ArticlePDF Available

NAD(P)H:Quinone Oxidoreductase 1 (NQO1) Localizes to the Mitotic Spindle in Human Cells

PLOS
PLOS ONE
Authors:

Abstract and Figures

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.
Content may be subject to copyright.
NAD(P)H:Quinone Oxidoreductase 1 (NQO1) Localizes to
the Mitotic Spindle in Human Cells
David Siegel*, Jadwiga K. Kepa, David Ross
Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado,
United States of America
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.
Citation: Siegel D, Kepa JK, Ross D (2012) NAD(P)H:Quinone Oxidoreductase 1 (NQO1) Localizes to the Mitotic Spindle in Human Cells. PLoS ONE 7(9): e44861.
doi:10.1371/journal.pone.0044861
Editor: Stefan Strack, University of Iowa, United States of America
Received April 12, 2012; Accepted August 8, 2012; Published September 11, 2012
Copyright: ß2012 Siegel et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Funding: This work was supported by National Institutes of Health grant R01ES108943. The funders had no role in study design, data collection and analysis,
decision to publish, or preparation of the manuscript.
Competing Interests: DS, DR and the University of Colorado are the licensor for the anti-NQO1 monoclonal antibody (clone A180). This does not alter the
authors’ adherence to all the PLOS ONE policies on sharing data and materials.
* E-mail: david.siegel@ucdenver.edu
Introduction
NAD(P)H:quinone oxidoreductase 1 (NQO1, DT-diaphorase,
EC 1.6.99.2) is a homodimeric flavoprotein that utilizes either
NADH or NADPH and catalyzes the 2-electron reduction of a
broad range of substrates most notably quinones [1]. The two-
electron reduction of quinones to hydroquinones by NQO1 is
believed to be primarily a detoxification reaction since it bypasses
the formation of the highly reactive semiquinone [1]. In many
cases, however, the reduction of quinones by NQO1 results in the
formation cytotoxic hydroquinones and the bioactivation of
quinone prodrugs by NQO1 has been utilized as a strategy to
target NQO1-rich cancer cells [2].
In normal tissues, NQO1 is expressed at relatively high levels in
epithelial tissues, vascular endothelium and adipocytes while in
cancer, NQO1 is expressed at high levels in many solid tumors
including lung (NSCLC), breast and pancreatic [3,4]. In humans,
the NQO1*2 polymorphism plays a major role in governing basal
protein levels of NQO1 [5]. The NQO1*2 polymorphism results
in a proline to serine amino acid substitution at position 187 in
NQO1 and this mutant protein undergoes rapid polyubiquitina-
tion by the E3 ubiquitin ligase STUB1/CHIP with subsequent
proteasomal degradation [6,7]. Individuals homozygous for the
NQO1*2 polymorphism are NQO1 null, while intermediate levels
of NQO1 protein are found in individuals with the heterozygous
genotype [5]. NQO1 is under transcriptional regulation by the
Keap1/NRF2 pathway and upregulation of NQO1 mRNA or
protein has been used extensively as a biomarker for NRF2
activation [8,9]. Upregulation of NQO1 may protect the cell from
oxidative damage due to the ability of NQO1 to reduce
superoxide to hydrogen peroxide and generate antioxidant forms
of vitamin E and co-enzyme Q [10,11,12].
In addition to its role as an antioxidant enzyme, NQO1 has
been shown to protect a wide range of proteins including p53 from
ubiquitin-independent 20 S proteasomal degradation [13,14]. The
protection of target proteins by NQO1 from 20 S proteasomal
degradation is dependent upon the redox state of NQO1 since
treatment with the NQO1 inhibitor dicoumarol has been shown to
enhance the 20 S proteasomal degradation of several target
proteins [13,14].
NQO1 is predominately located in the cytoplasm but low levels
of NQO1 have been found in the nucleus under normal conditions
[15]. Under conditions of stress NQO1 has been shown to migrate
to the nucleus where it is hypothesized that NQO1 may protect
PLOS ONE | www.plosone.org 1 September 2012 | Volume 7 | Issue 9 | e44861
p53 against 20 S proteasomal degradation [16]. In experiments
designed to monitor the subcellular distribution of NQO1 in
human cells by confocal microscopy we discovered that NQO1
could also be found in association with the mitotic spindle.
Results
Immunofluorescent staining of the human pancreatic adeno-
carcinoma cell line BxPc-3 for NQO1 revealed that NQO1 is
located primarily in the cytosol of these cells (Fig. 1). However, in
BxPc-3 cells undergoing mitosis intense immunostaining for
NQO1 was observed on the mitotic spindles (Fig. 1). Our source
of anti-NQO1 monoclonal antibody for these studies was
conditioned tissue culture supernatant from mouse hybridoma
clone A180. In control studies, we utilized unconditioned media
(RPMI1640 containing 10% fetal bovine serum) in place of
hybridoma clone A180 supernatant and in these studies no
immunofluorescent staining of NQO1in BxPc3 cells or spindles
could be observed (Fig. S1). In addition, no observable difference
in immunostaining of NQO1 including spindles could be detected
in BxPc-3 cells between tissue culture supernatant from hybridoma
clone A180 or commercially prepared affinity purified mouse IgG
from hybridoma clone A180 (Fig S2).
To confirm that NQO1 was the target of monoclonal antibody
A180 we generated a BxPc-3 cell line stably expressing a
doxycycline-inducible anti-NQO1shRNA expression vector. The
growth of this genetically modified BxPc3 cell line in doxycyline
for 7 days resulted in near complete knockdown of NQO1 protein
expression as confirmed by immunoblotting for NQO1 (Fig 2). As
expected, doxycycline treated cells showed no detectable immu-
nostaining for NQO1 either in the cytosol or on mitotic spindles
(Fig 2). These data confirm that NQO1 is the target of monoclonal
antibody clone A180 and that the immunostaining of mitotic
spindles by anti-NQO1 antibodies from clone A180 is not
observed when NQO1 protein levels are knockdown.
To confirm co-localization of NQO1 with the microtubule
containing mitotic spindles, doubling-labeling experiments were
performed using antibodies against NQO1 and alpha-tubulin and
results from these studies confirmed the co-localization of NQO1
with alpha-tubulin in the mitoic spindles (Fig 3). The association of
NQO1 with the mitotic spindles could be observed throughout
mitosis from metaphase to cytokinesis (Fig. 3). In cells not
undergoing division no obvious co-localization of NQO1 with
alpha-tubulin was observed, (see Fig. 3 D–F) suggesting that the
association between NQO1 and microtubules is specific for mitotic
spindles. Treatment of BxPc-3 cells with the competitive inhibitor
dicoumarol or the mechanism-based inhibitor ES936 did not
prevent the association of NQO1 with mitotic spindles suggesting
that the association of NQO1 with the mitotic spindle is not
dependent upon the redox state of NQO1 (Fig. 4).
The immunofluorescent staining of NQO1 was performed in
many different human cell lines (Fig. 5) including cancer cell lines
(pancreatic), immortalized cell lines (HBMEC, 16HBE) and
primary cell lines (astrocytes, HUVEC). The human pancreatic
cancer cell line Panc-1, which has previously been genotyped as
homozygous for the NQO1*2 polymorphism [17] and character-
ized as NQO1 null, did not demonstrate detectable immunoflu-
orescent staining for NQO1 either in the cytosol or mitotic
spindles. Stable expression of NQO1 in the Panc-1 cell line (clone
panc-1/C5) resulted in significant staining for NQO1 in the
cytosol and mitotic spindles (Fig 6).
