Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 235
Filter
1.
Open Neurol J ; 11: 48-58, 2017.
Article in English | MEDLINE | ID: mdl-29290835

ABSTRACT

BACKGROUND: Despite increased neuronal death, senile plaques, and neurofibrillary tangles observed in patients suffering from Alzheimer's disease (AD), the detailed mechanism of cell death in AD is still poorly understood. METHOD: We hypothesized that p38 kinase activates and then phosphorylates Bax, leading to its translocation to mitochondria in AD brains compared to controls. The aim of this study was to investigate the role of p38 kinase in phosphorylation and sub-cellular localization of pro-apoptotic Bax in the frontal cortex of the brains from AD and control subjects. Increased oxidative stress in AD individuals compared to control was evaluated by measuring the levels of carbonylated proteins and oxidized peroxiredoxin, an antioxidant enzyme. The relative amounts of p38 kinase and phospho-Bax in mitochondria in AD brains and controls were determined by immunoblot analysis using the respective antibody against each protein following immunoprecipitation. RESULTS: Our results showed that the levels of oxidized peroxiredoxin-SO3 and carbonylated proteins are significantly elevated in AD brains compared to controls, demonstrating the increased oxidative stress. CONCLUSION: The amount of phospho-p38 kinase is increased in AD brains and the activated p38 kinase appears to phosphorylate Thr residue(s) of Bax, which leads to its mitochondrial translocation, contributing to apoptosis and ultimately, neurodegeneration.

2.
Kidney Int Suppl ; (106): S3-8, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17653208

ABSTRACT

Peroxiredoxin (Prx) is a family of bifunctional proteins that exhibit peroxidase and chaperone activities. Prx proteins contain a conserved Cys residue that undergoes a redox change between thiol and disulfide states. 2-Cys Prx enzymes, a subgroup of Prx family, are intrinsically susceptible to reversible hyperoxidation to cysteine sulfinic acid during catalysis. Cysteine hyperoxidation of Prx was shown to result in loss of peroxidase activity and a concomitant gain of chaperone activity. Reduction of sulfinic Prx enzymes, the first known biological example of such a reaction, is catalyzed by sulfiredoxin (Srx) in the presence of ATP. Srx appears to exist solely to support the reversible sulfinic modification of 2-Cys Prx enzymes. Srx specifically binds to 2-Cys Prx enzymes by recognizing several critical surface-exposed residues of the Prxs, and transfer the gamma-phosphate of ATP to their sulfinic moiety, using its conserved cysteine as the phosphate carrier. The resulting sulfinic phosphoryl ester is reduced to cysteine after oxidation of four thiol equivalents.


Subject(s)
Oxidoreductases/physiology , Peroxidases/physiology , Cysteine/analogs & derivatives , Cysteine/metabolism , Diabetic Angiopathies/metabolism , Diabetic Angiopathies/physiopathology , Humans , Hydrogen Peroxide/metabolism , Molecular Chaperones/physiology , Oxidation-Reduction , Oxidoreductases Acting on Sulfur Group Donors , Peroxiredoxins
3.
J Pathol ; 196(3): 316-23, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11857495

ABSTRACT

Peroxiredoxins (Prxs) are a recently characterized group of thiol-containing proteins with efficient antioxidant capacity, capable of consuming hydrogen peroxide in living cells. Altogether six distinct Prxs have been characterized in mammalian tissues. Their expression was investigated in histological samples of mesothelioma and in cell lines established from the tumours of mesothelioma patients. Four cases with histopathologically healthy pleura from non-smokers were used as controls. Healthy pleural mesothelium was negative or very weakly positive for all Prxs. In mesothelioma, the most prominent reactivity was observed with Prxs I, II, V, and VI. Prx I was highly or moderately expressed in 25/36 cases, the corresponding figures for Prxs II-VI being 27/36 (Prx II), 13/36 (Prx III), 2/36 (Prx IV), 24/36 (Prx V), and 30/36 (Prx VI). Positive staining was observed both in the cytosolic and the nuclear compartment, with the exception of Prx III, which showed no nuclear reactivity. The staining pattern of Prxs III and V was granular. Immunoelectron microscopic localization of Prxs was in accordance with the immunohistochemical findings, showing diffuse cytoplasmic localization for Prxs I, II, IV, and VI and distinct mitochondrial labelling for Prxs III and V. There was no significant association between the extent of staining and different Prxs. It appeared that Prxs may not have prognostic significance, but being prominently expressed in most mesotheliomas these proteins, at least in theory, may play a role in the primary drug resistance of this disease.


