ArticlePDF Available

Osteoblast-induced EGFR/ERBB2 signaling in androgen-sensitive prostate carcinoma cells characterized by multiplex kinase activity profiling

Authors:

Abstract and Figures

Bone metastases in prostate cancer are predominantly osteoblastic. To study regulatory mechanisms underlying the establishment of prostate cancer within an osteoblastic microenvironment, human androgen-sensitive prostate carcinoma cells (LNCaP) were treated with culture medium conditioned by human osteoblast-derived sarcoma cells (OHS), and activated signalling pathways in the carcinoma cells were analyzed using microarrays with tyrosine kinase substrates. Network interaction analysis of substrates with significantly increased phosphorylation levels revealed that signalling pathways mediated by EGFR and ERBB2 were activated in LNCaP cells under OHS influence but also by androgen treatment. Activation of EGFR/ERBB2 signalling was also found in LNCaP cells in cocultures with OHS cells or osteoblastic cells that had been differentiated from human mesenchymal stem cells. Our experimental data suggests osteoblast-directed induction of signalling activity via EGFR and ERBB2 in prostate carcinoma cells and may provide a rationale for the use of EGFR or ERBB2 inhibition in systemic prevention or treatment of metastatic prostate cancer in the androgen-sensitive stage of the disease.
Content may be subject to copyright.
RESEARCH PAPER
Osteoblast-induced EGFR/ERBB2 signaling in androgen-sensitive
prostate carcinoma cells characterized by multiplex kinase
activity profiling
A
˚
se Bratland Æ Piet J. Boender Æ Hanne K. Høifødt Æ Ingrid H. G. Østensen Æ
Rob Ruijtenbeek Æ Meng-yu Wang Æ Jens P. Berg Æ Wolfgang Lilleby Æ
Øystein Fodstad Æ Anne Hansen Ree
Received: 4 June 2008 / Accepted: 27 February 2009 / Published online: 18 March 2009
Ó The Author(s) 2009. This article is published with open access at Springerlink.com
Abstract Bone metastases in prostate cancer are pre-
dominantly osteoblastic. To study regulatory mechanisms
underlying the establishment of prostate cancer within an
osteoblastic microenvironment, human androgen-sensitive
prostate carcinoma cells (LNCaP) were treated with culture
medium conditioned by human osteoblast-derived sarcoma
cells (OHS), and activated signalling pathways in the car-
cinoma cells were analyzed using microarrays with
tyrosine kinase substrates. Network interaction analysis of
substrates with significantly increased phosphorylation
levels revealed that signalling pathways mediated by
EGFR and ERBB2 were activated in LNCaP cells under
OHS influence but also by androgen treatment. Activation
of EGFR/ERBB2 signalling was also found in LNCaP cells
in cocultures with OHS cells or osteoblastic cells that had
been differentiated from human mesenchymal stem cells.
Our experimental data suggests osteoblast-directed induc-
tion of signalling activity via EGFR and ERBB2 in prostate
carcinoma cells and may provide a rationale for the use of
EGFR or ERBB2 inhibition in systemic prevention or
treatment of metastatic prostate cancer in the androgen-
sensitive stage of the disease.
Keywords Androgen EGFR ERBB2 Kinase array
Metastasis Osteoblast Prostate carcinoma
Abbreviations
FBS Fetal bovine serum
EGF Epidermal growth factor
Introduction
Metastatic prostate cancer is a leading cause of cancer
morbidity and mortality. The skeleton is the principal
organ for metastasis formation in prostate cancer, and bone
metastases are frequently painful and debilitating. Whilst
androgens are critical regulators of prostate carcinoma
growth and progression, most patients respond only
temporarily to androgen ablation therapy. Skeletal metas-
tases are predominantly osteoblastic, as apparent both
A
˚
. Bratland H. K. Høifødt M.-y. Wang Ø. Fodstad
A. H. Ree (&)
Department of Tumor Biology, Oslo University Hospital,
Montebello, 0310 Oslo, Norway
e-mail: a.h.ree@medisin.uio.no
A
˚
. Bratland W. Lilleby
Division of Cancer Medicine and Radiotherapy,
Oslo University Hospital, Oslo, Norway
P. J. Boender R. Ruijtenbeek
PamGene International BV, ‘s-Hertogenbosch, The Netherlands
I. H. G. Østensen
Norwegian Microarray Consortium, Oslo University Hospital,
Oslo, Norway
I. H. G. Østensen
Department of Molecular Biosciences, University of Oslo,
Oslo, Norway
J. P. Berg
Faculty Division Ulleva
˚
l University Hospital,
University of Oslo, Oslo, Norway
A
˚
. Bratland Ø. Fodstad
Faculty Division The Norwegian Radium Hospital,
University of Oslo, Oslo, Norway
A. H. Ree
Faculty Division Akershus University Hospital,
University of Oslo, Oslo, Norway
123
Clin Exp Metastasis (2009) 26:485–496
DOI 10.1007/s10585-009-9248-9
radiographically and histopathologically, and are associ-
ated with elevated serum level of bone-specific alkaline
phosphatase, a marker of osteoblast proliferation [1, 2].
These observations suggest that the biological interaction
between prostate carcinoma cells and osteoblasts contrib-
utes to the metastatic progression of prostate cancer.
In order to experimentally address how osteoblastic cells
may influence prostate carcinoma cell biology upon for-
mation of bone metastasis, the human, androgen-sensitive
prostate carcinoma cell line LNCaP [3] was treated with
culture medium conditioned by the human, osteoblast-
derived sarcoma cell line OHS [4], and subsequent multi-
plex profiling of LNCaP kinase activity was performed
using flow-through microarrays with peptide substrates
(Tyrosine Kinase PamChip
Ò
Arrays; PamGene Interna-
tional BV, ‘s-Hertogenbosch, The Netherlands), a novel
platform that allows rapid, real-time measurements of
phosphopeptide signatures generated by biological sam-
ples. As supplementary approaches, we used LNCaP cells
in cocultures with either OHS cells or with osteoblastic
cells that had been differentiated from human mesenchy-
mal stem cells.
Insight into regulatory mechanisms underlying the
establishment of prostate carcinoma cells within an osteo-
blastic microenvironment may eventually lead to more
effective therapies to prevent or treat metastatic disease.
Hence, from a therapeutic perspective, we compared the
intracellular LNCaP signalling pathways activated by
influence of osteoblastic cells with pathways induced by
androgen treatment, to identify signalling networks
potentially accessible for therapeutic targeting.
Materials and methods
Cell cultures
The LNCaP and OHS cell lines were routinely held in
RPMI 1640 medium supplemented with 10% fetal bovine
serum (FBS) and 2.0 mM glutamine, defined as growth
medium. Seventy-two hours before start of experimental
incubations, monocultures of LNCaP and OHS were see-
ded in a total number of 1.0 9 10
6
cells in 75 cm
2
cell
flasks in RPMI containing 2% charcoal-treated FBS and
glutamine. After 48 h, this medium was changed to RPMI
containing 0.5% charcoal-treated FBS and glutamine,
defined as experimental medium, for another 24 h before
experimental incubations were started (at time 0). All cell
cultures were invariably held in 10 ml medium throughout
different incubations.
At time 0 (start of the experimental incubations),
LNCaP cells were refed with experimental medium sup-
plemented with 100 nM of the synthetic androgen analog
R1881 (methyltrienolone; Biocompare, Inc., South San
Francisco, CA), giving rise to the LNCaP entity denoted
androgenic, or with medium conditioned by OHS cells.
The conditioned medium had been collected from OHS
monocultures that had been grown for 48 h (relative to
time 0) in experimental medium. This medium was sub-
sequently diluted 1:10 in either fresh experimental medium
or in medium obtained from standard LNCaP monocultures
after 48 h of incubation (relative to time 0) in experimental
medium, before the application onto monocultured LNCaP
cells, giving rise to the LNCaP entities denoted paracrine 1
and paracrine 2, respectively.
Cocultures of LNCaP/OHS were also seeded in a total
number of 1.0 9 10
6
cells in 75 cm
2
cell flasks in 10 ml
medium and were incubated identically to the monocul-
tured cell lines prior to and during experimental
incubations. Different ratios of LNCaP to OHS cells had
been tested in a series of cocultures to find the optimal
culturing conditions [5], and a 10:1 seeding ratio was
chosen.
Differentiated mesenchymal stem cells from human
bone marrow were prepared as previously described [6, 7].
Briefly, bone marrow (10–20 ml) was aspirated from the
posterior iliac crest of healthy, adult volunteers. Mononu-
clear cells were isolated by density gradient centrifugation
and resuspended in complete medium, consisting of MEMa
supplemented with 20% FBS and 2.0 mM glutamine. After
24 h, non-adherent cells were discarded, and the adherent
cells were thoroughly washed and further cultured in
complete medium for 7 days. Subsequently, the cells were
detached and replated in osteogenic differentiation med-
ium, consisting of MEMa with 10% FBS, 1.0 mM
glutamine, 10 nM dexamethasone, 10 lM b-glycerol
phosphate, and 200 lM ascorbic acid. This medium was
replaced every 3–4 days for about 20 days before LNCaP
cells (1.0 9 10
6
cells in 75 cm
2
cell flasks) were seeded on
top of the osteoblasts and the cocultures of LNCaP and
differentiated stem cells were incubated identically to the
other culture setups prior to and during experimental
incubations.
