Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Research Article

Estrogens and Human Papilloma Virus Oncogenes Regulate Human


Ether-à-go-go-1 Potassium Channel Expression
1 3 4 1 1
Lorenza Dı́az, Irais Ceja-Ochoa, Iván Restrepo-Angulo, Fernando Larrea, Euclides Avila-Chávez,
1 1 1 7 5
Rocı́o Garcı́a-Becerra, Elizabeth Borja-Cacho, David Barrera, Elı́as Ahumada, Patricio Gariglio,
5 5 5 4
Elizabeth Alvarez-Rios, Rodolfo Ocadiz-Delgado, Enrique Garcia-Villa, Elizabeth Hernández-Gallegos,
8 1 1 3
Ignacio Camacho-Arroyo, Angélica Morales, David Ordaz-Rosado, Ethel Garcı́a-Latorre,
4 4 2,8 2
Juan Escamilla, Luz Carmen Sánchez-Peña, Milena Saqui-Salces, Armando Gamboa-Dominguez,
4 6 9 4
Eunice Vera, Marisela Uribe-Ramı́rez, Janet Murbartián, Cindy Sharon Ortiz,
4 6 4
Claudia Rivera-Guevara, Andrea De Vizcaya-Ruiz, and Javier Camacho
1
Departamento de Biologı́a de la Reproducción and 2Departamento de Patologı́a, Instituto Nacional de Ciencias Médicas y Nutrición
Salvador Zubirán; 3Departamento de Inmunologı́a, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional; 4Pharmacology
Section, 5Department of Genetics and Molecular Biology, and 6Toxicology Section, Centro de Investigación y de Estudios Avanzados;

Downloaded from http://aacrjournals.org/cancerres/article-pdf/69/8/3300/2623357/3300.pdf by guest on 23 May 2023


7
Dirección Médica de la U.M.A.E. Hospital de Gineco-Obstetricia No. 3; 8Departamento de Biologı́a. Facultad de Quı́mica, Universidad
Nacional Autónoma de México, Ciudad Universitaria; 9Departamento de Farmacobiologı́a, Centro de Investigación y de Estudios
Avanzados, Mexico D.F., Mexico

Abstract Introduction
Ether-à-go-go-1 (Eag1) potassium channels are potential tools Ether à go-go (EAG) potassium channels possess oncogenic
for detection and therapy of numerous cancers. Here, we properties (1); human Eag1 (hEag1) mRNA shows restricted
show human Eag1 (hEag1) regulation by cancer-associated distribution in healthy tissues expressed mainly in brain; slightly
factors. We studied hEag1 gene expression and its regulation in placenta, testis, and adrenal gland; and transiently in myoblasts
by estradiol, antiestrogens, and human papillomavirus (HPV) (1–3). Interestingly, Eag1 is abundantly expressed in tumor cells,
oncogenes (E6/E7). Primary cultures from normal placentas including cervical, lung, breast, colon, and prostate cancer (2, 4);
and cervical cancer tissues; tumor cell lines from cervix, cho- therefore, Eag1 is suggested as a cancer marker. In addition, an
riocarcinoma, keratinocytes, and lung; and normal cell lines association of Eag1 amplification with reduced overall survival has
from vascular endothelium, keratinocytes, and lung were used. been observed in patients with colon carcinoma (5). Inhibition of
Reverse transcription-PCR (RT-PCR) experiments and South- Eag1 gene expression decreases tumor cell proliferation (1, 6, 7),
ern blot analysis showed Eag1 expression in all of the cancer and inhibition of Eag1 channel activity by imipramine or
cell types, normal trophoblasts, and vascular endothelium, in astemizole has been suggested to decrease cancer cell proliferation
contrast to normal keratinocytes and lung cells. Estradiol and (6–8). Specific block of Eag1 with monoclonal antibodies inhibits
antiestrogens regulated Eag1 in a cell type–dependent manner. tumor cell growth both in vitro and in vivo (9). Thus, Eag1 is also
Real-time RT-PCR experiments in HeLa cells showed that suggested as a cancer therapeutic target. Because no obvious side
Eag1 estrogenic regulation was strongly associated with the effects were observed in the animals treated with monoclonal
expression of estrogen receptor-A. Eag1 protein was detected antibodies (9), the potential therapeutic value of Eag1 channels
by monoclonal antibodies in normal placenta and placental seems to be very high.
blood vessels. Patch-clamp recordings in normal trophoblasts High-risk human papillomavirus (HR-HPV) is the major risk
treated with estradiol exhibited potassium currents resembling factor for cervical cancer (10), and estrogen use is suggested as a
Eag1 channel activity. Eag1 gene expression in keratinocytes likely factor contributing to this cancer (11). Actually, estrogen
depended either on cellular immortalization or the presence receptors (ER) or aromatase enzymes provide advantages for
of HPV oncogenes. Eag1 protein was found in keratinocytes proliferation of tumor cells derived from mammary gland, lung,
transfected with E6/E7 HPV oncogenes. Cell proliferation of and cervix (12, 13). Nevertheless, tamoxifen stimulates proliferation
E6/E7 keratinocytes was decreased by Eag1 antibodies inhibit-
of some cancer cells (14–16).
ing channel activity and by the nonspecific Eag1 inhibitors We reported Eag1 expression in normal cervical samples from
imipramine and astemizole; the latter also increased apoptosis.
women with negative pap smears, including a patient with HPV
Our results propose novel oncogenic mechanisms of estrogen/
infection (4). In addition, Eag1 mRNA was detected in the tumor-
antiestrogen use and HPV infection. We also suggest Eag1 as
free mammary gland surrounding the breast carcinoma tissue (2),
an early indicator of cell proliferation leading to malignancies
in human diverticulitis (which has the potential to change into
and a therapeutic target at early stages of cellular hyper-
colonic cancer), and in crypt cells of a colon cancer mouse model
proliferation. [Cancer Res 2009;69(8):3300–7]
(5). These observations suggest Eag1 expression as an early sign
of cellular hyperproliferation.
Despite the potential clinical relevance of Eag1, its regulation
Note: L. Diaz and I. Ceja-Ochoa contributed equally to this work. by cancer etiologic factors is poorly understood. Here, we studied
Requests for reprints: Javier Camacho, Centro de Investigación y de Estudios
Avanzados del I.P.N., Pharmacology Section, Avenida Instituto Politécnico Nacional hEag1 expression and regulation by some cancer-associated
2508, Mexico D.F., Mexico 07360. Phone: 52-55-57473800, ext. 5416; Fax: 52-55- factors, namely, estrogens, antiestrogens, and HPV oncogenes in
53430106; E-mail: fcamacho@cinvestav.mx.
I2009 American Association for Cancer Research. normal and tumor cells. We studied cervical cancer cells obtained
doi:10.1158/0008-5472.CAN-08-2036 from primary cultures and cell lines from different histogenesis.

