ArticlePDF Available

Independent role of phosphoinositol-3-kinase (PI3K) and Casein Kinase II (CK-2) in EGFR and Her-2-mediated constitutive NF-kappaB activation in prostate cancer cells

Authors:
  • Institut de Cancérologie de Libreville

Abstract and Figures

Recent research has highlighted the potential role of EGFR and Her-2 in the constitutive activation of NF-kappaB (NF-kappaB) in prostate cancer cells, although the mechanism by which these receptors activate NF-kappaB in these cells remains unclear. Using pharmacological and genetic approaches we show that in PC-3 cells, EGFR and Her-2 are involved in the constitutive activation of NF-kappaB through two different mechanisms. EGFR activates NF-kappaB through the PI3K/Akt pathway that leads to the phosphorylation of IkappaBalpha on serines 32 and 36, thereby promoting the nuclear translocation of the p65 subunit. In contrast, Her-2 activates NF-kappaB through Casein Kinase II (CK-2) activation independently of IkappaBalpha phosphorylation on serines 32 and 36. Our study not only directly clarifies the signaling pathways involved in NF-kappaB activation in prostate cancer cell lines and but also provides a framework for further studies in the clinical characterization and management of prostate cancer.
Content may be subject to copyright.
The Prostate 65:306 ^315 (2005)
Independent Role of PhosphoInositol-3-Kinase (PI3K)
and Casein Kinase II (CK-2) in EGFR and
Her-2-Mediated Constitutive NF-kappaB
Activation in Prostate Cancer Cells
Ce
´cile Le Page,
1
Ismael Herve
´Koumakpayi,
1
Laurent Lessard,
1
Fred Saad,
1,2
and Anne-Marie Mes-Masson
1,3
*
1
Centre de Recherche du Centre Hospitalier del’Universite
¤de Montre
¤al (CR- CHUM) and Institut du cancer de Montre
¤al,
1560 rue Sherbrooke est, Montre
¤al, Que
¤bec,Canada
2
De
¤par teme nt d’urologie, Universite
¤de Montre
¤al, Montre
¤al, Que
¤bec,Canada
3
De
¤partement de Me
¤decine,Universite
¤de Montre
¤al, Montre
¤al, Que
¤bec,Canada
BACKGROUND. Recent research has highlighted the potential role of EGFR and Her-2 in the
constitutive activation of NF-kappaB (NF-kB) in prostate cancer cells, although the mechanism
by which these receptors activate NF-kB in these cells remains unclear.
METHODS AND RESULTS. Using pharmacological and genetic approaches we show that in
PC-3 cells, EGFR and Her-2 are involved in the constitutive activation of NF-kB through two
different mechanisms. EGFR activates NF-kB through the PI3K/Akt pathway that leads to the
phosphorylation of IkBaon serines 32 and 36, thereby promoting the nuclear translocation of the
p65 subunit. In contrast, Her-2 activates NF-kB through Casein Kinase II (CK-2) activation
independently of IkBaphosphorylation on serines 32 and 36.
CONCLUSIONS. Our study not only directly clarifies the signaling pathways involved in NF-
kB activation in prostate cancer cell lines and but also provides a framework for further studies
in the clinical characterization and management of prostate cancer. Prostate 65: 306 –315, 2005.
#2005 Wiley-Liss, Inc.
KEY WORDS: ErbB-1; ErbB-2/c-neu; signaling; PC-3; LNCaP
INT RODUCTION
In men, prostate cancer is the most frequently
diagnosed cancer and a leading cause of cancer death.
Treatment of prostate cancer depends on its stage. If the
tumor is localized and the patient is healthy, aggressive
therapy is often recommended. However, some early
stage tumors will remain latent and will not require
aggressive therapy, while others at risk of progression
need to be treated early. Unfortunately, there are no
known molecular markers that reliably predict if a
cancer will progress to an aggressive disease. It is
unclear which genes are implicated in the initiation and
progression of prostate cancer and to date attention has
focused on several genes and pathways involved in this
mechanism although few of these players are fre-
quently altered in primary prostate cancer. Recently, an
alternate candidate marker, Nuclear Factor-kappaB
(NF-kB) has been shown to play a role in the devel-
opment of prostate cancer and the progression to an
advanced disease [13].
Grant sponsor: Canadian Prostate Cancer Research Foundation;
Grant sponsor: The Canadian Uro-Oncology/Astra Zeneca Award;
Grant sponsor: Chercheur National fellowship from the Fonds de la
Recherche en Sante
´du Que
´bec (to A.-M.M.-M.); Grant sponsor:
Canadian Institute of Health Research/Canadian Prostate Cancer
Research Initiative studentship (to L.L.); Grant sponsor: The
Canderel fund of the Institut du Cancer de Montre
´al (I.H.K.).
*Correspondence to: Dr. Anne-Marie Mes-Masson, Centre de
recherche CHUM/Institut du cancer de Montre
´al, 1560 Sherbrooke
Est, Montre
´al, Que
´bec, Canada, H2L 4M1.
E-mail: anne-marie.mes-masson@umontreal.ca
Received 9 February 2005; Accepted 7 April 2005
DOI 10.1002/pros.20291
Published online 13 July 2005 in Wiley InterScience
(www.interscience.wiley.com).
2005Wiley-Liss,Inc.
The Rel/NF-kB transcription factor family is com-
posed of structurally related members that possess a
N-terminal Rel-Homology (RH) domain involved in
proteinprotein interactions and DNA-binding. NF-kB
proteins exist as homo- and heterodimers expressed
in many cells but kept inactive and maintained in
the cytoplasm by IkB family of inhibitors. Classically,
the activation of NF-kB requires signals that converge
to the trimeric IkB kinase complex (IKK) resulting in
phosphorylation of IkBaon serine 32/36 and its subse-
quent ubiquitinylation and degradation allowing NF-
kB to translocate to the nucleus. The precise mechanism
by which NF-kB is constitutively activated in hormone-
independent prostate cancer cells is not well known.
Constitutive activation of PI3K/Akt pathway and IKK
has been implicated in this activation in the PC-3
hormone-independent prostate cancer cell line [1,4 –6].
PI3K activation results in the translocation of Akt from
the cytoplasm to the inner membrane where Akt is
phosphorylated on serine 473 and threonine 308 by
upstream kinases, PDK and ILK [7]. The activation of
Akt leads to the phosphorylation of downstream target
such as IKK, GSK3, Bad, caspase 9, and FOXO family of
forkhead transcription factors. However, it has yet to be
determined which initial event constitutively activate
PI3K/Akt pathway in prostate cancer cells.
In different epithelial cancer cell lines, evidence
points to a role of EGFR and Her-2 as upstream acti-
vators of the PI3K/Akt pathway and responsible for the
constitutive activation of NF-kB through the activation
of IKK [810]. EGFR and Her-2 are members of the
ErbB growth factor receptor family composed of four
distinct receptors: EGFR/ErbB1, Her-2/ErbB2/c-neu,
Her-3/ErbB3, and Her-4/ErbB4. In PC-3 cells, EGFR
and Her-2 overexpression and constitutive activation
modulate NF-kB signaling through two distinct mecha-
nisms: one dependent and one independent of the
phosphorylation of IkBaon serines 32/36 [11]. How-
ever the signaling intermediates that lead to this
constitutive NF-kB activation are presently not known.
Here, we investigated the role of PI3K/Akt pathway as
a downstream signaling component of EGFR and Her-2
in PC-3 cells. We observed that EGFR, but not Her-2,
activated NF-kB through the PI3K/Akt pathway,
which induces the phosphorylation of IkBaon serines
32/36 and thereby the nuclear translocation of NF-kB.
Then we further investigated the role of CK-2 in Her-2-
mediated signaling. CK-2 is an ubiquitous serine/
threonine kinase composed of two catalytic subunits
(aand a0) and one regulatory subunit (b). The regu-
lation and function of CK-2 is not well understood.
Originally CK-2 was defined as a constitutively active
protein kinase though recently several reports sug-
gest that CK-2 acts as a regulated kinase participa-
ting in signal transduction [12] and activation of NF-kB
[1316]. Here we demonstrate for the first time a role
for CK-2 in Her-2mediated NF-kB signaling.
MATERIALS AND METHODS
Cell Culture and Reagent
PC-3 and LNCaP cells were grown at 378C and in 5%
CO
2
in RPMI supplemented with 10% fetal bovine
serum (FBS) and 2 mg/ml gentamicin. Experiments
were performed on cells before passage 20. All reagents
used for cell culture media were from Gibco BRL. All
pharmacological inhibitors were from Calbiochem and
resuspended in dimethyl sulfoxide (DMSO).
Western Blot Analysis
Cells were lyzed with cold lysis buffer (10 mM Tris-
HCl, pH 7.4, 150 mM NaCl, 1 mM EDTA, 1 mM DTT/
1 mM NaF/0.5% NP-40/0.5 mM PMSF/0.2 mM
sodium orthovanadate/2 mg/ml of aprotinin, leupep-
tin, and pepstatin), boiled in loading buffer, separated
by SDSPAGE, and transferred on a nitrocellulose
membrane under refrigerated conditions (200 mA, 2 h).
Membranes were saturated with 5% milk/ PBS/0.1%
Tween 20. Immunodetection was done as described in
the protocol of the ECL kit (Amersham Biosciences UK
limited, England): i.e., incubated 2 hr at room tempera-
ture with the specific antibody (0.5 to 1 mg/ml), washed
two times with PBS/0.05% Tween 20 and incubated for
another 30 min at room temperature with peroxidase
conjugated antibodies (Santa-Cruz, CA). For Western-
blot polyclonal antibodies IkBa(B-8), c-myc (A-14), CK-
2a(C-18), CK-2a0(C-20), Akt-1 (D-17) from Santa Cruz
Biotechnology (CA), and phospho-IkBaand phospho-
Akt (Cell Signaling, MA), were used. Monoclonal
antibodies NF-kB p65 (F-6) and Ran (C-20) from Santa
Cruz, and beta-actin (ab6272, Abcam, MA) were used.
