You are on page 1of 12

IJC

International Journal of Cancer

Epigenetic dysregulation of KCa3.1 channels induces poor


prognosis in lung cancer
Etmar Bulk1, Anne-Sophie Ay2, Mehdi Hammadi2,3, Halima Ouadid-Ahidouch2, Sonja Schelhaas4, Antje Hascher5,
Christian Rohde5,6, Nils H. Thoennissen5,7, Rainer Wiewrodt5, Eva Schmidt5, Alessandro Marra8, Ludger Hillejan8,
Andreas H. Jacobs4,9, Hans-Ulrich Klein10, Martin Dugas10, Wolfgang E. Berdel5, Carsten Mu
€ller-Tidow5,6*
1
and Albrecht Schwab *
1
Institute of Physiology II, University of M€
unster, M€
unster, Germany
2
Laboratory of Cellular Physiology, EA 4667, SFR CAP-SANTE (FED4231), UFR Sciences, University of Picardie Jules Verne, Amiens, 80039, France
3
Inserm U916, Institut Bergonie, Bordeaux, 33076, France
4
European Institute for Molecular Imaging (EIMI), University of Mu €nster, Mu
€nster, Germany
5
Department of Medicine, Hematology, Oncology and Pneumology, University of Mu €nster, Mu
€nster, Germany
6
Department of Medicine, Hematology and Oncology, University Hospital of Halle (Saale), Halle (Saale), Germany
7
Department of Internal Medicine II and Clinic (Oncology Center), University Hospital Hamburg-Eppendorf, Hamburg, Germany
8
Department of Thoracic Surgery, Niels-Stensen Clinics, Ostercappeln, Germany
9
Department of Geriatric Medicine, Johanniter Hospital, Bonn, Germany
10
Institute of Medical Informatics, University of Mu€nster, Mu
€nster, Germany
Cancer Cell Biology

Epigenomic changes are an important feature of malignant tumors. How tumor aggressiveness is affected by DNA methylation
of specific loci is largely unexplored. In genome-wide DNA methylation analyses, we identified the KCa3.1 channel gene
(KCNN4) promoter to be hypomethylated in an aggressive non–small-cell lung carcinoma (NSCLC) cell line and in patient sam-
ples. Accordingly, KCa3.1 expression was increased in more aggressive NSCLC cells. Both findings were strong predictors for
poor prognosis in lung adenocarcinoma. Increased KCa3.1 expression was associated with aggressive features of NSCLC cells.
Proliferation and migration of pro-metastatic NSCLC cells depended on KCa3.1 activity. Mechanistically, elevated KCa3.1 expres-
sion hyperpolarized the membrane potential, thereby augmenting the driving force for Ca21 influx. KCa3.1 blockade strongly
reduced the growth of xenografted NSCLC cells in mice as measured by positron emission tomography–computed tomography.
Thus, loss of DNA methylation of the KCNN4 promoter and increased KCa3.1 channel expression and function are mechanisti-
cally linked to poor survival of NSCLC patients.

Lung cancer is a leading cause of cancer-related death.1 The 5- low as 10%.2–4 Unfortunately, many patients develop local or
year survival rate of patients with non–small-cell lung cancer distant metastasis relapse even after complete resection. Thus,
(NSCLC), which accounts for 80% of lung cancers, may be as high aggressiveness is an intrinsic feature of many NSCLC
tumors. Adjuvant and palliative chemotherapy are of limited
Key words: KCa3.1, lung cancer, aggressiveness benefit in NSCLC, and novel therapeutic targets are needed.
Additional Supporting Information may be found in the online Individual NSCLC cancers harbor multiple mutations in
version of this article. protein-coding genes, most of which cannot be targeted ther-
*A.S. contributed equally to this work apeutically.5 In contrast, epigenetic changes might be target-
Grant sponsor: Deutsche Krebshilfe; Grant number: 110261, able. Effector mechanisms, e.g., for metastatic spread, could
110262 and 109666; Grant sponsor: Wilhelm-Sander Stiftung; be promising therapeutic targets. Metastasis development
Grant number: 2009.041.2; Grant sponsor: Else-Kr€oner Fresenius originates from subpopulations of tumor cells that acquire
Stiftung; Grant number: 2012_A76; Grant sponsor: DAAD additional features for increased aggressiveness. These fea-
(PROCOPE) (to A.S. and H.O.A.); Grant sponsor: Interdisciplinary tures depend on changes in gene expression, including that
Centre for Clinical Research (IZKF, core unit PIX); Grant sponsor: of ion channels,6–8 presumably often by epigenetic mecha-
Cells-in-Motion Cluster of Excellence (EXC 1003–CiM), University nisms rather than metastasis-specific mutations.9–11 Altered
of M€ unster, Germany DNA methylation of specific loci as a stable epigenetic mark
DOI: 10.1002/ijc.29490 is a likely culprit in many instances. DNA hypermethylation
History: Received 21 July 2014; Accepted 26 Jan 2015; Online 20 and gene silencing occur in CpG islands and promoters of
Feb 2015 tumor suppressors.9 DNA hypomethylation is thought to
Correspondence to: Albrecht Schwab, Institute of Physiology II, occur mainly in intergenic regions, with effects mainly on
University of M€ unster, M€
unster, Germany, Tel.: 149 251 835 5329, genome stability.9 However, the potential pathophysiological
E-mail: aschwab@unimunester.de and clinical relevance of DNA hypomethylation of specific

Int. J. Cancer: 137, 1306–1317 (2015) V


C 2015 UICC
Bulk et al. 1307

What’s new?
Another possible avenue for thwarting metastasis has been found: the potassium channel KCa3.1. Aggressiveness in lung can-
cer (NSCLC) is linked to methylation of the potassium channel promoter gene. When the promoter is under-methylated, more
of the potassium channel protein is produced, and the cancer cells proliferate and spread more aggressively. The authors also
showed that blocking KCa3.1 stops the cells from dividing and spreading, both in vitro and in mice, suggesting that KCa3.1
could make a promising therapeutic target.

genes and promoters and their mechanistic link to aggres- clinical information were obtained from a pathology program
siveness of lung cancer cells remain unresolved. affiliated with the Osterkappeln Hospital. Ethics committee
Here, we describe the epigenetic dysregulation of the approval was obtained (4XM€ uller1 and 2006-150-f-S).
KCNN4 gene coding for the calcium-activated KCa3.1 channel
in lung cancer. High-level expression of KCa3.1 channels fre- Gene expression analyses by quantitative real-time
quently occurs in aggressive NSCLC cells. KCa3.1 channels reverse transcription polymerase chain reaction
increase migratory activity by elevating the driving force for Quantitative real-time reverse transcription polymerase chain
Ca21 influx. Their inhibition strongly impairs growth of tumor reaction (RT-PCR) was carried out to determine gene expres-
xenografts. KCNN4 DNA hypomethylation and KCa3.1 overex- sion in A549 cell lines and in fresh frozen patient samples as
pression are intimately linked to poor prognosis in NSCLC. described.14 KCa3.1 was detected with the following primers:
forward 50 -CCTTTCAGACACACTTTGGCTGATCC-30 and
Material and Methods reverse 50 -CAGTGCTAAGCAGCTCAGTCAGG-30 . KCa3.1

