Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Neurochem Res ; 38(9): 1838-49, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23743623

ABSTRACT

The antioxidant glutathione (GSH) plays a critical role in maintaining intracellular redox homeostasis but in tumors the GSH biosynthetic pathway is often dysregulated, contributing to tumor resistance to radiation and chemotherapy. Glutamate-cysteine ligase (GCL) catalyzes the first and rate-limiting reaction in GSH synthesis, and enzyme function is controlled by GSH feedback inhibition or by transcriptional upregulation of the catalytic (GCLC) and modifier (GCLM) subunits. However, it has recently been reported that the activity of GCLC and the formation of GCL can be modified by reactive aldehyde products derived from lipid peroxidation. Due to the susceptibility of GCLC to posttranslational modifications by reactive aldehydes, we examined the potential for 2-deoxy-D-ribose (2dDR) to glycate GCLC and regulate enzyme activity and GCL formation. 2dDR was found to directly modify both GCLC and GCLM in vitro, resulting in a significant inhibition of GCLC and GCL enzyme activity without altering substrate affinity or feedback inhibition. 2dDR-mediated glycation also inhibited GCL subunit heterodimerization and formation of the GCL holoenzyme complex while not causing dissociation of pre-formed holoenzyme. This PTM could be of particular importance in glioblastoma (GBM) where intratumoral necrosis provides an abundance of thymidine, which can be metabolized by thymidine phosphorylase (TP) to form 2dDR. TP is expressed at high levels in human GBM tumors and shRNA knockdown of TP in U87 GBM cells results in a significant increase in cellular GCL enzymatic activity.


Subject(s)
Brain Neoplasms/metabolism , Deoxyribose/metabolism , Drug Resistance, Neoplasm , Glioblastoma/metabolism , Glucose/metabolism , Glutamate-Cysteine Ligase/metabolism , Brain Neoplasms/enzymology , Brain Neoplasms/pathology , Catalysis , Cell Line, Tumor , Electrophoresis, Polyacrylamide Gel , Glioblastoma/enzymology , Glioblastoma/pathology , Humans , Kinetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
2.
PLoS One ; 7(9): e44861, 2012.
Article in English | MEDLINE | ID: mdl-22984577

ABSTRACT

NAD(P)H:quinone oxidoreductase 1 (NQO1) is an FAD containing quinone reductase that catalyzes the 2-electron reduction of a broad range of quinones. The 2-electron reduction of quinones to hydroquinones by NQO1 is believed to be a detoxification process since this reaction bypasses the formation of the highly reactive semiquinone. NQO1 is expressed at high levels in normal epithelium, endothelium and adipocytes as well as in many human solid tumors. In addition to its function as a quinone reductase NQO1 has been shown to reduce superoxide and regulate the 20 S proteasomal degradation of proteins including p53. Biochemical studies have indicated that NQO1 is primarily located in the cytosol, however, lower levels of NQO1 have also been found in the nucleus. In these studies we demonstrate using immunocytochemistry and confocal imaging that NQO1 was found associated with mitotic spindles in cells undergoing division. The association of NQO1 with the mitotic spindles was observed in many different human cell lines including nontransformed cells (astrocytes, HUVEC) immortalized cell lines (HBMEC, 16HBE) and cancer (pancreatic adenocarcinoma, BXPC3). Confocal analysis of double-labeling experiments demonstrated co-localization of NQO1with alpha-tubulin in mitotic spindles. In studies with BxPc-3 human pancreatic cancer cells the association of NQO1 with mitotic spindles appeared to be unchanged in the presence of NQO1 inhibitors ES936 or dicoumarol suggesting that NQO1 can associate with the mitotic spindle and still retain catalytic activity. Analysis of archival human squamous lung carcinoma tissue immunostained for NQO1 demonstrated positive staining for NQO1 in the spindles of mitotic cells. The purpose of this study is to demonstrate for the first time the association of the quinone reductase NQO1 with the mitotic spindle in human cells.


