Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Chem Res Toxicol ; 36(1): 23-31, 2023 01 16.
Article in English | MEDLINE | ID: mdl-36525601

ABSTRACT

Electrophilic quinones are produced during the combustion of gasoline in the atmosphere. Although these reactive species covalently bind to protein-based nucleophiles in cells, resulting in the formation of protein adducts involved in the modulation of redox signaling pathways and cytotoxicity, the extracellular regulation of quinones is not understood. In this study, incubation of 1,2-naphthoquinone (1,2-NQ) with the low-molecular-weight fraction of mouse plasma resulted in the consumption of cysteine (CysSH) in the plasma in a concentration-dependent manner. Covalent modification of albumin was markedly repressed by the addition of either the low-molecular-weight fraction of mouse plasma or CysSH, suggesting that CysSH protects by forming a conjugate with 1,2-NQ. Similar phenomena also occurred for other atmospheric quinones 1,4-NQ and 1,4-benzoquinone (1,4-BQ). The addition of cystine to a culture medium without amino acids enhanced the release of CysSH from A431 cells and blocked 1,2-NQ-mediated arylation of intracellular proteins, suggesting that 1,2-NQ interacts with extracellular CysSH. Liquid chromatography-tandem mass spectrometry analysis revealed that 1,2-NQ and 1,4-BQ undergoes nucleophilic attack by CysSH, yielding a 1,2-NQH2-SCys adduct and 1,4-BQH2-SCys adduct, respectively. Unlike 1,2-NQ and 1,4-BQ, the authentic 1,2-NQH2-SCys adduct and 1,4-BQH2-SCys adduct had little effect on the covalent modification of cellular proteins and viability of A431 cells. These results suggest that electrophilic quinones are readily trapped by CysSH released from A431 cells, forming less-toxic CysSH adducts and thereby repressing covalent modification of cellular proteins. These findings provide evidence for the existence of a "phase zero" reaction of electrophiles prior to their uptake by cells.


Subject(s)
Naphthoquinones , Quinones , Mice , Animals , Extracellular Space/metabolism , Naphthoquinones/chemistry , Proteins , Signal Transduction
2.
Sci Rep ; 7(1): 4814, 2017 07 06.
Article in English | MEDLINE | ID: mdl-28684787

ABSTRACT

Electrophiles can activate redox signal transduction pathways, through actions of effector molecules (e.g., kinases and transcription factors) and sensor proteins with low pKa thiols that are covalently modified. In this study, we investigated whether 1,4-naphthoquinone (1,4-NQ) could affect the phosphatase and tensin homolog (PTEN)-Akt signaling pathway and persulfides/polysulfides could modulate this adaptive response. Simultaneous exposure of primary mouse hepatocytes to Na2S4 and 1,4-NQ markedly decreased 1,4-NQ-mediated cell death and S-arylation of cellular proteins. Modification of cellular PTEN during exposure to 1,4-NQ was also blocked in the presence of Na2S4. 1,4-NQ, at up to 10 µM, increased phosphorylation of Akt and cAMP response element binding protein (CREB). However, at higher concentrations, 1,4-NQ inhibited phosphorylation of both proteins. These bell-shaped dose curves for Akt and CREB activation were right-shifted in cells treated with both 1,4-NQ and Na2S4. Incubation of 1,4-NQ with Na2S4 resulted in formation of 1,4-NQ-S-1,4-NQ-OH. Unlike 1,4-NQ, authentic 1,4-NQ-S-1,4-NQ-OH adduct had no cytotoxicity, covalent binding capability nor ability to activate PTEN-Akt signaling in cells. Our results suggested that polysulfides, such as Na2S4, can increase the threshold of 1,4-NQ for activating PTEN-Akt signaling and cytotoxicity by capturing this electrophile to form its sulfur adducts.