Cancer Cell Biology


Cell culture channel expression was normalized to that of GAPDH.
A549 lung adenocarcinoma cells were cultured at 37  C
under 5% CO2 in Dulbecco’s modified Eagle’s medium (Invi- Western blot analysis
trogen, Carlsbad, CA) supplemented with 10% fetal calf Cells were lysed in radioimmunoprecipitation assay buffer (50
serum. The generation of the highly aggressive A549 cell line, mmol/l Tris, 150 mmol/l NaCl, 0.1% sodium dodecyl sulfate,
designated as A549-3R, was described previously.12 0.5% sodium deoxycholate, 1% NP-40 and protease inhibitors
from Roche, Germany). Proteins were detected using the follow-
Gene expression analyses of published microarray ing antibodies: rabbit anti-KCa3.1 (# AV35098; Sigma, St. Louis,
expression data MO), mouse anti-GAPDH (# MA1-16757; Thermo Scientific,
Published gene expression data from Stage I and II lung can- Waltham, MA) and mouse anti-b-actin (# A2228, Sigma) as
cer patients and normal lung tissue were analyzed first antibodies, and goat anti-mouse and goat anti-rabbit
(GSE31210).13 The robust multiarray average normalized (Sigma, St. Louis, MO) antibodies as secondary antibodies.
data were analyzed for KCNN4 expression (Affymetrix probe:
204401_at) and its relation to clinical parameters. Prognostic Immunofluorescence
analysis for overall survival was carried out by log-rank test A549 cells were fixed in 3.5% paraformaldehyde (in
or Cox proportional hazard models for univariate and multi- phosphate-buffered saline) and blocked for 30 min with goat
variate analyses, respectively. serum. After incubation with the first antibody (anti-KCa3.1
1:1,000) for 1 hr at room temperature, cells were incubated
Microarray gene expression analyses of A549 cells with the secondary antibody (1:200; goat anti-rabbit labeled
For gene expression analyses of the parental and highly meta- with Alexa Fluor 488, Invitrogen) for 45 min. Cells were cov-
static A549 cell lines, we used the human Gene 1.0 ST Array ered in Vectashield (Vector Laboratories, Germany), and
(Affymetrix, Santa Clara, CA). In brief, total RNA was iso- images were taken as described.15
lated from both A549 cell lines using RNeasyV R Mini Kit

(QIAgen, Hilden, Germany). We used a class comparison DNA methylation analysis


tool in BRB ArrayTools (v4.1.0) to examine differential A549 and A549-3R were analyzed for DNA methylation dif-
expression in both A549 cell lines. ferences by using reduced representation bisulfite sequencing
(RRBS), which was performed and analyzed as described.12
NSCLC patient cohorts DNA methylation in paraffin-embedded and fresh frozen
Fresh frozen lung cancer tissue and matching normal lung matched tumor/normal primary lung cancer specimens was
tissue were obtained from patients at the time of surgery fol- analyzed using the Infinium HumanMethylation450 K Bead-
lowing informed consent. Samples were snap-frozen in liquid Chip (RevB, Illumina, San Diego, CA). DNA was extracted
nitrogen. Tumor cell content of >70% was verified before from paraffin-embedded specimens using 10-mm sections.
samples were prepared for DNA and RNA extraction. Ano- Every tenth section was stained with hematoxylin and eosin
nymized paraffin sections from NSCLC patients with related and analyzed for tumor cell content. Methylation levels

Int. J. Cancer: 137, 1306–1317 (2015) V


C 2015 UICC
1308 KCa3.1 channel and poor lung cancer prognosis

(termed b-values) were calculated using the Methylation adhere for 3 hrs under growth conditions. When applicable,
Module v1.0 of the GenomeStudio software package (Illu- cells were stimulated with 100 ng/ml epidermal growth factor
mina). Fresh frozen specimens from matched normal and (EGF) and/or treated with DMSO or TRAM-34 (10 mmol/l) 10
tumor (n 5 16 pairs) and paraffin-embedded specimens min before starting the experiment. Images were captured in
(n 5 186) were analyzed separately. Methylation levels for 10-min intervals for 10 hrs using the ZEISS microscope Axio-
CpG sites located at KCNN4 gene of matched normal and vert 40C (Carl Zeiss, Oberkochen, Germany), linked to a video
tumor samples were analyzed using beta regression models camera (Hamamatsu, Germany). The analysis of single cells was
(R package betareg). Methylation levels of paraffin-embedded performed as described previously.15 At least three independent
samples were imported into SPSS (SPSS, Chicago, IL) and experiments were performed for each experimental condition.
analyzed with regard to clinical parameters and survival.
Measurements of the intracellular Ca21 concentration
siRNA transfection Measurements of the intracellular calcium concentration
Transfection of A549 cells was performed using the Amax- ([Ca21]i) of A549 cells were performed by means of ratio imag-
aTM NucleofectorTM technology. The following sequences for ing with the fluorescent Ca21 indicator fura-2-AM. Cells were
siRNA were used to diminish KCa3.1 expression in both loaded with 3 mmol/l fura-2-AM for 45 min at 37  C. The
A549 cell lines: sense 50 -UGUAAAGCUUGGCCACGAAC-30 , experiment started with superfusion of Ringer’s solution (in
antisense 50 -GUUCGUGGCCAAGCUUUACA-30 . A nontar- mmol/l: NaCl 122.5, KCl 5.4, CaCl2 1.2, MgCl2 0.8, HEPES 10
geting siRNA (siGENOME #1, Dharmacon, Lafayette, CO) and D-glucose 5.5 at pH 7.4) on an inverse microscope (ZEISS
was used as a negative control. Axiovert TV 100, Carl Zeiss; equipped with a 340 objective),
followed by Ca21-free solution (in mmol/l: NaCl 122.5, KCl 5.4,
Cancer Cell Biology