Subject(s)
Antioxidants/metabolism , Mesothelioma/metabolism , Peroxidases/metabolism , Pleural Neoplasms/metabolism , Adult , Aged , Antioxidants/analysis , Apoptosis , Case-Control Studies , Cell Nucleus/chemistry , Cytosol/chemistry , Drug Resistance, Neoplasm , Female , Humans , Immunohistochemistry/methods , Male , Mesothelioma/pathology , Microscopy, Immunoelectron , Middle Aged , Mitochondria/chemistry , Peroxidases/analysis , Peroxiredoxin VI , Peroxiredoxins , Pleura/pathology , Pleural Neoplasms/pathology , Tumor Cells, Cultured
4.
Thorax ; 57(2): 157-64, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11828047

ABSTRACT

BACKGROUND: Six proteins of the peroxiredoxin (Prx) family have recently been characterised which have the capacity to decompose hydrogen peroxide in vivo and in vitro. These proteins may have an important role in the protection of human lung against endogenous and exogenous oxidant stress. However, the expression and distribution of these proteins in healthy human lung and diseased lung tissue is unknown. METHODS: The cell specific expression of Prxs in healthy lung tissue from four non-smokers and in parenchymal tissue from 10 subjects with pulmonary sarcoidosis was investigated by immunohistochemistry, and expression of these proteins in various cultured lung cells and cells of bronchoalveolar lavage (BAL) fluid of controls and patients with sarcoidosis was assessed by Western blot analysis. RESULTS: All six Prxs could be synthesised in cultured human lung cells. The bronchial epithelium showed moderate to high expression of Prxs I, III, V and VI, the alveolar epithelium expressed mainly Prxs V and VI, and alveolar macrophages expressed mainly Prxs I and III. Granulomas of subjects with sarcoidosis expressed mainly Prxs I and III. Samples of BAL fluid from controls and from subjects with sarcoidosis had very similar findings, except that Prxs II and III had a tendency for increased immunoreactivity in sarcoidosis tissue. CONCLUSIONS: Prxs I, III, V, and VI, in particular, have prominent and cell specific expression in human lung tissue. High expression of Prxs I and III in granulomas and alveolar macrophages of sarcoidosis parenchyma may have a significant effect on the oxidant burden and the progression of lung injury in this disease.


Subject(s)
Bronchi/enzymology , Peroxidases/metabolism , Sarcoidosis, Pulmonary/enzymology , Biopsy/methods , Blotting, Western , Bronchoalveolar Lavage Fluid/chemistry , Cells, Cultured , Epithelial Cells/enzymology , Humans , Immunohistochemistry , Peroxiredoxin VI , Peroxiredoxins
5.
Am J Physiol Cell Physiol ; 281(3): C1046-58, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11502583

ABSTRACT

The actin-regulatory protein villin is tyrosine phosphorylated and associates with phospholipase C-gamma(1) (PLC-gamma(1)) in the brush border of intestinal epithelial cells. To study the mechanism of villin-associated PLC-gamma(1) activation, we reconstituted in vitro the tyrosine phosphorylation of villin and its association with PLC-gamma(1). Recombinant villin was phosphorylated in vitro by the nonreceptor tyrosine kinase c-src or by expression in the TKX1 competent cells that carry an inducible tyrosine kinase gene. Using in vitro binding assays, we demonstrated that tyrosine-phosphorylated villin associates with the COOH-terminal Src homology 2 (SH2) domain of PLC-gamma(1). The catalytic activity of PLC-gamma(1) was inhibited by villin in a dose-dependent manner with half-maximal inhibition at a concentration of 12.4 microM. Villin inhibited PLC-gamma(1) activity by sequestering the substrate phosphatidylinositol 4,5-bisphosphate (PIP(2)), since increasing concentrations of PIP(2) reversed the inhibitory effects of villin on PLC activity. The inhibition of PLC-gamma(1) activity by villin was reversed by the tyrosine phosphorylation of villin. Further, we demonstrated that tyrosine phosphorylation of villin abolished villin's ability to associate with PIP(2). In conclusion, tyrosine-phosphorylated villin associates with the COOH-terminal SH2 domain of PLC-gamma(1) and activates PLC-gamma(1) catalytic activity. Villin regulates PLC-gamma(1) activity by modifying its own ability to bind PIP(2). This study provides biochemical proof of the functional relevance of tyrosine phosphorylation of villin and identifies the molecular mechanisms involved in the activation of PLC-gamma(1) by villin.


