ArticlePDF Available

CtIP Silencing as a Novel Mechanism of Tamoxifen Resistance in Breast Cancer

Authors:

Abstract and Figures

Acquired resistance to the antiestrogen tamoxifen constitutes a major clinical challenge in breast cancer therapy. However, the mechanisms involved are still poorly understood. Using serial analysis of gene expression, we identified CtIP, a BRCA1- and CtBP-interacting protein, as one of the most significantly down-regulated transcripts in estrogen receptor alpha-positive (ER+) MCF-7 tamoxifen-resistant breast cancer cells. We further confirmed the association of CtIP down-regulation with tamoxifen resistance in an additional ER+ breast cancer line (T47D), strengthening the relevance of the phenomenon observed. In additional studies, we found CtIP protein expression in a majority of ER+ breast cancer cell lines that we tested, but no or very little CtIP expression in ER-negative lines. Furthermore, CtIP protein expression status correlates with clinical response to neoadjuvant endocrine therapy, and patients with progressive disease express significantly lower CtIP protein in their primary breast carcinomas than those who respond. Meta-analysis of seven publicly available gene expression microarray data sets showed that CtIP expression is significantly associated with ER, disease-free survival, and breast cancer metastasis status. Importantly, we found that silencing endogenous CtIP in tamoxifen-sensitive breast cancer cells confers tamoxifen resistance. On the other hand, reexpression of CtIP in tamoxifen-resistant breast cancer cells restores sensitivity to the inhibitory growth effects of tamoxifen. Together, our findings indicate that CtIP silencing might be a novel mechanism for the development of tamoxifen resistance in breast cancer, suggesting that CtIP is likely associated with ER function, and that CtIP gene and protein expression may be useful biomarkers for breast cancer prognosis and clinical management.
Content may be subject to copyright.
CtIP Silencing as a Novel Mechanism of Tamoxifen
Resistance in Breast Cancer
Minhao Wu,1David Ramos Soler,2Martin C. Abba,1Maria I. Nunez,1
Richard Baer,3Christos Hatzis,4Antonio Llombart-Cussac,5
Antonio Llombart-Bosch,2and C. Marcelo Aldaz1
1
Department of Carcinogenesis, The University of Texas M. D. Anderson Cancer Center, Smithville,
Texas;
2
Department of Pathology, Medical School, The University of Valencia, Valencia, Spain;
3
Institute for Cancer Genetics, Columbia University Medical Center, New York, New York;
4
Nuvera Biosciences, Inc., Woburn, Massachusetts; and
5
Department of Oncology,
Fundacio´n Instituto Valenciano de Oncologı´a, Valencia, Spain
Abstract
Acquired resistance to the antiestrogen tamoxifen
constitutes a major clinical challenge in breast cancer
therapy. However, the mechanisms involved are still
poorly understood. Using serial analysis of gene
expression, we identified CtIP, a BRCA1- and CtBP-
interacting protein, as one of the most significantly
down-regulated transcripts in estrogen receptor
Apositive (ER+) MCF-7 tamoxifen-resistant breast
cancer cells. We further confirmed the association of
CtIP down-regulation with tamoxifen resistance in an
additional ER+ breast cancer line (T47D), strengthening
the relevance of the phenomenon observed. In
additional studies, we found CtIP protein expression in a
majority of ER+ breast cancer cell lines that we tested,
but no or very little CtIP expression in ER-negative lines.
Furthermore, CtIP protein expression status correlates
with clinical response to neoadjuvant endocrine therapy,
and patients with progressive disease express
significantly lower CtIP protein in their primary breast
carcinomas than those who respond. Meta-analysis of
seven publicly available gene expression microarray
data sets showed that CtIP expression is significantly
associated with ER, disease-free survival, and breast
cancer metastasis status. Importantly, we found that
silencing endogenous CtIP in tamoxifen-sensitive
breast cancer cells confers tamoxifen resistance. On the
other hand, reexpression of CtIP in tamoxifen-resistant
breast cancer cells restores sensitivity to the inhibitory
growth effects of tamoxifen. Together, our findings
indicate that CtIP silencing might be a novel mechanism
for the development of tamoxifen resistance in
breast cancer, suggesting that CtIP is likely associated
with ER function, and that CtIP gene and protein
expression may be useful biomarkers for breast cancer
prognosis and clinical management. (Mol Cancer Res
2007;5(12):1285 95)
Introduction
Estrogen plays a pivotal role in the etiology and progression
of human breast cancer. Therefore, for a long time, treatment of
breast cancer has been directed toward inhibiting the tumor-
promoting effects of estrogen. Tamoxifen, a nonsteroidal
antiestrogen, has been the gold standard for endocrine treatment
of all stages of estrogen receptor a(ER) positive breast cancer
for >25 years and was the first approved drug by the Food and
Drug Administration as a cancer chemopreventive agent for
reducing breast cancer incidence in both premenopausal and
postmenopausal women at high risk of breast cancer develop-
ment (1). As adjuvant therapy, tamoxifen reduces the risk of
recurrence and improves overall survival in early breast cancer
(2). It was also shown to be effective for patients with untreated
metastatic breast cancer (2). Despite the benefits of tamoxifen
in treating breast cancer, unfortunately, many tumors that
initially respond to tamoxifen therapy develop resistance. This
phenomenon has become a serious obstacle in breast cancer
treatment. In the clinic, almost all patients with advanced
metastatic disease and as many as 40% of patients receiving
adjuvant tamoxifen eventually relapse and die from their
disease (3). The mechanisms involved in the development of
tamoxifen resistance are still poorly understood. Numerous
mechanisms have been proposed to contribute to the develop-
ment of tamoxifen resistance, including altered drug metabo-
lism, loss of expression or mutation of ER, lack of expression of
progesterone receptor, increased expression of ERh, posttrans-
lational modifications of ER, altered expression of coregulators
(e.g., increased expression of the coactivator AIB1 or decreased
expression of the corepressor NCoR), and increased growth
factor signaling (e.g., HER2 signaling pathways; reviewed in
refs. 3-9). Interestingly, a recent study showed that DIBA, an
ER zinc finger inhibitor, restores the antagonistic action of
tamoxifen in tamoxifen-resistant breast cancer cells through
targeted disruption of the ER DNA-binding domain and its
interaction with the proximal NH
2
-terminal domain to suppress
ligand-dependent and ligand-independent ER transcription and
Received 3/15/07; revised 7/23/07; accepted 8/15/07.
Grant support: Susan G. Komen Breast Cancer Foundation Award, BC-
TR0402877 (C.M. Aldaz), and Department of Defense Concept Award,
W81XWH-04-1-0622-01 (C.M. Aldaz).
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.
Requests for reprints: C. Marcelo Aldaz, Department of Carcinogenesis, The
University of Texas M. D. Anderson Cancer Center, Science Park Research
Division, 1808 Park Road 1C, P.O. Box 389, Smithville, TX 78957. Phone: 512-
237-2403; Fax: 512-237-2475. E-mail: maaldaz@mdanderson.org
Copyright D2007 American Association for Cancer Research.
doi:10.1158/1541-7786.MCR-07-0126
Mol Cancer Res 2007;5(12). December 2007 1285
influence the recruitment of cofactor to the ER (10). These
findings strengthen the important role of ER in the development
of tamoxifen resistance and suggest a possible new approach
in modifying tamoxifen resistance (10). However, much work is
still needed to learn whether some of the postulated mechanisms
thus far can explain resistance to tamoxifen therapy in a majority
of patients, or simply each of the enumerated possibilities
account for minor portions of resistant cases. Thus, studies
geared at better understanding the most common mechanisms
involved in tamoxifen resistance are of considerable clinical
significance.
With the aim of identifying key genes involved in the
development of tamoxifen resistance, we defined the global
gene expression profiles of recently developed tamoxifen-
resistant MCF-7 breast cancer cell lines and compared them
with their tamoxifen-sensitive parental MCF-7 counterpart by
using serial analysis of gene expression (SAGE). We determined
that the mRNA expression of CtIP, a BRCA1- and CtBP-
interacting protein, was found to be 15-fold down-regulated in
the tamoxifen-resistant cells when compared with their
tamoxifen-sensitive counterparts. In this report, we describe
these observations and show the functional involvement of CtIP
in the development of tamoxifen resistance.
Results
CtIP Expression Is Significantly Down-Regulated in
Tamoxifen-Resistant Breast Cancer Cells
To identify genes implicated in the development of
tamoxifen resistance, we determined global gene expression
profiles of two independently derived isogenic MCF-7 breast
cancer cell line variants (TAMR1 and TAMR2) that are
resistant to tamoxifen and their parental tamoxifen-sensitive
MCF-7 line by using SAGE. As shown in Fig. 1A, in contrast
to their parental MCF-7 cells, both TAMR1 and TAMR2 cells
cultured in estrogen-depleted medium proliferated in the
presence of tamoxifen. Additionally, despite continuous
exposure to trans -4-hydroxytamoxifen (4-OH-TAM), both
tamoxifen-resistant variants are still estrogen responsive
(Fig. 1B) and express equivalent levels of ER protein as their
parental MCF-7 cells (Fig. 1D). Therefore, both TAMR1 and
TAMR2 cells express functional ER, as do their parental
MCF-7 cells. The tamoxifen-resistant phenotypes of these cells
seem not to be a consequence of changes in ER expression or
function. These data are also consistent with the clinical
findings that the majority of patients with acquired resistance to
tamoxifen still express functional ERs (11, 12).
By SAGE comparative analyses, we identified that the
transcript encoding for CtIP (also known as retinoblastoma
binding protein 8, RBBP8) was significantly down-regulated
(15-fold) in both tamoxifen-resistant cell lines when compared
with their tamoxifen-sensitive parental MCF-7 counterpart
(Fig. 1C). Real-time reverse transcription-PCR on the same
RNA samples used for SAGE analysis also confirmed
significantly lower CtIP mRNA expression in the tamoxifen-
resistant cells than in the tamoxifen-sensitive parental MCF-7
cells (data not shown). Next, we determined whether the
differential expression of CtIP detected at the mRNA level
could also be observed at the protein level. By Western blot
analysis, we detected CtIP protein expression in the tamoxifen-
sensitive parental MCF-7 line, but not in the two tamoxifen-
resistant lines (Fig. 1D). In fact, both TAMR1 and TAMR2 cells
seem to express little or no CtIP protein.