In the experimental setups, LNCaP cells were harvested
48 h after time 0, either directly from the various mono-
cultures or after immunomagnetic cell segregation of
cocultures (see below), and lysed in M-PER Mammalian
Extraction Reagent containing Halt Phosphatase Inhibitor
Cocktail and EDTA-free Halt Protease Inhibitor Cocktail
(Pierce Biotechnology, Inc., Rockford, IL). Reference
lysates were made from monocultured LNCaP grown for
48 h (relative to time 0) in experimental medium only.
In the experimental setup involving epidermal growth
factor (EGF; Sigma–Aldrich Norway AS, Oslo, Norway),
EGF was added to LNCaP cells in a final concentration of
50 ng/ml.
486 Clin Exp Metastasis (2009) 26:485–496
123
Kinase activity profiling
The kinase substrate array technology allows functional
comparison of biological samples without prior knowledge
of which activity pathways are influenced by the experi-
mental conditions. The high-throughput format of the
Tyrosine Kinase PamChip
Ò
Array is based on the use of a
porous, three-dimensional aluminum-oxide material as
solid support for the substrates. The sample lysates are
actively pumped through the interconnected capillary pores
of the arrays to allow contact with the reactive surface for
enzymatic reaction with the peptide substrates. The phos-
phorylation kinetics are therefore rapid and can be
completed within few minutes, allowing the generation of
spot images to be followed in real-time.
Each array contains 144 peptide substrates, and these
target peptides consist of 13 or 14 amino acids with sites
for phosphorylation, mainly tyrosine, representing 100
different proteins. Reaction mixtures consisted of Abl
Reaction Buffer (50 mM Tris–HCl pH 7.5, 10 mM MgCl
2
,
1 mM EGTA, 2 mM dithiothreitol, 0.01% Brij 35; New
England BioLabs, Inc., Ipswich, MA), 1 mg/ml bovine
serum albumin, 100 lM ATP, and 12.5 lg/ml of the
monoclonal, FITC-conjugated anti-phosphotyrosine anti-
body (Exalpha Biologicals, Inc., Maynard, MA), added to
each sample lysate, containing 2–5 lg total protein. The
arrays were blocked with 20 lg/ml bovine serum albumin
and subsequently washed twice with Abl Reaction Buffer
before the reaction mixtures were applied for initiation of
enzymatic reactions. Spot images were recorded after each
completed pumping cycle by a charge-coupled device
camera until the reactions were terminated.
Raw and processed data and investigation and array
designs can be obtained from the EMBL-EBI ArrayExpress
database (www.ebi.ac.uk) by accession number E-TABM-
626.
Microarray data adaptation and statistical analysis
Six independent LNCaP reference samples (denoted
LNCaP baseline), one of each LNCaP treated sample
(paracrine 1, paracrine 2, androgenic), one sample each of
LNCaP and OHS cells immunosegregated from coculture
(see below), and two independent samples of monocultured
OHS cells were analyzed, and each sample was measured
in three independent reactions. The image information was
converted using BioNavigator software (PamGene Inter-
national BV, www.pamgene.com). For each spot on the
array, signal intensity after background subtraction was
calculated and used for further analysis. Data normalization
of spot signal intensities and subsequent comparison
analyses were conducted using GeneSpring software
(Agilent Technologies, www.home.agilent.com). All values
were normalized to the calculated mean value of all sub-
strate phosphorylation intensities in the LNCaP baseline
sample. In the LNCaP treated samples, the criterion for
selecting a substrate with significantly increased phos-
phorylation level compared to the corresponding value in
the baseline sample was set at a statistical difference of
P \ 0.05 (Student’s t-test). Using this criterion, the fold-
increase (log
2
) of substrate phosphorylation levels was
within a range of 0.85 to [ 5.2.
Following the statistical analysis, two different pathway
visualization systems were used to create information about
pathway connectivity. Peptide names were generated from
corresponding gene name entries and protein sequences in
SwissProt (http://au.expasy.org/sprot) to enable unambigu-
ous annotation of the peptide sequences and phosphorylation
sites. It should be noted that, for some proteins, multiple
peptides have been derived and are present on the Tyrosine
Kinase PamChip
Ò
Array. For the pathway connectivity
analysis, one list of peptide identifications for each of the
treated samples was generated. First, those lists were loaded
into PathwayArchitect software (Stratagene Corp.,
www.stratagene.com; Strand Life Sciences Pvt. Ltd.,
www.avadis.strandgenomics.com), and the program was
used to generate a new list that contained all peptides in the
others, but where duplicates had been removed. The peptide
identifications in the final list were visualized through a
direct interaction network, defined to show all interactions
between peptides that were of the following types: binding,
expression, protein modification, and regulation. Some
peptides were omitted from the network because the inter-
actions were below maximum score/quality, which is
defined as an interaction from manually curated sources in
the database. The direct interaction network was exported,
all interactions were manually checked, and uninteresting
interactions were removed. These manual edits were incor-
porated into the interaction network. Subsequently, the
peptide list identified for each of the treated samples was
imported into PathwayStudio software (Ariadne Genomics,
www.ariadnegenomics.com), and pathways were created
directly from the ResNet database (Ariadne Genomics),
which comprises both curated and canonical pathways. The
peptide set was cross-checked with these pathways, and a
pathway list was generated and ranked by the hypergeo-
metric probability factor. All interactions were manually
inspected and selected on the criterion of the highest number
of proteins being involved in a linear pathway or sub-path-
way, by superposition of the peptide set to a cartoon
representation of the pathway reported.
Western blot analysis
Expression of phosphoproteins was measured by means of
standard western immunoblots. Primary, polyclonal
Clin Exp Metastasis (2009) 26:485–496 487
123
antibodies, selected against the same phosphorylated epi-
topes as the corresponding target peptides on the
microarrays, were anti-phospho(y1197)-EGFR (Abcam
Plc., Cambridge, UK) and anti-phospho(y1248)-Neu (Santa
Cruz Biotechnology, Inc., Santa Cruz, CA). Further, the
monoclonal anti-EGFR antibody 425.3 [8], a polyclonal
anti-ErbB2 antibody (R&D Systems Europe Ltd., Abing-
don, UK), and a monoclonal anti-a-tubulin antibody
(Calbiochem/Merck Biosciences Ltd., Nottingham, UK)
were used.
Immunomagnetic cell separation
MOC-31 (IQ Corporation BV, Groningen, The Nether-
lands) is an IgG1 class antibody that binds to the EPCAM
antigen, which is consistently expressed in most epithelial
cells [9]. The high-affinity monoclonal antibody 9.2.27,
which was originally developed against melanoma [10],
recognizes an epitope on the high molecular weight mel-
anoma-associated antigen and has also been shown to bind
some subgroups of sarcoma, including osteosarcoma [11,
12]. The antibodies were conjugated to superparamagnetic
particles coated with polyclonal sheep-antimouse IgG
particles (Dynabeads; Dynal A.S., Oslo, Norway).
At the end of experimental incubations, cocultures of
LNCaP cells with OHS cells and of LNCaP cells with
differentiated mesenchymal stem cells were detached, and
the resulting single cell suspensions were subjected to
immunomagnetic target cell isolation, essentially as pre-
viously described [1215]. The positive cell fractions were
examined by light microscopy for the principal presence of
cells with C5 immunobeads bound to their surface (bead
rosettes). For the purpose of cell quantification and quality
assurance, the immunosegregation procedure was repeated
with 9.2.27-coated beads added to the negative cell frac-
tions for isolation of OHS cells. The segregated cell
fractions from LNCaP/OHS coculture samples (10
6
–10
7
cells) contained 60–70% MOC-31-selected bead rosettes
and *30% 9.2.27-selected bead rosettes.
Results
Kinase activity evoked by osteoblastic and androgenic
influence
Monocultured LNCaP cells were treated with OHS-con-
ditioned medium to experimentally replicate the biological
context of paracrine osteoblastic influence. Moreover, to
simulate the complex processes involved in aberrant acti-
vation of the androgen signalling axis in prostate cancer
[16, 17], the experimental setup included LNCaP cells
treated with the synthetic androgen analog R1881 to
observe whether androgen receptor-mediated signalling
pathways might differ from pathways activated by OHS-
directed influence. Given the assumption that biologically
relevant signalling events implicated in the metastatic
phenotype require sustained activation, 48-h incubation
times were used for the experimental LNCaP contexts.
From the tyrosine kinase microarray analysis, the sub-
strate phosphorylation state generated by each LNCaP
entity (paracrine, androgenic) was calculated with respect
to baseline (untreated LNCaP cells). The identified sub-
strates showed increases in phosphorylation level within a
broad range (Table 1).
Each individual substrate phosphorylation signature was
considered to represent a subset of the information flow
through the globally activated signalling network of the
particular LNCaP entity. Tools for analyzing interconnec-
tivity of biological molecules have so far been used
primarily to explore system-level gene expression data.
However, such computational methods may also elucidate
how kinase activity information is directed, and by using
these algorithms, we assumed that phosphorylation events
that appeared simultaneously might be interlinked and
provide information about pathway connectivity [18, 19].
By applying these assumptions, the network interaction
analysis omitted phosphorylated substrates that did not
appear within any signalling pathway when defined by the
interaction types delineated in Materials and methods’’ .
As illustrated in Fig. 1, the resulting network connectivity
map indicated that signalling pathways involved in cell
adhesion and motility as well as cell proliferation were
activated in the paracrine LNCaP entity. Activation of
similar but also completely unrelated proliferation path-
ways was observed in the androgenic entity. Interestingly,
only the signalling pathway mediated by EGFR seemed to
be activated by the influence of both osteoblastic cells and
androgen treatment.