Subject(s)
Actins/metabolism , Carrier Proteins/metabolism , Isoenzymes/metabolism , Microfilament Proteins/metabolism , Type C Phospholipases/metabolism , CSK Tyrosine-Protein Kinase , Calcium-Binding Proteins/metabolism , Carrier Proteins/genetics , Enzyme Activation , Gene Expression Regulation/drug effects , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , HeLa Cells , Humans , Isoenzymes/genetics , Isopropyl Thiogalactoside/pharmacology , Kinetics , Mercaptoethanol/pharmacology , Microfilament Proteins/genetics , Models, Biological , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phospholipase C gamma , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/metabolism , Transfection , Type C Phospholipases/genetics , src Homology Domains , src-Family Kinases
6.
Annu Rev Biochem ; 70: 281-312, 2001.
Article in English | MEDLINE | ID: mdl-11395409

ABSTRACT

Eleven distinct isoforms of phosphoinositide-specific phospholipase C (PLC), which are grouped into four subfamilies (beta, gamma, delta, and epsilon), have been identified in mammals. These isozymes catalyze the hydrolysis of phosphatidylinositol 4,5-bisphosphate [PtdIns(4,5)P2] to inositol 1,4,5-trisphosphate and diacylglycerol in response to the activation of more than 100 different cell surface receptors. All PLC isoforms contain X and Y domains, which form the catalytic core, as well as various combinations of regulatory domains that are common to many other signaling proteins. These regulatory domains serve to target PLC isozymes to the vicinity of their substrate or activators through protein-protein or protein-lipid interactions. These domains (with their binding partners in parentheses or brackets) include the pleckstrin homology (PH) domain [PtdIns(3)P, beta gamma subunits of G proteins] and the COOH-terminal region including the C2 domain (GTP-bound alpha subunit of Gq) of PLC-beta; the PH domain [PtdIns(3,4,5)P3] and Src homology 2 domain [tyrosine-phosphorylated proteins, PtdIns(3,4,5)P3] of PLC-gamma; the PH domain [PtdIns(4,5)P2] and C2 domain (Ca2+) of PLC-delta; and the Ras binding domain (GTP-bound Ras) of PLC-epsilon. The presence of distinct regulatory domains in PLC isoforms renders them susceptible to different modes of activation. Given that the partners that interact with these regulatory domains of PLC isozymes are generated or eliminated in specific regions of the cell in response to changes in receptor status, the activation and deactivation of each PLC isoform are likely highly regulated processes.


Subject(s)
Phosphatidylinositols/metabolism , Type C Phospholipases/chemistry , Type C Phospholipases/metabolism , Cell Nucleus , Enzyme Activation , GTP-Binding Proteins/chemistry , GTP-Binding Proteins/metabolism , Isoenzymes/chemistry , Isoenzymes/metabolism , Protein Subunits , Protein-Tyrosine Kinases/metabolism , Signal Transduction
8.
Am J Physiol Lung Cell Mol Physiol ; 280(6): L1212-7, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11350800

ABSTRACT

Peroxiredoxin I (Prx I) and peroxiredoxin II (Prx II) are found in abundance in the cytoplasm of cells and catalyze the reduction of hydrogen peroxide with the use of electrons provided by thioredoxin. Here we examined Prx I and Prx II expression in rat lung during perinatal development and in response to hyperoxia. Prx I protein increased during late gestation and after birth fell to adult levels; conversely, Prx I mRNA increased after birth. Prx II protein concentration was unchanged in the perinatal period, but Prx II mRNA increased after birth. In response to hyperoxia begun on postnatal day 4, there was no change in Prx II expression; however, Prx I mRNA, protein, and enzymatic activity increased significantly. These data show that 1) Prx I and Prx II are developmentally regulated at the level of translational efficiency and 2) Prx I, but not Prx II, is inducible and is upregulated during the late-gestational preparation for the oxidative stress experienced by the lung at birth and during exposure to hyperoxia in the neonatal period.