Cancer Res 2009; 69: (8). April 15, 2009 3300 www.aacrjournals.org
Potassium Channels and Cancer Risk Factors

‘‘La Raza,’’ following the local ethical considerations. Tissue was placed in
liquid nitrogen to obtain mRNA or fixed in paraformaldehyde for
immunochemistry. For cytotrophoblast culturing, tissue was washed with
saline solution and immediately processed. Neonatal foreskins from
patients registered at Hospital ‘‘Gabriel Mancera’’ were collected following
the local ethical considerations and used to obtain normal keratinocytes
primary cultures.
Cell culture and reagents. Cytotrophoblasts were isolated and cultured
as described (18, 19). Villous tissue was enzymatically dispersed;
cytotrophoblasts were separated on a Percoll gradient, plated in DMEM-
HG with 20% fetal bovine serum (FBS) and incubated at 37jC (95%
humidity and 5% CO2 atmosphere). Human chorionic gonadotrophin
hormone (hCG) concentration was measured with an immunoassay kit
(Immunometrics Ltd.). Human cervical cancer cells were obtained from
previously established primary cultures (4). Tumor cell lines (HeLa, SiHa,
CaSki, INBL, and C33A from cervix; JEG-3 and A-549 from choriocarcinoma
and lung, respectively), as well as bronchial epithelial cells (BEAS) and
human umbilical vascular endothelial cells (HUVEC) from normal lung and
umbilical cord, respectively, were obtained from American Type Culture

Downloaded from http://aacrjournals.org/cancerres/article-pdf/69/8/3300/2623357/3300.pdf by guest on 23 May 2023


Collection and cultured according to manufacturer’s instructions. Cell
treatments were performed, as follows, to avoid participation of exogenous
estrogens present in serum or medium: cells were washed with HBSS
24 h after plating and incubated in supplemented medium (DMEM-HG
without phenol red plus 5% charcoal-stripped FBS and antibiotics). Cells
were incubated with 17h-estradiol (Sigma Chemical Co.) and/or the
antiestrogens ICI 182,780 (Zeneca Pharmaceuticals) or 4-hydroxy-tamoxifen
(Sigma Chemical Co.) during 24 h; ethanol was used as vehicle, and
incubations were stopped by aspirating the culture media and adding Trizol
reagent (Invitrogen) for RNA extraction. Normal keratinocytes were
prepared from neonatal foreskins and grown in the keratinocyte serum-
free medium (Invitrogen) supplemented with 20 mg/mL bovine pituitary
extract and 0.1 ng/mL of epidermal growth factor (Invitrogen); transformed
human keratinocytes (HaCat cells) provided by Dr. Lutz Gissmann [German
Cancer Research Center (DKFZ)] were cultured in the same medium.
Primary human keratinocytes immortalized by amphotropic retroviruses
encoding E6, E7, or E6/E7 oncoproteins of HPV-16 (provided by Dr. Frank
Rösl, DKFZ) were grown in a keratinocyte growth medium (Invitrogen) and
cultured as described (20).
Transient human ER-a transfection and ER Western blot. Expression
vector for human ER-a (pCMV5-hERa) to transfect HeLa cells was provided
Figure 1. Eag1 channels in human placenta. A, Eag1 protein was
immunolocalized (dark blue-violet staining ) in the cytoplasm, nucleus of the by Dr. A.J. Cooney (Baylor College of Medicine). Cells were transfected using
syncytiotrophoblast layer (SC ), and nucleus of vascular endothelial cells (VEC ). PolyFect (Qiagen, Inc.) as described (21). Transcriptional activity of
Magnification, 200. Insert, negative control in the absence of Eag1 transfected ERa was confirmed by a chloramphenicol acetyltransferase
antibody. Image is representative of seven placentas. In some other sections, (CAT) reporter assay (21). Western blots were performed to detect ERa in
Eag1 was also expressed in the plasma membrane (data not shown). B,
Southern blot analysis (top ) from RT-PCR experiments of seven different HeLa, A-549, BEAS, and cervical cancer cells with the primary antibody anti-
normal-term placentas. All of them displayed Eag1 expression. Columns, Eag1 ERa (Santa Cruz). HeLa cells expressing ERa (HeLa-ERa) were subjected to
mRNA relative expression after normalization with the constitutive gene the same treatments as described above.
cyclophilin (cyc ).
PCR amplifications and Southern blot analysis. Total RNA was
isolated from tissues and cell cultures with Trizol reagent. RNA was
reversed transcribed, and PCR amplifications were performed with the
In the case of normal cells/tissues, we selected some normal cell following primers: 5¶-GCTTTTGAGAACGTGGATGAG-3¶ and 5¶-CGAA-
lines and normal human placenta as a very interesting tissue to GATGGTGGCATAGAGAA-3¶, which yielded 475-bp hEag1 reverse tran-
study, because placental development resembles tumor growth (17) scription-PCR (RT-PCR) products. In some cases, Eag amplification was
and estrogens play a pivotal role in placental physiology, and performed using the following sense and antisense primers: 5¶-
noteworthy, because it expresses Eag1 mRNA and the potential role TGGTCCTGCTGGTGTGTG-3¶ and 5¶-ACAACGAGGAGATGTAGACAG-3¶.
of Eag1 in healthy tissues is almost unknown. Our regulation These amplifications yielded a 187-bp hEag1 product. Human Eag1-
transfected Chinese hamster ovary (CHO) cells (provided by Walter
studies unravel potential oncogenic mechanisms of HPV infection
Stühmer, Max-Planck-Institut für Experimentelle Medizin) were used as
and estrogen/antiestrogen use, emphasizing the potential use of positive control. Cyclophilin and b2-microglobulin genes were used as
Eag1 as an early indicator of cell proliferation. On the other hand, constitutive genes and amplified with the following primers: 5¶-CCC CAC
our pharmacologic experiments suggest Eag1 as a therapeutic CGT GTT CTT CGA CAT-3¶ and 5¶-AGG TCC TTA CCG TTC TGG TCG-3¶ for
target at the early stages of cell hyperproliferation. cyclophilin yielding a 453-bp product and 5¶-ACCCCCACTGAAAA-3¶ and 5¶-
ATGATGCTGCTTACATGTCTCGAT-3¶ for b2-microglobulin yielding a 100-
bp product. Identity of the 475-bp Eag RT-PCR product has been already
Materials and Methods determined by nucleotide sequence (4). For Southern blot analysis, PCR
Biological samples. Biopsies from normal human placentas (38–42 wk products were separated in agarose gels, blotted onto nylon membranes,
of gestation, n = 7) were obtained from patients registered at Centro Médico and hybridized with [32P]dCTP-labeled nested probes. Probes were obtained