Since the technology for multiplex kinase activity pro-
filing and computational tools for data analysis are at an
early stage of development [20], no general consensus
approach to data validation exists. We therefore performed
conventional western immunoblotting for selected, indi-
vidual phosphoproteins expressed by LNCaP cells [21, 22]
that in addition are representative of signalling pathways
and therapeutic targets under investigation in recently
conducted trials of metastatic prostate cancer [2326].
Markedly increased expression levels of EGFR phosphor-
ylated on tyrosine 1197 and ERBB2 phosphorylated on
tyrosine 1248 were found in both the paracrine and
androgenic LNCaP entity (Fig. 2a). In contrast, EGFR was
found to be non-phosphorylated following 48 h of incu-
bation with either EGF or 10% FBS, whereas short-
time EGF treatment caused transient increase in EGFR
phosphorylation (Fig. 2b), which is in accordance with
488 Clin Exp Metastasis (2009) 26:485–496
123
previously published data [21, 27, 28] and may indicate
that functional signalling in prostate carcinoma cells under
influence of osteoblastic cells is diverse and complex [21].
EGFR/ERBB2 phosphorylation in supplementary
biological models
We used additional experimental setups as biological
controls for the osteoblastic influence on prostate carci-
noma cells; the first to model the direct interaction between
LNCaP and OHS cells and the second to provide cells that
might be more representative of physiological osteoblasts.
Culturing LNCaP cells with OHS cells caused sub-
stantial change in LNCaP morphology. The spindle-shaped
feature of monocultured LNCaP cells (Fig. 3a) was rapidly
lost upon direct contact with the OHS cells. In coculture,
both cell types appeared rounded, although cytoplasmic
processes were still apparent on LNCaP cells (Fig. 3b).
Cellular morphology of the OHS cells (Fig. 3c), however,
remained independent of the culturing conditions.
Non-hematopoietic stem cells in the bone marrow are
capable of differentiating into a variety of tissue entities,
including osteogenic cells of bone tissue [29]. Incubation
of mononuclear cells isolated from adult, human bone
marrow with mesenchymal stem cell-stimulating medium
followed by osteogenic differentiation medium [6, 7] gave
rise to cells with osteoblastic characteristics, for example
mineral deposition (Fig. 3d, e) and alkaline phosphatase-
secreting activity (not shown), to be used in coculture with
LNCaP cells (Fig. 3f).
Table 1 Peptide substrates with increased phosphorylation levels
generated by the paracrine and androgenic LNCaP entities
Peptide substrate Paracrine
a
Androgenic
EGFR_y1197
b
1.84
c
1.94
EGFR_y1110 2.34
RB1_s807/s811 3.39 3.07
ERBB2_y877 1.67
ERBB2_y1248 1.56
CDK2_t14/y15 1.86
EPHB1_y778 2.11
IRS2_y919 5.56
JAK1_y1022/y1023 2.05
LAT_y200 3.49
LCK_y394 1.91
MET_y1230/y1234/y1235 1.57
MST1R_y1353 2.86
MST1R_y1356/y1360 3.38
PDPK1_y9 1.82
PDPK1_y373/y376 1.82
PTK2_y576/y577 1.39
PTK2B_y579/y580 1.21
RASA1_y460 1.63
RET_y1029 1.76
ZAP70_y492/y493 1.93
CREB1_y134/s133 1.47
ERBB4_y1284 3.31
GSK3B_y216 1.15
RAF1_s337/s338/y339/y340 1.14
CHRNB1_y390 1.57 1.57
LTK_y772/y776/y777 1.20
DDR1_y792/y796/y797 1.20
MAPK10_t221/y223 1.32
MAPK12_t183/y185
PECAM1_y713 2.06
PRRX2_y214 1.48
ANXA1_y20/t23 2.41
CD79A_y182/y188 1.75
CTTN1_y477/y483 2.01
CTTN1_y499 2.13
ENO2_y43 1.85
EPHA2_y772 1.99
EPHA7_y608/y614 2.27
EPOR_y368 2.08
EPOR_y426 2.29
FER_y714 1.66
FES_y713 2.20
FGFR2_y769 1.77
FGFR3_y760 2.23
FRK_y387 2.02
LAT_y255 1.54
Table 1 continued
Peptide substrate Paracrine
a
Androgenic
NTRK2_y702/y706/y707 1.43
PDGFRB_y579/y581 3.87
PDGFRB_y716 1.78
PIK3R1_y607/s608 2.09
PXN_y31 1.88
PXN_y118 2.07
TEC_y519 1.68
PFKFB1_s33 1.36
PTPN11_y542 2.94
SYN1_s9 1.20
‘–’ denotes that the change in substrate phosphorylation level was not
found to be significant (P \ 0.05, Student’s t-test)
a
Phosphopeptide values from the LNCaP paracrine 2 entity are given
b
For each substrate, position of phosphorylation sites within the
protein is indicated
c
Fold changes (log
2
) relative to LNCaP baseline samples are listed
Clin Exp Metastasis (2009) 26:485–496 489
123
We applied the immunomagnetic cell separation method
for selective isolation of LNCaP cells from the cocultured
osteoblastic cells (Fig. 3g) and subjected the isolated
carcinoma cells to western immunoblotting. Increased
expression levels of EGFR phosphorylated on tyrosine
1197 and ERBB2 phosphorylated on tyrosine 1248 were
found in LNCaP cells from coculture with both OHS cells
and osteoblastic cells that had been differentiated from
human mesenchymal stem cells (Fig. 2a).
Supplementary kinase activity profiling
To assess robustness of the biological models in the ana-
lytical application of multiplex kinase activity profiling,
three experimental setups were exploited.
Kinase activity was compared in LNCaP cells treated
with medium conditioned by OHS cells only and with the
addition of medium obtained from standard LNCaP
monocultures (LNCaP entities denoted paracrine 1 and
paracrine 2, respectively). In this experimental setting, the
profile of phosphorylated substrates generated by the
LNCaP cells did not significantly change by the addition of
medium conditioned by LNCaP monocultures (compare
Table 1 with Table 2), strongly indicating that the signal-
ling pathways of biological importance in the paracrine
setting were activated by factors secreted by the OHS cells
exclusively.
Kinase activity induced in LNCaP cells by direct contact
with OHS cells was analyzed following carcinoma cell
isolation from cocultures. The substrate phosphorylation
state generated by such LNCaP cells overlapped the
phosphopeptide signatures identified in the paracrine
LNCaP entities (Table 2).
ERBB4
EGFR
MST1R
PTK2B
LAT
ZAP70
RASA1
PTK2
PDPK1
EPHB1
CDK2
MET
RET
JAK1
IRS2
LCK
ERBB2
CREB1
GSK3B
RB1
RAF1
Fig. 1 Interconnected
signalling pathways activated in
LNCaP cells by influence of
osteoblastic cells or androgen
treatment. Two pathway
visualization systems were
applied to the data set (Table 1),
which resulted in almost
identical network connectivity
maps. The substrate annotations
are derived from gene name
entries in SwissProt. The lines
connecting nodes represent
interactions of the following
types: binding, expression,
protein modification, and
regulation. Yellow and blue
nodes symbolize substrates
phosphorylated by the paracrine
and androgenic LNCaP entities,
respectively. (Color figure
online)
α-tubulin
EGFR
p-EGFR
(170kDa)
b
a
12345
123456
α-tubulin
ERBB2
EGFR
p-ERBB2
(185 kDa)
p-EGFR
(170 kDa)
Fig. 2 Expression of phospho(y1197)-EGFR and phospho(y1248)-
ERBB2 in LNCaP cells. a Cells were harvested after 48 h of
experimental treatments. Lanes: (1) baseline, (2) paracrine entity, (3)
androgenic entity, (4) cells immunosegregated from coculture with
OHS cells, and (5) cells immunosegregated from coculture with
osteoblastic cells differentiated from human mesenchymal stem cells.
b Cells were treated with 50 ng/ml EGF or 10% FBS. Lanes: (1)
baseline, (2) EGF for 15 min, (3) EGF for 24 h, (4) EGF for 48 h, (5)
FBS for 48 h, and (6) 48 h untreated
490 Clin Exp Metastasis (2009) 26:485–496
123
We have previously evaluated the immunomagnetic cell
separation method for selective isolation of target cells and
demonstrated that target cell populations are highly enri-
ched [1215]. But because kinase activity profiling might
be sensitive to the possible presence of contaminants,
we screened for phosphopeptide signatures generated by
immunoselected as well as monocultured OHS cells. Nota-
bly, the resulting substrate phosphorylation patterns were
closely similar for the two conditions (Table 3) and also
clearly distinguishable from those generated by the LNCaP
entities, which argues against significant contamination of
OHS cells in MOC-31-positive LNCaP cell isolates.
