Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

Nitric Oxide 24 (2011) 1–7

Contents lists available at ScienceDirect

Nitric Oxide
journal homepage: www.elsevier.com/locate/yniox

Review

Dual role of NO donors in the reversal of tumor cell resistance and EMT:
Downregulation of the NF-jB/Snail/YY1/RKIP circuitry
Benjamin Bonavida ⇑, Stavroula Baritaki
Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, Johnson Comprehensive Cancer Center, University of California,
Los Angeles, CA 90095, United States

a r t i c l e i n f o a b s t r a c t

Article history: Several studies have implicated the role of Nitric Oxide (NO) in the regulation of tumor cell behavior
Received 18 August 2010 and have shown that NO either promotes or inhibits tumorigenesis. These conflicting findings have
Revised 29 September 2010 been resolved, in part, by the levels of NO used such that low levels promote tumor growth and high
Available online 8 October 2010
levels inhibit tumor growth. Our studies have focused on the use of high levels of NO provided pri-
marily by the NO donor, DETANONOate. We have shown that treatment of resistant tumor cells with
Keywords: DETANONOate sensitizes them to apoptosis by both chemotherapeutic drugs and cytotoxic immuno-
EMT
therapeutic ligands. The underlying mechanisms by which NO sensitizes tumor cells to apoptosis
Resistance
Therapeutic targets
were shown to be regulated, in part, by NO-mediated inhibition of the NF-jB survival/anti-apoptotic
NF-jB/Snail/YY1/RKIP circuitry pathways and downstream of NF-jB by inhibition of the transcription factor Yin Yang 1 (YY1). In
Apoptosis addition to NO-induced sensitization to apoptosis, we have also shown that NO induced the expres-
sion of the metastasis-suppressor/immunosurveillance cancer gene product, Raf-1 kinase inhibitor
protein (RKIP). Overexpression of RKIP mimics NO in tumor cells-induced sensitization to apoptosis.
The induction of RKIP by NO was the result of the inhibition of the RKIP repressor, Snail, downstream
of NF-jB. These findings established the presence of a dysregulated NF-jB/Snail/YY1/ RKIP circuitry in
resistance and that treatment with NO modifies this loop in tumor cells in favor of the inhibition of
tumor cell survival and the response to cytotoxic drugs. Noteworthy, the NF-jB/Snail/YY1/RKIP loop
consists of gene products that regulate the epithelial to mesenchymal transition (EMT) and, thus,
tumor metastasis. Hence, we have found that treatment of metastatic cancer cell lines with DETAN-
ONOate inhibited the EMT phenotype, through both the inhibition of the metastasis-inducers, NF-jB
and Snail and the induction of the metastasis-suppressor, RKIP. Altogether, the above findings estab-
lish, for the first time, the dual role of high levels of NO in the sensitization of tumor cells to apop-
totic stimuli as well as inhibition of EMT. Hence, NO donors may be considered as novel potential
therapeutic agents with dual roles in the treatment of patients with refractory cancer and in the pre-
vention of the initiation of the metastatic cascade via EMT.
Ó 2010 Elsevier Inc. All rights reserved.

Contents

Introduction. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
Establishment of the NF-jB/Snail/YY1/RKIP circuitry in cancer cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
Role of NF-jB . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
Role of Snail. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
Role of YY1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
Role of RKIP . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
The NO donor, DETANONOate, reverses tumor cell resistance for apoptosis and inhibits EMT. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3

Abbreviations: Bcl-xL, B-cell leukemia-xL; CDDP, cis-diammine-dichloro-platinum; DETANONOate, (Z)-1-[2-(2-aminoethyl)-N-(2-ammonio-ethyl) amino] diazen-1-ium-
1, 2-diolate; DHMEQ, dehydroxymethylepoxyquinomicin; DR, death receptor; ERK, extracellular signal-regulated kinase; FasL, Fas ligand; IAP, inhibitor of apoptosis protein;
LNCaP, lymph node carcinoma of prostate; MAPK, Mitogen-activated protein kinases; MEK, MAP kinase kinase; NF-jB, nuclear factor-jB; si, small interfering; TRAIL, TNF-
related apoptosis-inducing ligand; XIAP, X-linked inhibitor of apoptosis.
⇑ Corresponding author. Fax: +1 310 206 2791.
E-mail address: bbonavida@mednet.ucla.edu (B. Bonavida).

1089-8603/$ - see front matter Ó 2010 Elsevier Inc. All rights reserved.
doi:10.1016/j.niox.2010.10.001
2 B. Bonavida, S. Baritaki / Nitric Oxide 24 (2011) 1–7

NO-mediated sensitization of tumor cells to apoptosis by chemotherapeutic drugs and immunotherapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4


Role of NF-jB . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
Role of Snail . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
Role of YY1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
The role of RKIP . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
NO-mediated inhibition of EMT in metastatic cell lines. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
Role of NF-jB . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
Role of Snail . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
Role of YY1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
Role of RKIP . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
In vivo validation of in vitro findings obtained with DETANONOate . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
Overall conclusions and future directions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6