Subject(s)
NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/physiology , Spindle Apparatus , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry/methods , Lung Neoplasms/metabolism , Microscopy, Fluorescence/methods , NAD(P)H Dehydrogenase (Quinone)/metabolism , Pancreatic Neoplasms/metabolism , Superoxides/metabolism , Tumor Suppressor Protein p53/metabolism
3.
J Pharmacol Exp Ther ; 336(3): 874-80, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21156818

ABSTRACT

Previous work demonstrated that NAD(P)H:quinone oxidoreductase 1 (NQO1) metabolized the heat shock protein 90 (Hsp90) inhibitor 17-(allylamino)-17-demethoxygeldanamycin (17AAG) to the corresponding hydroquinone (17AAGH2). The formation of 17AAGH2 by NQO1 results in a molecule that binds with greater affinity to Hsp90 compared with the parent quinone. 17AAG induced substantial growth inhibition in human pancreatic cancer cell lines expressing NQO1. Growth inhibition induced by 17AAG could be reduced by pretreatment with 5-methoxy-1,2-dimethyl-3-[(4-nitrophenoxy)methyl]-indole-4,7-dione (ES936), a mechanism-based inhibitor of NQO1. After treatment with 17AAG, biomarkers of Hsp90 inhibition, including markers of cell-cycle arrest, were more pronounced in NQO1-expressing cells compared with NQO1-null cells. The intracellular concentrations of 17AAG and 17AAGH2 were measured in human pancreatic cancer cells, and it was observed that larger amounts of 17AAG and 17AAGH2 could be detected in cells with catalytically active NQO1 compared with cells lacking NQO1 activity or cells pretreated with ES936. These data demonstrate that, in addition to generating an inhibitor with greater affinity for Hsp90 (17AAGH2), reduction of 17AAG to 17AAGH2 by NQO1 leads to substantially greater intracellular concentrations of 17AAG and 17AAGH2. In addition, oxidation of 17AAGH2 could be prevented by superoxide dismutase (SOD), demonstrating that 17AAGH2 was sensitive to oxidation by superoxide. Stable transfection of manganese-dependent SOD into MiaPaCa-2 cells resulted in a significantly greater intracellular concentration of 17AAGH2 with a corresponding increase in growth inhibitory activity. These data confirm the role of NQO1 in sensitivity to 17AAG and demonstrate that SOD functions in conjunction with NQO1 to maintain intracellular levels of 17AAGH2, the active Hsp90 inhibitor derived from 17AAG.


Subject(s)
Benzoquinones/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Lactams, Macrocyclic/pharmacology , NAD(P)H Dehydrogenase (Quinone)/physiology , Pancreatic Neoplasms/metabolism , Superoxide Dismutase/physiology , Cell Line, Tumor , Humans , Pancreatic Neoplasms/pathology
4.
J Pharmacol Exp Ther ; 334(1): 260-8, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20378716

ABSTRACT

NAD(P)H:quinone oxidoreductase 1 (NQO1) deficiency resulting from a homozygous NQO1*2 polymorphism has been associated with an increased risk of benzene-induced myeloid toxicity and a variety of de novo and therapy-induced leukemias. Endothelial cells in human bone marrow form one of the two known hematopoietic stem cell microenvironments and are one of the major cell types that express NQO1 in bone marrow. We have used a transformed human bone marrow endothelial cell (TrHBMEC) line to study the potential impact of a lack of NQO1 activity on adhesion molecule [endothelial leukocyte adhesion molecule 1 (E-selectin), vascular cell adhesion molecule (VCAM)-1, and intercellular adhesion molecule (ICAM)-1] expression and functional adhesion to bone marrow progenitor cells. We used both 5-methoxy-1,2-dimethyl-3-[(4-nitrophenoxy)methyl]indole-4,7-dione (ES936), a mechanism-based inhibitor of NQO1, and anti-NQO1 small interfering RNA to abrogate NQO1 activity. Real-time reverse transcription-polymerase chain reaction data demonstrated a significant inhibition of tumor necrosis factor (TNF)alpha-induced E-selectin mRNA levels after ES936 pretreatment. Immunoblot assays demonstrated a significant reduction in TNFalpha-stimulated E-selectin, VCAM-1, and ICAM-1 proteins after inhibition or knockdown of NQO1. The mechanisms underlying this effect remain undefined, but modulation of nuclear factor-kappaB (p65), c-Jun, and activating transcription factor 2, transcriptional regulators of adhesion molecules, were observed after inhibition or knockdown of NQO1. Decreased level of E-selectin, VCAM-1, and ICAM-1 also resulted in a functional deficit in adhesion. A parallel plate flow chamber study demonstrated a marked reduction in CD34(+) cell (KG1a) adhesion to NQO1-deficient TrHBMECs relative to controls. The reduced adhesive ability of TrHBMECs may affect the function of the vascular stem cell niche and also may contribute to the increased susceptibility of polymorphic individuals lacking NQO1 to leukemias and hematotoxicants such as benzene.