Patch clamp experiments MgCl2 0.8, HEPES 10 and EGTA 5 at pH 7.4). To release Ca21
Untransfected or transfected A549 cells were seeded overnight from intracellular stores, 1 mmol/l thapsigargin was added until
at a density of 6,000 cells per cm2. Whole cell currents or the basal steady state ratio was reached. Then, the cells were
membrane potential were recorded with an Axopatch 200 B superfused with Ringer’s solution containing 5 mmol/l Ca21,
patch-clamp amplifier and a Digidata 1322 interface (Molecu- followed by Ringer’s solution containing 5 mmol/l Ca21 supple-
lar Devices, Sunnyvale, CA). PClamp 9 software (Molecular mented with 10 mmol/l TRAM-34. To test the contribution of
Devices) was used for voltage control, data acquisition and other K1 channels, cells were depolarized with modified Ring-
analysis. Resistance of the borosilicate fire-polished pipettes er’s solution containing 50 mmol/l KCl (substituting NaCl). Flu-
(HirschmannV R Laborger€
ate) was 3–5 MX. Whole cell currents orescence was excited alternately at 340 nm and 380 nm, using
were allowed to stabilize for 5 min before being measured. a monochromator. Data acquisition was controlled by Metafluor
Currents were recorded during ramps of 250 ms duration software (Visitron Systems, Puchheim, Germany). All experi-
from 2120 to 1 40 mV, starting from a holding potential of ments were carried out at room temperature.
240 mV. The bath and pipette solutions contained (in mmol/
l): bath: NaCl 145, KCl 5, CaCl2 2, MgCl2 2, HEPES 10 and Animal experiments
glucose 5 at pH 7.4 (NaOH); pipette: KCl 150, HEPES 10, All animal procedures were approved by the animal care com-
EGTA 0.1 and MgCl2 2 at pH 7.2 (KOH). The free Ca21 con- mittee of the local government (North Rhine-Westphalia State
centration of the pipette solution amounted to 100 nmol/l. Agency for Nature, Environment and Consumer Protection).
The cells were continuously superfused with control or test A549-3R cells at a concentration of 23 106 cells were
solutions at room temperature. subcutaneously inoculated at three positions into the
shoulder region of 8-weeks-old female NMRI nu/nu mice
Proliferation assay (Janvier Labs, Paris, France). Xenograft growth was deter-
The proliferation rate of A549 cells was determined after 24, mined by caliper measurements. When tumor size reached
48 and 72 hrs. A549 cells at a concentration of 3 3 105 cells 100 mm3, treatment was started. 120 mg/kg senicapoc or
were seeded on 35-mm dishes and starved in serum-free DMSO (total volume of 50 ml) was injected intraperitoneally
medium for 24 hrs. The next day, growth medium was added every second day. During the experimental procedures, body
(including different amounts of TRAM-34 or its solvent dime- weight and general health of the mice were monitored.
thylsulfoxide (DMSO)). Media were replaced after 24 hrs and
48 hrs, and proliferation was analyzed by counting, after the Positron emission tomography–computed tomography
cells were harvested. All experiments were repeated four times. imaging and analysis
Proliferation of tumors was assessed by 18F-fluorodeoxy thy-
Single cell migration analysis midine (FLT) positron emission tomography (PET) before
A549 cells were seeded on a collagen matrix (containing 13 and 7 days after treatment initiation. Radiotracer synthesis
RPMI, 10 mmol/l HEPES, 0.02 mg/ml laminin, 0.04 mg/ml and PET and computed tomography (CT) image acquisition
fibronectin, 0.01 mg/ml collagen IV, 0.01 mg/ml collagen III and coregistration were performed as described previously.16
and 0.8 mg/ml collagen I at pH 7.4) and were allowed to The anatomical information of the CT images was used to

Int. J. Cancer: 137, 1306–1317 (2015) V


C 2015 UICC
Bulk et al. 1309

Cancer Cell Biology


Figure 1. DNA hypomethylation of the KCNN4 promoter is associated with increased KCa3.1 expression in aggressive A549 cells. (a) RRBS of A549
cells. The three major regions covered by RRBS, phylogenetic conservation and major regulatory tracks from the UCSC browser are depicted. Transcrip-
tional start site (TSS) of the KCNN4 gene and the start of the first exon are indicated. The relevant CpGs are indicated below the tracks. (b) mRNA
expression of KCa3.1 channels in A549 cells was analyzed by real-time RT-PCR. (c, d) Western blot of KCa3.1 protein expression in A549 cells and its
quantification (n 5 5). *p<0.05. (e) Immunofluorescence of KCa3.1 channels in A549 cells. Each (green) dot indicates the localization of a single KCa3.1
channel protein. [Color figure can be viewed in the online issue, which is available at wileyonlinelibrary.com.]

delineate the contours of the tumors and determine the methylation analysis revealed that many of the altered pro-
respective tumor volumes. 18F-FLT uptake was calculated as moters and genes showed marked DNA hypomethylation.12
maximal percentage injected dose (%IDmax/ml). The comparison of DNA methylation with microarray gene
expression data revealed several genes with decreased pro-
Statistical analysis moter methylation and increased mRNA expression. One of
Experimental data are shown as mean 6 standard error of the top-ranking genes was KCNN4 that encodes the Ca21-
mean. The mean values of two groups were compared by the activated K1 channel KCa3.1, which was expressed 1.95 times
Student’s t-test. Mean values of more than two groups were higher in A549-3R than in A549-0R (p 5 0.00019; GSE52143).
tested using one-way analysis of variance. A p-value <0.05 Three regions upstream and downstream of its transcriptional
was considered significant. start site showed significant loss of DNA methylation in the
more aggressive A549-3R cell line (Fig. 1a). The most striking
Results difference in DNA methylation was observed within a 100-bp
Hypomethylation of the KCNN4 promoter and increased region 2,000 bp upstream of the transcriptional start site.
expression of the KCa3.1 channel in highly aggressive lung This highly conserved and CpG-rich region bears important
adenocarcinoma cells DNA regulatory activity as evidenced by the presence of
We recently developed a model of human lung adenocarci- DNAse clusters, frequent transcription factor binding sites
noma with increased aggressiveness.12 Genome-wide DNA and H3K27 acetylation in the Encode project (Fig. 1a).

Int. J. Cancer: 137, 1306–1317 (2015) V


C 2015 UICC
1310 KCa3.1 channel and poor lung cancer prognosis
Cancer Cell Biology

Figure 2. KCa3.1 expression is increased in aggressive A549 cells and in tumor samples of NSCLC patients. (a) Boxplot of microarray mRNA
expression data from GSE31210 from 226 NSCLC patients and 20 normal lung samples. (b) mRNA expression levels of KCa3.1 channels
from 15 primary NSCLC tumor samples (matched to normal tissue samples from the same patients) were analyzed by real-time RT-PCR. The
ratios of tumor/matched normal lung tissues are indicated. (c) Methylation status of the KCNN4 gene for 16 matched (tumor vs. normal)
DNA samples from NSCLC. The seven CpGs within the promoter region (indicated in Fig. 1a) are numbered. (d) Methylation levels (CpG#3)
of 16 matched (tumor vs. normal) DNA samples from NSCLC patients with regard to the clinical stage. [Color figure can be viewed in the
online issue, which is available at wileyonlinelibrary.com.]

