Sie sind auf Seite 1von 9

Cancer Letters 233 (2006) 1–9

www.elsevier.com/locate/canlet

Mini-review

Ether à go-go potassium channels and cancer


Javier Camacho*
Pharmacology Section, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional,
Avenida Instituto Politécnico Nacional 2508, México, DF 07360, México

Received 10 February 2005; accepted 14 February 2005

Abstract
Ion channels play important roles in health and disease. In the last few years, an interesting relationship between potassium
channels and cancer has evolved. Especially, members of the ether à go-go (EAG) potassium channels family have gained
interest as research tools for detection and therapy of different cancers. This review will summarize most of the findings relating
EAG channels and cancer, focusing on mRNA expression in tissues, oncogenic properties, modulation and pharmacology.
Plausible scenarios on the cellular mechanisms of EAG oncogenicity will be discussed.
q 2005 Elsevier Ireland Ltd. All rights reserved.

Keywords: Cancer; Tumor markers; Cervical cancer; Ion channels; Ether à go-go; Herg; Proliferation; Epsin; Human papilloma virus;
Tamoxifen; Patch-clamp; Potassium channels; Hypoxia; Von hippel-lindau protein

1. Introduction signals vary from voltage and neurotransmitters to pH


and temperature and are usually specific for a type of
Ion channels are membrane proteins governing channel [3]. Several ion channels have been related to
massive ion fluxes through the cells and participating cell proliferation and/or cancer including calcium,
in many and diverse physiological events such as chloride and sodium channels [4–6]; however, the ion
excitability, contraction, cell cycle progression and channels most commonly related to cell proliferation
metabolism [1] both in health and disease. Several and cancer are the potassium channels; actually some
channelopathies have been described and many of excellent general reviews on potassium channels and
these proteins are therapeutic targets, highlighting the cell proliferation have been recently published [7–9].
physiological relevance of ion channels [2]. In This review focus on the family of voltage-gated
addition to leak channels, which are opened at resting potassium channels ether à go-go (EAG) and its
membrane potential, there is a quite diverse group of interesting relationship to cancer. Because of their
channels needing a special signal to be gated. Gating oncogenic properties, distribution, modulation and
pharmacology, EAG channels have gained interest as
* Tel.: C52 55 50613800x5416; fax: C52 55 53430106.
potential diagnostic markers and membrane thera-
E-mail address: fcamacho@cinvestav.mx. peutic targets for cancer.

0304-3835/$ - see front matter q 2005 Elsevier Ireland Ltd. All rights reserved.
doi:10.1016/j.canlet.2005.02.016
2 J. Camacho / Cancer Letters 233 (2006) 1–9

2. Ether à go-go channels domain in the former and a cyclic nucleotide binding
domain in the latter [reviewed in Ref. [15]]. PAS
In 1969, several mutants from the fruit fly domain has been suggested to be an oxygen sensor
Drosophila melanogaster were produced following [19], actually, EAG channels are modulated by
the treatment of adult males by ethyl methane hypoxia or hypoxia-related proteins, as will be
sulfonate [10]. The mutants presented shaking of the discussed later.
legs under ether anesthesia and independent gene loci Particular voltage-dependent properties of eag and
were found to be involved. One of these mutants erg channels as well as pharmacological tools
exhibited a slow and rhythmic leg-shaking behaviour; [reviewed in Ref. [15]] allow to identify endogenous
the locus involved was then named ether à go-go EAG mediated currents. As expected for voltage-
(eag). gated channels, different roles have been described to
Further evidences including the observation that members of the EAG family in excitable cells. Erg
eag mutants presented a high frequency of spon- channels have a major role in the repolarization phase
taneous action potentials and enhanced transmitter of the cardiac action potential; mutations in this gene
release in motor neurons, and the comparison of eag are responsible for the long Q-T syndrome 2 (LQT2)
sequence with other genes [11,12], suggested that eag in humans and the channels are targeted by several
might encode for a protein involved in cellular anti-arrhythmic drugs [reviewed in Ref. [20]]. In
excitability, namely, a potassium channel. It was in addition, during a train of action potentials, a role in
1993 that expression studies in oocytes demonstrated spike-frequency adaptation has been suggested for erg
that Drosophila eag encodes a voltage-gated potass- [21]. In the case of the eag1, known physiological
ium channel [13]. Homology screening helped to roles in normal cells are limited to human myoblasts
identify other two sequences closely related to eag: differentiation [22] and will be described below.
the eag-related gene (erg) and the eag-like gene (elk); Specific roles for eag1 have not been described in
thus the EAG channels family comprises three nervous system, despite its major expression in brain.
subfamilies: EAG, ERG and ELK [14]. Two members Interestingly, different experimental approaches
for the eag subfamily, three for erg and two for elk are have related eag1 and erg channels to cell cycle and
differentially expressed in different species including cellular transformation, suggesting them as potential
rat, bovine and humans [reviewed in Ref. [15]]. In the diagnose markers and therapeutic targets for cancer.
present review, ‘EAG’ will refer to members belong-
ing to the EAG channels family; and only those
channels involved in cell proliferation and/or cancer 3. EAG channels, cell-cycle and cancer
will be discussed, it is ‘eag1’ (from the eag subfamily)
and ‘erg’ channels. 3.1. Cell-cycle and EAG
Distribution of EAG in normal tissues depends on
the subfamily. Erg channels are ubiquitous; tran- Indicative data that EAG might be related to or
scripts have been found in heart, brain, kidney, liver, modulated during cell-cycle appeared when electro-
testis, uterus and prostate, among other sites physiological studies showed that currents with
[reviewed in Ref. [16]]. Eag1 has a more restricted biophysical and pharmacological properties resem-
distribution, it has been found almost exclusively in bling those of erg channels were present in neuro-
brain, slightly in placenta and transiently in myoblasts blastoma cell lines; the voltage-dependent properties
[17,18]; in fact, this restricted distribution in normal of such currents varied widely in unsynchronized cells
tissues is one of the most attractive features of eag1 as but the variability was greatly reduced when the cells
a potential tumor marker. were synchronized in the G1 phase of the cell cycle
EAG channels form tetramers, each monomeric [23].
subunit has six putative transmembrane domains Later on, Walter Stühmer’s group described for the
including the voltage sensor (a segment enriched in first time that a channel with a defined molecular
positively charged amino-acids) and its intracellular entity, namely eag1, was ‘cell cycle-sensitive’. Frog
C- and N-terminus with a Per-Arnt-Sim (PAS) oocytes arrested in G2 phase are commonly used for
J. Camacho / Cancer Letters 233 (2006) 1–9 3