Subject(s)
Aging/metabolism , Gene Expression Regulation, Developmental/physiology , Hyperoxia/metabolism , Lung/enzymology , Peroxidases/metabolism , Animals , Animals, Newborn , Enzyme Induction/physiology , Isoenzymes/genetics , Isoenzymes/metabolism , Lung/growth & development , Male , Oxidative Stress/physiology , Peroxidases/genetics , Peroxiredoxins , RNA, Messenger/metabolism , Rats , Up-Regulation
9.
J Biol Chem ; 276(26): 24293-300, 2001 Jun 29.
Article in English | MEDLINE | ID: mdl-11294858

ABSTRACT

Regulator of G-protein signaling 3 (RGS3) enhances the intrinsic rate at which Galpha(i) and Galpha(q) hydrolyze GTP to GDP, thereby limiting the duration in which GTP-Galpha(i) and GTP-Galpha(q) can activate effectors. Since GDP-Galpha subunits rapidly combine with free Gbetagamma subunits to reform inactive heterotrimeric G-proteins, RGS3 and other RGS proteins may also reduce the amount of Gbetagamma subunits available for effector interactions. Although RGS6, RGS7, and RGS11 bind Gbeta(5) in the absence of a Ggamma subunit, RGS proteins are not known to directly influence Gbetagamma signaling. Here we show that RGS3 binds Gbeta(1)gamma(2) subunits and limits their ability to trigger the production of inositol phosphates and the activation of Akt and mitogen-activated protein kinase. Co-expression of RGS3 with Gbeta(1)gamma(2) inhibits Gbeta(1)gamma(2)-induced inositol phosphate production and Akt activation in COS-7 cells and mitogen-activated protein kinase activation in HEK 293 cells. The inhibition of Gbeta(1)gamma(2) signaling does not require an intact RGS domain but depends upon two regions in RGS3 located between acids 313 and 390 and between 391 and 458. Several other RGS proteins do not affect Gbeta(1)gamma(2) signaling in these assays. Consistent with the in vivo results, RGS3 inhibits Gbetagamma-mediated activation of phospholipase Cbeta in vitro. Thus, RGS3 may limit Gbetagamma signaling not only by virtue of its GTPase-activating protein activity for Galpha subunits, but also by directly interfering with the activation of effectors.


Subject(s)
GTP-Binding Protein beta Subunits , GTP-Binding Protein gamma Subunits , GTP-Binding Proteins/antagonists & inhibitors , GTPase-Activating Proteins , Heterotrimeric GTP-Binding Proteins/antagonists & inhibitors , Inositol Phosphates/biosynthesis , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/antagonists & inhibitors , RGS Proteins/physiology , Animals , Binding Sites , COS Cells , Cell Line , Enzyme Activation , Humans , Isoenzymes/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3 , Phospholipase C beta , Precipitin Tests , Proto-Oncogene Proteins c-akt , RGS Proteins/chemistry , RGS Proteins/genetics , Transfection , Type C Phospholipases/antagonists & inhibitors
10.
Free Radic Biol Med ; 30(4): 412-24, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11182297

ABSTRACT

Components of the thioredoxin system were localized in normal rat kidney using immunoperoxidase techniques at the light microscopic level and immunogold techniques at the ultrastructural level. Results from both methods were similar. Thioredoxin, thioredoxin reductases, and peroxiredoxins showed cell-type-specific localization, with the same cell types (proximal and distal tubular epithelial, papillary collecting duct, and transitional epithelial cells) previously identified as having high amounts of antioxidant enzyme immunoreactive proteins and oxidative damage products also having high levels of proteins of the thioredoxin system. In addition, peroxiredoxins II and IV were found in high levels in the cytoplasm of red blood cells, identified in kidney blood vessels. While thioredoxin and thioredoxin reductase 1 were found in all subcellular locations in kidney cells, thioredoxin reductase 2 was found predominantly in mitochondria. Thioredoxin reductase 1 was identified in rat plasma, suggesting it is a secreted protein. Peroxiredoxins often had specific subcellular locations, with peroxiredoxins III and V found in mitochondria and peroxiredoxin IV found in lysosomes. Our results emphasize the complex nature of the thioredoxin system, demonstrating unique cell-type and organelle specificity.