www.aacrjournals.org 3301 Cancer Res 2009; 69: (8). April 15, 2009
Cancer Research

as previously described and also confirmed by sequence (4). In all cases, EDTA buffer [0.01 mol/L (pH 8.0)] and boiled for antigen retrieval. Slides
filters were washed after 18 h of hybridization and exposed to X-ray films. were blocked for 2 h with levamisole and incubated in the presence of
Real-time PCR amplifications. SYBR Green real-time PCR was 1:50 anti-Eag1 antibody (Ifv001 single-chain antibody coupled with alkaline
performed with 1 AL of cDNA obtained as described above. SYBR Green phosphatase) for 2 h at room temperature. The slides were washed with
reaction was conducted using a QuantiTect SYBR Green PCR reagents kit TBS/Tween, and the specific staining reaction was completed by
(Qiagen) following the manufacturer’s instructions. The following primers incubating the slides in the presence of 5-bromo-4-chloro-3-indolyl
were used: 5¶-TGG TCC TGC TGG TGT GTG-3¶ ( forward), 5¶-ACA ACG AGG phosphate/nitroblue tetrazolium (Roche) in a buffer solution for 1 h at
AGA TGT AGA CAG-3¶ (reverse) for Eag1 (4) and 5¶-ACC CCC ACT GAA room temperature, protected from light, and observed as a dark blue-
AAA GAT GAG TAT-3¶ ( forward), 5¶-ATG ATG CTG CTT ACA TGT CTC violet staining. Sections were counterstained with eosin. Slides were
GAT-3¶ (reverse) for b2-microglobulin (used as constitutive gene and to observed with a Nikon Eclipse E600 microscope using a Nikon Plan Apo
analyze relative Eag1 expression). Data analysis was performed by the 10/0.45 objective lens. Images were acquired with a DMX1200 camera
2 DDCt method. and the Nikon ACT-1 acquisition software at room temperature.
Eag1 immunochemistry. Specific anti-Eag1 antibodies were kindly Keratinocytes expressing HPV oncoproteins (K-E6, K-E6/E7) were pro-
provided by Walter Stühmer and Luis Pardo (Max Planck Institute). This cessed in a very similar manner. After antigen retrieval with EDTA, slides
antibody has been validated and shown to be very specific; it does not were blocked with endogenous peroxidase and phospatase blocker (Dako)
bind to hErg channels and discriminates between Eag1 and Eag2 (9). With for 10 min and then incubated in the presence of 1:100 anti-Eag1 antibody
this antibody, we already reported Eag1 channel expression in human for 1 h at room temperature. The slides were then incubated with MACH
cervical biopsies, cervical cancer cells from primary cultures, and CHO 4 mouse probe (Biocare) in TBS/Tween for 10 min and then incubated
cells transfected with Eag1 in opposite to the absence of Eag1 in with MACH 4 horseradish peroxidase polymer (Biocare) in TBS/Tween for

Downloaded from http://aacrjournals.org/cancerres/article-pdf/69/8/3300/2623357/3300.pdf by guest on 23 May 2023


nontransfected CHO cells (4). Placental biopsies were fixed in a buffered 10 min. The specific staining reaction was completed by incubating the
4% formaldehyde solution, processed as standardized for histology, and slides in the presence of diaminobencidine in buffer reaction solution
embedded in paraffin. Serial sections (4 Am) were mounted on charged (Dako) and observed as a brown staining. Sections were counterstained
glass slides and deparaffinized using xylene and a decreasing series of with hematoxylin (Dako), observed with a 40 objective lens, and
ethanol. After washing with TBS/Tween (pH 7.4), slides were immersed in analyzed as above described for the placental sections.

Figure 2. Eag1 regulation by E2 in


cultured human syncytiotrophoblast and
vascular endothelial cells. Southern
blot analysis from RT-PCR experiments
shows that E2 induced Eag1 mRNA
expression in syncytiotrophoblasts with no
detectable basal Eag1 expression (A )
and antiestrogenic treatment with ICI
182,780 (ICI ) prevented such
up-regulation and decreased Eag1 levels
in a syncytiotrophoblast primary culture
with basal Eag1 expression (B). Eag1 is
endogenously expressed in HUVEC and
up-regulated by E2, and antiestrogenic
treatment prevented such up-regulation
(C) as in trophoblasts. Columns,
Eag1 mRNA relative expression after
normalization with cyclophilin (A–C); Eag1
expression in vehicle was given the
value of 1 in B and C. D, whole-cell
patch-clamp experiments showed
outward potassium currents in
syncytiotrophoblasts treated with E2 (top )
and untreated choriocarcinoma JEG-3
cells (bottom ). Currents elicited at +60 mV
were preceded by negative prepulses
( 140 or 60 mV) and displayed different
activation kinetics, suggesting Eag1
channel activity. Eh = 80 mV,
unsubtracted traces. Figures are
representative of at least three separate
experiments.

Cancer Res 2009; 69: (8). April 15, 2009 3302 www.aacrjournals.org
Potassium Channels and Cancer Risk Factors

thecin (apoptosis inductor) and methanol (necrosis inductor) were used as


controls. Experiments were carried out with the flow cytometer FACS-SYAN
ADP with nine colors (DAKO). Percentages of viable (FITC and PI negative),
apoptotic (FITC positive and PI negative), and late apoptotic (FITC and PI
positive) cells were obtained by quadrant analysis using WinDMI software
version 2.8.
Statistical analysis. Student’s t test or ANOVA, followed by Dunnett’s or
Tukey-Kramer test, was used to compare data between different groups;
P values of <0.05 or <0.01 for the respective tests were considered to be
statistically significant. Analysis was made using GraphPad Prism software
version 3.0.