Subject(s)
Antigens, CD34/metabolism , Bone Marrow Cells/physiology , Cell Adhesion Molecules/biosynthesis , Endothelial Cells/physiology , Hematopoietic Stem Cells/physiology , NAD(P)H Dehydrogenase (Quinone)/deficiency , Bone Marrow Cells/cytology , Bone Marrow Cells/enzymology , Cell Adhesion/physiology , Cell Line, Transformed , E-Selectin/biosynthesis , E-Selectin/genetics , Endothelial Cells/cytology , Endothelial Cells/enzymology , Enzyme Inhibitors/pharmacology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Immunoblotting , Indolequinones/pharmacology , Intercellular Adhesion Molecule-1/biosynthesis , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1/biosynthesis
5.
Mol Pharmacol ; 76(3): 579-87, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19525446

ABSTRACT

Bone marrow is a major target of benzene toxicity, and NAD-(P)H:quinone oxidoreductase (NQO1), an enzyme protective against benzene toxicity, is present in human bone marrow endothelial cells, which form the hematopoietic stem cell vascular niche. In this study, we have employed a transformed human bone marrow endothelial cell (TrHBMEC) line to study the adverse effects induced by the benzene metabolite hydroquinone. Hydroquinone inhibited TrHBMEC tube formation at concentrations that were not overtly toxic, as demonstrated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide or sulforhodamine B analysis. Hydroquinone was found to up-regulate chondromodulin-I (ChM-I), a protein that promotes chondrocyte growth and inhibits endothelial cell growth and tube formation. Recombinant human ChM-I protein inhibited tube formation in TrHBMECs, suggesting that up-regulation of ChM-I may explain the ability of hydroquinone to inhibit TrHB-MEC tube formation. To explore this possibility further, anti-ChM-I small interfering RNA (siRNA) was used to deplete ChM-I mRNA and protein. Pretreatment with anti-ChM-I siRNA markedly abrogated hydroquinone-induced inhibition of tube formation in TrHBMECs. Overexpression of the protective enzyme NQO1 in TrHBMECs inhibited the up-regulation of ChM-I and abrogated the inhibition of tube formation induced by hydroquinone. In summary, hydroquinone treatment up-regulated ChM-I and inhibited tube formation in TrHBMECs; NQO1 inhibited hydroquinone-induced up-regulation of ChM-I in TrHB-MECs and protected cells from hydroquinone-induced inhibition of tube formation. This study demonstrates that ChM-I up-regulation is one of the underlying mechanisms of inhibition of tube formation and provides a mechanism that may contribute to benzene-induced toxicity at the level of bone marrow endothelium.


Subject(s)
Benzene/metabolism , Bone Marrow Cells/drug effects , Endothelial Cells/drug effects , Hydroquinones/toxicity , Intercellular Signaling Peptides and Proteins/biosynthesis , Membrane Proteins/biosynthesis , Benzene/toxicity , Cell Line , Collagen , Drug Combinations , Humans , Hydroquinones/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Laminin , Membrane Proteins/genetics , NAD(P)H Dehydrogenase (Quinone)/biosynthesis , Proteoglycans , RNA, Messenger/biosynthesis , RNA, Small Interfering/genetics , Up-Regulation
6.
Mol Pharmacol ; 74(6): 1657-65, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18794327