Fig. 3 Morphologic characteristics of LNCaP and osteoblastic cells.
a Monocultured LNCaP cells, untreated (magnification 9300). b
LNCaP/OHS coculture, at start of experimental incubation (magni-
fication 9400). The fluorescent cells are stained with FITC-
conjugated 9.2.27 antibody; accordingly, representing OHS cells. c
Monocultured OHS cells (magnification 9300). d Mononuclear cells
isolated from adult human bone marrow were cultured in mesencymal
stem cell-stimulating medium followed by osteogenic differentiation
medium for 20 days to obtain in vitro-differentiated osteoblasts
(magnification 9300). e The differentiated osteoblasts were fixed in
ice-cold methanol for 30 min, incubated in dark with 5% silver nitrate
solution for 30 min, and washed thoroughly with deionized water and
then with 5% Na
2
CO
3
and 0.2% formaldehyde, according to the von
Kossa staining protocol for calcium salts. Mineral deposits are
visualized as dark brown stain (magnification 9300). f Coculture of
LNCaP cells and in vitro-differentiated osteoblasts, at start of
experimental incubation (magnification 9400). The fluorescent cells
are stained with FITC-conjugated MOC-31 antibody; accordingly,
representing LNCaP cells. g Suspended cells binding MOC-31-coated
beads; accordingly, representing LNCaP cells, after immunomagnetic
selection from LNCaP/OHS coculture (magnification 9400) (Color
figure online)
Clin Exp Metastasis (2009) 26:485–496 491
123
Discussion
Given the considerable health care challenges generated by
the prevalence of metastatic bone disease in prostate can-
cer, the apparent shortage of adequate experimental models
to study this aspect of disease progression is striking. From
a clinical point of view, prostate cancer metastasis to bone
is a lengthy and complex disease process, which makes it
difficult to establish adequate experimental models to
recreate all steps involved. As highlighted in a compre-
hensive review [30], most of the experimental systems
examining this phenomenon are based on rodent models.
Importantly, the model systems used in this study exclu-
sively utilize cell types of human origin, but caution should
be exercised regarding any compelling conclusion based on
specific in vitro experimental conditions, which can not
reflect an intact microenvironmental in vivo setting. Nev-
ertheless, given that regulatory mechanisms implicated in
the metastatic phenotype are evoked when carcinoma cells
settle within an osteoblastic microenvironment, the com-
bination of our experimental models and analytical
technology may provide relevant information about func-
tional signalling networks that facilitate this biological
process, and in addition may enable the identification of
new targets for therapeutic intervention.
The OHS cell line was originally established from a
patient with aggressive osteosarcoma [4], and it might be
argued that it is not representative for physiological
osteoblasts. However, formation of osteosclerotic (i.e.,
osteoblastic) lesions following intratibial OHS cell inocu-
lation has previously been demonstrated by radiographic,
scintigraphic, and morphologic assessments [31].
Table 2 Peptide substrates with increased phosphorylation levels
generated by the LNCaP paracrine 1 entity and LNCaP cells immu-
nosegregated from coculture with OHS cells (co-LNCaP)
Peptide substrate Paracrine 1 Co-LNCaP
EGFR_y1197
a
1.50
b
2.16
EGFR_y1110 1.86
RB1_s807/s811 3.72
ERBB2_y877 1.06
ERBB2_y1248
CDK2_t14/y15 1.47
EPHB1_y778 1.78
IRS2_y919 4.88
JAK1_y1022/y1023 1.81
LAT_y200 2.70 2.94
LCK_y394 1.38
MET_y1230/y1234/y1235 1.08
MST1R_y1353 2.29 2.20
MST1R_y1356/y1360 2.60 2.01
PDPK1_y9 1.50
PDPK1_y373/y376 1.50
PTK2_y576/y577 1.33 1.03
PTK2B_y579/y580 0.85 0.85
RASA1_y460 1.27 0.96
RET_y1029 1.24
ZAP70_y492/y493 1.68 1.21
CREB1_y134/s133
ERBB4_y1284
GSK3B_y216
RAF1_s337/s338/y339/y340
CHRNB1_y390 1.00 0.95
LTK_y772/y776/y777 0.96
DDR1_y792/y796/y797 1.11 0.91
MAPK10_t221/y223 1.28 0.98
MAPK12_t183/y185 2.23
PECAM1_y713 1.51 0.87
PRRX2_y214 1.23 0.93
ANXA1_y20/t23 1.93
CD79A_y182/y188 1.39
CTTN1_y477/y483 1.45
CTTN1_y499 1.64
ENO2_y43 1.43
EPHA2_y772 1.60
EPHA7_y608/y614 1.64
EPOR_y368 1.67
EPOR_y426 1.77
FER_y714 1.36
FES_y713 1.82
FGFR2_y769 1.28
FGFR3_y760 1.63
FRK_y387 1.47
LAT_y255 1.13
Table 2 continued
Peptide substrate Paracrine 1 Co-LNCaP
NTRK2_y702/y706/y707 1.08
PDGFRB_y579/y581 3.52
PDGFRB_y716 1.59
PIK3R1_y607/s608 1.59
PXN_y31 1.44
PXN_y118 1.61
TEC_y519 1.23
PFKFB1_s33
PTPN11_y542
SYN1_s9
‘–’ denotes that the change in substrate phosphorylation level was not
found to be significant (P \ 0.05, Student’s t-test)
a
For each substrate, position of phosphorylation sites within the
protein is indicated
b
Fold changes (log
2
) relative to LNCaP baseline samples are listed
492 Clin Exp Metastasis (2009) 26:485–496
123
Kinase activity microarrays represent an emerging
technology that may become a powerful tool for signal
transduction profiling of biological samples. A key
advantage of this technology lies in its ability to provide
signalling pathway maps that indicate the state of infor-
mation flow through intracellular networks. Essentially, the
peptide substrate array technology determines kinase
activity profiles, which is different from but complemen-
tary to mass spectrometry, which identifies phosphorylated
proteins that represent the end products of kinase activity.
An additional advantage of kinase activity microarrays is
their robustness with small sample quantities, typically 2–
5 lg total protein being sufficient for Tyrosine Kinase
PamChip
Ò
Array analysis, in contrast to several milligrams
of total protein usually required for mass spectrometry
analysis.
Of note, the phosphopeptide signatures generated by the
LNCaP paracrine 1 and paracrine 2 entities and by OHS
cells cultured alone or with LNCaP cells, respectively, were
essentially identical. This indicates low variation between
biologically similar samples and a high degree of repro-
ducibility using this peptide substrate array technology.
Computational methods for mapping of phosphorylation
networks are being developed, and reconstruction of
EGFR-mediated pathways have been used to test applica-
bility of such models [32]. Correct network definition may
be restricted by the influence of contextual factors, such as
subcellular compartmentalization or temporal expression
of the proteins involved [33]. Therefore, to predict pathway
connectivity with sufficient accuracy, the use of more than
one computational technique on the dataset of interest has
been recommended [20]. In this study, two different
pathway visualization systems identified signalling net-
works that were essentially identical.
The signalling pathway mediated by EGFR was found to
be activated in androgen-sensitive LNCaP cells under the
influence of either osteoblastic cells or androgen treatment.
Table 3 Phosphopeptides generated by monocultured OHS cells
(mono-OHS) and OHS cells immunosegregated from coculture with
LNCaP cells (co-OHS)
Peptide substrates Mono-OHS Co-OHS
ANXA1_y20/t23
a
-1.56
b
-2.84
CD79A_y182/y188 -3.70 -4.54
CDK2_t14/y15 -0.94 -1.32
CHRNB1_y390 -0.49 -0.64
CREB1_y134/s133 -0.81 -1.29
CTTN1_y477/y483 -0.86 -1.43
CTTN1_y499 -1.56 -2.00
DCX_y112/s116 -0.92 -0.86
DDR1_y792/y796/y797 -0.40 -0.79
EFS_y253 -0.86 -1.18
ENO2_y43 -3.72 -4.76
EPHA2_y772 -1.43 -2.32
EPHA7_y608/y614 -1.60 -2.18
EPHB1_y778 -1.60 -2.40
EPOR_y368 -0.94 -1.40
EPOR_y426 -0.94 -2.32
ERBB2_y877 -1.74 -2.18
ERBB2_y1248 -1.22 -2.06
FER_y714 -1.15 -1.64
FES_y713 -1.03 -2.25
FGFR2_y769 -1.89 -
2.06
FGFR3_y760 -1.56 -0.89
FRK_y387 -1.22 -1.84
GSK3B_y216 -1.25 -0.97
JAK1_y1022/y1023 -2.18 -2.32
KRT6E_s59 -0.92 -1.47
LAT_y255 -1.29 -2.40
LCK_y394 -1.60 -2.25
LTK_y772/y776/y777 -0.86 -1.03
MAPK10_t221/y223 -0.94 -0.62
MET_y1230/y1234/y1235 -0.89 -1.12
NCF1_s315/s320 -0.67 -0.74
NTRK1_y496 -1.94 -2.84
NTRK2_y702/y706/y707 -2.94 -3.84
PDGFRB_y579/y581 -4.32 -3.84
PDGFRB_y716 -1.51 -1.00
PDGFRB_y771/y775/y778 -1.00 -1.25
PDPK1_y9 -1.40 -1.25
PDPK1_y373/y376 -1.36 -2.00
PECAM1_y713 -1.06 -1.47
PFKFB1_s33 -0.79 -0.92
PIK3R1_y607/s608 -1.25 -2.56
PLCG1_y771 -4.57 -4.64
PRRX2_y214 -1.36 -1.25
PTK2_y576/y577 -1.15 -0.81
PTK2B_y579/y580 -0.71 -1.51
PXN_y31 -1.25 -1.84
Table 3 continued
Peptide substrates Mono-OHS Co-OHS
PXN_y118 -1.03 -1.29
RAF1_s337/s338/y339/y340 -0.89 -1.18
RASA1_y460 -1.36 -1.09
RET_y1029 -1.22 -2.18
SYN1_s9 -1.06 -1.40
TEC_y519 -1.51 -1.64
TYRO3_y686 -1.40 -1.29
ZAP70_y492/y493 -1.56 -1.74
a
For each substrate, position of phosphorylation sites within the
protein is indicated
b
Fold changes (log
2
) relative to LNCaP baseline sample are listed
Clin Exp Metastasis (2009) 26:485–496 493
123
Analysis by conventional western immunoblotting also
revealed that ERBB2-dependent signalling was activated in
both the paracrine and androgenic LNCaP entities,
although the latter did not seem to generate this particular
substrate phosphorylation profile on the microarray analy-
sis. The kinase activity and protein expression data together
strongly indicates a central involvement of both signalling
pathways in the interaction between prostate carcinoma
cells and osteoblasts.