To determine whether this was a phenomenon exclusive to
MCF-7 cells, we selected another estrogen-responsive and
tamoxifen-sensitive breast cancer cell line, T-47-D, and deve-
loped a T-47-D/TR variant that is resistant to tamoxifen. To this
end, we followed the same procedure as with MCF-7 cells,
growing parental tamoxifen-sensitive T-47-D cells under chronic
exposure to 4-OH-TAM (1 Amol/L). After an initial growth arrest
by tamoxifen, T-47-D cells regained exponential growth ability
even in the presence of tamoxifen. As seen in Fig. 1E, after
being cultured in tamoxifen for over 3 months, T-47-D/TR cells
proliferated in fetal bovine serum supplemented growth
medium even in the presence of tamoxifen, whereas the growth
of parental cells was significantly inhibited by tamoxifen.
FIGURE 1. CtIP expression in tamoxifen-sensitive and tamoxifen-
resistant breast cancer cells. Aand B. In vitro characterization of parental
MCF-7, TAMR1, and TAMR2 cells. Growth curves of parental MCF-7,
TAMR1, or TAMR2 cells cultured in the presence of 10 nmol/L 4-OH-TAM
(A) or 10 nmol/L E
2
(B). Points, mean of triplicate determinations; bars, SE.
C. CtIP expression levels in parental MCF-7, TAMR1, and TAMR2 cells as
determined by SAGE (tag frequencies, normalized to 200,000 tags per
library). D. CtIP and ER protein levels were determined by immunoblotting
in cell lines as indicated. h-Actin was used as a loading control. E. Parental
T-47-D and T-47-D tamoxifen-resistant cells (T-47-D/TR) were grown in full
serum medium and treated with 4-OH-TAM (1 Amol/L). Cell numbers were
determined at various time points as indicated. Points, mean; bars, SE.
F. Western blotting for CtIP in parental T-47-D and T-47-D cultured in
tamoxifen for 1, 2, and 3 mo.
Wu et al.
Mol Cancer Res 2007;5(12). December 2007
1286
Furthermore, we determined CtIP protein levels in these T-47-D
cells cultured in tamoxifen for 1, 2, and 3 months and compared
them with parental T-47-D cells. We observed that consistent
with the findings obtained from MCF-7 cells, T-47-D cells also
have significantly decreased CtIP protein expression as they
became tamoxifen resistant (Fig. 1F).
Together, the above data confirm that the expression of CtIP
is significantly decreased in tamoxifen-resistant cells at both
mRNA and protein levels and raise the possibility that CtIP
silencing could be a novel mechanism for the development of
tamoxifen resistance.
CtIP Expression in Various Human Breast Cancer Cell
Lines
To perform a comparative analysis of CtIP expression levels,
we next determined CtIP protein expression by Western blot
analysis in 10 human breast cancer lines. As can be observed in
Fig. 2, three of six ER-positive (ER+) breast cancer lines
express abundant CtIP (MCF-7, T-47-D, and ZR-75-1) and one
of these lines (BT-483) expresses some CtIP; that is, in total,
four of six (67%) ER+ cell lines express detectable CtIP. In
contrast, four of four (100%) ER-negative (ER) breast cancer
lines either do not express CtIP at all or very little (UAC 812,
MDA-MB-231, MDA-MB-435, SKBR3).
Poor Clinical Response to Endocrine Therapy Is Associ-
ated with CtIP Deficiency in Breast Cancer Patients
To determine whether there is a relationship between
CtIP status and endocrine therapy response in vivo, we evaluated
CtIP expression by immunohistochemistry in 59 ER+, non-
operable primary breast carcinomas from patients who received
endocrine therapy as single neoadjuvant therapy. Immunohisto-
chemistry was done of samples from tumors before initiation of
antiestrogen therapy. Based on the clinical response to the
therapy after 4 months of follow-up, patients were classified into
four groups: complete response, 4 cases (7%); partial response,
32 cases (54%); stable disease, 17 cases (29%), and progressive
disease, 6 cases (10%). These cases are representative of a larger
cohort previously reported in which complete response was
reported to be between 4% and 10% (13). Immunoreactive
scores (IRS) for CtIP were used to semiquantify immunohisto-
chemical staining intensity and percentage of positive cells
(14, 15). IRS ranging from 0 to 12 represents CtIP protein
staining from undetectable to the highest expression level,
respectively (Fig. 3A). One-way ANOVA analysis of CtIP IRS
revealed significantly different CtIP expression in the four
response groups (P= 0.01). Remarkably, we observed that
patients who had the worst response to endocrine therapy
(defined as progressive disease) had significantly lower CtIP
expression than those who had the best response to endocrine
therapy (defined as complete response; P= 0.006; Fig. 3B).
Moreover, Pearson’s correlation analysis showed a significant
correlation between CtIP status and clinical response to
endocrine therapy (P= 0.004). In this limited small study, data
seem to indicate that poor response to endocrine therapy is
associated with CtIP deficiency in breast cancer patients.
However, these observations have to be confirmed in larger
cohorts.
FIGURE 2. CtIP protein expression in breast cancer cell lines as
determined by Western blot analysis. ER expression status is also
indicated as positive (+) or negative (). As can be observed, all ER
breast cancer lines (100%) tested do not express CtIP protein, whereas
four of six ER+ lines (67%) express this protein.
FIGURE 3. Poor clinical response to endocrine therapy is significantly
associated with CtIP deficiency in primary breast carcinomas. A. CtIP
expression patterns in human breast carcinomas. CtIP expression was
evaluated by immunohistochemistry and semiquantified using IRS in 59
inoperable hormone receptor positive primary breast carcinomas (before
treatment) from patients who received endocrine therapy as single
neoadjuvant therapy. Representative tumor CtIP immunoreactivity (IRS
scores 0, 4, 8, and 12). B. CtIP status strongly correlates with clinical
response to endocrine therapy (Pearson correlation, P= 0.004), and
patients with progressive disease have significantly lower CtIP IRS than
those who completely respond to endocrine therapy. **, P= 0.006 by
ANOVA and Tukey’s post tests. CR, complete response; PR, partial
response; SD, stable disease; and PD, progressive disease.
CtIP and Tamoxifen Resistance
Mol Cancer Res 2007;5(12). December 2007
1287
CtIP Expression Is Associated with ER, Disease-Free
Survival, and Breast Cancer Metastasis Status
To further explore the clinical relevance of CtIP expression
in breast cancer, we evaluated information of seven publicly
available breast cancer gene expression microarray data sets
(16-22) through the use of the web-based Oncomine cancer
microarray database (23).
6
Clinicopathologic and gene expres-
sion data from a total of 828 breast carcinomas was obtained
using this publicly available resource. Because ER plays a
critical role in the clinical management of breast cancer
patients, we first analyzed levels of CtIP mRNA expression
in the mentioned microarray sets according to ER status of the
tumors (Fig. 4A). By using a meta-analysis approach, we
directly compared CtIP expression profiles between 590 ER+
and 238 ERbreast carcinomas by combining all seven
microarray data sets. We found a significant association
between high CtIP expression and ER (+) status in breast
carcinomas (P< 0.0001; Fig. 4A). Next, we analyzed CtIP
expression profiles versus disease-free survival in two micro-
array data sets that had at least 5 years of follow-up clinical
information available. Analysis from the study of van de Vijver
et al. (19) showed a statistically significant association between
loss of CtIP and disease relapse (P= 0.019; Fig. 4B, left ). A
trend was also found in the Sorlie et al. study (20) but did not
reach statistical significance (P= 0.069), possibly due to the
low number of samples (Fig. 4B, right). However, by using the
meta-analysis approach and pooling both studies together, we
found a highly significant association between decreased
CtIP expression and disease relapse (P= 0.004; Fig. 4B).
Furthermore, CtIP mRNA expression levels were significantly
lower in invasive breast carcinomas that had distant metastasis
when compared with breast cancer counterparts that did not
metastasize (P= 0.029; Fig. 4C).
Silencing Endogenous CtIP in Tamoxifen-Sensitive
MCF-7 Cells Confers Tamoxifen Resistance In vitro
To further explore the putative role of CtIP in the
development of tamoxifen resistance, we first examined
whether silencing the expression of endogenous CtIP in
tamoxifen-sensitive cells can induce a tamoxifen-resistant
phenotype. Knockdown of CtIP protein levels in tamoxifen-
sensitive parental MCF-7 cells was achieved using RNA
interference techniques. As shown in Fig. 5A, in the resulting
clone [MCF-7/CtIP; small interfering (siRNA)] stably trans-
fected with a vector expressing siRNA targeting CtIP mRNA,
the silencing of CtIP protein levels reproduces quite closely
the expression difference observed between the tamoxifen-
sensitive (parental MCF-7) and tamoxifen-resistant (TAMR1
and TAMR2) cells. The negative control siRNA clone
[MCF-7/() control siRNA] showed unchanged CtIP protein
levels when compared with parental MCF-7 cells (Fig. 5A).
In addition, we observed an equal level of ER expression in
all three clones, indicating that ER protein expression was
not affected by the siRNA intervention (Fig. 5A).
To test whether silenced CtIP expression in parental MCF-7
cells leads to the tamoxifen-sensitive to tamoxifen-resistant
transition, we next compared cell proliferation between MCF-7/
CtIP siRNA and MCF-7/() control siRNA cells under various
stimuli. Reduced CtIP expression increased MCF-7/CtIP
siRNA cell proliferation under estrogen-deprived conditions
compared with control siRNA cells (Fig. 5B). When hormone-
starved experimental and control siRNA cells were treated with
4-OH-TAM, growth of the control cells was inhibited by
exposure to tamoxifen, whereas growth of MCF-7/CtIP siRNA
cells was not inhibited by tamoxifen, indicating acquired
resistance to tamoxifen (P< 0.05; Fig. 5C). Furthermore, CtIP-
silenced MCF-7/CtIP siRNA cells grew significantly faster than
control cells in the presence of both estrogen plus tamoxifen
(P< 0.01), indicating that unlike in control cells, tamoxifen
indeed fails to inhibit the stimulatory effect of estrogen in CtIP-
silenced MCF-7 cells (Fig. 5D). Interestingly, the MCF-7/CtIP
siRNA cells still retain response to estrogen-stimulated cell
proliferation when exposed to estradiol (Fig. 5E), indicating
that ER is still functional and capable of regulating cell growth.
