Literature
首页医源资料库在线期刊动脉硬化血栓血管生物学杂志2007年第27卷第11期

Nuclear Redox-Signaling Is Essential for Apoptosis Inhibition in Endothelial Cells—Important Role for Nuclear Thioredoxin-

来源:《动脉硬化血栓血管生物学杂志》
摘要:NuclearTrxplaysanimportantroleinendothelialcellapoptosis。NuclearimportofTrxisrequiredfortheantiapoptoticpropertiesofH2O2。NuclearTrxbindstoseveraltranscriptionfactorsandincreasestranscriptionfactorbindingtoantioxidantresponsiveelements(AREs)。Separationo......

点击显示 收起

【摘要】  Objective— The redox regulator thioredoxin-1 (Trx) is a potent antioxidative enzyme and exerts important cellular functions. Physiological concentrations of reactive oxygen species (ROS) and of nitric oxide (NO) act as second messengers. Previously, we demonstrated that ROS and NO reduced apoptosis in a Trx-dependent manner. The aim of this study was to determine the underlying mechanisms.

Methods and Results— First, we investigated the localization of Trx after H 2 O 2 and NO. Both induced nuclear import of Trx, which required karyopherin-. siRNA against karyopherin- inhibited nuclear import of Trx. Analysis of the Trx amino acid sequence and subsequent immunoprecipitation studies revealed that Trx(K81/82E) is not imported into the nucleus under H 2 O 2 treatment and Trx(K81/82/85E) was retained in the cytosol and induced cell death. Trx(K81/82E) abolished the antiapoptotic capacity of H 2 O 2. Glutathione S-transferase P1 (GST-P1) was identified as one major target regulated by H 2 O 2. siRNA against GST-P1 abolished the antiapoptotic effect of H 2 O 2. Cysteine 69, but not cysteines 32 and 35, which are all required for the complete antiapoptotic function of Trx, is not imported into the nucleus.

Conclusion— H 2 O 2 -induced nuclear import of Trx depends on karyopherin- and NO. Trx-dependent induction of GST-P1 expression is required for apoptosis inhibition in endothelial cells.

Physiological concentrations of ROS and NO activate signaling pathways. ROS induce karyopherin –dependent nuclear import of Trx, which is enhanced by posttranslational modifications of Trx. Import of Trx modified antioxidant responsive element containing promoters and increased GST-P1 expression. Knocking down GST-P1 abolished the antiapoptotic properties of ROS. Nuclear Trx plays an important role in endothelial cell apoptosis.

【关键词】  antioxidants apoptosis reactive oxygen species glutathione Stransferase P thioredoxin


Introduction


Oxygen and nitric oxide (NO) are physiologically relevant molecules. In physiological concentrations reactive oxygen species (ROS) and NO play important roles in the regulation of a variety of cellular functions including vascular tone, migration, apoptosis, and proliferation. 1,2 One of these molecules, which is highly regulated by changes of the redox status in cells, is the oxidoreductase thioredoxin-1 (Trx). It is known that cells possess antioxidative enzymes, including superoxide dismutase, catalase, and Trx to maintain intracellular levels of ROS and reactive nitrogen species in physiological concentrations. 3,4 The thioredoxin family includes 3 proteins, thioredoxin-1, thioredoxin-2, and Sp-thioredoxin. 5–7 All of them contain a conserved -Cys-Gly-Pro-Cys- active site (cysteine 32 and cysteine 35 within thioredoxin-1), which is essential for the redox regulatory function of thioredoxins. 8,9


Trx is a 12-kDa protein, which is ubiquitously expressed in mammalian cells 8 and exerts its enzymatic activity as an oxidoreductase via cysteines 32 and 35 in the active site. 8,9 This site is conserved among species from bacteria to humans. 8,10 The active site cysteines are accessible on the surface of the protein and are oxidized to a disulfide upon reduction of a target protein. 9,11,12 Trx itself is reduced by thioredoxin-reductase. These 2 oxidoreductases form the thioredoxin system in mammalian cells. Besides its well described function as an oxidoreductase, Trx exerts several other functions. By binding to different proteins, it modulates their function: Inhibition of binding to the apoptosis signaling kinase 1 and to the transcription factors AP1, Ref1, and Nf B modulates the ability of Trx to regulate cellular functions. 13–16


Previously, we have demonstrated that physiological concentrations of H 2 O 2 and NO inhibit apoptosis in endothelial cells in a Trx dependent manner. 17,18 Although we were able to demonstrate that mutation of cysteine 69 in Trx reduced its antiapoptotic capacity, 17 the underlying mechanisms are not entirely clear.