Additionally, given that EGFR and ERBB2 are phos-
phorylated in androgen-sensitive LNCaP cells upon
influence of osteoblasts, a functional androgen signalling
axis [16, 17] may appear to be permissive for activity of
these particular pathways in prostate cancer. Not the less,
EGFR phosphorylation resulting from short-term incuba-
tion with bone stromal conditioned medium (containing 6%
FBS) and suppression of in vivo bone metastasis formation
following targeted inhibition of EGFR-dependent signalling
have been demonstrated in androgen-independent prostate
carcinoma PC3 cells [34]. Separately, EGFR and ERBB2
may facilitate androgen receptor-driven activity in prostate
cancer at the level of target gene transcription in the absence
or at low concentrations of androgens [35, 36]. Yet,
androgen receptor pathway genes, identified by system-
level analysis of gene expression in primary tumor speci-
mens from therapy-naı
¨
ve prostate cancer patients, were
reported to be down-regulated, with a few exceptions, in
lymph-node metastases from the patients [37]. This finding
further supports the assumption that the regulatory control
by the androgen receptor on carcinoma cell biology is lost in
the process of prostate cancer metastasis, even in the pres-
ence of activating receptor ligands.
Of importance, our experimental data suggests that tar-
geted inhibition of the signalling pathways directed by
EGFR or ERBB2 may simultaneously ablate androgen-
driven proliferation of prostate carcinoma cells and the
survival responses within an osteoblastic microenviron-
ment. It equally provides a biological rationale for the use
of EGFR or ERBB2 inhibition in systemic prevention or
treatment of metastatic prostate cancer in the androgen-
sensitive stage of the disease. Intriguingly, the therapeutic
concept of EGFR or ERBB2 inhibition in hormone-
refractory prostate cancer has recently been evaluated;
however, in initial studies addressing the use of single-
agent therapies in patients with androgen-resistant disease,
neither receptor-blocking antibodies nor small-molecular
tyrosine kinase inhibitors showed clinically significant
activity [2326].
Of note, the tumor suppressor PTEN phosphatase is
frequently found to be functionally inactivated in prostate
cancer, leading to increased activity of AKT kinase sig-
nalling independent of the up-stream EGFR/ERBB2 and to
insensitivity to EGFR/ERBB2 inhibitors [16, 3840]. The
LNCaP cells are deficient in PTEN [41, 42], questioning the
relevance of extrapolating from our data that EGFR/ERBB2
signalling is involved in the maintenance of osteoblast-
induced survival responses in prostate cancer. On the other
hand, development of androgen independence and more
advanced disease is associated with increased PTEN loss
[4345], favoring therapeutic EGFR/ERBB2 inhibition in
the initial, androgen-sensitive stage of the disease. Thus, if
exploitable in patients with therapy-naı
¨
ve prostate cancer,
inhibitory EGFR/ERBB2 targeting might be incorporated
into treatment schedules with a potential reduction in the
alternative requirement of long-term androgen depletion, a
reduction in related side effects, and, intriguingly, the
potential for an improvement in patient survival.
Acknowledgments We thank Dr. R. Reisfeld for the gift of the
425.3 and 9.2.27 antibodies. We are grateful to Ingrid J. Guldvik and
Heidi Rasmussen for technical assistance. This study was supported
by The Norwegian Cancer Society grant C-04083 and grants from
Faculty Division The Norwegian Radium Hospital (to A. H. Ree).
Bioinformatics service was provided by the Norwegian Microarray
Consortium at the national technology platform (http://microarray.no
), supported by the Functional Genomics Program of the Norwegian
Research Council.
Conflict of interest statements A. H. Ree, A
˚
. Bratland, P. J.
Boender, and R. Ruijtenbeek are owners of a patent for diagnostic
application of the Tyrosine Kinase PamChip
Ò
Array technology in
prostate cancer (WIPO no. WO/2008/125633). P. J. Boender and R.
Ruijtenbeek are employees of PamGene International BV.
Open Access This article is distributed under the terms of the
Creative Commons Attribution Noncommercial License which per-
mits any noncommercial use, distribution, and reproduction in any
medium, provided the original author(s) and source are credited.
References
1. Roodman GD (2004) Mechanisms of bone metastasis. N Engl J
Med 350:1655–1664. doi:10.1056/NEJMra030831
2. Logothetis CJ, Lin SH (2005) Osteoblasts in prostate cancer
metastasis to bone. Nat Rev Cancer 5:21–28. doi:10.1038/nrc1528
3. Horoszewicz JS, Leong SS, Kawinski E et al (1983) LNCaP
model of human prostatic carcinoma. Cancer Res 43:1809–1818
4. Fodstad Ø, Brøgger A, Bruland Ø et al (1986) Characteristics of a
cell line established from a patient with multiple osteosarcoma,
appearing 13 years after treatment for bilateral retinoblastoma.
Int J Cancer 38:33–40. doi:10.1002/ijc.2910380107
5. Bratland A
˚
, Ragnhildstveit E, Bjørnland K et al (2003) The
metalloproteinase inhibitor TIMP-2 is down-regulated by
androgens in LNCaP prostate carcinoma cells. Clin Exp Metas-
tasis 20:541–547. doi:10.1023/A:1025860214891
6. Colter DC, Class R, DiGirolamo CM et al (2000) Rapid expan-
sion of recycling stem cells in cultures of plastic-adherent cells
from human bone marrow. Proc Natl Acad Sci USA 97:3213–
3218. doi:10.1073/pnas.070034097
7. Peister A, Mellad JA, Wang M et al (2004) Stable transfection of
MSCs by electroporation. Gene Ther 11:224–228. doi:10.1038/
sj.gt.3302163
494 Clin Exp Metastasis (2009) 26:485–496
123
8. Mueller BM, Romerdahl CA, Trent JM et al (1991) Suppression
of spontaneous melanoma metastasis in scid mice with an anti-
body to the epidermal growth factor receptor. Cancer Res 51:
2193–2198
9. de Jonge MW, Kosterink JG, Bin YY et al (1993) Radioim-
munodetection of human small cells lung carcinoma xenografts
in the nude rat using
111
in-labelled monoclonal antibody MOC-
31. Eur J Cancer 29A:1885–1890. doi:10.1016/0959-8049(93)
90543-O
10. Morgan AC Jr, Galloway DR, Reisfeld RA (1981) Production and
characterization of a monoclonal antibody to a melanoma specific
glycoprotein. Hybridoma 1:27–36
11. Godal A, Bruland OS, Haug E et al (1986) Unexpected expres-
sion of the 250 kD melanoma-associated antigen in human
sarcoma cells. Br J Cancer 53:839–841
12. Bruland OS, Hoifodt H, Saeter G et al (2005) Hematogenous
micrometastases in osteosarcoma patients. Clin Cancer Res
11:4666–4673. doi:10.1158/1078-0432.CCR-05-0165
13. Forus A, Høifødt HK, Øverli GET et al (1999) Sensitive fluo-
rescent in situ hybridisation method for the characterisation of
breast cancer cells in bone marrow aspirates. J Clin Pathol Mol
Pathol 52:68–74
14. Fodstad Ø, Faye R, Høifødt HK et al (2001) Immunobead-based
detection and characterization of circulating tumor cells in mel-
anoma patients. Recent Results in Cancer Res 158:40–50
15. Tveito S, Maelandsmo GM, Hoifodt HK et al (2007) Specific iso-
lation of disseminated cancer cells: a new method permitting
sensitive detection of target molecules of diagnostic and therapeutic
value. Clin Exp Metastasis 24:317–327. doi:10.1007/s10585-
006-9052-8
16. Scher HI, Sawyers CL (2005) Biology of progressive, castration-
resistant prostate cancer: directed therapies targeting the andro-
gen-receptor signaling axis. J Clin Oncol 32:8253–8261. doi:
10.1200/JCO.2005.03.4777
17. Attard G, Sarker D, Reid A et al (2006) Improving the outcome of
patients with castration-resistant prostate cancer through rational
drug development. Br J Cancer 95:767–774. doi:10.1038/sj.bjc.