To determine if NQO1 could be found associated with mitotic
spindles in human tissues, we examined human NSCLC tissue
samples that had been immunostained for NQO1 in a previous
study [18]. Upon examination of squamous cell carcinomas
samples mitotic figures were located and immunostaining for
NQO1 was clearly visible on the mitotic spindles of these cells
(Fig 7). These data support our studies in cell culture that
demonstrate the association of NQO1 with mitotic spindles.
Discussion
These data demonstrate that the quinone oxidoreductase
NQO1 associates with the mitotic spindles and suggests that this
association with the mitotic spindle is independent of the redox
state of NQO1 since immunostaining for NQO1 on spindles was
not diminished in the presence of NQO1 inhibitors. While this is
the first report of localization of NQO1 with mitotic spindles,
previous studies using Xenopus egg extracts indentified NQO1 as
a target in a chemical screen of purine-based mitotic disrupters
[19]. In this study, diminutol (2-(1R-isopropyl-2-hydroxyethyla-
mino)-6-(3-aminophenylthio)-9-isopropylpurine), which prevents
microtubule polymerization, was also shown to inhibit NQO1
catalytic activity via competitive binding with NAD(P)H. These
same studies demonstrated immuno-depletion of NQO1 from
Xenopus egg extracts resulted in defects in spindle assembly [19].
Despite these findings that suggested NQO1 catalytic activity may
be linked to spindle assembly in Xenopus eggs, the authors did not
demonstrate localization of NQO1 with spindles [19]. Unlike
studies in Xenopus eggs, our studies in human cells have shown
Figure 1. Immunofluorescent staining of NQO1 in BxPc-3 human pancreatic cancer cells. Immunostaining for NQO1 reveals cytosolic
localization and intense staining of mitotic spindles in cells undergoing division (*).
doi:10.1371/journal.pone.0044861.g001
NQO1 Localizes to the Mitotic Spindle
PLOS ONE | www.plosone.org 2 September 2012 | Volume 7 | Issue 9 | e44861
that NQO1 can be found associated with the mitotic spindles;
however, treatment of human cells with the NQO1 mechanism-
based inhibitor ES936, which resulted in .95% inhibition of
NQO1 catalytic activity, did not alter the growth rate of these cells
[20]. These data suggest that in human cells despite the binding of
NQO1 to the mitotic spindles NQO1 most likely does not play a
direct role in regulating mitosis. These observations are reinforced
by studies in human cell lines that lack NQO1 activity and protein
due to homozygous expression of the NQO1*2 polymorphism.
Many commonly used human cells lines (BE, MDA231, MDA468,
Panc-1, Caco-2) lack NQO1 activity and protein due to
homozygous expression of the NQO1*2 polymorphism and for
many of these cell lines isogenic pairs have been created following
the stable transfection of wild type NQO1. The reintroduction of
wild type NQO1 back into these cell lines did not significantly alter
the growth rate when compared to their NQO1 null isogenic
parental cells [21]. Taken together, these data imply that in
human cells NQO1 does not play a major role in regulating
mitosis but instead NQO1 may act in a protective role.
Figure 2. Knockdown of NQO1 eliminates immunostaining of NQO1 in BxPc-3 cells. Knockdown of NQO1 in BxPc-3 cells was performed
using a doxycyline- inducible anti-NQO1 shRNA methodologies. BxPc-3 cells were immunostained for NQO1 (green) in the absence (panel A) and
presence (panel B) of doxycyline pretreatment. DAPI staining was included in panel B to aid in the location of cells. Right panel: immunoblot
demonstrating near complete knockdown of NQO1 protein in the presence of doxycycline. Arrows indicate mitotic cells.
doi:10.1371/journal.pone.0044861.g002
Figure 3. Co-localization of NQO1 and alpha-tubulin in mitotic spindles and midbody region in BxPc-3 human pancreatic cancer
cells. Triple immunofluorescent staining for NQO1 (red), alpha-tubulin (green) and nuclei (blue). Association of NQO1 with spindles can be seen in
different stages of mitosis including metaphase (*), telophase (**) and the midbody region (arrow) during cytokinesis.
doi:10.1371/journal.pone.0044861.g003
NQO1 Localizes to the Mitotic Spindle
PLOS ONE | www.plosone.org 3 September 2012 | Volume 7 | Issue 9 | e44861
The ability of NQO1 to associate with the mitotic spindles in
human cells while still maintaining catalytic activity has implica-
tions for both quinone detoxification and activation reactions in
close proximity to critical mitotic apparatus. The reduction of
quinones by NQO1 could result in protection of vulnerable
microtubules from attack by electrophilic quinones. The benzene
metabolite p-benzoquinone has been shown in cell free systems to
react readily with sulfhydryl groups on microtubule proteins and
inhibit assembly into microtubules [22]. Alternatively, NQO1
could be the source of high levels of reactive oxygen species due to
redox cycling of unstable hydroquinones formed in close proximity
to the mitotic apparatus.
An alternative catalytic function of NQO1 may be to aid in
DNA repair during mitosis through the generation of NAD
+
an
essential cofactor for proteins including the family of poly (ADP
ribose) polymerase (PARP) enzymes and the sirtuins. PARP
enzymes have been shown to play an important role in the repair
of single strand DNA breaks [23] while SIRT6 has been proposed
to aid in DNA repair under conditions of oxidative stress by
binding directly to PARP1 and stimulating PARP1 poly-ADP-
ribosylase activity [24].
From our studies it is unclear whether NQO1 binds directly to
microtubules in mitotic spindles or binds to a protein complex that
associates with the mitotic spindles. NQO1 has many character-
istics of a microtubule binding protein including a net positive
charge and multiple domains containing clusters of positively
charged amino acids [25,26,27]. Alternatively, NQO1 could be
binding to another protein or protein complex that is tethered to
the mitotic spindle. In addition to its major role in quinone
metabolism NQO1 has emerged as a regulator of 20 S
proteasomal degradation [28]. A growing number of proteins
have been shown to be protected from ubiquitin-independent 20 S
proteasome degradation by NQO1 including p53, p63, p73 and
ornithine decarboxylase [14,29,30,31]. Interestingly, immunoflu-
orescence studies have previously demonstrated co-localization of
p53 with centrosomes and mitotic spindles in human cell lines [32]
and co-localization of the 20 S proteasome to centrosomes and
mitotic spindles in rat Schwann cells [33]. Whether NQO1
interacts with these proteins on the spindle to facilitate in the
stabilization and redistribution of selected proteins such as p53
during mitosis is currently under investigation. Recently, it has
been shown that persistent expression of NQO1 via p62-mediated
Nrf2 activation facilitated p53-dependent mitotic catastrophe
supporting a role for NQO1 in the stabilization of p53 during
mitosis [34].
In summary, these data report the association of NQO1 with
mitotic spindles in human cells using immunofluorescent staining
and suggest that the association of NQO1 with spindles may
involve protection against electrophic quinones, aid in the
generation of oxidized pyridine nucleotides or shield selected
proteins from 20 S degradation.