Therefore, the aim of this study was to elucidate the mechanisms underlying the antiapoptotic properties of physiological concentrations of H 2 O 2 and to identify the role of Trx. Here, we demonstrate that on exposure of cells to physiological concentrations of H 2 O 2 and NO Trx is imported into the nucleus in a karyopherin- –dependent manner. Nuclear import of Trx is required for the antiapoptotic properties of H 2 O 2. Nuclear Trx binds to several transcription factors and increases transcription factor binding to antioxidant responsive elements (AREs). Glutathione S-transferase P1 (GST-P1) expression is increased. Genetic ablation of GST-P1 by siRNA completely abrogated H 2 O 2 -induced inhibition of apoptosis, demonstrating that increased expression of GST-P1 is 1 major downstream target for the Trx-dependent antiapoptotic properties of physiological concentrations of H 2 O 2.


Methods


Cell Culture


Human umbilical vein endothelial cells (HUVECs) were cultured in endothelial basal medium supplemented with hydrocortisone (1 µg/mL), bovine brain extract (12 µg/mL), gentamicin (50 µg/mL), amphotericin B (50 ng/mL), epidermal growth factor (10 ng/mL), and 10% fetal calf serum. After detachment with trypsin, cells were grown for at least 18 hours. Human embryonic kidney cells (HEK293) were cultured in DMEM basal medium with 10% heat-inactivated fetal calf serum. Neonatal rat cardiac myocytes were isolated as described previously. 19


Transfection


Endothelial cell single strand cDNA was prepared out of endothelial cell RNA using RT-PCR. Trx was cloned out of endothelial cell cDNA using the following primers: 5'-GTGGTACCTTGGTGAAGCAGATCGAGAGC (sense) and 5'-CTCTAGACTTAGACTAATTCATTAATGGTGGC (antisense) incorporating Kpn I and Xba I restriction sites. The amplified PCR product was subcloned into pcDNA4/His vector containing a Xpress-tag and an enterokinase (EK) recognition site (Invitrogen). TrxK85E, TrxK81E/K82E, TrxK81E/K82E/K85E, and TrxK94E/K96E were generated by site directed mutagenesis (Stratagene) out of Trx wt.


HUVECs were transfected with 3 µg plasmid and 25 µL Superfect with a transfection efficiency of 40%. For transfection of siRNA 6.6 µg double stranded DNA were transfected with JetSi (Eurogentec) according to the manufacturer?s instructions.


cDNA Generation and PCR


cDNA was generated out of 5 µg total RNA using random hexamer primers according to the manufacturers instruction (Invitrogen). Primers to human karyopherin- : 5'ATCCTGAGGCTTGGAGAACA-3' and 5'-GGTCCCGAAGTAATGCTCAA-3' to human glyceraldehyde-3-phosphate dehydrogenase, 5'-GAAGGTGAAGGTCGGAGTC-3' and 5'-GAAGATGGTGATGGGATTTC-3'.


Separation of Nuclear and Cytosolic Fractions


Nuclear and cytosolic fractions were separated as described previously. 20 Purity of the nuclear and cytosolic fractions were assured by immunoblotting with topoisomerase 1 (nuclear) and HSP70 (cytosolic) as demonstrated previously. 20


Immunoprecipitation


Lysates (500 µg) were immunoprecipitated with 5 µg Xpress-antibody overnight at 4°C. After incubation with G Sepharose (Amersham) for 2 hours at 4°C, resulting beads were washed, subjected to SDS-PAGE sample buffer, and resolved by a SDS-PAGE.


Immunoblot


After stimulation for the indicated times, endothelial cells were scraped off the plates and lysed in RIPA buffer (50 mmol/L Tris-HCl pH 8.0, 150 mmol/L NaCl, 1% Nonidet-P40, 0.5% deoxycholic acid, 0.1% sodium dodecyl sulfate). After removing cell debris (15 minutes, 4°C, 20 000 g ), proteins were resolved on SDS-polyacrylamide gels and blotted onto polyvinylidene fluoride (PVDF) membranes. For detection of protein expression, membranes were incubated with antibodies against Trx (1:500, overnight, Pharmingen), karyopherin- (1:250, overnight, Santa Cruz), topoisomerase I (1:250, overnight, Santa Cruz) or tubulin (1:1000, 2 hours, Neomarkers). After incubation for 2 hours with the corresponding secondary antibody tagged with horse radish peroxidase, signals were detected by the enhanced chemiluminescence system (Amersham).