cell-cycle studies and expression of exogenous [29], and suggested h-erg channel activity as a
proteins; addition of progesterone to the oocytes selective advantage for cancer cells. Ischemic con-
induces cell-cycle progression reentering the cells into ditions lead to failure of the NaC/KC pump inducing
the meiotic phase [24,25]. Stühmer’s group expressed cellular potassium loss, which is a prerequisite for
rat eag1 channels in the oocytes and found that apoptosis; the authors suggested that h-erg channels
channel activity was down-regulated in the presence might provide the hyperpolarization needed to avoid
of progesterone; because this effect was also observed such potassium loss preventing the cells from entering
when injecting the Mitosis Promoting Factor even in the apoptotic pathway.
the absence of progesterone, the conclusion was that The second report is a landmark in ion channel
eag1 was modulated in a cell-cycle dependent manner research and was communicated by Stühmer’s group.
[26]. Cloning of human eag1 was soon reported and a They demonstrated that eag1 posses oncogenic
physiological role in a cell-cycle-related event properties. Cell lines transfected with eag1 acquire
described [17]. In human myoblasts, cell-cycle is properties of cancerous cells, namely, they lose cell-
arrested by cell fusion and membrane hyperpolariz- contact inhibition, are able to grow in very low serum-
ation is needed. H-eag1 is transiently expressed in concentrations and, noteworthy, induce formation of
myoblasts slightly before fusion providing an early aggressive tumors when injected into immune-
hyperpolarization, then eag expression decreases and depressed mice [18]. Moreover, in contrast to its
it is not expressed in the multinucleate skeletal muscle restricted distribution in normal tissues (mainly in
cell [22]. Properties of eag1 channels during cell- brain, slightly in placenta and transiently in myo-
cycle were then studied in detail; during M-phase, rat blasts) they found that eag1 was expressed in several
and h-eag1 channels expressed in a cell line become
human somatic cancer cell lines including HeLa,
more selective to potassium ions, are sensitive to
MCF-7 and SHSY-5Y, from carcinoma of the cervix,
blockage by intracellular sodium and display a
breast tumor and neuroblastoma, respectively. The
smaller current density in comparison to G1, S or
immediately arising question was whether eag1
G2 phases [27,28]; this cell-cycle modulation was
expression in tumor cells was a consequence of the
suggested to be determined by cytoskeletal elements
abnormal growth or if the channels were necessary for
proposing that eag1 channels might be modulated in
cell proliferation. Antisense experiments against
other cellular events in which cytoskeletal rearrange-
ments occur, as in migration of cancer cells. It was h-eag1 decreased significantly DNA synthesis in the
also observed that erg is rapidly turned on in normal tumor cell lines mentioned above, suggesting that
CD34C cells from peripheral blood upon induction of eag1 channels are important in the proliferation of the
the mitotic cycle by cytokines/growth factors [33]. tumor cells lines. In a following communication, the
Different tumor cell lines and primary human same group reported that inhibition of eag1 (either
cancer cells have been used for expression, modu- expression or channel function) by small molecule,
lation, and pharmacological EAG studies creating a blocking antibody or short interfering RNA led to a
new atmosphere in the study of cancer and ion reduction in DNA synthesis and proliferation in
channels. Following, major findings on EAG and several human tumor cell lines [30]. Therefore,
cancer are described and summarized in Table 1. h-eag1 is considered as a potential tumor marker
and therapeutic target for cancer.
3.2. EAG and cancer Further investigations have shown EAG expression
and/or channel activity in additional tumor cell lines
There are two initial papers representing the including melanoma, neuroblastoma and rhabdomyo-
milestones of the link cancer-EAG. The first report sarcoma for eag1 [31,32,42], and hematopoietic
was communicated by Enzo Wanke’s group. They (Burkitt’s lymphoma, chronic myelogenous leukemia,
showed that human erg transcripts were expressed in acute lymphoblastic leukemia and acute promyelocy-
tumor cell lines from different histogenesis including tic leukemia), uterine cancer, retinoblastoma, mam-
rhabdomyosarcoma, neuroblastoma, mammary gland mary adenocarcinoma and colon cancer cells for erg
adenocarcinoma and monoblastic leukemia cell lines [33–36,42].
4 J. Camacho / Cancer Letters 233 (2006) 1–9

