ArticlePDF Available

Identification of Docetaxel Resistance Genes in Castration-Resistant Prostate Cancer

Authors:
  • Fundación Instituto Oncológico Dr. Rosell
  • Hospital Universitario Germans Trias i Pujol

Abstract and Figures

Docetaxel-based chemotherapy is the standard first-line therapy in metastatic castration-resistant prostate cancer (CRPC). However, most patients eventually develop resistance to this treatment. In this study, we aimed to identify key molecular genes and networks associated with docetaxel resistance in two models of docetaxel-resistant CRPC cell lines and to test for the most differentially expressed genes in tumor samples from patients with CRPC. DU-145 and PC-3 cells were converted to docetaxel-resistant cells, DU-145R and PC-3R, respectively. Whole-genome arrays were used to compare global gene expression between these four cell lines. Results showed differential expression of 243 genes (P < 0.05, Bonferroni-adjusted P values and log ratio > 1.2) that were common to DU-145R and PC-3R cells. These genes were involved in cell processes like growth, development, death, proliferation, movement, and gene expression. Genes and networks commonly deregulated in both DU-145R and PC-3R cells were studied by Ingenuity Pathways Analysis. Exposing parental cells to TGFB1 increased their survival in the presence of docetaxel, suggesting a role of the TGF-β superfamily in conferring drug resistance. Changes in expression of 18 selected genes were validated by real-time quantitative reverse transcriptase PCR in all four cell lines and tested in a set of 11 FFPE and five optimal cutting temperature tumor samples. Analysis in patients showed a noteworthy downexpression of CDH1 and IFIH1, among others, in docetaxel-resistant tumors. This exploratory analysis provides information about potential gene and network involvement in docetaxel resistance in CRPC. Further clinical validation of these results is needed to develop targeted therapies in patients with CRPC that can circumvent such resistance to treatment.
Content may be subject to copyright.
Therapeutic Discovery
Identication of Docetaxel Resistance Genes in
Castration-Resistant Prostate Cancer
Mercedes Marín-Aguilera
1
, Jordi Codony-Servat
1
, Susana G. Kalko
2
, Pedro L. Fern
andez
3
,
Raquel Bermudo
5
, Elvira Buxo
1
, María Jos
e Ribal
4
, Pedro Gasc
on
1
, and Bego~
na Mellado
1
Abstract
Docetaxel-based chemotherapy is the standard first-line therapy in metastatic castration-resistant prostate
cancer (CRPC). However, most patients eventually develop resistance to this treatment. In this study, we aimed
to identify key molecular genes and networks associated with docetaxel resistance in two models of docetaxel-
resistant CRPC cell lines and to test for the most differentially expressed genes in tumor samples from patients
with CRPC. DU-145 and PC-3 cells were converted to docetaxel-resistant cells, DU-145R and PC-3R,
respectively. Whole-genome arrays were used to compare global gene expression between these four cell
lines. Results showed differential expression of 243 genes (P<0.05, Bonferroni-adjusted Pvalues and log ratio >
1.2) that were common to DU-145R and PC-3R cells. These genes were involved in cell processes like growth,
development, death, proliferation, movement, and gene expression. Genes and networks commonly deregu-
lated in both DU-145R and PC-3R cells were studied by Ingenuity Pathways Analysis. Exposing parental cells
to TGFB1 increased their survival in the presence of docetaxel, suggesting a role of the TGF-bsuperfamily in
conferring drug resistance. Changes in expression of 18 selected genes were validated by real-time quantitative
reverse transcriptase PCR in all four cell lines and tested in a set of 11 FFPE and five optimal cutting temperature
tumor samples. Analysis in patients showed a noteworthy downexpression of CDH1 and IFIH1, among others,
in docetaxel-resistant tumors. This exploratory analysis provides information about potential gene and
network involvement in docetaxel resistance in CRPC. Further clinical validation of these results is needed
to develop targeted therapies in patients with CRPC thatcancircumventsuchresistancetotreatment.
Mol Cancer Ther; 11(2); 329–39. 2011 AACR.
Introduction
Prostate cancer is the second most common cancer
among men worldwide (1). Approximately 85% of newly
diagnosed prostate cancer cases are localized to the pros-
tate, whereas the remainder are invasive or metastatic
disease (2). Patients with metastatic prostate cancer
respond initially to antiandrogen therapy. However,
tumors eventually progress and transform themselves
into castration-resistant prostate cancer (CRPC). Doce-
taxel improves survival of patients with metastatic CRPC
and is considered a standard first-line therapy in such
cases (3; 4). However, only approximately 50% of patients
respond to docetaxel and most of them eventually devel-
op resistance to this therapy.
Taxanes bind b-tubulin, stabilizing microtubules
assembly and preventing depolymerization in the
absence of GTP (5). Furthermore, docetaxel leads to Bcl-
2 phosphorylation, which causes apoptosis of cancer cells
that had previously blocked the apoptotic-inducing
mechanism, leading to tumor regression (6). Some pro-
teins such as stathmin, Aurora-A, and b-III tubulin have
been described to be involved in resistance to antimicro-
tubule agents (7–9). However, mechanisms of resistance
to docetaxel in CRPC need to be further elucidated to
design targeted therapies that can circumvent treatment
resistance.
cDNA microarrays technology has given us the ability
to simultaneously examine the expression of thousands of
genes and determine the molecular profile of clinical
phenotypes, some of which can be involved in specific
cell profiles, such as chemotherapy resistance. This study
compared gene expression profiles of 2 models of doc-
etaxel-resistant CRPC cell lines, and identified a set of
genes involved in resistance to this chemotherapy. The
most differentially expressed genes were validated by
real-time quantitative reverse transcriptase PCR (qRT-
PCR) in cell lines and, in an exploratory way, in CRPC
tumor samples.
Authors' Afliations:
1
Laboratory and Medical Oncology Department,
2
Bioinformatics Support Unit, Departments of
3
Pathology and
4
Urology,
Hospital Clínic; and
5
Human and Experimental Functional Oncomorphol-
ogy Department, Institut d'Investigacions Biom
ediques August Pi i Sunyer,
Barcelona, Spain
Note: Supplementary data for this article are available at Molecular Cancer
Therapeutics Online (http://mct.aacrjournals.org/).
Corresponding Author: Bego~
na Mellado, Medical Oncology Department,
Hospital Clínic de Barcelona, Villarroel 170, Barcelona 08036, Spain.
Phone: 34-93-227-5400, ext. 2262; Fax: 34-93-454-6520; E-mail:
bmellado@clinic.ub.es
doi: 10.1158/1535-7163.MCT-11-0289
2011 American Association for Cancer Research.
Molecular
Cancer
Therapeutics
www.aacrjournals.org 329
on November 5, 2015. © 2012 American Association for Cancer Research. mct.aacrjournals.org Downloaded from
Published OnlineFirst October 25, 2011; DOI: 10.1158/1535-7163.MCT-11-0289
Materials and Methods
Cell culture
The human prostate carcinoma cell lines DU-145 and
PC-3 (obtained from American Tissue Culture Collec-
tion) were converted to docetaxel-resistant cells by
exposing them to an initial dose of 5 nmol/L doce-
taxel and culturing surviving cells during 1 year and
6 months, respectively, with increasing doses in an
intermittent regimen. DU-145 and DU-145R cell lines
were cultured in RPMI-1640 medium (Invitrogen), and
PC-3 and PC-3R in F-12K nutrient mixture medium
(Invitrogen), both supplemented with 10% FBS. No
further authentication of the cell lines was done by the
authors. Docetaxel (Sigma) was dissolved in dimethyl
sulfoxide (10 mmol/L). IC
50
values were estimated in
triplicate from the dose–response curves.
Microarray hybridizations and differential
expression analysis
Total RNAs were isolated from DU-145, DU-145R,
PC-3, and PC-3R cell lines (in triplicate) using the TRIzol
Reagent (Invitrogen) according to the manufacturer’s
instructions. RNAs were purified using RNeasy Micro
kit (Qiagen), and quality and quantity were assessed on
a spectrophotometer. Fragmented, labeled, and ampli-
fied cDNA was hybridized to the Affymetrix Human
Genome U133Plus2.0 array, which represents about
38,500 well-characterized genes. Washes and scanning
of the arrays were carried out according to manufac-
turer’s instructions.
Raw expression measures were summarized after
background correction and normalization steps using
the robust multiarray analysis (RMA) methodology in
the affy (10) package from the Bioconductor project (11).
Unsupervised cluster analysis of high variability genes
was done with dChip v1.3 software (12). Differential
expression analysis was carried out by a linear model
using the empirical Bayes method to moderate the
standard errors of the estimated log ratio changes with
the limma package (13), according to adjusted Pvalues
less than 0.05 for the comparison of interest. Special
attention was given to the statistically significant genes
that showed the largest changes ( logratio
>1:2). CEL
files and RMA values were deposited on Gene Expres-
sion Omnibus (GSE33455).
Network analysis
Gene interactions were studied using the Ingenuity
Pathway Analysis (IPA; Ingenuity Systems) software
(14), according to IPA instructions. First, genes differen-
tially expressed between resistant and parent cell lines
(DU-145R vs. DU-145 and PC-3R vs. PC-3) were listed.
Interactions between common deregulated genes in DU-
145R and PC-3R cells were then analyzed to understand
resistance mechanisms at a molecular level. Network-
eligible genes were placed by IPA into networks and
ranked by the relevance of the network-eligible molecules.
Patients and samples
Tumor samples from patients with metastatic prostate
cancer were collected to test microarray results in patients.
We selected patients who were prescribed docetaxel-
based therapy after the palliative transurethral deso-
bstructive resection, or after being biopsied once the
tumor had already spread. Samples were obtained before
the start of docetaxel treatment.
We were able to collect 11 formalin-fixed, paraffin-
embedded (FFPE) samples, and 5 tumor samples embed-
ded in 1 to 2 mL of optimal cutting temperature (OCT)
medium immediately after surgery. The OCT samples
were stored at 80C until processing. The institutional
review board approved this study and written informed
consent was obtained from all patients.