Protein-DNA Binding Array


Trx-Xpress was immunoprecipitated out of nuclear fractions after H 2 O 2 incubation for 6 hours. Protein/DNA arrays were performed in the washed immunoprecipitates according to the manufacturer's protocol (Panomics). Briefly, immunoprecipitate was incubated with the TranSignal Probe Mix. DNA/protein complexes were washed. Then, DNA was separated from protein and hybridized on the membranes at 42°C. Signal was detected using ECL-Hyperfilm. To semiquantitatively analyze the dot blots, spots were scanned and normalized to the internal controls on the membranes as suggested by the manufacturer?s instructions. Scanned spots of Trxwt + H 2 O 2 were set to 1 and intensity (=protein-DNA binding) of TrxK81E/K82E + H 2 O 2 was calculated using scion image.


Detection of Cell Death by Fluorescence-Activated-Cell Sorter (FACS)


Detection of cell death was performed by FACS analysis using annexin V-PE binding and 7-Amino-actinomycin (7AAD)-fluorescein isothiocyanate (FITC) staining (Pharmingen). In brief, cells were trypsinized of the dish and pelleted. After washing twice with annexin binding buffer, cell pellets were resuspended in 50 µL of annexin binding buffer and incubated with 2.5 ng/mL annexin V-PE and 2.5 ng/mL 7AAD-FITC for 20 minutes and analyzed using FACS.


Immunostaining


Cells were fixed with 4% paraformaldehyde for 15 minutes at room temperature. After permeabilization and blocking, cells were incubated with anti–Trx-antibody (1:50, BD Biosciences) or anti-Xpress antibody (1:50, Invitrogen) overnight at 4°C. After incubation with a Rhodamine Red X–conjugated anti-mouse antibody (1:300, Jackson ImmunoResearch), cells were incubated with RNase and nuclei were stained with Cytox-Blue (1:500, Invitrogen). For knockdown experiments against karyopherin-, cells were first incubated with anti Trx-antibody (1:50, BD Biosciences) overnight at 4°C. After incubation with a Rhodamine Red X–conjugated anti-mouse antibody (1:300, Jackson ImmunoResearch), cells were incubated with karyopherin- (1:25, Santa Cruz) antibody overnight at 4°C. After incubation with a FITC-conjugated anti-goat antibody (1:300, Molecular Probes), nuclei were stained with Tropro-3-iodide (1:1000, Invitrogen). Cells were visualized by confocal microscopy (Zeiss, LSM 510 META, Objective: Plan-Apochromat 63 x, 1,4 oil).


Statistics


Statistical analysis was performed with student t test or ANOVA followed by modified LSD (Bonferroni) test (SPSS-Software).


Results


Trx Is Imported Into the Nucleus Under Physiological Concentrations of H 2 O 2


Previously, we have demonstrated that low doses of H 2 O 2 protect endothelial cells from apoptosis in a Trx-dependent manner. However, the underlying mechanism is completely unclear. It is known that Trx can bind to several proteins, including transcription factors, and thereby modulate their functions. Therefore, we first investigated the localization of Trx after treatment with 10 µmol/L H 2 O 2 for 6 hours. H 2 O 2 induced an increase in nuclear localization of Trx in endothelial cells (supplemental Figure I, available online at http://atvb.ahajournals.org; Figure 1A). This phenomenon was also seen in other cell types including cardiomyocytes ( Figure 1 B), suggesting a general mechanism for the regulation of Trx by low doses of reactive oxygen species.


Figure 1. Nuclear Trx levels are increased under H 2 O 2. Endothelial cells (A) and cardiomyocytes (B) were treated with H 2 O 2 for 6 hours and nuclear Trx expression was detected (Topoisomerase I=Topo I).