Results
Regulation of Eag1 by Estradiol in Normal and Tumor
Cells
Eag1 channels in normal human placenta and vascular
endothelium and their regulation by estradiol. We investigated

Downloaded from http://aacrjournals.org/cancerres/article-pdf/69/8/3300/2623357/3300.pdf by guest on 23 May 2023


Eag1 protein expression in normal human placenta and studied the
likely regulation of Eag1 gene expression by 17h-estradiol (E2) in
primary cultures from normal human trophoblasts. Eag1 channels
were strongly expressed in the syncytiotrophoblast layer in both
the cytoplasm and the nucleus in normal placenta (Fig. 1A, dark
blue-violet staining ). Interestingly, we also observed Eag1 channels
in the vascular endothelium, especially in the nucleus (Fig. 1A).
Whereas Eag1 protein expression pattern was very similar among
placentas, relative Eag1 mRNA levels were very different. Figure 1B
shows some placentas with low Eag1 mRNA expression and some
with higher levels. Despite such differences in mRNA levels, we
observed both Eag1 protein and mRNA expression in all of the
placentas studied.
Unexpectedly, Eag1 mRNA was not detected in most of the
Figure 3. Estrogenic regulation of Eag1 expression in HeLa cells and
trophoblasts primary cultures but was induced by estradiol.
participation of ERa. A, real-time RT-PCR experiments show that wild-type HeLa Figure 2A shows Eag1 mRNA up-regulation by E2 at picomolar
cells display endogenous Eag1 expression (as previously reported in ref. 1), but concentrations whereas a higher E2 concentration originated
E2 up-regulates Eag1 expression at some concentrations without the effect of
antiestrogens. However, when HeLa cells were forced to express ERa, Eag1 only a faint Eag1 signal. The antiestrogen ICI 182,780 prevented
was strongly up-regulated by both E2 and antiestrogens (B). Columns, mean; such up-regulation. In cultures displaying endogenous Eag1
bars, SD. *, P < 0.05 compared with control (no E2 treatment). mRNA (Fig. 2B), E2 induced a slight increase in Eag1 expression
and antiestrogen treatment down-regulated endogenous Eag1
Electrophysiology. Whole-cell recordings were acquired from isolated expression both in the presence and absence of E2 (Fig. 2B). As
cells with the patch-clamp technique (22) using an EPC-9 amplifier expected, we also found endogenous Eag1 expression in the
(HEKA Electronics) and analyzed with Igor Pro (WaveMetrics). Patch placental choriocarcinoma cell line JEG-3; however, in this case, we
pipettes (2–3 MV) were obtained by double-pulling Kimax capillaries. did not find a significant estrogenic regulation (data not shown).
Internal solution contained (in mmol/L) 140 KCl, 10 EGTA, and 10 Eag1 channel expression in vascular endothelium prompted us to
HEPES/KOH (pH 7.2). External solution contained (in mmol/L) 2.8 KCl, study Eag1 mRNA expression and regulation by estradiol in
115 NaCl, 2 CaCl2, 2 MgCl2, and 10 HEPES/NaOH (pH 7.2); in some
endothelial cells. Endogenous Eag1 mRNA was found in normal
experiments, we used external solutions containing 10 mmol/L MgCl2.
HUVECs supplemented with 10% FBS (Fig. 2C). Eag1 was up-
Holding potential was 80 mV, unless indicated. Experiments were
performed at room temperature (20–22jC). regulated by E2 ; this effect was inhibited by ICI 182,780, which also
Metabolic activity and apoptosis. K-E6/E7 cells were seeded in 96-well down-regulated endogenous Eag1 levels (Fig. 2C). The effect of
plates, and cell proliferation was assayed by the colorimetric method based estrogens and antiestrogens on Eag1 expression was very similar in
on the conversion of the tetrazolium salts to formazan crystals by both normal cell types (trophoblasts and vascular endothelium).
dehydrogenase activity in active mitochondria [3-(4,5-dimethylthiazol-2-yl)- Endogenous Eag1 channel activity has not been reported for
2,5-diphenyltetrazolium bromide (MTT) cell proliferation kit I, Boehringer most of the normal tissues where it is expressed, including the
Mannheim GmbH]. Cells were incubated during 96 h in culture medium brain. Therefore, we investigated if Eag1 channel activity could be
alone or in the presence of astemizole (10 Amol/L), imipramine (10 Amol/L), recorded in placental-derived cells. Whole-cell patch-clamp record-
DMSO as vehicle (all of these reagents were purchased from Sigma
ings showed no outward voltage-activated potassium currents in
Chemical Co.), or the anti-Eag1 monoclonal antibody 56 (133 nmol/L, also
trophoblast primary cultures in the absence of E2 (data not shown).
provided by Walter Stühmer and Luis Pardo) known to inhibit Eag1 channel
activity (9). MTT (0.5 mg/mL) was added 4 h before completing the whole However, when incubated with E2 (1 pmol/L), normal human
incubation time. Absorbance data were obtained from the resulting colored trophoblasts displayed noninactivating outward currents resem-
solution with a microplate photometer (Sunrise Touchscreen). Apoptosis bling Eag1 channel activity (Fig. 2D, top). Eag1 channels display a
was determined with the Annexin V-FITC kit (Invitrogen Co.) binding to strong Cole-Moore shift (the current elicited with a depolarizing
phosphatidylserine and DNA staining by propidium iodide (PI). Campto- pulse is slower at a more negative prepulse voltage) and a very slow

www.aacrjournals.org 3303 Cancer Res 2009; 69: (8). April 15, 2009
Cancer Research

activation in the presence of extracellular magnesium (23). Figure cells (data not shown). In contrast, Eag1 was present in the lung
2D shows that outward currents elicited at +60 mV are slower cancer cell line A549 and up-regulated with most treatments,
when preceded by a very negative prepulse ( 140 mV) compared especially by antiestrogens (Table 1). Thus, E2 also regulates Eag1
with a more positive prepulse ( 60 mV), recordings were obtained expression in cancer cells in a cell type–dependent manner. Then,
in the presence of 10 mmol/L MgCl2. Because no other potassium we wondered whether a well-identified etiologic factor for some
channels have been reported to display such a strong Cole-Moore cancers could also affect Eag1 expression.
shift, our data suggest Eag1 channel activity in normal human
trophoblasts incubated with E2. Similar currents were elicited in Expression and Potential Role of Eag1 in Cells
the choriocarcinoma JEG-3 cells in the absence of estradiol (Fig. 2D, Expressing HPV Oncogenes
bottom), although, in this case, the current was not that accelerated Eag1 in cervical cancer cell lines and keratinocytes
with a more positive prepulse, as in normal human trophoblasts, expressing HPV oncogenes. We studied hEag1 gene expression
despite of a lower MgCl2 concentration (2 mmol/L) used. in normal keratinocytes and keratinocytes immortalized with the
Estrogenic regulation of Eag1 in cancer cells and the oncogenes E6, E7, or both (K-E6, K-E7, K-E6/E7), as well as in
participation of ERa. We studied estrogenic regulation of Eag1 immortalized keratinocytes lacking HPV and cervical cancer cell
in HeLa cells by real-time RT-PCR. We found Eag1 regulation by E2 lines presenting or lacking HR-HPV.
only at two-hormone concentrations (Fig. 3A) with no effect of the Normal keratinocytes did not express Eag1 (Fig. 4A); however, a
antiestrogens in these HeLa wild-type cells (Fig. 3A). ER-a mRNA prominent Eag1 expression was found in keratinocytes immortal-