Real-time RT-PCR (Fig. 1b) confirmed that DNA hypome- CpG sites within promoter/first exon). In contrast, increased
thylation was associated with the induction of KCa3.1 mRNA DNA methylation in NSCLC samples was observed in the
expression in the more aggressive A549-3R cell line. This cor- gene body, a situation which is often associated with increased
responded with increased protein expression as revealed by mRNA expression.17 In Figure 2d, we present the differences
immunoblot (Figs. 1cd) and by immunofluorescence staining of the methylation of CpG#3 (CG14066757) in normal and
(Fig. 1e). Importantly, KCa3.1 channel expression was also sig- tumor tissue samples. A significant hypomethylation was
nificantly increased in samples from NSCLC patients. Figure observed in tumor samples (p < 0.001; beta regression), which
2a compares mRNA expression data from published microar- tended to be more pronounced in advanced stages. An inverse
ray analyses (GSE31210) in normal lung and NSCLC tissue. correlation between methylation and expression was observed
Similar findings were obtained by real-time RT-PCR in our in matched (normal/tumor) specimens from NSCLC patients
own cohort of primary NSCLC tumors (Fig. 2b). In most of (Pearson’s correlation coefficient r 5 20.70).
the 15 primary tumor samples from NSCLC patients, the We next analyzed 186 samples of patients with NSCLC
KCa3.1 mRNA expression was higher than in the matched using the 450K DNA methylation arrays, focusing on the
normal tissue samples from the same patients. seven CpGs located within the KCNN4 promoter region
(Supporting Information Tables S1 and S2). CpG locations
KCNN4 hypomethylation and increased Kca3.1 channel are indicated in Figure 1a. Gender (145 male and 41 female
expression predict poor prognosis of NSCLC patients patients), clinical performance status (Eastern Cooperative
We next analyzed DNA methylation of the KCNN4 gene in Oncology Group performance status), age or smoking status
16 matched NSCLC tumor and normal lung specimens. The was not associated with altered DNA methylation of the
KCNN4 promoter region (Fig. 2c) was significantly DNA CpGs located in the KCNN4 promoter. This also applied for
hypomethylated in tumor samples (p < 0.000001 for all seven the clinical stage and the histological type. The clinical and

Int. J. Cancer: 137, 1306–1317 (2015) V


C 2015 UICC
Bulk et al. 1311

Cancer Cell Biology


Figure 3. Loss of methylation in the KCNN4 gene and increased KCa3.1 channel expression predict a poor outcome for NSCLC patients. (a)
DNA methylation of the KCNN4 promoter. Patients were dichotomized into two groups with low and high DNA methylation levels of the
KCNN4 promoter. The Kaplan–Meier plot shows progression-free survival for lung adenocarcinoma patients who underwent complete resec-
tion (n 5 118). (b) Overall survival analysis for patients with low versus high DNA methylation levels of the KCNN4 gene promoter. (c) A multi-
variate analysis (Cox-regression) for overall survival of lung cancer patients was performed that included KCNN4 promoter DNA methylation.
Hazard ratios with 95% confidence intervals are indicated as well as p-values. (d) Kaplan–Meier survival chart for progression-free survival of
204 lung adenocarcinoma patients with regard to KCa3.1 expression. mRNA levels were dichotomized, and high- versus low-level expressions
were analyzed. Microarray data were obtained from GSE31210. (e) Overall survival analysis with regard to low/high KCa3.1 (KCNN4) mRNA
expression in the same patient cohort. [Color figure can be viewed in the online issue, which is available at wileyonlinelibrary.com.]

pathological characteristics of the patients are listed in the The corresponding values after 5 years were 37% and 66%,
Supporting Information Table S3. respectively. Accordingly, overall survival was significantly
We next investigated potential association of CpG methyl- decreased for patients whose tumor samples showed
ation level and progression-free survival. The survival analysis decreased DNA methylation at CpG2 (p 5 0.001). Median
included all patients with a non-squamous cell histology survival was 923 days (2.5 years) for those with low DNA
(n 5 118) because the A549 cells were also of adenocarci- methylation, whereas 80% of patients whose tumor samples
noma origin. The relative DNA methylation level of the had high DNA methylation were still alive at this time point
KCNN4 region in lung adenocarcinoma was highly variable, (Fig. 3b). The corresponding 5-year survival rates were 37%
ranging from 7% to 93% (Supporting Information Table S4). and 72% for patients with low and high DNA methylation of
Methylation levels of CpG2 (CG01424145) positively corre- their tumors, respectively. The association of CpG
lated with that of other neighboring CpGs (Supporting Infor- (cg01424145) with progression-free and overall survival was
mation Table S5), and thus, this CpG2 was chosen for also evident when only patients with Stage I and II cancer
further analyses. Methylation levels of CpG2 (CG01424145) were analyzed (Supporting Information Fig. S1).
were dichotomized using the median methylation level (30%) The association of CpG2 (cg01424145) with survival was
as threshold. The methylation status was strongly associated evident in univariate and in multivariate analyses. A Cox-
with progression-free survival. Patients experienced a median regression analysis included CpG2 (cg01424145) as well as
progression-free survival of 756 days (2.1 years) when the Eastern Cooperative Oncology Group performance status,
DNA methylation of CpG2 in tumor biopsies was low, smoking status, grading, age, tumor stage, resection status
whereas 60% of the patients were disease-free even after 8 and gender. Loss of DNA methylation of the CpG2
years, when DNA methylation was high (p 5 0.002; Fig. 3a). (cg01424145) was an independent predictor of shortened