All experiments were repeated at least two times with
similar results.
Cytoplasmic and Nuclear Extracts
To prepare cellular extracts, 5 10
6
cells were wash-
ed twice in cold PBS and resuspended in 300 ml of lysis
buffer containing 10 mM Tris pH 7.9/10 mM NaCl/
5mMMgCl
2
/10 mM sodium orthovanadate/0.5 mM
PMSF/10 mg/ml of the protease inhibitors (PMSF,
pepstatin, leupeptin, and aprotinin). After swelling
cells for 30 min on ice, 0.1% Nonidet P-40, and 10%
glycerol (v/v) were added and lysed cells were centri-
fuged for 1 min at 48C and 5,000 rpm. Supernatants
were carefully decanted for cytoplasmic extracts.
Nuclei pellets were resuspended for 1 hr in 40 mlof
lysis buffer containing 10 mM Tris pH 7.9/400 mM
NaCl/0.1 EDTA/0.5 mM DTT/5% glycerol/0.5 mM
Mechanism of NF-kB Activation in Prostate Cancer Cells 307
PMSF/10 mg/ml protease inhibitors. Particulate matter
was eliminated by centrifugation for 10 min at
13,000 rpm at 48C. Protein concentration was deter-
mined using the Bradford method.
Electrophoretic Mobility Shift Assay (EMSA)
Nuclear cell extracts were prepared using 5
10
6
cells incubated with the indicated inhibitors or
with vehicle alone. Ten micrograms were subjected
to EMSA using a double-stranded (50-AGTTGAG-
GGGAGATTTGCAGGC-30)
32
P-labelled probe that
corresponds to the NF-kB-binding element from the
Igk-chain promoter. Binding reactions were carried out
on ice for 30 min in binding buffer (20 mM Tris pH 8.0,
5% glycerol, 0.1 KCl, 0.2 mM EDTA pH 8.0, 0.2 EGTA
pH 8.0, 0.5 mg poly(dIdC). The resulting protein DNA
complexes were separated on 5% non-denaturating
polyacrylamide gel in Tris-GlycineEDTA buffer. Gels
were dried and exposed for autoradiography. All ex-
periments were repeated two times with similar
results.
Transfection and Luciferase Reporter Assay
Cells were co-transfected in 96-well plates with
0.2 mg of DNA and 200 ng of a constitutively active
Renilla luciferase (pCMV-RL, Promega, WI) by lipo-
fectamine method (Invitrogen, CA). After 6 hr, cells
were washed in fresh medium and incubated over-
night. Cells were then stimulated for 16 hr with the
different inhibitors or DMSO. Cells were then assayed
for luciferase activity using the dual luciferase reporter
assay system (Promega, WI). The 3enh-kb-CONA-luc,
carrying a firefly luciferase gene under the control of a
trimer of kB consensus was a gift from Dr. Juana
Wietzerbin (Paris, France). The PCDNA3-HERCD533
plasmid was a kind gift from Dr. Alex Ullrich
(Germany) and Dr. Sylvain Meloche (Montreal, Qc,
Canada). PCDNA3.1-Myc-mut-Her-2 was a generous
gift from Dr. Ming-Fong Lin (Omaha, Nebraska, USA).
CK-2a0was provided by Dr. David Lichfield (Ontario,
Canada). All experiments were repeated three to four
times with similar results.
CK-2 Kina se Assays
The CK-2 kinase assay was performed following the
instruction of the manufacturer (Upstate Biotechnol-
ogy Inc.). Briefly, 5 10
6
cells were washed and resus-
pended in lysis buffer containing 10 mM HEPES
pH 7.5/250 mM NaCl/5 mM MgCl
2
/20 mM sodium
orthovanadate/0.5 mM PMSF/ b-glycerophosphate/
1 mM NaF/2 mg/ml of the protease inhibitors
(pepstatin, leupeptin, and aprotinin). Samples (5
10 mg) were resuspended in kinase buffer and the assay
was performed at 378C for 10 min, with 200 mMof
specific peptide in the presence of PKA inhibitors and
0.5 mCi [
32
P]g-ATP. The resulting products were anal-
yzed on P81 filters, washed, and counted in scintillation
liquid. Counts in assay containing sample without
peptide were subtracted as background to calculate the
specific activity in each sample. All experiments were
repeated two to three times with similar results.
RESULTS
EGFR but not Her-2 Activates the PI3K/Akt
Pathway in PC-3 and LNCaP Cells
The PI3K/Akt pathway can be activated by various
growth factors, including EGF through the activation of
ErbB receptors [17]. To investigate the role of EGFR
and Her-2 on PI3K/Akt pathway activation in prostate
cancer cells, we tested the effect of ErbB inhibitors on
the Akt phosphorylation in the PC-3 and LNCaP cell
lines. The efficiency of these inhibitors had already
been tested and described in our different prostate
cancer cell lines [11] and confirmed the results reported
by others [18,19]. Under appropriate conditions,
tyrphostins AG1517 and AG1478 selectively block the
phosphorylation of EGFR and do not affect the phos-
phorylation of Her-2 while tyrphostin AG879 selec-
tively blocks the phosphorylation of Her-2 but not
EGFR activation. In addition, we tested emodin, an
inhibitor of both Her-2 and CK-2 [20,21]. As shown in
Figure 1A, in PC-3 and LNCaP cells the inhibitors of
EGFR, AG1478 and AG1517, diminished the phospho-
rylation of Akt. The inhibition of PI3K by the AG1517
inhibitor was dose-dependent (Fig. 1B). The specificity
of the Akt phosphorylation observed was confirmed by
using LY294002, a specific inhibitor of PI3K, the kinase
responsible of Akt phosphorylation (Fig. 1A, second
lane). LY294002 completely abrogated the phospho-
rylation of Akt. In contrast, in PC-3 and LNCaP cells,
Her-2 inhibitors (AG879 and emodin) showed no effect
on constitutive Akt phosphorylation (Fig. 1A), even
with high doses of inhibitor (Fig. 1B), suggesting that
Her-2 is not involved in the constitutive activation of
PI3K in prostate cancer cell lines. Together, these
observations indicate that Akt activation is strongly
dependent of EGFR signaling in prostate cancer cells.
To confirm the role of EGFR in PI3K/Akt activation
of prostate cancer cells and to eliminate the role of
a potential side effect of AG1478 and AG1517, we also
tested by transient transfection the effect of HERCD533
and mut-mycHer-2, two dominant mutant forms of
EGFR and Her-2 respectively. These truncated recep-
tors lack the cytoplasmic domain containing the kinase
activity and reduce the tyrosine phosphorylation of
their respective endogenous receptor [18,22]. LNCaP
308 Page et al.
cells were chosen as a model in this assay as these cells,
in comparison to PC-3 cells, were readily transfectable
and provided the more appropriate model to monitor
transfected protein expression by western-blot analy-
sis. In addition, the transfection control for HERCD533
was assessed by its ability to inhibit the activation of
endogenous EGFR receptor as no antibody are com-
mercially available for the detection by Western blot-
ting of the EGFR N-terminal sites. As shown in
Figure 1C, cells transfected with HERCD533 showed
a decreased phosphorylation of EGFR and Akt com-
pared to cells transfected with the empty vector
PCDNA3.1 or PCDNA3.1-mut-mycHer-2 (Fig. 1D).
These results again support the notion that EGFR, but
not Her-2, activates NF-kB through the PI3K/Akt sig-
naling pathway in prostate cancer cells.
The PI3K/Akt Pathway is Involved in the
Phosphorylation of IjBaand the Nuclear
Translocation of p65 in PC-3 Cells
PI3K has been involved in the constitutive activation
of NF-kB in PC-3 cells however the precise role of PI3K
in these cells is controversial. In particular, while two
reports cite a reduced NF-kB DNA binding in presence
of PI3K inhibitors [5,23] it has also been reported that
PI3K does not affect the nuclear localization of NF-kB
but rather the transcriptional ability of p65 subunit [6].
We used LY294002, a specific inhibitor of PI3K, to dis-
criminate the role of PI3K in the constitutive activation
of NF-kB in our PC-3 cells. As shown in Figure 2A and
B, LY294002 strongly decreased the transactivational
ability as well as the DNA binding of NF-kB in these
cells. As expected, no effect was observed in LNCaP
cells that do not present constitutive activation of NF-
kB. Furthermore, in PC-3 LY294002 prevented the
phosphorylation of IkBaon serines 32/36 and the
nuclear translocation of the p65 subunit of NF-kB
(Fig. 2C) confirming the role of Akt in the constitutive
activation of NF-kB in PC-3 prostate cancer cell line.