FFPE samples belonged to 11 patients with CRPC, all of
whom had bone metastasis; 5 also had lymph node
lesions, 2 had lung metastasis, and 2 others had liver and
ureteral tumor growth. Six patients partially responded to
docetaxel and the other 5 progressed during the treat-
ment. On the other hand, OCT samples were extracted
from 5 patients with CRPC with bone metastasis, of whom
4 also had a lymph node lesion and 1 had liver metastasis.
Two patients partially responded to docetaxel and the
other progressed during the treatment.
Treatment response was evaluated by prostate-specific
antigen–based response criteria (15) and Response Eval-
uation Criteria in Solid Tumors (RECIST; ref. 16).
Gene validation in cell lines
Genes that could potentially be related with docetaxel
resistance were selected for further validation in cell lines
using qRT-PCR. Genes were selected according to their
change in degree of relative expression ( logratio
>1:2),
but also by function, that is, genes involved in known path-
ways such as cell adhesion, cell signaling, or regulation
of apoptosis, using DAVID (17; 18) and IPA softwares (14).
Total RNAsfrom cell lines were isolated using the TRIzol
Reagent (Invitrogen) according to the manufacturer’s
instructions. One microgram of total RNA was reverse
transcribed using the High Capacity cDNA Archive Kit
(Applied Biosystems), following manufacturer’s instruc-
tions. qRT-PCR was carried out in a 7500 Real Time PCR
system (Applied Biosystems), according to the manufac-
turer’s recommendations. Data were acquired using SDS
Software 1.4. Expression values were based on the quan-
tification cycle (C
q
) from target genes relative to ACTB
endogenous control gene (DC
q
). Relative expression with
respect to each reference group studied was reported as
log ratio. Commercial codes for primers and probes from
target genes were AREG (amphiregulin-Hs00950669_m1),
CDH1 (E-cadherin-Hs01023895_m1), CYBRD1 (cyto-
chrome b-reductase 1-Hs00227411_m1), DLC1 (deleted
in liver cancer 1-Hs00183436_m1), GJB2 (gap junction
protein, b2-Hs00955889_m1), GSPT2 (G
1
to S-phase tran-
sition 2-Hs00250696_s1), IFIH1 (IFN induced with heli-
case C domain 1-Hs01070332_m1), IL8 (interleukin
8-Hs00174103_m1), MAPK13 (mitogen-activated protein
Marín-Aguilera et al.
Mol Cancer Ther; 11(2) February 2012 Molecular Cancer Therapeutics
330
on November 5, 2015. © 2012 American Association for Cancer Research. mct.aacrjournals.org Downloaded from
Published OnlineFirst October 25, 2011; DOI: 10.1158/1535-7163.MCT-11-0289
kinase 13-Hs00559623_m1), MPZL2 (myelin protein zero-
like 2-Hs00170684_m1), MX1 (myxovirus resistance
1-Hs00895608_m1), MYO6 (myosin VI-Hs00192265_
m1), S100A4 (S100 calcium–binding protein A4-
Hs00243202_m1), SERPINA1 (serpin peptidase inhibitor,
clade A-Hs01097800_m1), SYK (spleen tyrosine kinase-
Hs00895377_m1), EPCAM (epithelial cell adhesion
molecule-Hs00901885_m1), NEAT1 (nuclear paraspeckle
assembly transcript 1-Hs01008264_s1), and TNFAIP3 (tu-
mor necrosis factor, a-induced protein 3-Hs00234713_m1).
Gene expression in tumor samples
Tissues from selected FFPE and OCT blocks were sec-
tioned at 10 and 20 mm thicknesses, respectively, imme-
diately before the RNA extraction. Total RNA from FFPE
samples was obtained using the RecoverAll Total Nucleic
Acid Isolation Kit for FFPE (Ambion). The RNAs from
OCT samples were extracted using TRIzol Reagent (Invi-
trogen) according to the manufacturer’s instructions.
Quality and quantity of total RNAs were measured by
ND-1000 Spectrophotometer (Nanodrop Technologies).
The genes selected for microarrays validation in cell
lines were also tested in tumor samples following the
same qRT-PCR protocol except that the expression values
of target genes were relative to the C
q
mean of ACTB,B2M,
and GUSB endogenous genes.
Viability assay
TGFB1 (Sigma-Aldrich) was dissolved in sterile 4
mmol/L HCl containing 0.1% endotoxin-free recombi-
nant human serum albumin (50 mg/mL). Parent cells
(DU-145 and PC-3) were maintained during 7 consecutive
days with TGFB1 (5 ng/mL) in the medium. Parental cells
with and without sustained treatment were then seeded at
a density of 3,200 cells per well in a 96-well microtiter plate
in the corresponding medium with 10% FBS. After 24
hours, cells were exposed to docetaxel with/without
TGFB1 for an additional 72 hours. Finally, cell prolifera-
tion/viability was assessed by using MTT colorimetric
assay (Promega).
Statistical analysis
Changes in gene expression comparing docetaxel-
resistant and docetaxel-sensitive cell lines and tumor
samples by qRT-PCR were analyzed with Wilcoxon
rank-sum test, considering as positively validated those
genes with significant expression changes (P<0.05).
SPSS 12.0 software was used for statistical analyses.
Results
Docetaxel-resistant cell lines
DU-145R and PC-3R cells acquired levels of resistance
to docetaxel that was 2 to 5 times higher than their parent
cells. IC
50
values for DU-145R and PC-3R ranged from 10
to 15 and 20 to 22 nmol/L, respectively, whereas DU-145
and PC-3 registered from 4 to 5 and 3 to 5 nmol/L,
respectively (data not shown).
Differentially expressed genes in resistant cells
Initially, unsupervised clustering analysis of microar-
rays was carried out using the probe sets that varied most
throughout the whole experiment, and this exploration
revealed a good segregation of the arrays in their respec-
tive classes on the basis of expression values (data not
shown). Subsequent differential expression analysis
revealed 1,064 and 1,361 differentially expressed genes
(Bonferroni <0.05 and log ratio >1.2), equivalent to 1,710
and 2,117 Affymetrix probe sets, between DU-145R versus
DU-145 and PC-3R versus PC-3, respectively (Supple-
mentary Table SI). Top 20 over- and downexpressed genes
from docetaxel-resistant cells with respect to their parent
cells according to log ratio expression values, and their
main function, are summarized in Supplementary Table
SII. When comparing differentially expressed genes
between both resistant cell lines (DU-145R and PC-3R),
243 genes overlapped (Supplementary Table SIII). From
those genes, 172 were over- or downexpressed in both
DU-145R and PC-3R cells versus their parent cells. The top
10 commonly over- and downexpressed genes, with their
respective main function, are summarized in Table 1.
Ingenuity network analysis
Only considering probe sets common to both resistant
cell lines, IPA qualified 252 genes as network and function
eligible. A core analysis showed 12 networks on the basis
of this gene selection with a score more than 2. The 2 top
networks were detected with scores of 51 and 39 respec-
tively, and were the same for DU-145R and PC-3R cell
lines (Fig. 1). Specifically, network 1 (Fig. 1A) was centered
on, amongst others, the nuclear receptor PPARA (perox-
isome proliferator-activated receptor a). This gene direct-
ly interacts with an important complex for cell survival
and proliferation, NFKB, which in turn interacts with
TNFAIP3,HMGA1 (high mobility group AT-hook 1), and
ISG15 (ISG15 ubiquitin-like modifier). Other deregulated
genes directly related to PPARA are SOCS2 (suppressor of
cytokine signaling 2), ASL (argininosuccinate lyase),
ASNS (asparagine synthetase), ELOVL6 (ELOVL family
member 6), IGFBP6 (insulin-like growth factor binding
protein 6) and CITED2 (Cbp/p300-interacting transacti-
vator, with Glu/Asp-rich carboxy-terminal domain 2).
Other genes commonly deregulated in network 1 are
SERPINA1 (serpin peptidase inhibitor, clade A), VDR
(vitamin D receptor), ITGB2 (integrin, b2), and TGFBR3
(TGF-breceptor III, also known as betaglycan; Fig. 1A).
The last is overexpressed in both DU-145R and PC-3R
cells. Interestingly, other TGF-bmembers appear deregu-
lated in the same network, such as TGFB2, which is
differentially regulated in both cell lines, and TGF-b
ligand, which is included by the software as a link
between the former TGF-belements and LTBP2 (latent
TGF-bbinding protein 2).
Network 2 (Fig. 1B) is mainly centered on the CDH1
gene, which codifies a classical calcium dependent cell–
cell adhesion glycoprotein. MYO6 (myosin VI), ID2
(inhibitor of DNA binding 2), PTPRM (protein tyrosine
Docetaxel Resistance in Castoration-Resistant Prostate Cancer
www.aacrjournals.org Mol Cancer Ther; 11(2) February 2012 331
on November 5, 2015. © 2012 American Association for Cancer Research. mct.aacrjournals.org Downloaded from
Published OnlineFirst October 25, 2011; DOI: 10.1158/1535-7163.MCT-11-0289
Table 1. Top 10 commonly over- and downexpressed genes in DU-145R and PC-3R versus their parent cells, with their respective main function
and log ratio expression
Overexpressed
DU-145R vs. DU-145 PC-3R vs. PC-3
Gene symbol Log ratio Main function Gene symbol Log ratio Main function
GSPT2 4.487 GTPase NEAT1 3.468 Nonprotein coding
ID2 4.050 Multicellular organism development;
regulation of transcription
FLJ27352 3.287 Undened
CXCR7 3.879 Signal transduction TGFBR3 2.960 Epithelial to mesenchymal transition; signal
transduction; cell cycle; cell migration
NDRG1 3.233 Stress and hormone responses,
cell growth and differentiation
PLSCR4 2.552 Phospholipid scrambling
FRMD3 3.191 Structural protein HTRA1 2.389 Regulation of cell growth
TP53INP1 2.797 Cell-cycle arrest; induction of apoptosis S100A4 2.217 Epithelial to mesenchymal transition;
cell-cycle progression and differentiation.