Int. J. Cancer: 137, 1306–1317 (2015) V


C 2015 UICC
1312 KCa3.1 channel and poor lung cancer prognosis

progression-free survival, with a hazard ratio of 2.24 KCa channel-dependent hyperpolarization of the mem-
(p 5 0.018). In a similar Cox-regression analysis for overall brane potential can have an important impact on the intra-
survival, DNA hypomethylation predicted increased risk of cellular Ca21 concentration of nonexcitable cells by
poor survival (Fig. 3c hazard ratio 2.19, p 5 0.031, Wald test). increasing the electrical driving force for Ca21 entry.18
Analysis of published mRNA expression data from another Because activation of KCa3.1 channels by 1-EBIO hyperpolar-
patient cohort (GSE31210) also revealed an association of ized the membrane potential of A549-3R cells (Fig. 4c), we
KCa3.1 with survival in NSCLC. We focused on patients with investigated whether KCa3.1 channel activity also impacted
Stage I or II adenocarcinoma who had undergone complete on [Ca21]i of A549 cells. Initially, we measured [Ca21]i
resection with negative margins (n 5 204). Tumor samples of under baseline conditions. Consistent with the differences of
patients with high level of KCa3.1 mRNA expression (high and the membrane potential, [Ca21]i was higher in prometastatic
low expression dichotomized at the median) experienced A549-3R than in parental A549-0R cells (43.2 6 0.5 nmol/l
shortened time to first progression (p 5 0.00001; Fig. 3d) and versus 30.3 6 0.3 nmol/l; n 5 20 cells each; data not shown).
reduced overall survival (p 5 0.003; Fig. 3e). This was evident We exposed the cells to thapsigargin (1 mmol/l) in Ca21-free
in both univariate and multivariate analyses. The multivariate solution to deplete intracellular Ca21 stores. On adding 5
analyses included KCNN4 gene expression (dichotomized at mmol/l Ca21, [Ca21]i rose immediately in both cell lines and
median), stage, gender and smoking history. remained almost stable (Figs. 4de). At this point, the 5
Thus, DNA hypomethylation correlated with increased mmol/l Ca21-containing solution was supplemented with 10
KCa3.1 channel mRNA and protein expression in cell lines mmol/l TRAM-34. In both A549 cell lines, the [Ca21]i ratio
and in primary tumor samples of NSCLC patients. DNA dropped by 22% (Figs. 4df A549-0R: Dratio 5 21.15 6 0.10;
hypomethylation of the KCa3.1 channel promoter and A549-3R: Dratio 5 21.25 6 0.13; n > 34 cells). Finally, we
Cancer Cell Biology

increased mRNA expression determined in independent superfused the cells with a modified Ringer’s solution con-
patient cohorts were associated with a poor prognosis in taining 50 mmol/l KCl. This led to a further reduction in
NSCLC. We next assessed the functional impact of elevated [Ca21]i (Figs. 4df A549-0R Dratio 5 21.27 6 0.11 and A549-
KCa3.1 channel expression on NSCLC cell aggressiveness. 3R Dratio 5 21.17 6 0.08), indicating that A549 cells express
other K1 channels as well.19
KCa3.1 channels are functionally active in highly
metastatic A549 cells KCa3.1 channel blockade reduces A549 cell proliferation
We performed patch clamp experiments to identify functional and migration
KCa3.1 channel expression (Fig. 4a, upper panels). Using the Next, we determined the role of KCa3.1 channels for tumor
KCa3.1channel activator 1-EBIO (200 mmol/l), KCa3.1 currents cell growth. Proliferation of A549 cells was dose-dependently
were activated in both A549 cell strains that were transfected inhibited by TRAM-34. Both cell strains responded with a
with the scrambled control siRNA. KCa3.1 currents were more slight inhibition of their growth rate when exposed to 10
than twice as high in aggressive A549-3R cells than in the mmol/l TRAM-34 (Fig. 5a). The treatment with 30 mmol/l
parental A549-0R cells. The activated KCa3.1 current had a TRAM-34 inhibited proliferation of both cell lines more
reversal potential of 279.1 6 1.6 mV (n 5 20) in A549-3R and effectively. Overall, DMSO-exposed A549-0R and A549-3R
276.0 6 2.4 mV (n 5 15) in A549-0R cells, respectively. The cells grew three times and four times faster, respectively,
activation of KCa3.1 currents was completely blocked with the compared with cells treated with TRAM-34.
KCa3.1 channel blocker TRAM-34. In cells transfected with We applied live cell imaging to monitor migration of
siRNA against KCa3.1 channels, 1-EBIO essentially failed to A549 cells. As shown in Figures 5ef, the maximum speed was
activate KCa3.1-mediated currents (Fig. 4a, lower panels and reached after 7 hrs of stimulation. The cell paths of single
Fig. 4b). At 140 mV, the amplitude of 1-EBIO-sensitive cur- cells, normalized to common starting points are presented in
rents was 1,605 6 228 pA in highly metastatic A549-3R cells Figure 5b. The more aggressive A549-3R cells migrated over
(n 5 20) and 778 6 152 pA in A549-0R cells with low meta- a longer distance compared with their parental cells
static potential (n 5 15; Fig. 4b). siRNA against KCa3.1 reduced (117 6 14 versus 75 6 8 mm, Figure 5c, Supporting Informa-
the amplitude to 542 6 111 pA in the A549-3R (n 5 20) and tion Videos S1 and S2). When treated with the KCa3.1 inhibi-
to 170 6 30 pA in the A549-0R cells (n 5 15). tor TRAM-34 (10 mmol/l), the migration of A549-3R cells
Figure 4c demonstrates that the resting membrane poten- resembled that of A549-0R cells (Supporting Information
tial of the pro-metastatic A549-3R cells was more negative Video S3). TRAM-34 reduced the translocation of A549-3R
than that of the parental cells. Hyperpolarization of the rest- cells by 33% (78 6 9 mm), whereas that of the parental A549-
ing membrane potential with 200 mmol/l 1-EBIO was largely 0R cells was reduced only by 25% (56 6 6 mm). Furthermore,
blunted in cells transfected with siRNA against KCa3.1 chan- during the last 3 hrs of the experiment, the speed of highly
nels (Fig. 4c). Taken together, highly metastatic A549-3R cells metastatic A549-3R cells was reduced from 0.55 6 0.03 to
expressed more functional KCa3.1 channels in the plasma 0.42 6 0.04 mm/min when treated with 10 mmol/l TRAM-34
membrane than the parental A549-0R cells, leading to a (224%, Figs. 1f). The speed of A549-0R cells was only mar-
hyperpolarized membrane potential in the former ones. ginally reduced from 0.47 6 0.03 to 0.42 6 0.03 mm/min with

Int. J. Cancer: 137, 1306–1317 (2015) V


C 2015 UICC
Bulk et al. 1313

Cancer Cell Biology

Figure 4. Functional expression of KCa3.1 channels in A549 cells. (a) Whole cell currents were elicited by voltage ramps from 2120 to 140 mV.
In control cells (si-ctrl, upper panels), the activation of KCa3.1 channels with 1-EBIO (200 mmol/l) is reversed by applying the KCa3.1 blocker
TRAM-34 (10 mmol/l). KCa3.1 channel activation is largely attenuated in cells transfected with siRNA against KCa3.1 channels (si-KCa3.1, lower
panel). (b) Summary of the experiments shown in (a) (n 5 15 for A549-0R and n 5 20 for A549-3R cells). (c) Membrane potential of A549 cells.
The resting cell membrane potential (Ctrl) of A549-3R cells is more hyperpolarized than that of A549-0R cells (n 5 15 for A549-0R and n 5 20 for
A549-3R cells). *p < 0.05. (d, e) Ca21 measurements with A549 cells. The pictures depict representative original recordings made with both A549
cell strains. DTRAM represents the change in the intracellular Ca21 concentration in response to the application of TRAM-34, DKCl corresponds to
the change elicited by the application of 50 mmol/l KCl. (f) Influence of TRAM-34 and 50 mmol/l KCl on the intracellular Ca21 concentration in
both A549 cell strains. The bars correspond to the mean values of DTRAM and DKCl from n 5 34 cells for A549-0R and n 5 40 for A549-3R cells.
[Color figure can be viewed in the online issue, which is available at wileyonlinelibrary.com.]