Table 1
Major findings relating cancer to human EAG channels

Channel Finding Refs.


Eag1 Displays oncogenic potential. Induces aggressive tumor formation in SCID mice. mRNA expressed in [17,18,30]
several somatic tumor cell lines including HeLa, MCF-7 and SHSY-5Y, but in normal tissues only in
brain, placenta and transiently in myoblasts. Inhibition of expression or channel activity by different
experimental approaches decreased cell proliferation in tumor cell lines
Transcript present in melanoma and neuroblastoma cells and channel activity inhibited by imipramine [31,32,52]
mRNA expressed in gliomas depending on the malignancy of the tumor [39]
mRNA expression in 100% of cervical cancer. Channel activity found in cancer cells. Expression in some [40]
normal cervical samples (from patients with negative pap smears) suggested as potential early indicator
of tumor development. Probably associated to HPV infection
Inhibited by the hypoxia-related and tumor suppressor gene von Hippel-Lindau in neuroblastoma cells [45]
Erg mRNA expression and channel activity in several tumor cell lines from different histogenesis including [29,33–35,42]
neuroblastoma, rhabdomyosarcoma, hemopoietic, retinoblastoma, mammary adenocarcinoma, uterine
and colon cancer cells. Suggested as an advantage for tumor cells by preventing apoptosis
More frequently expressed in human endometrial cancer as compared to non-cancerous endometrium [33,36]
and not found in endometrial hyperplasia. Constitutively activated in neoplastic hematopoietic cells
while turned-off in normal cells
mRNA expression in gliomas and channel activity present in lactotrophs from prolactinoma tissue [38,39]
mRNA expression and channel activity in primary colon cancer with a higher incidence in metastatic [36]
cancers. No expression found in normal colonic mucosa or adenomas. Suggested as a potential
prognostic factor
Modulated by hypoxic conditions and Src tyrosine kinase which is commonly overexpressed in tumors. [43,47,49]
Regulates both apoptosis and proliferation
Inhibited by chemotherapeutic drugs (tamoxifen and adrenergic antagonists) [50,51]

EAG expression and activity have been demon- expression. Erg transcripts and protein were found to
strated not only in tumor cell lines but also in several be expressed in a high percentage of primary human
primary human cancers; comparative studies in the colorectal cancers, with the highest incidence occur-
corresponding normal tissues have stressed the ring in metastatic cancers [36]. However, when
potential clinical relevance of EAG. looking for erg in normal colonic mucosa or in
Erg channels were found to be more frequently adenomas, no expression was found. Erg channels
expressed in human endometrial cancer as compared were then suggested as potential prognostic factors for
to non-cancerous endometrium, while it was not colorectal cancer. Investigations in human gliomas
found in endometrial hyperplasia [37]; the authors have also been made [39] that suggest a differential
suggested h-erg channels as potential tools discrimi- expression of erg and eag1 depending on the
nating endometrial cancer from simple hyperplasia. malignancy grade and nature of tumor cells; such
Studies of erg in leukemias and normal blood cells by expression seems to be inversely related to the
two different groups, led to the conclusion that erg is malignancy of the tumor. Because EAG channels
constitutively activated in the neoplastic cells while are expressed in normal brain, the precise relationship
turned-off in normal cells [33,34], suggesting it as a between EAG expression and brain tumors remains to
novel molecular marker for human neoplastic hema- be elucidated.
topoietic cells. Human lactotroph cells from prolacti- Finally, a recent report relates eag1 expression and
noma tissue have also been reported to express erg channel activity to cervical cancer [40], the leading
channels, its activity was suggested to participate in cause of women death in some developing countries.
the control of prolactin secretion, in accordance to its The group reported eag1 mRNA expression in 100%
role in normal rat lactotrophs [38]. of the cervical cancer samples studied and only in
Colorectal cancer, one of the leading causes of 33% of the normal control samples. Interestingly, they
cancer death, has also been subject of study for erg observed that in one of the patients submitted to
J. Camacho / Cancer Letters 233 (2006) 1–9 5

