ClC-3 expression enhances etoposide resistance by increasing acidification of the late

publicité
979
ClC-3 expression enhances etoposide resistance by
increasing acidification of the late
endocytic compartment
Karsten H. Weylandt,1 Maxim Nebrig,1,2
Nils Jansen-Rosseck,1,2 Joanna S. Amey,2
David Carmena,2 Bertram Wiedenmann,1
Christopher F. Higgins,2 and Alessandro Sardini2
1
Department of Gastroenterology, Charité University Medicine,
Campus Virchow-Klinikum, Berlin, Germany and 2Medical
Research Council Clinical Sciences Centre, Imperial College
Faculty of Medicine, Hammersmith Hospital Campus, London,
United Kingdom
Abstract
Resistance to anticancer drugs and consequent failure of
chemotherapy is a complex problem severely limiting
therapeutic options in metastatic cancer. Many studies
have shown a role for drug efflux pumps of the ATPbinding cassette transporters family in the development of
drug resistance. ClC-3, a member of the CLC family of
chloride channels and transporters, is expressed in
intracellular compartments of neuronal cells and involved
in vesicular acidification. It has previously been suggested
that acidification of intracellular organelles can promote
drug resistance by increasing drug sequestration. Therefore, we hypothesized a role for ClC-3 in drug resistance.
Here, we show that ClC-3 is expressed in neuroendocrine
tumor cell lines, such as BON, LCC-18, and QGP-1, and
localized in intracellular vesicles colabeled with the late
endosomal/lysosomal marker LAMP-1. ClC-3 overexpression increased the acidity of intracellular vesicles, as
assessed by acridine orange staining, and enhanced
resistance to the chemotherapeutic drug etoposide by
almost doubling the IC50 in either BON or HEK293 cell
lines. Prevention of organellar acidification, by inhibition of
the vacuolar H+-ATPase, reduced etoposide resistance.
No expression of common multidrug resistance transporters, such as P-glycoprotein or multidrug-related protein-1, was detected in either the BON parental cell line or
Received 8/8/06; revised 11/15/06; accepted 1/11/07.
The costs of publication of this article were defrayed in part by the
payment of page charges. This article must therefore be hereby marked
advertisement in accordance with 18 U.S.C. Section 1734 solely to
indicate this fact.
Note: K.H. Weylandt and M. Nebrig contributed equally to this work.
Requests for reprints: Alessandro Sardini, Medical Research Council
Clinical Sciences Centre, Imperial College Faculty of Medicine,
Hammersmith Hospital Campus, Du Cane Road, London W12 0NN,
United Kingdom. Phone: 44-20-8383-8270; Fax: 44-20-8383-8337.
E-mail: [email protected]
Copyright C 2007 American Association for Cancer Research.
doi:10.1158/1535-7163.MCT-06-0475
the derivative clone overexpressing ClC-3. The probable
mechanism of enhanced etoposide resistance can be
attributed to the increase of vesicular acidification as
consequence of ClC-3 overexpression. This study therefore provides first evidence for a role of intracellular CLC
proteins in the modulation of cancer drug resistance. [Mol
Cancer Ther 2007;6(3):979 – 86]
Introduction
Metastatic cancers are usually treated using chemotherapy
regimens to target tumor cells disseminated throughout the
organism. Yet, although the initial response to treatment
might be good, multidrug resistance eventually emerges,
leading to treatment failure and death. Several mechanisms
are responsible for the development of drug resistance at a
cellular level. Although it has been shown that target
modification can lead to drug resistance, it is frequently the
emergence of expression of ATP-binding cassette transporters, responsible for drug efflux from cancer cells, that
actually leads to resistance (1). Proteins of this family have
also been shown to sequester drugs in intracellular
membrane compartments, thereby decreasing effective
drug concentrations and inducing resistance (2).
Intracellular acidic compartments can sequester basic
anticancer drugs, which accumulate passively in response
to the pH gradient, and thus contribute to drug resistance.
Several studies have implied a link between drug resistance
and intravesicular acidity either by comparison of drugsensitive and drug-resistant cell lines (3, 4) or through
treatment with agents disrupting organellar pH (5). A
recent study showed that omeprazole, a proton pump
inhibitor used in antiacid treatment of peptic disease, and
the vacuolar proton pump (v-H+-ATPase) inhibitor bafilomycin A increased cytotoxicity of the basic chemotherapeutic drugs doxorubicin and mitoxantrone (6). Similarly,
in vivo studies of human melanoma xenografts in severe
combined immunodeficient mice showed that proton
pump inhibitors affected sensitivity to cisplatin (7).