Constitutive Activation of CK-2
in Prostate Cancer Cells
As Her-2 does not appear to signal through the
PI3K/Akt pathway that leads to the phosphorylation of
IkBaon serines 3236, we addressed which pathway
may be the downstream target of Her-2 and involved in
the constitutive activation of NF-kB in PC-3 cells. We
have previously shown that Her-2 activates NF-kB
through a mechanism that does not involve the phos-
phorylation of IkBaon serine 3236 [11]. Interestingly,
in other tumor cell types, CK-2 has also been involved
in the constitutive activation of NF-kB by a mechanism
independent of IkBaphosphorylation on serine 32 36
[1316]. CK-2 is constitutively activated in hormone-
insensitive PC-3 cells and to a lower extent in LNCaP
cells as measured by specific peptide phosphorylation
(Fig. 3A and [24,25]). This activity correlated with
the level of the catalytic subunits CK-2aand CK-2a0
(Fig. 3B) and was enhanced by the transient transfec-
tion of a wild type subunit CK-2a0construct (Fig. 3C)
while it was decreased by two CK-2 inhibitors, 4,5,6,7-
tetrabromobenzotriazole (TBB) and apigenin. We chose
these pharmacologic inhibitors since TBB is the most
Fig. 1. Activation of PI3K pathway by EGFR. A:InhibitionofPI3K
activation by EGFR inhibitors. PI3K activity was tested based on its
ability to phosphorylate Akt on serine 473. Thirty micrograms o f
total extracts from PC-3 and LNCaP cells treated for 6 hr with
DMSO (con trol) or tyrpho stin inhibitors. Cell extrac ts were loade d
on a 10% SDS PAGE and subjected to Western-blotting using anti-
phospho-serine473-Akt or anti-Akt antibodies. In all experiments
concentration of tyrphostininhibitors were 20 mMLY294002(LY),
40 mMemodin,10mMAG879,10mM AG1517, 250 nM AG1478, or
10 mMAG1296.B: Dosedependentinhibition of PI3Kby AG1517.Fifty
micrograms of totalextractsfromLNCaPcells treated for6 hr with
DMSO(control)orincreasingconcentrationof tyrphostin inhibitors
wereloadedona10%SDS ^PAGEandsubjectedtoWestern-blotting
with anti-phospho ser473-Akt, anti-Her-2 (control) or anti-Akt-1
(not shown) antibodies. Concentrations of tyrphostin inhibitors
were em odin 20, 4 0, 8 0 mM(lanes 2,6,and10 respectively), AG879
5,10, 20 mM(lane s 4,8,12 respectively),AG15172,10,20mM(lanes 3,7,
11 respectively).Inhibition of PI3K by dominant negative mutant of
EGFR but notby a dominant negative mutant of Her-2. LNCaPcells
were transfected with PCDNA3.1, HERCD533 a dominant negative
mut ant o f EGFR, (C) or a domina nt neg ative of Her- 2, Mut-Her- 2 (D).
After 4 8 hr, 50 mg of total extracts were subjectedto Western-blot-
tingusing an antibody recognizingthe transfectedconstructs (panel
C: anti-myc and panel D: anti-phosphoEGFR) or anti-phospho-
ser273Aktor anti-Akt1 antibodies.
Mechanism of NF-kB Activation in Prostate Cancer Cells 30 9
specific and selective CK-2 inhibitor [26,27] whereas,
though less specific, apigenin is more commonly used
as CK-2 inhibitor. Altogether these results show that
CK-2 is constitutively activated in PC-3 prostate cancer
cells line and to a lesser extent in LNCaP cells.
Role of CK-2 in the Constitutive
Activation of NF-jB
To ascertain a role of CK-2 in the constitutive
activation of NF-kB in PC-3 cells we used known in-
hibitors of CK-2, TBB, and apigenin, over a time period
that does not affect the phosphorylation status or the
expression of Her-2 (data not shown). Both inhibitors
significantly decreased the constitutive transactiva-
tional ability of NF-kB (Fig. 4A) in a dose dependent
manner (Fig. 4B) while the overexpression of CK-2a0
was correlated with this transcriptional activity. Inter-
estingly, the phosphorylation of IkB-aon serine 32/36
and the nuclear translocation of p65 was neither
affected by apigenin nor by TBB (Fig. 4C) supporting
the fact that Her-2 may activate the transactivational
ability of NF-kB through CK-2 activation in PC-3 cells.
Altogether, these results show that Her-2 activates NF-
kB, at least in part, through the activation of CK-2 in a
process independent of the phosphorylation of IkBaon
serines 32/36 and p65 nuclear translocation.
Her-2 Activates CK-2 in Prostate Cancer Cells
To determine whether EGFR and/or Her-2 are in-
volved in the constitutive activation of CK-2 in prostate
cancer cells, we tested the effect of tyrphostin inhibitors
on the CK-2 activity in PC-3 and LNCaP cells (Fig. 5A).
Fig. 2. Activation of NF-kB by PI3Kpathway. A:InhibitionofNF-
kB byinhibitors of PI3K.PC- 3 andLNCaPcells were co-transfected
with th e 3kB -c onA-luc a nd Renilla ve ctor s. Sixte en hour s followi ng
transfection, cells wereincubated for 24 hr with DMSOor 20 mMof
LY29004(LY).Cellswerethenassayedfor luciferase activities.Rela-
tive firefly luciferase activity (expressedin % control) is theratio of
luciferase activity in treated cells to that in control cells (DMSO).
Transfectionefficiency was normalizedto that of Renillaluciferase.
Values represent the mean SEM. *P<0.05 versus control (Stu-
dent’s t-test). B:InhibitionofNF-kB binding activity by PI3K inhibi-
tors. PC-3 and LNCaP cells were incubated for 6 hr with 20 mM
LY2940 02 (LY) and nuclear extracts were prepared and tested for
NF-kB DNAbinding activity a s describedin Material and Methods.
ns: not specific. C: Inhibition of p65 nuclear localization and IkBa
phosphorylation by PI3K inhibitor. Fifty micrograms of nuclear
extracts or cytoplasmic extracts from PC-3 cells treated for 6 hr
with DMSO (con trol) or 2 0 mM of LY294002 were loaded on a10%
SDS ^PAGE and subjected to Western-blotting using an anti-phos-
phoIkBa,anti-IkBa, anti-p65 or anti-Ran (a loading control for
nuclearextract) antibodies.
Fig. 3. A: Activity of CK-2 in prostate cancer cell lines. CK-2
activity was tested with10 mg of totalprotein and the activity was
determined as described in Materialand Methods. B:Westernblot
analysisofCK-2aandCK-2a.Celllysatesweresubjected toWestern
blotting and revealed using CK2 subunit specific antibodies.C:Inhi-
bition of CK-2 activity by apigenin and TBB.PC-3 cells were stimu-
lated for 6 hr with 50 mMapigeninor25mM of TBB or transfected
for 24 hr with empty ve ctor or CK-2awild type construction.CK-2
activity was tested with10 mg of totalprotein and the activity was
determinedas described (Materials and Methods).Values (panels A
and C) represent themean SEM. Significance, as c ompared to the
control, was defined as *P<0.05 using a S tuden t’s t-test.
310 Page et al.
In PC-3 cells, the Her-2 inhibitor AG879 decreased the
CK-2 activation by 75% while EGFR inhibitors AG1517
had a weaker inhibitory effect. As AG1478 had no
significant effect, this would argue against a role of
EGFR in constitutive activation of CK-2 in PC-3 cells.
Similarly, in LNCaP cells only AG879 showed a signi-
ficant effect on the constitutive activation of CK-2. This
inhibition was weaker than in PC-3 cells and may be
related the high expression of Her-2 in LNCaP cells
compared to PC-3 cells [11]. We verified that the inhi-
bitory effect of tyrphostin inhibitors on CK-2 activation
was not due to a downregulation of the CK-2 protein
expression (Fig. 5B), supporting the notion that Her-2 is
involved in the activation of CK-2. To confirm this
result we also tested the effect of mut-mycHer-2 ex-
pression by transient transfection assays. As shown in
Figure 5C, mut-mycHer-2 almost completely abroga-
ted CK-2 activity, again supporting the idea that Her-2
is involved in the constitutive activation of CK-2 in
prostate cancer cells.
Fig. 4. Activation of NF-kBbyCK-2kinase.A: PC-3 cells were
co-transfectedwithRenilla and3kB-conA-luc vectorsanda control
or a CK-2a0encoding vector. Sixteenhours after transfection, cells
were incubatedin media with either DMSO, an appropriate control,
50 mM apigenin (Api) or 25 mM TBB for 24 hr.Cells were then as sayed
for luciferase activity. Relative firefly luciferase ac tivity (expressed
in % control) is theratio of luciferase activity in treated cells to that
in control cells (DMSO or control vector). Values represent the
mean SEM. *P<0.05 versus control(Student’st-test).B:PC-3cells
were co-transfected with the Renilla and 3kB-conA-luc vectors.
Sixteenhours after transfection, cells were incubatedin media with
either DMSO, or indicated doses of apigenin (Api) orTBB for 24 hr.
Cellswere then assayed forluciferaseac tivity.Relativefireflylucifer-
ase activity(expressedin % control) is theratio of luciferase activity
in treated cells to that in control cells (DMSO or control vector).
Values represent the mean SEM. *P<0.05, **P<0.10 versus con-
trol (Stude nt’s t-test).C:50mgof nuclear extracts (NE) or cytoplas-
micextracts (CE)fromPC-3 cells treatedfor6 hr witheitherDMSO,
50 mM apigenin, or 25 mM TBB wereloaded ona 10% SDS ^PAGEand
subjec ted to Western blotting using either an anti-p65, anti-phos-
phoserines 32 and36 IkBaoranti-IkBaantibodies. Anti-Ranserved
as theloading controlfor nuclear extracts.
Fig. 5. Constitutive activation of CK-2 by Her-2. A:Inhibitionof
CK- 2 ac tivity by tyrphos tin.Pros tate ca ncer cells were treated with
tyrphos tin inhibitor s and 10 mg of totalcell extracts were tested for
CK-2 activity.Relative CK-2 activity(expressedas %) was calculated
as theratio of CK2ac tivity inDMSO-treated cells (control,10 0%)to
that in cells treated with tyrphostins inhibitors. Concentrations
were10 mMAG879,10mMAG1517,or250nMAG1478.B: Expression
of CK-2 inthe presence of tyrphostininhibitors.Fourtymicrograms
of total extract from PC-3 cells used in Figure 4Awere loaded on
SDS ^PAGE and subjected to Western-blotting using anti-CK-2a
anti-CK-2a0,actin(control)antibodies.C: InhibitionofCK-2activity
by Her-2 mutant.Cells were transfected with PCDNA3.1 or mut-
Her-2 and 24 hr later 2.5 mg of total extracts were tested for CK-2
activity.Relative CK-2 activity(expresseda s %)is the ratios of CK- 2
activity in control PCDNA3.1 cells (100%) to thatin mut-Her-2 cells.
Values (panels A and B) represent themean SEM. Significance, as
compared to the control, was defined as *P<0.05 using a Student
t-test.