Materials and Methods
Reagents
Anti-NQO1 mouse monoclonal antibody (IgG
1
, clone A180)
was developed in our laboratory against full-length recombinant
human NQO1 protein and hybridoma supernatant (RPMI1640
containing 10% fetal bovine serum) from clone A180 was used
for these studies (dilution 1:5–1:20). Affinity purified anti-
NQO1 monoclonal antibody from clone A180 was obtained
commercially from Cell Signaling Technologies (Danvers, MA)
and used at a dilution of 1:200. Anti-alpha tubulin rabbit
polyclonal antibody was purchased from Abcam, (Cambridge
MA, Cat#15246, dilution 1:10,000). DyLight 488 and DyLight
544 conjugated goat anti-mouse IgG and DyLight 488
conjugated goat anti-rabbit IgG antibodies were purchased
from Jackson ImmunoResearch Laboratories (West Grove PA).
49,6-diamidino-2-phenylindole (DAPI,) dicoumarol and doxy-
cycline were obtained from Sigma (St. Louis MO). The NQO1
mechanism-based inhibitor ES936 (5-methoxy-1,2-dimethyl-3-
[(4-nitrophenoxy)methyl]indole-4,7-dione) was synthesized in
the laboratory of Dr. Christopher J. Moody, School of
Chemistry, University of Nottingham, U.K [35].
Cell Lines and Tissues
BXPC3 and Panc-1 human pancreatic cancer cell lines were
obtained from ATCC (Manassas, VA) and grown in RPMI 1640
containing 5% (v/v) fetal bovine serum and 1% (v/v) penicillin/
streptomycin. Panc-1 cells have previously been genotyped as
homozygous for the NQO1*2 polymorphism and are NQO1null
[17]. Panc-1/C5 cells stably express wildtype NQO1 and were
created from parental Panc-1 cells as described [17]. Primary
cultures of human umbilical cord vein endothelial cells (HUVEC)
Figure 4. NQO1 inhibitors do not prevent binding of NQO1 to mitotic spindles. Immunostaining for NQO1 (red) in BXPC3 cells pretreated
with DMSO (panel A), ES936 (500 nM, panel B) or dicoumarol (50mM, panel C). Cells were pretreated with inhibitors 2 hr before immunostaining.
Arrows indicate mitotic cells.
doi:10.1371/journal.pone.0044861.g004
NQO1 Localizes to the Mitotic Spindle
PLOS ONE | www.plosone.org 4 September 2012 | Volume 7 | Issue 9 | e44861
were obtained from Cell Applications (San Diego, CA) and grown
in Endothelial Cell Growth Media (Cell Applications). Primary
human astrocytes were obtained from ScienCell Research Labs
(Carlsbad, CA) and grown in Astrocyte Medium (ScienCell
Research Labs). Immortalized human bone marrow endothelial
cells (HBMEC) were provided by Dr. B.B. Weksler (Cornell
University) and grown as previously described [36]. Immortalized
human bronchial epithelial cells [37] (16HBE) were provided by
Dr. Brian Day (National Jewish Health, Denver CO) and grown in
RPMI 1640 containing 5% fetal bovine serum and 1% penicillin/
Figure 5. Co-localization of NQO1 and alpha-tubulin with mitotic spindles in human cells. Triple immunofluorescent staining for NQO1
(red, panel A); alpha-tubulin (green, panel B); merged (with DAPI,panel C).
doi:10.1371/journal.pone.0044861.g005
NQO1 Localizes to the Mitotic Spindle
PLOS ONE | www.plosone.org 5 September 2012 | Volume 7 | Issue 9 | e44861
streptomycin. All cells were maintained as monolayers at 37uCin
5% CO
2
.
For immunocytochemical studies all cells were plated onto glass
coverslips (18 mm60.15 mm circle, Fisher Scientific) in 100 mm
tissue cultures dishes 3 days prior to analysis. For studies using
NQO1 inhibitors ES936 and dicoumarol cells were incubated in
fresh media (2 ml) containing inhibitors (500 nM ES936 or 50 mM
dicoumarol) for 2 hr. The generation of a doxycycline-inducible
NQO1 knock-down cell line was performed using a TRIPZ
inducible shRNA plasmid for human NQO1 (Open Biosystems,
Lafayette, CO). To create a stable knockdown cell line, 2610
6
BxPc3 cells were electroporated with 20 ug of TRIPZ inducible
shRNA plasmid for human NQO1 and transfected cells were
selected in normal culture media containing 3 mg/ml puromycin.
The antibiotic resistant cells were induced with 300 ng/ml
doxycyclin for 48 h and then cells were sorted for turbo red
fluorescence (553excitation/574emission) using the Legacy MoFlo
sorter (University of Colorado Cancer Center). Sorted cells were
maintained in complete media containing 500 ng/ml doxycycline
and knockdown of NQO1 was confirmed by immunoblot analysis.
Coincubation studies with Panc-1 cells and Panc-1/C5 cells
were performed using the following method. Panc-1 cells (1610
6
)
Figure 6. Co-localization of NQO1 with mitotic spindles is not observed in cells homozygous for the NQO1*2 polymorphism. Panc-1
cells which are homozygous for the NQO1*2 polymorphism (NQO1 null), were prelabeled with cell tracker green then mixed with unlabled Panc-1/C5
cells which stably express wild type hNQO1. Following a 24hr coincubation period cells were fixed and immunostained for NQO1 followed by
confocal microscopy. Cells were searched until a mitotic Panc-1 cell (green) and a mitotic Panc-1/C5 cell (non-staining) were observed in the same
field. Panels A and C, data was collected using 488nm excitation wavelength for cell tracker green fluorescence (green Panc-1 cells). Panels B and D,
data was collected using 561nm excitation wavelength (red/NQO1). Co-localization of NQO1 to the mitotic spindles was not observed in NQO1-null
Panc-1 cells (green) while co-localization of NQO1 to the mitotic spindle was seen in NQO1 overexpressing Panc-1/C5 cells.
doi:10.1371/journal.pone.0044861.g006
Figure 7. NQO1 localizes to mitotic spindles in human lung cancer cells. Immunohistochemical analysis of NQO1 (DAB, brown) in archival
formalin-fixed human squamous lung carcinoma tissue samples. Mitotic figures are visible due to their intense chromatin staining (hemotoxylin).
doi:10.1371/journal.pone.0044861.g007
NQO1 Localizes to the Mitotic Spindle
PLOS ONE | www.plosone.org 6 September 2012 | Volume 7 | Issue 9 | e44861
were treated with 5 mM Cell Tracker Green (Invitrogen, Carlsbad,
CA) in serum-free media in suspension at 37uC. After 1 hr Panc-1
cells were collected by centrifugation, washed in PBS and then
mixed with unlabeled Panc-1/C5 cells (1610
6
) and plated onto
glass coverslips in complete media. After 24 hr the coverslips were
fixed for immunocytochemisty as described below.
Achieved formalin fixed human squamous lung cancer tissue
was obtained from the University of Colorado Cancer Center and
immunostained for NQO1 as previously described [18].