Introduction of the resistance (through NO-mediated sensitization to apoptotic


stimuli) and inhibition of the EMT phenotype. This review will
During the last decades, several therapeutics strategies have briefly discuss the above circuitry and the role of each of the in-
been developed for the treatment of patients with cancer. Such ap- volved gene products in the regulation of resistance and EMT.
proaches resulted in significant clinical responses and, sometimes,
prolongation of survival. However, subsets of patients do not ini-
tially respond or develop resistance, thus, becoming refractory to Establishment of the NF-jB/Snail/YY1/RKIP circuitry in cancer
subsequent treatments. Further, the non-responsive patients usu- cells
ally develop metastasis and are also unresponsive to treatments
[1]. Hence, there is an urgent need to develop new therapeutic Based on studies reported by us and others, we have estab-
interventions to overcome both the resistance and metastasis. lished in tumor cells the presence of a dysregulated NF-jB/
One of the main underlying mechanisms in the development of Snail/YY1/RKIP loop [9–13]. This loop was found to be pivotal in
resistance in cancer patients is the acquisition by the tumor cells the regulation of tumor cell sensitivity to cytotoxic apoptotic
of strategies to inhibit the apoptotic pathways. Dysregulation of stimuli (for example, chemotherapeutics drugs and cytotoxic lym-
the apoptotic signaling pathways is common in cancer and it has phocytes/ligands). In addition, we have established that this loop
been implicated in the pathogenesis and progression of tumors. is also involved in the regulation of EMT [9]. Below, we briefly de-
These include (a) cell accumulation caused by failure of normal scribe how the above loop was established and demonstrate that
cells turnover mechanisms (programmed cell death) and, thus, each gene product of this loop regulates the remaining gene
creating a permissive environment for genetic instability and products.
oncogene activation (b) promotion of tumor cell-resistance to
immune attack (c) contribution of tumor cell-resistance to chemo- Role of NF-jB
therapy and radiation (d) tumor cells’ ability to survive in the face
of hypoxia and (e) fostering tumor metastasis by allowing the cells Cancer cells have been shown to exhibit a constitutively hyper-
to survive in a detached state [2–6]. activated NF-jB survival signaling pathways [14]. This hyperacti-
In metastasis, the tumor cells invade the basement membrane vation results downstream in the transcriptional activation of
and circulatory system, extravasate through the endothelial lining various anti-apoptotic gene products including Bcl-xL, XIAP and
of the blood vessels and form colonies in tissue sites. The migration Yin Yang 1 (YY1) [15,16]. In addition, NF-jB has been reported to
of the epithelial layer into the mesenchymal layer before reaching regulate positively EMT and metastasis [17–19]. EMT is induced,
the basement membrane is known as ‘‘epithelial to mesenchymal in part, directly via NF-jB-mediated activation of the transcription
transition’’ (EMT) [7]. During EMT, the polarized epithelial cells ac- of the EMT-inducer Snail [20], and indirectly via downregulation of
quire certain attributes of mesenchymal cells and, thus, are able to the expressions of the metastasis-suppressor cancer gene products,
penetrate the mesenchymal layer through the basement mem- RKIP and E-cadherin [21,22]. We and others have shown that inhi-
brane and invade neighboring tissues. Breaching of the mesenchy- bition of NF-jB activity by different agents (e.g. drugs, specific
mal layers leads to infiltration into the blood vessels and migration inhibitors, siRNA silencing, etc.) results in the reversal of tumor
to other points of the body [8]. resistance to apoptotic stimuli and inhibition of cancer cell inva-
The objective of our current studies has been to delineate the sion and migration [23–32]. We have reported that one of the
underlying biochemical, molecular, and genetic mechanisms that underlying mechanisms by which NF-jB inhibition by NO sup-
are responsible for the resistance in cancer and the development presses tumor cell resistance and metastasis is through inhibition
of the EMT phenotype and subsequent metastasis. These studies of the downstream targets Snail and YY1 and induction of RKIP
may lead to the discovery of novel targets for therapeutic interven- [9,31–33] (see scheme Fig. 1A).
tions in the reversal of resistance and inhibition of EMT. We have
established in model tumor cell lines the existence of a dysregu- Role of Snail
lated NF-jB/Snail/YY1/RKIP circuitry. This circuitry was found to
regulate both tumor cell resistance and EMT. Treatment of the Snail is a member of the Snail superfamily of zinc finger tran-
tumor cells with the NO donor, DETANONOate, modifies this scription factors. It has been shown to play a role in embryonic
circuitry and associated gene products and results in the reversal development, neural differentiation, cell division and cell survival
B. Bonavida, S. Baritaki / Nitric Oxide 24 (2011) 1–7 3

tumor resistance to the above apoptotic stimuli has been shown


by direct silencing of YY1 using siRNA, which resensitized the
resistant tumor cells to apoptosis [43]. In contrast, YY1 has been
reported to activate the transcription of the EMT-inducer Snail
[36], therefore, suggesting that YY1 might be also involved in the
regulation of EMT during cancer metastasis (see scheme Fig. 1C).