Mechanism of NF-kB Activation in Prostate Cancer Cells 311
DISCUSSION
In prostate cancer cells, the sequential signaling
events involved in the constitutive activation of NF-kB
are not clearly defined. It has previously been shown
that in hormone-independent PC-3 cells EGFR and
Her-2 induce the activation of NF-kB through two
distinct mechanisms [11]. We have previously demons-
trated using selective tyrphostin inhibitors that EGFR
activates NF-kB through a mechanism involving the
phosphorylation of IkBaon serine 32/36 thereby alte-
ring the nuclear translocation of p65 [11]. In contrast,
Her-2 activates a signaling pathway that neither affects
the nuclear translocation of p65 nor the phosphoryla-
tion of IkBaon N-terminal serines 32/36 [11]. Here we
extend these observation to show that in PC-3 cells,
EGFR, but not Her-2, is involved in the constitutive
activation of the PI3K/Akt signaling pathway. Inhibi-
tion of the PI3K/Akt signaling pathway leads to a
decreased phosphorylation of IkBaon serines 32 36
and the nuclear translocation of p65. The similarity of
effects between EGFR and PI3K/Akt inhibitors, on the
constitutive NF-kB activation in PC-3 cells, strongly
suggests a model where EGFR is involved in the acti-
vation of NF-kB through the PI3K/Akt activation
(Fig. 6). Furthermore, in line with our results obtained
in PC-3 prostate cancer cells, PI3K has also been impli-
cated in the constitutive activation of NF-kB in several
cancer cell type through stimulation by various growth
factors, including ErbB ligands [810].
In addition, here we demonstrate for the first time
that Her-2 also participates in the constitutive NF-kB
activation through the activation of CK-2. CK-2 has
previously been identified as a kinase responsible for
the constitutive phosphorylation of C-terminal sites of
IkBa[1315,28,29]. This phosphorylation regulates its
intrinsic stability but does not affect its stimulated
degradation [28]. Moreover, the inhibition of CK-2 acti-
vation does not substantially impair either the degra-
dation of IkBaor the nuclear translocation of the p65
subunit of NF-kB [28,30]. Similarly, we have demons-
trated that in PC-3 cells, inhibition of CK-2 neither
impairs the phosphorylation of IkBaon serine 32/36
nor the nuclear translocation of the p65 subunit of NF-
kB. In addition we also observed that Her-2 is involved
in the constitutive activation of CK-2 in PC-3 cells, and
this provides an explanation for the observation that
Her-2 signaling does not affect the phosphorylation
of IkBaon serine 32/36 (Le Page et al. in press).
Altogether, these results strongly implicate Her-2 in the
constitutive activation of NF-kB through the activation
of CK-2 (Fig. 6). This is the first direct evidence show-
ing CK-2 as a component of the constitutive signaling
mediated by Her-2. It would be interesting to extend
this observation to other epithelial cancer cell type
presenting a constitutive activation of Her-2 such as
breast cancer cells.
Surprisingly, in LNCaP cells the overexpression and
activation of Her-2 [18,19] (Le Page et al. in press), as
well as the constititutive activation of PI3K [5,6,23]
(Fig. 1), is not sufficient for inducing a constitutive NF-
kB activation [1,5,6,23]. Several studies have attempted
to explain the reason for the lack of constitutive NF-kB
activity in LNCaP cells. One possibility is that this
phenomenon is due to the presence of the androgen
receptor (AR). Indeed, the absence of androgen in the
culture media of LNCaP cells induces NF-kB activa-
tion, while addition of androgen inhibits this activation
[24,31,32]. The precise mechanism of such inhibition is
still unknown, although protein interaction between
p65 and AR as well as AR and EGFR have been pro-
posed to be responsible for this interference [33,34]. In
addition, the expression of REPS2, a recently described
protein expressed in LNCaP cells, has also been shown
Fig. 6. Model representing mechanisms of constitutive NF-kB
activation in prostate ca ncer cells. In prostate ca ncer cells, we pro-
pose that EGFRac tivates the PI3K/Aktpathway leading to IKK acti-
vation, phosphorylation of IkBaon serine 32/36 and subsequent
nuclear translocation of NF-kB. Her-2 signaling through CK-2
enhances the transactivationalabilityof NF-kB. Additional signaling
mediators such as MAPK and NIK have alsobeen shown to partici-
pa te to the c ons titutive NF-kB activation[4].Severalpharmacologic
and endogenous inhibitors can interfere at each step of this
activation.
312 P age et al .
to act as an inhibitor of p65 [34] and may also explain
the lack of NF-kB activation in LNCaP cells.
In prostate cancer patients, overexpression of EGFR
and Her-2 as well as activation of Akt, CK-2, and NF-kB
correlates with poor prognosis and/or high Gleason
score [3541]. Consequently inhibition of both EGFR
and Her-2 may be an interesting therapeutic strategy.
Indeed the presence of these two independent activa-
tion pathways suggests that strategies to inhibit NF-kB
in prostate cancer cells through selective inhibition of
one receptor may not be completely efficient. In addi-
tion, our study does not exclude the involvement of
other active components of such as MAPK and NIK,
which have already been shown to be constitutively
activated in prostate cancer cell lines [4246] and
potentially involved in the constitutive activation of
NF-kB in these cells [4]. Interventions that target
intracellular proteins, in conjunction with receptor in-
hibitors, are promising but require the precise iden-
tification of such signaling pathway in order to act
optimally. One of the lessons learned with this class of
targets is that we currently do not know how to opti-
mally apply them to the treatment of cancer due to the
lack of knowledge about the specific role of each ErbB
and their signaling in particular cancers. For example,
in contrast to the mechanism observed in breast cancer
cell lines [47], Her-2 is not involved in the activation of
PI3K and CK-2 is not implicated in IkBadegradation or
p65 translocation in PC-3 prostate cancer cells (Figs. 1
and 4C). In addition, negative effects on normal cells
with more closely matched receptor binding capacities
may bring dramatic side effects. Interventions that
target the intracellular protein, in conjunction with a
receptor inhibitor may be more promising. Here we
have demonstrated the additional implication of Akt
and CK-2 as independent signaling pathways consti-
tutively activated by EGFR and Her-2 respectively.
As their constitutive activities revealed here appear
to be present in the native cell lines without the need
of genetic manipulation associated with transfection
studies, this suggests that they are biologically relevant
and may more closely represent what occurs during
disease progression.
CONCLUSION
In conclusion, this study improves our understan-
ding of the ErbB-mediated signaling and shows for the
first time CK-2 as a specific component of Her-2 sig-
naling. We also demonstrate the independent implica-
tion of PI3K and CK-2 in the constitutive activation NF-
kB by EGFR and Her-2 respectively. This allows us to
propose a more complete model about the mechanism
of constitutive activation of NF-kB in prostate cancer
cells (Fig. 6) which appears different from the mechan-
ism previously observed in cancer cell lines such as
breast cancer, where EGFR and Her-2 act as hetero-
dimers. It will be important to extend the present
observations to cancer tissues, in the presence of thera-
peutics that target ErbB members, to determine the
extent to which these signaling pathways are modu-
lated in prostate cancer patients. In this way it will be
possible to determine whether the dual activation of
NF-kB through both EGFR and Her-2 is an important
consideration when attempting to devise novel anti-
cancer therapies for androgen-independent and/or re-
fractory prostate cancer.
ACKNOWLEDGMENT S
The authors thank members of Institute of Cancer of
Montreal for helpful discussion and technical support.
We particularly thank Nathalie Delvoye for technical
assistance and Jean-Simon Diallo for critical reading of
the manuscript. We are very grateful to Dr Ming-Fong
Lin for providing Her-2 and mut-Her-2 plasmids, to
Dr Sylvain Meloche and Dr Alex Ullrich for HERCD533
construction and to Dr. Lichfield for CK-2a0construc-
tion. We also thank Dr Juana Wietzerbin for providing
ConA-luc and kB-ConALuc vector.
REFERENCES
1. Gasparian AV, Yao YJ, Kowalczyk D, Lyakh LA, Karseladze A,
Slaga TJ, Budunova IV. The role of IKK in constitutive activation
of NF-kappaB transcription factor in prostate carcinoma cells.
J Cell Sci 2002;115(Pt 1):141– 151.
2. Ismail HA, Lessard L, Mes-Masson AM, Saad F. Expression of
NF-kappaB in prostate cancer lymph node metastases. Prostate
2004;58(3):308– 313.
3. Shukla S, MacLennan GT, Fu P, Patel J, Marengo SR, Resnick MI,
Gupta S. Nuclear factor-kappaB/p65 (Rel A) is constitutively
activated in human prostate adenocarcinoma and correlates
with disease progression. Neoplasia 2004;6(4):390– 400.
4. Suh J, Payvandi F, Edelstein LC, Amenta PS, Zong WX, Gelinas
C, Rabson AB. Mechanisms of constitutive NF-kappaB activa-
tion in human prostate cancer cells. Prostate 2002;52(3):183
200.
5. Gustin JA, Maehama T, Dixon JE, Donner DB. The PTEN tumor
suppressor protein inhibits tumor necrosis factor-induced
nuclear factor kappa B activity. J Biol Chem 2001;276(29):
27740– 27744.
6. Mayo MW, Madrid LV, Westerheide SD, Jones DR, Yuan XJ,
Baldwin AS Jr., Whang YE. PTEN blocks tumor necrosis factor-
induced NF-kappa B-dependent transcription by inhibiting the
transactivation potential of the p65 subunit. J Biol Chem 2002;
277(13):11116– 11125.
7. Fresno Vara JA, Casado E, de Castro J, Cejas P, Belda-Iniesta C,
Gonzalez-Baron M. PI3K/Akt signalling pathway and cancer.
Cancer Treat Rev 2004;30(2):193– 204.
8. Bhat-Nakshatri P, Sweeney CJ, Nakshatri H. Identification of
signal transduction pathways involved in constitutive NF-
kappaB activation in breast cancer cells. Oncogene 2002;21(13):
2066– 2078.
