation (IP). A, co-immunoprecipitation of RdCVFL and chicken TAU tagged with HA. Anti-HA and anti-RdCVFL were used for the precipitation (lanes 2 and 3), and the Western blot of the immunoprecipitates was probed for HA and RdCVFL. B, co-immunoprecipitation of TRX1 and TAU. Both proteins were tagged with HA. Anti-HA antibody was used for immunoprecipitation (lane 3), and anti-TRX1 was used for the co-immunoprecipitation (lane 2). The Western blot of the immunoprecipitates was probed for HA. C, co-immunoprecipitation of RdCVF and TAU. Anti-RdCVF antibody was used for immunoprecipitation (lane 2), and anti-HA (lane 3) was used for the co-immunoprecipitation. The Western blot of the immunoprecipitates was probed for RdCVF. D, co-immunoprecipitations of RdCVFL and HA-TAU C-terminal (microtubule binding domain) and N-terminal (projection domain) fragments (lanes 1–3 and lanes 4–6, respectively). Anti-RdCVF was used for immunoprecipitation (lanes 2 and 5), and anti-HA antibody was used for the co-immunoprecipitation (lanes 3 and 6). The Western blot of the immunoprecipitates was probed for RdCVFL.

TAU is composed of two main domains, a projection domain in the N terminus and a microtubule binding domain in the C terminus. Both domains bind to a subset of specific proteins. The sequences of 1–188 and 1–154 aa correspond to the projection domain in isomers 1 and 2, respectively, and the sequences 189–348 and 155–345 aa correspond to the microtubule binding domain in isomers 1 and 2, respectively. Fragments containing the projection domain and the microtubule binding domain were cloned into HA vector, and the interaction between the different domains and RdCVFL was tested by co-immunoprecipitation (Fig. 6D). RdCVFL was co-immunoprecipitated by anti-HA in the presence of co-transfected TAU microtubule binding domain (lane 3) but not TAU projection domain (lane 6; see also supplemental Fig. 1). These results show that RdCVFL interacts with the microtubule binding domain of TAU. Identical results were obtained using isoform 2 of TAU (results not shown).

TAU Is Hyperphosphorylated in the Retina of the Nxnl1−/− Mouse—

The TAU protein was identified to be in a hyperphosphorylated form in protein aggregates in the brains of patients suffering from AD (22). It has been shown that oxidative stress caused by the inactivation of the gene coding for superoxide dismutase-2 or in a mitochondrial aldehyde dehydrogenase 2 dominant mutant increases the level of TAU phosphorylation (35, 36). Because RdCVFL has a potential thiol oxidoreductase activity, we analyzed the level of phosphorylated TAU in retina of Nxnl1−/− mice. Nxnl1−/− mice were created by homologous recombination of the entire exon 1 that encodes the short isoform RdCVF and the N-terminal part of RdCVFL on a BALB/c background.2 RdCVFL was consequently absent in the retina of the Nxnl1−/− mouse as analyzed by Western blotting (Fig. 7A). Whereas the antibody Tau5 recognizes all forms of TAU independently of the phosphorylation state, AT8 recognizes TAU phosphorylated at residues Ser-202 and Thr-205. Retinas from Nxnl1−/− and Nxnl1+/+ mice aged 11 days, 4 months, and 12 months were analyzed as well as brains from animals at 5 months. The analysis showed an increase in the expression of TAU (Tau5) at 5 and 12 months when compared with actin that is independent of the genotype. In the Nxnl1−/− retina TAU is hyperphosphorylated (AT8) compared with the aged-matched Nxnl1+/+ retina (Fig. 7A). The increase in phosphorylation was also observed for the fetal isoform of TAU (labeled with an asterisk). No difference was detected in the level of TAU phosphorylation in the brain of the Nxnl1−/− mouse. This is consistent with the restricted expression of RdCVFL in the retina (14). We quantified the phosphorylated TAU (AT8) and normalized it to Tau5 and actin. There is a statistically significant increase in the excess of phosphorylation of TAU with age between 11 days and 5 and 12 months (Fig. 7B). To localize the hyperphosphorylated TAU, we performed immunohistochemistry with both TAU antibodies on retinal sections at 10 months. No difference was observed with Tau5 antibody, whereas the retina layers of the Nxnl1−/− mice had higher immunoreactivity for phosphorylated TAU (AT8; Fig. 7C). The inner nuclear layer, the outer nuclear layer, and to a limited extent the ganglion layer had a higher level of TAU phosphorylation consistent with the secretion of RdCVFL by rod photoreceptors.