GOSR2 2.434 Intracellular protein transport CYBRD1 2.158 Transport; oxidation reduction
CITED2 2.372 Vasculogenesis; response to hypoxia;
positive regulation of cellcell adhesion;
positive regulation of cell cycle
PPP2R2C 2.052 Signal transduction
KLHL24 2.309 Undened GSPT2 2.006 GTPase
NEAT1 2.267 Nonprotein coding PC 1.979 Metabolism, insulin secretion, and synthesis
of the neurotransmitter glutamate
Downexpressed
SCEL 6.428 Epidermis development; assembly
or regulation of structural proteins
ESRP1 6.612 Regulation of RNA splicing
EPCAM 5.357 Calcium-independent cell adhesion TACSTD2 6.309 Signal transduction
C1orf116 5.292 Undened EFEMP1 6.079 Extracellular matrix glycoprotein
TACSTD2 5.240 Signal transduction GJB2 5.984 Transport; cellcell signaling
SERPINA1 5.153 Response to hypoxia MPZL2 5.088 Cell adhesion
AIM1 4.876 Undened RBM47 4.333 Undened
AREG 4.864 EGF receptor signaling pathway;
cell proliferation; cellcell signaling
JPH1 4.312 Calcium ion transport into cytosol
JPH1 3.992 Calcium ion transport into cytosol TXNIP 4.224 Transcription; response to oxidative
stress; regulation of cell proliferation
GPR87 3.648 Signal transduction GPR87 4.192 Signal transduction
GALNT3 3.501 Carbohydrate metabolic process KAP2.1B 4.181 Keratin-associated protein
Marín-Aguilera et al.
Mol Cancer Ther; 11(2) February 2012 Molecular Cancer Therapeutics
332
on November 5, 2015. © 2012 American Association for Cancer Research. mct.aacrjournals.org Downloaded from
Published OnlineFirst October 25, 2011; DOI: 10.1158/1535-7163.MCT-11-0289
A
B
Figure 1. Gene networks deregulated in resistant cell lines with respect to their parent cells. A, the most signicant (P<0.05) gene network deregulated in
resistant cells, scored 51 by IPA software. B, the second most signicant (P<0.05) gene network deregulated in resistant cells, scored 39 by IPA software.
Docetaxel Resistance in Castoration-Resistant Prostate Cancer
www.aacrjournals.org Mol Cancer Ther; 11(2) February 2012 333
on November 5, 2015. © 2012 American Association for Cancer Research. mct.aacrjournals.org Downloaded from
Published OnlineFirst October 25, 2011; DOI: 10.1158/1535-7163.MCT-11-0289
phosphatase, receptor type M), and OCLN (occludin)
are other deregulated genes directly related with
CDH1.
No overlapping genes were found between networks 1
and 2, although both are involved in gene expression.
Functional in silico analysis
IPA detected several biologic functions with overrep-
resentation, when considering together the differentially
expressed genes in docetaxel-resistant versus their parent
cells. Such functions were related with elemental metab-
olism of cell survival like gene expression, cell growth and
proliferation, cell cycle, cell death, and cell movement.
Specifically, and as expected, the 2 main networks were
involved in cell growth and proliferation, but also in
gene expression, cell movement and development, and
the skeletal and muscular system.
The top overexpressed individual genes were enzymes,
cell surface receptors, transcription regulators, and struc-
tural proteins involved in cell transport, signaling, bind-
ing, and cytoskeletal organization. Similarly, proteins
from downexpressed genes were structural proteins, sig-
nal transducers, enzymes, growth factors, cell receptors,
and transcription factors involved in cell structure, adhe-
sion, transport, and cycle regulation.
Cell viability assay for TGF-bfamily
The TGF-bsuperfamily was represented at network 1
and was overexpressed in resistant cell lines. We selected
this target for a functional study of docetaxel resistance.
The results of MTT experiments showed that continuous
treatment with TGFB1 (5 ng/mL) increased DU-145 and
PC-3 cell viability until 16.5% and 15.6%, respectively,
with respect to cells that were not cultured with the ligand.
Furthermore, when parental cells were exposed to TGFB1
(10 ng/mL) for 72 hours, cell proliferation was until 27%
greater in PC-3 cells at 7.5 nmol/L of docetaxel than in
cells without TGFB1 treatment. Cell viability of DU-145
cells was not affected by TGFB1 under these experimental
conditions (Fig. 2).
Validation by qRT-PCR in cell lines and test in tumor
samples
A panel of 18 of the top 20 with the highest or lowest
expression range in both DU-145R and PC-3R versus
parent cells lines (see materials and methods) was selected
for being commonly over- or downexpressed.
It was possible to confirm by qRT-PCR significant dif-
ferences in expression in all 18 (Wilcoxon test, P<0.05)
in the 4 cell lines studied (Fig. 3). Moreover, their expres-
sion was analyzed in docetaxel-resistant (n¼5) versus
docetaxel-sensitive (n¼6) FFPE tumor samples, and
docetaxel-resistant (n¼3) versus docetaxel-sensitive
(n¼2) OCT tumor samples from patients with metastatic
CRPC. This analysis showed a significant downexpres-
sion of NEAT1 (P¼0.044) in FFPE tumors. Of the 18
markers studied, 10 markers in the FFPE samples and 13
in OCT samples were deregulated in the same way as in
the in vitro models. Interestingly, as shown in Fig. 4, 7 of
these marker genes were commonly downexpressed in
docetaxel-resistant cell lines and in FFPE and OCT sam-
ples from patients with CRPC resistant to docetaxel:
AREG,CDH1,DLC1,GJB2,IFIH1,MX1, and EPCAM
(Fig. 4).
Discussion
This study revealed potential genes and networks
involved in resistance to docetaxel in 2 CRPC cell lines,
DU-145 and PC-3. Both cell lines were converted to
A
B
DU-145 (sustained TGFB1)
DU-145 (TGFB1, 72 h) PC-3 (TGFB1, 72 h)
PC-3 (sustained TGFB1)
120
100
80
60
40
20
0
Docetaxel
Docetaxel + TGFB1
Docetaxel
Docetaxel + TGFB1
Docetaxel
Docetaxel + TGFB1
Docetaxel
Docetaxel + TGFB1
Ctrl. 0 1 1.8 2.5
% cell viability
120
100
80
60
40
20
0
% cell viability
120
100
80
60
40
20
0
% cell viability
120
100
80
60
40
20
0
% cell viability
5 7.5 10 20
Docetaxel (nmol/L)
Ctrl. 0 1 1.8 2.5 5 7.5 10 20
Docetaxel (nmol/L)
Ctrl. 0 1 1.8 2.5 5 7.5 10 20
Docetaxel (nmol/L)
Ctrl. 0 1 1.8 2.5 5 7.5 10 20
Docetaxel (nmol/L)
TGFB1 (5 ng/mL) TGFB1 (5 ng/mL)
TGFB1 (10 ng/mL) TGFB1 (10 ng/mL)
Figure 2. Cell viability assay of
docetaxel treatment for 72 hours in
DU-145 and PC-3 cells. A, after a
sustained treatment with TGFB1
5 ng/mL during 7 days plus
additional 72 hours and after
72 hours of treatment with TGFB1
10 ng/mL (B). Cell viability is
expressed as the relative
colorimetric signal calculated from
the untreated wells (Ctrl.). Each
experimental condition was carried
out in triplicate (SD is shown).
Marín-Aguilera et al.
Mol Cancer Ther; 11(2) February 2012 Molecular Cancer Therapeutics
334
on November 5, 2015. © 2012 American Association for Cancer Research. mct.aacrjournals.org Downloaded from
Published OnlineFirst October 25, 2011; DOI: 10.1158/1535-7163.MCT-11-0289
docetaxel-resistant and then microarrays analysis of par-
ent and resistant cells was carried out.
Docetaxel resistance was achieved with nanomolar
concentrations of the drug (10–15 and 20–22 nmol/L for
DU-145R and PC-3R, respectively). The fact that docetaxel
IC
50
in resistant cells was not substantially higher than in
sensitive cells is consistent with other in vitro docetaxel
resistance studies (19; 20). This issue may be partially
explained by the fact that microtubule alterations induced
by taxanes can occur even at concentration of 1 nmol/L
(21).
Microarrays analysis was focused on the identification
of commonly deregulated genes in the 2 different cell line
models. We found 243 eligible genes (Bonferroni <0.05
and log ratio >1.2) that were similarly deregulated in DU-
145R and PC-3R with respect to their parent cell lines.
These genes were involved in survival functions such as
gene expression and cell growth, proliferation, death, and
movement. Interestingly, in both cell lines GSPT2 and
NEAT1 were within the top 10 overexpressed genes, and
TACSTD2 (tumor-associated calcium signal transducer
2), JPH1 (junctophilin 1), and GPR87 (G-protein–coupled
receptor 87) within the top 10 downexpressed genes.
Among these genes, only TACSTD2 and GPR87 have
already been related with cancer in the literature (22; 23).
Briefly, GSPT2 encodes a GTPase that may be involved
in mRNA stability and NEAT1 is a non–protein-coding
RNA that seems to regulate mRNA export. Such involve-
ment of docetaxel with mRNA regulation is not surprising
because, as previously described, taxanes can promote
transcription and mRNAs stabilization of some genes.
Such stabilizing effects can be achieved by the stimulation
of proteins that bind the AU-rich region of the 30UTR
region of target genes (24).
On the other hand, TACSTD2 encodes a carcinoma-
associated antigen that is a cell surface receptor that
transduces calcium signals. It has been described to be
unmethylated in normal prostate cells and prostatic
intraepithelial neoplasia but hypermethylated in primary
prostate tumors (25), suggesting that methylation could
be one of the mechanisms for silencing the expression of
crucial genes, thus inactivating the apoptotic pathway in
CRCP. The real significance of TACSTD2 intraexpression
in resistant cell lines and the potential role of docetaxel in
methylation-based regulation need to be further explored.
0.5
0
–0.5
–1
–1.5
–2
Genes
AREG
CDH1
CYBRD1
DLC1
GJB2
GSPT2
IFIH1
IL8
MAPK13
MPZL2
MX1
MYO6
S100A4
SERPINA1
SYK
EPCAM
NEAT1
TNFAIP3
Genes
AREG
CDH1
CYBRD1
DLC1
GJB2
GSPT2
IFIH1
IL8
MAPK13
MPZL2
MX1
MYO6
S100A4
SERPINA1
SYK
EPCAM
NEAT1
TNFAIP3
FFPE tumor samples
OCT tumor samples
Log ratio
1.5
1
0.5
0
–0.5
–1
–1.5
–2
–2.5
–3
–3.5
Log ratio
Figure 4. Differential gene expression by qRT-PCR in docetaxel-resistant
versus docetaxel-sensitive FFPE and OCT tumor samples from patients
with metastatic CRPC. Expression data are represented by a log ratio
calculated comparing DC
q
from docetaxel-resistant patients with the
median of DC
q
from docetaxel-sensitive patients. DC
q
was calculated as
the difference between C
q
of target genes and the mean of C
q
of the
endogenous control genes ACTB,B2M,andB-GUS.,P<0.05.