Int. J. Cancer: 137, 1306–1317 (2015) V


C 2015 UICC
1314 KCa3.1 channel and poor lung cancer prognosis
Cancer Cell Biology

Figure 5. Proliferation and migration of A549 cells are inhibited by KCa3.1 channel blockade. (a) A549 cells were treated with TRAM-34 (10,
20 and 30 mmol/l) for 24–72 hrs. *p < 0.05. (b) Cells were migrating on a collagen-based matrix. Ten minutes before the start of the experi-
ment, 100 ng/ml EGF and 10 mM TRAM-34 or DMSO were added. Trajectories of individual cells normalized to a common starting point.
The circle represents the mean distance covered during the course of the experiment. (c) Statistical evaluation of migration experiments for
distance of translocation. (d) Statistical evaluation of migration experiments for speed. *p<0.05. (e) Migration speed plotted as a function
of time for parental A549 cells. EGF (100 ng/ml) was added at t 5 0. Mean values of three independent experiments are shown. (f) Migra-
tion speed was plotted for the highly aggressive A549 cells.

TRAM-34 (211%, Figs. 5e). In both cell types, KCa3.1 block- KCa3.1 channel inhibition results in reduced proliferation
ade inhibited translocation more effectively than the speed of of A549 xenografts in vivo
migration. This is an indication for the involvement of Finally, we analyzed the impact of KCa3.1 channel inhibition
KCa3.1 channels in controlling the directionality of migrating on tumor growth in vivo. A549-3R tumor-bearing mice were
A549 cells. treated with the KCa3.1 channel blocker senicapoc

Int. J. Cancer: 137, 1306–1317 (2015) V


C 2015 UICC
Bulk et al. 1315

Cancer Cell Biology


Figure 6. KCa3.1 blocker senicapoc impairs proliferation of lung cancer xenografts. A549-3R tumor-bearing mice were treated with senicapoc
every day. (a) Growth of n 5 12 senicapoc-treated and n 5 14 DMSO-treated tumors was determined by caliper measurements. (b) 18F-FLT PET-
CT was performed before and 7 days after treatment initiation to assess proliferation of the tumors. CT allowed for delineation of the tumor
margins (marked with a white line) and determination of the tumor volume. Transverse slices of PET and CT images of representative mice are
depicted here. Because of nonuniform tumor growth, not all slices show three tumors. Scale bar 5 1 mm. (c) n 5 12 senicapoc-treated and
n 5 9 DMSO-treated tumors were analyzed quantitatively with respect to variations in 18F-FLT uptake and tumor size determined by CT.
*p<0.05. [Color figure can be viewed in the online issue, which is available at wileyonlinelibrary.com.]

(structurally similar to TRAM-34, but with a greatly primary NSCLC tumors from patients. DNA hypomethyla-
increased metabolic stability).20,21 Tumor growth was signifi- tion was associated with increased expression and function of
cantly inhibited after 4 days of treatment when compared the KCa3.1 channel as well as enhanced metastatic potential
with DMSO-treated mice (Fig. 6a relative change in size as and proliferation. Importantly, KCNN4 hypomethylation and
assessed by caliper (day 4/day 0): senicapoc: 0.95 6 0.04; increased expression of the KCa3.1 channel were strong and
DMSO: 1.21 6 0.07. p 5 0.005). Further, proliferation of the independent predictors for poor survival in NSCLC patients.
tumors was reduced as assessed by PET using 18F-FLT as in Functionally, DNA hypomethylation of the promoter and
vivo proliferation marker (Figs. 6bc relative change in 18F- increased expression of KCa3.1 channels are linked to
FLT uptake (day 7/day 0): senicapoc: 0.85 6 004; DMSO: increased calcium influx, representing a direct mechanistic
1.10 6 0.07; p 5 0.0041). Accumulation of this thymidine link between epigenetic alterations and calcium-dependent
analog has been shown to reflect the proliferation rate of the tumor aggressiveness.23 Thus, inhibition of KCa3.1 channels
respective tissue.22 CT provides more precise anatomical in vivo in an experimental mouse model resulted in reduced
information on tumor volume than caliper measurements, NSCLC proliferation, and hence decreased tumor volume as
and it confirmed the aforementioned results: senicapoc measured by 18F-FLT PET/CT.
strongly impaired A549 xenograft growth (relative change in Metastatic spread is a major reason for cancer-related death.
size as assessed by CT (day 7/day 0): senicapoc: 1.44 6 0.04; Despite intensive genome sequencing efforts, no metastasis-
DMSO: 1.96 6 0.17; p 5 0.0061). specific mutations have been discovered.11 Intriguingly, epige-
netic changes are likely to be major causal contributors for
Discussion metastatic capability. The resulting functional changes leading
The main findings of our study are as follows: Loss of DNA to a metastatic phenotype of cancer cells might be easier to tar-
methylation in the promoter region of the KCNN4 gene is a get than the mutations that provide the genetic landscape of
frequent feature of NSCLC. Promoter hypomethylation cancers. The molecular characterization of NSCLC has been
occurred in cells with increased aggressiveness as well as in advanced with improved patient stratification that takes into