Role of RKIP

The Raf-1 kinase inhibitory protein (RKIP) is a member of the


phosphatidylethanolamine-binding protein family (PEBP). It is a
small cytosolic protein that was initially shown to play a role in
lipid metabolism and phospholipid membrane biogenesis [44]. Al-
most a decade ago, RKIP was first reported to function as an inhib-
itor of both the Raf-1/MEK/ERK and NF-jB survival pathways
[45–47]. Later, Lorenz et al. [48] demonstrated that RKIP also
regulates negatively the activation of G-protein coupled receptors
(GPCRs), resulting in attenuation of GPCR-mediated signaling. RKIP
has been further shown to regulate tumor cell sensitivity to apop-
totic stimuli including chemotherapy [49]. In addition, our studies
have identified RKIP as an immunosurveillance gene product
based on RKIP involvement in the regulation of tumor cell resis-
Fig. 1. Effect of DETANONOate on the expression and activity of each of the gene
products involved in the NF-jB/Snail/YY1/RKIP loop and their intrarelationships.
tance to cytotoxic immune ligands, such as TRAIL [9,33]. RKIP
(A) The inhibition of NF-jB activity by DETANONOate results in the transcriptional expression is significantly downregulated in several malignancies
repression of its downstream targets Snail and YY1. In addition, the DETANONOate- and almost absent in metastatic tumors [50–55]. RKIP overexpres-
mediated NF-jB inhibition also induces RKIP expression via de-repression of its sion in in vitro and in vivo tumor models has shown significant
promoter by Snail. (B) DETANONOate inhibits the expression and activity of Snail
anti-metastatic effects, thus supporting the role of RKIP as a
through NF-jB inhibition and S-nitrosylation, respectively. Snail inhibition, in turn,
derepresses the RKIP promoter and increases the RKIP levels. The RKIP induction ‘‘metastasis-suppressor’’ gene product [44,50,54]. We have further
results in further inhibition of NF-jB and its downstream targets YY1 and Snail. (C) demonstrated that RKIP overexpression directly correlates with
In addition to NF-jB and Snail, DETANONOate directly S-nitrosylates and deacti- the inhibition of EMT and reversal of tumor resistance to apoptotic
vates YY1. Since YY1 functions as a transcriptional activator of Snail, the
stimuli, namely, through a common mechanism which involves
DETANONOate-mediated inhibition of YY1 results in Snail downregulation and
subsequent induction of RKIP. The overexpression of RKIP, in turn, further inhibits
inhibition of the expressions and activities of NF-jB, Snail, and
the NF-jB activity. (D) As mentioned above, RKIP induction by DETANONOate has a YY1 [9,31,33] (see scheme Fig. 1D).
direct inhibitory effect on NF-jB activation which in turn leads to decreased Overall, the above findings clearly establish the tightly con-
expression of Snail and YY1. trolled NF-jB/Snail/YY1/RKIP loop that is dysregulated in cancer
cells and that is responsible, in part, for tumor cell survival and
resistance and the initiation of EMT.
[34]. It also plays an important role in the acquisition of invasion
and migratory properties by cancer cells during tumor progression
by triggering EMT [35]. Snail transcription is activated by NF-jB The NO donor, DETANONOate, reverses tumor cell resistance for
and YY1 [20,36], while it acts directly as a transcriptional repressor apoptosis and inhibits EMT
of the metastasis-suppressor gene products E-cadherin and RKIP
[21,22]. Our findings demonstrate that DETANONOate-mediated We and others have reported that treatment of tumor cells with
inhibition of Snail and de-repression of E-cadherin is only one high levels of NO [56] derived by DETANONOate at the concentra-
mechanism by which DETANONOate upregulates E-cadherin tions of 500–1000 mM inhibited NF-jB activity through S-nitrosy-
expression. We do not rule out other pathways regulated by lation of its p50 subunit [10,11,13,57]. Thus, we hypothesized that
DETANONOate in the regulation of E-cadherin. Snail silencing has treatment of tumor cells with NO will result in the modification of
been shown to result not only in reversal of the mesenchymal can- the NF-jB/Snail/YY1/RKI loop by inhibiting downstream the
cer cell phenotype, but also in resensitization of resistant tumor expressions and activities of Snail and YY1 and resulting in the
cells to apoptotic stimuli [9,31,37]. We have reported that inhibi- induction of RKIP. RKIP, in turn, will further potentiate the inacti-
tion of both EMT and resistance by Snail silencing is partially vation of NF-jB and its downstream gene targets. Hence, such ef-
attributed to induction of RKIP, which in turn results in inhibition fects by NO will result in the inhibition of the anti-apoptotic
of NF-jB and YY1 [9,31,33] (see scheme Fig. 1B). mechanisms as well as inhibition of EMT (see general scheme of
NO effects in Fig. 2).
Role of YY1 Below, we will describe briefly the experimental data that dem-
onstrated, following treatment of resistant and metastatic prostate
Yin Yang 1 (YY1) is a multifunctional DNA-binding protein that tumor cell lines, that the NO donor, DETANONOate, results in the
can activate, repress, or initiate transcription depending on the sensitization of tumor cells to apoptosis and inhibits the EMT phe-
context in which it binds (directly or indirectly through complex- notype. The role of each of the gene products that is implicated in
ing with other DNA-binding proteins) [38]. YY1 has been the NF-jB/Snail/YY1/RKIP circuitry has been examined and our
documented in the repression of several genes including IFN-c, data demonstrate that each gene product regulates both tumor cell
GM-CSF and IL-3 [39–41]. We have also identified YY1 as a resistance and EMT.
transcriptional repressor of the death receptors Fas and DR5, thus DETANONOate is an NO donor which belongs to the family of
suggesting that YY1 might have a critical role in the regulation of diazoniumdiolates (formerly NONOates). It spontaneously dissoci-
tumor cell resistance to cytotoxic immune ligands, such as FasL ates in a pH-dependent first order process with a half life of 20 h
and TRAIL [11,42,43]. The direct role of YY1 in the regulation of and 56 h at 37 °C and 22–25 °C, pH 7.4, respectively, to liberate
4 B. Bonavida, S. Baritaki / Nitric Oxide 24 (2011) 1–7

Fig. 2. Schematic diagram of the crosstalks among the members of the NF-jB/Snail/YY1/RKIP loop in the regulation of tumor resistance to apoptotic stimuli and EMT by
DETANONOate. NO mediates its biological effect on reversal of tumor cell resistance and EMT mainly by interfering with components of the NF-jB/Snail/YY1/RKIP circuit.
Tumor cells with highly resistant and metastatic phenotypes show constitutive activation of the NF-jB pathway and its downstream transcriptional targets YY1, Snail and
several anti-apoptotic gene products such as Bcl-xL, XIAP and survivin. The above gene products have been linked directly and indirectly with acquisition of tumor cell
resistance to both chemotherapy and immunotherapy and induction of EMT. Snail and YY1 act as transcriptional repressors of RKIP and death receptor genes such as DR5 and
Fas, respectively. Snail also acts as an essential initiator of EMT via inhibiting the transcription of the metastasis-suppressor genes RKIP and E-cadherin, while it induces
directly and/or indirectly the expression of mesenchymal markers. We show that NO inhibits the expressions and activities of NF-jB, Snail and YY1 and induces RKIP.
Induction of RKIP has been shown to resensitize resistant tumors to apoptosis mediated by drugs and/or cytotoxic death ligands such as TRAIL, and to reverse EMT. RKIP
induction also further facilitates the inhibition of NF- jB and its targeted genes Snail and YY1. Overall, the NO-mediated regulation of the NF-jB/Snail/YY1/RKIP loop via the
above mechanism results in reversal of tumor cell resistance to apoptotic stimuli and inhibits the migratory and invasive properties of metastatic tumor cells, thus it inhibits
EMT. Green lines correspond to the NO-mediated effects on the indicated gene products, while red lines correspond to the constitutive basil levels in tumor cells in the
absence of NO.