6
4
2
0
–2
–4
–6
–8
Validated genes
AREG
CDH1
CYBRD1
DLC1
GJB2
GSPT2
IFIH1
IL8
MAPK13
MPZL2
MX1
MYO6
S100A4
SERPINA1
SYK
EPCAM
NEAT1
TNFAIP3
DU-145R vs. DU-145
Log ratio
6
4
2
0
–2
–4
–6
–8
Validated genes
AREG
CDH1
CYBRD1
DLC1
GJB2
GSPT2
IFIH1
IL8
MAPK13
MPZL2
MX1
MYO6
S100A4
SERPINA1
SYK
EPCAM
NEAT1
TNFAIP3
PC-3R vs. PC-3
Log ratio
Microarray s
qRT-P CR
Microarray s
qRT-P CR
Figure 3. Validation of microarrays data by qRT-PCR in DU-145,
DU-145R, PC-3, and PC-3R cell lines. Expression data are represented
by a log ratio calculated by comparing DC
q
from resistant cells with
DC
q
from parent cells. DC
q
was calculated as the difference between C
q
of target genes and C
q
of the endogenous control gene ACTB.
Docetaxel Resistance in Castoration-Resistant Prostate Cancer
www.aacrjournals.org Mol Cancer Ther; 11(2) February 2012 335
on November 5, 2015. © 2012 American Association for Cancer Research. mct.aacrjournals.org Downloaded from
Published OnlineFirst October 25, 2011; DOI: 10.1158/1535-7163.MCT-11-0289
Another downexpressed gene was JPH1, which med-
iates cross-talk between the cell surface and intracellular
ion channels, modulating electrochemical gradients that
are vital to the cell and a potentially strong influence on
drug activity, according to previous work (26). According
to the authors, the expression of several genes that encode
subunits of sodium, chloride, potassium, and other cation
channels also is correlated with drug activity.
GPR87 encodes a G-protein–coupled receptor that
plays an essential role in many physiologic processes,
including neurotransmission, immunity, and inflamma-
tion. This gene seems to be overexpressed in diverse
carcinomas and plays an essential role in tumor cell
survival. A recent study revealed that a lack of GPR87
triggers an increase in p53, concomitant with a decrease in
AKT, which results in the sensitization of tumor cells to
DNA damage–induced apoptosis and growth suppres-
sion (23). However, these results are in disagreement with
the fact that resistant cells downregulate the expression of
GPR87 but remain able to survive and proliferate. The
role of several other genes that contribute to docetaxel
resistance must account for such discrepancy.
Interestingly, several well-known molecules already
related with docetaxel resistance have been found to be
deregulated in the present work. ABCA13,ABCA8, and
ABCC2, all from the ABC (ATP-binding cassette) family of
drug transporters, are significantly deregulated in DU-
145R cells; only ABCC2 was already known to be involved
in multidrug resistance (27). None of these genes has been
found to be deregulated in both resistant cell lines. On the
other hand, there are growing evidences about the role of
b-tubulin isotypes in resistance to taxanes in CRPC, as
they are the primary target of these drugs (9; 28). In the
present study we observed that TUBB2B (tubulin, b2B)
was downexpressed in PC-3R cells versus their parent cell
line (see Supplementary Table SI), but no differences were
observed in the expression of b-tubulins in the DU-145
model. Furthermore, tubulin is one of the molecules in-
cluded by the Ingenuity software as a link between EML1
and MAP2, actin and calmodulin proteins (Fig. 1B);
however, its expression was not significantly deregulated.
These results suggest that alternative pathways may be
more important in generating docetaxel resistance than
those directly related to tubulin alterations.
Apart from an individual view of deregulated genes, in
this study we focused on IPA analysis that allowed us to
obtain global and integrated molecular information about
interactions between significant differentially expressed
genes. Results from such analysis showed that the most
significant network was centered on PPARA, which was
overexpressed in docetaxel-resistant cells (Fig. 1A).
PPARA is a nuclear receptor that regulates the expression
of multiple genes involved in cell proliferation, differen-
tiation, and immune and inflammation responses. The
alpha form of this gene is functional in human prostate
and is downregulated by androgens. It has been sug-
gested that its overexpression in advanced prostate cancer
indicates a role in tumor progression, with the potential
involvement of dietary factors (29). As shown in Fig. 1A,
PPARA interacts with CITED2 (Cbp/p300-interacting
transactivator, with Glu/Asp-rich carboxy-terminal
domain 2), which has been involved in cisplatin resistance
(30), and notably with the transcription regulators ETS1
(v-ets erythroblastosis virus E26 oncogene homolog 1).
Inappropriate expression of ETS1 has been observed in a
variety of human cancers and might play an important
role in carcinogenesis and/or the progression of human
prostate cancer (31). However, no link between ETS1
expression and docetaxel resistance has been described
to date.
ETS1 directly interacts with VDR and ITGB2,which
also are downexpressed. VDR is a transacting transcrip-
tional regulatory factor involved in a variety of meta-
bolic pathways, but also in antineoplastic activities. In
fact, calcitriol, the most active metabolite of vitamin D,
showed to enhance antitumor activity of docetaxel,
although an effect on patients survival has not been
shown (32; 33). Downregulation of ITGB2 has been
previously associated with transition between prostatic
intraepithelial neoplasia and prostate cancer, playing a
role in cell adhesion of invasive prostate cancer cells
(34).
A second focus of interest in network 1 is NFKB. This
protein complex has been widely associated with onco-
genesis due to its ability to regulate cell proliferation and
protect cells from apoptosis (35). CRPC cell lines like DU-
145 and PC-3 exhibit constitutive activation of NFKB,
whereas its activity is low in the androgen-sensitive
LNCaP and LAPC-4 cells, which is consistent with the
role of NFKB in progression of prostate cancer. Moreover,
higher levels of NFkB protein can be further enhanced in
response to certain types of chemotherapy (36; 37). Our
group previously found that the inhibition of this complex
may be an attractive strategy to enhance docetaxel
response in PC (38). This strategy has also been clinically
tested through the use of bortezomib (39; 40).
The present study specifically explored the role of the
TGF-bsuperfamily, which is also represented at network
1. The signaling pathway derived from this family of
proteins has a principal role in growth control (41). As
a member of this family, TGFBR3 acts as a coreceptor of
TGF-bligand, being a positive or negative regulator of
TGF-bsignals depending on the cellular context (42; 43).
Recently, it has been shown that TGFBR3 may act as a
protective factor in the apoptotic process of fibroblasts by
negative regulation of TGF-bsignaling (44). Our results
further suggest that TGFB1 acts as a protective factor
against docetaxel for DU-145 and PC-3 cells and has a
partial role in the development of docetaxel resistance in
cultured cells. The specific mechanism through which
TGF-b/TGFBR3 protects DU-145 and PC-3 cells from this
drug remains to be elucidated.
The epithelial cell adhesion molecule CDH1, which is
significantly downexpressed in docetaxel-resistant cells,
is included in the second most significant network
obtained in this analysis (Fig. 1B). The loss of CDH1 in
Marín-Aguilera et al.
Mol Cancer Ther; 11(2) February 2012 Molecular Cancer Therapeutics
336
on November 5, 2015. © 2012 American Association for Cancer Research. mct.aacrjournals.org Downloaded from
Published OnlineFirst October 25, 2011; DOI: 10.1158/1535-7163.MCT-11-0289
metastatic cells has been shown in a variety of in vitro and
in vivo models, and has been related to the epithelial-
mesenchymal transition process (45–47). However, its
role in chemotherapy resistance should be further stud-
ied. The mere loss of CDH1 probably does not directly
confer chemoresistance properties to the tumor cell, but
signals may be conveyed that induce resistance to che-
motherapy. According to network 2, CDH1 directly inter-
acts with the transcription regulator ID2 (inhibitor of
DNA binding 2), which is significantly overexpressed
in resistant cells, and enzymes like OCLN, which is also
involved in cell–cell adhesion and is downexpressed in
resistant cells. Other CDH1-related genes in this network
are the phosphatase PTPRM, the plakophilin PKP2, and
the myosin MYO6.
Another interesting focal gene downexpressed in net-
work 2 is IFI16 (interferon, gamma-inducible protein 16).
The encoded protein contains domains involved in DNA
binding, transcriptional regulation, and protein–protein
interactions. It is known that this protein modulates P53
function and inhibits cell growth in the RAS/RAF signal-
ing pathway, an antitumor activity. Downexpression of
this gene in resistant cells seems to be related to their
ability to grow despite the presence of docetaxel in the
medium.
A recent study used microarray analysis to compare
the PC-3R cell line with and without docetaxel in culture
media, to identify genes responsible for the multinucle-
ated process that PC-3 cells suffer as a result of doc-
etaxel exposure. The authors also compared DU-145
with docetaxel-resistant DU-145 cells and showed a set
of 10 genes present in both PC-3R and DU-145R cells
(19). However, in contrast to our study only LAMC2,
which increases more than 10% during hormone escape
in prostate cancer (48), was represented in both models.
Methodologic factors and differences in bioinformatics
analysis between these studies could account for such
high variability of commonly deregulated genes in
docetaxel-resistant cells.
Eighteen genes were selected according to their func-
tion and degree of relative expression in resistant cells
versus parent cells. They were further validated in cell
lines and tested in docetaxel-sensitive and resistant CRPC
tumor samples (11 FFPE and 5 OCT samples) by qRT-PCR.
Seven of the 18 marker genes were deregulated in the
same way in cell lines, FFPE and OCT samples. These
included the CDH1 gene discussed above and also AREG
(amphiregulin), DLC1,GJB2 (gap junction protein, b2),
IFIH1,MX1, and EPCAM, all of them consistently down-
expressed in docetaxel-resistant tumor samples.
