Click
here to close Hello! We notice that
you are using Internet Explorer, which is not supported by Echinobase
and may cause the site to display incorrectly. We suggest using a
current version of Chrome,
FireFox,
or Safari.
PLoS One
2013 Dec 16;812:e84588. doi: 10.1371/journal.pone.0084588.
Show Gene links
Show Anatomy links
Nitrosative/oxidative stress conditions regulate thioredoxin-interacting protein (TXNIP) expression and thioredoxin-1 (TRX-1) nuclear localization.
Ogata FT
,
Batista WL
,
Sartori A
,
Gesteira TF
,
Masutani H
,
Arai RJ
,
Yodoi J
,
Stern A
,
Monteiro HP
.
Abstract
Thioredoxin (TRX-1) is a multifunctional protein that controls the redox status of other proteins. TRX-1 can be found in the extracellular milieu, cytoplasm and nucleus, and it has distinct functions in each environment. Previously, we studied the intracellular localization of TRX-1 and its relationship with the activation of the p21Ras-ERK1/2 MAP Kinases signaling pathway. In situations where this pathway was activated by stress conditions evoked by a nitrosothiol, S-nitroso-N-acetylpenicillamine (SNAP), TRX-1 accumulated in the nuclear compartment due to nitrosylation of p21Ras and activation of downstream ERK1/2 MAP kinases. Presently, we demonstrate that ERK1/2 MAP Kinases activation and spatial distribution within cells trigger TRX-1 nuclear translocation through down-regulation of the physiological inhibitor of TRX-1, Thioredoxin Interacting Protein (TXNIP). Once activated by the oxidants, SNAP and H₂O₂, the ERK1/2 MAP kinases migrate to the nucleus. This is correlated with down-regulation of TXNIP. In the presence of the MEK inhibitors (PD98059 or UO126), or in cells transfected with the Protein Enriched in Astrocytes (PEA-15), a cytoplasmic anchor of ERK1/2 MAP kinases, TRX-1 nuclear migration and TXNIP down-regulation are no longer observed in cells exposed to oxidants. On the other hand, over-expression of TXNIP abolishes nuclear migration of TRX-1 under nitrosative/oxidative stress conditions, whereas gene silencing of TXNIP facilitates nuclear migration even in the absence of stress conditions. Studies based on the TXNIP promoter support this regulation. In conclusion, changes in TRX-1 compartmentalization under nitrosative/oxidative stress conditions are dependent on the expression levels of TXNIP, which are regulated by cellular compartmentalization and activation of the ERK1/2 MAP kinases.
Figure 1. HeLa cells submitted to oxidative and nitrosative stress conditions.(A) HeLa cells were treated with increasing concentrations of SNAP (0 - 1.0 mM) during 2 h. After this period medium was replaced for complete medium (MEM supplemented with 10% FBS). Cell viability was determined 24 h later using the MTT assay as described in Materials and Methods. Values are reported in the bar graphs and expressed as means ± S.D. (n = 3, *p < 0.05). (B) Intracellular NO detection in HeLa cells after incubation with 0.5 mM SNAP for 2 h at 37°C. DAF fluorescence is directly related to NO generation by intracellular metabolism of SNAP and was determined by flow cytometry. (C) HeLa cells were treated with increasing concentrations of H2O2 (0 - 1.0 mM) during 2 h. After this period medium was replaced for complete medium (MEM supplemented with 10% FBS). Cell viability was determined 24 h later using the MTT assay as described in Materials and Methods. Values are reported in the bar graphs and expressed as means ± S.D. (n = 3, *p < 0.05). (D) Catalase and Glutathione Peroxidase enzymatic activity assays (described in Materials and Methods) were performed after exposure of HeLa cells to 0.5 mM H2O2 for 2 h at 37°C. (E) HeLa cells were exposed to 0.5 mM SNAP or to 0.5 mM H2O2 for 2 h at 37°C. After cell lysis, 50 μg total protein were subjected to Western blotting with a rabbit polyclonal anti-phospho-Akt antibody and with a mouse monoclonal anti-Akt antibody. Western blot results are representative of three independent experiments. Histogram represents the ratio between the densitometric values of the protein bands corresponding to the phosphorylated form of Akt and of Akt protein of one representative experiment (Lower panel).