Immunocytochemistry
Immunocytochemistry was performed on cell lines grown on
glass coverslips using the following methods. All steps were
performed at room temperature. Cells were washed in phosphate
buffered saline (PBS) then fixed in 3.7% (v/v) formaldehyde in
PBS for 12 min, rinsed with PBS, then permeabilize with 0.15%
(v/v) Triton-X100 in PBS for 10 min. Cells were then blocked in
RPMI1640 containing 5% fetal bovine serum for 1 hr at room
temperature. Coverslips were then transferred to a 6-well plate
(single coverslip/well) and primary antibodies diluted in TBST
(10 mM Tris-HCl, pH 8 containing 150 mM NaCl and 0.4% (v/
v) Tween-20), were added (2 ml/well). After 90 min the primary
antibodies were removed, the cells were gently rinsed with TBST,
and secondary antibodies (diluted in TBST) were then added
(2 ml/well) for 30 min. DAPI (1 mg/ml) was included with the
secondary antibody. After 30 min coverslips were rinsed in TBST,
briefly immersed in deionized/distilled H
2
0, inverted, and
mounted on to glass microscope slides using SuperMount
(Biogenex, San Ramon, CA). Stained cells were examined using
a Nikon TE2000E2 inverted microscope equipped with a C1-Plus
confocal system (lasers 402 nm, 488 nm, 514 nm, Nikon Instru-
ments, Melville, NY).
Supporting Information
Figure S1 No fluorescent immunostaining of mitotic
spindles in the absence of anti-NQO1 primary antibody.
When unconditioned RPMI1640 containing 10% fetal bovine
serum was substituted for conditioned hybridoma clone A180
supernatant no immunostaining of cells or mitotic spindles could
be observed. Panel A, acquired with confocal settings used for
Figure 1; Panel B, acquired with confocal settings greatly
enhanced for detection of Dylight 549 nm (red). Arrows indicate
mitotic cells.
(TIF)
Figure S2 NQO1 immunostaining in BxPc-3 cells using
hybridoma supernatant or purified antibody. Identical
immunostaining of NQO1 in BxPc-3 cells was observed using
either (A) hybridoma tissue culture media from clone A180 diluted
1:10 or (B) affinity purified mouse IgG from clone A180 (Cell
Signaling Technologies) diluted 1:200.
(TIF)
Author Contributions
Conceived and designed the experiments: DS JK DR. Performed the
experiments: DS. Analyzed the data: DS JK DR. Contributed reagents/
materials/analysis tools: JK. Wrote the paper: DS JK DR.
References
1. Lind C, Cadenas E, Hochstein P, Ernster L (1990) DT-diaphorase: purification,
properties, and function. Methods Enzymol 186: 287–301.
2. Siegel D, Yan C, Ross D (2012) NAD(P)H:quinone oxidoreductase 1 (NQO1) in
the sensitivity and resistance to antitumor quinones. Biochem Pharmacol 83:
1033–1040.
3. Siegel D, Ross D (2000) Immunodetection of NAD(P)H:quinone oxidoredu ctase
1 (NQO1) in human tissues. Free Radic Biol Med 29: 246–253.
4. Schlager JJ, Powis G (1990) Cytosolic NAD(P)H:(quinone-ac ceptor)oxidoreduc-
tase in human normal and tumor tissue: effects of cigarette smoking and alcohol.
Int J Cancer 45: 403–409.
5. Siegel D, McGuinness SM, Winski SL, Ross D (1999) Genotype-phenotype
relationships in studies of a polymorphism in NAD(P)H:quinone oxidoreductase
1. Pharmacogenetics 9: 113–121.
6. Tsvetkov P, Adamovich Y, Elliott E, Shaul Y (2011) E3 ligase STUB1/CHIP
regulates NAD(P)H:quinone oxidoreductase 1 (NQO1) accumulation in aged
brain, a process impaired in certain Alzheimer disease patients. J Biol Chem 286:
8839–8845.
7. Siegel D, Anwar A, Winski SL, Kepa JK, Zolman KL, et al. (2001) Rapid
polyubiquitination and proteasomal degradation of a mutant form of
NAD(P)H:quinone oxidoreductase 1. Mol Pharmacol 59: 263–268.
8. Nioi P, Hayes JD (2004) Contribution of NAD(P)H:quinone oxidoreductase 1 to
protection against carcinogenesis, and regulation of its gene by the Nrf2 basic-
region leucine zipper and the arylhydrocarbon receptor basic helix-loop-helix
transcription factors. Mutat Res 555: 149–171.
9. Ade N, Leon F, Pallardy M, Peiffer JL, Kerdine-Romer S, et al. (2009) HMOX1
and NQO1 genes are upregulated in response to contact sensitizers in dendritic
cells and THP-1 cell line: role of the Keap1/Nrf2 pathway. Toxicol Sci 107:
451–460.
10. Siegel D, Gustafson DL, Dehn DL, Han JY, Boonchoong P, et al. (2004)
NAD(P)H:quinone oxidoreductase 1: role as a superoxide scavenger. Mol
Pharmacol 65: 1238–1247.
11. Siegel D, Bolton EM, Burr JA, Liebler DC, Ross D (1997) The redu ction of
alpha-tocopherolquinone by human NAD(P)H: quinone oxidoreductase: the
role of alpha-tocopherolhydroquinone as a cellular antioxidant. Mol Pharmacol
52: 300–305.
12. Chan TS, Teng S, Wilson JX, Galati G, Khan S, et al. (2002) Coenzyme Q
cytoprotective mechanisms for mitochondrial complex I cytopathies involves
NAD(P)H: quinone oxidoreductase 1(NQO1). Free Radic Res 36: 421–427.
13. Asher G, Lotem J, Cohen B, Sachs L, Shaul Y (2001) Regulation of p53 stability
and p53-dependent apoptosis by NADH quinone oxidoreductase 1. Proc Natl
Acad Sci U S A 98: 1188–1193.
14. Asher G, Tsvetkov P, Kahana C, Shaul Y (2005) A mechanism of ubiquitin-
independent proteasomal degradation of the tumor suppressors p53 and p73.
Genes Dev 19: 316–321.
15. Winski SL, Koutalos Y, Bentley DL, Ross D (2002) Subcellular localization of
NAD(P)H:quinone oxidoreductase 1 in human cancer cells. Cancer Res 62:
1420–1424.
16. Khutornenko AA, Roudko VV, Chernyak BV, Vartapetian AB, Chumakov PM,
et al. (2010) Pyrimidine biosynthesis links mitochondrial respiration to the p53
pathway. Proc Natl Acad Sci U S A 107: 12828–12833.
17. Siegel D, Shieh B, Yan C, Kepa JK, Ross D (2011) Role for NAD(P)H:quinone
oxidoreductase 1 and manganese-dependent superoxide dismutase in 17-
(allylamino)-17-demethoxygeldanamycin-induced heat shock protein 90 inhibi-
tion in pancreatic cancer cells. J Pharmacol Exp Ther 336: 874–880.
18. Siegel D, Franklin WA, Ross D (1998) Immunohistochemical detection of
NAD(P)H:quinone oxidoreductase in human lung and lung tumors. Clin Cancer
Res 4: 2065–2070.
19. Wignall SM, Gray NS, Chang YT, Juarez L, Jacob R, et al. (2004) Identification
of a novel protein regulating microtubule stability through a chemical approach.
Chem Biol 11: 135–146.
20. Winski SL, Swann E, Hargreaves RH, Dehn DL, Butler J, et al. (2001)
Relationship between NAD(P)H:quinone oxidoreductase 1 (NQO1) levels in a
series of stably transfected cell lines and susceptibility to antitumor quinones.
Biochem Pharmacol 61: 1509–1516.