In fact, after 10 days in culture in the presence of estrogen,
the cell number of both control and MCF-7/CtIP siRNA cells
increased f145-fold (Fig. 5E), which is five to eight times
more than that of the MCF-7/CtIP siRNA cells cultured in the
presence of tamoxifen (Fig. 5C) or in the absence of estrogen
(Fig. 5B), respectively (over the same time period), suggesting
that estrogen still seems to stimulate cell proliferation in MCF-7
cells regardless of CtIP status. Taken together, these data
indicated that CtIP silencing leads to resistance to the inhibitory
growth effects of tamoxifen in vitro.
Reexpression of CtIP in Tamoxifen-Resistant Cells
Restores Sensitivity to the Inhibitory Growth Effects of
Tamoxifen
Next, we addressed the reciprocal question of whether
reexpression of CtIP in tamoxifen-resistant cells abrogates
resistance to tamoxifen. Because both tamoxifen-resistant lines
have similar proliferation profiles in the presence of tamoxifen,
we selected the TAMR1 cell line for further functional studies.
To this end, a Tet-off inducible gene expression system was
used to reexpress CtIP in the tamoxifen-resistant TAMR1 cells.
TAMR1 cells were transiently cotransfected with Tet-off–
inducible vectors containing NH
2
-terminal Flag-tagged full-
length human CtIP cDNA. After transfection, cells were
immediately divided equally into two batches. The first batch
was treated with 4-OH-TAM or vehicle, and cultured in the
presence of doxycycline. The second batch was treated with
4-OH-TAM or vehicle, but cultured in the absence of
doxycycline. Expression of FLAG-CtIP was analyzed by
immunoblotting after 72 h of transfection. FLAG-CtIP was only
detected in TAMR1 cells cultured without doxycycline (Fig. 6A,
left), indicating the restoration of CtIP is tightly controlled by
doxycycline. Cell proliferation was determined in TAMR1 cells
with or without CtIP reexpression. This experiment showed
that cells with restored CtIP protein expression displayed a
significant growth inhibition by tamoxifen in comparison with
control cells having no CtIP restoration (Fig. 6A, middle ).
Without tamoxifen treatment (vehicle control), transient CtIP
restoration had no significant effect on TAMR1 cell proliferation
(Fig. 6A, right ). Therefore, restoration of CtIP abrogates
resistance to tamoxifen in tamoxifen-resistant cells.
6
http://www.oncomine.org
Wu et al.
Mol Cancer Res 2007;5(12). December 2007
1288
To further confirm the observations derived from the
transient transfection experiments, we developed double stably
transfected TAMR1 Tet-off FLAG-CtIP cells. Among the
various stably transfected clones obtained, clone 32 was
selected for further study. This clone showed no CtIP expression
in the presence of doxycycline but similar CtIP protein levels to
those produced in parental MCF-7 cells upon doxycycline
withdrawal, as determined by Western blot analysis (Fig. 6B).
FIGURE 4. CtIP expression is associated with ER status and prognosis in breast cancer. CtIP gene expression profiles and clinicopathologic data of 828
breast carcinomas were obtained from seven published and publicly available breast cancer microarray data sets as described in Materials and Methods.
Data were collected and visualized using the Oncomine cancer microarray resource. A. Oncomine database output and meta-analysis showing CtIP
expression patterns relative to ER status across seven breast cancer microarray studies. Columns, CtIP transcripts represented as normalized expression
units; bars, SE (95% CI). B. Oncomine database output of CtIP expression patterns relative to disease-free survival in two of the seven data sets that have
5 y follow-up information available. The van de Vijver et al. data set (left ) shows a significant association between loss of CtIP expression and relapse
(P= 0.019). The Sorlie et al. study (right ) shows a trend that does not reach statistical significance (P= 0.069). Meta-analysis (pooling both studies together)
shows an excellent statistical inverse correlation between decreased CtIP expression and breast cancer relapse (P= 0.004). C. Oncomine database output
of CtIP expression patterns related to metastasis status (left, the van de Vijver et al. study; right, the Sorlie et al. study). The analysis shows a statistically
significant decrease of CtIP expression in association with metastatic breast carcinomas (P= 0.029).
CtIP and Tamoxifen Resistance
Mol Cancer Res 2007;5(12). December 2007
1289
Next, we measured the growth of the double-stably transfected
TAMR1 Tet-off FLAG-CtIP cells under conditions in which
CtIP restoration was either induced or repressed. Cells from
clone 32 were cultured in two different conditions. Half of the
cells were grown in hormone-free medium containing doxycy-
cline whereas the other half was cultured in the same medium
but devoid of doxycycline. After 3 days of incubation, cells
were treated with either 17-h-estradiol (E
2
), 4-OH-TAM, or
vehicle control. As shown in Fig. 6C, the growth of TAMR1
Tet-off FLAG-CtIP cells was significantly inhibited by
tamoxifen when doxycycline was removed from the medium
compared with cells from the same clone but cultured in the
presence of doxycycline (P< 0.05). Cells still responded well to
estrogen regardless of doxycycline status (Fig. 6D). Taken
together, these results show that sensitivity to the inhibitory
growth effects of tamoxifen in previously tamoxifen-resistant
cells is restored, at least partially by CtIP reexpression.
Discussion
SAGE studies identified CtIP as one of the most
significantly down-regulated transcripts in two independently
developed tamoxifen-resistant breast cancer cell lines when
compared with their tamoxifen-sensitive parental MCF-7 line.
Immunoblotting analyses not only validated the SAGE
observations but also showed a dramatic difference in CtIP
FIGURE 5. Silencing of
CtIP protein expression in ta-
moxifen-sensitive MCF-7 cells
confers tamoxifen resistance
in vitro.A. Western blot anal-
ysis of CtIP and ER proteins in
parental MCF-7 cells, MCF-7/
() control siRNA, and MCF-7/
CtIP siRNA stable clones.
Growth curves of control and
MCF-7/CtIP siRNA cells
treated with vehicle control (B)
or 4-OH-TAM (C) for 2 wk. D.
Growth curves of MCF-7/CtIP
siRNA and () control cells
grown in hormone-free medium
and treated with E
2
(10 nmol/L)
and 4-OH-TAM (1 Amol/L) for 6
d. E. Growth curves of control
and MCF-7/CtIP siRNA cells
exposed to E
2
for 10 d. All
experiments were done in trip-
licate. Points, mean; bars, SE. *
and **, P<0.05andP< 0.01 by
ttest, respectively. Note that
Y-axis scale for cell number in
Dis different from that in B,
C, and E.
Wu et al.
Mol Cancer Res 2007;5(12). December 2007
1290
protein expression levels, with high levels of protein
expression in parental ER+ MCF-7 breast cancer cells but
very little or no CtIP protein product in the ER+ tamoxifen-
resistant derivative isogenic cell lines. In addition to the
observations in MCF-7 cells, we also found significantly
reduced CtIP protein expression in ER+ T-47-D breast cancer
cells when they became tamoxifen resistant. Furthermore,
consistent with the in vitro observations, we found significant
association of CtIP deficiency in breast cancers from patients
with poor clinical response to endocrine therapy. By meta-
analysis of gene expression data sets, we also found an
association of CtIP expression levels with poor outcome.
These in vivo data suggest that CtIP gene and protein
expression may be useful biomarkers for breast cancer
prognosis and clinical management, although it would require
further studies in larger patient cohorts. In addition, we found
a significant reverse association of CtIP expression with ER
status, suggesting that CtIP may function in a pathway or
pathways associated with ER signaling and perhaps regulate
ER-mediated cell proliferation. All ERbreast cancer cells
FIGURE 6. Tamoxifen-resistant cells regain sensitivity to the inhibitory growth effects of tamoxifen upon restoration of CtIP protein expression. A. Left,
expression of FLAG-CtIP protein in TAMR1 cells transiently cotransfected with inducible FLAG-CtIP expression vectors and cultured in the presence or
absence of doxycycline (DOX) for 3 d. Middle and right, transient CtIP restoration partially abrogates resistance to tamoxifen in TAMR1 cells. TAMR1 cells
were transiently cotransfected with inducible FLAG-CtIP expression vectors and treated with 4-OH-TAM (middle ) or vehicle control (right ) in the presence or
absence of doxycycline for 3 d. Cell proliferation was determined as shown. Percent cell number (middle) represents cell numbers relative to vehicle control
treated cells. Columns, mean of triplicate determinations; bars, SE. B. Total CtIP protein expression in the double-stably transfected TAMR1 Tet-off FLAG-
CtIP clone 32 cells in the presence or absence of doxycycline. CtIP expression in parental MCF-7 cells is shown for comparative purposes. C. CtIP
reexpression upon doxycycline withdrawal restores sensitivity to the inhibitory growth effects of tamoxifen in TAMR1 Tet-off FLAG-CtIP clone 32 cells. The
proliferation of clone 32 cells under the treatment of 4-OH-TAM was determined in the presence (black columns ) or absence (white columns) of doxycycline.
*, P< 0.05 by ttest. D. Effect of E
2
on the proliferation of clone 32 cells cultured with or without doxycycline. Note that Yaxis scales for cell number in Cand
Dare different from each other.
CtIP and Tamoxifen Resistance
Mol Cancer Res 2007;5(12). December 2007
1291
tested (intrinsically resistant to tamoxifen) express either none or
very little CtIP protein. Interestingly, BT-474, an ER+ but
tamoxifen-resistant breast cancer cell line as reported by Wang
et al. (10), also expresses nearly undetectable CtIP protein when
compared with ER (+) and tamoxifen-sensitive lines such as
MCF-7 and T-47-D, which further supports the important role of
CtIP in the development of tamoxifen resistance. Finally, based
on RNA interference and reciprocal reexpression studies, we
were able to reproduce the tamoxifen-resistant phenotype
simply by shutting down the expression of one gene
(i.e., CtIP). These data show that CtIP silencing is critical for
the development of tamoxifen resistance in breast cancer in vitro
models and suggest that CtIP silencing may be a novel
mechanism by which cells can circumvent the inhibitory effects
of tamoxifen to resume proliferation and ultimately acquire
resistance to the antiestrogen tamoxifen.