Fig. 7.

Fig. 7.

TAU is hyperphosphorylated in the retina of the Nxnl1−/− mouse. A, the experiment was repeated on five different preparations. The results from a representative experiment are presented here. Retinas were dissected from 11-day-old, 5-month-old, and 12-month-old Nxnl1+/+ and Nxnl1−/− (BALB/c background) mice and lysed in Nonidet P-40 lysis buffer. The extracts were analyzed by Western blotting using anti-AT8, anti-Tau5, anti-actin, and anti-RdCVFL antibodies. Extracts from brains of 5-month-old mice were also analyzed. The asterisk indicates the fetal form of TAU. B, quantitative analysis. The anti-AT8 bands were quantified using the lab-tool software. For normalization, we quantified the bands corresponding to AT8, Tau5, and actin. We divided the quantification value of AT8 by Tau5. The results were then divided by the value of actin. The ratio between Nxnl1−/−/Nxnl1+/+ at different ages is 1.75 (11 days), 3.00 (5 months), and 2.94 (12 months). Statistical analysis was preformed using a two-tailed paired t test. The p values are provided in the figure. C, distribution of phosphorylated TAU (AT8) and TAU (Tau5) in 10-month-old mouse retina. Immunostaining of Nxnl1+/+ and Nxnl1−/− mice retina was carried out using AT8 and Tau5 antibodies. The two panels represents the same exposure time. GC, ganglion cells; INL, inner nuclear layer; ONL, outer nuclear layer. Scale bar, 10 μm.

RdCVFL Inhibits the Phosphorylation of TAU—

Glycogen synthase kinase-3β (GSK-3β) is a kinase abundant in the brain and in the retina that phosphorylates TAU in transfection assays (37). Isoform 0N4R of human TAU (383 aa) corresponds to chicken isoform 1 (0N4R has no N-terminal inserts (N) and four microtubule binding repeats (R)). To examine the activity of RdCVFL on TAU phosphorylation by GSK-3β, we transfected HEK 293 cells with plasmids encoding RdCVFL, GSK-3β, and human TAU (Fig. 8A). Transfection of GSK-3β led to an increase in phosphorylation of TAU as detected with AT8 antibody (lanes 2 and 7). The level of TAU phosphorylation was reduced by transfecting increasing amounts of RdCVFL (lanes 7–10). The expression of TAU and actin was only marginally affected. To demonstrate that the inhibition of TAU phosphorylation was a consequence of its specific interaction with RdCVFL, we repeated the experiment using TRX1, an enzymatically active thioredoxin that does not interact with TAU (Fig. 6B). Increasing the concentration of TRX1 did not reduce the level of phosphorylated TAU (lanes 2–5). The quantification of AT8 immunoreactivity normalized to Tau5 and actin demonstrated that RdCVFL inhibits TAU phosphorylation in a dose-dependent manner (Fig. 8B).

Fig. 8.

Fig. 8.

RdCVFL inhibits the phosphorylation of TAU. The experiment was repeated five times independently and further verified in COS-1 cells. The results from a representative experiment are presented here. A, HEK 293 cells were transfected with a fixed amount of TAU and GSK-3β (700 and 450 ng, respectively) and increasing concentrations of RdCVFL (450, 900, and 1350 ng) (lanes 8–10). As a control, RdCVFL was replaced by TRX1 (used in the same range of concentrations as RdCVFL) (lanes 3–5). Cells were lysed, and the protein extraction was subjected to Western blotting against anti-AT8, anti-Tau5, and anti-actin. B, quantitative analysis. The AT8 bands were quantified using the lab-tool software. The values were normalized to total TAU (Tau5) and to actin (AT8/Tau5/actin). The white columns represent the level of phosphorylated TAU with GSK-3β addition in the absence of RdCVFL or TRX1.