Int. J. Cancer: 137, 1306–1317 (2015) V


C 2015 UICC
1316 KCa3.1 channel and poor lung cancer prognosis

account the presence or absence of driver mutations in impor- provided evidence that NSCLC proliferation can be inhibited
tant signaling pathways.24,25 Most of these pathways, however, with a KCa3.1 channel blocker, senicapoc, which had already
cannot be targeted easily by current therapies. been tested clinically.42
In contrast, ion channels and other ion transport proteins Our experiments indicated that KCa3.1 channels contrib-
are well-established and validated therapeutic targets in multiple ute to the aggressiveness of NSCLC cells by regulating prolif-
diseases (e.g., cardiovascular, renal and neuropsychiatric dis- eration and migration as in other tumor cells.43 Tumor
eases). Ion channels could also be very attractive therapeutic growth of aggressive NSCLC cells in mice could be inhibited
targets in oncology because they are usually blocked from their almost completely within the first week of treatment using
extracellular side so that drug resistance due to the expression the KCa3.1 blocker senicapoc. The impact of KCa3.1 channels
of multidrug resistance proteins appears unlikely.26 Indeed, on [Ca21]i, by setting the membrane potential, is a likely
recent data indicate that membrane proteins involved in ion explanation for the dual role in proliferation and migration.
transport also play crucial roles in tumor progression because It is noteworthy that KCa3.1 channels are required for migra-
they contribute essentially to all hallmarks of cancer.7,23,27–31 tion of EGF/fetal calf serum-stimulated cells but not for that
They are expressed aberrantly in almost all tumor types.32 Being of unstimulated cells. This raises the question whether other
central players in proliferation, apoptosis, angiogenesis, cell– ion channels upstream to KCa3.1 channels are required to
matrix interaction and migration or invasion, ion channels provide them with Ca21 necessary for their activation.
shape the aggressive phenotype of metastatic cancer cells.6,8 TRPC1 could be a potential candidate channel. In A549 cells,
Thus, ion channels are potentially new therapeutic, diagnostic TRPC1 channels are activated by EGF, and they are central
and/or prognostic targets in oncology. The Ca21-sensitive K1 constituents of the EGFR signaling cascade.44 Blocking
channel KCa3.1 is upregulated in many tumors, including pan- KCa3.1 channels impairs Ca21 influx and thereby EGF signal-
Cancer Cell Biology

creas, prostate and breast cancer33–35 as well as melanoma,36 ing. Because our experiments showed that KCa3.1 channel
where it plays important roles in proliferation and migration. blockade leads to decreased Ca21 influx into A549 cells,
However, so far, there is only very little information on the role impaired migration could be a consequence of a disturbed,
of K1 channels in NSCLC progression (e.g., Refs. [19 and 27). Ca21-dependent disassembly of focal adhesions.45–47 In such
KCa3.1 is one of the channels whose expression correlates a scenario, cells would tend to migrate “on the spot.”
with tumor grade and metastatic spread, cell cycle progres- Taken together, our experiments provide evidence for
sion and cell proliferation in several cancer types.34,35,37–39 increased KCa3.1 channel expression by epigenetic mecha-
Another important observation is that this channel is nisms in NSCLC. DNA hypomethylation as well as high-level
expressed in nearly all migrating cells.40 Its activity is mRNA expression are independently associated with poor
required for efficient cell migration.41 In view of the above, survival. We provide functional evidence that KCa3.1 chan-
the marked dependence of NSCLC patient outcome on the nels contribute to the aggressive behavior of NSCLC cells
expression of KCa3.1 channels led us to hypothesize that they and, thereby, to the poor prognosis of NSCLC patients with
contribute to disease progression and to the metastatic behav- high channel expression.
ior of NSCLC cells. If this were the case, its elevated expres-
sion in NSCLC would not only be a predictor of poor Acknowledgements
outcome. Then, abundantly expressed KCa3.1 channels could The authors are grateful for the excellent technical assistance of Sarah Sargin,
also constitute a therapeutic target with clinical potential. Stefanie Bouma and Christine B€atza. TRAM-34 and Senicapoc were kind
gifts from Heike Wulff, Davis, CA, USA.
This idea is also supported by our in vivo experiments that

References
1. Jemal A, Bray F, Center MM, et al. Global cancer antineoplastic therapy. Curr Med Chem 2009;16: 12. Hascher A, Haase AK, Hebestreit K, et al. DNA
statistics. CA Cancer J Clin 2011;61:69–90. 66–93. methyltransferase inhibition reverses epigeneti-
2. Jemal A, Siegel R, Xu J, et al. Cancer statistics, 7. Pedersen SF, Stock C. Ion channels and trans- cally embedded phenotypes in lung cancer prefer-
2010. CA Cancer J Clin 2010;60:277–300. porters in cancer: pathophysiology, regulation, entially affecting polycomb target genes. Clin
3. Goldstraw P, Crowley J, Chansky K, et al. The and clinical potential. Cancer Res 2013;73: Cancer Res 2014;20:814–26.
IASLC lung cancer staging project: proposals for 1658–61. 13. Okayama H, Kohno T, Ishii Y, et al. Identifica-
the revision of the TNM stage groupings in the 8. Prevarskaya N, Skryma R, Shuba Y. Ion channels tion of genes upregulated in ALK-positive and
forthcoming (seventh) edition of the TNM classi- and the hallmarks of cancer. Trends Mol Med EGFR/KRAS/ALK-negative lung adenocarcino-
fication of malignant tumours. J Thorac Oncol 2010;16:107–21. mas. Cancer Res 2012;72:100–11.
2007;2:706–14. 9. Baylin SB, Jones PA. A decade of exploring the 14. Bulk E, Hascher A, Liersch R, et al. Adjuvant
4. Ballas MS, Chachoua A. Rationale for targeting cancer epigenome—biological and translational therapy with small hairpin RNA interference pre-
VEGF, FGF, and PDGF for the treatment of implications. Nat Rev Cancer 2011;11:726–34. vents non-small cell lung cancer metastasis devel-
NSCLC. Onco Targets Ther 2011;4:43–58. 10. Brevet M, Fucks D, Chatelain D, et al. Deregula- opment in mice. Cancer Res 2008;68:1896–904.
5. Lawrence MS, Stojanov P, Polak P, et al. Muta- tion of 2 potassium channels in pancreas adeno- 15. Schwab A, Wulf A, Schulz C, et al. Subcellular
tional heterogeneity in cancer and the search for carcinomas: implication of KV1.3 gene promoter distribution of calcium-sensitive potassium chan-
new cancer-associated genes. Nature 2013;499: methylation. Pancreas 2009;38:649–54. nels (IK1) in migrating cells. J Cell Physiol 2006;
214–8. 11. Vogelstein B, Papadopoulos N, Velculescu VE, 206:86–94.
6. Arcangeli A, Crociani O, Lastraioli E, et al. Tar- et al. Cancer genome landscapes. Science 2013; 16. Viel T, Schelhaas S, Wagner S, et al. Early assess-
geting ion channels in cancer: a novel frontier in 339:1546–58. ment of the efficacy of temozolomide chemother-