hysterectomy without any previous evidence of MCF-7 cells, from human breast cancer, eag1 mRNA
cervical malignancy (negative pap smears), post- expression is significantly increased in cells in the G1
surgery pathological studies showed an unexpected phase of the cell cycle but then lowered in the S phase
endocervical adenocarcinoma expressing eag1. This [41]. Very fine regulation of EAG channels during cell
case, although unique in their study, emphasizes the cycle was reported in tumor cell lines from different
potential significance of eag1 as a tumor marker. In histogenesis [42]; erg isoforms are differentially
addition, despite all of their control cervical samples regulated during cell cycle; one of these isoforms
came from patients with negative pap smears, they lacks the PAS domain, an oxygen sensor. Actually,
also found eag1 expression in some of such biopsies. erg channels are modulated by hypoxia, a key element
Quite interesting, diagnosis of one of those samples in tumor progression. Neuroblastoma cells exposed to
was correlated to human papilloma virus infection, the hypoxia undergo profound changes in its gating
most important etiological factor for cervical cancer; properties [43].
other control sample presented atypical adenomatous Eag1 has also been somehow related to hypoxia.
hyperplasia of the endometrium, a predisposing The von Hippel-Lindau (VHL) tumor suppressor
condition of endometrial cancer and in other control gene, linked to the development of sporadic heman-
eag-positive case, hystopathological studies reported gioblastomas and clear-cell renal carcinomas, plays a
a paratubaric serous cystadenoma, a benign epithelial central role in the oxygen sensing pathway because of
ovarian tumor. A plausible scenario is that the its close interaction with the hypoxia-inducible factor
increase of eag1 expression in normal cervix could [HIF, reviewed in Ref. [44]]. In the absence of the
be an early sign of tumor development. In addition, VHL protein, HIF becomes stabilized and is free to
they found channel expression and activity in cervical induce the expression of cancer related genes such as
cancer cells. The authors suggest that eag1 might be those regulating angiogenesis, cell growth or cell
not only a molecular marker and a potential survival. Expression of the VHL protein in human
membrane therapeutic target for cervical cancer but neuroblastoma cells switched-off drastically eag1
probably also an early indicator of the disease. mediated currents; accordingly, VHL inhibition
In addition to the expression and channel activity increased eag1 channel activity in comparison to
studies, modulation and pharmacological experiments control cells [45].
have provided important clues for the potential Other modulation studies have also provided very
therapeutic use of EAG in cancer treatment and are interesting information. Intracellular calcium at nano-
described below. molar concentrations inhibits eag1 currents through
calmodulin binding to the eag1 C-terminus [46], the
3.3. EAG modulation and cancer binding is calcium dependent with an apparent KD of
480 nM. Because of calcium has a regulatory role
Cell-cycle regulation of proteins, signal transduc- during cell cycle, the authors proposed to elucidate
tion pathways, extracellular matrix components, whether this calcium regulation of eag1 is an
tumor suppressor genes and hypoxia play important important link between these channels and tumor
roles in tumor development and have been key progression. Src tyrosine kinases are overexpressed in
elements when designing anticancer therapies. Modu- a variety of human tumors and are an integral part of
lation of EAG channels by such elements has been tumor signaling pathways associated with migration,
studied in different cancer cells. proliferation, adhesion and angiogenesis; co-immu-
Induction of human neuroblastoma cells differen- noprecipitation studies suggest that rat erg channels
tiation by retinoic acid reduces eag1 current in more appears to exist in a signaling complex with Src
than 95% and also changes the voltage-dependent tyrosine kinase. Erg channel activity was reduced by
properties of erg channels [32]; in other differen- the Src-selective inhibitory peptide and the current
tiation study, it was shown that in a human leukemic was increased by the Src-activating peptide [47].
cell line undergoing differentiation after adhesion to Studies of protein associations revealed that epsin
fibronectin, erg current was early activated in binds to eag1 channels modulating its gating proper-
adhering cells [48]. In comparison to unsynchronized ties [54]. Epsins are proteins that function at
6 J. Camacho / Cancer Letters 233 (2006) 1–9