Protection of TAU Protein from Oxidation by RdCVFL—

To evaluate the functional consequence of this interaction on the redox status of TAU in vitro, we studied the oxidation of TAU by H2O2 in the presence of RdCVFL. Free thiols in purified TAU were alkylated by fluorescent IAF (Fig. 9A). Incubation with H2O2 induced TAU oxidation and consequently the decrease in fluorescence (lanes 4–5). This decrease was of lower amplitude in the presence of GST-RdCVFL (lane 6). Oxidation of TAU led to a decrease of the 62-kDa protein band (TAU), which results from oxidative damage of the protein (Fig. 9B, lanes 4–5). Interestingly GST-RdCVFL protected the TAU protein from this damage, and the level of TAU was no longer changed by the H2O2 oxidation (lane 6). We next analyzed the subsequent reduction of TAU by the thioredoxin system (TRX1 + thioredoxin reductase), the main cellular protein disulfide reduction system (17). The thioredoxin system reduced the oxidized TAU protein, resulting in an increase of fluorescent IAF (Fig. 9A, lane 9) as reported previously (38). The level of fluorescence was even higher with GST-RdCVFL addition (Fig. 9A, lane 8), and the level of undamaged TAU protein (Fig. 9B, lane 8) was increased. These results show that the interaction of RdCVFL with TAU protects this protein from oxidative damage, and this protection assists in the reduction of TAU by the thioredoxin system.

Fig. 9.

Fig. 9.

Protection of TAU protein from oxidation by RdCVFL in vitro. Oxidation of TAU by H2O2 in the presence of RdCVFL (lanes 1–6) and in the presence of the thioredoxin system (lanes 7–9) is shown. A, free thiols in purified TAU were alkylated by fluorescent IAF. The reaction was subjected to different conditions as indicated (lanes 1–9). B, Coomassie Blue-stained gel of the reactions under the same conditions. The migration of the different proteins is indicated on the right. IAM, iodoacetamide. Lane M, molecular mass markers.

DISCUSSION

RdCVF is a trophic factor belonging to the thioredoxin family. It is a spliced isoform of the Nxnl1 gene that additionally encodes RdCVFL, a protein that contains a complete thioredoxin fold domain and most likely carries a thiol oxidoreductase activity. Nevertheless this enzymatic activity could only be demonstrated in its distant orthologue, the arthropod Carcinoscorpius, and not for the human protein (18, 19). The role of RdCVFL in photoreceptor survival was speculative. The functional interaction between RdCVFL and TAU described here demonstrates that in addition to the activity of RdCVF on cones the second isoform of the Nxnl1 gene, RdCVFL, might participate in the protection of photoreceptors against oxidative stress.

To unravel the signaling pathway associated with RdCVFL, we used a proteomics approach to identify the RdCVFL-interacting proteins. The analysis of the RdCVFL interactome revealed three main clusters of proteins: ribosomal proteins, spliceosome proteins, and actin-binding proteins. Within the cluster of actin-binding proteins, the presence of microtubule-associated protein TAU, which is involved in neurodegenerative diseases, raises questions regarding the involvement of the TAU protein in photoreceptor degeneration.

TAU is a microtubule-associated protein that has a role in assembly and stabilization of microtubules. Twenty years ago, interest in the TAU protein arose when it was identified as a component of the paired helical filaments (39). Paired helical filaments contribute to neurofibrillary tangles and protein aggregates, the aberrant structures that were found in the brains of patients with AD; hence we focused our study on the interaction of RdCVFL with TAU. We demonstrated that RdCVFL as well as two newly described isoforms of TAU are expressed in the cone-enriched cultures and that RdCVFL interacts with TAU through the region containing a microtubule binding domain located at the C terminus of the protein. This region of the human TAU protein is enriched in phosphorylation sites that are grouped in two clusters located on both sides of the microtubules binding domain with the exception of Ser-262/Ser-356 (located inside the binding domain itself) (23). TAU was demonstrated to exist in a hyperphosphorylated form in neurofibrillary tangles in the brains of patients suffering from AD (22) that leads to a decrease in the binding of TAU to microtubules and makes microtubules dynamically unstable (23). In addition, there is evidence that hyperphosphorylated TAU is associated with the induction of apoptotic cell death (24). This led us to assess the level of hyperphosphorylation of TAU in the retina of mice carrying a deletion of the Nxnl1 gene. Both Western blotting and immunohistochemistry analyses demonstrated an increase in TAU phosphorylation in the neural retina in the absence of both products of the Nxnl1 gene, RdCVF and RdCVFL. The fact that the TAU immunoreactivity was localized into the photoreceptor layer and to other inner retinal cells may be due to the secretion of RdCVFL as observed previously (14). The observation of an increase in phosphorylated TAU between 11 days (1.75-fold) and 5 months (3.00-fold) indicates that it is not related to any abnormality in the maturation of the retina but to a process involving environmental factors, most likely oxidative stress. The relation between hyperphosphorylation of TAU and RdCVFL was further demonstrated in a cell-based assay. The inhibition of TAU phosphorylation is specifically mediated by the interaction with RdCVFL and not by a general redox mechanism as shown by the lack of TRX1 activity in that assay. The mechanism of action of RdCVFL was addressed in vitro. RdCVFL was able to prevent the oxidative damage to the TAU protein induced by H2O2 and assisted in the reduction of TAU by the thioredoxin system. Our results demonstrate that RdCVFL plays a role in the inhibition of TAU phosphorylation and protects TAU against oxidative stress. One hypothesis is that RdCVFL, the protein with a thioredoxin catalytic site (CPQC), possesses an enzymatic activity toward its interacting substrate TAU. This activity controls the redox status of cysteine residues in the C-terminal domain of the TAU protein, and as reported previously, oxidized TAU is more accessible to phosphorylation than the reduced form (38, 40). The hyperphosphorylation of TAU promotes its aggregation (22), a consequence that may represent the first step toward the induction of neuronal cell death (Fig. 10). We propose that RdCVFL, which displays oxidoreductase-like features, is capable of chemically reducing TAU under oxidative stress conditions, stabilizing it and thereby inhibiting its phosphorylation. The role of RdCVFL in protecting retinal cells from apoptosis mediated by oxidative stress is supported by the fact that transfection of the cone-like cell line 661W with RdCVFL leads to cell protection against the death induced by photooxidative damage (19).