Moreover, a study with a novel v-H+-ATPase inhibitor
showed an increased effectiveness of topotecan chemotherapy when combined with this inhibitor, supporting the
hypothesis of a role for intracellular acidic compartments in
mediating chemotherapy resistance (8). Vesicular proton
pump inhibition might therefore be a future target option
for increasing the effectiveness of cancer chemotherapy.
Although these studies have focused on the role of
intracellular proton pumps and transporters for chemotherapy resistance, the presence also of an anion shunt
current is a requisite for the accumulation of protons in
intracellular membrane compartments. This current is
necessary to dissipate the charge gradient generated by
Mol Cancer Ther 2007;6(3). March 2007
Downloaded from mct.aacrjournals.org on July 8, 2017. © 2007 American Association for Cancer Research.
980 ClC-3 and Cancer Drug Resistance
the electrogenic activity of the v-H+-ATPase during the H+
transport. Chloride channels/transporters of the CLC
family have been suggested to provide the anion shunt
current (9). We therefore tested the hypothesis of an
involvement of intracellular CLC channels/transporters in
drug resistance.
ClC-3, a member of the CLC family of chloride channels
and transporters, is expressed predominantly in the cells
of the nervous system and is located in acidic intracellular
compartments (10, 11). ClC-3 has been suggested to
generate a shunt current of chloride for v-H+-ATPases,
thereby aiding the acidification of endosomes and synaptic
vesicles (10) as well as lysosomes (12). This hypothesis was
confirmed in experiments directly showing a role for ClC3 in endosomal acidification (13). The homologous CLC
proteins ClC-5 and ClC-7 play also a role in modulating
the pH of intracellular compartments and vesicle trafficking in the cell (9, 14). Because ClC-3 is expressed in
pheochromocytoma cells (15), we hypothesized the presence of ClC-3 also in gastrointestinal neuroendocrine
tumor cells.
Etoposide, in combination with a platin substance, is the
mainstay of chemotherapy treatment of undifferentiated,
high-grade neuroendocrine tumors (16 – 19) and small cell
lung cancers (20). At the time of diagnosis, these tumors are
mostly disseminated so that systemic chemotherapy
remains as the only applicable treatment option. Although
initial responses to chemotherapy treatment are usually
good with significant reduction of tumor size, most of these
tumors rapidly develop chemotherapy resistance. As a
consequence, nearly all patients with these cancers eventually succumb to progressive disease.
As for most other anticancer drugs, etoposide has a basic
pKa (9.8; ref. 21), predisposing it to trapping in acidic
compartments. We therefore investigated the hypothesis
that expression of ClC-3 in a neuroendocrine tumor cell line
increases the acidity of intracellular compartments and
thereby etoposide drug resistance.
Materials and Methods
Cell Culture and Generation of Clonal Cell Lines
BON is a neuroendocrine cell line established from a
human pancreatic neuroendocrine carcinoma (22, 23).
BON cells were cultured at 37jC in a 5% CO2 and
water-saturated atmosphere and grown in DMEM and
F12 (1:1), 25 mmol/L HEPES, 10% fetal bovine serum
(FBS), and 10 Ag/mL ciprofloxacin (Ciprobay). LCC-18 is a
cell line established from a human neuroendocrinedifferentiated colonic carcinoma (24). LCC-18 cells were
cultured in RPMI 1640, 10% FBS, insulin, transferrin, and
selenium liquid medium supplement (Sigma, St. Louis,
MO), and 10 Ag/mL ciprofloxacin (Ciprobay). QGP-1 cells,
a human pancreatic carcinoma cell line of islet origin
(25), were cultured in DMEM and 10% FBS. PC12 cells are
an established line derivative of a rat pheochromocytoma
and were cultured in high-glucose DMEM, 10% FBS,
and 5% horse serum. CaCo-2 is a cell line established
from an adenocarcinoma of the colon and was cultured in
MEM plus 10% FCS. The BON cell clone permanently
overexpressing ClC-3-green fluorescent protein (GFP)
was generated by transfection with the pCIneo plasmid
coding for the fusion protein and selection in G418
following a similar procedure used to generate the
HEK293 clone expressing ClC-3-GFP (11). HEK293 clone
expressing ClC-3-GFP was cultured in DMEM, 10% FBS,
and 500 Ag/mL G418. NIH3T3-MDR1 cell line (26), a
derivative of the mouse fibroblast NIH3T3 line, permanently transfected with the human MDR1 gene coding
for P-glycoprotein, was grown in DMEM, 10% FBS, and
1 Ag/mL colchicine. SW-620 cell line, derived from a
human colon adenocarcinoma (27), was grown in RPMI
1640 and 10% FBS.