Figure 2. Confocal microscopy and western blot analysis to determine TRX-1 and phospho ERK 1/2 MAP Kinases sub-cellular location in wild-type HeLa cells.(A) Cells were treated with SNAP (0.5 mM), or H2O2 (0.5 mM) for 2 h at 37°C. Detection of TRX-1 and phospho-ERK 1/2 MAP Kinases was performed as described in Materials and Methods. Bar = 20µm. (B) Cells were treated with SNAP (0.5mM) or H2O2 (0.5 mM) for 2 h at 37°C. HeLa cells were pre-treated with 50μM PD98059, a MEK inhibitor, for 30 min and incubated with 0.5mM SNAP or 0.5 mM H2O2 for 2 h at 37°C. Detection of TRX-1 was performed on a fluorescence microscope as described in Materials and Methods. Images shown are representative from three independent experiments. Bar = 20µm. (C) HeLa cells were incubated with SNAP and H2O2 at the indicated concentrations for 2 h and nuclear extracts were subjected to Western blotting with the anti-human TRX-1 mouse monoclonal antibody and with the anti-human phospho-ERK1/2 MAP Kinases rabbit polyclonal antibody. Western blot results are representative of three independent experiments. Purity of nuclear fractions was estimated by LDH detection as described in Materials and Methods. Values are means ± S.D. of triplicate cultures.
Figure 3. Stress-induced sub-cellular localization and activation of TRX-1 and phospho ERK 1/2 MAP Kinases in HeLa cells expressing PEA-15.(A) HeLa cells and HeLa cells expressing PEA-15 were incubated with SNAP (0.5 mM) or H2O2 (0.5 mM) for 2 h at 37°C. Cells were lysed and phosphorylation of the ERK1/2 MAP kinases was examined by western blotting. Western blot results are representative of four independent experiments. Histogram represents the ratio between the densitometric values of the protein bands corresponding to the phosphorylated form and of the total ERK 1/2 MAP kinases (Lower panel). (B) HeLa cells expressing PEA-15, were treated with SNAP (0.5 mM), or H2O2 (0.5 mM) for 2 h at 37°C. Sub-cellular localization of TRX-1 and phospho-ERK 1/2 MAP Kinases was performed as described in Materials and Methods. Bar = 20µm. (C) HeLa cells and HeLa cells expressing PEA-15 were incubated with SNAP (0.5 mM) or H2O2 (0.5 mM) for 2 h at 37°C. Nuclear extracts were subjected to Western blotting with the anti-human TRX-1 mouse monoclonal antibody. Western blot results are representative of three independent experiments.
Figure 5. SNAP and H2O2 -modulated TXNIP mRNA and protein levels in wild-type Hela cells and in HeLa cells expressing PEA-15.(A) Relative levels of TXNIP mRNA determined by quantitative real-time PCR in HeLa cells wild-type, in HeLa cells expressing PEA-15, and in HeLa cells wild-type pre-treated with 50μM of PD98059 for 30 min as indicated (see Materials and Methods for details). All cell lines were treated with 0.5mM SNAP or 0.5mM H2O2 for 2 h at 37°C. Values are reported in the bar graphs and expressed as means ± S.D. (n = 3, *p < 0.05; **p < 0.01). (B) wild-type HeLa cells and HeLa cells expressing PEA-15 were incubated with SNAP (0.5 mM) or H2O2 (0.5 mM) for 2 h at 37°C. Cells were lysed and expression levels of TXNIP were examined by western blotting. Western blot results are representative of four independent experiments. Histogram represents the ratio between the densitometric values of the protein bands corresponding to TXNIP and of β-actin (Lower panel).
Figure 6. TXNIP silencing and TRX-1 intracellular localization.(A) Western blot analysis to determine the effects of mRNA TXNIP knockdown on TXNIP protein levels in HeLa cells permanently transfected with a shRNA for TXNIP. The blot presented is representative of three independent experiments. (B) Indirect immunofluorescence to determine TRX-1 sub-cellular localization in HeLa cells permanently transfected with a shRNA for TXNIP. Images shown are representative from three independent experiments. Blue arrows indicate TRX-1 nuclear accumulation. Bar = 20µm.
Figure 7. Confocal microscopy based analysis to determine a time-dependent sub-cellular localization of TRX-1 in HeLa cells.(A) Wild-type HeLa cells, HeLa cells expressing PEA-15, and HeLa cells expressing TXNIP were plated two days prior to experiment. Transient transfections with pEGFP-C1_TRX-1 were performed on day one. On day two, cells were assayed for TRX-1 sub-cellular localization. Cells were kept in a heated stage chamber (37°C) of the confocal microscope, and a first scanning was recorded to serve as control. After that, media was replaced and H2O2 was added to a final concentration of 0.5 mM. Subsequent scanning were performed at every 15 min up to 2 h. Bar = 20µm. (B) Cells were transfected with pEGFP-C1-TRX-1, treated with SNAP (0.5 mM) for 2 h at 37°C and submitted to indirect immunofluorescence for GFP, to show the localization of TRX-1 in HeLa cells. Red arrows indicate TRX-1 nuclear accumulation.