Fig. 10.

Fig. 10

A model describing the role of RdCVFL-TAU interaction in the oxidative stress-induced retinal degeneration. red, reduced; ox, oxidation.

The contribution of this interaction to RdCVF signaling from rods to cones is still an open question. Because we showed that RdCVFL is expressed in the cone-enriched culture system, its position would be expected to be downstream of the activation of a putative RdCVF cell surface receptor. Alternatively and as reported earlier, Nxnl1 is a bifunctional gene with the longer isoform RdCVFL participating in an intracellular mechanism of photoreceptor protection against oxidative stress. The participation of RdCVFL in cone rescue may be connected to its specific association with the spliceosome (Table II) that may reveal a role of RdCVFL in regulating the splicing. It is worth noting that the splicing protein U5-15K/DIM1 has a thioredoxin fold (41). Under oxidative stress condition this association may inhibit the splicing of the Nxnl1 premessenger RNA and consequently enhance the production of the unspliced isoform RdCVF (42, 43). Oxidative stress has been implicated in the pathogeny of AD (44) and was shown at least in tauopathy models to proceed through the phosphorylation of TAU (45). Oxidative stress leads to the production of 4-hydroxynonenal and acrolein, which are found in the brain tissue of AD patients and favor TAU phosphorylation (46, 47). In animal models, the deficit in proteins controlling the production of free radicals such as superoxide dismutase-2 and aldehyde dehydrogenase 2 causes hyperphosphorylation of TAU (35, 36). All this combined evidence supports the role of oxidative stress in mediating TAU phosphorylation. The observation of abnormal TAU phosphorylation in a model lacking a thioredoxin family member strengthens the role of thioredoxin signaling in controlling the redox status of TAU and oxidative stress in neuronal tissues. Indeed this is the second time that an abnormal phosphorylation of TAU has been observed in the retina: hyperphosphorylated TAU was recently detected in the retina of patients suffering from glaucoma (48). The hyperphosphorylation of TAU in the retina of the Nxnl1−/− mouse (Fig. 7C) suggests that this mechanism is involved in triggering photoreceptor cell death in that model.2 The description of the hyperphosphorylation of TAU in the retina, a tissue particularly vulnerable to photooxidative damage, may extend its role in neurodegeneration and the relevance of oxidative stress. Although RdCVF expression is restricted to the retina, its paralogue, RdCVF2, is also expressed in the brain (31). This opens the possibility that the RdCVF family members may be involved in regulating TAU redox status and therefore its phosphorylation/aggregation in the brain of aging humans. This novel signaling pathway may be implicated in neurodegenerative diseases such as AD in addition to retinal degeneration. In such cases, RdCVF proteins might be used broadly for treating patients suffering from these disorders by regulating oxidative stress (49).

Acknowledgments

We thank E. Clerin, W. Raffelsberger, D. Thiersé, S. Fouquet, and B. Kinzel.