Mechanism of NF-kB Activation in Prostate Cancer Cells 313
9. de Bono JS, Rowinsky EK. Therapeutics targeting signal
transduction for patients with colorectal carcinoma. Br Med
Bull 2002;64:227– 254.
10. Navolanic PM, Steelman LS, McCubrey JA. EGFR family
signaling and its association with breast cancer development
and resistance to chemotherapy (review). Int J Oncol 2003;22(2):
237– 252.
11. Le Page C, Koumakpayi IH, Lessard L, Mes-Masson AM, Saad F.
EGFR and Her-2 regulate the constitutive activation of NF-
KappaB in PC-3 prostate cancer cells. Prostate 2005; (in press).
12. Litchfield DW. Protein kinase CK2: Structure, regulation and
role in cellular decisions of life and death. Biochem J 2003;
369(Pt 1):1– 15.
13. Barroga CF, Stevenson JK, Schwarz EM, Verma IM. Constitutive
phosphorylation of I kappa B alpha by casein kinase II. Proc Natl
Acad Sci USA 1995;92(17):7637– 7641.
14. McElhinny JA, Trushin SA, Bren GD, Chester N, Paya CV.
Casein kinase II phosphorylates I kappa B alpha at S-283, S-289,
S-293, and T-291 and is required for its degradation. Mol Cell Biol
1996;16(3):899– 906.
15. Schwarz EM, Van Antwerp D, Verma IM. Constitutive phos-
phorylation of IkappaBalpha by casein kinase II occurs prefer-
entially at serine 293: Requirement for degradation of free
IkappaBalpha. Mol Cell Biol 1996;16(7):3554– 3559.
16. Lin A, Karin M. NF-kappaB in cancer: A marked target. Semin
Cancer Biol 2003;13(2):107– 114.
17. Yarden Y. The EGFR family and its ligands in human cancer.
Signalling mechanisms and therapeutic opportunities. Eur J
Cancer 2001;37(Suppl 4):S3– S8.
18. Lee MS, Igawa T, Yuan TC, Zhang XQ, Lin FF, Lin MF. ErbB-2
signaling is involved in regulating PSA secretion in androgen-
independent human prostatecancer LNCaP C-81 cells.Oncogene
2003;22(5):781– 796.
19. Meng TC, Lee MS, Lin MF. Interaction between protein tyrosine
phosphatase and protein tyrosine kinase is involved in andro-
gen-promoted growth of human prostate cancer cells. Oncogene
2000;19(22):2664– 2677.
20. Zhang L, Chang CJ, Bacus SS, Hung MC. Suppressed trans-
formation and induced differentiation of HER-2/neu-over-
expressing breast cancer cells by emodin. Cancer Res 1995;
55(17):3890– 3896.
21. Battistutta R, Sarno S, De Moliner E, Papinutto E, Zanotti G,
Pinna LA. The replacement of ATP by the competitive inhibitor
emodin induces conformational modifications in the catalytic
site of protein kinase CK2. J Biol Chem 2000;275(38):29618
29622.
22. Gschwind A, Prenzel N, Ullrich A. Lysophosphatidic acid-
induced squamous cell carcinoma cell proliferation and motility
involves epidermal growth factor receptor signal transactiva-
tion. Cancer Res 2002;62(21):6329– 6336.
23. Li X, Stark GR. NFkappaB-dependent signaling pathways. Exp
Hematol 2002;30(4):285– 296.
24. Hessenauer A, Montenarh M, Gotz C. Inhibition of CK2 activity
provokes different responses in hormone-sensitiveand hormone-
refractory prostate cancer cells. Int J Oncol 2003;22(6):1263
1270.
25. Guo C, Yu S, Davis AT, Ahmed K. Nuclear matrix targeting of the
protein kinase CK2 signal as a common downstream response to
androgen or growth factor stimulation of prostate cancer cells.
Cancer Res 1999;59(5):1146– 1151.
26. Sarno S, Moro S, Meggio F, Zagotto G, Dal Ben D, Ghisellini P,
Battistutta R, Zanotti G, Pinna LA. Toward the rational design of
protein kinase casein kinase-2 inhibitors. Pharmacol Ther 2002;
93(2-3):159– 168.
27. Sarno S, Reddy H, Meggio F, Ruzzene M, Davies SP, Donella-
Deana A, Shugar D, Pinna LA. Selectivity of 4,5,6,7-tetrabromo-
benzotriazole, an ATP site-directed inhibitor of protein kinase
CK2 (‘casein kinase-2’). FEBS Lett 2001;496(1):44– 48.
28. Lin R, Beauparlant P, Makris C, Meloche S, Hiscott J.
Phosphorylation of IkappaBalpha in the C-terminal PEST do-
main by casein kinase II affects intrinsic protein stability. Mol
Cell Biol 1996;16(4):1401– 1409.
29. Janosch P, Schellerer M, Seitz T, Reim P, Eulitz M, Brielmeier M,
Kolch W, Sedivy JM, Mischak H. Characterization of IkappaB
kinases. IkappaB-alpha is not phosphorylated by Raf-1 or
protein kinase C isozymes, but is a casein kinase II substrate.
J Biol Chem 1996;271(23):13868– 13874.
30. Farah M, Parhar K, Moussavi M, Eivemark S, Salh B. 5,6-
dichloro-ribifuranosylbenzimidazole- and apigenin-induced
sensitization of colon cancer cells to TNF-alpha-mediated
apoptosis. Am J Physiol Gastrointest Liver Physiol 2003;285(5):
G919– G928.
31. Keller ET, Chang C, Ershler WB. Inhibition of NFkappaB activity
through maintenance of IkappaBalpha levels contributes to
dihydrotestosterone-mediated repression of the interleukin-6
promoter. J Biol Chem 1996;271(42):26267– 26275.
32. Altuwaijri S, Lin HK, Chuang KH, Lin WJ, Yeh S, Hanchett LA,
Rahman MM, Kang HY, Tsai MY, Zhang Y, Yang L, Chang C.
Interruption of nuclear factor kappaB signaling by the androgen
receptor facilitates 12-O-tetradecanoylphorbolacetate-induced
apoptosis in androgen-sensitive prostate cancer LNCaP cells.
Cancer Res 2003;63(21):7106– 7112.
33. Palvimo JJ, Reinikainen P, Ikonen T, Kallio PJ, Moilanen A,
Janne OA. Mutual transcriptional interference between RelA
and androgen receptor. J Biol Chem 1996;271(39):24151– 24156.
34. Penninkhof F, Grootegoed JA, Blok LJ. Identification of REPS2 as
a putative modulator of NF-kappaB activity in prostate cancer
cells. Oncogene 2004;23(33):5607– 5615.
35. Hernes E, Fossa SD, Berner A, Otnes B, Nesland JM. Expression
of the epidermal growth factor receptor family in prostate
carcinoma before and during androgen-independence. Br J
Cancer 2004;90(2):449– 454.
36. Di Lorenzo G, Tortora G, D’Armiento FP, De Rosa G, Staibano S,
Autorino R, D’Armiento M, De Laurentiis M, De Placido S,
Catalano G, Bianco AR, Ciardiello F. Expression of epidermal
growth factor receptor correlates with disease relapse and
progression to androgen-independence in human prostate
cancer. Clin Cancer Res 2002;8(11):3438– 3444.
37. Liao Y, Grobholz R, Abel U, Trojan L, Michel MS, Angel P, Mayer
D. Increase of AKT/PKB expression correlates with Gleason pat-
tern in human prostate cancer. Int J Cancer 2003;107(4):676–680.
38. Ahmed K. Significance of the casein kinase system in cell growth
and proliferation with emphasis on studies of the androgenic
regulation of the prostate. Cell Mol Biol Res 1994;40(1):1–11.
39. Yenice S, Davis AT, Goueli SA, Akdas A, Limas C, Ahmed K.
Nuclear casein kinase 2 (CK-2) activity in human normal, benign
hyperplastic, and cancerous prostate. Prostate 1994;24(1):11– 16.
40. Lessard L, Mes-Masson AM, Lamarre L, Wall L, Lattouf JB, Saad
F. NF-kappa B nuclear localization and its prognostic signi-
ficance in prostate cancer. BJU Int 2003;91(4):417– 420.
41. Kreisberg JI, Malik SN, Prihoda TJ, Bedolla RG, Troyer DA,
Kreisberg S, Ghosh PM. Phosphorylation of Akt (Ser473) is an
excellent predictor of poor clinical outcome in prostate cancer.
Cancer Res 2004;64(15):5232– 5236.
314 Page et al .
42. Yeh S, Lin HK, Kang HY, Thin TH, Lin MF, Chang C. From
HER2/Neu signal cascade to androgen receptor and its co-
activators: A novel pathway by induction of androgen target
genes through MAP kinase in prostate cancer cells. Proc Natl
Acad Sci USA 1999;96(10):5458– 5463.
43. Qiu Y, Ravi L, Kung HJ. Requirement of ErbB2 for signalling by
interleukin-6 in prostate carcinoma cells. Nature 1998;393(6680):
83– 85.
44. Jones HE, Dutkowski CM, Barrow D, Harper ME, Wakeling
AE, Nicholson RI. New EGF-R selective tyrosine kinase
inhibitor reveals variable growth responses in prostate carci-
noma cell lines PC-3 and DU-145. Int J Cancer 1997;71(6):1010
1018.
45. Grasso AW, Wen D, Miller CM, Rhim JS, Pretlow TG, Kung HJ.
ErbB kinases and NDF signaling in human prostate cancer cells.
Oncogene 1997;15(22):2705– 2716.
46. Shimada K, Nakamura M, Ishida E, Kishi M, Yonehara S, Konishi
N. Contributions of mitogen-activated protein kinase and
nuclear factor kappa B to N-(4-hydroxyphenyl)retinamide-
induced apoptosis in prostate cancer cells. Mol Carcinog 2002;
35(3):127– 137.