Downloaded from http://aacrjournals.org/cancerres/article-pdf/69/8/3300/2623357/3300.pdf by guest on 23 May 2023


expression in these cells was detected by RT-PCR; however, ized with HR-HPV oncogenes, with K-E7 showing the highest
Western blot experiments failed to show the presence of ERa or expression. Eag1 was also found in spontaneously transformed
ERh (data not shown). Therefore, we transfected these cells with keratinocytes lacking HR-HPV (HaCat cells). These data suggest
human ERa (HeLa-ERa cells) and studied again Eag1 regulation by that Eag1 might be regulated by different oncogenic pathways. In
E2. ER-mediated transcription was confirmed by CAT assays (21) in accordance, Eag1 expression was found in cervical cancer cell lines
HeLa-ERa cells cotransfected with a CAT reporter plasmid (data expressing either HPV-16 (CaSki, SiHa) or HPV-18 (INBL) and in a
not shown). Figure 3B shows a remarkable Eag1 up-regulation by cell line lacking HPV (C33A).
E2 in HeLa-ERa cells. ICI 182,780 and tamoxifen, alone or in Potential role of Eag1 channels in cell proliferation and
combination with E2, produced a very high Eag1 up-regulation apoptosis of keratinocytes expressing HPV oncogenes. Immu-
(Fig. 3B). These results suggest ERa activation as one of the nocytochemistry experiments showed an important Eag1 channel
mechanisms of the estrogenic regulation of Eag1. protein expression in keratinocytes immortalized with HPV
Eag1 regulation was also studied in cervical cancer cells from oncogenes (Fig. 4B, brown staining ). We wondered whether Eag1
three human cervical biopsies. Lung cell lines A549 and BEAS-2B channels might have a role in the proliferation and apoptosis of
from cancer and normal tissue, respectively, were used to study these cells. Cell proliferation (assayed by metabolic activity studies)
both Eag1 expression and estrogenic regulation in cells from a very was decreased in K-E6/E7 cells by astemizole and imipramine, two
common cancer influenced by estrogenic signaling (12) but widely used nonspecific Eag1 inhibitors (Fig. 4C). Specific Eag1
histogenically different from placental or cervical tissue. Western inhibition can be achieved by incubating the cells with the
blot analysis showed that cervical and lung cells express ERa (data monoclonal Eag1 antibody 56. Because this antibody does not have
not shown). Cervical cancer cells from primary cultures were an effect on hErg channels or even on the more closely Eag1-related
previously characterized to express Eag1, HPV, and cytokeratins (4), protein, namely Eag2 (9), this is a very fine and specific tool to
supporting their cancer epithelial nature. Table 1 shows screening study potential physiologic roles of Eag1 channels. This antibody
of Eag1 estrogenic regulation in these cells. Effect of E2 and also decreased metabolic activity of K-E6/E7 cells (Fig. 4C).
antiestrogens on Eag1 expression in primary cultures from cervical Astemizole increased K-E6/E7 apoptosis (Fig. 4D).
cancer was similar between two patients (cervical cancer 1 and 2);
interestingly, in a sample from another patient (cervical cancer 3),
such regulation occurred in the opposite direction in all of the Discussion
treatments studied. As expected, Southern blot analysis showed the Eag1 is a potential tool for cancer detection and therapy (1, 2,
absence of Eag1 gene expression in the normal human lung cells 4, 9), and several findings suggest Eag1 as an early marker for
(BEAS-2B). Eag1 was not induced by E2 or antiestrogens in these breast, cervical, and colon cancer (2, 4, 5).

Table 1. Relative Eag-1/cyclophilin expression in cancer cells treated with E2 and/or antiestrogens (from Southern blot
analysis)

Cell type E2 (10 nmol/L) E2 + ICI 182,780 E2 + Tx ICI 182,780 (100 nmol/L) Tx (100 nmol/L)

Cervical (1) 1.42 1.24 0.62 1.44 0.93


Cervical (2) 1.21 2.95 0.08 1.26 0.95
Cervical (3) 0.21 0.61 1.11 0.74 1.03
Lung (A549) 1.59 0.94 1.48 2.83 2.35

NOTE: Relative Eag-1/cyclophilin expression, in comparison with vehicle-treated cells which were given an arbitrary value of 1. Abbreviation: Tx,
tamoxifen.

Cancer Res 2009; 69: (8). April 15, 2009 3304 www.aacrjournals.org
Potassium Channels and Cancer Risk Factors

Figure 4. Eag1 in cervical cancer cells and


effect of Eag1 inhibitors in cell proliferation and
apoptosis of keratinocytes expressing HPV
oncogenes. A, duplex PCR experiments
and densitometric analysis for Eag1 and
b2-microglobulin showed no Eag1 expression
in normal keratinocytes (NK ); in contrast,
keratinocytes expressing HPV oncogenes
(K-E6, K-E7 , or both, K-E6/E7 ) expressed
Eag1 . Eag1 was also found in transformed
keratinocytes without HPV (HaCat and C33A )
and in cervical cancer cell lines with either
HPV-16 (CaSki and SiHa ) or HPV-18 (HeLa
and INBL ). Columns, mean of Eag1 mRNA
relative expression after normalization with
h2-microglobulin (b2-m); bars, SD. Eag1

Downloaded from http://aacrjournals.org/cancerres/article-pdf/69/8/3300/2623357/3300.pdf by guest on 23 May 2023


expression in HeLa cells was given the value
of 1. B, Eag1 protein (brown staining ) in
K-E6/E7 and K-E6 cells can be observed in the
plasma membrane (CM ), nuclear membrane
(Nuc ), or cytoplasm (Cyt ). Negative controls in
the absence of the Eag1 antibody are shown
in the insets. Magnification, 800. C,
nonspecific Eag1 inhibitors astemizole (Astem )
and imipramine (Imip ; 10 Amol/L, 96 h)
decreased cell proliferation of keratinocytes
expressing both HPV oncogenes (K-E6/E7 ),
assayed by metabolic activity; the anti-Eag1
monoclonal antibody 56 (mAb56 ), a very
specific inhibitor of Eag1, also decreased
metabolic activity. D, astemizole increased
apoptosis in these K-E6/E7 cells. Columns,
mean; bars, SD. *, P < 0.05 compared with
control (medium or vehicle).