Footnotes

Published, MCP Papers in Press, March 11, 2009, DOI 10.1074/mcp.M800406-MCP200

1

The abbreviations used are: TRX, thioredoxin; AD, Alzheimer disease; CDS, coding DNA sequence; GSK-3β, glycogen synthase kinase-3; RdCVF, rod-derived cone viability factor; RdCVFL, rod-derived cone viability factor long isoform; Nxnl1, nucleoredoxin-like 1; STRING, Search Tool for the Retrieval of Interacting Genes/Proteins; GO, gene ontology; aa, amino acids; HEK, human embryonic kidney; API5, apoptosis inhibitor 5; PARP1, poly(ADP-ribose) polymerase 1; HA, hemagglutinin; IAF, 5-(iodoacetamido)fluorescein.

2

T. Cronin, W. Raffelsberger, I. Lee-Rivera, M.-L. Niepon, B. Kinzel, E. Clerin, A. Petrosian, S. Picaud, O. Poch, J. A. Sahel, and T. Leveillard, manuscript submitted.

*

This work was supported by INSERM, Agence Nationale de Recherches, European Vision Institute-GENORET, European Union Grant MRTN-CT-2003-504003, and Retina France.

S

The on-line version of this article (available at http://www.mcponline.org) contains supplemental material.

The nucleotide sequence(s) reported in this paper has been submitted to the GenBank™EBI Data Bank with accession number(s) EU380277 and EU380278.