21. Sharp SY, Kelland LR, Valenti MR, Brunton LA, Hobbs S, et al. (2000)
Establishment of an isogenic human colon tumor model for NQO1 gene
expression: application to investigate the role of DT-diaphorase in bioreductive
drug activation in vitro and in vivo. Mol Pharmacol 58: 1146–1155.
22. Pfeiffer E, Metzler M (1996) Interaction of p-benzoquinone and p-biphenoqui-
none with microtubule proteins in vitro. Chem Biol Interact 102: 37–53.
23. Durkacz BW, Omidiji O, Gray DA, Shall S (1980) (ADP-ribose)n partic ipates in
DNA excision repair. Nature 283: 593–596.
24. Mao Z, Hine C, Tian X, Van Meter M, Au M, et al. (2011) SIRT6 promotes
DNA repair under stress by activating PARP1. Science 332: 1443–1446.
25. Wu N, Hanson SM, Francis DJ, Vishnivetskiy SA, Thibonnier M, et al. (2006)
Arrestin binding to calmodulin: a direct interaction between two ubiquitous
signaling proteins. J Mol Biol 364: 955–963.
26. Cao JN, Shafee N, Vickery L, Kaluz S, Ru N, et al. (2010) Mitogen-activated
protein/extracellular signal-regulated kinase kinase 1act/tubulin interaction is
an important determinant of mitotic stability in cultured HT1080 human
fibrosarcoma cells. Cancer Res 70: 6004–6014.
NQO1 Localizes to the Mitotic Spindle
PLOS ONE | www.plosone.org 7 September 2012 | Volume 7 | Issue 9 | e44861
27. Aizawa H, Emori Y, Murofushi H, Kawasaki H, Sakai H, et al. (1990)
Molecular cloning of a ubiquitously distributed microtubule-associated protein
with Mr 190,000. J Biol Chem 265: 13849–13855.
28. Sollner S, Macheroux P (2009) New roles of flavoproteins in molecular cell
biology: an unexpected role for quinone reductases as regulators of proteasomal
degradation. FEBS J 276: 4313–4324.
29. Asher G, Bercovich Z, Tsvetkov P, Shaul Y, Kahana C (2005) 20S proteasomal
degradation of ornithine decarboxylase is regulated by NQO1. Mol Cell 17:
645–655.
30. Hershkovitz Rokah O, Shpilberg O, Granot G (2010) NAD(P)H quinone
oxidoreductase protects TAp63gamma from proteasomal degradation and
regulates TAp63gamma-dependent growth arrest. PLoS One 5: e11401.
31. Patrick BA, Gong X, Jaiswal AK (2011) Disruption of NAD(P)H:quinone
oxidoreductase 1 gene in mice leads to 20S proteasomal degradation of p63
resulting in thinning of epithelium and chemical-induced skin cancer. Oncogene
30: 1098–1107.
32. Morris VB, Brammall J, Noble J, Reddel R (2000) p53 localizes to the
centrosomes and spindles of mitotic cells in the embryonic chick epiblast, human
cell lines, and a human primary culture: An immunofluorescence study. Exp Cell
Res 256: 122–130.
33. Lafarga M, Fernandez R, Mayo I, Berciano MT, Castano JG (2002) Proteasome
dynamics during cell cycle in rat Schwann cells. Glia 38: 313–328.
34. Bui CB, Shin J (2011) Persistent expression of Nqo1 by p62-mediated Nrf2
activation facilitates p53-dependent mitotic catastrophe. Biochem Biophys Res
Commun 412: 347–352.
35. Beall HD, Winski S, Swann E, Hudnott AR, Cotterill AS, et al. (1998)
Indolequinone antitumor agents: correlation between quinone structure, rate of
metabolism by recombinant human NAD(P)H:quinone oxidoreductase, and in
vitro cytotoxicity. J Med Chem 41: 4755–4766.
36. Schweitzer KM, Vicart P, Delouis C, Paulin D, Drager AM, et al. (1997)
Characterization of a newly established human bone marrow endothelial cell
line: distinct adhesive properties for hematopoietic progenitors compared with
human umbilical vein endothelial cells. Lab Invest 76: 25–36.
37. Cozens AL, Yezzi MJ, Kunzelmann K, Ohrui T, Chin L, et al. (1994) CFTR
expression and chloride secretion in polarized immortal human bronchial
epithelial cells. Am J Respir Cell Mol Biol 10: 38–47.
NQO1 Localizes to the Mitotic Spindle
PLOS ONE | www.plosone.org 8 September 2012 | Volume 7 | Issue 9 | e44861

Supplementary resources (2)

Data
September 2012
David Siegel · Jadwiga K Kepa · David Ross
Data
September 2012
David Siegel · Jadwiga K Kepa · David Ross
... NQO1 detoxifies the electrophilic quinones by a two-electron reduction to a redox-stable hydroquinone (Park et al., 2011;Ross & Siegel, 2021), ultimately preventing the generation of semiquinone radicals and ROS. Furthermore, NQO1 provides additional functions in maintaining the plasma membrane redox system, stabilizing proteins (Maeda et al., 2016), microtubules (Siegel et al., 2012), and mRNAs (Di Francesco et al., 2016), as well as exerting limited superoxide reductase activity (Zhu et al., 2007). ...
Article
Oxidative stress is implicated in the initiation, pathogenesis, and progression of various gastric inflammatory diseases (GID). The prevalence of these diseases remains a concern along with the increasing risks of adverse effects in current clinical interventions. Hence, new gastroprotective agents capable of inhibiting oxidative stress by modulating cellular defense systems such as the nuclear factor erythroid 2‐related factor 2 (Nrf2)/antioxidant response element (ARE) signaling pathway are critically needed to address these issues. A candidate to solve the present issue is xanthone, a natural compound that reportedly exerts gastroprotective effects via antioxidant, anti‐inflammatory, and cytoprotective mechanisms. Moreover, xanthone derivatives were shown to modulate the Nrf2/ARE signaling pathway to counter oxidative stress in both in vitro and in vivo models. Thirteen natural xanthones have demonstrated the ability to modulate the Nrf2/ARE signaling pathway and have high potential as lead compounds for GID as indicated by their in vivo gastroprotective action–particularly mangiferin ( 2 ), α‐mangostin ( 3 ), and γ‐mangostin ( 4 ). Further studies on these compounds are recommended to validate the Nrf2 modulatory ability in relation to their gastroprotective action.
... It has been proposed that the key role played by BDNF as an inducer of neuronal antioxidant responses entails crosstalk between RyRmediated Ca 2+ release and ROS [93]. One of the targets of Nrf2 signaling is the expression of Nqo1, an oxidoreductase that catalyzes the 2-electron reduction of quinones to hydroquinones, which is a cellular detoxification process [97]. Here, we show that the activity dependent increase in Bdnf expression mediated by GBZ is accompanied by an increase in the expression of Nqo1. ...