Total Membrane Preparation and Western Blotting
Cells, cultured as described above, were homogenized
and crude membrane preparation was obtained (see
Supplementary Data for a detailed description).3 Membrane
proteins were resolved by SDS-PAGE and transferred onto
polyvinylidene difluoride membranes (Immobilon-P, Millipore, Billiberia, MA). ClC-3 proteins were detected with the
D1-specific polyclonal antibody against ClC-3 (1:100 overnight at 5jC; ref. 11), P-glycoprotein was detected with the
monoclonal antibody C219 (1:1,000 overnight; Dako, Carpinteria, CA), and the multidrug-related protein-1 (MRP-1)
was detected with the monoclonal antibody MRPm5 (1:50
overnight; Abcam, Cambridge, United Kingdom) followed
by appropriate horseradish peroxidase secondary antibodies (1:1,000; Dako). Signal was visualized by chemiluminescence (Amersham ECL system; Amersham, Little
Chalfont, United Kingdom). A detailed description of
the Western blotting procedure is available in the Supplementary Data.3
Immunocytochemistry, Life Stains, and Confocal
Imaging
For immunocytochemistry on BON cells permanently
transfected with ClC-3-GFP, cells were plated on poly-Llysine – coated (Sigma) glass coverslips and cultured in sixwell plates for 24 to 48 h before fixation with a solution of
4% formaldehyde/4% sucrose in PBS buffer or methanol at
20jC. Cells were then permeabilized with 0.1% Triton
X-100 for 4 min and costained with antibodies against
chromogranin A (DAK-A3; Dako), synaptophysin (gift from
C. Groetzinger, Charité University Medicine, Campus
Virchow-Klinikum, Berlin, Germany), EEA-1 (1:50; Santa
Cruz Biotechnology, Santa Cruz, CA), and LAMP-1 (1:50;
Santa Cruz Biotechnology) for 12 h at 4jC after blocking
with 0.2% fish skin gelatin in PBS. All primary antibodies
were detected by appropriate secondary antibody conjugated to the fluorophore Alexa Fluor 568 (1:600; Molecular
Probes/Invitrogen, Carlsbad, CA). Cells were also stained
with 4¶,6-diamidino-2-phenylindole (DAPI; 15 mg/mL;
1:10,000 dilution; Molecular Probes) for nuclear DNA.
3
Supplementary material for this article are available at Molecular Cancer
Therapeutics Online (http://mct.aacrjournals.org/).
Mol Cancer Ther 2007;6(3). March 2007
Downloaded from mct.aacrjournals.org on July 8, 2017. © 2007 American Association for Cancer Research.
Molecular Cancer Therapeutics
Coverslips were mounted in Vectashield (Vector Laboratories, Burlingame, CA) for examination and cells were
imaged using a Leica SP confocal microscope equipped
with a 100/1.4 NA PlanApoChromat oil immersion
objective lens (Leica, Wetzlar, Germany) as described in
the Supplementary Data.3 Analysis of LAMP-1 signal at the
fluorescence-activated cell sorting was conducted by fixing
the resuspended cells in 70% ice-cold methanol and staining
for LAMP-1 with the above described antibody (1:50) for
2 h at room temperature in 0.2% fish skin gelatin, and then,
the primary antibody was recognized by goat anti-mouse
Cy5-conjugated antibody (1:200; 30 min at room temperature; Abcam; see Supplementary Data for details of
fluorescence-activated cell sorting settings).3 Control was
obtained by exposure of the preparation to the secondary
antibody only. To identify acidic compartments, live BON
cells expressing ClC-3-GFP were stained for 30 min at 37jC
with 50 nmol/L DND-99/LysoTracker Red, a low pHspecific fluorophore (Molecular Probes). Cells were imaged
in a perfusion chamber with a Leica SP confocal microscope
equipped with 63 1.32 NA PlanApoChromat oil immersion objective, and to avoid bleed through, the fluorophores
were excited sequentially as described in the Supplementary Data.3 Similarly, identification of acidic compartments
was achieved by staining the cells in the perfusion chamber
for 5 min with 10 Amol/L acridine orange (Molecular
Probes) until organelle steady-state accumulation was
reached (see Supplementary Data and the next paragraph
for further details).3 All images were analyzed with MetaMorph 5.0v1 software (Universal Imaging, Downingtown,
PA). Figures were assembled for publication with Adobe
Photoshop 6.0 software (Adobe Systems, San Jose, CA).
Quantification of Acridine Orange Fluorescence from
Acidic Compartments
Acridine orange (AO) was used to measure acidic
compartments. The fluorophore accumulates in acidic
compartments of cells where it remains trapped following
protonation. On increasing concentrations of AO, the
spontaneous formation of dimeric AO molecules leads to
a shift in its fluorescence emission maximum from green to
far red, thereby labeling acidic compartments in red (28).