REFERENCES

  • 1.Jones, D. P. ( 2006) Extracellular redox state: refining the definition of oxidative stress in aging. Rejuvenation Res. 9, 169–181 [DOI] [PubMed] [Google Scholar]
  • 2.Reynolds, A., Laurie, C., Mosley, R. L., and Gendelman, H. E. ( 2007) Oxidative stress and the pathogenesis of neurodegenerative disorders. Int. Rev. Neurobiol. 82, 297–325 [DOI] [PubMed] [Google Scholar]
  • 3.Lundmark, P. O., Pandi-Perumal, S. R., Srinivasan, V., Cardinali, D. P., and Rosenstein, R. E. ( 2007) Melatonin in the eye: implications for glaucoma. Exp. Eye Res. 84, 1021–1030 [DOI] [PubMed] [Google Scholar]
  • 4.Beatty, S., Koh, H., Phil, M., Henson, D., and Boulton, M. ( 2000) The role of oxidative stress in the pathogenesis of age-related macular degeneration. Surv. Ophthalmol. 45, 115–134 [DOI] [PubMed] [Google Scholar]
  • 5.Cingolani, C., Rogers, B., Lu, L., Kachi, S., Shen, J., and Campochiaro, P. A. ( 2006) Retinal degeneration from oxidative damage. Free Radic. Biol. Med. 40, 660–669 [DOI] [PubMed] [Google Scholar]
  • 6.Yu, D. Y., Cringle, S., Valter, K., Walsh, N., Lee, D., and Stone, J. ( 2004) Photoreceptor death, trophic factor expression, retinal oxygen status, and photoreceptor function in the P23H rat. Investig. Ophthalmol. Vis. Sci. 45, 2013–2019 [DOI] [PubMed] [Google Scholar]
  • 7.Komeima, K., Rogers, B. S., Lu, L., and Campochiaro, P. A. ( 2006) Antioxidants reduce cone cell death in a model of retinitis pigmentosa. Proc. Natl. Acad. Sci. U. S. A. 103, 11300–11305 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Yoshida, T., Oka, S., Masutani, H., Nakamura, H., and Yodoi, J. ( 2003) The role of thioredoxin in the aging process: involvement of oxidative stress. Antioxid. Redox Signal. 5, 563–570 [DOI] [PubMed] [Google Scholar]
  • 9.Tanito, M., Masutani, H., Nakamura, H., Oka, S., Ohira, A., and Yodoi, J. ( 2002) Attenuation of retinal photooxidative damage in thioredoxin transgenic mice. Neurosci. Lett. 326, 142–146 [DOI] [PubMed] [Google Scholar]
  • 10.Akterin, S., Cowburn, R. F., Miranda-Vizuete, A., Jimenez, A., Bogdanovic, N., Winblad, B., and Cedazo-Minguez, A. ( 2006) Involvement of glutaredoxin-1 and thioredoxin-1 in β-amyloid toxicity and Alzheimer's disease. Cell Death Differ. 13, 1454–1465 [DOI] [PubMed] [Google Scholar]
  • 11.Lovell, M. A., Xie, C., Gabbita, S. P., and Markesbery, W. R. ( 2000) Decreased thioredoxin and increased thioredoxin reductase levels in Alzheimer's disease brain. Free Radic. Biol. Med. 28, 418–427 [DOI] [PubMed] [Google Scholar]
  • 12.Saitoh, M., Nishitoh, H., Fujii, M., Takeda, K., Tobiume, K., Sawada, Y., Kawabata, M., Miyazono, K., and Ichijo, H. ( 1998) Mammalian thioredoxin is a direct inhibitor of apoptosis signal-regulating kinase (ASK) 1. EMBO J. 17, 2596–2606 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Lillig, C. H., and Holmgren, A. ( 2007) Thioredoxin and related molecules—from biology to health and disease. Antioxid. Redox Signal. 9, 25–47 [DOI] [PubMed] [Google Scholar]
  • 14.Leveillard, T., Mohand-Said, S., Lorentz, O., Hicks, D., Fintz, A. C., Clerin, E., Simonutti, M., Forster, V., Cavusoglu, N., Chalmel, F., Dolle, P., Poch, O., Lambrou, G., and Sahel, J. A. ( 2004) Identification and characterization of rod-derived cone viability factor. Nat. Genet. 36, 755–759 [DOI] [PubMed] [Google Scholar]
  • 15.Cronin, T., Leveillard, T., and Sahel, J. A. ( 2007) Retinal degenerations: from cell signaling to cell therapy; pre-clinical and clinical issues. Curr. Gene Ther. 7, 121–129 [DOI] [PubMed] [Google Scholar]
  • 16.Sahel, J. A. ( 2005) Saving cone cells in hereditary rod diseases: a possible role for rod-derived cone viability factor (RdCVF) therapy. Retina 25, S38–S39 [DOI] [PubMed] [Google Scholar]
  • 17.Arner, E. S., and Holmgren, A. ( 2000) Physiological functions of thioredoxin and thioredoxin reductase. Eur. J. Biochem. 267, 6102–6109 [DOI] [PubMed] [Google Scholar]
  • 18.Wang, X. W., Liou, Y. C., Ho, B., and Ding, J. L. ( 2007) An evolutionarily conserved 16-kDa thioredoxin-related protein is an antioxidant which regulates the NF-κB signaling pathway. Free Radic. Biol. Med. 42, 247–259 [DOI] [PubMed] [Google Scholar]
  • 19.Wang, X. W., Tan, B. Z., Sun, M., Ho, B., and Ding, J. L. ( 2008) Thioredoxin-like 6 protects retinal cell line from photooxidative damage by upregulating NF-κB activity. Free Radic. Biol. Med. 45, 336–344 [DOI] [PubMed] [Google Scholar]