47. Pianetti S, Arsura M, Romieu-Mourez R, Coffey RJ, Sonenshein
GE. Her-2/neu overexpression induces NF-kappaB via a PI3-
kinase/Akt pathway involving calpain-mediated degradation
of IkappaB-alpha that can be inhibited by the tumor suppressor
PTEN. Oncogene 2001;20(11):1287– 1299.
Mechanism of NF-kB Activation in Prostate Cancer Cells 315
... The net result of this process is translation to the nucleus of NF-kB (Gasparian et al, 2002). A constitutive activation of the PI3K/Akt pathway through IKK has been implicated in this activation of NF-kB in the PC-3 hormone-independent prostate cancer cell line (Gustin et al, 2001;Gasparian et al, 2002;Mayo et al, 2002;Le Page et al, 2005b). The activation of PI3K results in the translocation of Akt from the cytoplasm to the inner membrane, where Akt is phosphorylated by upstream kinases, PDK and ILK. ...
... The activation of Akt leads to the phosphorylation of downstream targets such as IKK, GSK3, Bad, mTOR, caspase 9 and the FOXO family of forkhead transcription factors (Vivanco and Sawyers, 2002). More recently, we provided evidence that the epidermal growth factor receptor (EGFR) signalling pathway is implicated as an early event that constitutively activates the PI3K/Akt/NF-kB pathway in PC-3 prostate cancer cells (Le Page et al, 2005b). ...
... Epidermal growth factor receptor and Her-2 are members of the ErbB growth factor receptor family composed of four distinct receptors: EGFR/ErbB1, Her-2/ErbB2/c-neu, Her-3/ErbB3 and Her-4/ ErbB4. In PC-3 cells, EGFR and Her-2 overexpression and constitutive activation modulate NF-kB signalling through two distinct mechanisms: one dependent and one independent of the phosphorylation of IkBa on ser32/ser36 (Le Page et al, 2005b). ...
... Although CKII is not considered an oncoprotein, studies have shown that its dysregulation can contribute to its oncogenic potential [47,51]. Moreover, high CKII levels are associated with cell proliferation: it has been found to be greatly increased during malignant transformation in a variety of tumours, raising the possibility of its having a key role in the tumoural phenotype [47,[51][52][53][54]. Targeting CKII with specific inhibitors has shown considerable therapeutic potential, and some inhibitors are currently in clinical trials with apparently promising outcomes [42,[55][56][57]. ...
Article
Full-text available
Background Oncogenic Human Papillomaviruses (HPVs) base their transforming potential on the action of both E6 and E7 viral oncoproteins, which perform cooperative or antagonistic actions and thus interfere with a variety of relevant cellular targets. Among them, the expression of some PDZ-containing polarity proteins, as DLG1 and hScrib, is altered during the HPV life cycle and the consequent malignant transformation. Together with the well-established interference of E6 with PDZ proteins, we have recently shown that E7 viral oncoprotein is also responsible for the changes in abundance and localization of DLG1 observed in HPV-associated lesions. Given that the mechanisms involved remained only partially understood, we here thoroughly analyse the contribution of a crucial E7 post-translational modification: its CKII-dependent phosphorylation. Moreover, we extended our studies to hScrib, in order to investigate possible conserved regulatory events among diverse PDZ targets of HPV. Methods We have acutely analysed the expression of DLG1 and hScrib in restrictive conditions for E7 phosphorylation by CKII in epithelial culture cells by western blot and confocal fluorescence microscopy. We made use of genome-edited HPV-positive cells, specific inhibitors of CKII activity and transient expression of the viral oncoproteins, including a mutant version of E7. Results We here demonstrate that the functional phosphorylation of E7 oncoprotein by the CKII cellular kinase, a key regulatory event for its activities, is also crucial to counteract the E6-mediated degradation of the PDZ-polarity protein DLG1 and to promote its subcellular redistribution. Moreover, we show that the CKII-dependent phosphorylation of E7 is able to control the expression of another PDZ target of HPV: hScrib. Remarkably, we found this is a shared feature among different oncogenic HPV types, suggesting a common path towards viral pathogenesis. Conclusions The present study sheds light into the mechanisms behind the misexpression of PDZ-polarity proteins during HPV infections. Our findings stress the relevance of the CKII-mediated regulation of E7 activities, providing novel insights into the joint action of HPV oncoproteins and further indicating a conserved and most likely crucial mechanism during the viral life cycle and the associated transformation.
... Of note, NF-jB may be activated upstream by other oncogenic signaling pathways widely implicated in tumorigenesis, including PI3K/AKT and Wnt signaling (16,177,185,329). Interestingly, NDRG1 is able to inhibit activation of PI3K/AKT signaling by decreasing p-AKT (activator of LYRIC) and c-Myc levels, while also increasing expression of the tumor suppressor, PTEN, which acts as an antagonist to PI3K/AKT signaling (80, 171, 357) (Fig. 10). ...
Article
Significance: Glutaredoxin (Grx1), an evolutionarily conserved and ubiquitous enzyme, regulates redox signal transduction and protein redox homeostasis by catalyzing reversible S-glutathionylation. Grx1 plays different roles in different cell types. In Parkinson's disease (PD), Grx1 regulates apoptosis signaling in dopaminergic neurons, so that loss of Grx1 leads to increased cell death; in microglial cells, Grx1 regulates pro-inflammatory signaling, so that upregulation of Grx1 promotes cytokine production. Here we examine the regulatory roles of Grx1 in PD with a view toward therapeutic innovation. Recent advances: In postmortem midbrain PD samples, Grx1 was decreased relative to controls, specifically within dopaminergic neurons. In C. elegans models of PD, loss of the Grx1 homolog led to exacerbation of the neurodegenerative phenotype. This effect was partially relieved by overexpression of neuroprotective DJ-1, consistent with regulation of DJ-1 content by Grx1. Increased GLRX copy number in PD patients was associated with earlier PD onset; and Grx1 levels correlated with levels of pro-inflammatory TNF- in mouse and human brain samples. In vitro studies showed Grx1 to be upregulated upon pro-inflammatory activation of microglia. Direct overexpression of Grx1 increased microglial activation; silencing Grx1 diminished activation. Grx1 upregulation in microglia corresponded to increased neuronal cell death in co-culture. Overall, these studies identify competing roles of Grx1 in PD etiology. Critical issues: The dilemma regarding Grx1 as a PD therapeutic target is whether to stimulate its upregulation for neuroprotection or inhibit its pro-inflammatory activity. Future directions: Further investigation is needed to understand the preponderant role of Grx1 regarding dopaminergic neuronal survival.
... Although EGFR kinase inhibitors, such as erlotinib and gefitinib, inhibit EGFR, the EGFRvIII-bearing cell lines are resistant to these inhibitors [10][11][12]. NF-κB can be activated by EGFR via an AKT-dependent [13] or AKT-independent pathway [14,15] and in response to therapy. The constitutive activation of NF-κB has been detected widespread in human cancers [16,17], including GBM. ...
Article
Worldwide, glioblastoma (GBM) is the most lethal and frequent intracranial tumor. Despite decades of study, the overall survival of GBM patients remains unchanged. epidermal growth factor receptor (EGFR) amplification and gene mutation are thought to be negatively correlated with prognosis. In this study, we used proteomics to determine that UBXN1 is a negative downstream regulator of the EGFR mutation vIII (EGFRvIII). Via bioinformatics analysis, we found that UBXN1 is a factor that can improve glioma patients’ overall survival time. We also determined that the down-regulation of UBXN1 is mediated by the upregulation of H3K27me3 in the presence of EGFRvIII. Because NF-κB can be negatively regulated by UBXN1, we believe that EGFRwt/vIII activates NF-κB by suppressing UBXN1 expression. Importantly, we used the latest genomic editing tool, CRISPR/Cas9, to knockout EGFRwt/vIII on exon 17 and further proved that UBXN1 is negatively regulated by EGFRwt/vIII. Furthermore, knockout of EGFR/EGFRvIII could benefit GBM in vitro, indicating that CRISPR/Cas9 is a promising therapeutic strategy for both EGFR amplification and EGFR mutation-bearing patients.
... These mechanisms of mTOR promoter regulation suggested by our results do not explain, however, how PI3k activation selectively regulates p65 binding to MTOR promoter after Nrf2 induction. It was shown for example that EGFR activates NF-kB (p65) through the PI3K/Akt pathway, by promoting the nuclear translocation of p65 (41). Also, in PI3k-activated prostate cells, MTOR was shown to control p65 activity by increasing its translocation to the nucleus to induce its target genes (42). ...
Article
Full-text available
Nrf2 is a master transcription factor that regulates a wide variety of cellular proteins by recognizing and binding to antioxidant response elements (AREs) in their gene promoter regions. In this study we show that increasing cellular Nrf2 results in transcriptional activation of the gene for mTOR, which is central to the PI3kinase signaling pathway. This is the case in cells with normal physiological PI3kinase. However, in cells with abnormally active PI3K, increased cellular Nrf2 levels have no effect on mTOR. ChIP assays results show that increased Nrf2 binding is associated with decreased p65 binding and H3-K27me3 signal (marker of gene repression), as well as increased H3-K4me3 signal (marker of gene activation). However, in cells with PI3k-activation, no effect of cellular Nrf2 increase on mTOR transcription was observed. In these cells, increasing Nrf2 levels increases Nrf2 promoter binding marginally, while p65 binding and H3-K27me3 mark were significantly increased and H3-K4me3 signal is reduced. Together, these data show for the first time that Nrf2 directly regulates mTOR transcription when the PI3kinase pathway is intact, while this function is lost when PI3k is activated. We have identified a link between the Nrf2 system of sensing environmental stress and mTOR, which is a key cellular protein in metabolism. Studies in cells with activating mutations in the PI3K pathway suggest that Nrf2 transcriptional regulation of mTOR is related to promoter binding of p65 and of methylation of histone residues permissive of transcription.