Discordant results were observed in the expression of
other genes such as NEAT1 (nuclear paraspeckle assem-
bly transcript 1), which was significantly downexpressed
in docetaxel-resistant tumors but was within the top 10
overexpressed genes in resistant cells. These conflicting
results could be due to the limitations of the present study.
First, gene panels derived from in vitro study cannot
represent the complex biology and heterogeneity of tumor
cells in patients. Moreover, molecular changes derived
from tumor–stroma interaction are lost in in vitro models.
On the other hand, the range of docetaxel concentrations
in cultured cells differs from docetaxel levels in patients, a
fact that also may cause differences in gene expression. Of
note, we found molecular alterations in docetaxel-resis-
tant cells lines after long-term exposure to docetaxel. It is
not clear whether genetic alterations responsible for doc-
etaxel resistance were already present in the initial cell
population or were induced after docetaxel exposure.
Future comparison of the present gene expression ana-
lysis with gene expression data from a de novo docetaxel-
resistant cell line will be useful to further elucidate
pathways involved in different resistance patterns. In
patients, docetaxel resistance may exist before drug
exposure (primary resistance) or may be developed after
a number of chemotherapy cycles (acquired resistance).
In our series, where tumor samples were obtained before
chemotherapy exposure, we observed a concordance
between some of the deregulated genes in the resistant
tumors and previously observed results in resistant cell
lines. These data suggest that basal gene expression altera-
tions, not induced by treatment, may be responsible for
the survival of cancer cells in the presence of docetaxel.
Finally, in the present work we carried out an exploratory
analysis in tumor samples that need to be further vali-
dated in a larger cohort of patients.
In summary, this exploratory analysis provides infor-
mation about potential genes and networks involved in
docetaxel resistance in CRPC, as well as a basis for the
investigation of the specific mechanism through which
the TGF-bfamily protects cultured cells from docetaxel.
The identification of docetaxel resistance genes may be
useful to select patients who may not benefit from therapy
or to develop targeted therapies to overcome docetaxel
resistance. Further clinical validation of these results is
needed in patients with CRPC.
Disclosure of Potential Conicts of Interest
No potential conflicts of interest were disclosed.
Acknowledgments
The authors thank the Tumor Bank of the Department of Pathology of
the Hospital Clı
´nic for providing the tumor samples; M
onica Marı
´n for her
excellent technical assistance; Ana Rovira for scientific advice; and Elaine
Lilly, Ph.D., for correction of the English text.
Grant Support
This study was supported by the Fondo de Investigaciones Sanitarias
(FIS 07/0388 and 08/0274) and RD07/0020/2014 grants. M. Marı
´n-Agui-
lera was supported by Cellex Foundation. Fondo de Investigaci
on Sani-
taria, Instituto de Salud Carlos III (Spain), project grants PI070388,
PI080274, and RD07/0020/2014.
The costs of publication of this article were defrayed in part by the
payment of page charges. This article must therefore be hereby marked
advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate
this fact.
Received April 18, 2011; revised September 14, 2011; accepted October
12, 2011; published OnlineFirst October 25, 2011.
Docetaxel Resistance in Castoration-Resistant Prostate Cancer
www.aacrjournals.org Mol Cancer Ther; 11(2) February 2012 337
on November 5, 2015. © 2012 American Association for Cancer Research. mct.aacrjournals.org Downloaded from
Published OnlineFirst October 25, 2011; DOI: 10.1158/1535-7163.MCT-11-0289
References
1. Kamangar F, Dores GM, Anderson WF. Patterns of cancer
incidence, mortality, and prevalence across ve continents: den-
ing priorities to reduce cancer disparities in different geographic
regions of the world. J Clin Oncol 2006;24:213750.
2. Cooperberg MR, Broering JM, Litwin MS, Lubeck DP, Mehta
SS, Henning JM, et al. The contemporary management of
prostate cancer in the United States: lessons from the can-
cer of the prostate strategic urologic research endeavor
(CapSURE), a national disease registry. J Urol 2004;171:
1393401.
3. Petrylak DP, Tangen CM, Hussain MH, Lara PN Jr, Jones JA, Taplin
ME, et al. Docetaxel and estramustine compared with mitoxantrone
and prednisone for advanced refractory prostate cancer. N Engl J Med
2004;351:151320.
4. Tannock IF, de WR, Berry WR, Horti J, Pluzanska A, Chi KN, et al.
Docetaxel plus prednisone or mitoxantrone plus prednisone for
advanced prostate cancer. N Engl J Med 2004;351:150212.
5. Eisenhauer EA, Vermorken JB. The taxoids. Comparative clinical
pharmacology and therapeutic potential. Drugs 1998;55:530.
6. Haldar S, Chintapalli J, Croce CM. Taxol induces bcl-2 phosphor-
ylation and death of prostate cancer cells. Cancer Res 1996;56:
12535.
7. Alli E, Bash-Babula J, Yang JM, Hait WN. Effect of stathmin on the
sensitivity to antimicrotubule drugs in human breast cancer. Cancer
Res 2002;62:68649.
8. Anand S, Penrhyn-Lowe S, Venkitaraman AR. AURORA-A amplica-
tion overrides the mitotic spindle assembly checkpoint, inducing
resistance to Taxol. Cancer Cell 2003;3:5162.
9. Kamath K, Wilson L, Cabral F, Jordan MA. BetaIII-tubulin induces
paclitaxel resistance in association with reduced effects on microtu-
bule dynamic instability. J Biol Chem 2005;280:129027.
10. Irizarry RA, Bolstad BM, Collin F, Cope LM, Hobbs B, Speed TP.
Summaries of Affymetrix GeneChip probe level data. Nucleic Acids
Res 2003;31:e15.
11. Gentleman RC, Carey VJ, Bates DM, Bolstad B, Dettling M, Dudoit S,
et al. Bioconductor: open software development for computational
biology and bioinformatics. Genome Biol 2004;5:R80.
12. Li C, Wong WH. Model-based analysis of oligonucleotide arrays:
expression index computation and outlier detection. Proc Natl Acad
Sci U S A 2001;98:316.
13. Smyth GK. Limma: linear models for microarray. In: Gentleman R,
Carey V, Dudoit S, Irizarry R, Huber W, editors. Bioinformatics and
computational biology solutions Using R and Bioconductor. New York:
Springer; 2005. p. 397465.
14. Ingenuity systems. Available from: http://www.ingenuity.com.
15. Bubley GJ, Carducci M, Dahut W, Dawson N, Daliani D, Eisenberger M,
et al. Eligibility and response guidelines for phase II clinical trials in
androgen-independent prostate cancer: recommendations from the
Prostate-Specic Antigen Working Group. J Clin Oncol 1999;17:
34617.
16. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford
R, et al. New response evaluation criteria in solid tumours: revised
RECIST guideline (version 1.1). Eur J Cancer 2009;45:22847.
17. Huang dW, Sherman BT, Lempicki RA. Systematic and integrative
analysis of large gene lists using DAVID bioinformatics resources. Nat
Protoc 2009;4:4457.
18. Huang dW, Sherman BT, Lempicki RA. Bioinformatics enrichment
tools: paths toward the comprehensive functional analysis of large
gene lists. Nucleic Acids Res 2009;37:113.
19. Desarnaud F, Geck P, Parkin C, Carpinito G, Makarovskiy AN. Gene
expression proling of the androgen independent prostate cancer cells
demonstrates complex mechanisms mediating resistance to doce-
taxel. Cancer Biol Ther 2011;11:20412.
20. Gan L, Wang J, Xu H, Yang X. Resistance to docetaxel-induced
apoptosis in prostate cancer cells by p38/p53/p21 signaling. Prost ate
2011;71:115866.
21. Torres K, Horwitz SB. Mechanisms of Taxol-induced cell death are
concentration dependent. Cancer Res 1998;58:36206.
22. Bignotti E, Todeschini P, Calza S, Falchetti M, Ravanini M, Tassi RA,
et al. Trop-2 overexpression as an independent marker for poor
overall survival in ovarian carcinoma patients. Eur J Cancer 2010;
46:94453.
23. Zhang Y, Scoumanne A, Chen X. G protein-coupled receptor 87: a
promising opportunity for cancer drug discovery. Mol Cell Pharmacol
2010;2:1116.
24. Subbaramaiah K, Marmo TP, Dixon DA, Dannenberg AJ. Regulation of
cyclooxgenase-2 mRNA stability by taxanes: evidence for involvement
of p38, MAPKAPK-2, and HuR. J Biol Chem 2003;278:3763747.
25. Ibragimova I, Ibanez dC I, Hoffman AM, Potapova A, Dulaimi E, Al-
Saleem T, et al. Global reactivation of epigenetically silenced genes in
prostate cancer. Cancer Prev Res (Phila) 2010;3:108492.
26. Huang Y, Anderle P, Bussey KJ, Barbacioru C, Shankavaram U, Dai Z,
et al. Membrane transporters and channels: role of the transportome in
cancer chemosensitivity and chemoresistance. Cancer Res 2004;64:
4294301.
27. Gottesman MM, Fojo T, Bates SE. Multidrug resistance in cancer:
role of ATP-dependent transporters. Nat Rev Cancer 2002;2:
4858.
28. Ranganathan S, Benetatos CA, Colarusso PJ, Dexter DW, Hudes GR.
Altered beta-tubulin isotype expression in paclitaxel-resistant human
prostate carcinoma cells. Br J Cancer 1998;77:5626.
29. Collett GP, Betts AM, Johnson MI, Pulimood AB, Cook S, Neal DE,
et al. Peroxisome proliferator-activated receptor alpha is an androgen-
responsive gene in human prostate and is highly expressed in prostatic
adenocarcinoma. Clin Cancer Res 2000;6:32418.
30. Wilson LA, Yamamoto H, Singh G. Role of the transcription factor Ets-1
in cisplatin resistance. Mol Cancer Ther 2004;3:82332.
31. Alipov G, Nakayama T, Ito M, Kawai K, Naito S, Nakashima M, et al.
Overexpression of Ets-1 proto-oncogene in latent and clinical prostatic
carcinomas. Histopathology 2005;46:2028.