Trx Is Imported Into the Nucleus via Karyopherins


To understand the nuclear import mechanism of Trx, we first analyzed the amino acid sequence of Trx and determined a potential nuclear import sequence for the import receptors, karyopherin- and karyopherin-β, at lysines 81, 82, and 85 (supplemental Figure II). To verify whether Trx is indeed imported into the nucleus in a karyopherin- – and karyopherin-β–dependent manner, we mutated the lysines to glutamates, to block nuclear import of Trx. We created several mutants: TrxK85E, TrxK81E/K82E, and TrxK81E/K82E/K85E. Overexpression of TrxK81E/K82E/K85E resulted in cell death of endothelial cells. Only after treatment with a pan-caspase inhibitor ZVAD and cell-permeable pepstatin A, we were able to detect endothelial cells overexpressing TrxK81E/K82E/K85E. TrxK81E/K82E/K85E is not detectable in the nucleus and shows a punctual staining (supplemental Figure III), demonstrating that lysines 81, 82, and 85 are required for nuclear import of Trx. Therefore, we did not pursue any further experiments with this mutant. We assured that Trxwt and TrxK81EK82E are overexpressed to a similar extent in endothelial cells (supplemental Figure II). Moreover, TrxK81E/K82E is in the nucleus under basal conditions, but H 2 O 2 did not increase nuclear import of TrxK81E/K82E (supplemental Figure IV and Figure 2 ). In certain cases after H 2 O 2 incubation, TrxK81E/K82E was only detected in the cytoplasm (supplemental Figure IV). In contrast, TrxK85E was imported under H 2 O 2 treatment as efficiently as Trxwt ( Figure 2 ).


Figure 2. Import of Trx into the nucleus dependent on lysines 81, 82, and 85. Endothelial cells were transfected with Trxwt, TrxK81E/K82E, or TrxK85E and treated with H 2 O 2. Trx expression was detected in nuclear extracts. Bar graphs show densitometric analysis (n=4).


To verify that karyopherins are indeed the import receptors for Trx, we overexpressed Trxwt or TrxK81E/K82E in endothelial cells, treated the cells for 6 hours with 10 µmol/L H 2 O 2, and immunoprecipitated Trxwt and the mutant with a Xpress antibody or for reverse Immunoprecipitation with a karyopherin- antibody. As expected, Trx associated with karyopherin- ( Figure 3 A and data not shown). The association increased after H 2 O 2 treatment in Trxwt transfected cells, but not in cells overexpressing TrxK81E/K82E ( Figure 3 A). Next, we wanted to investigate a direct link between the association of Trx with karyopherin- and the nuclear import of Trx. Therefore, we knocked down karyopherin- by siRNA transfection in endothelial cells ( Figure 3B and 3 C). The maximal reduction of karyopherin-, which did not kill the cells, was around 50% ( Figure 3 B and data not shown). Using this reduction in karyopherin-, H 2 O 2 -induced nuclear import of Trx was strongly inhibited ( Figure 3 C).


Figure 3. Karyopherin- –dependent nuclear import of Trx. A, Trxwt or TrxK81E/K82E-transfected cell lysates were immunoprecipitated. Semi quantitative analyses were performed. The diagram shows the ratio of karyopherin- to Trx-Xpress. B and C, Knockdown of karyopherin-. C, Left panels: Trx staining (red), second panels: karyopherin- (green), third panels: nuclear staining (blue); right panels represent the merge.


Nuclear Import of Trx Is Required for the Antiapoptotic Properties of H 2 O 2


To test whether nuclear import is indeed required for the antiapoptotic functions of low doses of H 2 O 2, we overexpressed Trxwt and TrxK81E/K82E in endothelial cells. As a structural control, we also cloned TrxK94E/K96E, in which the putative second nuclear import motif is destroyed (see supplemental Figure II). As expected, Trxwt inhibited basal apoptosis 17 ( Figure 4 A). Moreover, the antiapoptotic effect of low doses of H 2 O 2 is completely abrogated in endothelial cells overexpressing TrxK81E/K82E ( Figure 4 A). In contrast, TrxK94E/K96E did not influence the antiapoptotic properties of low doses of H 2 O 2 ( Figure 4 A). Serum-deprivation–induced apoptosis was also dependent on nuclear import of Trx. Overexpression of Trxwt, but not of TrxK81E/K82E, reduced apoptosis induction ( Figure 4 B) demonstrating that nuclear import of Trx is indeed required to protect from basal and stimulus-induced apoptosis.


Figure 4. The antiapoptotic effects of H 2 O 2 depend on nuclear import of Trx and the activation of ARE containing promoters. A-C, Endothelial cells were transfected and treated as indicated. Data are means±SEM. A and B, Apoptosis was measured by FACS analysis. A, * P <0.05 vs EV + H 2 O 2; ** P <0.05 vs Trxwt+H 2 O 2, n=5 (EV=empty vector). B, * P <0.05 vs Trxwt+serum-deprivation. ** P <0.05 vs Trxwt+serum-deprivation+H 2 O 2, n=3. C, Transcription factors bound to Trx were analyzed by protein-DNA binding assay. Bar graphs show densitometric analysis (n=3).