ABSTRACT

2,5-Diaziridinyl-3-(hydroxymethyl)-6-methyl-1,4-benzoquinone (RH1) is a novel antitumor diaziridinyl benzoquinone derivative designed to be bioactivated by the two-electron reductase NAD(P)H:quinone oxidoreductase (NQO1) and is currently in clinical trials. NQO1 is expressed at high levels in many solid tumors. RH1 cytotoxicity has been shown previously to be NQO1-dependent. The purpose of this study was to investigate whether other reducing enzymes such as cytochrome b(5) reductase (b5R), cytochrome P450 reductase (P450R), dihydronicotinamide riboside:quinone oxidoreductase 2 (NQO2), and xanthine oxidase/xanthine dehydrogenase (XO/XDH) also contribute to the bioactivation and cytotoxicity of RH1 in human tumor cells. For these studies, we established a series of stable MDA468 breast cancer cell lines overexpressing various levels of NQO1, b5R, P450R, and NQO2 and compared RH1-induced growth inhibition [3-(4,5-dimethylthiazol-2,5-diphenyl)tetrazolium and sulforhodamine B analysis] and interstrand DNA cross-linking (comet analysis) in both parental MDA468 cells and transfected clones. RH1 toxicity correlated with NQO1 and NQO2 but not with either b5R or P450R activity levels in the respective series of transfected MDA468 cell clones. Enzymatic assays showed that RH1 was an in vitro substrate for xanthine oxidase. However, XO/XDH protein and activity could not be detected in a variety of human tumor cell lines. These studies suggest that NQO1 and NQO2 are the principal enzymatic determinants of RH1 bioactivation in MDA468 tumor cells and that b5R, P450R, and XDH/XO are unlikely to play major roles. Our studies also suggest that NQO2 may be particularly relevant as a bioactivation system for RH1 in NQO1-deficient tumors such as leukemias and lymphomas.


Subject(s)
Antineoplastic Agents/pharmacology , Aziridines/pharmacology , Benzoquinones/pharmacology , NAD(P)H Dehydrogenase (Quinone)/metabolism , Oxidoreductases/metabolism , Antineoplastic Agents/metabolism , Aziridines/metabolism , Benzoquinones/metabolism , Cell Line, Tumor , Cytochromes b5/metabolism , Humans , NADPH-Ferrihemoprotein Reductase/metabolism , Quinone Reductases/metabolism , Xanthine Dehydrogenase/metabolism , Xanthine Oxidase/metabolism
7.
Curr Protoc Toxicol ; Chapter 4: Unit4.22, 2007.
Article in English | MEDLINE | ID: mdl-23045144

ABSTRACT

NAD(P)H:quinone oxidoreductase 1 (NQO1, DT-diaphorase, E.C. 1.6.99.2) is an FAD containing obligate two-electron reductase that catalyzes the reduction of a broad range of substrates. This unit will describe methods for the detection of NQO1 protein in formalin-fixed, paraffin-embedded tissues by immunohistochemistry; detection of NQO1 protein in fresh tissues or cell lines by immunoblot analysis; measurement of NQO1 catalytic activity in fresh and frozen tissues and cell lines using spectrophotometric assays based upon the reduction of 2,6-dichlorophenol-indophenol (DCPIP) or coupled menadione-cytochrome reduction; and determination of the NQO1*2 polymorphism by PCR-RFLP.


Subject(s)
NAD(P)H Dehydrogenase (Quinone) , Base Sequence , Catalysis , Cell Line , Humans , Immunoblotting , Immunohistochemistry , Molecular Sequence Data , NAD(P)H Dehydrogenase (Quinone)/chemistry , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , Pharmaceutical Preparations/metabolism , Polymorphism, Restriction Fragment Length , Reverse Transcriptase Polymerase Chain Reaction , Spectrophotometry
8.
Biochem Biophys Res Commun ; 311(2): 446-53, 2003 Nov 14.
Article in English | MEDLINE | ID: mdl-14592434

ABSTRACT

To determine whether the human (h) NQO1 (NAD(P)H: quinone oxidoreductase 1) gene contains DNA sequences that directly mediate its high and low expression in non-small cell lung carcinoma (NSCLC) and small cell lung carcinoma (SCLC), respectively, a series of deletion constructs spanning up to -4kb of the 5(') flanking region of hNQO1 was used in transient transfection assays. The antioxidant response element (ARE) was found to be critical to the elevated expression of NQO1 in NSCLC. However, the ability of both heterologous and deletion promoter constructs to confer ARE responsiveness demonstrated that SCLC contains the necessary program/menu of transcription factors responsible to drive hNQO1 expression via the ARE. By examining the regulatory region of the hNQO1 gene, we identified a proximal repressor region between -259 and -131. These results provide the first evidence of a proximal repressor region exerting a negative role on the regulation of the hNQO1 promoter in SCLC.