Preprint
Full-text available
Hippocampal neuronal activity generates dendritic and somatic Ca2+ signals, which depending on stimulus intensity, rapidly propagate to the nucleus and induce the expression of transcription factors and genes with crucial roles in cognitive functions. Soluble Amyloid-beta Oligomers (AβOs), the main synaptotoxins engaged in the pathogenesis of Alzheimer's disease, generate aberrant Ca2+ signals in primary hippocampal neurons, increase their oxidative tone and disrupt structural plasticity. Here, we explored the effects of sub-lethal AβOs concentrations on activity-generated nuclear Ca2+ signals and on the Ca2+-dependent expression of neuroprotective genes. To induce neuronal activity, neuron-enriched primary hippocampal cultures were treated with the GABAA receptor blocker gabazine (GBZ), and nuclear Ca2+ signals were measured in AβOs-treated or control neurons transfected with a genetically encoded nuclear Ca2+ sensor. Incubation (6 h) with AβOs significantly reduced the nuclear Ca2+ signals and the enhanced phosphorylation of cyclic AMP response element binding protein (CREB) induced by GBZ. Likewise, incubation (6 h) with AβOs significantly reduced the GBZ-induced increases in the mRNA levels of Neuronal Per Arnt Sim domain protein 4 (Npas4), Brain-derived Neurotrophic Factor (Bdnf), Ryanodine Receptor type-2 (RyR2), and the antioxidant enzyme NADPH-Quinone-Oxidoreductase (Nqo1). Based on these findings we propose that AβOs, by inhibiting the generation of activity induced nuclear Ca2+ signals, disrupt key neuroprotective gene expression pathways required for hippocampal-dependent learning and memory processes.
... It has been proposed that the key role played by BDNF as an inducer of neuronal antioxidant responses entails crosstalk between RyR-mediated Ca 2+ release and ROS [102]. One of the targets of Nrf2 signaling is the expression of Nqo1, an oxidoreductase that catalyzes the 2-electron reduction of quinones to hydroquinones, which is a cellular detoxification process [106]. Here, we show that the activity dependent increase in BDNF expression mediated by GBZ is accompanied by an increase in the expression of Nqo1. ...
Article
Full-text available
Hippocampal neuronal activity generates dendritic and somatic Ca2+ signals, which, depending on stimulus intensity, rapidly propagate to the nucleus and induce the expression of transcription factors and genes with crucial roles in cognitive functions. Soluble amyloid-beta oligomers (AβOs), the main synaptotoxins engaged in the pathogenesis of Alzheimer’s disease, generate aberrant Ca2+ signals in primary hippocampal neurons, increase their oxidative tone and disrupt structural plasticity. Here, we explored the effects of sub-lethal AβOs concentrations on activity-generated nuclear Ca2+ signals and on the Ca2+-dependent expression of neuroprotective genes. To induce neuronal activity, neuron-enriched primary hippocampal cultures were treated with the GABAA receptor blocker gabazine (GBZ), and nuclear Ca2+ signals were measured in AβOs-treated or control neurons transfected with a genetically encoded nuclear Ca2+ sensor. Incubation (6 h) with AβOs significantly reduced the nuclear Ca2+ signals and the enhanced phosphorylation of cyclic AMP response element-binding protein (CREB) induced by GBZ. Likewise, incubation (6 h) with AβOs significantly reduced the GBZ-induced increases in the mRNA levels of neuronal Per-Arnt-Sim domain protein 4 (Npas4), brain-derived neurotrophic factor (BDNF), ryanodine receptor type-2 (RyR2), and the antioxidant enzyme NADPH-quinone oxidoreductase (Nqo1). Based on these findings we propose that AβOs, by inhibiting the generation of activity-induced nuclear Ca2+ signals, disrupt key neuroprotective gene expression pathways required for hippocampal-dependent learning and memory processes.
... Some specific enzymes produced by ARE genes are ferritin, heme oxygenase 1 (HO-1), NAD(P)H quinone oxidoreductase (NQO1), thioredoxin reductase (TrxR), and glutathionemetabolizing genes (glutathione peroxidase (GPx), glutathione s-transferase (GST), γglutamyl transpeptidase (GGT), glutathione-disulfide reductase (GSR), and glutamyl cysteine ligase (GCL)) [103]. NAD(P)H quinone oxidoreductase 1 (NQO1) catalyzes the 2e − reduction of quinones to hydroquinones, catechols, and a broad range of organic compounds [104]. Its physiological function is to reduce the free radical load in cells and the detoxification of xenobiotics [105,106]. ...
Article
Full-text available
In this review, we have focused mainly on the study of their antioxidant and pro-oxidant capacity, and the analysis of the oxidation of the catechol group to o-quinone. The redox balance established between the different oxidase and reductase enzymes generates reducing species, H + and e − , and allows the oxidation of polyphenolic groups to quinones to be reversible. This continuous balance between these nucleophilic and electrophilic substances allows the activation of the NRF2/ARE axis, which regulates cellular antioxidant responses against oxidative stress, as well as cell proliferation. Understanding the ambivalent character of polyphenols, which can act simultaneously as antioxidants and pro-oxidants, will allow the design of specific therapies that can serve science and medicine in their tasks.
... Previously, our group reported that NQO1 in cancer cells induces rapid degradation of Aurora-A during mitotic progression, specifically demonstrating that an NQO1 deficiency leads to aneuploidy during mitotic progression in irradiated cancer cells [41]. In human cells, NQO1 is associated with mitotic spindles during mitotic progression [42] and has been further shown to regulate mitotic progression and response to mitotic stress through modulation of SIRT2 activity [43]. However, the mechanisms underlying the activity of NQO1 in cancer cell proliferation, in particular, cell cycle progression, have yet to be clarified. ...
Article
Full-text available
Rationale: Overexpression of NAD(P)H:quinone oxidoreductase 1 (NQO1) is associated with tumor cell proliferation and growth in several human cancer types. However, the molecular mechanisms underlying the activity of NQO1 in cell cycle progression are currently unclear. Here, we report a novel function of NQO1 in modulation of the cell cycle regulator, cyclin-dependent kinase subunit-1 (CKS1), at the G2/M phase through effects on the stability of c‑Fos. Methods: The roles of the NQO1/c-Fos/CKS1 signaling pathway in cell cycle progression were analyzed in cancer cells using synchronization of the cell cycle and flow cytometry. The mechanisms underlying NQO1/c-Fos/CKS1-mediated regulation of cell cycle progression in cancer cells were studied using siRNA approaches, overexpression systems, reporter assays, co-immunoprecipitation, pull-down assays, microarray analysis, and CDK1 kinase assays. In addition, publicly available data sets and immunohistochemistry were used to investigate the correlation between NQO1 expression levels and clinicopathological features in cancer patients. Results: Our results suggest that NQO1 directly interacts with the unstructured DNA-binding domain of c-Fos, which has been implicated in cancer proliferation, differentiation, and development as well as patient survival, and inhibits its proteasome-mediated degradation, thereby inducing CKS1 expression and regulation of cell cycle progression at the G2/M phase. Notably, a NQO1 deficiency in human cancer cell lines led to suppression of c-Fos-mediated CKS1 expression and cell cycle progression. Consistent with this, high NQO1 expression was correlated with increased CKS1 and poor prognosis in cancer patients. Conclusions: Collectively, our results support a novel regulatory role of NQO1 in the mechanism of cell cycle progression at the G2/M phase in cancer through effects on c‑Fos/CKS1 signaling.