The human CtIP (also known as RBBP8) encodes an 897-
amino-acid nuclear protein that is widely expressed in various
human tissues (24-27). It was initially identified as a cofactor of
transcriptional corepressor CtBP (24). CtIP is also known to
interact with tumor suppressors, Rb family proteins (Rb and
p130; refs. 25, 28) and BRCA1 (26, 29-31), as well as the
transcriptional repressors such as LIM-only protein LMO4 (30)
and Ikaros family members (32). Recent studies suggest that
CtIP plays an important role in cell cycle regulation and DNA
damage response (33-37). Emerging evidence also suggests that
CtIP may itself be a tumor susceptibility gene. Analysis of CtIP
cDNA from 89 human tumor cell lines revealed 5 missense and
11 silent mutations (26). In a more recent screening study of
109 colon cancers, CtIP was found to be a frequent target for
microsatellite instability (38). More importantly, it has been
shown that inactivation of CtIP in mice leads to early
embryonic lethality, and the life span of Ctip
+/
heterozygotes,
which have Ctip haploid insufficiency, was shortened due to
the development of multiple types of tumors. These findings
show that CtIP is a critical protein in early embryogenesis and
implicates an important role of CtIP in tumorigenesis (39). In
addition, CtIP interacts with the BRCT domains of BRCA1
where most mutations occur in BRCA1 breast cancer patients,
and such protein-protein interaction is abolished by tumor-
associated mutations in the BRCT domains (26, 29, 31),
suggesting that interaction between CtIP and BRCA1 is
of functional relevance in the breast cancer suppressor activity.
It has been shown that amino acid residues 299 to 345 of
CtIP mediate its interaction with the BRCT domains of BRCA1
(26, 27, 37). Recently, it was also reported that phosphorylation
at Ser
327
in CtIP seemed to be critical for its interaction with
BRAC1 BRCT domains (37, 40).
Available evidence suggests that CtIP is involved in
transcriptional repression (reviewed in refs. 41, 42). Signifi-
cantly, recent studies showed that BRCA1, CtIP, and ZBRK1
form a repressor complex at a recognition site of ZBRK1 in
ANG1 promoter and a defect of this complex formation
derepresses ANG1 transcription, promoting endothelial cell
survival and vascular enlargement (43). Interestingly, other
studies showed that BRCA1 physically interacts with ER and
inhibits transcriptional activity of the receptor (44, 45). In this
study, we found that high CtIP expression is significantly
associated with ER+ status. Thus, it is possible that CtIP may
functionally be linked with ER signaling via its interaction with
BRCA1. Moreover, CtIP was also shown to form a complex
with BRCA1 and the transcriptional corepressor CtBP, which is
important for the repression of p21 promoter activity (29).
Together, it raises the possibility that in physiologic conditions,
CtIP could bridge BRCA1 and CtBP to form a transcriptional
repressor complex, which in turn may modulate ER signaling
pathways through the interaction between BRCA1 and ER.
Therefore, we hypothesize that in tamoxifen-sensitive cells,
BRCA1, CtIP, and CtBP form a transcriptional repressor
complex that leads to inhibition of a full ER+ transcriptional
response, accounting for the inhibitory growth effects of
tamoxifen. To circumvent the transcriptional inhibitory effects
of tamoxifen, tamoxifen-resistant cells silence CtIP expression,
which, in turn, disrupts the repressor complex and allows breast
cancer cells to resume proliferation. Further characterization of
the CtIP signaling pathways may provide insights into how CtIP
silencing leads to the development of tamoxifen resistance.
Materials and Methods
Cell Lines and Chemicals
Parental MCF-7 cells used in this study have been described
previously (46). The tamoxifen-resistant MCF-7 isogenic cell
line variants (termed TAMR1 and TAMR2) were generated
in our laboratory by culturing MCF-7 cells under continuous
4-OH-TAM (the active metabolite of tamoxifen; 1 Amol/L)
exposure for f2 years. These cells were maintained in phenol
red-free IMEM medium containing 5% fetal bovine serum
(TAMR1) or 5% charcoal-stripped fetal bovine serum
(TAMR2), glutamine (2 mmol/L), gentamicin (50 Ag/mL)
and 4-OH-TAM (1 Amol/L). The characterization of parental
MCF-7 and TAMR1 cells has been previously described
(47, 48). Tamoxifen-resistant T-47-D cells (T-47-D/TR) were
generated by growing regular ER+ T-47-D cells in the presence
of 4-OH-TAM for over 3 months. These cells were maintained
in DMEM (Cambrex Bio Science) medium containing 10%
fetal bovine serum and 4-OH-TAM (1 Amol/L). Other breast
cancer cell lines used, including SUM-44-PE, ZR-75-1, MDA-
MB-231, MDA-MB-435, SKBR3, and BT-474, were main-
tained in DMEM (Cambrex Bio Science) supplemented with
10% fetal bovine serum. The BT-483 cell line was maintained
in RPMI (Cambrex Bio Science) supplemented with 10% fetal
bovine serum. The UACC-812 breast cancer cell line was
grown in L-15 medium (Invitrogen) supplemented with 10%
fetal bovine serum. We purchased 4-OH-TAM and E
2
from
Sigma-Aldrich.
Serial Analysis of Gene Expression
SAGE was done on tamoxifen-sensitive parental MCF-7 line
and tamoxifen-resistant TAMR1 and TAMR2 lines. SAGE
library generation, sequencing, and tag extraction were done as
previously described (46, 49, 50). SAGE data were analyzed
using an ANOVA-based multivariate approach called multiple
linear contrast analysis. Two contrasts were defined to identify
differentially expressed genes in the tamoxifen-resistant cells,
one comparing the average expression level in the two resistant
cell lines to that of the parental strain and the second one
comparing the expression levels between the two resistant
Wu et al.
Mol Cancer Res 2007;5(12). December 2007
1292
strains. The null hypothesis for first contrast tests for lack of
differential expression between resistant and parental strains
and the second tests for consistent expression between the two
resistant cell lines. Significance of the null hypotheses for these
tests was set at the 95% level after a Bonferroni-type adjustment
for the multiplicity of comparisons.
Western Blot Analysis
Cells were washed twice with ice-cold PBS and then lysed
with radioimmunoprecipitation assay buffer [10 mmol/L Tris,
5 mmol/L EDTA, 150 mmol/L NaCl, 0.1% SDS, 1% Triton
X-100, 1% deoxycholate, (pH 7.2), 1protease inhibitor
cocktail (Roche)]. Cell lysates were flushed 10 times through
21-gauge needle and microcentrifuged at 21,000 gfor
10 min at 4jC. Supernatants were collected and protein
concentration was measured with a Pierce Protein Assay Kit,
according to the manufacturer’s instruction. Equal amount
(30 50 Ag) of protein from each sample was separated on 6%
to 10% SDS-PAGE and transferred to polyvinylidene difluoride
membranes by electroblotting. Blots were first treated with
blocking buffer [5% milk in 1TBS containing 0.1% Tween
20 (TBS-T)] for 1 h and then incubated with primary antibodies
for 1 to 2 h. After washing thrice in TBS-T, blots were
incubated with horseradish peroxidaselabeled secondary
antibodies for 1 h. Labeled proteins were detected using KPL
Protein Detector chemiluminescence detection reagents and
exposed to X-ray films. All procedures described were carried
out at room temperature. Antibodies used were as follows:
CtIP (14-1; ref. 27; T-16, Santa Cruz Biotechnology; 19E8,
GeneTex), ER (HC-20, Santa Cruz Biotechnology), anti-Flag
(M2, Sigma), and h-actin (AC-15, Sigma).
Human Samples
Primary breast cancer formalin-fixed, paraffin-embedded
tissue samples were collected retrospectively from 59 postmen-
opausal patients with stage II to III ER (+) breast carcinomas
(median age 78 years; range 60 to 92 years). The 59 patients
were treated at a single institution (Instituto Valenciano
Oncologı´a, Valencia, Spain) between 1999 and 2002 with 4
months neoadjuvant endocrine therapy consisting of tamoxifen
(23 patients) or letrozole (36 patients) for large nonoperable or
locally advanced ER+ breast cancers. These patients were part of
a larger study published elsewhere (13). All patients gave written
informed consent before the submission of tumor samples for
CtIP analyses, and the local ethic committee approved the study
protocol and informed consent form. Determination of response
to the referred endocrine adjuvant therapy was made after
4 months of patient follow-up. We used one-way ANOVA
followed by Tukey’s test post hoc comparisons, and Pearson’s
correlation test to assess the association between CtIP protein
status (raw IRS scores) and clinical response to endocrine
therapy. All statistical analyses were two side, and P< 0.05 was
considered as statistically significant. Analyses were conducted
using SPSS version 11.5 software (SPSS, Inc.).
Immunohistochemistry Analysis
Immunohistochemical staining was done as previously
described (51) with a rabbit polyclonal CtIP antibody (H-300,
1:100 dilution, Santa Cruz Biotechnology). CtIP expression
levels was scored blindly by two independent pathologists
(D.R.S. and M.I.N.) using the IRS method as previously
described (14, 15). In brief, the IRS was calculated by
multiplying the percentage of CtIP-positive cells (scored 0 to
4: 0, 0%; 1, 0-25%; 2, 26-50%; 3, 51-75%; 4, >75%) with the
CtIP staining intensity (scored 0 to 3: 0, none; 1, weak; 2,
moderate; 3, strong).
Meta-analysis of Breast Cancer Microarray Data Sets
CtIP gene expression profiles and clinicopathologic data of
828 breast carcinomas were obtained from seven published and
publicly available breast cancer microarray data sets (16-22).
The Oncomine cancer microarray database was used for data
collection, processing, and visualization (23).