two moles of NO per mole of the parent compound, DETANONOate Role of NF-jB
[58,59]. The decomposition is not catalyzed by thiols or biological Treatment of our cell lines with DETANONOate resulted in the
tissues, unless specifically designed to and because NO release fol- inhibition of NF-jB activity as a result of its inhibition of its
lows simple first-order kinetics [60]. The rate of NO release can be DNA-binding activity [10,11]. DETANONOate inhibited the NF-jB
accurately predicted. This can be achieved via specific modification activity part, via DETANONOate-induced S-nitrosylation of p50
of the NONOate structure, which can stabilize the drug in solution [10]. DETANONOate also resulted in significant inhibition of NF-
and potentially engender a selective NO release in different organs, jB regulated anti-apoptotic gene products such as Bcl-xL and XIAP
vascular beds, or specific cell types [61,62]. [10,11,13]. The direct role of NF-jB in DETANONOate-mediated
At 37 degree this is equal to 28 uM NO relesaed each second, one might use this as a tumor cell sensitization was corroborated with experiments dem-
comparison instead or together with SNAP. If the peak concentratrion is of importance it might
be possible to accieve similar leverls with PDNO infusions in vivo. 20 = 72 000 sek. onstrating that treatment of the tumor cell lines with the specific
NO-mediated sensitization of tumor cells to apoptosis by NF-jB inhibitor, DHMEQ, mimicked DETANONOate in tumor cell
chemotherapeutic drugs and immunotherapy sensitization to both CDDP and TRAIL and inhibition of its down-
stream anti-apoptotic targets XIAP, Bcl-xL and survivin [10,11]
We have selected to utilize the chemotherapeutic drug, cis- (unpublished data) (Fig. 1A).
platinum (CDDP) and immunotherapeutic ligand TRAIL as repre-
sentatives for chemotherapy and immunotherapy, respectively. Role of Snail
We demonstrate that treatment of the resistant cells with Treatment of resistant cells with DETANONOate had a signifi-
DETANONOate resulted in tumor cell sensitization to apoptosis cant negative impact on the expression of Snail mRNA and protein
by CDDP and TRAIL, as assessed by activation of the effector cas- levels [unpublished data]. The DETANONOate-mediated inhibition
pase 3. The reversal of tumor resistance to the apoptotic stimuli of Snail expression may be, in part, attributed to inhibition of its
used was a function of both the concentrations of DETANONOate transcriptional activator, NF-jB. In addition, DETANONOate caused
and either CDDP or TRAIL. The DETANONOate-mediated cell sen- a rapid S-nitrosylation of the Snail protein which resulted in inhi-
sitization to TRAIL and CDDP was shown to be synergistic as bition of its activity as assessed by DNA-binding assays [unpub-
determined by isobologran analysis [10] (unpublished data) In lished data]. The direct role of NO-mediated inhibition of Snail in
addition, the reversal of tumor resistance to TRAIL was shown tumor cell sensitization was demonstrated by Snail silencing using
to involve both the type I and type II apoptotic pathways specific siRNA assays. Our findings demonstrate that, in contrast to
[10,11]. We further analyzed the underlying mechanism of untreated or cells treated with control siRNA, treatment with Snail
DETANONOate-mediated cell sensitization focusing specifically siRNA mimicked DETANONOate in terms of inducing significant
on the role of each of the gene products of the NF-jB/Snail/ potentiation of apoptosis following treatment with either TRAIL
YY1/RKIP loop, as described below. or CDDP [9] (Fig. 1B).
B. Bonavida, S. Baritaki / Nitric Oxide 24 (2011) 1–7 5

Role of YY1 data]. To examine further the underlying mechanism of DETANON-


In addition to Snail inhibition, cell treatment with DETANONO- Oate-mediated EMT inhibition we investigated selectively the con-
ate also inhibited the expression levels of another NF-jB targeted tribution of each of the members of the NF-jB/Snail/YY1/RKIP
gene product, YY1 [11,13]. In addition, DETANONOate was shown loop.
to abolish YY1 DNA-binding activity through direct S-nitrosylation
[11,13,57]. Since YY1 is a transcriptional activator of Snail, the inhi- Role of NF-jB
bition of YY1 activity might, therefore, contribute to further sup- The constitutive activation of NF-jB observed in various malig-
pression of Snail expression. Since YY1 is a negative regulator of nant tumors is known to promote angiogenesis and invasion, thus,
the transcription of the TRAIL receptor DR5, we examined whether increasing the metastatic cell potential [17]. In addition, NF-jB is
the DETANONOate-mediated YY1 inhibition results in upregula- critical for the transcriptional activation of the EMT-inducer Snail
tion of DR5 expression, thus contributing to cell sensitization to in metastatic tumors. Our studies with metastatic cell line models
TRAIL apoptosis. Indeed, we have confirmed that cell treatment were also characterized by their constitutively activated NF-jB
with DETANONOate led to selective upregulation of DR5 with no [63], which was significantly deactivated following cell treatment
effect on the TRAIL receptor DR4 or the decoy receptors DcR1 with DETANONOate, as mentioned above. The direct role of
and DcR2 [11] (unpublished data). The direct role of YY1-induced DETANONOate-mediated inhibition of NF-jB in the regulation of
inhibition by DETANONOate was further demonstrated following EMT was corroborated by the NF-jB inhibitor, DHMEQ. Cell treat-
treatment of tumor cells with YY1 siRNA. Such treated cells, in con- ment with DHMEQ mimicked DETANONOate in the induction of
trast to untreated or treated cells with control siRNA, resulted in E-cadherin and inhibition of both vimentin and fibronectin, thus
significant induction of apoptosis following cell treatment with supporting the critical role of NF-jB in the regulation of EMT
either TRAIL or CDDP [11,32,33] (unpublished data) (Fig. 1C). [unpublished data].