32. Beer TM, Hough KM, Garzotto M, Lowe BA, Henner WD. Weekly high-
dose calcitriol and docetaxel in advanced prostate cancer. Semin
Oncol 2001;28:4955.
33. Scher HI, Chi KN, De Wit R, Berry WR, Albers P, Henick B, et al.
Docetaxel (D) plus high-dose calcitriol versus D plus prednisone (P) for
patients (Pts) with progressive castration-resistant prostate cancer
(CRPC): results from the phase III ASCENT2 trial. J Clin Oncol 28:15s,
2010 (suppl; abstr 4509).
34. Ashida S, Nakagawa H, Katagiri T, Furihata M, Iiizumi M, Anazawa
Y, et al. Molecular features of the transition from prostatic intrae-
pithelial neoplasia (PIN) to prostate cancer: genome-wide gene-
expression proles of prostate cancers and PINs. Cancer Res 2004;
64:596372.
35. Karin M. Nuclear factor-kappaB in cancer development and progres-
sion. Nature 2006;441:4316.
36. Domingo-Domenech J, Mellado B, Ferrer B, Truan D, Codony-Servat
J, Sauleda S, et al. Activation of nuclear factor-kappaB in human
prostate carcinogenesis and association to biochemical relapse. Br J
Cancer 2005;93:128594.
37. Baldwin AS. Control of oncogenesis and cancer therapy resistance by
the transcription factor NF-kappaB. J Clin Invest 2001;107:2416.
38. Domingo-Domenech J, Oliva C, Rovira A, Codony-Servat J, Bosch M,
Filella X, et al. Interleukin 6, a nuclear factor-kappaB target, predicts
resistance to docetaxel in hormone-independent prostate cancer and
nuclear factor-kappaB inhibition by PS-1145 enhances docetaxel
antitumor activity. Clin Cancer Res 2006;12:557886.
39. Dreicer R, Petrylak D, Agus D, Webb I, Roth B. Phase I/II study of
bortezomib plus docetaxel in patients with advanced androgen-inde-
pendent prostate cancer. Clin Cancer Res 2007;13:120815.
40. Hainsworth JD, Meluch AA, Spigel DR, Barton J Jr, Simons L, Meng C,
et al. Weekly docetaxel and bortezomib as rst-line treatment for
patients with hormone-refractory prostate cancer: a Minnie Pearl
Cancer Research Network phase II trial. Clin Genitourin Cancer
2007;5:27883.
41. Schuster N, Krieglstein K. Mechanisms of TGF-beta-mediated apo-
ptosis. Cell Tissue Res 2002;307:114.
Marín-Aguilera et al.
Mol Cancer Ther; 11(2) February 2012 Molecular Cancer Therapeutics
338
on November 5, 2015. © 2012 American Association for Cancer Research. mct.aacrjournals.org Downloaded from
Published OnlineFirst October 25, 2011; DOI: 10.1158/1535-7163.MCT-11-0289
42. Vilchis-Landeros MM, Montiel JL, Mendoza V, Mendoza-Hernandez
G, Lopez-Casillas F. Recombinant soluble betaglycan is a potent and
isoform-selective transforming growth factor-beta neutralizing agent.
Biochem J 2001;355:21522.
43. Blobe GC, Schiemann WP, Pepin MC, Beauchemin M, Moustakas
A, Lodish HF, et al. Functional roles for the cytoplasmic domain of
the type III transforming growth factor beta receptor in regulating
transforming growth factor beta signaling. J Biol Chem 2001;
276:2462737.
44. ChuW,LiX,LiC,WanL,ShiH,SongX,etal.TGFBR3,apotential
negative regulator of TGF-beta signaling, protects cardiac bro-
blasts from hypoxia-induced apoptosis. J Cell Physiol 2011;
226:258694.
45. Frixen UH, Behrens J, Sachs M, Eberle G, Voss B, Warda A, et al.
E-cadherin-mediated cell-cell adhesion prevents invasiveness of
human carcinoma cells. J Cell Biol 1991;113:17385.
46. Perl AK, Wilgenbus P, Dahl U, Semb H, Christofori G. A causal role for
E-cadherin in the transition from adenoma to carcinoma. Nature
1998;392:1903.
47. Lombaerts M, van WT, Philippo K, Dierssen JW, Zimmerman RM,
Oosting J, et al. E-cadherin transcriptional downregulation by promot-
er methylation but not mutation is related to epithelial-to-mesenchy-
mal transition in breast cancer cell lines. Br J Cancer 2006;94:66171.
48. Edwards J, Krishna NS, Witton CJ, Bartlett JM. Gene amplications
associated with the development of hormone-resistant prostate
cancer. Clin Cancer Res 2003;9:527181.
www.aacrjournals.org Mol Cancer Ther; 11(2) February 2012 339
Docetaxel Resistance in Castoration-Resistant Prostate Cancer
on November 5, 2015. © 2012 American Association for Cancer Research. mct.aacrjournals.org Downloaded from
Published OnlineFirst October 25, 2011; DOI: 10.1158/1535-7163.MCT-11-0289
2012;11:329-339. Published OnlineFirst October 25, 2011.Mol Cancer Ther
Mercedes Marín-Aguilera, Jordi Codony-Servat, Susana G. Kalko, et al.
Castration-Resistant Prostate Cancer
Identification of Docetaxel Resistance Genes in
Updated version
10.1158/1535-7163.MCT-11-0289doi:
Access the most recent version of this article at:
Material
Supplementary
http://mct.aacrjournals.org/content/suppl/2011/10/14/1535-7163.MCT-11-0289.DC1.html
Access the most recent supplemental material at:
Cited articles
http://mct.aacrjournals.org/content/11/2/329.full.html#ref-list-1
This article cites 45 articles, 20 of which you can access for free at:
Citing articles
http://mct.aacrjournals.org/content/11/2/329.full.html#related-urls
This article has been cited by 3 HighWire-hosted articles. Access the articles at:
E-mail alerts related to this article or journal.Sign up to receive free email-alerts
Subscriptions
Reprints and
.pubs@aacr.org
To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at
Permissions
.permissions@aacr.org
To request permission to re-use all or part of this article, contact the AACR Publications Department at
on November 5, 2015. © 2012 American Association for Cancer Research. mct.aacrjournals.org Downloaded from
Published OnlineFirst October 25, 2011; DOI: 10.1158/1535-7163.MCT-11-0289
... For WNT7B and WNT9A, WNT expression showed an inverse correlation with PHB levels. WNT10B showed an inverse correlation Interestingly, another dataset (GDS3973) [19] showed that PHB gene expression was downregulated in two docetaxel-resistant prostate cancer cell lines (Supplemental Figure S1B). PHB was also downregulated in an invasive gastric adenocarcinoma (dataset GDS4198 [20] when compared to proliferative gastric adenocarcinoma, suggesting PHB's role in cancer progression is not PC-specific (Supplemental Figure S1C). ...
... accessed on 1 February 2023), we examined the disease-free survival of the prostate cancer dataset (PRAD). Although, no significant difference was seen for PHB, higher expression of all Interestingly, another dataset (GDS3973) [19] showed that PHB gene expression was downregulated in two docetaxel-resistant prostate cancer cell lines (Supplemental Figure S1B). PHB was also downregulated in an invasive gastric adenocarcinoma (dataset GDS4198 [20] when compared to proliferative gastric adenocarcinoma, suggesting PHB's role in cancer progression is not PC-specific (Supplemental Figure S1C). ...
Article
Full-text available
Prohibitin (PHB) is a tumour suppressor gene with several different molecular activities. PHB overexpression leads to G1/S-phase cell cycle arrest, and PHB represses the androgen receptor (AR) in prostate cancer cells. PHB interacts with and represses members of the E2F family in a manner that may also be AR-linked, therefore making the AR:PHB:E2F interaction axis highly complex. PHB siRNA increased the growth and metastatic potential of LNCaP mouse xenografts in vivo. Conversely, PHB ectopic cDNA overexpression affected several hundred genes in LNCaP cells. Furthermore, gene ontology analysis showed that in addition to cell cycle regulation, several members of the WNT family were significantly downregulated (WNT7B, WNT9A and WNT10B), as well as pathways for cell adhesion. Online GEO data studies showed PHB expression to be decreased in clinical cases of metastatic prostate cancer, and to be correlated with higher WNT expression in metastasis. PHB overexpression reduced prostate cancer cell migration and motility in wound-healing assays, reduced cell invasion through a Matrigel layer and reduced cellular attachment. In LNCaP cells, WNT7B, WNT9A and WNT10B expression were also upregulated by androgen treatment and downregulated by androgen antagonism, indicating a role for AR in the control of these WNT genes. However, these WNTs were strongly cell cycle regulated. E2F1 cDNA ectopic expression and PHB siRNA (both cell cycle promoting effects) increased WNT7B, WNT9A and WNT10B expression, and these genes were also upregulated as cells were released from G1 to S phase synchronisation, indicating further cell cycle regulation. Therefore, the repressive effects of PHB may inhibit AR, E2F and WNT expression and its loss may increase metastatic potential in human prostate cancer.
... Based on a combination of genetic alterations, gene expression patterns, and methylation profiles, precision targeted therapy might serve as a promising strategy to balance racial disparities in the mCRPC setting [10]. One study showed that TGF-b expression, which differs by race, modulated taxane and docetaxel sensitivity in PCa cells [32]. Moreover, SPHKAP/SPHK1 was identified as a predictor of clinical benefit based on ancestry, and modulated the efficacy of chemotherapy and radiotherapy and influenced the proliferation of tumor cells [18,33,34]. ...