... To our knowledge, there are no published reports relating CK2 and NF-jB protein expression in BPH and PCa human tissue samples. Two studies in cultured breast and prostate cells determined that CK2 phosphorylation and the subsequent degradation of IjB-a causes classical NF-jB (e.g., p50/p65) activation through the EGFR2/ HER2 pathway [29,31]. We examined the coexpression of CK2a and NF-jB p65 proteins in human PCa and BPH specimens to determine how their expression levels in the nuclear compartment relate to one another in the context of benign and malignant proliferative signaling. ...
Article
Full-text available
Protein kinase CK2 plays a critical role in cell growth, proliferation, and suppression of cell death. CK2 is overexpressed, especially in the nuclear compartment, in the majority of cancers, including prostate cancer (PCa). CK2-mediated activation of transcription factor nuclear factor kappa B (NF-κB) p65 is a key step in cellular proliferation, resulting in translocation of NF-κB p65 from the cytoplasm to the nucleus. As CK2 expression and activity are also elevated in benign prostatic hyperplasia (BPH), we sought to increase the knowledge of CK2 function in benign and malignant prostate by examination of the relationships between nuclear CK2 and nuclear NF-κB p65 protein expression. The expression level and localization of CK2α and NF-κB p65 proteins in PCa and BPH tissue specimens was determined. Nuclear CK2α and NF-κB p65 protein levels are significantly higher in PCa compared with BPH, and these proteins are positively correlated with each other in both diseases. Nuclear NF-κB p65 levels correlated with Ki-67 or with cytoplasmic NF-κB p65 expression in BPH, but not in PCa. The findings provide information that combined analysis of CK2α and NF-κB p65 expression in prostate specimens relates to the disease status. Increased nuclear NF-κB p65 expression levels in PCa specifically related to nuclear CK2α levels, indicating a possible CK2-dependent relationship in malignancy. In contrast, nuclear NF-κB p65 protein levels related to both Ki-67 and cytoplasmic NF-κB p65 levels exclusively in BPH, suggesting a potential separate impact for NF-κB p65 function in proliferation for benign disease as opposed to malignant disease.
... 5 In addition, protein kinases, including mitogen-activated protein kinases (MAPKs) and Akt, are known to be upstream regulators of NF-κB and also play important roles in controlling MMPs. [6][7][8] Curcumol ( Figure 1) is one of the major active components in the essential oil of Rhizoma Curcumae, which is traditionally used for the treatment of gynecological tumors in China. It is reported that curcumol inhibited the proliferation and attenuated the total RNA synthesis of MCF-7, MDA-MB-231, HeLa, and OV-UL-2 gynecological cancer cells with negligible effect on normal cells. ...
Article
Full-text available
Curcumolhas been reported to possess antitumor activity. However, its effect and mechanisms against tumor metastasis are still unclear. This study is to investigate the inhibitory effect of curcumol on breast cancer cell metastasis and elucidate the underlying molecular mechanisms. Our results showed that noncytotoxicity was caused by curcumol within 10 to 40 µg/mL in MDA-MB-231 and 4T1 cells for 24 hours, whereas sustained treatment with curcumol for 14 days significantly suppressed the clonogenic activity of cells. Importantly, curcumol at noncytotoxic concentrations suppressed the migration ability of both MDA-MB-231 and 4T1 cells. Moreover, curcumol suppressed the migration and invasion of MDA-MB-231 cells in the Boyden chamber migration and invasion assay and inhibited the adhesion of MDA-MB-231 cells onto the matrigel. Further investigations revealed that curcumol decreased the enzyme activity and protein expression of matrix metalloproteinase (MMP-9) in MDA-MB-231 cells. Moreover, curcumol inhibited the activation of c-Jun N-terminal kinase (JNK) 1/2 and Akt (Ser473). Meanwhile, it also inhibited the nuclear translocation and transcriptional activity of nuclear factor κB (NF-κB). Furthermore, JNK inhibitor SP600125 and Akt (Ser473) inhibitor LY294002 enhanced the inhibition of curcumol on NF-κB p65 nuclear translocation. Finally, supplementation with SP600125, LY294002, or NF-κB inhibitor Ammonium pyrrolidinedithiocarbamate (PDTC) significantly enhanced the inhibitory effect of curcumol on MMP-9 expression and cell migration, invasion, and adhesion in MDA-MB-231 cells. Our findings provide evidence for the suppression of breast cancer cell metastasis by curcumol and suggest that the inhibition of MMP-9 via JNK1/2 and Akt (Ser473)-dependent NF-κB signaling pathways may be the underlying mechanisms.
... We also observed increased levels of cytoplamic IκBα and decreased levels of cytoplasmic phosphorylated IκBα and nuclear NF-κB after re-expression of miR-146a or EGFR siRNA transfection, suggesting that miR-146a could regulate IκB/NF-κB signaling through the inhibition of both IRAK-1 and EGFR (Fig. 5D). Our results are also consistent with the findings reported by other investigators showing that IRAK-1 could inhibit IκBα leading to the activation of NF-κB (32,33), and that EGFR or Her-2/neu are known activators of NF-κB through the phosphorylation of IκBα mediated via by Akt (34) and the degradation of IκBα by calpain (35). Therefore, the inactivation of NF-κB could be another molecular mechanism by which miR-146a inhibits cancer cell invasion and metastasis. ...
Article
Pancreatic cancer (PC) is an aggressive malignancy with high mortality and is believed to be in part due to its highly invasive and metastatic behavior, which is associated with over-expression of EGFR and activation of NF-κB. Emerging evidence also suggest critical roles of microRNAs (miRNAs) in the regulation of various pathobiological processes including metastasis in PC and in other human malignancies. In the present study, we found lower expression of miR-146a in PC cells compared to normal human pancreatic duct epithelial (HPDE) cells. Interestingly, re-expression of miR-146a inhibited the invasive capacity of Colo357 and Panc-1 PC cells with concomitant down-regulation of EGFR and IRAK-1. Mechanistic studies including miR-146a re-expression, anti-miR-146 transfection, and EGFR knock-down experiment showed that there was a crosstalk between EGFR, MTA-2, IRAK-1, IκBα and NF-κB. Most importantly, we found that the treatment of PC cells with "natural agents" [3,3'-diinodolylmethane (DIM) or isoflavone] led to an increase in the expression of miR-146a and consequently down-regulated the expression of EGFR, MTA-2, IRAK-1 and NF-κB, resulting in the inhibition of invasion of Colo357 and Panc-1 cells. These results provide experimental evidence in support of the role of DIM and isoflavone as potential non-toxic agents as regulators of miRNA, which could be useful for the inhibition of cancer cell invasion and metastasis, and further suggesting that these agents could be important for designing novel targeted strategy for the treatment of PC.
Article
Significance: Nuclear factor kappa B (NF-κB) signaling is essential under physiologically relevant conditions. However, aberrant activation of this pathway plays a pertinent role in tumorigenesis and contributes to resistance. Recent Advances: The importance of the NF-κB pathway means that its targeting must be specific to avoid side effects. For many currently used therapeutics and those under development, the ability to generate reactive oxygen species (ROS) is a promising strategy. Critical issues: As cancer cells exhibit greater ROS levels than their normal counterparts, they are more sensitive to additional ROS, which may be a potential therapeutic niche. It is known that ROS are involved in (i) the activation of NF-κB signaling, when in sublethal amounts; and (ii) high levels induce cytotoxicity resulting in apoptosis. Indeed, ROS-induced cytotoxicity is valuable for its capabilities in killing cancer cells, but establishing the potency of ROS for effective inhibition of NF-κB signaling is necessary. Indeed, some cancer treatments, currently used, activate NF-κB and may stimulate oncogenesis and confer resistance. Future directions: Thus, combinatorial approaches using ROS-generating agents alongside conventional therapeutics may prove an effective tactic to reduce NF-κB activity to kill cancer cells. One strategy is the use of thiosemicarbazones, which form redox-active metal complexes that generate high ROS levels to deliver potent antitumor activity. These agents also upregulate the metastasis suppressor, N-myc downstream regulated gene 1 (NDRG1), which functions as an NF-κB signaling inhibitor. It is proposed that targeting NF-κB signaling may proffer a new therapeutic niche to improve the efficacy of anticancer regimens. Antioxid. Redox Signal. 00, 000-000.
Article
A novel and the shortest route, thus far, for preparing cytosporone B (Csn-B) is reported. Csn-B and two analogs were used to probe the importance of hydroxyl groups at the 3- and 5-positions of the Csn-B benzene ring in inhibiting the viability of human H460 lung cancer and LNCaP prostate cancer cells, inducing H460 cell apoptosis, and interacting with the NR4A1 (TR3) ligand-binding domain (LBD). These studies indicate that Csn-B and 5-Me-Csn-B, having a phenolic hydroxyl at the 3-position of their aromatic rings, had similar activities in inhibiting cancer cell viability and in inducing apoptosis, whereas 3,5-(Me)2-Csn-B was unable to do so. These results are in agreement with ligand-binding experiments showing that the interaction with the NR4A1 LBD required the presence of the 3-hydroxyl group.
Article
Full-text available
BACKGROUND The mechanism through which NF-kappaB (NF-κB) is constitutively activated in prostate cancer cells remains unclear. We investigated whether members of the ErbB family of epidermal growth factor receptors (EGFR) are involved in the constitutive activation of NF-κB in prostate cancer cell lines.METHODS AND RESULTSEGFR, Her-2, and ErbB3 are expressed and constitutively activated in PC-3, DU145, and LNCaP prostate cancer cells lines. Using several pharmacological ErbB inhibitors, we demonstrate that EGFR and Her-2 are involved in the constitutive activation of NF-κB in PC-3 cells through two different mechanisms. EGFR activates NF-κB through the phosphorylation of IκBα on serines 32/36 thereby influencing the nuclear translocation of the p65 subunit. In contrast, Her-2 activates NF-κB independently of IκBα phosphorylation on serines 32/36.CONCLUSION This study directly implicates ErbB receptors in the activation of NF-κB in PC-3 prostate cancer cells. © 2005 Wiley-Liss, Inc.