The role of RKIP Role of Snail


In contrast to DETANONOated-mediated inhibition of NF-jB, The direct role of DETANONOate-mediated inhibition of Snail
Snail and YY1, cell treatment with DETANONOate had rather an expression and activity in the inhibition of EMT was examined in
inducing effect on RKIP expression, that was attributed to DETAN- cells treated with Snail siRNA. In contrast to untreated or cells
ONOate-mediated inhibition of its direct and indirect suppressors treated with control siRNA, transfection with Snail siRNA re-
NF-jB, Snail and YY1 (10, 11, submitted data). The direct role of sulted in the reversal of EMT to MET (mesenchymal to epithelial
DETANONOate-mediated RKIP induction in the regulation of tumor transition), accompanied by induction of the metastasis supres-
cell resistance to both CDDP and TRAIL was demonstrated by treat- sors E-cadherin and RKIP and inhibition of both vimentin and
ment of tumor cells with an RKIP-expression vector. In comparison fibronectin [31]. In addition, overexpression of Snail in the
with untreated or cells treated with an empty vector, cell over- non-metastatic prostate carcinoma cell line, LnCaP, resulted in
expressing RKIP were significantly sensitized tumor cells to CDDP the induction of EMT [31]. These studies demonstrate the direct
and TRAIL-apoptosis, similar to the one observed after cell treat- role of Snail inhibition by DETANONOate and the resulting inhi-
ment with DETANONOate. In addition, overexpression of RKIP re- bition of EMT.
sulted in inhibition of YY1 and Snail expression which might be
possibly attributed to the suppression of NF-jB activity by RKIP
Role of YY1
[9] (Fig. 1D).
The role of YY1 in metastasis has not been clearly elucidated in
the literature. However, given the positive transcriptional regula-
Conclusions tion of the EMT-inducer Snail by YY1 [36], we examined whether
DETANONOate-mediated inhibition of YY1 expression and activity
The above findings demonstrate that treatment of resistant has a direct role in the inhibition of EMT. Our preliminary findings
tumor cells, which exhibit a dysregulated NF-jB/Snail/YY1/RKIP show that YY1 silencing by siRNA mimics DETANONOate in terms
loop, with DETANONOate resulted in the modification of this of inhibiting the mesenchymal cell phenotype through downregu-
loop, and that each gene product was shown to directly regulate lation of the expressions of Snail, vimentin and fibronectin and
tumor cell resistance, as would have been expected from a loop. induction of the metastasis suppressors RKIP and E-cadherin. The
Our findings with normal human derived peripheral blood leuko- direct transcriptional regulation of RKIP by YY1 has been further
cytes and treated with DETANONOate, alone or in combination studied and our preliminary evidence supports a direct suppressive
with chemotherapy, did not result in any significant toxicity effect of YY1 on the RKIP promoter activity.
however additional studies should be performed using normal
bone marrow derived cells and hematopoietic cells for analysis Role of RKIP
of toxicity. In addition, the findings demonstrate potential novel The expression of the metastasis-suppressor RKIP has been
therapeutic targets whose modifications can reverse tumor cell reported to be diminished up to barely detected in several
resistance. metastatic tumor tissues compared to their normal counterparts.
Accordingly the basal levels of RKIP expression in our studied
NO-mediated inhibition of EMT in metastatic cell lines metastatic cell line models were low and could be induced after
cell treatment with DETANONOate. The direct role of RKIP induc-
Besides the sensitizing effects of DETANONOate for apoptosis in tion in the regulation of EMT by DETANONOate was demon-
resistant tumor cells, our studies also identified DETANONOate as a strated by RKIP overexpression assays. In contrast to untreated
potent inhibitor of cell invasion and migration, thus resulting in or cells treated with an empty vector, treatment with the RKIP-
reversal of the EMT cell phenotype. Specifically, treatment of the expression vector resulted in downregulation of Snail, vimentin
metastatic tumor cells with DETANONOate inhibited the expres- and fibronectin and induction of E-cadherin [31]. The above find-
sion of mesenchymal gene products such as fibronectin and vimen- ings suggest that one mechanism by which RKIP mediates its
tin while it upregulated the levels of the metastasis-suppressor anti-metastatic effects is through inhibition of EMT and its induc-
E-cadherin, as assessed by western blot analysis. The above find- tion by DETANONOate has a pivotal role in the NO-mediated
ings were also confirmed by immunohistochemistry [submitted supression of EMT.
6 B. Bonavida, S. Baritaki / Nitric Oxide 24 (2011) 1–7