the plasma membrane in the internalization steps of efflux through potassium channels induces hyperpol-
endocytosis [reviewed in Ref. [55]], thus epsin arization and changes in cell volume. Hyperpolariz-
binding to eag1 represents a potential link between ation increases the driving force for calcium ions—
cell growth and eag1 channels that deserves further which might interact with cell cycle proteins—and on
investigations. Finally, it has been shown that human the other hand, cell size has been closely related to
erg channels might be regulators of both apoptosis cell proliferation; however, it has been confirmed in
and proliferation. On one hand, erg channel activity numerous studies that cancer cells are more depolar-
markedly promotes H2O2-induced apoptosis of var- ized than non-tumor cells. Clearly, additional clues
ious tumor cells, blockade of erg inhibited such must be found and alternative mechanisms uncovered.
induced apoptosis. On the other hand, erg expression Probably, particular characteristics like special struc-
facilitated the tumor cell proliferation caused by tural features, protein–protein interactions and modu-
tumor necrosis factor [49]. lation of potassium channels by cell cycle—or cancer-
related factors might provide such clues; EAG
3.4. EAG pharmacology and cancer drugs channels possess some of these particular properties,
hence more hypothetical scenarios might be suggested
Specific blockers for erg channels have been in addition to the described above (Fig. 1).
extensively used, mainly because of their pivotal EAG interacts with and is modulated by proteins
role in the LQT2 syndrome [reviewed in Refs. related to cell cycle and/or cancer like epsin, Src
[15,20]] but the specific block has been also shown tyrosine kinase, calmodulin and integrins. In addition,
to inhibit cell proliferation in tumor cell lines from modulation of EAG also includes important elements
different histogenesis [34]. Interestingly, chemother- involved in tumor progression like hypoxia, tumor
apeutic drugs like tamoxifen (widely used in breast suppressor genes (also related to hypoxic conditions),
cancer treatment) and a1-adrenoceptors antagonists cytoskeletal elements and differentiation-inducing
(used in the treatment of benign prostatic hyperplasia) factors. An intriguing question remains: what happens
also inhibit erg channel activity [50,51]. Unfortu- in the opposite direction? Inhibition of EAG (either
nately, there are no specific blockers for eag1, expression or channel activity) decreases cell pro-
however, as in the case for erg, some non-specific liferation. How does changes in channel activity
blockers have been shown to inhibit eag1 and modify the interaction of EAG with the cell cycle—or
decrease cell proliferation in tumor cell lines. cancer-related proteins? Is EAG channel activity
Imipramine, a tryciclic antidepressant and the anti- necessary to maintain such interactions? Does EAG
histamine astemizole inhibit eag1 [52,53]; imi- channel activity modify the intrinsic activity of its
pramine impairs cell proliferation in human binding proteins, controlling indirectly tumor pro-
melanoma cell lines expressing eag1 [52]. gression? How is such interaction modulated for
Non-specific EAG channel inhibition by che- example, by hypoxic conditions? Ubiquitous EAG
motherapeutic drugs offers alternative mechanisms channels expression in tumors might be compatible
explaining the anti-proliferative effect of some with the idea of considering EAG as modulators of
chemotherapeutic compounds and provide insights cell cycle—and cancer-related proteins. Actually it
for the design of new anti-cancer drugs. has been recently demonstrated that human erg
channels and integrins co-precipitate in tumor cells
and it was suggested that the protein and its channel
4. Hypothetical scenarios on oncogenicity activity can modulate integrin downstream signaling
mechanisms [57]. Another unsolved question is how EAG channels
expression is induced. Is HPV infection related to
Potassium channels activity has been related to cell EAG expression? or, do other viruses, oncogenes or
cycle progression, oncogenesis and proliferation, and hormones induce EAG expression? Addressing some
although the precise mechanism involved has not of these issues will help for a better understanding of
been elucidated, several hypothesis have been the relationship between EAG and cancer. We are
suggested [reviewed in Refs. [7–9,56]]. Potassium already trying to solve some of these questions.
J. Camacho / Cancer Letters 233 (2006) 1–9 7

Mitogenic signals Oncogenes Anti-apoptotic


(hormones, growth factors) signals

Chemical E AG exp re ss ion?


Human papilloma or
carcinogens other viruses

H y p o x i a , d if f e re n t ia ti o n
an d drug s
eag1 erg
Apoptosis
Membrane potential
? changes. K+ efflux ?
?
?
Ca2+ ? Cell volume
influx changes
Epsin ? ? Src tyrosine
Calmodulin kinase
Cytoskeleton Integrins
VHL protein Tumor Necrosis
(HIF-related) Proliferation Factor
Metastasis
Apoptosis
prevention

Fig. 1. Hypothetical scenarios on the mechanisms of EAG oncogenicity: EAG channels as modulators of cell cycle and cancer-related proteins?
It has been suggested that potassium channels activity would relate to cell proliferation by increasing calcium influx and modifying cell volume.
However, since EAG channels are modulated by several cell cycle and cancer-related proteins as well as hypoxic conditions and inhibited by
some anti-cancer drugs, alternative explanations would arise if at least some of such elements are in turn modulated by the EAG protein and/or
channel activity. In such hypothetical model, EAG channels would be indirect but more general inducers of proliferation.