Article
Full-text available
Context Data on racial disparities among patients with metastatic castration-resistant prostate cancer (mCRPC) are limited and there is no uniform conclusion on differences by race in this setting. Objective To provide the latest evidence on racial disparities in survival outcomes between Black and White patients receiving systemic therapies for mCRPC. Evidence acquisition Our study was performed according to the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guidelines. We systematically searched the PubMed, Web of Science, and Cochrane Library databases up to September 2023 to identify potentially relevant studies. Overall survival (OS) and progression-free survival (PFS) were the outcomes of interest. Pooled hazard ratios (HRs) with 95% confidence intervals (CIs) were evaluated. Evidence synthesis Nine studies involving 9462 patients with mCRPC (2058 Black and 7404 White men) met the eligibility criteria and were included. Pooled estimates demonstrated significantly better OS for Black than for White men (HR 0.75, 95% CI 0.70–0.80; p < 0.0001). The results were similar in a subgroup of men receiving androgen receptor–targeted therapies (HR 0.72, 95% CI 0.66–0.78; p < 0.0001) and a subgroup of men receiving other treatments (HR 0.79, 95% CI 0.71–0.88; p < 0.0001). Likewise, significantly favorable PFS was observed for Black men receiving ARTs in comparison to their White counterparts (HR 0.84, 95% CI 0.71–0.99; p = 0.0373). Conclusions Overall, our meta-analysis of survival outcomes for men with mCRPC stratified by race revealed a significant survival benefit for Black men in comparison to their White counterparts, regardless of systemic therapeutic agent. Patient summary Both biological and nonbiological factors could account for racial differences in the efficacy of systemic treatments for metastatic prostate cancer that is resistant to hormone therapy. Our review provides the latest reliable evidence showing better survival outcomes for Black than for White men. The results will be helpful in further understanding the molecular mechanisms that might explain racial differences in this disease stage and in planning treatment.
... Docetaxel-resistant DU145-DR and PC3-DR cell lines had previously been generated from the PTEN-wild-type (PTEN-wt) DU145 (RRID: CVCL_0105) and the PTEN-loss PC3 (RRID: CVCL_0035) human mCRPC cell lines, respectively (Marin-Aguilera et al., 2012). Docetaxel IC50 doses were previously determined (Ruiz de Porras et al., 2021b) and are shown in Supplementary Table S1. ...
Article
Full-text available
Background: Docetaxel remains the standard treatment for metastatic castration-resistant prostate cancer (mCRPC). However, resistance frequently emerges as a result of hyperactivation of the PI3K/AKT and the MEK/ERK pathways. Therefore, the inhibition of these pathways presents a potential therapeutic approach. In this study, we evaluated the efficacy of simultaneous inhibition of the PI3K/AKT and MEK/ERK pathways in docetaxel-resistant mCRPC, both in vitro and in vivo. Methods: Docetaxel-sensitive and docetaxel-resistant mCRPC cells were treated with selumetinib (MEK1/2 inhibitor), AZD8186 (PI3Kβ/δ inhibitor) and capivasertib (pan-AKT inhibitor) alone and in combination. Efficacy and toxicity of selumetinib+AZD8186 were tested in docetaxel-resistant xenograft mice. CRISPR-Cas9 generated a PTEN-knockdown docetaxel-resistant cell model. Changes in phosphorylation of AKT, ERK and downstream targets were analyzed by Western blot. Antiapoptotic adaptations after treatments were detected by dynamic BH3 profiling. Results: PI3K/AKT and MEK/ERK pathways were hyperactivated in PTEN-wild-type (wt) docetaxel-resistant cells. Selumetinib+AZD8186 decreased cell proliferation and increased apoptosis in PTEN-wt docetaxel-resistant cells. This observation was further confirmed in vivo, where docetaxel-resistant xenograft mice treated with selumetinib+AZD8186 exhibited reduced tumor growth without additional toxicity. Conclusion: Our findings on the activity of selumetinib+AZD8186 in PTEN-wt cells and in docetaxel-resistant xenograft mice provide an excellent rationale for a novel therapeutic strategy for PTEN-wt mCRPC patients resistant to docetaxel, in whom, unlike PTEN-loss patients, a clinical benefit of treatment with single-agent PI3K and AKT inhibitors has not been demonstrated. A phase I-II trial of this promising combination is warranted.
... The design ideas of this study can be seen in Fig. 1. In this study, the genesets bioinformatics analysis of prostate cancer docetaxel-resistant (GSE33455 [23] and GSE36135 [24]) and enzalutamide-resistant (GSE78201 [25], GSE104935 [26] and GSE143408 [27]) are performed to identify potential target genes. The study is expected to provide insight into the molecular mechanisms of drug resistance in prostate cancer and enable the exploration of its prognostic biomarkers and potential therapeutic targets for drug resistance. ...
Article
Full-text available
Background Prostate cancer is currently the second most lethal malignancy in men worldwide due to metastasis and invasion in advanced stages. Studies have revealed that androgen deprivation therapy can induce stable remission in patients with advanced prostate cancer, although most patients will develop castration-resistant prostate cancer (CRPC) in 1–2 years. Docetaxel and enzalutamide improve survival in patients with CRPC, although only for a short time, eventually patients develop primary or secondary resistance, causing disease progression or biochemical relapse. Methods The gene expression profiles of docetaxel-sensitive or -resistant prostate cancer cell lines, namely GSE33455, GSE36135, GSE78201, GSE104935, and GSE143408, were sequentially analyzed for differentially expressed genes and progress-free interval significance. Subsequently, the overall survival significance and clinic-pathological features were analyzed by the R package. The implications of hub genes mutations, methylation in prostate cancer and the relationship with the tumor immune cell infiltration microenvironment were assessed with the help of cBioPortal, UALCAN and TISIDB web resources. Finally, effects of the hub genes on the progression and drug resistance in prostate cancer were explored using reverse transcription-polymerase chain reaction (RT-PCR), immunohistochemistry, cell phenotype, and drug sensitivity. Result Glutamate decarboxylase 1 (GAD1) was tentatively identified by bioinformatic analysis as an hub gene for the development of drug resistance, including docetaxel and enzalutamide, in prostate cancer. Additionally, GAD1 expression, mutation and methylation were significantly correlated with the clinicopathological features and the tumor immune microenvironment. RT-PCR, immunohistochemistry, cell phenotype and drug sensitivity experiments further demonstrated that GAD1 promoted prostate cancer progression and decreased the therapeutic effect of docetaxel or enzalutamide. Conclusion This research confirmed that GAD1 was a hub gene in the progression and development of drug resistance in prostate cancer. This helped to explain prostate cancer drug resistance and provides new immune-related therapeutic targets and biomarkers for it.
... It has also been reported that EMT is related to drug resistance [49]. Docetaxel is the main chemotherapy drug for patients with metastatic castration-resistant PCa [50]; thus, we next compared the docetaxel sensitivity of hybrid cells and parental RM1 cells in vitro. The IC 50 values of docetaxel for hybrid and RM1 cells were determined by cytotoxicity assay based on the CCK8 cell proliferation assay following drug treatment for 48 h. ...
Article
Full-text available
Background Bone metastasis is the leading cause of death in patients with prostate cancer (PCa) and currently has no effective treatment. Disseminated tumor cells in bone marrow often obtain new characteristics to cause therapy resistance and tumor recurrence. Thus, understanding the status of disseminated prostate cancer cells in bone marrow is crucial for developing a new treatment. Methods We analyzed the transcriptome of disseminated tumor cells from a single cell RNA-sequencing data of PCa bone metastases. We built a bone metastasis model through caudal artery injection of tumor cells, and sorted the tumor hybrid cells by flow cytometry. We performed multi-omics analysis, including transcriptomic, proteomic and phosphoproteomic analysis, to compare the difference between the tumor hybrid cells and parental cells. In vivo experiments were performed to analyze the tumor growth rate, metastatic and tumorigenic potential, drug and radiation sensitivity in hybrid cells. Single cell RNA-sequencing and CyTOF were performed to analyze the impact of hybrid cells on tumor microenvironment. Results Here, we identified a unique cluster of cancer cells in PCa bone metastases, which expressed myeloid cell markers and showed a significant change in pathways related to immune regulation and tumor progression. We found that cell fusion between disseminated tumor cells and bone marrow cells can be source of these myeloid-like tumor cells. Multi-omics showed the pathways related to cell adhesion and proliferation, such as focal adhesion, tight junction, DNA replication, and cell cycle, were most significantly changed in these hybrid cells. In vivo experiment showed hybrid cells had a significantly increased proliferative rate, and metastatic potential. Single cell RNA-sequencing and CyTOF showed tumor-associated neutrophils/monocytes/macrophages were highly enriched in hybrid cells-induced tumor microenvironment with a higher immunosuppressive capacity. Otherwise, the hybrid cells showed an enhanced EMT phenotype with higher tumorigenicity, and were resistant to docetaxel and ferroptosis, but sensitive to radiotherapy. Conclusion Taken together, our data demonstrate that spontaneous cell fusion in bone marrow can generate myeloid-like tumor hybrid cells that promote the progression of bone metastasis, and these unique population of disseminated tumor cells can provide a potential therapeutic target for PCa bone metastasis.
... uk/) [21]. Subsequently, the microarray data of DR and docetaxel-sensitive (DS) DU-145 cell lines as well as DR and DS PC3 cell lines were obtained from GSE33455 [22]. GEO2R, an online R programming language-based program for microarray data analysis, was utilized to process the data and identify the differentially expressed genes (DEGs) using the default settings. ...
Article
Full-text available
Background: Prostate cancer (PC) is a silent but potent killer among men. In 2018, PC accounted for more than 350, 000 death cases while more than 1.2 million cases were diagnosed. Docetaxel, a chemotherapeutic drug belonging to the taxane family of drugs, is one of the most potent drugs in combating advanced PC. However, PC cells often evolve resistance against the regimen. Hence, necessitating the search for complementary and alternative therapies. Quercetin, a ubiquitous phytocompound with numerous pharmacological properties, has been reported to reverse docetaxel resistance (DR) in docetaxel-resistant prostate cancer (DRPC). Therefore, this study aimed to explore the mechanism via which quercetin reverses DR in DRPC using an integrative functional network and exploratory cancer genomic data analyses. Results: The putative targets of quercetin were retrieved from relevant databases, while the differentially expressed genes (DEGs) in docetaxel-resistant prostate cancer (DRPC) were identified by analysing microarray data retrieved from the Gene Expression Omnibus (GEO) database. Subsequently, the protein-protein interaction (PPI) network of the overlapping genes between the DEGs and quercetin targets was retrieved from STRING, while the hub genes, which represent the key interacting genes of the network, were identified using the CytoHubba plug-in of Cytoscape. The hub genes were further subjected to a comprehensive analysis aimed at identifying their contribution to the immune microenvironment and overall survival (OS) of PC patients, while their alterations in PC patients were also revealed. The biological roles played by the hub genes in chemotherapeutic resistance include the positive regulation of developmental process, positive regulation of gene expression, negative regulation of cell death, and epithelial cell differentiation among others. Conclusion: Further analysis revealed epidermal growth factor receptor (EGFR) as the most pertinent target of quercetin in reversing DR in DRPC, while molecular docking simulation revealed an effective interaction between quercetin and EGFR. Ultimately, this study provides a scientific rationale for the further exploration of quercetin as a combinational therapy with docetaxel.