Genes With an ARE Containing Promoter Are Necessary for Apoptosis Inhibition by Nuclear Trx


Trx has been demonstrated to regulate transcription factors, such as NF- B, by protein binding. Thus, we hypothesized that nuclear import of Trx may influence the activity of transcription factors. To get insights in the underlying mechanisms, we investigated protein-DNA binding of 400 transcription factors. To only investigate the transcription factors bound to Trx, we first immunoprecipitated Trxwt and TrxK81E/K82E after stimulation with H 2 O 2 out of nuclear extracts. The immunoprecipitates were then subjected to protein-DNA binding studies. The most pronounced differences were found by genes containing an ARE motif in their promoters (supplemental Figure V and Figure 4 C). We also detected slight differences in AP1 and AP2, whereas activity of Nf B was unaltered ( Figure 4 C).


Glutathione S-Transferase P1 (GST-P1) Is Required for the Antiapoptotic Effects of Low Doses of H 2 O 2


One important protein which contributes to the redox status of cells is GST-P1. GST-P1 contains 4 ARE motifs in its promoter. Therefore, we next investigated the regulation of GST-P1 protein expression by low doses of H 2 O 2 in endothelial cells. Low doses of H 2 O 2 increased protein expression of GST-P1 in cells expressing Trxwt or TrxK85E ( Figure 5 A). Interestingly, in endothelial cells overexpressing TrxK81E/K82E, H 2 O 2 was unable to upregulate GST-P1 protein expression. To elucidate whether the protein expression of GST-P1 is indeed required as a downstream target of nuclear Trx for the antiapoptotic effects of H 2 O 2, we genetically ablated GST-P1 expression by siRNA ( Figure 5 B). Genetic knock down of GST-P1 protein expression inversed the antiapoptotic effects of low doses of H 2 O 2 ( Figure 5 C), demonstrating that GST-P1 is one of the targets regulated by nuclear Trx which plays an important role for the antiapoptotic effects of low doses of H 2 O 2.


Figure 5. GST-P1 is required for the antiapoptotic effects of H 2 O 2. A, GST-P1 protein expression was detected, bar graph shows the densitometric analysis (n=4). B and C, GST-P1 expression was knocked down in endothelial cells. C, Apoptosis was measured by FACS. Data are means±SEM. (n=4). * P <0.05 vs GST-P1 scrambled control; ** P <0.05 vs GST-P1 scrambled+H 2 O 2.


Posttranslational Modification by Nitric Oxide Is Required for Nuclear Import of Thioredoxin


Finally, we investigated the signals responsible to import Trx into the nucleus. Trx has been described to be posttranslationally modified by oxidation, nitros(yl)ation, or glutathionylation (for review see 21 ). Therefore, we determined whether one of these posttranslational modifications has an impact on nuclear import of Trx. Oxidation of Trx occurs on the active site cysteines 32 and 35. Therefore, we isolated nuclear extracts from mouse hearts overexpressing either Trxwt or TrxC32S/C35S. 22 We did not detect any difference between nuclear localization of Trxwt and TrxC32S/C35S in mouse hearts as well as in endothelial cells ( Figure 6 A and data not shown). This is in accordance to findings by Hirota et al who excluded modifications on cysteine 32 and cysteine 35 for nuclear import of Trx in their cell system. 23 It has been demonstrated in cells and in a mouse model that S-nitros(yl)ation of Trx enhanced its activity, its antiapoptotic capacity, and its cardioprotective properties. 17,24 Thus, it is tempting to speculate that NO influences the nuclear import of Trx. Therefore, we stimulated endothelial cells with different NO-donors and measured nuclear Trx protein. Trx protein levels are increased in the nucleus after stimulation with NO ( Figure 6 B). Moreover, TrxC69S, a Trx mutant which cannot be S-nitros(yl)ated in endothlelial cells, was barely detected in the nucleus, demonstrating that posttranslational modification by NO is required for nuclear import of Trx ( Figure 6 C).


Figure 6. Cystein 69 is required for nuclear import. A, Nuclear fractions were isolated from mouse hearts overexpressing Trxwt or TrxC32S/C35S. Immunoblot was performed. B, Endothelial cells were treated as indicated (SNP:10 µmol/L; NOC-15:100 µmol/L) and nuclear extracts isolated. C, Endothelial cells were transfected with Trxwt or TrxC69S and stimulated with NO or H 2 O 2. Immunoblot analysis was performed in nuclear extracts.


Discussion


The findings of our present study demonstrate that nuclear Trx is required for the antiapoptotic effects of low doses of H 2 O 2. We determined for the first time that (1) Trx is imported into the nucleus by karyopherin alpha, (2) GST-P1 is one important downstream target activated by nuclear Trx, and (3) posttranslational modification of Trx by NO is necessary for nuclear import of Trx.