6
CtIP gene
expression was log-transformed, median centered for each
gene expression data set, and SD was normalized to one per
array. The gene module application was used for differential
expression analysis (two-sided ttest). We used a meta-analysis
approach to determine and summarize the CtIP mRNA
expression pattern from the seven independent studies. We
computed summary estimates (effect sizes) of CtIP expression
changes by the standardized mean difference method using the
exact tvalues and sample size for each groups. To calculate the
pooled effects of CtIP profile, each study was weighed by the
inverse of the individual and between-study variance according
to a random-effects model. Meta-analysis was carried out using
comprehensive meta-analysis software (Biostat, Inc.). All effect
sizes were presented with 95% confidence interval –based on
the estimated variances.
In vitro Cell Proliferation Assays
Cells were cultured in estrogen-free medium for 48 h. On
day 0, 1 10
4
cells in estrogen-free medium were plated
in triplicate in 12-well plates. E
2
(10 nmol/L), 4-OH-TAM
(10 nmol/L), or ethanol (vehicle control, 1 AL/mL) was added
directly into the medium at the same time. Fresh medium
with the adequate treatment was changed every 2 days. Cell
counts were done at various time points as indicated in the
figures.
RNA Interference
siRNA expression cassette (SEC) encoding siRNA targeting
CtIP mRNA (from 2,492 to 2,512, NM_002894; AATGA-
TAGCTTGGAAGATATG) was generated using the Silencer
Express siRNA Expression Cassette Kits (Ambion). A negative
control SEC expressing siRNA with no significant homology to
human, mouse, or rat genome sequences was also generated
by the same method. The SECs were then cloned into the
mammalian expression pSEC-puro vector (Ambion). To obtain
cell clones that stably expressed siRNAs targeting CtIP, MCF-7
cells were transfected with either pSEC-CtIP-puro or pSEC-
Control-puro by electroporation. One day after, puromycin
(600 ng/mL) was added into the culture medium. After 3 weeks
of selection, puromycin-resistant clones were picked up,
expanded, and analyzed for CtIP expression levels by
immunoblotting. The stable clone displaying the lowest level
of CtIP protein expression was chosen for further studies.
CtIP and Tamoxifen Resistance
Mol Cancer Res 2007;5(12). December 2007
1293
Generation of Double-Stably Transfected Tet-off TAMR1
Cells with Doxycycline-Inducible Restoration of CtIP
The Tet-off gene expression system was purchased from BD
Biosciences Clontech. Full-length human CtIP cDNA with
three NH
2
-terminal Flag epitope tags was cloned into
pTRE2hyg response vector (pTRE2hyg-FLAG-CtIP). To
generate double-stable Tet-off TAMR1 cell clones, cells were
cotransfected with pTet-Off and pTRE2hyg-FLAG-CtIP vec-
tors. After electroporation, cells were plated in 10-cm dishes
and allowed to grow in regular medium containing 4-OH-TAM
(1 Amol/L) plus doxycycline (1 Ag/mL) for 48 h. Cells were
then selected for resistance to G418 (800 Ag/mL) and
hygromycin B (200 Ag/mL). Fresh doxycycline (1 Ag/mL)
was added to tamoxifen-containing medium (regular culture
medium for TAMR1 cells) every 2 days to maintain a constant
suppression of CtIP expression during the selection process.
Hygromycin and G418 double-resistant colonies began to
appear after 3 to 4 weeks of selection. Thirty-seven large and
healthy colonies were isolated using cloning cylinders and
transferred to individual wells for expansion. Each clone was
first screened by immunoblotting using anti-Flag M2 antibody
for doxycycline-responsive CtIP expression in the presence or
absence of 1 Ag/mL doxycycline. We then used anti-CtIP (14-1)
antibody to assess the total CtIP protein in the positive clones.
Clone 32, in which the level of total CtIP protein expressed
upon the withdrawal of doxycycline was similar to that
produced in the parental MCF-7 cells, was chosen for further
studies.
Acknowledgments
We thank Dr. John H. Ludes-Meyers for helpful discussions.
References
1. Jordan VC. Tamoxifen: a most unlikely pioneering medicine. Nat Rev Drug
Discov 2003;2:205 13.
2. Osborne CK. Tamoxifen in the treatment of breast cancer. N Engl J Med 1998;
339:1609 18.
3. Normanno N, Di Maio M, De Maio E, et al. Mechanisms of endocrine
resistance and novel therapeutic strategies in breast cancer. Endocr Relat Cancer
2005;12:721 47.
4. Clarke R, Leonessa F, Welch JN, Skaa r TC. Cellular and molec ular
pharmacology of antiestrogen action and resistance. Pharmacol Rev 2001;53:
25 71.
5. Clarke R, Skaar TC, Bouker KB, et al. Molecular and pharmacological aspects
of antiestrogen resistance. J Steroid Biochem Mol Biol 2001;76:71– 84.
6. Ali S, Coombes RC. Endocrine-responsive breast cancer and strategies for
combating resistance. Nat Rev Cancer 2002;2:101 12.
7. Clarke R, Liu MC, Bouker KB, et al. Antiestrogen resistance in breast cancer
and the role of estrogen receptor signaling. Oncogene 2003;22:7316 39.
8. Ring A, Dowsett M. Mechanisms of tamoxifen resistance. Endocr Relat
Cancer 2004;11:643 58.
9. Osborne CK, Shou J, Massarweh S, Schiff R. Crosstalk between estrogen
receptor and growth factor receptor pathways as a cause for endocrine therapy
resistance in breast cancer. Clin Cancer Res 2005;11:865 70s.
10. Wang LH, Yang XY, Zhang X, et al. Disruption of estrogen receptor DNA-
binding domain and related intramolecular communication restores tamoxifen
sensitivity in resistant breast cancer. Cancer Cell 2006;10:487 99.
11. Gutierrez MC, Detre S, Johnston S, et al. Molecular changes in tamoxifen-
resistant breast cancer: relationship between estrogen receptor, HER-2, and p38
mitogen-activated protein kinase. J Clin Oncol 2005;23:2469 76.
12. Johnston SR, Saccani-Jotti G, Smith IE, et al. Changes in estrogen receptor,
progesterone receptor, and pS2 expression in tamoxifen-resistant human breast
cancer. Cancer Res 1995;55:3331 8.
13. Eiermann W, Paepke S, Appfelstaedt J, et al. Preoperative treatment of
postmenopausal breast cancer patients with letrozole: a randomized double-blind
multicenter study. Ann Oncol 2001;12:1527 32.
14. Chui X, Egami H, Yamashita J, et al. Immunohistochemical expression of the
c-kit proto-oncogene product in human malignant and non-malignant breast
tissues. Br J Cancer 1996;73:1233 6.
15. Friedrichs K, Gluba S, Eidtmann H, Jonat W. Overexpression of p53 and
prognosis in breast cancer. Cancer 1993;72:3641 7.
16. Perou CM, Jeffrey SS, van de Rijn M, et al. Disti nctive gene expression
patterns in human mammary epithelial cells and breast cancers. Proc Natl Acad
Sci U S A 1999;96:9212 7.
17. West M, Blanchette C, Dressman H, et al. Predicting the clinical status of
human breast cancer by using gene expression profiles. Proc Natl Acad Sci U S A
2001;98:11462 7.
18. Gruvberger S, Ringner M, Chen Y, et al. Estrogen receptor status in breast
cancer is associated with remarkably distinct gene expression patterns. Cancer
Res 2001;61:5979 84.
19. van de Vijver MJ, He YD, van’t Veer LJ, et al. A gene-expression
signature as a predictor of survival in breast cancer. N Engl J Med 2002;347:
1999 2009.
20. Sorlie T, Tibshirani R, Parker J, et al. Repeated observation of breast tumor
subtypes in independent gene expression data sets. Proc Natl Acad Sci U S A
2003;100:8418 23.
21. Zhao H, Langerod A, Ji Y, et al. Different gene expression patterns in
invasive lobular and ductal carcinomas of the breast. Mol Biol Cell 2004;15:
2523 36.
22. Wang Y, Klijn JG, Zhang Y, et al. Gene-expression profiles to predict
distant metastasis of lymph-node-negative primary breast cancer. Lancet 2005;
365:671 9.
23. Rhodes DR, Yu J, Shanker K, et al. ONCOMINE: a cancer microarray
database and integrated data-mining platform. Neoplasia 2004;6:1 6.
24. Schaeper U, Subramanian T, Lim L, Boyd JM, Chinnadurai G. Interaction
between a cellular protein that binds to the C-terminal region of adenovirus E1A
(CtBP) and a novel cellular protein is disrupted by E1A through a conserved
PLDLS motif. J Biol Chem 1998;273:8549 52.
25. Fusco C, Reymond A, Zervos AS. Molecular cloning and charact erization of
a novel retinoblastoma-binding protein. Genomics 1998;51:351 8.
26. Wong AK, Ormonde PA, Pero R, et al. Characterization of a carboxy-
terminal BRCA1 interacting protein. Oncogene 1998;17:2279 85.
27. Yu X, Baer R. Nuclear localization and cell cycle-specific expression of CtIP,
a protein that associates with the BRCA1 tumor suppressor. J Biol Chem 2000;
275:18541 9.
28. Meloni AR, Smith EJ, Nevins JR. A mechanism for Rb/p130-mediated
transcription repression involving recruitment of the CtBP corepressor. Proc Natl
Acad Sci U S A 1999;96:9574 9.
29. Li S, Chen PL, Subramanian T, et al. Binding of CtIP to the BRCT repeats of
BRCA1 involved in the transcription regulation of p21 is disrupted upon DNA
damage. J Biol Chem 1999;274:11334 8.
30. Sum EY, Peng B, Yu X, et al. The LIM domain protein LMO4 interacts with
the cofactor CtIP and the tumor suppressor BRCA1 and inhibits BRCA1 activity.
J Biol Chem 2002;277:7849 56.
31. Yu X, Wu LC, Bowcock AM, Aronheim A, Baer R. The C-terminal (BRCT)
domains of BRCA1 interact in vivo with CtIP, a protein implicated in the CtBP
pathway of transcriptional repression. J Biol Chem 1998;273:25388 92.