Subject(s)
Peroxidases/analysis , Rats , Thioredoxin-Disulfide Reductase/analysis , Thioredoxins/analysis , Animals , Immunoenzyme Techniques , Kidney/chemistry , Kidney/ultrastructure , Kidney Glomerulus/chemistry , Kidney Tubules, Proximal/chemistry , Lysosomes/chemistry , Male , Mitochondria/chemistry , Peroxiredoxins , Rats, Sprague-Dawley , Tissue Distribution
12.
J Immunol ; 165(4): 2190-7, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10925306

ABSTRACT

Stimulation of human lung fibroblast cells with TGF-beta1 resulted in a transient burst of reactive oxygen species with maximal increase at 5 min after treatment. This reactive oxygen species increase was inhibited by the antioxidant, N-acetyl-l -cysteine (NAC). TGF-beta1 treatment stimulated IL-6 gene expression and protein synthesis in human lung fibroblast cells. Antioxidants including NAC, glutathione, and catalase reduced TGF-beta1-induced IL-6 gene expression, and direct H2O2 treatment induced IL-6 expression in a dose-dependent manner. NAC also reduced TGF-beta1-induced AP-1 binding activity, which is involved in IL-6 gene expression. It has been reported that Ca2+ influx is stimulated by TGF-beta1 treatment. EGTA suppressed TGF-beta1- or H2O2-induced IL-6 expression, and ionomycin increased IL-6 expression, with simultaneously modulating AP-1 activity in the same pattern. PD98059, an inhibitor of mitogen-activated protein kinase (MAPK) kinase/extracellular signal-related kinase kinase 1, suppressed TGF-beta1- or H2O2-induced IL-6 and AP-1 activation. In addition, TGF-beta1 or H2O2 increased MAPK activity which was reduced by EGTA and NAC, suggesting that MAPK is involved in TGF-beta1-induced IL-6 expression. Taken together, these results indicate that TGF-beta1 induces a transient increase of intracellular H2O2 production, which regulates downstream events such as Ca2+ influx, MAPK, and AP-1 activation and IL-6 gene expression.


Subject(s)
Calcium/physiology , Fibroblasts/metabolism , Hydrogen Peroxide/metabolism , Interleukin-6/biosynthesis , Lung/metabolism , Signal Transduction/immunology , Transforming Growth Factor beta/physiology , Egtazic Acid/pharmacology , Enzyme Activation/drug effects , Enzyme Activation/immunology , Enzyme Inhibitors/pharmacology , Fibroblasts/drug effects , Fibroblasts/immunology , Flavonoids/pharmacology , Humans , Hydrogen Peroxide/pharmacology , Interleukin-6/genetics , Intracellular Fluid/immunology , Intracellular Fluid/metabolism , Lung/cytology , Lung/drug effects , Lung/immunology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Protein Binding/drug effects , Protein Binding/immunology , RNA, Messenger/biosynthesis , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Transcription Factor AP-1/metabolism , Tumor Cells, Cultured
13.
Anal Biochem ; 283(2): 214-21, 2000 Aug 01.
Article in English | MEDLINE | ID: mdl-10906242

ABSTRACT

A procedure for detecting proteins that contain H(2)O(2)-sensitive cysteine (or selenocysteine) residues was developed as a means with which to study protein oxidation by H(2)O(2) in cells. The procedure is based on the facts that H(2)O(2) and biotin-conjugated iodoacetamide (BIAM) selectively and competitively react with cysteine residues that exhibit a low pK(a), and that the decrease in the labeling of cell lysate proteins with BIAM caused by prior exposure of cells to H(2)O(2) or to an agent that induces H(2)O(2) production can be monitored by streptavidin blot analysis. This procedure was applied to rat pheochromocytoma PC12 cells directly treated with H(2)O(2), mouse hippocampal HT22 cells in which H(2)O(2) production was induced by glutamate, and human erythroleukemia K562 cells in which H(2)O(2) production was induced by phorbol myristate acetate. It revealed that several cell proteins contain cysteine or selenocysteine residues that are selectively oxidized by H(2)O(2). Three of these H(2)O(2)-sensitive proteins were identified as a member of the protein disulfide isomerase family, thioredoxin reductase, and creatine kinase, all of which were previously known to contain at least one reactive cysteine or selenocysteine at their catalytic sites. This procedure should thus prove useful for the identification of proteins that are oxidized by H(2)O(2) generated in response to a variety of extracellular agents.