Subject(s)
Cyclic AMP Response Element-Binding Protein/genetics , Naphthoquinones/antagonists & inhibitors , PTEN Phosphohydrolase/genetics , Protective Agents/pharmacology , Proto-Oncogene Proteins c-akt/genetics , Sodium Compounds/pharmacology , Sulfides/pharmacology , Animals , Cell Death/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Female , Gene Expression Regulation , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Mice , Mice, Inbred C57BL , Naphthoquinones/toxicity , PTEN Phosphohydrolase/metabolism , Phosphorylation/drug effects , Primary Cell Culture , Protective Agents/chemistry , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Sodium Compounds/chemistry , Sulfides/chemistry
4.
Free Radic Biol Med ; 104: 118-128, 2017 03.
Article in English | MEDLINE | ID: mdl-28049024

ABSTRACT

The current consensus is that environmental electrophiles activate redox signal transduction pathways through covalent modification of sensor proteins with reactive thiol groups at low concentrations, while they cause cell damage at higher concentrations. We previously exposed human carcinoma A431 cells to the atmospheric electrophile 1,4-naphthoquinone (1,4-NQ) and found that heat shock protein 90 (HSP90), a negative regulator of heat shock factor 1 (HSF1), was a target of 1,4-NQ. In the study presented here, we determined whether 1,4-NQ activates HSF1. We also examined whether such redox signaling could be regulated by nucleophilic sulfur species. Exposure of A431 cells to 1,4-NQ covalently modified cellular HSP90, resulting in repression of the association between HSF1 with HSP90, thereby enhancing HSF1 translocation into the nuclei. Liquid chromatography-tandem mass spectrometry analysis with recombinant HSP90 revealed that the modifications site were Cys412 and Cys564. We found that HSF1 activation mediated by 1,4-NQ upregulated downstream genes, such as HSPA6. HSF1 knockdown accelerated 1,4-NQ-mediated cytotoxicity in the cells. While simultaneous treatment with reactive persulfide and polysulfide, Na2S2 and Na2S4, blocked 1,4-NQ-dependent protein modification and HSF1 activation in A431 cells, the knockdown of Cys persulfide producing enzymes cystathionine ß-synthase (CBS) and/or cystathionine γ-lyase (CSE) enhanced these phenomena. 1,4-NQ-thiol adduct and 1,4-NQ-S-1,4-NQ adduct were produced during the enzymatic reaction of recombinant CSE in the presence of 1,4-NQ. The results suggest that activation of the HSP90-HSF1 signal transduction pathway mediated by 1,4-NQ protects cells against 1,4-NQ and that per/polysulfides can diminish the reactivity of 1,4-NQ by forming sulfur adducts.


Subject(s)
HSP70 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Heat Shock Transcription Factors/genetics , Naphthoquinones/administration & dosage , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/metabolism , Chromatography, Liquid , Cysteine/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation/drug effects , HSP90 Heat-Shock Proteins/genetics , Humans , Naphthoquinones/metabolism , Oxidation-Reduction , Signal Transduction/drug effects , Sulfhydryl Compounds/metabolism , Sulfur/metabolism , Tandem Mass Spectrometry
5.
J Toxicol Sci ; 37(5): 891-8, 2012.
Article in English | MEDLINE | ID: mdl-23037999

ABSTRACT

While metabolic activation of naphthalene, yielding 1,2-naphthoquinone (1,2-NQ) and 1,4-NQ that can covalently bind to cellular proteins, has been recognized to be associated with its toxicity, the current consensus is that such electrophile-mediated covalent modification of sensor proteins with thiolate ions is also involved in activation of cellular signal transduction pathways for cellular protection against reactive materials. In the present study, we developed an immunochemical assay to detect cellular proteins adducted by 1,4-NQ. Dot blot analysis indicated that the antibody prepared against 1,4-NQ recognized the naphthalene moiety with the para-dicarbonyl group, rather than with the ortho-dicarbonyl group. Furthermore, little cross-reactivity of para-quinones with either a different number of aromatic rings (n = 1) or substituent groups was observed. With this specific antibody against 1,4-NQ, we identified nine target proteins of 1,4-NQ following exposure of human epithelial carcinoma cell line A431 to 1,4-NQ. Among them, heat shock protein 90 (HSP90) and HSP70 are of interest because covalent modification of these chaperones causes activation of heat shock factor-1, which plays a role in the cellular response against electrophiles such as 1,4-NQ. Thus, our method, which does not use radiolabeled compounds, would be applicable for exploring activation of electrophilic signal transduction pathways coupled to covalent modification of sensor proteins during exposure to naphthalene as well as 1,4-NQ.