32. Koipally J, Georgopoulos K. Ikaros-CtI P interactions do not require
C-terminal binding protein and participate in a deacetylase-independent mode
of repression. J Biol Chem 2002;277:23143 9.
33. Foray N, Marot D, Gabriel A, et al. A subset of ATM- and ATR-dependent
phosphorylation events requires the BRCA1 protein. EMBO J 2003;22:2860 –71.
34. Li S, Ting NS, Zheng L, et al. Functional link of BRCA1 and ataxia
telangiectasia gene product in DNA damage response. Nature 2000;406:210 5.
35. Liu F, Lee WH. CtIP activates its own and cyclin D1 promoters via the E2F/
RB pathway during G1/S progr ession. Mol Cell Biol 2006;26:3124 34.
36. Wu-Baer F, Baer R. Effect of DNA damage on a BRCA1 complex. Nature
2001;414:36.
37. Yu X, Chen J. DNA damage-induced cell cycle checkpoint control requires
CtIP, a phosphorylation-depende nt binding partner of BR CA1 C-terminal
domains. Mol Cell Biol 2004;24:9478 86.
38. Vilkki S, Launonen V, Karhu A, Sistonen P, Vastrik I, Aaltonen LA.
Screening for microsatellite instability target genes in colorectal cancers. J Med
Genet 2002;39:785 9.
39. Chen PL, Liu F, Cai S, et al. Inactivation of CtIP leads to early embryonic
Wu et al.
Mol Cancer Res 2007;5(12). December 2007
1294
lethality mediated by G
1
restraint and to tumorigenesis by haploid insufficiency.
Mol Cell Biol 2005;25:3535 42.
40. Varma AK, Brown RS, Birrane G, Ladias JA. Structural basis for cell cycle
checkpoint control by the BRCA1-CtIP comple x. Biochemistry 2005;44:
10941 6.
41. Chinnadurai G. CtIP, a candidate tumor susceptibility gene is a team player
with luminaries. Biochim Biophys Acta 2006;1765:67 73.
42. Wu G, Lee WH. CtIP, a multivalent adaptor connecting transcriptional
regulation, checkp oint control and tumor suppression. Cell Cycle 2006;5:1592 6.
43. Furuta S, Wang JM, Wei S, et al. Removal of BRCA1/CtIP/ZBRK1 repressor
complex on ANG1 promoter leads to accelerated mammary tumor growth
contributed by prominent vasculature. Cancer Cell 2006;10:13 24.
44. Zheng L, Annab LA, Afshari CA, Lee WH, Boyer TG. BRCA1 mediates
ligand-independent transcriptional repression of the estrogen receptor. Proc Natl
Acad Sci U S A 2001;98:9587 92.
45. Fan S, Ma YX, Wang C, et al. Role of direct interaction in BRCA1 inhibition
of estrogen receptor activity. Oncogene 2001;20:77 87.
46. Charpentier AH, Bednarek AK, Daniel RL, et al. Effects of estrogen on
global gene expression: identification of novel targets of estrogen action. Cancer
Res 2000;60:5977 83.
47. Hodges LC, Cook JD, Lobenhofer EK, et al. Tamoxifen functions as a
molecular agonist inducing cell cycle-associated genes in breast cancer cells. Mol
Cancer Res 2003;1:300 11.
48. Rayala SK, Talukder AH, Balasenthil S, et al. P21-activated kinase 1
regulation of estrogen receptor-aactivation involves serine 305 activation linked
with serine 118 phos phorylation. Cancer Res 2006;6 6:1694 701.
49. Hu Y, Sun H, Drake J, et al. From mice to humans: identification of
commonly deregulated genes in mammary cancer via comparative SAGE studies.
Cancer Res 2004;64:7748 55.
50. Velculescu VE, Zhang L, Vogelstein B, Kinzler KW. Serial analysis of gene
expression. Science 1995;270:484 7.
51. Nunez MI, Ludes-Meyers J, Abba MC, et al. Frequent loss of WWOX
expression in breast cancer: correlation with estrogen receptor stat us. Breast
Cancer Res Treat 2005;89:99 105.
CtIP and Tamoxifen Resistance
Mol Cancer Res 2007;5(12). December 2007
1295
... Consistently, CtIP is frequently amplified in oesophageal carcinoma (ESCA) and stomach adenocarcinoma (STAD) (Fig. 3A). Other studies, however, have found that CtIP expression is frequently downregulated in breast cancer patients and that low expression is combined with unfavourable prognosis and metastasis formation [97][98][99], which is markedly different from what is observed in pancreatic cancer patients (Fig. 3C). Thus, the relationship between CtIP expression and disease progression appears to be intricately linked with tumour type, possibly reflecting the relative importance of distinct CtIP functions in the pathogenesis of different cancers. ...
... High CtIP expression correlates with unfavourable prognosis in PAAD, both for overall (p-value = 0.0032) and progression-free (p-value = 0.0092) survival. For this analysis, patient samples were divided into two RBBP8 expression groups, separated by the median: "High" (expression values ≥ 9.350) and "Low" (expression values cancer aggressiveness and poor response to hormonal therapy in breast cancer [97,99]. However, reduced CtIP protein levels in luminal breast cancers are associated with better response to DNA-damaging chemotherapy [99], presumably due to a decreased DSB repair capacity. ...
Article
Full-text available
Human CtIP was originally identified as an interactor of the retinoblastoma protein and BRCA1, two bona fide tumour suppressors frequently mutated in cancer. CtIP is renowned for its role in the resection of DNA double-strand breaks (DSBs) during homologous recombination, a largely error-free DNA repair pathway crucial in maintaining genome integrity. However, CtIP-dependent DNA end resection is equally accountable for alternative end-joining, a mutagenic DSB repair mechanism implicated in oncogenic chromosomal translocations. In addition, CtIP contributes to transcriptional regulation of G1/S transition, DNA damage checkpoint signalling, and replication fork protection pathways. In this review, we present a perspective on the current state of knowledge regarding the tumour-suppressive and oncogenic properties of CtIP and provide an overview of their relevance for cancer development, progression, and therapy.
... Studies have shown that Rbbp8 −/− homozygosity leads to early embryo death in mice but that Rbbp8 +/− heterozygosity renders mice prone to multiple malignant tumors, mainly large B-cell lymphoma [34]. Accordingly, expression of RBBP8-WT is decreased in breast cancer patients, correlating positively with survival [35], and is associated with tamoxifen resistance [36]. In our research, RBBP8-WT germline mutation may be the biomarker for precise treatment and synthetic lethal treatment of this familial hereditary cancer syndrome. ...
Article
Full-text available
Screening tumor susceptibility genes helps in identifying powerful biomarkers for hereditary cancer monitoring, prevention, and diagnosis, providing opportunities for understanding potential molecular mechanisms and biomarkers for the precise treatment of hereditary cancer syndromes. Whole-exome sequencing of blood and bioinformatics analysis uncovered a novel RBBP8(p.E281*) germline mutation in a family with hereditary cancer syndrome, which was verified by Sanger sequencing. Cell proliferation, colony formation, cell migration, and in vivo tumorigenesis were investigated by CCK8, colony formation, Transwell, and in vivo xenograft assays. Protein localization and interaction were detected by immunofluorescence, nuclear and cytoplasmic protein extraction kits, and Co-IP. A new heterozygous germline mutation of the RBBP8(p.E281*) gene was found to be associated with familial hereditary cancer syndrome. RBBP8-WT was mainly detected in the nucleus and interacts with BRCA1. In contrast, RBBP8(p.E281*) is mainly located in the cytoplasm, with no interaction with BRCA1. RBBP8(p.E281*) variant plays an oncogenic role in the cytoplasm in addition to its loss of function in the nucleus, which promotes breast cancer proliferation, in vivo tumorigenesis, and migration. Compared with the control group, RBBP8(p.E281*) showed elevated cell death in response to cisplatin and olaparib treatment. A novel RBBP8(p.E281*) germline mutation was identified from familial hereditary cancer syndrome. RBBP8(p.E281*) is not able to enter the nucleus or interact with BRCA1 through the lost binding motif, and RBBP8(p.E281*) variant appears to promote tumorigenesis in the cytoplasm in addition to its loss of function in the nucleus. RBBP8(p.E281*) variant may promote tumor susceptibility and serve as a precision medicine biomarker in familial hereditary cancer syndrome. Key messages RBBP8(p.E281*) is a susceptibility gene in this familial hereditary cancer syndrome RBBP8(p.E281*) lost its ability to enter the nucleus and the BRCA1 binding motif A novel RBBP8(p.E281*) germline mutation promotes breast cancer tumorigenesis Patients with RBBP8(p.E281*) germline mutation may benefit from Olaparib, Cisplatin
... Studies have shown that the expression of RBBP8-WT was decreased in breast cancer patients and positively correlated with the survival rate [36] . and also associated with tamoxifen resistance [37] . Similarly, in our research, RBBP8-WT inhibited breast cancer proliferation, clone formation, and migration by induce E-cadherin expression. ...
Preprint
Full-text available
Background: Screening tumor susceptibility genes will be found powerful biomarkers for hereditary cancer monitoring, prevention and diagnosis, which providing opportunities for understanding the potential molecular mechanism and biomarker for precise treatment of hereditary cancer syndromes. Methods: Whole blood exon sequencing and bioinformatics analysis uncovers a novel RBBP8 (c.841G>T) germline mutation in this familial hereditary cancer syndrome, and verified by sanger sequencing. Cell proliferation, clone formation, cell migration, in vivo tumorigenesis were investigated by CCK8, clone formation assays, transwell, in vivo xenograft. The protein localization and interaction were detected by immunofluorescence, nuclear and cytoplasmic protein extraction kit and Co-IP . Results: A new heterozygous germline mutation of RBBP8 (c.841G>T) gene was associated with familial hereditary cancer syndrome. Unlike RBBP8-WT is mainly detected in the nucleus and could interact with BRCA1. Opposite, RBBP8(c.841G>T) is mainly located in the cytoplasm and lost interact with BRCA1, which may gain some new functions in the cytoplasm to promote breast cancer proliferation, in vivo tumorigenesis and migration. Meanwhile, patients with RBBP8(c.841G>T) germline mutation maybe benefit from PARP inhibitors (Olaparib) and platinum (Cisplatin). Conclusions: A de novo RBBP8 (c.841G>T) germline mutation was identified from familial hereditary cancer syndrome. RBBP8 (c.841G>T) lost its ability to enter the nucleus and the BRCA1 binding motif, which may gain some new functions to promote tumorigeneses in the cytoplasm. RBBP8 (c.841G>T) is a tumor susceptibility gene and candidate precision medicine biomarker in this familial hereditary cancer syndrome.