Placental development resembles malignant tumor behavior that Eag1 promotes vascular endothelial growth factor secretion
(17). We found Eag1 protein in the syncytiotrophoblast layer and and angiogenesis in tumors (29). The effect of other angiogenic
vascular endothelium of placental villi at the plasma membrane, in factors on Eag1 expression in both normal and cancer cells remains
the cytoplasm, and in the nucleus. This distribution was expected to be elucidated. We show the first recordings suggesting Eag1
because Eag1 has a bipartite nuclear targeting signal in its carboxy channel activity in normal human trophoblasts, but more
terminus (24) and perinuclear localization of Eag1 channels has pharmacologic studies are needed to determine the potential
been observed in tumors (2, 6). A COOH terminal fragment of a contribution of other potassium channels to the outward currents
voltage-gated calcium channel translocates to the nucleus and recorded in placental cells.
regulates transcription (25), a similar role for Eag1 remains elusive. Human ERa is necessary for a clear estrogenic regulation of Eag1
Eag1 mRNA was rarely detected in trophoblast primary cultures gene expression in HeLa cells. Only when forced to express ER-a did
despite cell viability confirmation by basal and 8-bromo-cyclic estradiol and antiestrogens clearly up-regulated Eag1. Actually, this
AMP–stimulated placenta hCG secretion (data not shown). is a potential explanation for the lack of estrogenic regulation of
Nevertheless, Eag1 mRNA expression was induced by E2. Chorio- Eag1 in JEG-3 cells, because these cells express very low amounts of
carcinoma JEG-3 cells displayed endogenous Eag1 gene expression; ER-a (30, 31). Thus, concerning ER-a expression, JEG-3 cells could be
however, Eag1 expression was only slightly regulated by estrogens compared with HeLa wild-type cells not transfected with ER-a.
and unaffected by ICI 182,780. Our results suggest that Eag1 might When screening estrogenic regulation of Eag1 mRNA in cells from
participate in the proliferation and/or fusion of trophoblast (as in cervical cancer primary cultures derived from three patients, we
myoblasts fusion; ref. 3), but this deserves further investigations. found similar effects in two cases but opposite effects in another
Eag1 protein was found in placental blood vessel endothelium, culture. Accordingly, tamoxifen opposite effects have been reported
accordingly; endogenous Eag1 mRNA was found in normal in cervical cancer. Tamoxifen treatment inhibited growth of cervical
HUVECs, up-regulated by E2 and down-regulated by ICI 182,780. cancer cell lines, including HeLa cells (32); but in the cervical cancer
This is important because estradiol is also considered as an cell line SFR lacking ERs, tamoxifen stimulated HPV-16 gene
angiogenic factor (26–28). Interestingly, it has been recently found expression and cell proliferation (16). Cervical cancer biopsies

www.aacrjournals.org 3305 Cancer Res 2009; 69: (8). April 15, 2009
Cancer Research

(16%) from patients treated with tamoxifen had a significant at early stages of cell hyperproliferation, for example, in cells initially
decrease in the number of mitotic figures (33). Thus, the variable transformed by HPV.
estrogenic regulation of Eag1 in cervical cancer primary cultures Eag1 channel activity is increased by arachidonic acid in IGR1
might have several explanations, including treatment conditions, melanoma cells (46), and insulin-like growth factor-I increases both
differences in HPV copy number, and different amounts of ERs or ER channel activity and gene expression of Eag1 in MCF-7 breast
phosphorylation; actually, tamoxifen can switch from ERa antago- cancer cells (47). Here, we show for the first time Eag regulation by
nist to ERa agonist if ERa is phosphorylated (34). cancer risk factors in normal cells. A highlighted feature in this
Estrogens stimulate growth and progression of lung tumors (35), study was that Eag1 can be regulated by both estrogens and HPV
and aromatase inhibitors suppress tumor growth in mice with oncoproteins. Eag1 up-regulation by E2 in HeLa cells and some
A-549 lung tumor xenografts (12). Eag1 was not found in BEAS-2B cervical cancer primary cultures suggest that estrogens and HPV
cells in accordance with the absence of Eag1 in normal bronchial might have a synergistic effect on Eag1 expression. Actually, it has
epithelium (2). We found Eag1 expression in A-549 cells in been shown that estrogens contribute to the onset, persistence,
compliance with the findings in lung cancer biopsies (2); in these and malignant progression of cervical cancer in a HPV-transgenic
cells, almost all treatments up-regulated Eag1. A combined therapy mouse model (48). A very interesting issue to address is to know
of antiestrogens and epidermal growth factor receptor (EGFR) the potential participation of Eag1 channels in the mouse cervical
inhibitor has been suggested for lung cancer (36) because of the cancer model mentioned. Another interesting issue is to investigate
functional linkage between estrogens and EGFRs (37). Therefore, the differential concentration-dependent effect of estrogens.

Downloaded from http://aacrjournals.org/cancerres/article-pdf/69/8/3300/2623357/3300.pdf by guest on 23 May 2023


Eag1 up-regulation by antiestrogens might be related to the cross- Concentration-dependent dual effects of estrogens have been
talk between different signal transduction pathways. already observed, for example, on growth hormone synthesis and
Different cervical cancer cell types and keratinocytes expressing interleukin-induced proteoglycan synthesis (49, 50).
HPV oncogenes displayed Eag1 expression in contrast to normal Our experiments unravel novel potential oncogenic mechanisms
keratinocytes. HPV oncoproteins E6 and E7 influence cell prolifer- of estrogen/antiestrogen use and HPV infection. This proposes that
ation by targeting p53 and Rb proteins, respectively (38). The cell Eag1 might be expressed in early stages potentially leading to
lines used present different molecular alterations. HeLa cells cervical transformation, for example, in patients infected with HPV
overexpress c-Myc and Bcl-2 proteins, SiHa cells have high levels or those undergoing estrogen treatment or subjects with low-grade
of pRb and BclXL proteins (39), and keratinocytes expressing E7 intraepithelial lesions.
display high c-Myc levels (20). Eag1 was also found in transformed The data presented herein suggest Eag1 as an early marker of
keratinocytes lacking HPV (HaCat and C33A cells). Interestingly, p53 cellular proliferation leading to malignancies, enhancing potential
and Rb are mutated in C33A cells (40) and HaCat cells have two benefits of using Eag1 in early detection programs. Because cancer is
mutations in p53 (41). Probably Eag1 is down-regulated, at least, a multifactorial disease, detecting the expression of oncogenic genes
through p53 and Rb pathways. Because Eag1 was found in cells or proteins regulated by several cancer risk factors, like Eag1, might
presenting or lacking HPV, these results suggest that Eag1 gene lead to improved diagnosis methods and early detection of
expression is regulated by different factors, which is in accordance malignancies. Finally, our pharmacologic experiments propose
with the Eag1 overexpression observed in very different types of Eag1 as a therapeutic target at early stages of cell hyperproliferation.
tumors (2, 4–6); our results also offer possible alternative
explanations for the oncogenic mechanisms of HPV infection.
Disclosure of Potential Conflicts of Interest
Eag1 protein was found in K-E6/E7 and K-E6 cells, and Eag1 channel
inhibitors affected cell proliferation and apoptosis. Astemizole was No potential conflicts of interest were disclosed.
more effective than impramine in inhibiting cell proliferation and
increasing apoptosis in keratinocytes expressing HPV oncogenes. Acknowledgments
Both drugs have other targets different from Eag1; thus, a plausible Received 6/2/08; revised 1/14/09; accepted 2/16/09; published OnlineFirst 4/7/09.
explanation is a higher affinity of other proteins participating in cell Grant support: CONACyT grants 45753 (J. Camacho) and 45953 (P. Gariglio).
proliferation/apoptosis to astemizole. One of such proteins might be E. Garcı́a-Latorre is a fellow of EDI and COFFA-IPN.
The costs of publication of this article were defrayed in part by the payment of page
hErg channels, which are strongly inhibited by astemizole and have charges. This article must therefore be hereby marked advertisement in accordance
also been suggested to play a role in tumorigenesis (42–45). Further with 18 U.S.C. Section 1734 solely to indicate this fact.
We thank Walter Stühmer and Luis Pardo (Max Planck Institute) for providing
mechanistic studies are required to understand the effect of Eag1-transfected CHO cells and the Eag1 antibodies and for their comments on the
astemizole on cell proliferation and apoptosis. Nevertheless, despite manuscript; Dr. Lutz Gissmann and Dr. Frank Rösl (DKFZ) for providing HaCat cells
of some nonspecific effects of imipramine and astemizole that might and immortalized human keratinocytes expressing E6, E7, or E6/E7 HPV oncoproteins;
Dr. Austin J. Cooney (Baylor College of Medicine) for providing the expression vector
be involved, the effect of these drugs and the monoclonal Eag1 for ERA (pCMV5-hERA); Verónica Cadena for her secretarial work; and Guadalupe
antibody on proliferation and/or apoptosis suggests Eag1 as a target Montiel for her technical assistance.