Subject(s)
Antioxidants/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Small Cell/genetics , Carcinoma, Small Cell/metabolism , Gene Expression Regulation, Neoplastic/genetics , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , Cell Line, Tumor , Enzyme Activation , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Promoter Regions, Genetic
9.
J Pharmacol Exp Ther ; 305(3): 1079-86, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12649308

ABSTRACT

The bioreductive activation of the antitumor quinone mitomycin C (MMC) by NAD(P)H: quinone oxidoreductase 1 (NQO1) is complicated by the ability of MMC to also act as a mechanism-based inhibitor of NQO1 in a pH dependent manner. Inhibition of NQO1 by MMC has been studied in purified enzyme preparations and in cultured cells but has not determined in vivo. In the studies presented here, NQO1 activity was measured in mouse tissues following treatment with MMC or the potent mechanism-based human NQO1 inhibitor 5-methoxy-1,2-dimethyl-[(4-nitrophenoxy)methyl]indole-4,7-dione (ES936). NQO1 activity was significantly decreased at 1, 2, and 4 h following MMC (10 or 20 mg/kg) treatment in kidney and lung but was unchanged in brain, heart, liver, and bladder. ES936 (1 mg/kg) treatment led to a significant and much more potent inhibition of NQO1 in all murine tissues analyzed except for bladder. To extrapolate these in vivo results from mice to humans, the species-specific kinetics of NQO1 inactivation by MMC was determined in vitro using mouse, rat, and human recombinant NQO1 proteins. Results showed the inactivation kinetics of mouse and human proteins by MMC were similar. Treatment of human and murine endothelial cells with MMC or ES936 showed similar inhibition of NQO1 activity. The aforementioned results clearly demonstrate that MMC can serve as a substrate for NQO1 in vivo; however, the metabolism resulting in enzyme inactivation is possibly tissue-specific. Furthermore, the kinetic similarities for inactivation between murine and human forms of NQO1 show these results are apropos to clinical use of MMC.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Indolequinones , Mitomycin/pharmacology , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , Animals , Drug Interactions , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Female , Humans , Indoles/pharmacology , Kinetics , Mice , Mice, Inbred BALB C , Rats , Species Specificity , Tumor Cells, Cultured
10.
J Biol Chem ; 278(12): 10368-73, 2003 Mar 21.
Article in English | MEDLINE | ID: mdl-12529318

ABSTRACT

NAD(P)H:quinone oxidoreductase 1 (NQO1) has been proposed to stabilize p53 via a redox mechanism involving oxidation of NAD(P)H as a consequence of the catalytic activity of NQO1. We report that treatment of HCT-116 human colon carcinoma cells with the NQO1 inhibitor ES936 had no effect on the levels of p53 protein. ES936 is a mechanism-based inhibitor of NQO1 that irreversibly blocks the catalytic function of the enzyme. This suggests that a redox mechanism involving NQO1-mediated NAD(P)H oxidation is not responsible for the stabilization of p53. We also examined the ability of the NQO1 protein to associate with p53 using co-immunoprecipitation experiments. Results from these experiments demonstrated co-immunoprecipitation of NQO1 with p53 and vice versa. The association between p53 and NQO1 was not affected by treatment of HCT-116 cells with ES936, demonstrating that the association was not dependent on the catalytic activity of NQO1. A comparison of isogenic HCT-116 p53+/+ and HCT-116 p53-/- cells demonstrated an interaction of NQO1 and p53 only in the p53+/+ cells. Experiments performed in an in vitro transcription/translation system utilizing rabbit reticulocyte lysates confirmed the interaction of NQO1 and p53. In these experiments a full-length p53 coding region was used to express p53 in the presence of recombinant NQO1 protein. An association of p53 and NQO1 was also observed in primary human keratinocytes and mammary epithelial cells. In studies where mdm-2 co-immunoprecipitated with p53, no association of mdm-2 with NQO1 was observed. These data demonstrate an association between p53 and NQO1 that may represent an alternate mechanism of p53 stabilization by NQO1 in a wide variety of human cell types.