ROS are formed and degraded by all aerobic organisms, leading to either physiological concentrations required for normal cell function, or excessive quantities resulting in oxidative stress. Ideally, a metabolically active cell should strike a balance between ROS production and the cellular antioxidant defense system. Different studies, predominantly in smooth muscle cells, show that on stimulation with EGF or PDGF, proliferation is induced in a ROS-dependent manner. 25,26 Furthermore, ROS can inhibit apoptosis and induce angiogenesis in endothelial cells and other cell types. 18,27,28 Therefore, intracellular ROS can be seen as a signal transduction messenger involved in several intracellular mechanisms. Here, we extend these findings and demonstrate that the antiapoptotic effects of H 2 O 2 depend on nuclear Trx. Nuclear Trx regulates ARE containing promoters of genes and thereby acts antiapoptotic. It has to be noted that the promoter of Trx contains ARE binding sites and that expression of the Trx gene itself can be induced through ARE. 29 This activation is dependent on the NF-E2–related factor (Nrf2). Kim et al postulated that Trx served as a factor facilitating DNA binding of Nrf2 to the ARE. Thereby, the ARE-triggered Trx gene activation may enhance the ARE-mediated induction of ARE-controlled enzymes. Indeed, we demonstrate here that nuclear Trx is required to induce GST-P1, which contains AREs in its promoter. This suggested that a feedback loop exists for balancing oxidant and antioxidant systems, which is essential for a cell to survive, because a gain of ROS product formation or a loss in antioxidative capacity can disturb the equilibrium and subsequently lead to cellular destruction.


In this study, we identified the nuclear import mechanism of Trx. The nuclear import receptors karyopherin- and -β have been described for nuclear import of several targets. 30 Proteins, which are smaller than 40 kDa have been proposed to freely diffuse through the nuclear pores. Here, we provide evidence that the 12-kDa protein Trx is imported into the nucleus by karyopherin- under certain conditions in endothelial cells. Thus, one may speculate that depending on the cellular conditions, a regulated nuclear import of small proteins exists.


In this study we identified GST-P1 as one major downstream target for nuclear Trx signaling. GST-P1 is an antioxidative enzyme, which was recently been implicated to protect from oxidative stress–induced cardiac injury. 31 The underlying mechanisms are not well understood. Recently, Zhao et al demonstrated that GST-P1 inhibited etoposide-induced apoptosis by inhibiting MEKK-1 activity, pro–caspase-3 activation, and PARP cleavage. 32 Here, we demonstrate an upstream mediator for GST-P1, namely nuclear Trx. Our findings provide evidence for the underlying mechanism, because induction of nuclear Trx did not only occur in endothelial cells but also in cardiac myocytes. Therefore, it is tempting to speculate that nuclear Trx is essential to inhibit oxidative stress induced cardiac injury. Indeed, it has been shown that specific overexpression of the catalytically inactive TrxC32S/C35S in the heart led to hypertrophy and oxidative stress–induced DNA damage in vivo. 22 Our data clearly demonstrate that TrxC32S/C35S can still be imported into the nucleus. However, cysteines 32 and 35 are required for the ability of Trx to bind to transcription factors and thereby modulate their protein functions.


Interestingly, nuclear import of Trx depends on its posttranslational modification by NO in endothelial cells. We have demonstrated that binding of NO by Trx increased its antiapoptotic function. 17 NO is a crucial factor for the integrity and function of the endothelium. Besides its role in blood pressure regulation, NO acts antithrombotically and antiapoptotically. Moreover, the cardioprotective and antiapoptotic effects of Trx against ischemia-reperfusion injury were more pronounced when S-nitros(yl)ated Trx was infused into mice. 24 Thus, Trx in concert with NO seems to be crucial for protection in endothelial cells and cardiac myocytes. Moreover, it seems that a balance between antioxidative and oxidative systems also exists in the nucleus, because we demonstrate here that Trx localized in the nucleus. Another important question is where is Trx S-nitros(yl)ated in the cell? Because the nuclear import of Trx is greatly impaired when cysteine 69 is mutated to serine, one may speculate that modification of Trx has to occur in the cytoplasm to allow Trx to be imported into the nucleus. However, further studies are needed to determine the compartment of S-nitros(yl)ation of Trx.