Subject(s)
Biotin/analogs & derivatives , Cysteine/analysis , Hydrogen Peroxide/chemistry , Proteins/analysis , Alkylation , Amino Acid Sequence , Animals , Biotin/chemistry , Creatine Kinase/analysis , Creatine Kinase/chemistry , Cysteine/chemistry , Ethylenediamines/chemistry , Hydrogen-Ion Concentration , Molecular Sequence Data , Oxidation-Reduction , PC12 Cells , Proteins/chemistry , Rabbits , Rats , Streptavidin/chemistry
14.
J Immunol ; 164(12): 6287-95, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10843682

ABSTRACT

As a result of identifying the regulatory proteins of thioredoxin (TRX), a murine homologue for human vitamin D3 up-regulated protein 1 (VDUP1) was identified from a yeast two-hybrid screen. Cotransfection into 293 cells and precipitation assays confirmed that mouse VDUP1 (mVDUP1) bound to TRX, but it failed to bind to a Cys32 and Cys35 mutant TRX, suggesting the redox-active site is critical for binding. mVDUP1 was ubiquitously expressed in various tissues and located in the cytoplasm. Biochemical analysis showed that mVDUP1 inhibited the insulin-reducing activity of TRX. When cells were treated with various stress stimuli such as H2O2 and heat shock, mVDUP1 was significantly induced. TRX is known to interact with other proteins such as proliferation-associated gene and apoptosis signal-regulating kinase 1. Coexpression of mVDUP1 interfered with the interaction between TRX and proliferation-associated gene or TRX and ASK-1, suggesting its roles in cell proliferation and oxidative stress. To investigate the roles of mVDUP1 in oxidative stress, mVDUP1 was overexpressed in NIH 3T3 cells. When cells were exposed to stress, cell proliferation was declined with elevated apoptotic cell death compared with control cells. In addition, c-Jun N-terminal kinase activation and IL-6 expression were elevated. Taken together, these results demonstrate that mVDUP1 functions as an oxidative stress mediator by inhibiting TRX activity.


Subject(s)
Carrier Proteins/physiology , Cholecalciferol/physiology , Oxidative Stress/physiology , Peroxidases , Thioredoxins/antagonists & inhibitors , Thioredoxins/pharmacology , Up-Regulation/physiology , 3T3 Cells , Amino Acid Sequence , Animals , Binding, Competitive , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Cell Line , Gene Expression Regulation , Heat-Shock Proteins/antagonists & inhibitors , Heat-Shock Proteins/metabolism , Humans , Hydrogen Peroxide/pharmacology , Mice , Molecular Sequence Data , Organ Specificity , Oxidative Stress/drug effects , Peroxiredoxins , Subcellular Fractions/metabolism , Thioredoxins/metabolism , Up-Regulation/drug effects
16.
FEBS Lett ; 473(2): 177-82, 2000 May 12.
Article in English | MEDLINE | ID: mdl-10812070

ABSTRACT

The involvement of reactive oxygen species in Ca(2+)-induced mitochondrial membrane permeabilization and cell viability was studied using yeast cells in which the thioredoxin peroxidase (TPx) gene was disrupted and/or catalase was inhibited by 3-amino-1,2, 4-triazole (ATZ) treatment. Wild-type Saccharomyces cerevisiae cells were very resistant to Ca(2+) and inorganic phosphate or t-butyl hydroperoxide-induced mitochondrial membrane permeabilization, but suffered an immediate decrease in mitochondrial membrane potential when treated with Ca(2+) and the dithiol binding reagent phenylarsine oxide. In contrast, S. cerevisiae spheroblasts lacking the TPx gene and/or treated with ATZ suffered a decrease in mitochondrial membrane potential, generated higher amounts of hydrogen peroxide and had decreased viability under these conditions. In all cases, the decrease in mitochondrial membrane potential could be inhibited by ethylene glycol-bis(beta-aminoethyl ether) N,N, N',N'-tetraacetic acid, dithiothreitol or ADP, but not by cyclosporin A. We conclude that TPx and catalase act together, maintaining cell viability and protecting S. cerevisiae mitochondria against Ca(2+)-promoted membrane permeabilization, which presents similar characteristics to mammalian permeability transition.