... Studies have shown that the expression of RBBP8-WT was decreased in breast cancer patients and positively correlated with the survival rate [36] . and also associated with tamoxifen resistance [37] . Similarly, in our research, RBBP8-WT inhibited breast cancer proliferation, clone formation, and migration by induce E-cadherin expression. ...
Preprint
Full-text available
Background: Screening tumor susceptibility genes will be found powerful biomarkers for hereditary cancer monitoring, prevention and diagnosis, which providing opportunities for understanding the potential molecular mechanism and biomarker for precise treatment of hereditary cancer syndromes. Methods: Whole blood exon sequencing and bioinformatics analysis uncovers a novel RBBP8 (c.841G>T) germline mutation in this familial hereditary cancer syndrome, and verified by sanger sequencing. Cell proliferation, clone formation, cell migration, in vivo tumorigenesis were investigated by CCK8, clone formation assays, transwell, in vivo xenograft. The protein localization and interaction were detected by immunofluorescence, nuclear and cytoplasmic protein extraction kit and Co-IP. Results: A new heterozygous germline mutation of RBBP8 (c.841G>T) gene was associated with familial hereditary cancer syndrome. Unlike RBBP8-WT is mainly detected in the nucleus and could interact with BRCA1. Opposite, RBBP8(c.841G>T) is mainly located in the cytoplasm and lost interact with BRCA1, which may gain some new functions in the cytoplasm to promote breast cancer proliferation, in vivo tumorigenesis and migration. Meanwhile, patients with RBBP8(c.841G>T) germline mutation maybe benefit from PARP inhibitors (Olaparib) and platinum (Cisplatin). Conclusions: A de novo RBBP8 (c.841G>T) germline mutation was identified from familial hereditary cancer syndrome. RBBP8 (c.841G>T) lost its ability to enter the nucleus and BRCA1 binding motif, which may gain some new functions to promote tumorigeneses in the cytoplasm. RBBP8 (c.841G>T) is a tumor susceptibility gene and candidate precision medicine biomarker in this familial hereditary cancer syndrome.
... Another lead SNP is located in RBBP8 which encodes Retinoblastoma-binding protein 8 (RBBP8, CtIP), an endonuclease involved in the repair of doublestranded DNA breaks through homologous recombination as well as regulation of G2/M cell-cycle checkpoints as part of the BRCA1-RBBP8 complex 36 . Variation in RBBP8 genotype and regulation has been reported in multiple types of cancer 37,38 . A third SNP was a missense variant in LINC01324, a lncRNA which has been established as a predictor for melanoma progression 39 . ...
Article
Full-text available
Context Estradiol is the primary female sex hormone and plays an important role for skeletal health in both sexes. Several enzymes are involved in estradiol metabolism but few genome-wide association studies (GWAS) have been performed to characterize the genetic contribution to variation in estrogen levels. Objective Identify genetic loci affecting estradiol levels and estimate causal effect of estradiol on bone mineral density (BMD). Design We performed GWAS for estradiol in males (N = 147,690) and females (N = 163,985) from UK Biobank (UKB). Estradiol was analyzed as a binary phenotype; above/below detection limit (175 pmol/L). We further estimated the causal effect of estradiol on BMD using Mendelian randomization. Results We identified 14 independent loci associated (P<5x10 -8) with estradiol levels in males, of which one (CYP3A7) was genome-wide and seven nominally (P<0.05) significant in females. In addition, one female specific locus was identified. Most loci contain functionally relevant genes that have not been discussed in relation to estradiol levels in previous GWAS. For example, SRD5A2, which encodes a steroid 5-alpha reductase that is involved in processing androgens, and UGT3A1 and UGT2B7 which encode enzymes likely to be involved in estradiol elimination. The allele that tags the O blood group, at the ABO locus, was associated with higher estradiol levels. We identified a causal effect of high estradiol levels on increased BMD in both males (P=1.58x10 -11) and females (P=7.48x10 -6). Conclusion Our findings further support the importance of the body's own estrogen to maintain skeletal health in males and in females.
... A survival analysis for this work was impractical given the small set of patient-matched pairs, but future metaanalyses are warranted to determine if gene expression signatures in of overall survival in the advanced setting, especially given the significant transcriptomic shifts observed in this study. The most significant gene set enriched in bone metastasis was an experimental perturbation gene set involving the knockdown of the tumor suppressor RBBP8 42 .RBBP8 (also known as CtIP) binds directly to Rb, mediates cell cycle regulation, helps maintain genomic stability and loss of RBBP8 incurs tamoxifen resistance and sensitizes breast cancer cells to PARP inhibition in vitro[43][44][45][46] . Concordant with the GSEA analysis, bone metastases have significant expression loss of RBBP8, with 45% of cases showing a greater than 2-fold decrease in expression. ...
Preprint
Full-text available
Bone metastases (BoM) are a significant cause of morbidity in patients with Estrogen-receptor (ER)-positive breast cancer, yet characterizations of human specimens are limited. In this study, exome-capture RNA-sequencing (ecRNA-seq) on aged (8-12 years), formalin-fixed paraffin-embedded (FFPE) and decalcified cancer specimens was first evaluated. Gene expression values and RNA-seq quality metrics from FFPE or decalcified tumor RNA showed minimal differences when compared to matched flash-frozen or non-decalcified tumors. ecRNA-seq was then applied on a longitudinal collection of 11 primary breast cancers and patient-matched de novo or recurrent BoM. BoMs harbored shifts to more Her2 and LumB PAM50 intrinsic subtypes, temporally influenced expression evolution, recurrently dysregulated prognostic gene sets and altered expression of clinically actionable genes, particularly in the CDK-Rb-E2F and FGFR-signaling pathways. Taken together, this study demonstrates the use of ecRNA-seq on decade-old and decalcified specimens and defines expression-based tumor evolution in long-term, estrogen-deprived metastases that may have immediate clinical implications. Grant Support Research funding for this project was provided in part by a Susan G. Komen Scholar award to AVL and to SO, the Breast Cancer Research Foundation (AVL and SO), the Fashion Footwear Association of New York, the Magee-Women’s Research Institute and Foundation, and through a Postdoctoral Fellowship awarded to RJW from the Department of Defense (BC123242). NP was supported by a training grant from the NIH/NIGMS (2T32GM008424-21) and an individual fellowship from the NIH/NCI (5F30CA203095). Conflicts of Interest Disclosure No relevant conflicts of interest disclosed for this study. Author Contributions Study concept and design (NP, RJW, SO, AVL); acquisition, analysis, or interpretation of data (all authors); drafting of the manuscript (NP, RJW, SO, AVL); critical revision of the manuscript for important intellectual content (all authors); administrative, technical, or material support (PCL, AB, RB, KRW, WH, JK, MR, ZF, AMB).
... These findings suggest that defects in BRCA1 and RBBP8 are mutually exclusive. Moreover, a high expression level of RBBP8 is maintained in hormone-positive BC, and reduced RBBP8 expression is a mechanism of resistance to tamoxifen 50,51 . These findings predict that RBBP8 may have a role in the transcription and signalling regulation of hormone genes, which may rely on a BRCA1-dependent mechanism. ...
Article
Full-text available
Breast cancer (BC) in patients with germline mutations of BRCA1/BRCA2 are associated with benefit from drugs targeting DNA damage response (DDR), but they account for only 5–7% of overall breast cancer. To define the characteristics of these tumors and also to identify tumors without BRCA mutation but with homologous recombination deficiency (HRD) is clinically relevant. To define characteristic features of HRD tumors and analyze the correlations between BRCA1/BRCA2 and BC subtypes, we analyzed 981 breast tumors from the TCGA database using the signature analyzer. The BRCA signature was strongly associated with the HRD score top 10% (score ≥ 57) population. This population showed a high level of mutations in DDR genes, including BRCA1/BRCA2. HRD tumors were associated with high expression levels of BARD1 and BRIP1. Besides, BRCA1/2 mutations were dominantly observed in basal and luminal subtypes, respectively. A comparison of HRD features in BC revealed that BRCA1 exerts a stronger influence inducing HRD features than BRCA2 does. It reveals genetic differences between BRCA1 and BRCA2 and provides a basis for the identification of HRD and other BRCA-associated tumors.
Article
Tamoxifen (TMX), a selective estrogen receptor modulator, is commonly used in the treatment of hormone-responsive cancers. However, the effects of TMX in anabolic tissues harboring estrogen receptors, such as skeletal muscle, are poorly understood. We report a tandem mass-tag approach to TMX-treated myogenesis in C2C12 cells, a well-characterized model of in vitro murine skeletal muscle differentiation. A longitudinal analysis of >10,000 proteins identified in untreated C2C12 myogenesis revealed a novel subset of 1,062 myogenically regulated proteins. These proteins clustered into five distinct longitudinal expression trends which significantly overlap those obtained in similar analyses performed in human myocytes. We document a specific functional enrichment for adiponectin-signaling unique to TMX-treated myogenesis, as well as a subset of 198 proteins that are differentially expressed in TMX-treated cells relative to controls at one or more stages of myogenesis, the majority of which were involved in steroid and lipid metabolism. Further analysis highlights metallothionein-1 as a novel target of TMX treatment at each stage of C2C12 myogenesis. Finally, we present a powerful, self-validating pipeline for analyzing the total proteomic response to in vitro treatment across every stage of muscle cell development which can be easily adapted to study the effects of other drugs on myogenesis.