Subject(s)
NAD(P)H Dehydrogenase (Quinone)/chemistry , Nuclear Proteins , Tumor Suppressor Protein p53/chemistry , Cell-Free System , Humans , NAD(P)H Dehydrogenase (Quinone)/analysis , Precipitin Tests , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins c-mdm2 , Tumor Cells, Cultured , Tumor Suppressor Protein p53/analysis
11.
J Biol Chem ; 277(16): 14060-7, 2002 Apr 19.
Article in English | MEDLINE | ID: mdl-11821413

ABSTRACT

NAD(P)H:quinone oxidoreductase 1 (EC; DT-Diaphorase, NQO1) is predominantly a cytosolic flavoenzyme that catalyzes a two-electron reduction. Using human tumor cell lines devoid of NQO1 enzymatic activity, we have previously identified a single nucleotide polymorphism (NQO1*2 allele) in the human NQO1 gene. This mutation has been characterized as a genetic polymorphism (NQO1*2), which leads to greatly diminished levels of protein due to rapid degradation of the NQO1*2 protein by the ubiquitin proteasomal pathway (UPP). In an attempt to decipher the mechanism responsible for the differential stability of wild-type NQO1*1 and mutant NQO1*2 proteins, we have investigated the interactions of these proteins with molecular chaperones of the Hsp family. Using co-immunoprecipitation studies (co-IPs), no association was observed between Hsp90 and either wild-type NQO1*1 or mutant NQO1*2 proteins. Hsp70, however, was found to associate with NQO1*1 protein in cells when co-IPs were performed with an anti-NQO1 antibody followed by immunoblotting with an anti-Hsp70 antibody or vice versa. Hsp40 could also be detected in the immunoprecipitated protein complex. Experiments were also performed using either the NQO1*1 or NQO1*2 coding regions in an in vitro transcription/translation system employing rabbit reticulocyte lysates (RRLs). Consistent with the cellular data, co-IP experiments in RRLs demonstrated an association of Hsp70 with wild-type NQO1*1 protein but not with NQO1*2 protein. To further elucidate the role of the association of Hsp70 with the NQO1*1 protein, site-directed mutagenesis was used to modify a proposed Hsp70 binding site near the N terminus of the NQO1 protein. We generated a plasmid containing an NQO1*1 coding region with a mutated Hsp70 binding site (isoleucine to aspartic acid at position 8, NQO1*1/I8D). In contrast to the NQO1*1 protein translated in RRLs, the NQO1*1/I8D protein did not associate with Hsp70, as demonstrated by co-IP, was catalytically inactive, and was degraded by the UPP. These data suggest that the association of Hsp70 with NQO1*1 may play an important role in the stability and functionality of the NQO1 protein.


Subject(s)
HSP70 Heat-Shock Proteins/metabolism , NADH, NADPH Oxidoreductases/metabolism , Alleles , Amino Acid Sequence , Catalysis , Cell Line , Cysteine Endopeptidases , Electrophoresis, Polyacrylamide Gel , FMN Reductase , HSP40 Heat-Shock Proteins , HSP70 Heat-Shock Proteins/chemistry , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/metabolism , Humans , Immunoblotting , Molecular Sequence Data , Multienzyme Complexes/antagonists & inhibitors , Mutagenesis, Site-Directed , Mutation , NADH, NADPH Oxidoreductases/chemistry , Plasmids/metabolism , Precipitin Tests , Proteasome Endopeptidase Complex , Protein Binding , Protein Biosynthesis , Protein Folding , Protein Structure, Tertiary , Time Factors , Transcription, Genetic , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...