5. Conclusions tissues—among many other studies needed—will


help to elucidate how EAG channels become turned-
Expression and channel activity studies have on and their precise participation in regulating or
highlighted the tight association between EAG and promoting proliferation in health and disease.
cancer; furthermore, modulation and pharmacological
experiments have provided important clues for the
potential therapeutic use of EAG in cancer treatment.
Because of its restricted distribution in normal Acknowledgements
tissues and its more ubiquitous distribution in cancer
The author is deeply grateful to Walter Stühmer
cells, EAG channels represent promising tools for the
and Luis Pardo (MPI, Göttingen Germany) for
development of improved diagnose cancer methods.
supporting significantly his scientific career. The
In addition, since channel inhibition impairs cell
author’s laboratory has received financial support by
proliferation in tumor cell lines, EAG channels also
the Consejo Nacional de Ciencia y Tecnologı́a.
represent interesting targets for the directed design of
anti-cancer therapies.
The study of the relationship between molecularly
identified EAG channels and cancer is a relatively References
young field of research and many questions have to be
solved. Studies of the time-course expression of EAG [1] B. Hille, Ion Channels of Excitable Membranes, third ed.,
in carcinogenesis models, association between EAG Sinauer Associates Inc., Sunderland, MA, USA, 2001.
[2] B.A. Niemeyer, L. Mery, C. Zawar, A. Suckow, F. Monje,
expression and hormone consumption or virus infec- L.A. Pardo, et al., Ion channels in health and disease, Eur. Mol.
tion like HPV, as well as the role of EAG in normal Biol. Org. Rep. 2 (2001).
8 J. Camacho / Cancer Letters 233 (2006) 1–9