Subject(s)
Calcium/pharmacology , Catalase/physiology , Intracellular Membranes/physiology , Mitochondria/physiology , Neoplasm Proteins , Peroxidases/physiology , Saccharomyces cerevisiae/physiology , Adenosine Diphosphate/pharmacology , Amitrole/pharmacology , Calcium/metabolism , Catalase/antagonists & inhibitors , Cell Division/drug effects , Dithiothreitol/pharmacology , Egtazic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Membrane Potentials/drug effects , Mutation , Permeability/drug effects , Peroxidases/genetics , Peroxiredoxins , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics
17.
J Biol Chem ; 275(25): 18751-8, 2000 Jun 23.
Article in English | MEDLINE | ID: mdl-10764771

ABSTRACT

Two distinct proteins inhibiting phospholipase D (PLD) activity in rat brain cytosol were previously purified and identified as synaptojanin and AP180, which are specific to nerve terminals and associate with the clathrin coat. Two additional PLD-inhibitory proteins have now been purified and identified as the amphiphysins I and II, which forms a heterodimer that also associates with the clathrin coat. Bacterially expressed recombinant amphiphysins inhibited both PLD1 and PLD2 isozymes in vitro with a potency similar to that of brain amphiphysin (median inhibitory concentration of approximately 15 nm). Expressions of either amphiphysin in COS-7 cells reduced activity of endogenous PLD as well as exogenously expressed PLD1 and PLD2. Coprecipitation experiments suggested that the inhibitory effect of amphiphysins results from their direct interaction with PLDs. The NH(2) terminus of amphiphysin I was critical for both inhibition of and binding to PLD. Phosphatidic acid formed by signal-induced PLD is thought to be required for the assembly of clathrin-coated vesicles during endocytosis. Thus, the inhibition of PLD by amphiphysins, synaptojanin, and AP180 might play an important role in synaptic vesicle trafficking.


Subject(s)
Enzyme Inhibitors/pharmacology , Isoenzymes/antagonists & inhibitors , Nerve Tissue Proteins/pharmacology , Phospholipase D/antagonists & inhibitors , Amino Acid Sequence , Animals , COS Cells , Cell Line , Chromatography, DEAE-Cellulose , Chromatography, High Pressure Liquid , Clathrin/metabolism , Cricetinae , Endocytosis , Glutathione Transferase/chemistry , Molecular Sequence Data , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/isolation & purification , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacology
18.
J Biol Chem ; 275(27): 20346-54, 2000 Jul 07.
Article in English | MEDLINE | ID: mdl-10751410

ABSTRACT

Peroxidases of the peroxiredoxin (Prx) family contain a Cys residue that is preceded by a conserved sequence in the NH(2)-terminal region. A new type of mammalian Prx, designated PrxV, has now been identified as the result of a data base search with this conserved Cys-containing sequence. The 162-amino acid PrxV shares only approximately 10% sequence identity with previously identified mammalian Prx enzymes and contains Cys residues at positions 73 and 152 in addition to that (Cys(48)) corresponding to the conserved Cys. Analysis of mutant human PrxV proteins in which each of these three Cys residues was individually replaced with serine suggested that the sulfhydryl group of Cys(48) is the site of oxidation by peroxides and that oxidized Cys(48) reacts with the sulfhydryl group of Cys(152) to form an intramolecular disulfide linkage. The oxidized intermediate of PrxV is thus distinct from those of other Prx enzymes, which form either an intermolecular disulfide or a sulfenic acid intermediate. The disulfide formed by PrxV is reduced by thioredoxin but not by glutaredoxin or glutathione. Thus, PrxV mutants lacking Cys(48) or Cys(152) showed no detectable thioredoxin-dependent peroxidase activity, whereas mutation of Cys(73) had no effect on activity. Immunoblot analysis revealed that PrxV is widely expressed in rat tissues and cultured mammalian cells and is localized intracellularly to cytosol, mitochondria, and peroxisomes. The peroxidase function of PrxV in vivo was demonstrated by the observations that transient expression of the wild-type protein, but not that of the Cys(48) mutant, in NIH 3T3 cells inhibited H(2)O(2) accumulation and activation of c-Jun NH(2)-terminal kinase induced by tumor necrosis factor-alpha.


Subject(s)
Disulfides/chemistry , Oxidoreductases , Peroxidases/genetics , Amino Acid Sequence , Animals , Cell Line , Cloning, Molecular , Conserved Sequence , Cysteine/genetics , Enzyme Activation , Glutaredoxins , Humans , Hydrogen Peroxide/metabolism , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , Mutation , Peroxidases/chemistry , Peroxiredoxins , Protein Isoforms/metabolism , Proteins/metabolism , Recombinant Proteins/metabolism , Sequence Alignment , Thioredoxins/metabolism
19.
J Biol Chem ; 275(14): 10527-31, 2000 Apr 07.
Article in English | MEDLINE | ID: mdl-10744745