Conclusions Huerta-Yepez, Mario Vega, Fumiya Hongo, Kam Yeung, and Michael
Palladino. We also acknowledge the Jonsson Comprehensive Cancer
The above findings demonstrate the direct role of the NF-jB/ Center for their valuable assistance. The assistance of Daphne Liang
Snail/YY1/RKIP circuitry in the regulation of EMT. Cell treatment in the preparation of this manuscript is also acknowledged.
with DETANONOate modifies the loop and results in the inhibition
of EMT. Each of the gene products in the loop was shown to be di-
rectly involved in the regulation of EMT. The findings also demon- References
strate novel therapeutic targets in the inhibition of EMT and
metastasis. [1] A. Singh, J. Settleman, EMT, cancer stem cells and drug resistance. an emerging
axis of evil in the war on cancer, Oncogene 29 (2010) 4741–4751.
[2] D.S. Tan, M. Gerlinger, B.T. Teh, C. Swanton, Anti-cancer drug resistance.
In vivo validation of in vitro findings obtained with Understanding the mechanisms through the use of integrative genomics and
functional RNA interference, Eur. J. Cancer 46 (2010) 2143–2144.
DETANONOate [3] J.C. Reed, Dysregulation of apoptosis in cancer, J. Clin. Oncol. 17 (1999) 2941–
2953.
The above findings demonstrate that treatment of resistant and [4] S. Raguz, E. Yague, Resistance to chemotherapy: new treatments and novel
insights into an old problem, Br. J. Cancer 99 (2008) 387–391.
metastatic cell lines with DETANONOate resulted in tumor cell [5] M.A. Shah, G.K. Schwartz, Cell cycle-mediated drug resistance. an emerging
sensitization to apoptosis by both chemotherapeutic drugs and concept in cancer therapy, Clin. Cancer Res. 7 (2001) 2168–2181.
immunotherapy. In addition, the above data also show that treat- [6] K. DeClerck, R.C. Elble, The role of hypoxia and acidosis in promoting
metastasis and resistance to chemotherapy, Front Biosci. 15 (2010) 213–225.
ment with DETANONOate inhibited the EMT phenotype. We have [7] H. Acloque, J.P. Thiery, M.A. Nieto, The physiology and pathology of the EMT
analyzed whether our in vitro findings may be validated in vivo Meeting on the epithelial–mesenchymal transition, EMBO Rep. 9 (2008) 322–
in pre-clinical models using mice bearing tumor xenografts. In- 326.
[8] J.P. Thiery, J.P. Sleeman, Complex networks orchestrate epithelial–
deed, ex vivo tumor tissues derived from mice bearing the human mesenchymal transitions, Nat. Rev. Mol. Cell Biol. 7 (2006) 131–142.
metastatic prostate cancer cell line, PC-3, and treated with DETAN- [9] S. Baritaki, K. Yeung, M. Palladino, J. Berenson, B. Bonavida, Pivotal roles of snail
ONOate, had decreased expression of YY1 expression, upregulation inhibition and RKIP induction by the proteasome inhibitor NPI-0052 in tumor
cell chemoimmunosensitization, Cancer Res. 69 (2009) 8376–8385.
of DR5, and reversal of the EMT phenotype [11] (unpublished data).
[10] S. Huerta-Yepez, M. Vega, A. Jazirehi, H. Garban, F. Hongo, G. Cheng, B.
Similarly, mice bearing tumor xenografts and treated with the Bonavida, Nitric oxide sensitizes prostate carcinoma cell lines to TRAIL-
combination of DETANONOate and CDDP showed significant tumor mediated apoptosis via inactivation of NF-kappa B and inhibition of Bcl-xl
regression in contrast to those treated with either DETANONOate expression, Oncogene 23 (2004) 4993–5003.
[11] S. Huerta-Yepez, M. Vega, S.E. Escoto-Chavez, B. Murdock, T. Sakai, S. Baritaki,
or CDDP alone [11] (unpublished data). These preliminary findings B. Bonavida, Nitric oxide sensitizes tumor cells to TRAIL-induced apoptosis via
demonstrate that the findings obtained in vitro are validated in vivo inhibition of the DR5 transcription repressor Yin Yang 1, Nitric Oxide 20 (2009)
in mice. In addition, clinical data in human cancer patients have 39–52.
[12] A.R. Jazirehi, M.I. Vega, D. Chatterjee, L. Goodglick, B. Bonavida, Inhibition of
been reported on either the systemic administration of nitroglyc- the Raf-MEK1/2-ERK1/2 signaling pathway Bcl-xL down-regulation, and
erin in patients with lung cancer [64] and in patches in patients chemosensitization of non-Hodgkin’s lymphoma B cells by Rituximab,
with prostate cancer [65] and both studies showed significant clin- Cancer Res. 64 (2004) 7117–7126.
[13] M.I. Vega, A.R. Jazirehi, S. Huerta-Yepez, B. Bonavida, Rituximab-induced
ical efficacy. inhibition of YY1 and Bcl-xL expression in Ramos non-Hodgkin’s lymphoma
cell line via inhibition of NF-kappa B activity: role of YY1 and Bcl-xL in Fas
resistance and chemoresistance, respectively, J. Immunol. 175 (2005) 2174–
Overall conclusions and future directions 2183.
[14] Y. Lin, L. Bai, W. Chen, S. Xu, The NF-kappaB activation pathways, emerging
We have demonstrated that treatment of tumor cells with molecular targets for cancer prevention and therapy, Expert Opin. Ther.
Targets 14 (2010) 45–55.
DETANONOate simultaneously sensitizes tumor cells to apoptotic [15] H. Wang, E. Hertlein, N. Bakkar, H. Sun, S. Acharyya, J. Wang, M. Carathers, R.
stimuli induced by both chemotherapy and immunotherapy and Davuluri, D.C. Guttridge, NF-kappaB regulation of YY1 inhibits skeletal
also inhibits the induction of the EMT phenotype. The findings of myogenesis through transcriptional silencing of myofibrillar genes, Mol. Cell.
Biol. 27 (2007) 4374–4387.
the role of DETANONOate in the regulation of EMT were also [16] L. Kleinberg, B. Davidson, Cell survival and apoptosis-related molecules in
shown to be manifested in other tumors such as Non-Hodgkin’s cancer cells in effusions: a comprehensive review, Diagn. Cytopathol. 37
B-cell lymphoma cell lines. Analysis of the underlying mechanisms (2009) 613–624.
[17] C. Min, S.F. Eddy, D.H. Sherr, G.E. Sonenshein, NF-kappaB and epithelial to
by which DETANONOate mediates its activities was shown to be
mesenchymal transition of cancer, J. Cell. Biochem. 104 (2008) 733–744.
due, in part, to modification of the dysregulated NF-jB/Snail/ [18] M.A. Huber, N. Azoitei, B. Baumann, S. Grunert, A. Sommer, H. Pehamberger, N.
YY1/RKIP circuitry. We have also demonstrated that each gene Kraut, H. Beug, T. Wirth, NF-kappaB is essential for epithelial–mesenchymal
transition and metastasis in a model of breast cancer progression, J. Clin.
product in this circuitry is directly involved in the regulation in
Invest. 114 (2004) 569–581.
both the resistance and the EMT phenotype (Fig. 2). [19] S.R. Shin, N. Sanchez-Velar, D.H. Sherr, G.E. Sonenshein, 7, 12-
The implications of our present findings are the demonstration dimethylbenz(a)anthracene treatment of a c-rel mouse mammary tumor cell
of the potential therapeutic application of NO donors in the treat- line induces epithelial to mesenchymal transition via activation of nuclear
factor-kappaB, Cancer Res. 66 (2006) 2570–2575.
ment of patients with refractory cancer and metastasis. In addition, [20] S. Julien, I. Puig, E. Caretti, J. Bonaventure, L. Nelles, F. van Roy, C. Dargemont,
the dysregulated gene products in the circuitry are potential diag- A.G. de Herreros, A. Bellacosa, L. Larue, Activation of NF-kappaB by Akt
nostic biomarkers as well as targets for the generation of new ther- upregulates Snail expression and induces epithelium mesenchyme transition,
Oncogene 26 (2007) 7445–7456.
apeutic agents. [21] S. Beach, H. Tang, S. Park, A.S. Dhillon, E.T. Keller, W. Kolch, K.C. Yeung, Snail is
Future directions include additional pre-clinical studies in ani- a repressor of RKIP transcription in metastatic prostate cancer cells, Oncogene
mal models for validation of our in vitro findings as well as analysis 27 (2008) 2243–2248.
[22] A. Cano, M.A. Perez-Moreno, I. Rodrigo, A. Locascio, M.J. Blanco, M.G. Del
of different NO donors for their therapeutic effects. In addition, the Barrio, F. Portillo, M.A. Nieto, The transcription factor snail controls epithelial–
establishment of the prognostic significance of the circuitry using mesenchymal transitions by repressing E-cadherin expression, Nat. Cell Biol. 2
patient-derived tumor tissues could also be further exploited. (2000) 76–83.
[23] A. Katsman, K. Umezawa, B. Bonavida, Reversal of resistance to cytotoxic
cancer therapies: DHMEQ as a chemo-sensitizing and immuno-sensitizing
Acknowledgments agent, Drug Resist. Updat. 10 (2007) 1–12.
[24] M. Brown, J. Cohen, P. Arun, Z. Chen, C. Van Waes, NF-kappaB in carcinoma
therapy and prevention, Expert. Opin. Ther. Targets 12 (2008) 1109–1122.
The authors acknowledge the various investigators who partici- [25] D.J. McConkey, K. Zhu, Mechanisms of proteasome inhibitor action and
pated in the described studies in this review, namely, Doctors Sara resistance in cancer, Drug Resist. Updat. 11 (2008) 164–179.
B. Bonavida, S. Baritaki / Nitric Oxide 24 (2011) 1–7 7