[3] J. Koester, S.A. Siegelbaum, in: E.R. Kandel, J.E. Schwartz, [21] N. Chiesa, B. Rosati, A. Arcangeli, M. Olivotto, E. Wanke, A
T. Jessel (Eds.), Membrane Potential in Principles of novel role for HERG KC channels: spike-frequency adap-
Neuroscience fourth ed., Mc Graw Hill, New York, 2000. tation, J. Physiol. 501 (1997) 313–318.
[4] I. Latour, D.F. Louw, A.M. Beedle, J. Hamid, G.R. Sutherland, [22] P. Bijlenga, T. Occhiodoro, J.H. Liu, C.A. Bader,
G.W. Zamponi, Expression of T-type calcium channels splice L. Bernheim, J. Fischer-Lougheed, An ether à go-go KC
variants in human glioma, Glia 48 (2004) 112–119. current, Ih-eag, contributes to the hyperpolarization of human
[5] M. Abdel-Ghany, H. Cheng, R.C. Elble, H. Lin, J. Dibiasio, fusion-competent myoblasts, J. Physiol. 512 (1998) 317–323.
B.U. Pauli, The interaction binding domains of the b4 integrin [23] A. Arcangeli, L. Bianchi, A. Becchcetti, L. Faravelli,
and calcium-activated chloride channels (CLCAs) in metas- M. Coronnello, E. Mini, et al., A novel inward-rectifying
tasis, J. Biol. Chem. 278 (2003) 49406–49416. KC current with a cell-cycle dependence governs the resting
[6] E.S. Bennet, B.A. Smith, J.M. Harper, Voltage-gated NaC potential of neuroblastoma cells, J. Physiol. 489 (1995)
channels confer invasive properties on human prostate cancer 455–471.
cells, Pflügers Arch. 447 (2004) 908–914. [24] W. Stühmer, Electrophysiological recordings from Xenopus
[7] L.A. Pardo, Voltage-gated potassium channels in cell oocytes, Meth. Enzymol. B 293 (1998) 280–300.
proliferation, Physiol. 19 (2004) 285–292. [25] P. Hausen, M. Riebesell, The Early Development of Xenopus
[8] Z. Wang, Roles of KC channels in regulating tumor cell Laevis, Springer, Berlin, 1990.
proliferation and apoptosis, Pflügers Arch. 448 (2004) [26] A. Brüggemann, W. Stühmer, L.A. Pardo, Mitosis-promoting
274–286. factor-mediated suppression of a cloned delayed rectifier
[9] A.J. Patel, M. Lazdunski, The 2P-domain KC channels: role in potassium channel expressed in Xenopus oocytes, Proc. Natl
apoptosis and tumorigenesis, Pflügers Arch. 448 (2004) Acad. Sci. USA 94 (1997) 537–542.
261–273. [27] L.A. Pardo, A. Brüggemann, J. Camacho, W. Stühmer, Cell
[10] W.D. Kaplan, W.E. Trout III, The behavior of four cycle-related changes in the conducting properties of r-eag KC
neurological mutants of Drosophila, Genetics 61 (1969) channels, J. Cell. Biol. 143 (1998) 767–775.
[28] J. Camacho, A. Sánchez, L.A. Pardo, W. Stühmer, Cyto-
399–409.
skeletal interactions determine the electrophysiological prop-
[11] B. Ganetzky, C.F. Wu, Neurogenetic analysis of potassium
erties of human EAG potassium channels, Pflügers Arch. 441
currents in Drosophila: synergic effects on neuromuscular
(2000) 167–174.
transmission in double mutants, J. Neurogenet. 1 (1983)
[29] L. Bianchi, B. Wible, A. Arcangeli, M. Taglialatela, F. Morra,
17–28.
P. Castaldo, et al., herg encodes a KC current highly
[12] J.W. Warmke, R. Drysdale, B. Ganetzky, A distinct potassium
conserved in tumors of different histogenesis: a selective
channel polypeptide encoded by the Drosophila eag locus,
advantage for cancer cells? Cancer Res. 58 (1998) 815–822.
Science 252 (1991) 551–555.
[30] L.A. Pardo, B. Hemmerlein, A. Sánchez, A. Suckow,
[13] A. Brüggemann, L.A. Pardo, W. Stühmer, O. Pongs, Ether à
H. Knötgen, T. Schliephacke, et al., Eag1 potassium channel
go-go encodes a voltage-gated channel permeable to KC and
as cancer therapy target, Eur. J. Cancer 38 (2002) 104.
Ca2C and modulated by cAMP, Nature 365 (1993) 445–448.
[31] R. Meyer, R. Schönherr, O. Gavrilova-Ruch, W. Wohlrab,
[14] J.W. Warmke, B. Ganetzky, A family of potassium channel S.H. Heinemann, Identification of ether à go-go and calcium-
genes related to eag in Drosophila and mammals, Proc. Natl activated potassium channels in human melanoma cells,
Acad. Sci. USA 91 (1994) 3438–3442. J. Membr. Biol. 171 (1999) 107–115.
[15] C.K. Bauer, J.R. Schwarz, Physiology of EAG KC channels, [32] R. Meyer, S.H. Heinemann, Characterization of an eag-like
J. Membr. Biol. 182 (2001) 1–15. potassium channel in human neuroblastoma cells, J. Physiol.
[16] W. Caterall, G. Chandy, G. Gutman, The IUPHAR Compen- 508 (1998) 49–56.
dium on Voltage-Gated Ion Channels, IUPHAR Media, UK, [33] S. Pillozzi, M.F. Brizzi, M. Balzi, O. Crociani, A. Cherubini,
2002. L. Guasti, et al., HERG potassium channels are constitutively
[17] T. Occhiodoro, L. Bernheim, J.H. Liu, P. Bijlenga, expressed in primary human acute myeloid leukemias and
M. Sinnreich, C.R. Bader, J. Fischer-Lougheed, Cloning of a regulate cell proliferation of normal and leukemic hemopoietic
human ether à go-go potassium channel expressed in progenitors, Leukemia 16 (2002) 1791–1798.
myoblasts at the onset of fusion, Fed. Eur. Bioch. Soc. Lett. [34] G.A.M. Smith, H. Tsui, E.W. Newell, X. Jiang, X.P. Zhu,
434 (1998) 177–182. F.W.L. Tsui, L.C. Schlichter, Functional up-regulation of
[18] L.A. Pardo, D. Del Camino, A. Sánchez, F. Alves, HERG KC channels in neoplastic hematopoietic cells, J. Biol.
A. Brüggemann, S. Beckh, W. Stühmer, Oncogenic potential Chem. 277 (2002) 18528–18534.
of EAG KC channels, Eur. Mol. Biol. Org. J. 18 (1999) [35] T. Suzuki, K. Takimoto, Selective expression of HERG and
5540–5547. Kv2 channels influences proliferation of uterine cancer cells,
[19] J.L. Pellequer, R. Brudler, E.D. Getzhoff, Biological sensors. Int. J. Oncol. 25 (2004) 153–159.
More than one way to sense oxygen, Curr. Biol. 9 (1999) [36] E. Lastraioli, L. Guasti, O. Crociani, S. Polvani, G. Hofmann,
R416–R418. H. Witchel, et al., herg1 and HERG1 protein are over-
[20] J.I. Vandenberg, B.D. Walker, T.J. Campbell, HERG KC expressed in colorectal cancers and regulate cell invasion of
channels: friend and foe, TIPS 22 (2001) 240–246. tumor cells, Cancer Res. 64 (2004) 606–611.
J. Camacho / Cancer Letters 233 (2006) 1–9 9