Article
The ability to gain spatiotemporal information, and in some cases achieve spatiotemporal control, in the context of drug delivery makes theranostic fluorescent probes an attractive and intensely investigated research topic. This interest is reflected in the steep rise in publications on the topic that have appeared over the past decade. Theranostic fluorescent probes, in their various incarnations, generally comprise a fluorophore linked to a masked drug, in which the drug is released as the result of certain stimuli, with both intrinsic and extrinsic stimuli being reported. This release is then signaled by the emergence of a fluorescent signal. Importantly, the use of appropriate fluorophores has enabled not only this emerging fluorescence as a spatiotemporal marker for drug delivery but also has provided modalities useful in photodynamic, photothermal, and sonodynamic therapeutic applications. In this review we highlight recent work on theranostic fluorescent probes with a particular focus on probes that are activated in tumor microenvironments. We also summarize efforts to develop probes for other applications, such as neurodegenerative diseases and antibacterials. This review celebrates the diversity of designs reported to date, from discrete small-molecule systems to nanomaterials. Our aim is to provide insights into the potential clinical impact of this still-emerging research direction.
Article
Full-text available
NAD(P)H Quinone Dehydrogenase 1 (NQO1) plays a pivotal role in the regulation of neuronal function and synaptic plasticity, cellular adaptation to oxidative stress, neuroinflammatory and degenerative processes, and tumorigenesis in the central nervous system (CNS). Impairment of the NQO1 activity in the CNS can result in abnormal neurotransmitter release and clearance, increased oxidative stress, and aggravated cellular injury/death. Furthermore, it can cause disturbances in neural circuit function and synaptic neurotransmission. The abnormalities of NQO1 enzyme activity have been linked to the pathophysiological mechanisms of multiple neurological disorders, including Parkinson's disease, Alzheimer's disease, epilepsy, multiple sclerosis, cerebrovascular disease, traumatic brain injury, and brain malignancy. NQO1 contributes to various dimensions of tumorigenesis and treatment response in various brain tumors. The precise mechanisms through which abnormalities in NQO1 function contribute to these neurological disorders continue to be a subject of ongoing research. Building upon the existing knowledge, the present study reviews current investigations describing the role of NQO1 dysregulations in various neurological disorders. This study emphasizes the potential of NQO1 as a biomarker in diagnostic and prognostic approaches, as well as its suitability as a target for drug development strategies in neurological disorders.
Article
Pancreatic cancer is among the top five leading causes of cancer-related deaths worldwide, with low survival rates. Current therapies for pancreatic cancer lack tumor specificity, resulting in harmful effects on normal tissues. Therefore, developing tumor-specific agents for the treatment of pancreatic cancer is critical. NAD(P)H:quinone oxidoreductase 1 (NQO1), highly expressed in pancreatic cancers but not in normal tissues, makes NQO1 bioactivatable drugs a potential therapy for selectively killing NQO1-positive cancer cells. Our previous studies have revealed that novel NQO1 bioactivatable drug deoxynyboquinone (DNQ) is ten-fold more potent than the prototypic NQO1 bioactivatable drug β-lapachone in killing of NQO1-positive cancer cells. However, DNQ treatment results in high-grade methemoglobinemia, a significant side effect that limits clinical development. Here, we report for the first time on a DNQ derivative, isopentyl-deoxynboquinone (IP-DNQ), which selectively kills pancreatic ductal adenocarcinoma cells in an NQO1-dependent manner with equal potency to the parent DNQ. IP-DNQ evokes massive ROS production and oxidative DNA lesions that results in PARP1 hyperactivation, mitochondrial catastrophe and G2/M-phase arrest, leading to apoptotic and necrotic programmed cell death. Importantly, IP-DNQ treatment causes mild methemoglobinemia in vivo, with a three-fold improvement in the maximum tolerated dose compared to DNQ, while significantly suppresses tumor growth and extends the lifespan of mice in subcutaneous and orthotopic pancreatic cancer xenograft models. Our study demonstrates that IP-DNQ is a promising therapy for NQO1-positive pancreatic cancers and may enhance the efficacy of other anticancer drugs. IP-DNQ represents a novel approach to treating pancreatic cancer with the potential to improve patient outcomes.
Article
Quinone compounds have been the subject of extensive research due to their remarkable efficiency and prospective use as drugs and in a variety of fields. In this study, we report the spectroscopic characterization, electronic structure, ADMET evaluation, and molecular docking assessment of 2,6- Bis(phenylamino)-4-(iminophenyl)benzoquinone as an anticancer drug. By using DFT investigations, the vibrational wavenumbers were calculated and utilized to assign vibrational bands, which were found to be in good accordance with the experimentally observed data. The analysis of the UV-Vis spectra revealing an absorption peak from electronic transitions HOMO→LUMO at 547 nm is found to be in good conformity with its experimental value. The HOMO and LUMO frontier molecular orbitals and their associated energies highlighted the mechanism of charge transfer within the molecule and revealed a small energy gap. The chemically reactive sites identified by the MEP surface helped predict the spots of the molecule’s biological activity. According to NBO analysis, the π C9-C10 → π∗ C11-C12 interaction has the maximum energy stability with 23.74 Kcal/mol., due to π electron delocalization within the ring. At the same level of theory, third-order NLO polarizability was found to be 4-fold stronger than the third-order of P-NA (a prototype NLO molecule). The potential for a safe oral bioavailability drug was identified by computing ADMET parameters and evaluating drug-likeness based on Lipinski’s rule of five. The molecular docking study found that the molecule binding to NQO1 receptor protein with superior binding energy -8.69 kcal/mol than previously studied Quinone derivatives. Additionally, molecular dynamics simulations were performed to test the dynamic behavior of the ligand-NQO1 complex. The complex was stable in the binding pocket of the receptor proteins, according to the analysis of the simulation outcomes such as RMSD and RMSF. These results suggests using this molecule as a potential anticancer drug due to its high capability to inhibit the NQO1 enzyme.
Article
Full-text available
Sirtuin 6 (SIRT6) is a mammalian homolog of the yeast Sir2 deacetylase. Mice deficient for SIRT6 exhibit genome instability. Here, we show that in mammalian cells subjected to oxidative stress SIRT6 is recruited to the sites of DNA double-strand breaks (DSBs) and stimulates DSB repair, through both nonhomologous end joining and homologous recombination. Our results indicate that SIRT6 physically associates with poly[adenosine diphosphate (ADP)-ribose] polymerase 1 (PARP1) and mono-ADP-ribosylates PARP1 on lysine residue 521, thereby stimulating PARP1 poly-ADP-ribosylase activity and enhancing DSB repair under oxidative stress.
Article
Full-text available
NAD(P)H:quinone oxidoreductase 1 (NQO1) is a flavoenzyme that is important in maintaining the cellular redox state and regulating protein degradation. The NQO1 polymorphism C609T has been associated with increased susceptibility to various age-related pathologies. We show here that NQO1 protein level is regulated by the E3 ligase STUB1/CHIP (C terminus of Hsc70-interacting protein). NQO1 binds STUB1 via the Hsc70-interacting domain (tetratricopeptide repeat domain) and undergoes ubiquitination and degradation. We demonstrate here that the product of the C609T polymorphism (P187S) is a stronger STUB1 interactor with increased susceptibility to ubiquitination by the E3 ligase STUB1. Furthermore, age-dependent decrease of STUB1 correlates with increased NQO1 accumulation. Remarkably, examination of hippocampi from Alzheimer disease patients revealed that in half of the cases examined the NQO1 protein level was undetectable due to C609T polymorphism, suggesting that the age-dependent accumulation of NQO1 is impaired in certain Alzheimer disease patients.