References myoblasts at the onset of fusion. FEBS Lett 1998;434: 6. DeQueiroz FM, Suarez-Kurtz G, Stühmer W, Pardo LA.
177–82. Ether à go-go potassium channel expression in soft
1. Pardo LA, Del Camino D, Sánchez A, et al. Oncogenic 4. Farias LMB, Ocaña DB, Dı́az L, et al. Ether à go-go tissue sarcoma patients. Mol Cancer 2006;5:42.
potential of EAG K+ channels. EMBO J 1999;18:5540–7. potassium channels as human cervical cancer markers. 7. Spitzner M, Ousingsawat J, Scheidt K, Kunzelmann K,
2. Hemmerlein B, Weseloh RM, DeQueiroz FM, et al. Cancer Res 2004;64:6996–7001. Schreiber R. Voltage-gated K+ channels support prolifer-
Overexpression of Eag1 potassium channels in clinical 5. Ousingsawat J, Spitzner M, Puntheeranurak S, et al. ation of colonic carcinoma cells. FASEB J 2007;21:35–44.
tumours. Mol Cancer 2006;5:41. Expression of voltage-gated potassium channels in 8. Gavrilova-Ruch O, Schönherr K, Gessner G, et al.
3. Occhiodoro TL, Bernheim L, Liu JH, et al. Cloning of a human and mouse colonic carcinoma. Clin Cancer Res Effects of imipramine on ion channels and proliferation
human ether-a-go-go potassium channel expressed in 2007;13:824–31. of IGR1 melanoma cells. J Membr Biol 2002;188:137–49.

Cancer Res 2009; 69: (8). April 15, 2009 3306 www.aacrjournals.org
Potassium Channels and Cancer Risk Factors

9. Gómez-Varela D, Zwick-Wallasch E, Knötgen H, et al. 23. Terlau H, Ludwig J, Steffan R, Pongs O, Stühmer W, non-small cell lung cancer shows enhanced antiprolifer-
Monoclonal antibody blockade of the human Eag1 Heinemann SH. Extracellular Mg2+ regulates activation ative effects. Cancer Res 2005;65:1459–70.
potassium channel function exerts antitumor activity. of rat eag potassium channel. Pflügers Arch Eur J 37. Levin ER. Bidirectional signaling between the estro-
Cancer Res 2007;67:7343–9. Physiol 1996;432:301–12. gen receptor and the epidermal growth factor receptor.
10. Walboomers JMM, Jacobs MV, Manos MM, et al. 24. Pardo LA, Contreras-Jurado C, Zientkowska M, Alves Mol Endocrinol 2003;17:309–17.
Human papillomavirus is a necessary cause of invasive F, Stühmer W. Role of voltage-gated potassium channels 38. zur Hausen H. Papillomaviruses and cancer: from
cervical cancer worldwide. J Pathol 1999;189:12–9. in cancer. J Membr Biol 2005;205:115–24. basic studies to clinical applications. Nat Rev Cancer
11. Moreno V, Bosch FX, Munoz N, et al. Effect of oral 25. Gomez-Ospina N, Tsuruta F, Barreto-Chang O, Hu L, 2002;2:342–50.
contraceptives on risk of cervical cancer in women with Doltmesch R. The C terminus of the L-type voltage- 39. Aguilar-Lemarroy A, Kirchhoff S, Whitaker N, et al.
human papillomavirus infection: the IARC multicentric gated calcium channel Cav1.2 encodes a transcription Differential sensitivity of human papillomavirus type
case-control study. Lancet 2002;359:1085–92. factor. Cell 2006;127:591–606. 16(+) and type 18(+) cervical carcinoma cells to CD95-
12. Weinberg OK, Marquez-Garban DC, Fishbein M, et al. 26. Gagliardi C, Collins DC. Inhibition of angiogenesis by mediated apoptosis. Int J Cancer 2001;93:823–31.
Aromatase inhibitors in human lung cancer therapy. antiestrogens. Cancer Res 1993;53:533–5. 40. Scheffner M, Munger K, Byrne JC, Howley PM. The
Cancer Res 2005;65:11287–91. 27. Haran EF, Maretzek AF, Goldberg I, Horowitz A, state of the p53 and retinoblastoma genes in human
13. Nair HB, Luthra R, Kirma N, et al. Induction of Degani H. Tamoxifen enhances cell-death in implanted cervical-carcinoma cell-lines. Proc Natl Acad Sci U S A
aromatase expression in cervical carcinomas: effects of MCF7 breast-cancer by inhibiting endothelium growth. 1991;88:5523–7.
endogenous estrogen on cervical cancer cell prolifera- Cancer Res 1994;54:5511–4. 41. Lehman TA, Modali R, Boukamp P, et al. p53
tion. Cancer Res 2005;65:11164–73. 28. Dabrosin C, Palmer K, Muller WJ, Gauldie J. Estradiol mutations in human immortalized epithelial-cell lines.
14. Osipo C, Gajdos C, Liu H, Chen B, Jordan VC. promotes growth and angiogenesis in polyoma middle T Carcinogenesis 1993;14:833–9.
Paradoxical actions of fulvestrant in estradiol-induced transgenic mouse mammary tumor explants. Br Cancer 42. Suesbrich H, Waldegger S, Lang F, et al. Blockade of
regression of tamoxifen-stimulated breast cancer. J Natl Treat 2003;78:1–6. HERG channels expressed in Xenopus oocytes by the
Cancer Inst 2003;95:1597–608. 29. Downie BR, Sánchez A, Knotgen H, et al. Eag1 histamine receptor antagonists terfenadine and astemi-
15. Coiret G, Borowiec AS, Mariot P, Ouadid-Ahidouch H, expression interferes with hypoxia homeostasis and zole. FEBS Lett 1996;385:77–80.
Matifat F.The antiestrogen tamoxifen activates BK induces angiogenesis in tumors. J Biol Chem 2008;283: 43. Garcı́a-Ferreiro RE, Kerschensteiner D, Major F,