[26] V. Baud, M. Karin, Is NF-kappaB a good target for cancer therapy? Hopes and [46] K.C. Yeung, D.W. Rose, A.S. Dhillon, D. Yaros, M. Gustafsson, D. Chatterjee, B.
pitfalls, Nat. Rev. Drug Discovery 8 (2009) 33–40. McFerran, J. Wyche, W. Kolch, J.M. Sedivy, Raf kinase inhibitor protein
[27] F. Li, G. Sethi, Targeting transcription factor NF-kappaB to overcome interacts with NF-kappaB-inducing kinase and TAK1 and inhibits NF-kappaB
chemoresistance and radioresistance in cancer therapy, Biochim. Biophys. activation, Mol. Cell. Biol. 21 (2001) 7207–7217.
Acta 1805 (2010) 167–180. [47] H. Tang, S. Park, S.C. Sun, R. Trumbly, G. Ren, E. Tsung, K.C. Yeung, RKIP inhibits
[28] C. Mauro, F. Zazzeroni, S. Papa, C. Bubici, G. Franzoso, The NF-kappaB NF-kappaB in cancer cells by regulating upstream signaling components of the
transcription factor pathway as a therapeutic target in cancer: methods for IkappaB kinase complex, FEBS Lett. 584 (2010) 662–668.
detection of NF-kappaB activity, Methods Mol. Biol. 512 (2009) 169–207. [48] K. Lorenz, M.J. Lohse, U. Quitterer, Protein kinase C switches the Raf kinase
[29] L. Zhang, W. Chen, X. Li, A novel anticancer effect of butein: inhibition of inhibitor from Raf-1 to GRK-2, Nature 426 (2003) 574–579.
invasion through the ERK1/2 and NF-kappa B signaling pathways in bladder [49] D. Chatterjee, Y. Bai, Z. Wang, S. Beach, S. Mott, R. Roy, C. Braastad, Y. Sun, A.
cancer cells, FEBS Lett. 582 (2008) 1821–1828. Mukhopadhyay, B.B. Aggarwal, J. Darnowski, P. Pantazis, J. Wyche, Z. Fu, Y.
[30] K. Tozawa, S. Sakurada, K. Kohri, T. Okamoto, Effects of anti-nuclear factor Kitagwa, E.T. Keller, J.M. Sedivy, K.C. Yeung, RKIP sensitizes prostate and breast
kappa B reagents in blocking adhesion of human cancer cells to vascular cancer cells to drug-induced apoptosis, J. Biol. Chem. 279 (2004) 17515–
endothelial cells, Cancer Res. 55 (1995) 4162–4167. 17523.
[31] S. Baritaki, A. Chapman, K. Yeung, D.A. Spandidos, M. Palladino, B. Bonavida, [50] Z. Fu, Y. Kitagawa, R. Shen, R. Shah, R. Mehra, D. Rhodes, P.J. Keller, A.
Inhibition of epithelial to mesenchymal transition in metastatic prostate Mizokami, R. Dunn, A.M. Chinnaiyan, Z. Yao, E.T. Keller, Metastasis suppressor
cancer cells by the novel proteasome inhibitor, NPI-0052: pivotal roles of Snail gene Raf kinase inhibitor protein (RKIP) is a novel prognostic marker in
repression and RKIP induction, Oncogene 28 (2009) 3573–3585. prostate cancer, Prostate 66 (2006) 248–256.
[32] S. Baritaki, E. Suzuki, K. Umezawa, D.A. Spandidos, J. Berenson, T.R. Daniels, [51] S. Hagan, F. Al-Mulla, E. Mallon, K. Oien, R. Ferrier, B. Gusterson, J.J. Garcia, W.
M.L. Penichet, A.R. Jazirehi, M. Palladino, B. Bonavida, Inhibition of Yin Yang 1- Kolch, Reduction of Raf-1 kinase inhibitor protein expression correlates with
dependent repressor activity of DR5 transcription and expression by the novel breast cancer metastasis, Clin. Cancer Res. 11 (2005) 7392–7397.
proteasome inhibitor NPI-0052 contributes to its TRAIL-enhanced apoptosis in [52] F. Al-Mulla, S. Hagan, A.I. Behbehani, M.S. Bitar, S.S. George, J.J. Going, J.J.
cancer cells, J. Immunol. 180 (2008) 6199–6210. Garcia, L. Scott, N. Fyfe, G.I. Murray, W. Kolch, Raf kinase inhibitor protein
[33] S. Baritaki, A. Katsman, D. Chatterjee, K.C. Yeung, D.A. Spandidos, B. Bonavida, expression in a survival analysis of colorectal cancer patients, J. Clin. Oncol. 24
Regulation of tumor cell sensitivity to TRAIL-induced apoptosis by the (2006) 5672–5679.
metastatic suppressor Raf kinase inhibitor protein via Yin Yang 1 inhibition [53] M.M. Schuierer, F. Bataille, S. Hagan, W. Kolch, A.K. Bosserhoff, Reduction in
and death receptor 5 up-regulation, J. Immunol. 179 (2007) 5441–5453. Raf kinase inhibitor protein expression is associated with increased Ras-
[34] Y. Wu, B.P. Zhou, Snail: More than EMT, Cell Adh. Migr. 4 (2010) 199–203. extracellular signal-regulated kinase signaling in melanoma cell lines, Cancer
[35] M.A. Nieto, The snail superfamily of zinc-finger transcription factors, Nat. Rev. Res. 64 (2004) 5186–5192.
Mol. Cell Biol. 3 (2002) 155–166. [54] Z. Fu, P.C. Smith, L. Zhang, M.A. Rubin, R.L. Dunn, Z. Yao, E.T. Keller, Effects of
[36] M.B. Palmer, P. Majumder, J.C. Cooper, H. Yoon, P.A. Wade, J.M. Boss, Yin yang 1 raf kinase inhibitor protein expression on suppression of prostate cancer