Article
Acquired resistance to cytotoxic antineoplastic agents is a major clinical challenge in tumor therapy; however, the mechanisms involved are still poorly understood. In this study, we show that knockdown of CtIP, a corepressor of CtBP, promotes cell proliferation and alleviates G2/M phase arrest in etoposide (Eto)‐treated HCT116 cells. Although the expression of p21 and growth arrest and DNA damage inducible α (GADD45a), which are important targets of p53, was downregulated in CtIP‐deficient HCT116 cells, p53 deletion did not affect G2/M arrest after Eto treatment. In addition, the phosphorylation levels of Ser317 and Ser345 in Chk1 and of Ser216 in CDC25C were lower in CtIP‐deficient HCT116 cells than in control cells after Eto treatment. Our results indicate that CtIP may enhance cell sensitivity to Eto by promoting G2/M phase arrest, mainly through the ATR‐Chk1‐CDC25C pathway rather than the p53‐p21/GADD45a pathway. The expression of CtIP may be a useful biomarker for predicting the drug sensitivity of colorectal cancer cells. CtIP can modulate the etoposide sensitivity of HCT116 cells by promoting G2/M phase arrest, which mainly through the ATR‐Chk1‐CDC25C pathway rather than the p53‐p21/GADD45a pathway.
Article
The ginsenoside Rg1, the primary pharmacologically active ingredient of the traditional Chinese herb ginseng, is widely used in the clinical treatment of diseases of the immune and nervous systems. Recent studies have shown that it also has an antitumor effect. In this study, we explored the effects of Rg1 on hepatoblastoma (HB) and its underlying mechanisms. We demonstrated that Rg1 significantly inhibited HB cell growth both in vivo and in vitro. Mechanistic studies revealed that Rg1 impaired homologous recombination and triggered double-strand breaks in HB cells by directly targeting CtBP-interacting protein (CtIP), a key double-strand break repair factor, which is highly expressed in HB tissues. Moreover, we also demonstrated that Rg1 sensitized HB cells to DNA-damaging agents both in vitro and in vivo. In conclusion, our data not only demonstrate the potential clinical application of Rg1 as a novel chemotherapeutic candidate but also offer a mechanism-based therapeutic option by which DNA-damaging agents can be used in combination with Rg1 to target HB.
Article
Full-text available
cDNA microarrays and a clustering algorithm were used to identify patterns of gene expression in human mammary epithelial cells growing in culture and in primary human breast tumors. Clusters of coexpressed genes identified through manipulations of mammary epithelial cells in vitro also showed consistent patterns of variation in expression among breast tumor samples. By using immunohistochemistry with antibodies against proteins encoded by a particular gene in a cluster, the identity of the cell type within the tumor specimen that contributed the observed gene expression pattern could be determined. Clusters of genes with coherent expression patterns in cultured cells and in the breast tumors samples could be related to specific features of biological variation among the samples. Two such clusters were found to have patterns that correlated with variation in cell proliferation rates and with activation of the IFN-regulated signal transduction pathway, respectively. Clusters of genes expressed by stromal cells and lymphocytes in the breast tumors also were identified in this analysis. These results support the feasibility and usefulness of this systematic approach to studying variation in gene expression patterns in human cancers as a means to dissect and classify solid tumors.
Article
Full-text available
Antiestrogens include agents such as tamoxifen, toremifene, raloxifene, and fulvestrant. Currently, tamoxifen is the only drug approved for use in breast cancer chemoprevention, and it remains the treatment of choice for most women with hormone receptor positive, invasive breast carcinoma. While antiestrogens have been available since the early 1970s, we still do not fully understand their mechanisms of action and resistance. Essentially, two forms of antiestrogen resistance occur: de novo resistance and acquired resistance. Absence of estrogen receptor (ER) expression is the most common de novo resistance mechanism, whereas a complete loss of ER expression is not common in acquired resistance. Antiestrogen unresponsiveness appears to be the major acquired resistance phenotype, with a switch to an antiestrogen-stimulated growth being a minor phenotype. Since antiestrogens compete with estrogens for binding to ER, clinical response to antiestrogens may be affected by exogenous estrogenic exposures. Such exposures include estrogenic hormone replacement therapies and dietary and environmental exposures that directly or indirectly increase a tumor's estrogenic environment. Whether antiestrogen resistance can be conferred by a switch from predominantly ER to ER expression remains unanswered, but predicting response to antiestrogen therapy requires only measurement of ER expression. The role of altered receptor coactivator or corepressor expression in antiestrogen resistance also is unclear, and understanding their roles may be confounded by their ubiquitous expression and functional redundancy. We have proposed a gene network approach to exploring the mechanistic aspects of antiestrogen resistance. Using transcriptome and proteome analyses, we have begun to identify candidate genes that comprise one component of a larger, putative gene network. These candidate genes include NFB, interferon regulatory factor-1, nucleophosmin, and the X-box binding protein-1. The network also may involve signaling through ras and MAPK, implicating crosstalk with growth factors and cytokines. Ultimately, signaling affects the expression/function of the proliferation and/or apoptotic machineries.Keywords: tamoxifen, Faslodex, ICI 182,780, estrogen receptor, coregulator
Article
The BRCA1 tumor suppressor encodes a polypeptide with two recognizable protein motifs: a RING domain near the N terminus and two tandem BRCT domains at the C terminus. Studies of tumor-associated mutations indicate that the RING and BRCT sequences are required for BRCA1-mediated tumor suppression. In addition, recent work has shown that BRCA1 is a potent regulator of RNA transcription and that the BRCT domains are also essential for this activity. Therefore, we used the Sos recruitment system to screen for proteins that bind this critical region of BRCA1. Our results show that the BRCT domains interact in vivo with CtIP, a protein originally identified on the basis of its association with the CtBP transcriptional co-repressor. This finding suggests that BRCA1 regulates gene expression, at least in part, by modulating CtBP-mediated transcriptional repression. Moreover, the in vivo interaction between BRCA1 and CtIP is completely ablated by each of three independent tumor-associated mutations affecting the BRCT motifs of BRCA1. These results indicate that the BRCA1-CtIP interaction may be required for tumor suppression by BRCA1.
Article
Loss of oestrogen-receptor (ER) expression and function may explain the development of tamoxifen resistance in breast cancer. In 72 paired biopsies the immunohistochemical expression of ERs was reduced from 51% before tamoxifen treatment to 29% at progression or relapse, with a reduction in mean H-score from 90 to 61 (P < 0.001, paired t-test). However, there was no significant change in progresterone receptor (PgR) or pS2 expression, markers of ER function. Tumours which developed acquired resistance after primary tamoxifen treatment frequently remained ER+ve at relapse (61%) and continued to express PgR or pS2. In contrast, those which progressed with de novo resistance were all ER-ve, although 6 of these tumours expressed high levels of PgR and/or pS2. In tumours which recurred during adjuvant tamoxifen therapy, the frequency and quantitative level both of ERs and of PgR were significantly reduced. These data imply that separate mechanisms of tamoxifen resistance may exist in these clinical subgroups.
Article
The tumour-suppressor protein BRCA1 mediates its biological functions by interacting with cellular factors1, 2 such as the CtIP polypeptide3, 4, a substrate for the ATM (for 'ataxia telangiectasia mutated') protein kinase5. Li et al.6 report that the BRCA1–CtIP interaction is disrupted by ionizing radiation and by other genotoxic stresses that induce phosphorylation of CtIP by ATM kinase, and that this dissociation of the BRCA1–CtIP complex in turn modulates the transcription of DNA-damage-response genes6. We have shown that the BRCA1-binding domain of CtIP (amino-acid residues 133–369) is distal to the sites that are phosphorylated by ATM kinase (residues S664 and S745)7. We now show that the BRCA1–CtIP complex is stable in irradiated cells, and that the phosphorylated isoforms of CtIP that are induced by ionizing radiation still interact in vivo with BRCA1. We conclude that disruption of the BRCA1–CtIP complex cannot account for induction of DNA-damage-response genes in the way proposed by Li et al.6.
Article
BRCA1 encodes a familial breast cancer suppressor that has a critical role in cellular responses to DNA damage. Mouse cells deficient for Brca1 show genetic instability, defective G2-M checkpoint control and reduced homologous recombination. BRCA1 also directly interacts with proteins of the DNA repair machinery and regulates expression of both the p21 and GADD45 genes. However, it remains unclear how DNA damage signals are transmitted to modulate the repair function of BRCA1. Here we show that the BRCA1-associated protein CtIP becomes hyperphosphorylated and dissociated from BRCA1 upon ionizing radiation. This phosphorylation event requires the protein kinase (ATM) that is mutated in the disease ataxia telangiectasia. ATM phosphorylates CtIP at serine residues 664 and 745, and mutation of these sites to alanine abrogates the dissociation of BRCA1 from CtIP, resulting in persistent repression of BRCA1-dependent induction of GADD45 upon ionizing radiation. We conclude that ATM, by phosphorylating CtIP upon ionizing radiation, may modulate BRCA1-mediated regulation of the DNA damage-response GADD45 gene, thus providing a potential link between ATM deficiency and breast cancer.
Article
Endocrine therapy is effective in approximately one-third of all breast cancers and up to 80% of tumors that express both estrogen and progesterone receptors. Despite the low toxicity, good overall response rates, and additional benefits associated with its partial agonist activity, most Tamoxifen-responsive breast cancers acquire resistance. The development of new antiestrogens, both steroidal and non-steroidal, provides the opportunity for the development of non-cross-resistant therapies and the identification of additional mechanisms of action and resistance. Drug-specific pharmacologic mechanisms may confer a resistance phenotype, reflecting the complexities of both tumor biology/pharmacology and the molecular endocrinology of steroid hormone action. However, since all antiestrogens will be effective only in cells that express estrogen receptors (ER), many mechanisms will likely be directly related to ER expression and signaling. For example, loss of ER expression/function is likely to confer a cross-resistance phenotype across all structural classes of antiestrogens. Altered expression of ERα and ERβ, and/or signaling from transcription complexes driven by these receptors, may produce drug-specific resistance phenotypes. We have begun to study the possible changes in gene expression that may occur as cells acquire resistance to steroidal and non-steroidal antiestrogens. Our preliminary studies implicate the altered expression of several estrogen-regulated genes. However, resistance to antiestrogens is likely to be a multigene phenomenon, involving a network of interrelated signaling pathways. The way in which this network is adapted by cells may vary among tumors, consistent with the existence of a highly plastic and adaptable genotype within breast cancer cells.