Accumulation in the acidic compartment is dependent on
extracellular acridine orange concentration [AO]ext and
proportional to the magnitude of the DpH across the
compartment membrane. When the external pH is kept
constant, the [AO]int of the acidic compartment is linearly
proportional to the [AO]ext. This was exploited to quantify
the acidic compartment in large numbers of cells by using
flow cytometry (see Supplementary Data3 and ref. 29 for
further details). Data so acquired were analyzed off-line by
FlowJo (TriStar, Inc., San Carlos, CA). In brief, the mean
average intensity fluorescence signal in the FL-3 channel
from a sample population of 10,000 cells was assessed for
each [AO]ext to which cells were exposed. The mean FL-3
intensity values were plotted against the [AO]ext and fitted
with a linear regression of the first order by using the
Marquardt-Levenberg algorithm based on the least-square
method (SigmaPlot version 8). The slopes of the relation-
Figure 1. Expression and localization of ClC-3 in neuroendocrine cell
lines. A, Western blot analysis of ClC-3 expression in cell membrane
preparations. Lane 1, PC12 cells; lane 2, BON cell line; lane 3, LCC-18 cell
line; lane 4, CaCo-2 cell line; lane 5, QGP-1 cell line. In each lane, 50 Ag of
total protein were loaded. Molecular weights are in kDa. B, confocal
images of BON cells overexpressing ClC-3-GFP. i, GFP fluorescence in
green ; ii, staining of the same cells with LAMP-1 in red; iii, overlay of (i )
and (ii) with colocalization in yellow . Nuclei are labeled in blue by DAPI
staining. Bar, 10 Am.
ship obtained for different treatments were statistically
compared by the parallelism test (F test; significantly
different for P V 0.05). For each treatment, at least three
independent measurements were done.
Cell Proliferation Assay
Drug cytotoxicity was assessed with the Cell Proliferation
Kit II as described by the manufacturer (Roche Diagnostics,
Mol Cancer Ther 2007;6(3). March 2007
Downloaded from mct.aacrjournals.org on July 8, 2017. © 2007 American Association for Cancer Research.
981
982 ClC-3 and Cancer Drug Resistance
Indianapolis, IN). Cells were seeded in 96-well tissue
culture plates with 100 AL of appropriate tissue culture
medium. After 24 h, culture medium was exchanged with
fresh medium without phenol red (to avoid interference
with the absorption readings) and containing appropriate
concentration of etoposide (stock solution in DMSO).
Percentage of cell survival was expressed as normalized
average absorption values from four replicates per each
drug concentration F SD. The concentration of drug
required to decrease cell proliferation by 50% (IC50) was
Figure 2. ClC-3 localizes in an acidic compartment of BON cells, increases its acidity, and enhances resistance to etoposide. A, confocal images of live
BON cells overexpressing ClC-3-GFP (green, left ) before staining acidic compartments for 5 min with 10 Amol/L acridine orange (red, middle). Right,
yellow, colocalization of GFP and acridine orange. Blue, nuclei were labeled with DAPI at the end of the experiment following permeabilization. Bar, 10 Am.
B, confocal images of live BON cells overexpressing ClC-3-GFP (green, left ) after 30 min of incubation with 50 nmol/L LysoTracker Red (red, middle).
Right, yellow, colocalization overlay of GFP and LysoTracker. Bar, 10 Am. C, assessment of acidity of intracellular compartments by flow cytometry
showing mean acridine orange fluorescence intensity of BON cells sorted for high (red trace ) and low (black trace ) expression of ClC-3-GFP. The slopes
are significantly different (P < 0.01, F test). D, cell survival after 48 h of exposure to etoposide. BON cells were sorted for high (red trace ) or low (black
trace ) expression of ClC-3-GFP. The IC50 of the two tested cell populations are significantly different (P < 0.01, Student’s t test); points, data average;
bars, SD.
Mol Cancer Ther 2007;6(3). March 2007
Downloaded from mct.aacrjournals.org on July 8, 2017. © 2007 American Association for Cancer Research.
Molecular Cancer Therapeutics
Figure 3.
Expression of ClC-3 in HEK293 cells increases the acidity of
intracellular compartment and enhances resistance to etoposide.
A, assessment of acidity of intracellular compartments by flow cytometry
showing mean acridine orange fluorescence intensity of HEK293 cells
sorted for high (red trace ) and low (black trace ) expression of ClC-3-GFP.
Slopes are significantly different (P < 0.01, F test). B, cell survival after
48 h of exposure to etoposide. HEK293 cells were sorted for high (red
trace ) and low (black trace ) expression of ClC-3-GFP. The IC50 of the two
tested cell populations are significantly different (P < 0.01, Student’s
t test); points, data average; bars, SD.
determined for each treatment from the concentrationresponse curve of the 2,3-bis[2-methoxy-4-nitro-5-sulfophenyl]-2H-tetrazolium-5-carboxanilide inner salt assay
and statistically compared by Student’s t test. The concentration-response curves were obtained by fitting the
experimental data by nonlinear regression with a logistic
curve (SigmaPlot version 8.0).