Article
Full-text available
Aim: Docetaxel is a microtubule-stabilizing drug used for the treatment of several cancers, including prostate cancer. Resistance to docetaxel can either occur through intrinsic resistance or develop under therapeutic pressure, i.e., acquired resistance. A possible explanation for the occurrence of acquired resistance to docetaxel is increased drug efflux via P-glycoprotein (P-gp) drug transporters. Methods: We have generated docetaxel-resistant cell lines DU-145DOC10 and 22Rv1DOC8 by exposing parental cell lines DU-145DOC and 22Rv1 to increasing levels of docetaxel. Gene expression levels between DU-145DOC10 and 22Rv1DOC8 were compared with those of their respective originator cell lines. Both parental and resistant cell lines were treated with the taxane drugs docetaxel and cabazitaxel in combination with the P-gp/CYP3A4 inhibitor ritonavir and the P-gp inhibitor elacridar. Results: In the docetaxel-resistant cell lines DU-145DOC10 and 22Rv1DOC8, the ABCB1 (P-gp) gene was highly up-regulated. Expression of the P-gp protein was also significantly increased in the docetaxel-resistant cell lines in a Western blotting assay. The addition of ritonavir to docetaxel resulted in a return of the sensitivity to docetaxel in the DU-145DOC10 and 22Rv1DOC8 to a level similar to the sensitivity in the originator cells. We found that these docetaxel-resistant cell lines could also be re-sensitized to cabazitaxel in a similar manner. In a Caco-2 P-gp transporter assay, functional inhibition of P-gp-mediated transport of docetaxel with ritonavir was demonstrated. Conclusion: Our results demonstrate that ritonavir restores sensitivity to both docetaxel and cabazitaxel in docetaxel-resistant cell lines, most likely by inhibiting P-gp-mediated drug efflux.
Article
Full-text available
Prostate cancer (PoC) is currently one of the most frequent cancers in males. Though a lot of work has been done on it, the biomarkers in the progression of prostate cancer have not been fully identified. In this research, we used the GSE70466 and GSE104131 for RNA-Seq gene expression datasets to reveal the important biomarkers and related pathways of prostate cancer. Furthermore, Differentially Expressed Genes (DEGs) were extracted from the dataset with the R statistical language tools, and to investigate the functional enrichment of the DEGs, an online repository Enrichr was used. Then, another database, STRING was used in this work to generate the protein-protein interaction (PPI) network and represented the network using the well-known Cytoscape software. After that, a Cystoscape plug-in MCODE identified clusters from the PPI network. In the end, some drug compounds that may be useful in the therapy of PoC have been revealed. A total of 83 common DEGs (43 upregulated and 40 downregulated) were exposed using some statistical criteria. Overexpressed DEGs were engaged in the metabolism of glutamine family amino acids and alpha-amino acids, while underexpressed DEGs were mostly involved in the regulation of the transmembrane receptor protein serine/threonine kinase signaling pathway and positive regulation of potassium ion transport. In addition, the overexpressed DEGs were highly involved with Arginine and Proline metabolism, whereas the underexpressed DEGs were connected with Proteoglycans in cancer, according to the KEGG pathway analysis. A total of 154 nodes and 2068 connections were used to construct the PPI. Using the connectivity method, we conclude 20 genes (INS; VEGFA; CDH1; IGF1; ITGB1; IGF1R; CAV1; ESR1; KDR; TGFB1; CD44; MMP9; PIK3R1; SHC1; IGF2; SMAD4; TGFBR1; ENG; IRS1; VWF) as hub genes. Finally, we identified and validated two important biomarkers as well as certain pharmacological compounds that could be effective in the therapy of PoC.
Article
Full-text available
DAVID bioinformatics resources consists of an integrated biological knowledgebase and analytic tools aimed at systematically extracting biological meaning from large gene/protein lists. This protocol explains how to use DAVID, a high-throughput and integrated data-mining environment, to analyze gene lists derived from high-throughput genomic experiments. The procedure first requires uploading a gene list containing any number of common gene identifiers followed by analysis using one or more text and pathway-mining tools such as gene functional classification, functional annotation chart or clustering and functional annotation table. By following this protocol, investigators are able to gain an in-depth understanding of the biological themes in lists of genes that are enriched in genome-scale studies.
Article
Full-text available
Development of malignant tumours is in part characterized by the ability of a tumour cell to overcome cell-cell adhesion and to invade surrounding tissue. E-cadherin is the main adhesion molecule of epithelia, and it has been implicated in carcinogenesis because it is frequently lost in human epithelial cancers. Re-establishing the functional cadherin complex in tumour cell lines results in a reversion from an invasive to a benign epithelial phenotype. However, it remained unresolved whether the loss of E-cadherin-mediated cell adhesion was a cause or a consequence of tumour progression in vivo. Here we report that the loss of E-cadherin expression coincides with the transition from well differentiated adenoma to invasive carcinoma in a transgenic mouse model of pancreatic beta-cell carcinogenesis (Rip1Tag2). Intercrossing Rip1Tag2 mice with transgenic mice that maintain E-cadherin expression in beta-tumour cells results in arrest of tumour development at the adenoma stage, whereas expression of a dominant-negative form of E-cadherin induces early invasion and metastasis. The results demonstrate that loss of E-cadherin-mediated cell adhesion is one rate-limiting step in the progression from adenoma to carcinoma.
Article
High density oligonucleotide array technology is widely used in many areas of biomedical research for quantitative and highly parallel measurements of gene expression. Affymetrix GeneChip arrays are the most popular. In this technology each gene is typically represented by a set of 11–20 pairs of probes. In order to obtain expression measures it is necessary to summarize the probe level data. Using two extensive spike‐in studies and a dilution study, we developed a set of tools for assessing the effectiveness of expression measures. We found that the performance of the current version of the default expression measure provided by Affymetrix Microarray Suite can be significantly improved by the use of probe level summaries derived from empirically motivated statistical models. In particular, improvements in the ability to detect differentially expressed genes are demonstrated.
Article
Paclitaxel and docetaxel are 2 compounds from the new taxoid class of anti-cancer agents. Both drugs are very similar in preclinical activity, mechanism of action and spectrum of clinical activity. Some subtle differences in the intracellular retention of docetaxel may account for its lack of schedule-related myelosuppression and greater potency, and may be relevant to the skin toxicity and oedema which it produces. Early data suggest that there may be differing behaviour of anthracycline/taxoid combinations with respect to cardiotoxicity. Paclitaxel has been studied in several first-line combination therapy trials in ovarian cancer. Here, paclitaxel in combination with a platinum compound seems to have proven itself as a standard regimen. It is uncertain if docetaxel will be evaluated in this context. An abundance of clinical data is available for both analogues in the advanced, metastatic setting of breast cancer. Both also have been compared as single agents with doxorubicin with the results suggesting paclitaxel in a 3-hour infusion is inferior to the anthracycline (in terms of response rate), and those of docetaxel suggesting it is superior to the same dose of doxorubicin. This indirect comparison favours the activity of docetaxel; however, it is clear that in the dose/schedules studied, the taxoid compounds are not equitoxic. Either agent by itself, in the treatment of metastatic breast cancer, remains appropriate; however, lack of cumulative toxicity may make paclitaxel more attractive in some situations where prolonged administration is foreseen. Lung cancer trials have also confirmed the activity of both agents, although docetaxel appears to have slightly more promising activity in previously treated patients than paclitaxel. Paclitaxel in combination with cisplatin has been evaluated in randomised trials as first-line treatment of non-small-cell lung cancer (NSCLC). The results of these trials taken together suggest that this combination has an impact on survival similar to other new regimens now considered ‘standard’ in the front-line setting in this disease. Unfortunately, despite all the phase II data generated in numerous tumour types, little else can be said about the role of either taxoid in the ‘standard’ management of malignant disease. It will be some years yet before taxoid-based combinations have been evaluated sufficiently in randomised trials such that the impact of this novel class can be adequately assessed in terms of survival and cure rates.
Chapter
A survey is given of differential expression analyses using the linear modeling features of the limma package. The chapter starts with the simplest replicated designs and progresses through experiments with two or more groups, direct designs, factorial designs and time course experiments. Experiments with technical as well as biological replication are considered. Empirical Bayes test statistics are explained. The use of quality weights, adaptive background correction and control spots in conjunction with linear modelling is illustrated on the β7 data.
Article
A lot of evidence indicates that cardiac fibroblasts are essential for maintaining the structure and function of heart. The present study examined whether TGFBR3 (transforming growth factor type III receptor, also known as betaglycan) could prevent hypoxia-induced injury in neonatal mice cardiac fibroblasts, if so, its possible molecular targets. MTT, electron microscopy and TUNEL assay were used to identify cell viability and apoptosis in neonatal mice cardiac fibroblasts. Results showed that hypoxia for 24 h markedly reduce cell viability by 49.8 ± 8.9%, largely via apoptosis. However, hypoxia-induced apoptosis in cardiac fibroblasts were almost completely prevented by overexpression of TGFBR3. In the present study, hypoxia also induced TGF-β1, p-Smad2/3 expression, TGFBR1-TGFBR2 complex formation and collagen production in cardiac fibroblasts, which were attenuated substantially by TGFBR3 overexpression. TGFBR3 also reversed Bax up-regulation, Bcl-2 down-regulation and Caspase-3 activation induced by hypoxia in cardiac fibroblasts. Hypoxia or TGF-β1 itself triggered an increase of [Ca(2+) ](i) in cardiac fibroblasts, which were both inhibited by TGFBR3 overexpression. Taken together, our results indicate that TGFBR3 may act as a protective factor in apoptotic process of cardiac fibroblasts by negative regulation of TGF-β signaling and represent a potential therapeutic target for heart remodeling after hypoxia injury.