[37] A. Cherubini, G.L. Taddei, O. Crociani, M. Paglierani, [48] G. Hofmann, P.A. Bernabei, O. Crociani, A. Cherubini,
A.M. Buccoliero, L. Fontana, et al., HERG potassium L. Guasti, S. Pillozzi, et al., HERG KC channel activation
channels are more frequently expressed in human endometrial during b1 integrin-mediated adhesion to fibronectin induces
cancer as compared to non-cancerous endometrium, Br. an up-regulation of avb3 integrin in the preosteoclastic
J. Cancer 83 (2000) 1722–1729. leukemia cell line FLG 29.1, J. Biol. Chem. 276 (2001)
[38] C.K. Bauer, I. Wulfsen, R. Schafer, G. Glassmeier, 4923–4931.
S. Wimmers, J. Flitsch, et al., HERG KC currents in human [49] H. Wang, Y. Zhang, L. Cao, H. Han, J. Wang, B. Yang, et al.,
prolactin-secreting adenoma cells, Pflügers Arch. 445 (2003) HERG KC channel, a regulator of tumor cell apoptosis and
589–600. proliferation, Cancer Res. 62 (2002) 4843–4848.
[39] S. Patt, K. Preußat, C. Beetz, R. Kraft, M. Schrey, R. Kalff, [50] D. Thomas, B. Gut, S. Karsai, A.B. Wimmer, K. Wu,
et al., Expression of ether à go-go potassium channels in G. Wendt-Nordhal, et al., Inhibition of cloned HERG
human gliomas, Neurosc. Lett. 368 (2004) 249–253. potassium channels by antiestrogen tamoxifen, Naunyn-
[40] L.M.B. Farias, D.B. Ocaña, L. Dı́az, F. Larrea, E. Avila- Schmiedeberg’s Arch. Pharmacol. 368 (2003) 41–48.
Chávez, A. Cadena, et al., Ether à go-go potassium channels as [51] D. Thomas, A.B. Wimmer, K. Wu, B.C. Hammerling,
human cervical cancer markers, Cancer Res. 64 (2004) E.K. Ficker, Y.A. Kuryshev, et al., Inhibition of humean
6996–7001. ether à go-go potassium channels by a1-adrenoceptor
[41] H. Ouadid-Ahidouch, X. Le Bourhis, M. Roudbaraki,
antagonists prazosin, doxazosin, and terazosin, Naunyn-
R.A. Toillon, P. Delcourt, N. Prevarskaya, Changes in the
Schmiedeberg’s Arch. Pharmacol. 369 (2004) 462–472.
KC current density of MCF-7 cells during progression through
[52] O. Gavrilova-Ruch, K. Schönherr, G. Gessner, R. Schönherr,
the cell cycle: possible involvement of a h-ether à go-go KC
T. Klapperstück, W. Wohlrab, S.H. Heinemann, Effects of
channel, Receptors Channels 7 (2001) 345–356.
imipramine on ion channels and proliferation of IGR1
[42] O. Crociani, L. Guasti, M. Balzi, A. Becchetti, E. Wanke,
melanoma cells, J. Membr. Biol. 188 (2002) 137–149.
M. Olivotto, et al., Cell cycle-dependent expression of HERG1
[53] R.E. Garcı́a-Ferreiro, D. Kerschensteiner, F. Major, F. Monje,
and HERG1B isoforms in tumor cells, J. Biol. Chem. 278
W. Stühmer, L.A. Pardo, Mechanism of block of hEag1 KC
(2003) 2947–2955.
channels by imipramine and astemizole, J. Gen. Physiol. 124
[43] L. Fontana, M. D’Amico, O. Crociani, T. Biagiotti,
M. Solazzo, B. Rosati, et al., Long-term modulation of (2004) 301–317.
HERG channel gating in hypoxia, Biochem. Biophys. Res. [54] E.T. Piros, L.J. Shen, X.Y. Huang, Purification of an EH
Commun. 286 (2001) 857–862. domain-binding protein from rat brain that modulates the
[44] W.Y. Kim, W.G. Kaelin, Role of VHL gene mutation in gating of the rat ether à go-go channel, J. Biol. Chem. 274
human cancer, J. Clin. Oncol. 22 (2004) 4991–5004. (1999) 33677–33683.
[45] H. Murata, N. Tajima, Y. Nagashima, M. Yao, M. Baba, [55] B. Wendland, Epsins: adaptors in endocytosis?, Nature Rev.
M. Goto, et al., Von Hippel-Lindau tumor supresor protein Mol. Cell. Biol. 3 (2002) 971–977.
transforms human neuroblastoma cells into functional neuron- [56] L. Pei, O. Wiser, A. Slavin, D. Mu, S. Powers, L.Y. Jan,
like cells, Cancer Res. 62 (2002) 7004–7011. T. Hoey, Oncogenic potential of TASK3 (Kcnk9) depends on
[46] R. Schönherr, K. Löber, S.H. Heinemann, Inhibition of human KC channel function, Proc. Natl Acad. Sci. USA 100 (2003)
ether à go-go potassium channels by Ca2C/calmodulin, Eur. 7803–7807.
Mol. Biol. Org. J. 19 (2000) 3263–3271. [57] A. Arcangeli, A. Becchetti, A. Cherubini, O. Crociani,
[47] F.S. Cayabyab, L.C. Schlichter, Regulation of an ERG KC P. Defilipi, L. Guasti, et al., Physical and functional interaction
current by Src tyrosine kinase, J. Biol. Chem. 277 (2002) between integrins and hERG potassium channels, Biochem.
13673–13681. Soc. Trans. 32 (2004) 826–827.

Das könnte Ihnen auch gefallen