Results
Western blot analysis (Fig. 1A), using a polyclonal antibody
specific against ClC-3 (11), showed endogenous expression
of ClC-3 in the BON cell line derived from a human
pancreatic neuroendocrine tumor and well established as
a model for neuroendocrine cells (23). ClC-3 was also
expressed in other human neuroendocrine cell lines
derived from tumors of the gastrointestinal tract, such as
LCC-18 and QGP-1. ClC-3 expression was confirmed in
PC12 cells, a cell line derived from rat pheochromocytoma
and previously shown to express ClC-3 (15). In contrast, the
gastrointestinal tumor cell line CaCo-2, established from a
human colon adenocarcinoma, showed significantly lower
levels of ClC-3 expression.
To study the intracellular localization of ClC-3 in
neuroendocrine cells, a clone of BON cells constitutively
expressing a C-terminally GFP-tagged ClC-3 protein was
generated (see Materials and Methods). ClC-3-GFP was
primarily expressed in intracellular vesicles colabeled by
the late endosomal/lysosomal marker LAMP-1 (Fig. 1B).
No colocalization of ClC-3 was observed with markers of
neuroendocrine secretory vesicles, such as chromogranin A
or synaptophysin (data not shown). The late endosomal/
lysosomal compartments are acidic and play a critical role
in cellular metabolism as the site of protein degradation
and of specialized autolytic processes. A v-type H+-ATPase
in the vesicular membrane is responsible for establishment
of the acidic intracellular pH. Because this protein is an
electrogenic pump, it is generally assumed that efficient
acidification also requires an anionic shunt current to
abolish the charge buildup generated by the transport of H+
(9). Live BON cells expressing ClC-3-GFP were stained for
intracellular acidic compartments using either the weak
amine base acridine orange (Fig. 2A) or the red fluorescent
cyanine dye DND-99/LysoTracker Red (Fig. 2B). The
colocalization with fluorescent ClC-3-GFP showed that
ClC-3 resides in the membrane of acidic vesicles.
To assess the acidity of vesicles overexpressing ClC-3, we
used a titration approach using acridine orange as a
fluorophore and flow cytometry analysis (ref. 29; see also
Materials and Methods and Supplementary Data).3 Briefly,
changes in acidity of intracellular organelles can be
followed by measuring the change in slope of the
relationship between the intensity of AO fluorescence
emission in the far red (FL-3 channel) and the concentration
of [AO]ext to which the cells are exposed. To maintain ClC-3GFP expression, cells were grown in selective medium
containing G418. This has the drawback that any detectable
functional difference between the overexpressing clone and
the parental cell line could be either associated to the
expression of ClC-3 or to unavoidable and unpredictable
changes due to the selection process. To avoid this, we
exploited the naturally occurring difference in ClC-3-GFP
level expression within cells of the same clone. Clonal BON
cells expressing ClC-3-GFP were fluorescence-sorted for
populations with high and low ClC-3-GFP expression and
compared for acidity of intracellular organelles. Higher
ClC-3-GFP expression conferred an increase of the acidity of
intracellular organelles as shown by a significantly steeper
slope, in comparison with BON cells expressing low ClC-3GFP (Fig. 2C). Previous studies have suggested that
intracellular acidic compartments may sequester basic
drugs, thereby decreasing effective concentrations of drug
in the cell nucleus and cytoplasm and, consequently,
cytotoxicity. BON cells sorted for high ClC-3-GFP expression showed, in comparison with BON cells sorted for low
ClC-3-GFP expression, a significantly increased resistance
to etoposide, a chemotherapeutic drug with a basic pKa
Mol Cancer Ther 2007;6(3). March 2007
Downloaded from mct.aacrjournals.org on July 8, 2017. © 2007 American Association for Cancer Research.
983
984 ClC-3 and Cancer Drug Resistance
(Fig. 2D). The cell growth rate was unaffected by ClC-3
expression (see Supplementary Fig. S1).3 To validate these
results in a different cell model, a clone of HEK293 cells
permanently expressing ClC-3-GFP was used (11). As for
the neuroendocrine BON cells, HEK293 cell populations
sorted for high expression of ClC-3-GFP showed a
significant increase in intravesicular acidity and etoposide
resistance (Fig. 3A and B, respectively).
The intensity of the red fluorescent acridine orange signal,
as determined by flow cytometry, is dependent on both the
acidity and number of intracellular organelles. We assessed
whether ClC-3 overexpression leads to an up-regulation of
the formation of late endosomes and lysosomes, which
could potentially account for the differences observed.