  • 20.Pekkari, K., Avila-Carino, J., Gurunath, R., Bengtsson, A., Scheynius, A., and Holmgren, A. ( 2003) Truncated thioredoxin (Trx80) exerts unique mitogenic cytokine effects via a mechanism independent of thiol oxido-reductase activity. FEBS Lett. 539, 143–148 [DOI] [PubMed] [Google Scholar]
  • 21.Drubin, D. G., and Kirschner, M. W. ( 1986) Tau protein function in living cells. J. Cell Biol. 103, 2739–2746 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Grundke-Iqbal, I., Iqbal, K., Tung, Y. C., Quinlan, M., Wisniewski, H. M., and Binder, L. I. ( 1986) Abnormal phosphorylation of the microtubule-associated protein τ (tau) in Alzheimer cytoskeletal pathology. Proc. Natl. Acad. Sci. U. S. A. 83, 4913–4917 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Buee, L., Bussiere, T., Buee-Scherrer, V., Delacourte, A., and Hof, P. R. ( 2000) Tau protein isoforms, phosphorylation and role in neurodegenerative disorders. Brain Res. Brain Res. Rev. 33, 95–130 [DOI] [PubMed] [Google Scholar]
  • 24.Brandt, R., Hundelt, M., and Shahani, N. ( 2005) Tau alteration and neuronal degeneration in tauopathies: mechanisms and models. Biochim. Biophys. Acta 1739, 331–354 [DOI] [PubMed] [Google Scholar]
  • 25.Fintz, A. C., Audo, I., Hicks, D., Mohand-Said, S., Leveillard, T., and Sahel, J. ( 2003) Partial characterization of retina-derived cone neuroprotection in two culture models of photoreceptor degeneration. Investig. Ophthalmol. Vis. Sci. 44, 818–825 [DOI] [PubMed] [Google Scholar]
  • 26.Elias, J. E., and Gygi, S. P. ( 2007) Target-decoy search strategy for increased confidence in large-scale protein identifications by mass spectrometry. Nat. Methods 4, 207–214 [DOI] [PubMed] [Google Scholar]
  • 27.Peng, J., Elias, J. E., Thoreen, C. C., Licklider, L. J., and Gygi, S. P. ( 2003) Evaluation of multidimensional chromatography coupled with tandem mass spectrometry (LC/LC-MS/MS) for large-scale protein analysis: the yeast proteome. J. Proteome Res. 2, 43–50 [DOI] [PubMed] [Google Scholar]
  • 28.von Mering, C., Jensen, L. J., Kuhn, M., Chaffron, S., Doerks, T., Kruger, B., Snel, B., and Bork, P. ( 2007) STRING 7—recent developments in the integration and prediction of protein interactions. Nucleic Acids Res. 35, D358–D362 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Shannon, P., Markiel, A., Ozier, O., Baliga, N. S., Wang, J. T., Ramage, D., Amin, N., Schwikowski, B., and Ideker, T. ( 2003) Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 13, 2498–2504 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Sherman, B. T., Huang, D. W., Tan, Q., Guo, Y., Bour, S., Liu, D., Stephens, R., Baseler, M. W., Lane, H. C., and Lempicki, R. A. ( 2007) DAVID Knowledgebase: a gene-centered database integrating heterogeneous gene annotation resources to facilitate high-throughput gene functional analysis. BMC Bioinformatics 8, 426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Chalmel, F., Leveillard, T., Jaillard, C., Lardenois, A., Berdugo, N., Morel, E., Koehl, P., Lambrou, G., Holmgren, A., Sahel, J. A., and Poch, O. ( 2007) Rod-derived Cone Viability Factor-2 is a novel bifunctional-thioredoxin-like protein with therapeutic potential. BMC Mol. Biol. 8, 74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Tewari, M., Yu, M., Ross, B., Dean, C., Giordano, A., and Rubin, R. ( 1997) AAC-11, a novel cDNA that inhibits apoptosis after growth factor withdrawal. Cancer Res. 57, 4063–4069 [PubMed] [Google Scholar]
  • 33.Solomon, S., Xu, Y., Wang, B., David, M. D., Schubert, P., Kennedy, D., and Schrader, J. W. ( 2007) Distinct structural features of caprin-1 mediate its interaction with G3BP-1 and its induction of phosphorylation of eukaryotic translation initiation factor 2α, entry to cytoplasmic stress granules, and selective interaction with a subset of mRNAs. Mol. Cell. Biol. 27, 2324–2342 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Goedert, M., Spillantini, M. G., Jakes, R., Rutherford, D., and Crowther, R. A. ( 1989) Multiple isoforms of human microtubule-associated protein tau: sequences and localization in neurofibrillary tangles of Alzheimer's disease. Neuron 3, 519–526 [DOI] [PubMed] [Google Scholar]