ABSTRACT

Autophosphorylation of the platelet-derived growth factor (PDGF) receptor triggers intracellular signaling cascades as a result of recruitment of Src homology 2 domain-containing enzymes, including phosphatidylinositol 3-kinase (PI3K), the GTPase-activating protein of Ras (GAP), the protein-tyrosine phosphatase SHP-2, and phospholipase C-gamma1 (PLC-gamma1), to specific phosphotyrosine residues. The roles of these various effectors in PDGF-induced generation of H(2)O(2) have now been investigated in HepG2 cells expressing various PDGF receptor mutants. These mutants included a kinase-deficient receptor and receptors in which various combinations of the tyrosine residues required for the binding of PI3K (Tyr(740) and Tyr(751)), GAP (Tyr(771)), SHP-2 (Tyr(1009)), or PLC-gamma1 (Tyr(1021)) were mutated to Phe. PDGF failed to increase H(2)O(2) production in cells expressing either the kinase-deficient mutant or a receptor in which the two Tyr residues required for the binding of PI3K were replaced by Phe. In contrast, PDGF-induced H(2)O(2) production in cells expressing a receptor in which the binding sites for GAP, SHP-2, and PLC-gamma1 were all mutated was slightly greater than that in cells expressing the wild-type receptor. Only the PI3K binding site was alone sufficient for PDGF-induced H(2)O(2) production. The effect of PDGF on H(2)O(2) generation was blocked by the PI3K inhibitors LY294002 and wortmannin or by overexpression of a dominant negative mutant of Rac1. These results suggest that a product of PI3K is required for PDGF-induced production of H(2)O(2) in nonphagocytic cells, and that Rac1 mediates signaling between the PI3K product and the putative NADPH oxidase.


Subject(s)
Hydrogen Peroxide/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Platelet-Derived Growth Factor/pharmacology , Receptor, Platelet-Derived Growth Factor beta/physiology , Binding Sites , Carcinoma, Hepatocellular , Enzyme Activation , Humans , Liver Neoplasms , Phosphorylation , Receptor, Platelet-Derived Growth Factor beta/chemistry , Receptor, Platelet-Derived Growth Factor beta/drug effects , Recombinant Proteins/chemistry , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Transfection , Tumor Cells, Cultured
20.
Proc Natl Acad Sci U S A ; 97(6): 2521-6, 2000 Mar 14.
Article in English | MEDLINE | ID: mdl-10688911

ABSTRACT

Mammalian cytosolic thioredoxin reductase (TrxR) has a redox center, consisting of Cys(59)/Cys(64) adjacent to the flavin ring of FAD and another center consisting of Cys(497)/selenocysteine (SeCys)(498) near the C terminus. We now show that the C-terminal Cys(497)-SH/SeCys(498)-Se(-) of NADPH-reduced enzyme, after anaerobic dialysis, was converted to a thioselenide on incubation with excess oxidized Trx (TrxS(2)) or H(2)O(2). The Cys(59)-SH/Cys(64)-SH pair also was oxidized to a disulfide. At lower concentrations of TrxS(2), the Cys(59)-SH/Cys(64)-SH center was still converted to a disulfide, presumably by reduction of the thioselenide to Cys(497)-SH/SeCys(498)-Se(-). Specific alkylation of SeCys(498) completely blocked the TrxS(2)-induced oxidation of Cys(59)-SH/Cys(64)-SH, and the alkylated enzyme had negligible NADPH-disulfide oxidoreductase activity. The effect of replacing SeCys(498) with Cys was determined by using a mutant form of human placental TrxR1 expressed in Escherichia coli. The NADPH-disulfide oxidoreductase activity of the purified Cys(497)/Cys(498) mutant enzyme was 6% or 11% of that of wild-type rat liver TrxR1 with 5, 5'-dithiobis(2-nitrobenzoic acid) or TrxS(2), respectively, as substrate. Disulfide formation induced by excess TrxS(2) in the mutant form was 12% of that of the wild type. Thus, SeCys has a critical redox function during the catalytic cycle, which is performed poorly by Cys.


Subject(s)
Cysteine/chemistry , Selenium/chemistry , Selenocysteine/chemistry , Sulfur/chemistry , Thioredoxin-Disulfide Reductase/chemistry , Thioredoxin-Disulfide Reductase/metabolism , Animals , Binding Sites , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Escherichia coli/metabolism , Humans , Liver/enzymology , Models, Chemical , Mutagenesis , Oxidation-Reduction , Placenta/enzymology , Protein Binding , Rats , Spectrophotometry , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...