However, BON cells overexpressing ClC-3 were indistinguishable from untransfected BON cells in expression levels
of the lysosomal marker LAMP-1 (Fig. 4A, right hand side
peaks). Similar results were obtained in HEK293 cells
overexpressing ClC-3-GFP (data not shown).
To confirm the role of acidification of vesicular compartments in etoposide resistance, BON cells overexpressing
ClC-3-GFP were exposed to concanamycin A, a specific
inhibitor of v-H+-ATPase, and then cultured in the
presence of etoposide. Although concanamycin A treatment alone did not affect cell survival (see Supplementary
Fig. S2),3 pretreatment with concanamycin A significantly
increased cells sensitivity to etoposide (Fig. 4B). We also
excluded that either the parental cell line BON cells or its
derivative clone overexpressing ClC-3-GFP were expressing the multidrug resistance transporter P-glycoprotein
(Fig. 5A) or MRP-1 (Fig. 5B).
ized to the late endocytic compartment in this tumor cell
line. This is consistent with previous data showing
expression of ClC-3 in intracellular membrane compartments of other cell types (10, 12, 13). Previous studies have
also implicated ClC-3 expression in the acidification of
intracellular membrane compartments, a function similar
to that described for ClC-5 and ClC-7 (14, 35 – 37). ClC-7
knockout mouse shows a decreased bone resorption and
osteopetrosis. This is similar to the effect of SB242784, a
potent and selective inhibitor of the osteoclast vesicular
proton pump that inhibits bone resorption in vitro (38) and
Discussion
Many solid tumors have an acidic extracellular environment probably due to glycolytic and anaerobic metabolism
of tumor cells. This leads to intracellular acidification and
increased extrusion of protons from intracellular compartments to prevent cytoplasmic acidosis (30).
Several studies have implied a link between endosomal
and lysosomal acidity and chemotherapy resistance, hypothesizing that weakly basic drugs can be sequestered to
acidic intracellular membrane compartments (31). However, all these studies have focused their view on the role of
the vesicular proton pump, establishing strong evidence for
the vesicular proton pumps as mediators of chemotherapy
resistance due to sequestration and extrusion of chemotherapeutic drugs. Modulation of pH-dependent cellular
resistance mechanisms by targeting of the vacuolar H+ATPase may thus be a novel approach to increase the
therapeutic efficacy of antitumor agents (7, 8, 32 – 34).
The results presented here show for the first time that
increased intracompartmental acidification as a result of
overexpression of an intracellular chloride channel/transporter of the CLC protein family enhances drug resistance.
ClC-3 is expressed in the neuroendocrine gastrointestinal
tumor cell line BON as well as in other gastrointestinal
neuroendocrine cell lines. Overexpressed ClC-3-GFP local-
Figure 4.
Expression of ClC-3 in BON cells does not up-regulate the
endocytic compartment, and reduction of its acidification induces
sensitization to etoposide A, flow cytometry analysis of cells sorted for
amount of LAMP-1 staining. Right hand side peaks, BON cells (black
trace ) and BON cells overexpressing ClC-3-GFP (red trace ); left hand
peaks, dashed lines, control staining with secondary antibody alone.
B, cell survival after 48 h of exposure to etoposide of BON cells
overexpressing ClC-3-GFP. Red trace, cells were pretreated for 30 min
with exposure to 10 nmol/L concanamycin A; black trace, no pretreatment. The IC50 of the two tested cell populations are significantly different
(P < 0.05, Student’s t test); points , data average; bars, SD.
Mol Cancer Ther 2007;6(3). March 2007
Downloaded from mct.aacrjournals.org on July 8, 2017. © 2007 American Association for Cancer Research.
Molecular Cancer Therapeutics
Figure 5.
Analysis of expression of the multidrug resistance transporters P-glycoprotein and MRP-1 in BON cells. A, Western blot analysis of
P-glycoprotein. Lane 1, BON cells; lane 2, BON clone overexpressing ClC3-GFP; lane 3, BON clone overexpressing ClC-3-GFP sorted for high
expression; lane 4, BON clone overexpressing ClC-3 sorted for low
expression; lane 5, NIH-3T3-MDR1 cell line overexpressing P-glycoprotein
as positive control. In each lane, 30 Ag of total membrane protein were
loaded. Molecular weights are in kDa. B, Western blot analysis of MRP-1.
Lanes 1, 2, 3, and 4, as above. As positive controls for MRP-1: SW-620
human colon adenocarcinoma cell line (lane 6), A-549 human adenocarcinoma epithelial cell line (lane 7 ), and T98G human glioblastoma
multiforme cell line (lane 8). In each lane, 25 Ag of total membrane
protein were loaded, except for lanes 7 and 8, where 25 Ag of total protein
from whole homogenate were loaded. Molecular weights are in kDa.
in vivo (39). These findings support further our hypothesis
of intracellular chloride channels/transporters as alternative targets in the modulation of intracellular and intracompartmental pH regulation and function.