Subject(s)
Naphthoquinones/toxicity , Proteins/metabolism , Animals , Cell Line, Tumor , Electrophoresis, Polyacrylamide Gel , Female , Humans , Immunochemistry , Naphthoquinones/immunology , Protein Binding , Proteins/immunology , Rabbits , Signal Transduction/drug effects
6.
Free Radic Biol Med ; 51(11): 2082-9, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21963991

ABSTRACT

1,2-Naphthoquinone (1,2-NQ) is electrophilic, and forms covalent bonds with protein thiols, but its two-electron reduction product 1,2-dihydroxynaphthalene (1,2-NQH(2)) is not, so enzymes catalyzing the reduction with reduced pyridine nucleotides as cofactors could protect cells from electrophile-based chemical insults. To assess this possibility, we examined proteins isolated from the 9000g supernatant from mouse liver for 1,2-NQ reductase activity using an HPLC assay procedure for the hydroquinone of 1,2-NQ and Cibacron Blue 3GA column chromatography and Western blot analysis with specific antibody to determine 1,2-NQ-bound proteins. Among the proteins with high affinities for pyridine nucleotides that also inhibited 1,2-NQ-protein adduct formation in the presence of NADH, a 37-kDa protein was found and identified as glyceraldehyde-3-phosphate dehydrogenase (GAPDH). Using recombinant human GAPDH, we found that this glycolytic enzyme indeed catalyzes the two-electron reduction of 1,2-NQ accompanied by extensive NADH consumption under 20% oxygen conditions. When either 1,2-NQH(2) or 1,2-NQ was incubated with GAPDH in the presence of NADH, minimal covalent bonding to the enzyme occurred compared to that in its absence. These results indicate that GAPDH can inhibit 1,2-NQ-based electrophilic protein modification by conversion to the nonelectrophilic 1,2-NQH(2) via an NADH-dependent process.


Subject(s)
Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , NAD(P)H Dehydrogenase (Quinone)/metabolism , Naphthoquinones/metabolism , Amino Acid Sequence , Animals , Biocatalysis , Glyceraldehyde-3-Phosphate Dehydrogenases/isolation & purification , Humans , Male , Mice , Mice, Inbred ICR , Molecular Sequence Data , NAD/metabolism , NAD(P)H Dehydrogenase (Quinone)/isolation & purification , Naphthoquinones/chemistry
7.
Chem Res Toxicol ; 24(11): 1836-44, 2011 Nov 21.
Article in English | MEDLINE | ID: mdl-21827172

ABSTRACT

Many cellular proteins with reactive thiols form covalent bonds with electrophiles, thereby modifying their structures and activities. Here, we describe the recovery of a glycolytic protein, glyceraldehyde-3-phosphate dehydrogenase (GAPDH), from such an electrophilic attack by 1,2-napthoquinone (1,2-NQ). GAPDH readily formed a covalent bond with 1,2-NQ through Cys152 at a low concentration (0.2 µM) in a cell-free system, but when human epithelial A549 cells were exposed to this quinone at 20 µM, only minimal binding was observed although extensive binding to numerous other cellular proteins occurred. Depletion of cellular glutathione (GSH) with buthionine sulfoximine (BSO) resulted in some covalent modification of cellular GAPDH by 1,2-NQ and a significant reduction of GAPDH activity in the cells. Incubation of native, but not boiled, human GAPDH that had been modified by 1,2-NQ with GSH resulted in a concentration-dependent removal of 1,2-NQ from the GAPDH conjugate, accompanied by partial recovery of lost catalytic activity and formation of a 1,2-NQ-GSH adduct (1,2-NQ-SG). While GAPDH is recognized as a multifunctional protein, our results show that GAPDH also has a unique ability to recover from electrophilic modification by 1,2-NQ through a GSH-dependent S-transarylation reaction.


Subject(s)
Epithelial Cells/enzymology , Glutathione/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Naphthoquinones/metabolism , Recombinant Proteins/metabolism , Sulfhydryl Compounds/metabolism , Buthionine Sulfoximine/adverse effects , Buthionine Sulfoximine/pharmacology , Cell Line , Cell-Free System , Cloning, Molecular , Epithelial Cells/drug effects , Escherichia coli , Glyceraldehyde-3-Phosphate Dehydrogenases/chemistry , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Glycolysis/drug effects , Glycolysis/genetics , Humans , Mutation , Naphthoquinones/chemistry , Oxidation-Reduction/drug effects , Plasmids , Protein Denaturation , Protein Processing, Post-Translational , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Sulfhydryl Compounds/chemistry , Transformation, Bacterial
SELECTION OF CITATIONS
SEARCH DETAIL
...