6603223
18. Petricoin EFIII, Bichsel VE, Calvert VS et al (2005) Mapping
molecular networks using proteomics: a vision for patient-tai-
lored combination therapy. J Clin Oncol 23:3614–3621. doi:
10.1200/JCO.2005.02.509
19. Sevecka M, MacBeath G (2006) State-based discovery: a multi-
dimensional screen for small-molecule modulators of EGF
signaling. Nat Methods 3:825–831. doi:10.1038/nmeth931
20. Janes KA, Lauffenburger DA (2006) A biological approach to
computational models of proteomic networks. Curr Opin Chem
Biol 10:73–80. doi:10.1016/j.cbpa.2005.12.016
21. Angelucci A, Festuccia C, Gravina GL et al (2004) Osteopontin
enhances the cell proliferation induced by the epidermal growth
factor in human prostate cancer cells. Prostate 59:157–166. doi:
10.1002/pros.20008
22. Berger R, Lin DI, Nieto M et al (2006) Androgen-dependent
regulation of Her-2/neu in prostate cancer cells. Cancer Res
66:5723–5728. doi:10.1158/0008-5472.CAN-05-3928
23. de Bono JS, Bellmunt J, Attard G et al (2007) Open-label phase II
study evaluating the efficacy and safety of two doses of pert-
uzumab in castrate chemotherapy-naı
¨
ve patients with hormone-
refractory prostate cancer. J Clin Oncol 25:257–262. doi:10.1200/
JCO.2006.07.0888
24. Agus DB, Sweeney CJ, Morris MJ et al (2007) Efficacy and safety
of single-agent pertuzumab (rhuMAb 2C4), a human epidermal
growth factor dimerization inhibitor, in castration-resistant pros-
tate cancer after progression from taxane-based therapy. J Clin
Oncol 25:675–681. doi:10.1200/JCO.2006.07.0649
25. Canil CM, Moore MJ, Winquist E et al (2005) Randomized phase
II study of two doses of gefitinib in hormone-refractory prostate
cancer: a trial of the National Cancer Institute of Canada-Clinical
Trials Group. J Clin Oncol 23:455–460. doi:10.1200/JCO.2005.
02.129
26. Ziada A, Barqawi A, Glode LM et al (2004) The use of trast-
uzumab in the treatment of hormone refractory prostate cancer;
phase II trial. Prostate 60:332–337. doi:10.1002/pros.20065
27. El Sheikh SS, Domin J, Abel P et al (2004) Phosphorylation of
both EGFR and ErbB2 is a reliable predictor of prostate cancer
cell proliferation in response to EGF. Neoplasia (New York, NY)
6:846–853. doi:10.1593/neo.04379
28. Martı
´
n-Orozco RM, Almaraz-Pro C, Rodrı
´
guez-Ubreva FJ et al
(2007) EGF prevents the neuroendocrine differentiation of
LNCaP cells induced by serum deprivation: the modulator role of
PI3 K/Akt. Neoplasia (New York, NY) 9:614–624. doi:10.1593/
neo.07337
29. Giordano A, Galderisi U, Marino IR (2007) From the laboratory
bench to the patient’s bedside: an update on clinical trials with
mesenchymal stem cells. J Cell Physiol 211:27–35. doi:10.1002/
jcp.20959
30. Singh AS, Figg WD (2005) In vivo models of prostate cancer
metastasis to bone. J Urol 174:820–826. doi:10.1097/01.ju.0000
169133.82167.aa
31. Kjønniksen I, Winderen M, Bruland Ø et al (1994) Validity and
usefulness of human tumor models established by intratibial cell
inoculation in nude rats. Cancer Res 54:1715–1719
32. Oda K, Matsuoka Y, Funahashi A et al. (2005) A comprehensive
pathway map of epidermal growth factor receptor signaling. Mol
Syst Biol 1:2005.0010
33. Linding R, Jensen LJ, Ostheimer GJ et al (2007) Systematic
discovery of in vivo phosphorylation networks. Cell 129:1415–
1426. doi:10.1016/j.cell.2007.05.052
34. Angelucci A, Gravina GL, Rucci N et al (2006) Suppression of
EGF-R signaling reduces the incidence of prostate cancer
metastasis in nude mice. Endocr Relat Cancer 13:197–210. doi:
10.1677/erc.1.01100
35. Gregory CW, Fei X, Ponguta LA et al (2004) Epidermal growth
factor increases coactivation of the androgen receptor in recurrent
prostate cancer. J Biol Chem 279:7119–7130. doi:10.1074/jbc.
M307649200
36. Mellinghoff IK, Vivanco I, Kwon A et al (2004) HER2/neu
kinase-dependent modulation of androgen receptor function
through effects on DNA binding and stability. Cancer Cell 6:517–
527. doi:10.1016/j.ccr.2004.09.031
37. Hendriksen PJ, Dits NF, Kokame K et al (2006) Evolution of the
androgen receptor pathway during progression of prostate cancer.
Cancer Res 66:5012–5020. doi:10.1158/0008-5472.CAN-05-3082
38. Feldman BJ, Feldman D (2001) The development of androgen-
independent prostate cancer. Nat Rev Cancer 1:34–45. doi:
10.1038/35094009
39. Bianco R, Shin I, Ritter CA et al (2003) Loss of PTEN/MMAC1/
TEP in EGF receptor-expressing tumor cells counteracts the
antitumor action of EGFR tyrosine kinase inhibitors. Oncogene
22:2812–2822. doi:10.1038/sj.onc.1206388
40. Wu Z, Gioeli D, Conaway M et al (2008) Restoration of PTEN
expression alters the sensitivity of prostate cancer cells to EGFR
inhibitors. Prostate 68:935–944. doi:10.1002/pros.20745
41. Li J, Yen C, Liaw D et al (1997) PTEN, a putative protein tyrosine
phosphatase gene mutated in human brain, breast, and prostate
cancer. Science 275:1943–1947. doi:10.1126/science.275.5308.
1943
42. Vlietstra RJ, van Alewijk DC, Hermans KG et al (1998) Frequent
inactivation of PTEN in prostate cancer cell lines and xenografts.
Cancer Res 58:2720–2723
Clin Exp Metastasis (2009) 26:485–496 495
123
43. Rubin MA, Gerstein A, Reid K et al (2000) 10q23.3 loss of
heterozygosity is higher in lymph node-positive (pT2-3,N ?)
versus lymph node-negative (pT2-3,N0) prostate cancer. Hum
Pathol 31:504–508. doi:10.1053/hp.2000.6713
44. Jiao J, Wang S, Qiao R et al (2007) Murine cell lines derived
from Pten null prostate cancer show the critical role of PTEN in
hormone refractory prostate cancer development. Cancer Res
67:6083–6091. doi:10.1158/0008-5472.CAN-06-4202
45. McCall P, Witton CJ, Grimsley S et al (2008) Is PTEN loss
associated with clinical outcome measures in human prostate
cancer? Br J Cancer 99:1296–1301. doi:10.1038/sj.bjc.6604680
496 Clin Exp Metastasis (2009) 26:485–496
123
... Apart from that, the role of EGFR/Erb-B2 receptor tyrosine kinase 2 (ERBB2) signaling in prostate cancer metastasis into the bone microenvironment has also been enumerated [55]. The study suggested that osteoblast-directed induction of signaling activity involving EGFR and ERBB2 in prostate carcinoma cells might be culpable in bone metastasis. ...
... EGFR and ERBB2 activation in LNCaP cells under the influence of osteoblast-derived sarcoma cells (OHS) has been reported to activate EGFR/ERBB2 signaling pathways in LNCaP cells co-cultured with osteoblastic cells that had been differentiated from human mesenchymal stem cells or OHS cells. This finding supported the rationale for the use of EGFR or ERBB2 inhibitors for prophylaxis or cure of prostate cancer metastasis in the androgen-sensitive stage [55]. ...
Article
Full-text available
Patients with advanced prostate cancer often develop bone metastases, leading to bone pain, skeletal fracture, and increased mortality. Bone provides a hospitable microenvironment to tumor cells. The disease manifestation is driven by the interaction between invading tumor cells, bone-forming osteoblasts, and bone-resorbing osteoclasts. The increased level of osteoclast-activating factor (parathyroid hormone-related peptide, PTHrP) is believed to induce bone resorption by upregulating receptor activator of nuclear factor-kappa B ligand (RANKL) and the release of various growth factors into the bone microenvironment to enhance cancer cell growth. However, the underlying molecular mechanisms remain poorly understood. This review outlines the possible molecular mechanisms involved in governing bone metastases driven by prostate cancer, which further provide the basis in searching for new molecular targets for the development of potential therapy.
... Further, EGFR/ERBB2 kinase activity was revealed to be significantly up-regulated in LNCaP cells co-cultured with osteoblastic cells as determined by multiplex kinase activity profiling. This study hints that EGFR activity is stimulated by tumor-associated bone cells (Bratland et al. 2009). Activation of EGFR is also mediated by type 1 insulin-like growth factor (IGF) and extracellular matrixes, which are produced by the tumor-associated microenvironment during PCa metastases in the bone marrow (Chott et al. 1999). ...
... It has been revealed that EGFR-mediated activation of AKT occurs in part through dimerization of EGFR with HER3 or alternatively, through enhanced HER3 activity and in part via interaction of EGFR with the intracellular adaptor protein (Craft et al. 1999;Di Lorenzo et al. 2002;Turke et al. 2012). Simultaneous occurrence of EGFR and phosphatase and tensin homolog (PTEN) alterations as well as an interplay between these two factors can be observed in various cancers such as cancers of the brain, lung and prostate (Bratland et al. 2009;Chott et al. 1999;Wozniak et al. 2017). Our data provides evidence suggesting that AR is functionally linked to EGFR and its associated AKT pathways. ...