We have shown that the observed increase of resistance
to etoposide following overexpression of ClC-3 in BON
cells is probably due to a mechanism of sequestration of the
drug in an acidic compartment because exposure to an
inhibitor of v-H+-ATPase, such as concanamycin A,
reduces the resistance. The observed effect, approximate
doubling of the IC50 by overexpression of ClC-3 (Figs. 2D
and 3B), is comparable with the sensitizing effect of
concanamycin A, which was able to reduce IC50 to half
(Fig. 4B). A similar effect has been reported for a resistant
cell line, derived from proximal tubule cells, exposed to
daunomycin (33), supporting biological relevance of these
results. The magnitude of the effect obtained by either
exposure to concanamycin A or overexpression of ClC-3 is
probably limited by the extent the pH of intracellular
vesicles can be modified. This is supported by the fact
that exposure to 100 nmol/L concanamycin A did not
modify the pH of the vesicles any more than exposure to
10 nmol/L (see Supplementary Fig. S3).3
For the studies presented here, a stable overexpression
system of ClC-3-GFP was used. As the selection procedure
for stably expressing ClC-3 involved selection in G418 for
extended times, it was necessary to control for the fact that
this selection process itself might have changed resistance
to etoposide. This was done by exploiting different levels of
ClC-3 expression in a single ClC-3-GFP – expressing clone,
comparing populations with low ClC-3-GFP expression
(as determined and sorted by the GFP signal) with those
with high ClC-3 GFP expression from the same clone.
Interclonal heterogeneity could thus be minimized, adding
validity to our observations.
The activity of multidrug resistance transporters, such as
ATP-binding cassette transporters, is unlikely to play a role
in the observed enhancement of etoposide resistance
because the parental BON cell line and its derivative overexpressing ClC-3 were found not to express P-glycoprotein,
a common culprit for multidrug resistance (1). Furthermore, there is no expression of MRP-1, an ATP-binding
cassette multidrug resistance transporter expressed also in
the lysosomal compartment and implicated in the establishment of resistance by intralysosomal drug sequestration (2).
Our results may be of relevance to the pharmacokinetics
of chemotherapy regimens in neuroendocrine and neuronal tumors because these tissues endogenously express
high amounts of ClC-3 protein as shown here and
elsewhere (10, 11).
However, the data presented here cannot provide a
correlation between level of ClC-3 expression and etoposide resistance, as ClC-3 presence might not be the ratelimiting step of intravesicular acidification, and a direct
relationship between intravesicular pH and ClC-3 expression is unlikely because pH cannot be lowered beyond a
certain threshold.
In spite of these limitations, our study provides proof of
principle of a role for intracellular chloride channels in
establishing intracompartmental acidity and subsequent
increased resistance to a basic chemodrug, etoposide.
Other proteins of the CLC family could confer a similar
effect. Further studies will be necessary to evaluate these
findings with regard to other proteins of the CLC family as
well as in vivo models of cancer drug resistance and to
determine whether intracellular CLC proteins might be a
target to modulate drug sensitivity in tumors.
References
1. Szakacs G, Paterson JK, Ludwig JA, Booth-Genthe C, Gottesman MM.
Targeting multidrug resistance in cancer. Nat Rev Drug Discov 2006;5:
219 – 34.
2. Rajagopal A, Simon SM. Subcellular localization and activity of
multidrug resistance proteins. Mol Biol Cell 2003;14:3389 – 99.
3. Schindler M, Grabski S, Hoff E, Simon SM. Defective pH regulation of
acidic compartments in human breast cancer cells (MCF-7) is normalized in
adriamycin-resistant cells (MCF-7adr). Biochemistry 1996;35:2811 – 7.
4. Altan N, Chen Y, Schindler M, Simon SM. Defective acidification in
human breast tumor cells and implications for chemotherapy. J Exp Med
1998;187:1583 – 98.
5. Altan N, Chen Y, Schindler M, Simon SM. Tamoxifen inhibits
acidification in cells independent of the estrogen receptor. Proc Natl Acad
Sci U S A 1999;96:4432 – 7.
6. Lee CM, Tannock IF. Inhibition of endosomal sequestration of basic
anticancer drugs: influence on cytotoxicity and tissue penetration. Br
J Cancer 2006;94:863 – 9.
7. Luciani F, Spada M, De Milito A, et al. Effect of proton pump inhibitor
Mol Cancer Ther 2007;6(3). March 2007
Downloaded from mct.aacrjournals.org on July 8, 2017. © 2007 American Association for Cancer Research.
985
986 ClC-3 and Cancer Drug Resistance
pretreatment on resistance of solid tumors to cytotoxic drugs. J Natl
Cancer Inst 2004;96:1702 – 13.