In conclusion, our data provide compelling evidence that nuclear Trx is one important signaling molecule, which modulates transcription factor activities. Trx is imported into the nucleus in a karyopherin- –dependent manner, and this nuclear import of Trx is required for apoptosis inhibition. Moreover, one major downstream target, which is activated by nuclear Trx, is GST-P1. GST-P1 also plays an important role as an antioxidant. Strikingly, the nuclear import of Trx depends mainly on its S-nitros(yl)ation. Thus, S-nitros(yl)ated Trx could be one important strategy to enhance nuclear Trx and to protect from cardiac disorders.


Acknowledgments


We thank Diane Schmiegelt for expert technical assistance.


Sources of Funding


This work was supported by a grant from the Deutsche Forschungsgemeinschaft to J.H. (HA2868/3-2).


Disclosures


None.


P.S. and R.P. contributed equally to this study.


Original received June 6, 2007; final version accepted August 28, 2007.

【参考文献】
  Harrison DG. Endothelial dysfunction in atherosclerosis. Basic Res Cardiol. 1994; 1: 87–102.

Girerd XJ, Hirsch AT, Cooke JP, Dzau VJ, Creager MA. L-arginine augments endothelium-dependent vasodilation in cholesterol-fed rabbits. Circ Res. 1990; 67: 1301–1308.

Orr WC, Sohal RS. Extension of life-span by overexpression of superoxide dismutase and catalase in Drosophila melanogaster. Science. 1994; 263: 1128–1130.

Nordberg J, Arner ES. Reactive oxygen species, antioxidants, and the mammalian thioredoxin system. Free Radic Biol Med. 2001; 31: 1287–1312.

Taniguchi Y, Taniguchi-Ueda Y, Mori K, Yodoi J. A novel promoter sequence is involved in the oxidative stress-induced expression of the adult T-cell leukemia-derived factor (ADF)/human thioredoxin (Trx) gene. Nucleic Acids Res. 1996; 24: 2746–2752.

Spyrou G, Enmark E, Miranda-Vizuete A, Gustafsson J. Cloning and expression of a novel mammalian thioredoxin. J Biol Chem. 1997; 272: 2936–2941.

Miranda-Vizuete A, Ljung J, Damdimopoulos AE, Gustafsson JA, Oko R, Pelto-Huikko M, Spyrou G. Characterization of Sptrx, a novel member of the thioredoxin family specifically expressed in human spermatozoa. J Biol Chem. 2001; 276: 31567–31574.

Holmgren A. Thioredoxin and glutaredoxin systems. J Biol Chem. 1989; 264: 13963–13966.

Martin JL. Thioredoxin–a fold for all reasons. Structure. 1995; 3: 245–250.

Eklund H, Gleason FK, Holmgren A. Structural and functional relations among thioredoxins of different species. Proteins. 1991; 11: 13–28.

Qin J, Clore GM, Gronenborn AM. The high-resolution three-dimensional solution structures of the oxidized and reduced states of human thioredoxin. Structure. 1994; 2: 503–522.

Holmgren A. Thioredoxin structure and mechanism: conformational changes on oxidation of the active-site sulfhydryls to a disulfide. Structure. 1995; 3: 239–243.

Liu H, Nishitoh H, Ichijo H, Kyriakis JM. Activation of apoptosis signal-regulating kinase 1 (ASK1) by tumor necrosis factor receptor-associated factor 2 requires prior dissociation of the ASK1 inhibitor thioredoxin. Mol Cell Biol. 2000; 20: 2198–2208.

Fernandez Pujol B, Lucibello FC, Gehling UM, Lindemann K, Weidner N, Zuzarte ML, Adamkiewicz J, Elsasser HP, Muller R, Havemann K. Endothelial-like cells derived from human CD14 positive monocytes. Differentiation. 2000; 65: 287–300.

Tanaka T, Nakamura H, Nishiyama A, Hosoi F, Masutani H, Wada H, Yodoi J. Redox Regulation by Thioredoxin Superfamily; Protection Against Oxidative Stress and Aging. Free Radic Res. 2001; 33: 851–855.

Malik G, Gorbounov N, Das S, Gurusamy N, Otani H, Maulik N, Goswami S, Das DK. Ischemic preconditioning triggers nuclear translocation of thioredoxin and its interaction with Ref-1 potentiating a survival signal through the PI-3-kinase-Akt pathway. Antioxid Redox Signal. 2006; 8: 2101–2109.

Haendeler J, Hoffmann J, Tischler V, Berk BC, Zeiher AM, Dimmeler S. Redox regulatory and anti-apoptotic functions of thioredoxin depend on S-nitrosylation at cysteine 69. Nat Cell Biol. 2002; 4: 743–749.