Article
Full-text available
Background Metastatic Prostate cancer (PCa) cells have gained survival and invasive advantages. Epidermal growth factor (EGF) receptor is a receptor tyrosine kinase, which may mediate signalling to promote progression and invasion of various cancers. In this study, we uncovered the molecular mechanisms underlying the interconnection among the androgen receptor (AR), matrix metalloproteinase-9 (MMP9) and EGFR in promoting PCa progression. Methods Immunohistochemical analysis of the tissue microarrays consisting of primary and metastatic PCa tissues was performed. The clinical importance of EGFR and its association with survivals were analyzed using three cohorts from MSKCC Prostate Oncogenome Project dataset (For primary tumors, n = 181; for metastatic tumors n = 37) and The Cancer Genome Atlas Prostate Adenocarcinoma Provisional dataset (n = 495). Targeted overexpression or inhibition of the proteins of interests was introduced into PCa cell lines. Treatment of PCa cell lines with the compounds was conducted. Immunoblot analysis was performed. ResultsWe showed that AR, MMP-9 and EGFR are interconnect factors, which may cooperatively promote PCa progression. Altered EGFR expression was associated with poor disease-free survival in PCa patients. Induced overexpression of AR led to an increase in the expression of EGFR, p-GSK-3β and decrease in p27 expression in PCa cell lines in the presence of androgen stimulation. Overexpression of MMP9 significantly induced EGFR expression in PCa cells. Inhibition of PIP5K1α, a lipid kinase that acts upstream of PI3K/AKT greatly reduced expressions of AR, MMP-9 and EGFR. Conclusions Our findings also suggest that PCa cells may utilize AR, EGFR and MMP-9 pathways in androgen-dependent as well as in castration-resistant conditions. Our data suggest a new therapeutic potential to block cancer metastasis by targeting AR, EGFR and MMP-9 pathways in subsets of PCa patients.
... A previous study indicated that FGF signalling could control many aspects of osteoblast differentiation through induction of Sox2 and regulation of the Wnt-beta-catenin pathway (Mansukhani et al., 2005). Moreover, focal adhesion, the PI3K-Akt signalling pathway, EGFR tyrosine kinase inhibitor resistance, the TNF signalling pathway, the MAPK signalling pathway, the Jak-STAT signalling pathway, and the calcium signalling pathway might have important roles in the differential regulation of MC3T3-E1 cells after microgravity stimulation (Tamura et al., 2001;Bratland et al., 2009;Lee et al., 2010;Xiao et al., 2000;Wu et al., 2019;Mccann et al., 1997). The results also predicted the existence of signal transduction crosstalk in osteogenic differentiation of MC3T3-E1 cells after microgravity stimulation; for example, downregulated FGFR 2 is involved in the PI3K-Akt signalling pathway, the FGFR/Erk signalling pathway and the Jak-STAT signalling pathway (Nakano et al., 2015;Maehara et al., 2017;Li et al., 2019). ...
Article
Background Circular RNAs (circRNAs) are a new class of non-coding RNA with a stable structure formed by special loop splicing. Research increasingly suggests that circRNAs play a vital role in the pathogenesis and progression of various diseases. However, the roles of circRNAs in osteoblast differentiation under microgravity remain largely unknown. Here, we investigated the roles and mechanobiological response of circRNAs in osteoblasts under simulated microgravity Methods Differential circRNA and mRNA expression profiles of MC3T3-E1 cells during exposure to microgravity were screened by RNA transcriptome sequencing technology (RNA-seq). The selected RNAs were validated using quantitative real-time polymerase chain reaction (qRT-PCR). Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) were applied for gene function analyses. Results A total of 427 circRNAs and 1912 mRNAs were differentially expressed along with osteogenic differentiation in the simulated microgravity group (SMG) compared to the control group (CON). Of these, 232 circRNAs and 991 mRNAs were upregulated, whereas 95 circRNAs and 921 mRNAs were downregulated (fold change ≥ 2, p < 0.05). The results showed that the parental genes of circRNAs and mRNAs were mainly enriched in anatomical structure morphogenesis, anchoring junction and protein binding. KEGG analysis results showed that the differentially expressed mRNAs were enriched in the regulation of the actin cytoskeleton, focal adhesion, and Ras signalling pathway. Subsequently, 9 core regulatory genes, including 6 mRNAs and 3 circRNAs, were identified based on their possible function in osteoblast differentiation. Based on this analysis, circ_014154 was selected as the target circRNA, which likely plays important roles in osteogenic differentiation processes under microgravity. The circRNA-miRNA-mRNA network showed that circRNAs might act as miRNA sponges to regulate osteoblast differentiation. Conclusion By presenting a better understanding of the molecular mechanisms of genes and circRNAs in simulated microgravity, the present study will provide a novel view of circRNAs in the regulation of osteogenic differentiation and bone formation.
... An advantage of the PamChip ® kinase activity microarrays (PamGene International BV) is their robustness and the small amount of protein that is required (i.e., typically 2.5-5 g of total protein for one PTK array and 0.25-1 g per STK array). This is in contrast to much higher amounts required for other proteomics technologies (e.g., several milligrams of total protein required for mass spectrometry analysis [30]). The PamChip ® 96 peptide microarray platform produced highly reproducible data (Fig. 2). ...
Article
Full-text available
Alzheimer's disease (AD) is characterized by a long pre-clinical phase (20-30 years), during which significant brain pathology manifests itself. Disease mechanisms associated with pathological hallmarks remain elusive. Most processes associated with AD pathogenesis, such as inflammation, synaptic dysfunction, and hyper-phosphorylation of tau are dependent on protein kinase activity. The objective of this study was to determine the involvement of protein kinases in AD pathogenesis. Protein kinase activity was determined in postmortem hippocampal brain tissue of 60 patients at various stages of AD and 40 non-demented controls (Braak stages 0-VI) using a peptide-based microarray platform. We observed an overall decrease of protein kinase activity that correlated with disease progression. The phosphorylation of 96.7% of the serine/threonine peptides and 37.5% of the tyrosine peptides on the microarray decreased significantly with increased Braak stage (p-value <0.01). Decreased activity was evident at pre-clinical stages of AD pathology (Braak I-II). Increased phosphorylation was not observed for any peptide. STRING analysis in combination with pathway analysis and identification of kinases responsible for peptide phosphorylation showed the interactions between well-known proteins in AD pathology, including the Ephrin-receptor A1 (EphA1), a risk gene for AD, and sarcoma tyrosine kinase (Src), which is involved in memory formation. Additionally, kinases that have not previously been associated with AD were identified, e.g., protein tyrosine kinase 6 (PTK6/BRK), feline sarcoma oncogene kinase (FES), and fyn-associated tyrosine kinase (FRK). The identified protein kinases are new biomarkers and potential drug targets for early (pre-clinical) intervention.
... A successful therapeutic approach should provide suppression of not only cancer cell proliferation, but also of the metastatic potential of any cell escaping from the primary tumor site to colonize the bone marrow [38]. Our study shows that both gold(III) complexes reduce EGFR expression, whose impaired endocytic downregulation contributes to oncogenic metastatic phenotype [39] and whose ligands are paracrine and autocrine growth factors [40][41][42]. Treatment of PC3 cells with AuD8 or AuD9 resulted in a significant decrease of the surface expression of both EGFR and its phosphorylated form (pEGFR), as assessed by flow cytometry (Figure 5A & 5B), whereas cisplatin was proved almost ineffective. EGFR phosphorylation is known to affect prostate cancer cells migration [43]. ...
Article
Background: Recently, novel gold(III)-dithiocarbamato peptidomimetics, designed to target peptide transporters upregulated in several tumor cells have shown promise as anticancer agents. Results: The biological behavior of the most promising derivatives AuD8 and AuD9 was studied in PC3 and DU145 prostate cancer cells. They exert higher cytotoxicity in vitro than the reference drug cisplatin and induce apoptosis, promoting mitochondrial membrane permeabilization and stimulating reactive oxygen species generation. Moreover, they inhibit both selenoenzyme thioredoxin reductase and proteasome activity. Additionally, AuD8 effectively reduces tumor growth in prostate tumor-bearing nude mice with minimal systemic toxicity. Conclusion: Altogether, our results provide insights into the anticancer activity of these gold(III)-dithiocarbamato peptidomimetics and support their potential as new agents for prostate cancer treatment.
Article
Androgens and androgen receptors are vital factors involved in prostate cancer progression, and androgen ablation therapies are commonly employed to treat advanced prostate cancer. Previously, FUsed in Sarcoma (FUS) was identified as an AR-interacting protein that enhances AR transcriptional activity. In the present study, we attempted to identify miRNAs that might target both FUS and AR to inhibit FUS and AR expression. Based on TCGA data and the online tools UALCAN, Kaplan Meier-plotter (KMplot), LncTar and miRWalk prediction, miR-133a-5p was selected. MiR-133a-5p expression was significantly downregulated in prostate cancer, and low miR-133a-5p expression was correlated with low survival probability. As predicted by LncTar and miRWalk, miR-133a-5p could bind to the 3′UTR of FUS and AR to inhibit their expression. MiR-133a-5p overexpression significantly suppressed the cell viability of the AR-positive prostate cancer cell lines VCaP and LNCaP, inhibited the expression of FUS, AR, as well as AR downstream targets IGF1R and EGFR. More importantly, miR-133a inhibition increased cancer cell proliferation as well as the expression of AR and AR downstream factors, while FUS knockdown exerted an opposite effect; the effect of miR-133a on cancer cell proliferation and AR could be significantly reversed by FUS knockdown. Moreover, IGF1R and EGFR knockdown reversed the effect of the miR-133a-5p inhibition. In summary, miR-133a-5p inhibits AR-positive prostate cancer cell proliferation by targeting FUS/AR, thus improving the resistance of prostate cancer to androgen ablation therapies, which requires further in vivo validation. We provided a novel miRNA regulation mechanism for proliferation regulation in AR-positive prostate cancer cells.