Int. J. Cancer: 137, 1306–1317 (2015) V


C 2015 UICC
Bulk et al. 1317

apy in experimental glioblastoma using decreases cell proliferation of human small cell cancer cell proliferation through a close control
[18F]FLT-PET imaging. PLoS One 2013;8:e67911. lung cancer cells. Pflugers Arch 2011;463:365–76. of calcium entry. Oncogene 2009;28:1792–806.
17. Ball MP, Li JB, Gao Y, et al. Targeted and 29. Hanahan D, Weinberg RA. Hallmarks of cancer: 39. Wang ZH, Shen B, Yao HL, et al. Blockage of
genome-scale strategies reveal gene-body methyl- the next generation. Cell 2011;144:646–74. intermediate-conductance-Ca21 -activated K1
ation signatures in human cells. Nat Biotechnol 30. Jang SH, Ryu PD, Lee SY. Dendrotoxin-kappa channels inhibits progression of human endome-
2009;27:361–8. suppresses tumor growth induced by human lung trial cancer. Oncogene 2007;26:5107–14.
18. Gao YD, Hanley PJ, Rinne S, et al. Calcium-acti- adenocarcinoma A549 cells in nude mice. J Vet 40. Schwab A, Fabian A, Hanley PJ, et al. Role of ion
vated K1 channel (KCa3.1) activity during Ca21 Sci 2011;12:35–40. channels and transporters in cell migration. Phys-
store depletion and store-operated Ca21 entry in 31. Roger S, Rollin J, Barascu A, et al. Voltage-gated iol Rev 2012;92:1865–913.
human macrophages. Cell Calcium 2010;48:19–27. sodium channels potentiate the invasive capacities 41. Schwab A, Wojnowski L, Gabriel K, et al. Oscil-
19. Roth B, Gibhardt CS, Becker P, et al. Low-dose of human non-small-cell lung cancer cell lines. lating activity of a Ca21-sensitive K1 channel. A
photon irradiation alters cell differentiation via Int J Biochem Cell Biol 2007;39:774–86. prerequisite for migration of transformed Madin-
activation of hIK channels. Pflugers Arch 2014. 32. Diss JK, Stewart D, Pani F, et al. A potential Darby canine kidney focus cells. J Clin Invest
DOI: 10.1007/s00424-014-1601-4. novel marker for human prostate cancer: voltage- 1994;93:1631–6.
20. Stocker JW, De Franceschi L, McNaughton-Smith gated sodium channel expression in vivo. Prostate 42. Ataga KI, Reid M, Ballas SK, et al. Improvements
GA, et al. ICA-17043, a novel Gardos channel Cancer Prostatic Dis 2005;8:266–73. in haemolysis and indicators of erythrocyte sur-
blocker, prevents sickled red blood cell dehydra- 33. Jager H, Dreker T, Buck A, et al. Blockage of vival do not correlate with acute vaso-occlusive
tion in vitro and in vivo in SAD mice. Blood intermediate-conductance Ca21-activated K1 crises in patients with sickle cell disease: a phase
2003;101:2412–8. channels inhibit human pancreatic cancer cell III randomized, placebo-controlled, double-blind
21. Wulff H, Castle NA. Therapeutic potential of growth in vitro. Mol Pharmacol 2004;65:630–8. study of the Gardos channel blocker senicapoc
KCa3.1 blockers: recent advances and promising 34. Ohya S, Kimura K, Niwa S, et al. Malignancy (ICA-17043). Br J Haematol 2011;153:92–104.
trends. Expert Rev Clin Pharmacol 2010; grade-dependent expression of K1-channel sub- 43. Ruggieri P, Mangino G, Fioretti B, et al. The
3:385–96. types in human prostate cancer. J Pharmacol Sci inhibition of KCa3.1 channels activity reduces cell
22. Shields AF, Grierson JR, Dohmen BM, et al. Imag- 2009;109:148–51. motility in glioblastoma derived cancer stem cells.
ing proliferation in vivo with [F-18]FLT and posi- 35. Haren N, Khorsi H, Faouzi M, et al. Intermediate PLoS One 2012;7:e47825.
tron emission tomography. Nat Med 1998;4:1334–6. conductance Ca21 activated K1 channels are 44. Tajeddine N, Gailly P. TRPC1 protein channel is
23. Prevarskaya N, Skryma R, Shuba Y. Calcium in expressed and functional in breast adenocarcino- major regulator of epidermal growth factor

Cancer Cell Biology


tumour metastasis: new roles for known actors. mas: correlation with tumour grade and metasta- receptor signaling. J Biol Chem 2012;287:
Nat Rev Cancer 2011;11:609–18. sis status. Histol Histopathol 2010;25:1247–55. 16146–57.
24. Petrosyan F, Daw H, Haddad A, et al. Gene 36. Tajima N, Schonherr K, Niedling S, et al. Ca21- 45. Giannone G, Ronde P, Gaire M, et al. Calcium
expression profiling for early-stage NSCLC. Am J activated K1 channels in human melanoma cells oscillations trigger focal adhesion disassembly in
Clin Oncol 2015;38:103–7. are up-regulated by hypoxia involving hypoxia- human U87 astrocytoma cells. J Biol Chem 2002;
25. Pao W, Iafrate AJ, Su Z. Genetically informed inducible factor-1alpha and the von Hippel- 277:26364–71.
lung cancer medicine. J Pathol 2011;223:230–40. Lindau protein. J Physiol 2006;571:349–59. 46. Schafer C, Rymarczyk G, Ding L, et al. Role of
26. Wulff H, Castle NA, Pardo LA. Voltage-gated 37. Ouadid-Ahidouch H, Roudbaraki M, Delcourt P, molecular determinants of store-operated Ca21
potassium channels as therapeutic targets. Nat et al. Functional and molecular identification of entry (Orai1, phospholipase A2 group 6, and
Rev Drug Discov 2009;8:982–1001. intermediate-conductance Ca21-activated K1 STIM1) in focal adhesion formation and
27. Jang SH, Choi SY, Ryu PD, et al. Anti-prolifera- channels in breast cancer cells: association with cell migration. J Biol Chem 2012;287:
tive effect of Kv1.3 blockers in A549 human lung cell cycle progression. Am J Physiol Cell Physiol 40745–57.
adenocarcinoma in vitro and in vivo. Eur J Phar- 2004;287:C125–34. 47. Svensson L, McDowall A, Giles KM, et al. Cal-
macol 2011;651:26–32. 38. Lallet-Daher H, Roudbaraki M, Bavencoffe A, pain 2 controls turnover of LFA-1 adhesions on
28. Glassmeier G, Hempel K, Wulfsen I, et al. Inhibi- et al. Intermediate-conductance Ca21-activated migrating T lymphocytes. PLoS One 2010;5:
tion of HERG1 K1 channel protein expression K1 channels (IKCa1) regulate human prostate e15090.

Int. J. Cancer: 137, 1306–1317 (2015) V


C 2015 UICC

You might also like