Article
Despite the extensive interest in NADPH:quinone oxidoreductase (NQO1, DT-diaphorase), there is little immunohistochemical information regarding its distribution in either normal human tissues or in human tumors. Using immunohistochemistry (IHC), we have examined cell-specific expression of NQO1 in many normal tissues and tumors as a step toward defining the distribution of NQO1 in humans. NQO1 was detected by IHC in respiratory, breast duct, thyroid follicle, and colonic epithelium, as well as in the corneal and lens epithelium of the eye. NQO1 was also detected by IHC in vascular endothelium in all tissues examined. NQO1 could also readily be detected in the endothelial lining of the aorta but was not detected using immunoblot analysis in the myocardium. Adipocytes stained positive for NQO1, and the enzyme was also detected by both IHC and immunoblot analysis in parasympathetic ganglia in the small intestine and in the optic nerve and nerve fibers. NQO1 was not highly expressed in five different human liver samples using immunoblot analysis, whereas studies using IHC demonstrated only trace NQO1 staining in isolated bile duct epithelium. NQO1 expresion was also examined by IHC in a variety of solid tumors. Marked NQO1 staining was detected in solid tumors from thyroid, adrenal, breast, ovarian, colon, and cornea and in non–small cell lung cancers. The NQO1 content of many solid tumors supports the use of NQO1-directed anticancer agents for therapeutic purposes, but the distribution of NQO1 in normal tissues suggests that potential adverse effects of such agents need to be carefully monitored in preclinical studies.
Article
The NAD(P)H:quinone oxidoreductase 1 (NQO1) genotype-phenotype relationship was examined in individuals with a polymorphism in NQO1. The polymorphism comprises a C to T base change at position 609 of the human NQO1 cDNA (C609T) and codes for a proline to serine substitution in the amino acid structure of the NQO1 protein. Genotyping was performed by polymerase chain reaction-restriction fragment length polymorphism analysis of genomic DNA. Phenotyping was performed using enzyme activity assays and/or immunoblotting of human tumor cell lines and of saliva and bone marrow samples from healthy donors. Phenotyping of uninvolved lung and lung tumors from archived biopsy material was performed by immunohistochemistry. NQO1 activity and protein could be detected in wild-type (C/C) human tumor cells (HT-29) under conditions where NQO1 protein could not be detected in cells (BE) homozygous for the C609T change (T/T). Trace levels of NQO1 protein could be detected in BE cells; however, when immunoblots were subjected to chemiluminescence detection for prolonged periods. In saliva samples from 11 individuals carrying the homozygous C609T change (T/T), no NQO1 protein could be detected even after prolonged chemiluminescence detection. The amount of NQO1 protein present in saliva was quantified and found to be significantly less in heterozygous individuals (C/T) than in wild-type individuals (C/C). In bone marrow stromal cultures, both NQO1 activity and protein could be detected in heterozygotes (C/T) and in wild-type (C/C) samples. In a bone marrow stromal culture from an individual genotyped as T/T at position 609, no NQO1 protein or activity could be detected. NQO1 is elevated in non-small cell lung cancers and could be readily observed as intense immunostaining throughout lung adenocarcinomas genotyped as C/C but no immunostaining could be detected in adenocarcinomas genotyped as T/T at position 609. NQO1 is expressed in normal human lung but is localized to respiratory epithelium and to vascular endothelium. In normal lung tissue from individuals genotyped as T/T, no or faint immunostaining for NQO1 could be detected in either respiratory epithelium or vascular endothelium. These results demonstrate that tissues from individuals homozygous for the C609T change have no detectable or, at best, only trace amounts of NQO1 protein and are devoid of NQO1 activity. Pharmacogenetics 9:113-121 (C) 1999 Lippincott Williams & Wilkins (C) 1999 Lippincott Williams & Wilkins, Inc.
Article
Quinones represent a large and diverse class of antitumor drugs and many quinones are approved for clinical use or are currently undergoing evaluation in clinical trials. For many quinones reduction to the hydroquinone has been shown to play a key role in their antitumor activity. The two-electron reduction of quinones by NQO1 has been shown to be an efficient pathway to hydroquinone formation. NQO1 is expressed at high levels in many human solid tumors making this enzyme ideally suited for intracellular drug activation. Cellular levels of NQO1 are influenced by the NQO1*2 polymorphism. Individuals homozygous for the NQO1*2 allele are NQO1 null and homozygous NQO1*2*2 cell lines have been shown to be more resistant to antitumor quinones when compared to isogenic cell lines overexpressing NQO1. In this review we will discuss the role of NQO1 in the sensitivity and resistance of human cancers to the quinone antitumor drugs mitomycin C, β-lapachone and the benzoquinone ansamycin class of Hsp90 inhibitors including 17-AAG. The role of NQO1 in the bioreductive activation of mitomycin C remains controversial but pre-clinical data strongly suggests a role for NQO1 in the activation of β-lapachone and the benzoquinone ansamycin class of Hsp90 inhibitors. Despite a large volume of preclinical data demonstrating that NQO1 is an important determinant of sensitivity to these antitumor quinones there is little information on whether the clinical response to these agents is influenced by the NQO1*2 polymorphism. The availability of simple assays for the determination of the NQO1*2 polymorphism should facilitate clinical testing of this hypothesis.
Article
Prolonged mitosis due to aberrant chromosome segregation permits cells to enter the G1 phase without cytokinesis and subsequently triggers the p53-dependent cell death program, known as mitotic catastrophe. Cells which fail to go through mitotic catastrophe create aneuploidy, posing a risk of oncogenesis. In the present report, we show that p62-mediated non-canonical activation of Nrf2 leads to the persistent expression of Nqo1, which plays a critical role for p53 stabilization during mitotic catastrophe. With prolonged exposure to nocodazole, a microtubule-depolymerizing agent, p62-deficient HCT116 cells exhibited an accumulation of a polyploid population with a limited appearance of apoptotic cells, which was attributable to the attenuated stabilization of p53. Combinatorial gene manipulation analysis verified that the regulatory cascade with a hierarchy of p62-Keap1-Nrf2-Nqo1 is required for p53 stabilization for mitotic catastrophe. This is consistent with the role of Nqo1 as a gatekeeper for proteasomal degradation of p53. Thus, we demonstrate for the first time the functional connection between the non-canonical Nrf2 pathway and p53-dependent cell death program upon prolonged mitosis.
Article
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.
Article
NAD(P)H:quinone oxidoreductase 1 (NQO1) is a cytosolic enzyme that protects cells against chemical and radiation-induced oxidative stress and skin cancer. Disruption of NQO1 gene in mice showed thinning of skin epithelium and loss of cytokeratin 14, an early marker of skin differentiation. Immunohistochemistry and western analysis demonstrated downregulation of p63 in NQO1-/- mouse skin, as compared with wild-type (WT) mouse. Further analysis including modulation of NQO1 expression revealed a direct correlation between the levels of NQO1 and p63 in skin-derived keratinocytes and dermal fibroblasts. Modulation of proteasomal activity revealed that p63 is degraded by 20S proteasome and that this degradation is significantly rescued by NQO1. Coimmunoprecipitation studies showed that NQO1 interacts directly with p63 but not 20S to protect against this degradation. In addition, benzo[a]pyrene treatment led to induction of NQO1 and stabilization of p63 in WT but not in NQO1-/- mouse skin and keratinocytes. These data suggest that NQO1 controls stabilization of p63 and progression towards keratinocyte differentiation leading to normal skin development and presumably skin carcinogenesis.