Article
There is increasing appreciation among researchers and clinicians of the value of investigating biology and pathobiology at the level of cellular kinase (kinome) activity. Kinome analysis provides valuable opportunity to gain insights into complex biology (including disease pathology), identify biomarkers of critical phenotypes (including disease prognosis and evaluation of therapeutic efficacy), and identify targets for therapeutic intervention through kinase inhibitors. The growing interest in kinome analysis has fueled efforts to develop and optimize technologies that enable characterization of phosphorylationmediated signaling events in a cost-effective, high-throughput manner. In this review, we highlight recent advances to the central technologies currently available for kinome profiling and offer our perspectives on the key challenges remaining to be addressed.
Article
In the last decade, we have been developing some gold(III) derivatives showing interesting antitumor properties and reduced systemic and renal toxicity, compared to the clinically-established reference drug cisplatin. Starting from the rationale at the base of our investigations, this review has been divided into two sections, with respect to our patented first- (aminoderivatives) and second-generation (peptidomimetics) potential drugs. Every section describes the in vitro and in vivo anticancer activity of the compounds, chosen as models, towards different types of tumor. In particular, we summarize the results achieved so far, in particular taking into account the latest in-depth studies related to their activity, mechanism of action and toxicological profile. Taken together, our data could open up new prospects for further advanced preclinical pharmacological testing.
Article
Locally advanced rectal cancer (LARC) comprises heterogeneous tumors with predominant hypoxic components, a hallmark of the tumor microenvironment and determinant of resistance to cytotoxic therapies, local recurrence, and metastatic progression. A rational integration of molecularly targeted agents in established combined-modality treatment regimens may improve local and systemic disease control, but will require a clear definition of functional biomarkers for patient stratification. In a prospective study of LARC patients given neoadjuvant chemotherapy and radiation, we applied a kinase substrate array technology to analyze the patients’ tumor biopsies sampled at the time of diagnosis, and observed that receptor tyrosine kinase activities integrated by high phosphatidylinositol 3-kinase signaling were correlated both with poor tumor response to the neoadjuvant treatment and adverse progression-free survival. Conceptually, the specific tumor signature of phosphatidylinositol 3-kinase signaling activity may point to actionable therapy targets in LARC patients with unfavorable disease features. Clinical trial registration number NCT00278694.
Article
Full-text available
Inactivating PTEN mutations are commonly found in prostate cancer, resulting in an increased activation of Akt. In this study, we investigate the role of PTEN deletion and protein expression in the development of hormone-refractory prostate cancer using matched hormone-sensitive and hormone-refractory tumours. Fluorescent in situ hybridisation and immunohistochemistry was carried out to investigate PTEN gene deletion and PTEN protein expression in the transition from hormone-sensitive to hormone-refractory prostate cancer utilising 68 matched hormone sensitive and hormone-refractory tumour pairs (one before and one after hormone relapse). Heterogeneous PTEN gene deletion was observed in 23% of hormone sensitive tumours. This increased significantly to 52% in hormone-refractory tumours (P=0.044). PTEN protein expression was observed in the membrane, cytoplasm and the nucleus. In hormone sensitive tumours, low levels of cytoplasmic PTEN was independently associated with shorter time to relapse compared to high levels of PTEN (P=0.028, hazard ratio 0.51 (95%CI 0.27-0.93). Loss of PTEN expression in the nucleus of hormone sensitive tumours was independently associated with disease-specific survival (P=0.031, hazard ratio 0.52, 95%CI 0.29-0.95). The results from this study demonstrate a role for both cytoplasmic and nuclear PTEN in progression of prostate cancer to the hormone-refractory state.
Article
Full-text available
The human melanoma cell line M24met metastasizes spontaneously from s.c. tumors to multiple distant sites in mice with severe combined immunodeficiency. Metastasis to lymph nodes and lungs is found in 100% of the animals. M24met has an undifferentiated phenotype and extra copies of the short arm of chromosome 7. This cell line expresses the epidermal growth factor receptor, and 425.3, a monoclonal antibody to the epidermal growth factor receptor, binds to 291,000 receptor molecules per M24met cell with a KD of 2.3 x 10(-10) M. This antibody has no effect on the proliferation of M24met cells under tissue culture conditions and does not mediate effector cell or complement-dependent cytotoxicity of these cells in vitro. However, treatment of established s.c. M24met tumors in mice with severe combined immunodeficiency with monoclonal antibody 425.3 specifically suppresses spontaneous metastasis of these tumors. Total doses of 4, 2, and 1 mg antibody per mouse decrease the number and size of melanoma metastases and prolong the life span of treated animals. Treatment with 4 mg of the F(ab')2 fragment of monoclonal antibody 425.3 does not influence M24met melanoma metastasis, implying a significant contribution of the Fc portion to the antimetastatic effect of this antibody.
Article
Castration-resistant prostate cancer (CRPC) is now the second most common cause of male cancer-related mortality. Recent evidence confirms that androgen receptor signalling continues to drive a significant proportion of progressing prostate cancers despite castrate serum levels of testosterone and multiple hormonal interventions. An increased understanding of the molecular biology underlying CRPC is informing on therapeutic targets for this disease, and we hypothesise that this will lead to the development of more efficacious drugs.
Article
The epidermal growth factor receptor (EGFR) signaling pathway is one of the most important pathways that regulate growth, survival, proliferation, and differentiation in mammalian cells. Reflecting this importance, it is one of the best-investigated signaling systems, both experimentally and computationally, and several computational models have been developed for dynamic analysis. A map of molecular interactions of the EGFR signaling system is a valuable resource for research in this area. In this paper, we present a comprehensive pathway map of EGFR signaling and other related pathways. The map reveals that the overall architecture of the pathway is a bow-tie (or hourglass) structure with several feedback loops. The map is created using CellDesigner software that enables us to graphically represent interactions using a well-defined and consistent graphical notation, and to store it in Systems Biology Markup Language (SBML).
Article
Loss of heterozygosity (LOH) in the region of 1Og23.3 has beenassociated with multiple tumors, including glioblastoma multiforme, melanoma, endometrial carcinoma, and prostate carcinoma. The tumor suppressor gene, PTEN/MMAC1, is also located in this region, and, in addition to other tumor types (eg, glioblastoma multiforme, endometrial, and melanoma), PTEN/MMAC1 mutations have been found in prostate cancer cell lines, xenografts, and hormone refractory prostate cancer tissue specimens. The aim of this study was to evaluate LOH at 1Oq23.3 as a marker of cancer progression in node-positive prostate cancer. Genetic alterations in the region of 1Oq23.3 were assessed in 23 node-positive (pT2-3, N+) and 44 node-negative prostate (pT2-3, NO) cancers with D10S532, D10S1687, D10S541, and D10S583 flanking polymorphic genetic markers; PTENCA, a genetic marker within PTEN/MMAC1, was also tested. Using DNA from paired normal and microdissected tumor samples, LOH at microsatellite loci was determined after polymerase chain reaction amplification. LOH in at least 1 marker was identified in 14% (6 of 44) of lymph node-negative and 43% (10 of 23) of lymph node-positive prostate cancers (chi-square test, P = .007). This increase in genetic alterations in node-positive prostate cancer suggests that 1Oq23.3 is a marker for metastatic progression.
Article
Extract: Cancer frequently spreads to bone, a process termed bone metastasis. Up to 70% of patients with breast cancer or prostate cancer, and 15 to 30% of patients with lung, colon, bladder or kidney cancer develop bone metastasis. Once tumors go to bone, such as in patients with breast cancer or prostate cancer, they are incurable, and only 20% of patients with breast cancer are still alive five years after they are found to have bone metastasis. It is estimated that about 350,000 people die with bone metastasis each year in the United States. Bone metastasis causes severe bone pain and can result in fractures without any injury, as well as other life-threatening conditions. There are two major types of bone metastasis, one in which bone destruction is the predominant feature and the other one in which new bone formation is predominant. Bone metastasis where bone destruction is the predominant feature is known as osteolytic, and that in which new bone formation is the primary feature is called osteoblastic. This classification for metastasis is really two extremes of a continuum because many patients can have both osteolytic and osteoblastic or mixtures of both in their bone metastasis. In fact, patients with prostate cancer who usually have bone metastasis that shows increased new bone formation also have increased bone destruction in the same lesions.
Article
An osteosarcoma cell line, OHS, was established from a patient with multiple skeletal manifestations of osteosarcoma, developing after bilateral retinoblastoma. The tumor cells expressed sarcoma-associated antigens and showed rapid growth in monolayers and as multicellular spheroids. They formed distinct colonies in soft agar, and subcutaneous tumors in nude mice. Morphological studies indicated that OHS cells had retained important characteristics of the cells of origin. No deletion of the retinoblastoma genes on chromosome 13q14 could be demonstrated with the banding techniques used. However, cytogenetic studies revealed double minute chromosomes, as evidence of gene amplification, as well as translocations involving chromosomes 1,6,11 and 13. The OHS line can be used to study the genetic basis of tumor initiation and growth, and to elucidate factors predisposing for second primary cancers in retinoblastoma patients.