Downloaded from http://aacrjournals.org/cancerres/article-pdf/69/8/3300/2623357/3300.pdf by guest on 23 May 2023


channels and stimulates proliferation of MCF-7 breast 36234–40. et al. Mechanism of block of hEag1 K+ channels by
cancer cells. Mol Pharmacol 2007;71:843–51. 30. Pepe GJ, Davies WA, Dong KW, et al. Cloning of the imipramine and astemizole. J Gen Physiol 2004;124:
16. Hwang JY, Lin BY, Tang FM, Yu WCY. Tamoxifen 11h-hydroxysteroid dehydrogenase (11h-SD)-gene in the 301–17.
stimulates human papillomavirus type 16 gene expres- baboon: effects of estradiol on promoter activity of 11h- 44. Roy J, Vantol B, Cowley EA, et al. Pharmacological
sion and cell proliferation in a cervical cancer cell line. HSD-1 and -2 in placental JEG-3 cells. Biochim Biophys separation of hEAG and hERG K+ channel function in
Cancer Res 1992;52:6848–52. Acta 1999;1444:101–10. the human mammary carcinoma cell line MCF-7. Oncol
17. Soundararajan R, Rao AJ. Trophoblast ‘pseudo- 31. O’Neil JS, Burrow ME, Green AE, et al. Effect of Rep 2008;19:1511–6.
tumorigenesis’: significance and contributory factors. estrogen on leptin gene promoter activation in MCF-7 45. Bianchi L, Wible B, Arcangeli A, et al. herg encodes
Reprod Biol Endocrinol 2004;2:15. breast cancer and JEG-3 choriocarcinoma cells: selective a K+ current highly conserved in tumors of different
18. Kliman HJ, Nestler JE, Sermasi E, Sanger JM, Strauss regulation via estrogen receptors a and h. Mol Cell histogenesis: a selective advantage for cancer cells?
JF. Purification, characterization, and in vitro differen- Endocrinol 2001;176:67–75. Cancer Res 1998;58:815–22.
tiation of cytotrophoblasts from human term placentae. 32. Grenman S, Shapira A, Carey TE. In vitro response of 46. Gavrilova-Ruch O, Schönherr R, Heinemann SH.
Endocrinology 1986;118:1567–82. cervical cancer cell lines CaSki, HeLa and ME-180 to the Activation of hEAG1 potassium channels by arachidonic
19. Diaz L, Arranz C, Avila E, Halhali A, Vilchis F, Larrea antiestrogen tamoxifen. Gynecol Oncol 1988;30:228–38. acid. Pflügers Arch Eur J Physiol 2007;453:891–903.
F. Expression and activity of 25-hydroxyvitamin D-1 33. Roig LMV, Lotfi H, Olcese JE, Castro GL, Ciocca DR. 47. Borowiec AS, Hague F, Harir N, et al. IGF-1 activates
a-hydroxylase are restricted in cultures of human Effects of short-term tamoxifen administration in hEAG K+ channels through an Akt-dependent signaling
syncytiotrophoblast cells from preeclamptic pregnan- patients with invasive cervical carcinoma. Anticancer pathway in breast cancer cells: role in cell proliferation.
cies. J Clin Endocrinol Metab 2002;87:3876–82. Res 1993;13:2457–63. J Cell Physiol 2007;212:690–701.
20. Aguilar-Lemarroy A, Gariglio P, Whitaker NJ, et al. 34. Michalides R, Griekspoor A, Balkenende A, et al. 48. Brake T, Lambert PF. Estrogen contributes to the
Restoration of p53 expression sensitizes human papil- Tamoxifen resistance by a conformational arrest of the onset, persistence, and malignant progression of cervi-
lomavirus type 16 immortalized human keratinocytes to estrogen receptor a after PKA activation in breast cal cancer in a human papillomavirus-transgenic mouse
CD95-mediated apoptosis. Oncogene 2002;21:165–75. cancer. Cancer Cell 2004;5:597–605. model. Proc Natl Acad Sci U S A 2005;102:2490–5.
21. Larrea F, Garcia-Becerra R, Lemus AE, et al. A-ring 35. Stabile LP, Davis ALG, Gubish CT, et al. Human non- 49. Childs G, Iruthayanathan M, Akhter N, et al.
reduced metabolites of 19-nor synthetic progestins as small cell lung tumors and cells derived from normal Bipotential effects of estrogen on growth hormone
subtype selective agonists for ERa. Endocrinology 2001; lung express both estrogen receptor a and h and show synthesis and storage in vitro . Endocrinology 2005;146:
142:3791–9. biological responses to estrogen. Cancer Res 2002;62: 1780–8.
22. Hamill OP, Marty A, Neher E, Sakmann B, Sigworth FJ. 2141–50. 50. Richette P, Dumontier MF, Francois M, et al. Dual
Improved patch-clamp techniques for high-resolution 36. Stabile LP, Lyker JS, Gubish CT, Zhang W, Grandis JR, effects of 17h-oestradiol on interleukin 1h-induced
current recording from cells and cell-free membrane Siegfried JM. Combined targeting of the estrogen proteoglycan degradation in chondrocytes. Ann Rheum
patches. Pflügers Arch Eur J Physiol 1981;391:85–100. receptor and the epidermal growth factor receptor in Dis 2004;63:191–9.

www.aacrjournals.org 3307 Cancer Res 2009; 69: (8). April 15, 2009

You might also like