regulates the expression of snail through a distal enhancer, Mol. Cancer Res. 7 metastasis, J. Natl Cancer Inst. 95 (2003) 878–889.
(2009) 221–229. [55] P. Minoo, I. Zlobec, K. Baker, L. Tornillo, L. Terracciano, J.R. Jass, A. Lugli,
[37] D. Olmeda, G. Moreno-Bueno, J.M. Flores, A. Fabra, F. Portillo, A. Cano, SNAI1 is Loss of raf-1 kinase inhibitor protein expression is associated with tumor
required for tumor growth and lymph node metastasis of human breast progression and metastasis in colorectal cancer, Am. J. Clin. Pathol. 127
carcinoma MDA-MB-231 cells, Cancer Res. 67 (2007) 11721–11731. (2007) 820–827.
[38] S. Gordon, G. Akopyan, H. Garban, B. Bonavida, Transcription factor YY1: [56] D. Wink, L. Ridnour, S. Hussain, C. Harris, The reemergence of nitric oxide and
structure, function, and therapeutic implications in cancer biology, Oncogene cancer, Nitric Oxide 19 (2008) 65–67.
25 (2006) 1125–1142. [57] F. Hongo, H. Garban, S. Huerta-Yepez, M. Vega, A.R. Jazirehi, Y. Mizutani, T.
[39] J. Guo, X. Lin, M.A. Williams, Q. Hamid, S.N. Georas, Yin-Yang 1 regulates Miki, B. Bonavida, Inhibition of the transcription factor Yin Yang 1 activity by
effector cytokine gene expression and T(H)2 immune responses, J. Allergy Clin. S-nitrosation, Biochem. Biophys. Res. Commun. 336 (2005) 692–701.
Immunol. 122 (2008) 195–201. [58] D.J. Smith, D. Chakravarthy, S. Pulfer, M.L. Simmons, J.A. Hrabie, M.L. Citro, J.E.
[40] J. Ye, H.A. Young, X. Zhang, V. Castranova, V. Vallyathan, X. Shi, Regulation of a Saavedra, K.M. Davies, T.C. Hutsell, D.L. Mooradian, S.R. Hanson, L.K. Keefer,
cell type-specific silencer in the human interleukin-3 gene promoter by the Nitric oxide-releasing polymers containing the [N(O)NO]-group, J. Med. Chem.
transcription factor YY1 and an AP2 sequence-recognizing factor, J. Biol. Chem. 39 (1996) 1148–1156.
274 (1999) 26661–26667. [59] L.K. Keefer, R.W. Nims, K.M. Davies, D.A. Wink, NONOates (1-substituted
[41] J. Ye, X. Zhang, Z. Dong, Characterization of the human granulocyte– diazen-1-ium-1, 2-diolates) as nitric oxide donors: convenient nitric oxide
macrophage colony-stimulating factor gene promoter: an AP1 complex and dosage forms, Methods Enzymol. 268 (1996) 281–293.
an Sp1-related complex transactivate the promoter activity that is suppressed [60] M. Kavdia, R.S. Lewis, Nitric oxide delivery in stagnant systems via nitric oxide
by a YY1 complex, Mol. Cell. Biol. 16 (1996) 157–167. donors: a mathematical model, Chem. Res. Toxicol. 16 (2003) 7–14.
[42] H.J. Garban, B. Bonavida, Nitric oxide inhibits the transcription repressor Yin- [61] J.G. Diodati, A.A. Quyyumi, N. Hussain, L.K. Keefer, Complexes of nitric oxide
Yang 1 binding activity at the silencer region of the Fas promoter: a pivotal with nucleophiles as agents for the controlled biological release of nitric oxide:
role for nitric oxide in the up-regulation of Fas gene expression in human antiplatelet effect, Thromb. Haemost. 70 (1993) 654–658.
tumor cells, J. Immunol. 167 (2001) 75–81. [62] V.G. Nielsen, Nitric oxide decreases coagulation protein function in rabbits as
[43] S. Baritaki, S. Huerta-Yepez, T. Sakai, D.A. Spandidos, B. Bonavida, assessed by thromboelastography, Anesth. Analg. 92 (2001) 320–323.
Chemotherapeutic drugs sensitize cancer cells to TRAIL-mediated apoptosis: [63] J. Suh, A.B. Rabson, NF-kappaB activation in human prostate cancer: important
up-regulation of DR5 and inhibition of Yin Yang 1, Mol. Cancer Ther. 6 (2007) mediator or epiphenomenon?, J Cell. Biochem. 91 (2004) 100–117.
1387–1399. [64] H. Yasuda, Solid tumor physiology and hypoxia-induced chemo/radio-
[44] A.E. Granovsky, M.R. Rosner, Raf kinase inhibitory protein: a signal transduction resistance. Novel strategy for cancer therapy: nitric oxide donor as a
modulator and metastasis suppressor, Cell Res. 18 (2008) 452–457. therapeutic enhancer, Nitric Oxide 19 (2008) 205–216.
[45] K. Yeung, T. Seitz, S. Li, P. Janosch, B. McFerran, C. Kaiser, F. Fee, K.D. Katsanakis, [65] D.R. Siemens, Phase II study of nitric oxide donor for men with increasing
D.W. Rose, H. Mischak, J.M. Sedivy, W. Kolch, Suppression of Raf-1 kinase prostate-specific antigen level after surgery or radiotherapy for prostate
activity and MAP kinase signalling by RKIP, Nature 401 (1999) 173–177. cancer, Urology 74 (2009) 878–883.

You might also like