  • 35.Melov, S., Adlard, P. A., Morten, K., Johnson, F., Golden, T. R., Hinerfeld, D., Schilling, B., Mavros, C., Masters, C. L., Volitakis, I., Li, Q. X., Laughton, K., Hubbard, A., Cherny, R. A., Gibson, B., and Bush, A. I. ( 2007) Mitochondrial oxidative stress causes hyperphosphorylation of tau. PLoS ONE 2, e536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Ohsawa, I., Nishimaki, K., Murakami, Y., Suzuki, Y., Ishikawa, M., and Ohta, S. ( 2008) Age-dependent neurodegeneration accompanying memory loss in transgenic mice defective in mitochondrial aldehyde dehydrogenase 2 activity. J. Neurosci. 28, 6239–6249 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Lovestone, S., Reynolds, C. H., Latimer, D., Davis, D. R., Anderton, B. H., Gallo, J. M., Hanger, D., Mulot, S., Marquardt, B., Stabel, S., Woodgett, J. R., and Miller, C. C. J. ( 1994) Alzheimer's disease-like phosphorylation of the microtubule-associated protein tau by glycogen synthase kinase-3 in transfected mammalian cells. Curr. Biol. 4, 1077–1086 [DOI] [PubMed] [Google Scholar]
  • 38.Landino, L. M., Skreslet, T. E., and Alston, J. A. ( 2004) Cysteine oxidation of tau and microtubule-associated protein-2 by peroxynitrite: modulation of microtubule assembly kinetics by the thioredoxin reductase system. J. Biol. Chem. 279, 35101–35105 [DOI] [PubMed] [Google Scholar]
  • 39.Grundke-Iqbal, I., Iqbal, K., Quinlan, M., Tung, Y. C., Zaidi, M. S., and Wisniewski, H. M. ( 1986) Microtubule-associated protein tau. A component of Alzheimer paired helical filaments. J. Biol. Chem. 261, 6084–6089 [PubMed] [Google Scholar]
  • 40.Schweers, O., Mandelkow, E. M., Biernat, J., and Mandelkow, E. ( 1995) Oxidation of cysteine-322 in the repeat domain of microtubule-associated protein tau controls the in vitro assembly of paired helical filaments. Proc. Natl. Acad. Sci. U. S. A. 92, 8463–8467 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Zhang, Y. Z., Gould, K. L., Dunbrack, R. J., Cheng, H., Roder, H., and Golemis, E. A. ( 1999) The evolutionarily conserved Dim1 protein defines a novel branch of the thioredoxin fold superfamily. Physiol. Genomics 1, 109–118 [DOI] [PubMed] [Google Scholar]
  • 42.Schmidt-Kastner, R., Yamamoto, H., Hamasaki, D., Yamamoto, H., Parel, J. M., Schmitz, C., Dorey, C. K., Blanks, J. C., and Preising, M. N. ( 2008) Hypoxia-regulated components of the U4/U6.U5 tri-small nuclear riboprotein complex: possible role in autosomal dominant retinitis pigmentosa. Mol. Vis. 14, 125–135 [PMC free article] [PubMed] [Google Scholar]
  • 43.Takahashi, N., Kariya, S., Hirano, M., and Ueno, S. ( 2003) Two novel spliced presenilin 2 transcripts in human lymphocyte with oxidant stress and brain. Mol. Cell. Biochem. 252, 279–283 [DOI] [PubMed] [Google Scholar]
  • 44.Nunomura, A., Perry, G., Aliev, G., Hirai, K., Takeda, A., Balraj, E. K., Jones, P. K., Ghanbari, H., Wataya, T., Shimohama, S., Chiba, S., Atwood, C. S., Petersen, R. B., and Smith, M. A. ( 2001) Oxidative damage is the earliest event in Alzheimer disease. J. Neuropathol. Exp. Neurol. 60, 759–767 [DOI] [PubMed] [Google Scholar]
  • 45.Fulga, T. A., Elson-Schwab, I., Khurana, V., Steinhilb, M. L., Spires, T. L., Hyman, B. T., and Feany, M. B. ( 2007) Abnormal bundling and accumulation of F-actin mediates tau-induced neuronal degeneration in vivo. Nat. Cell Biol. 9, 139–148 [DOI] [PubMed] [Google Scholar]
  • 46.Gomez-Ramos, A., Diaz-Nido, J., Smith, M. A., Perry, G., and Avila, J. ( 2003) Effect of the lipid peroxidation product acrolein on tau phosphorylation in neural cells. J. Neurosci. Res. 71, 863–870 [DOI] [PubMed] [Google Scholar]
  • 47.Mattson, M. P., Fu, W., Waeg, G., and Uchida, K. ( 1997) 4-Hydroxynonenal, a product of lipid peroxidation, inhibits dephosphorylation of the microtubule-associated protein tau. Neuroreport 8, 2275–2281 [DOI] [PubMed] [Google Scholar]
  • 48.Gupta, N., Fong, J., Ang, L. C., and Yucel, Y. H. ( 2008) Retinal tau pathology in human glaucomas. Can. J. Ophthalmol. 43, 53–60 [DOI] [PubMed] [Google Scholar]
  • 49.Yang, Y., Mohand-Said, S., Danan, A., Simonutti, M., Fontaine, V., Clerin, E., Picaud, P., Léveillard, T., and Sahel, J. A. ( 2009) Functional cone rescue by RdCVF protein in a dominant model of retinitis pigmentosa. Mol. Ther., in press [DOI] [PMC free article] [PubMed]

Articles from Molecular & Cellular Proteomics : MCP are provided here courtesy of American Society for Biochemistry and Molecular Biology

RESOURCES