Figure 8. Determination of sub-cellular compartmentalization of TRX-1 in HeLa cells transiently over-expressing PEA-15; TXNIP; TRX-1 and exposed to 0.5 mM H2O2.HeLa wild-type cells were seeded in complete medium and 24 h later were transiently transfected with the following plasmids pcDNA3_PEA-15, pcDNA3_TXNIP, and pEGFP-C1_TRX-1. After 16 h, transiently transfected cultures were divided to perform either western blot analysis, to confirm over expression of designated inserts, or to perform immunofluorescence-based analysis to determine TRX-1 sub cellular localization after exposure to 0.5 mM H2O2. (A) HeLa cells transfected with pcDNA3_PEA-15. Left panel: western blot analysis of PEA-15 expression. Right panel: Immunofluorescence image of TRX-1 cellular location. (B) HeLa cells transfected with pcDNA3_TXNIP. Left panel: western blot analysis of TXNIP expression. Right panel: Immunofluorescence image of TRX-1 cellular location (C) HeLa cells transfected with pEGFP-C1_TRX-. Left panel: western blot analysis of TRX-1 expression. Right panel: Immunofluorescence image of TRX-1 cellular location. Red arrows indicate TRX-1 nuclear accumulation.
Figure 9. Schematic representation of TRX-1 and the ERK1/2 MAP kinases nuclear translocation stimulated by nitrosative/oxidative stress conditions.(A) The Ras-Raf-MEK-ERK 1/2 signaling pathway is activated in HeLa wild-type cells after exposure to 0.5 mM SNAP or 0.5 mM H2O2 (1). Under these conditions expression of TXNIP is down-regulated at mRNA and protein levels (2) and TRX-1 and the ERK 1/2 MAP kinases, independently of each other, migrate to the nuclear compartment (3). (B) Nuclear migration of TRX-1 and the ERK 1/2 MAP kinases is prevented in three situations: HeLa cells pre-incubated with the MEK inhibitor PD98059 (4), HeLa cells over-expressing the cytoplasmic anchor of ERK 1/2 MAP kinases – PEA-15 (5), HeLa cells over-expressing the physiological inhibitor of TRX-1, TXNIP (6).
Aebi,
Catalase in vitro.
1984,
Pubmed
Akamatsu,
Redox regulation of the DNA binding activity in transcription factor PEBP2. The roles of two conserved cysteine residues.
1997,
Pubmed
Arai,
Nitric oxide induces thioredoxin-1 nuclear translocation: possible association with the p21Ras survival pathway.
2006,
Pubmed
Arnér,
Physiological functions of thioredoxin and thioredoxin reductase.
2000,
Pubmed
Bai,
Critical roles of thioredoxin in nerve growth factor-mediated signal transduction and neurite outgrowth in PC12 cells.
2003,
Pubmed
Batista,
S-nitrosoglutathione and endothelial nitric oxide synthase-derived nitric oxide regulate compartmentalized ras S-nitrosylation and stimulate cell proliferation.
2013,
Pubmed
,
Echinobase
Bradford,
A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding.
1976,
Pubmed
Butler,
The histone deacetylase inhibitor SAHA arrests cancer cell growth, up-regulates thioredoxin-binding protein-2, and down-regulates thioredoxin.
2002,
Pubmed
Chen,
Isolation and characterization of a novel cDNA from HL-60 cells treated with 1,25-dihydroxyvitamin D-3.
1994,
Pubmed
Chen,
Prediction of interface residues in protein-protein complexes by a consensus neural network method: test against NMR data.
2006,
Pubmed
Chen,
Lack of TXNIP protects against mitochondria-mediated apoptosis but not against fatty acid-induced ER stress-mediated beta-cell death.
2010,
Pubmed
Deora,
Recruitment and activation of Raf-1 kinase by nitric oxide-activated Ras.
2000,
Pubmed
Eu,
An apoptotic model for nitrosative stress.
2000,
Pubmed
Ferrè,
DiANNA 1.1: an extension of the DiANNA web server for ternary cysteine classification.
2006,
Pubmed
Flohé,
Assays of glutathione peroxidase.
1984,
Pubmed
Formstecher,
PEA-15 mediates cytoplasmic sequestration of ERK MAP kinase.
2001,
Pubmed
Haling,
Phosphoprotein enriched in astrocytes 15 kDa (PEA-15) reprograms growth factor signaling by inhibiting threonine phosphorylation of fibroblast receptor substrate 2alpha.