Article
OBJECTIVE To detect the subcellular localization of NF-κB (p65) in human prostate cancer tissues of different histological grades, and to test whether NF-κB localization alone, or combined with the histological grade, can be used to predict patient outcome.PATIENTS AND METHODS Prostate cancer tissues were obtained from radical prostatectomy specimens; the histological grade was determined using the Gleason grading system. Clinical outcomes were defined as good (5-year disease-free survival with undetectable levels of prostate specific antigen) or poor (progression to bone metastases). The subcellular localization of NF-κB was visualized by immunohistochemistry using an anti-p65 antibody.RESULTSThe NF-κB subcellular localization was initially assessed in 45 specimens; in these samples a nuclear localization of NF-κB was specific to cancer tissues, but did not correlate with the Gleason score (P = 0.089). NF-κB was then assessed as a prognostic marker to complement Gleason score in predicting cancer progression. Tumour tissues from 30 men with a known clinical outcome were included; 10 of 17 patients who had a poor outcome were positive for NF-κB nuclear staining, whereas only two of 13 with a good outcome were positive (P = 0.026). When NF-κB subcellular localization and Gleason score were combined, two risk categories of progression were defined. Eleven of 13 specimens from those with a good outcome were in the low-risk category (Gleason 2–4 or Gleason 5–7 with negative nuclear NF-κB) and 12 of 17 in the poor outcome group were in the high-risk category (Gleason 8–10 or Gleason 5–7 with positive nuclear NF-κB; P = 0.004).CONCLUSIONNF-κB is detectable in the nucleus in prostate cancer tissues and positivity can be used to help predict patient outcome. Multivariate analyses using other clinical and molecular variables are underway, and will validate the usefulness of NF-κB as a prognostic factor.
Article
Growth factors and their transmembrane receptor tyrosine kinases play important roles in cell proliferation, survival, migration and differentiation. One group of growth factors, comprising epidermal growth factor (EGF)-like proteins and neuregulins, stimulates cells to divide by activating members of the EGF receptor (EGFR) family, which consists of the EGFR itself and the receptors known as HER2–4. This highly conserved signalling module plays a fundamental role in the morphogenesis of a diverse spectrum of organisms, ranging from humans to nematodes, and has also been implicated in the development and growth of many types of human tumour cells. In humans, more than 30 ligands and the EGFR family of four receptors lie at the head of a complex, multi-layered signal-transduction network. Different activated receptor–ligand complexes vary in both the strength and type of cellular responses that they induce. Analysis of the multiple processes that modulate EGFR signal transduction, such as receptor heterodimerisation and endocytosis, has revealed new therapeutic opportunities and elucidated mechanisms contributing to the efficacy of existing anticancer treatments.
Article
The imbalance between proliferation and programmed cell death (apoptosis) is one of the critical cellular events that lead to oncogenesis. While there is no doubt that uncontrolled cell proliferation is essential for the development of cancer, deregulation of apoptosis may play an equally important role in this process. Inhibition of apoptosis prevents the death of tumor cells with DNA damage either associated with carcinogenic initiation or cancer therapy. The transcription factor NF-κB is a key regulator in oncogenesis. By promoting proliferation and inhibiting apoptosis, NF-κB tips the balance between proliferation and apoptosis toward malignant growth in tumor cells.
Article
The transcription factor NFκB is activated by numerous stimuli. Once NFκB is fully activated, it participates in the regulation of various target genes in different cells to exert its biological functions. NFκB has often been referred to as a central mediator of the immune response, since a large variety of bacteria and viruses can lead to the activation of NFκB, which in turn controls the expression of many inflammatory cytokines, chemokines, immune receptors, and cell surface adhesion molecules. Recent studies have shown that NFκB may function more generally as a central regulator of stress responses, since different stressful conditions, including physical stress, oxidative stress, and exposure to certain chemicals, also lead to NFκB activation. Furthermore, NFκB blocks cell apoptosis in several cell types. Taken together, these findings make it clear that NFκB plays an important role in cell proliferation and differentiation. It is the intention of this review to cover the various NFκB-dependent signaling pathways, thereby to achieve a better understanding of the mechanisms of NFκB activation and the physiological functions of activated NFκB.
Article
In previous work, we had observed that chromatin-associated nonhistone protein phosphorylation, catalyzed by intrinsic protein kinase reaction in chromatin preparations from human benign prostatic hyperplasia (BPH) prostate samples was markedly elevated, compared with the normal prostate chromatin samples [Rayan et al: Cancer Res 45:2277-2282, 1985]. The properties of this protein kinase reaction were suggestive of the involvement of casein kinase(s). By employing the specific synthetic substrate for casein kinase 2 (CK-2) for assays in cellular fractions, we have shown that this protein kinase is present in human prostate chromatin. Its activity is increased in BPH chromatin by about 25-fold, as compared with its activity in the normal prostate chromatin. This suggests that CK-2 is a possible mediator of the enhanced phosphorylation of chromosomal proteins in BPH chromatin. By comparison, CK-2 activity in chromatin preparations from prostatic carcinoma samples was markedly less elevated than that of the BPH chromatin. Immunohistochemical analysis of the enzyme in human frozen sections of prostate tissue samples showed that the enzyme immunostaining was diffuse in the cytoplasm, but more intense in the nucleus, especially in the nucleoli. In general, the staining corresponded with the enzymic data. However, sections from prostatic carcinoma samples appeared to show differential staining, depending on the Gleason's grade of the sample. The samples with higher Gleason's grade showed less intense immunostain in the nucleus, compared with samples of lower Gleason's grade. Further, regions of sections in samples with higher Gleason's grade did not show any immunostaining. These differences in the characteristics of CK-2 expression in prostatic carcinoma samples may be potentially significant, but need to be evaluated further for their significance to the pathobiology of prostatic neoplasia.
Article
Protein phosphorylation is a significant mechanism in many cellular functions, including genomic regulation and control of cell proliferation. Thus, investigations of protein kinases (PKs) in appropriate experimental models are of intense current interest. Employing androgenic regulation of the rat ventral prostate (RVP) as an experimental model, we have investigated certain nuclear PK reactions with the aim of defining their role in androgen-mediated genomic regulation in the prostate and their implications for human prostate pathobiology. The author's laboratory identified androgen-sensitive PKs in the prostatic cell nucleus that might be involved in regulation of the gland. These PKs are analogous to casein kinase 1 (CK-1) and casein kinase 2 (CK-2), which are important multipotential enzymes in growth regulation and cell proliferation. Because CK-2 demonstrated a greater androgen sensitivity, we investigated the mechanism of its regulation at the level of transcription in the RVP, finding that androgens exert a substantial tissue-specific effect on its expression. However, regulation of CK-2 gene transcription does not appear to be an early event in androgen action, an observation that did not accord with our previous documentation of an early androgenic modulation of nuclear CKs in the prostate. Studies to uncover alternative mechanisms of regulation of CK-2 yielded a potentially important observation that androgenic regulation involves changes in the association of the enzyme with subnuclear compartments as an early event in its growth control function. Additionally, chromatin preparations from human benign prostatic hyperplasia (BPH) demonstrate a large enhancement in intrinsic CK-2 activity compared with normal tissue. An increase also is observed in specimens of prostatic carcinoma, although the change is less substantial than that in BPH. There appears to be a correlation between the localization of CK-2 in sections of prostatic carcinoma and the Gleason grade, a measure of the degree of differentiation. Further investigations of the cellular as well as the molecular mechanisms of CK-2 regulation in the prostate as an experimental model of the response to influences on growth may yield new insight into the function of this PK.
Article
The amplification and overexpression of the HER-2/neu proto-oncogene, which encodes the tyrosine kinase receptor p185neu, have been observed frequently in tumors from human breast cancer patients and are correlated with poor prognosis. To explore the potential of chemotherapy directed at the tyrosine kinase of p185neu, we have found that emodin (3-methyl-1,6,8-trihydroxyanthraquinone), a tyrosine kinase inhibitor, suppresses autophosphorylation and transphosphorylation activities of HER-2/neu tyrosine kinase, resulting in tyrosine hypophosphorylation of p185neu in HER-2/neu-overexpressing breast cancer cells. Emodin, at a 40-microM concentration, which repressed tyrosine kinase of p185neu, efficiently inhibited both anchorage-dependent and anchorage-independent growth of HER-2/neu-overexpressing breast cancer cells. However, the inhibition was much less effective for those cells expressing basal levels of p185neu under the same conditions. Emodin also induced differentiation of HER-2/neu-overexpressing breast cancer cells by exhibiting a morphological maturation property of large lacy nuclei surrounded by sizable flat cytoplasm and by showing a measurable production of large lipid droplets, which is a marker of mature breast cells. Therefore, our results indicate that emodin inhibits HER-2/neu tyrosine kinase activity and preferentially suppresses growth and induces differentiation of HER-2/neu-overexpressing cancer cells. These results may have chemotherapeutic implications for using emodin to target HER-2/neu-overexpressing cancer cells.
Article
The NF-kappa B/Rel proteins are sequestered in the cytoplasm in association with the phosphorylated form of I kappa B alpha. Upon induction with a wide variety of agents, the activity of NF-kappa B/Rel proteins is preceded by the rapid degradation of I kappa B alpha protein. We report the identification and partial purification of a cellular kinase from unstimulated or stimulated murine cells, which specifically phosphorylates the C terminus of I kappa B alpha. There are several consensus sites for casein kinase II (CKII) in the C-terminal region of I kappa B alpha. Additionally, the activity of the cellular kinase is blocked by antibodies against the alpha subunit of CKII. No phosphorylation of the C-terminal region of I kappa B alpha can be detected if the five possible serine and threonine residues that can be phosphorylated by CKII are mutated to alanine. A two-dimensional tryptic phosphopeptide map of I kappa B alpha from unstimulated cells was identical to that obtained by in vitro phosphorylation of I kappa B alpha with the partially purified cellular kinase. We propose that constitutive phosphorylation of I kappa B alpha is carried out by CKII.