Haendeler J, Tischler V, Hoffmann J, Zeiher AM, Dimmeler S. Low doses of reactive oxygen species protect endothelial cells from apoptosis by increasing thioredoxin-1 expression. FEBS Lett. 2004; 577: 427–433.

Badorff C, Brandes RP, Popp R, Rupp S, Urbich C, Aicher A, Fleming I, Busse R, Zeiher AM, Dimmeler S. Transdifferentiation of blood-derived human adult endothelial progenitor cells into functionally active cardiomyocytes. Circulation. 2003; 107: 1024–1032.

Haendeler J, Hoffmann J, Brandes RP, Zeiher AM, Dimmeler S. Hydrogen peroxide triggers nuclear export of telomerase reverse transcriptase via Src kinase family-dependent phosphorylation of tyrosine 707. Mol Cell Biol. 2003; 23: 4598–4610.

Haendeler J. Thioredoxin-1 and posttranslational modifications. Antioxid Redox Signal. 2006; 8: 1723–1728.

Yamamoto M, Yang G, Hong C, Liu J, Holle E, Yu X, Wagner T, Vatner SF, Sadoshima J. Inhibition of endogenous thioredoxin in the heart increases oxidative stress and cardiac hypertrophy. J Clin Invest. 2003; 112: 1395–1406.

Hirota K, Murata M, Sachi Y, Nakamura H, Takeuchi J, Mori K, Yodoi J. Distinct roles of thioredoxin in the cytoplasm and in the nucleus. A two-step mechanism of redox regulation of transcription factor NF-kappaB. J Biol Chem. 1999; 274: 27891–27897.

Tao L, Gao E, Bryan NS, Qu Y, Liu HR, Hu A, Christopher TA, Lopez BL, Yodoi J, Koch WJ, Feelisch M, Ma XL. Cardioprotective effects of thioredoxin in myocardial ischemia and reperfusion: role of S-nitrosation . Proc Natl Acad Sci U S A. 2004; 101: 11471–11476.

Heeneman S, Haendeler J, Saito Y, Ishida M, Berk BC. Angiotensin II induces transactivation of two different populations of the platelet-derived growth factor beta receptor. Key role for the p66 adaptor protein Shc. J Biol Chem. 2000; 275: 15926–15932.

Ushio-Fukai M, Griendling KK, Becker PL, Hilenski L, Halleran S, Alexander RW. Epidermal growth factor receptor transactivation by angiotensin II requires reactive oxygen species in vascular smooth muscle cells. Arterioscler Thromb Vasc Biol. 2001; 21: 489–495.

Li PF, Dietz R, von Harsdorf R. Reactive oxygen species induce apoptosis of vascular smooth muscle cell. FEBS Lett. 1997; 404: 249–252.

Yasuda M, Ohzeki Y, Shimizu S, Naito S, Ohtsuru A, Yamamoto T, Kuroiwa Y. Stimulation of in vitro angiogenesis by hydrogen peroxide and the relation with ETS-1 in endothelial cells. Life Sci. 1999; 64: 249–258.

Kim YC, Yamaguchi Y, Kondo N, Masutani H, Yodoi J. Thioredoxin-dependent redox regulation of the antioxidant responsive element (ARE) in electrophile response. Oncogene. 2003; 22: 1860–1865.

Xu L, Massague J. Nucleocytoplasmic shuttling of signal transducers. Nat Rev Mol Cell Biol. 2004; 5: 209–219.

Cao Z, Zhu H, Zhang L, Zhao X, Zweier JL, Li Y. Antioxidants and phase 2 enzymes in cardiomyocytes: Chemical inducibility and chemoprotection against oxidant and simulated ischemia-reperfusion injury. Exp Biol Med (Maywood). 2006; 231: 1353–1364.

Zhao X, Fan Y, Shen J, Wu Y, Yin Z. Human glutathione S-transferase P1 suppresses MEKK1-mediated apoptosis by regulating MEKK1 kinase activity in HEK293 cells. Mol Cells. 2006; 21: 395–400.


作者单位:IUF (Institut fuer Umweltmedizinische Forschung) at the University of Duesseldorf gGmbH (P.S., J.H.), Duesseldorf, Germany; the Department of Physiology (R.P.), University of Frankfurt, Germany; the Department of Molecular Cardiology (B.W.), University of Giessen, Germany; and the Department of Mole

作者: Peter Schroeder; Ruediger Popp; Barbara Wiegand; J
医学百科App—中西医基础知识学习工具
  • 相关内容
  • 近期更新
  • 热文榜
  • 医学百科App—健康测试工具