2010,
Pubmed
Hirota,
AP-1 transcriptional activity is regulated by a direct association between thioredoxin and Ref-1.
1997,
Pubmed
Hirota,
Distinct roles of thioredoxin in the cytoplasm and in the nucleus. A two-step mechanism of redox regulation of transcription factor NF-kappaB.
1999,
Pubmed
Holmgren,
Thioredoxin.
1985,
Pubmed
Junn,
Vitamin D3 up-regulated protein 1 mediates oxidative stress via suppressing the thioredoxin function.
2000,
Pubmed
Kaadige,
Glutamine-dependent anapleurosis dictates glucose uptake and cell growth by regulating MondoA transcriptional activity.
2009,
Pubmed
Livak,
Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method.
2002,
Pubmed
Liyanage,
Regulation of the bioavailability of thioredoxin in the lens by a specific thioredoxin-binding protein (TBP-2).
2007,
Pubmed
Masaki,
Deficiency of thioredoxin binding protein-2 (TBP-2) enhances TGF-β signaling and promotes epithelial to mesenchymal transition.
2012,
Pubmed
Meuillet,
Thioredoxin-1 binds to the C2 domain of PTEN inhibiting PTEN's lipid phosphatase activity and membrane binding: a mechanism for the functional loss of PTEN's tumor suppressor activity.
2004,
Pubmed
Minn,
Thioredoxin-interacting protein is stimulated by glucose through a carbohydrate response element and induces beta-cell apoptosis.
2005,
Pubmed
Murphy,
MAPK signal specificity: the right place at the right time.
2006,
Pubmed
Nakamura,
Redox regulation of cellular activation.
1997,
Pubmed
Nishinaka,
Importin alpha1 (Rch1) mediates nuclear translocation of thioredoxin-binding protein-2/vitamin D(3)-up-regulated protein 1.
2004,
Pubmed
Nishiyama,
Identification of thioredoxin-binding protein-2/vitamin D(3) up-regulated protein 1 as a negative regulator of thioredoxin function and expression.
1999,
Pubmed
Nishizawa,
Thioredoxin-interacting protein suppresses bladder carcinogenesis.
2011,
Pubmed
Oliveira,
The low molecular weight S-nitrosothiol, S-nitroso-N-acetylpenicillamine, promotes cell cycle progression in rabbit aortic endothelial cells.
2008,
Pubmed
Oliveira,
Nitric oxide and cGMP activate the Ras-MAP kinase pathway-stimulating protein tyrosine phosphorylation in rabbit aortic endothelial cells.
2004,
Pubmed
Pouysségur,
Fidelity and spatio-temporal control in MAP kinase (ERKs) signalling.
2002,
Pubmed
Schonhoff,
The Ras-ERK pathway is required for the induction of neuronal nitric oxide synthase in differentiating PC12 cells.
2001,
Pubmed
Schulze,
Vitamin D3-upregulated protein-1 (VDUP-1) regulates redox-dependent vascular smooth muscle cell proliferation through interaction with thioredoxin.
2002,
Pubmed
Schulze,
Hyperglycemia promotes oxidative stress through inhibition of thioredoxin function by thioredoxin-interacting protein.
2004,
Pubmed
Schulze,
Nitric oxide-dependent suppression of thioredoxin-interacting protein expression enhances thioredoxin activity.
2006,
Pubmed
Sheth,
Hepatocellular carcinoma in Txnip-deficient mice.
2006,
Pubmed
Stoltzman,
MondoA senses non-glucose sugars: regulation of thioredoxin-interacting protein (TXNIP) and the hexose transport curb.
2012,
Pubmed
Stoltzman,
Glucose sensing by MondoA:Mlx complexes: a role for hexokinases and direct regulation of thioredoxin-interacting protein expression.
2008,
Pubmed
Turturro,
Hyperglycemia-induced thioredoxin-interacting protein expression differs in breast cancer-derived cells and regulates paclitaxel IC50.
2007,
Pubmed
Ueno,
Thioredoxin-dependent redox regulation of p53-mediated p21 activation.
2000,
Pubmed
Wang,
Vitamin D(3)-up-regulated protein-1 is a stress-responsive gene that regulates cardiomyocyte viability through interaction with thioredoxin.
2002,
Pubmed
Yu,
Adenosine-containing molecules amplify glucose signaling and enhance txnip expression.
2009,
Pubmed
Yu,
Rapid and direct quantitative RT-PCR method to measure promoter activity.
2007,
Pubmed