8. Petrangolini G, Supino R, Pratesi G, et al. Effect of a novel vacuolar-H+ATPase inhibitor on cell and tumor response to camptothecins.
J Pharmacol Exp Ther 2006;318:939 – 46.
9. Jentsch TJ, Poet M, Fuhrmann JC, Zdebik AA. Physiological functions
of CLC Cl channels gleaned from human genetic disease and mouse
models. Annu Rev Physiol 2005;67:779 – 807.
10. Stobrawa SM, Breiderhoff T, Takamori S, et al. Disruption of ClC-3,
a chloride channel expressed on synaptic vesicles, leads to a loss of the
hippocampus. Neuron 2001;29:185 – 96.
25. Kaku M, Nishiyama T, Yagawa K, Abe M. Establishment of a
carcinoembryonic antigen-producing cell line from human pancreatic
carcinoma. Gann 1980;71:596 – 601.
26. Pastan I, Gottesman MM, Ueda K, Lovelace E, Rutherford AV,
Willingham MC. A retrovirus carrying an MDR1 cDNA confers multidrug
resistance and polarized expression of P-glycoprotein in MDCK cells. Proc
Natl Acad Sci U S A 1988;85:4486 – 90.
27. Leibovitz A, Stinson JC, McCombs WB III, McCoy CE, Mazur KC,
Mabry ND. Classification of human colorectal adenocarcinoma cell lines.
Cancer Res 1976;36:4562 – 9.
11. Weylandt KH, Valverde MA, Nobles M, et al. Human ClC-3 is not the
swelling-activated chloride channel involved in cell volume regulation.
J Biol Chem 2001;276:17461 – 7.
28. Millot C, Millot JM, Morjani H, Desplaces A, Manfait M. Characterization of acidic vesicles in multidrug-resistant and sensitive cancer cells
by acridine orange staining and confocal microspectrofluorometry.
J Histochem Cytochem 1997;45:1255 – 64.
12. Li X, Wang T, Zhao Z, Weinman SA. The ClC-3 chloride channel
promotes acidification of lysosomes in CHO-K1 and Huh-7 cells. Am
J Physiol Cell Physiol 2002;282:C1483 – 91.
29. Dzekunov SM, Ursos LM, Roepe PD. Digestive vacuolar pH of intact
intraerythrocytic P. falciparum either sensitive or resistant to chloroquine.
Mol Biochem Parasitol 2000;110:107 – 24.
13. Hara-Chikuma M, Yang B, Sonawane ND, Sasaki S, Uchida S,
Verkman AS. ClC-3 chloride channels facilitate endosomal acidification
and chloride accumulation. J Biol Chem 2005;280:1241 – 7.
30. Vaupel P, Kallinowski F, Okunieff P. Blood flow, oxygen and nutrient
supply, and metabolic microenvironment of human tumors: a review.
Cancer Res 1989;49:6449 – 65.
14. Kasper D, Planells-Cases R, Fuhrmann JC, et al. Loss of the chloride
channel ClC-7 leads to lysosomal storage disease and neurodegeneration.
EMBO J 2005;24:1079 – 91.
31. Raghunand N, Martinez-Zaguilan R, Wright SH, Gillies RJ. pH and
drug resistance. II. Turnover of acidic vesicles and resistance to
weakly basic chemotherapeutic drugs. Biochem Pharmacol 1999;57:
1047 – 58.
15. Salazar G, Love R, Styers ML, et al. AP-3-dependent mechanisms
control the targeting of a chloride channel (ClC-3) in neuronal and nonneuronal cells. J Biol Chem 2004;279:25430 – 9.
16. Moertel CG. Karnofsky memorial lecture. An odyssey in the land of
small tumors. J Clin Oncol 1987;5:1502 – 22.
32. McSheehy PM, Troy H, Kelland LR, Judson IR, Leach MO, Griffiths
JR. Increased tumour extracellular pH induced by Bafilomycin A1 inhibits
tumour growth and mitosis in vivo and alters 5-fluorouracil pharmacokinetics. Eur J Cancer 2003;39:532 – 40.
17. Fjallskog ML, Granberg DP, Welin SL, et al. Treatment with cisplatin
and etoposide in patients with neuroendocrine tumors. Cancer 2001;92:
1101 – 7.
33. Ouar Z, Bens M, Vignes C, et al. Inhibitors of vacuolar H+-ATPase
impair the preferential accumulation of daunomycin in lysosomes and
reverse the resistance to anthracyclines in drug-resistant renal epithelial
cells. Biochem J 2003;370:185 – 93.
18. O’Toole D, Hentic O, Corcos O, Ruszniewski P. Chemotherapy for
gastroenteropancreatic endocrine tumours. Neuroendocrinology 2004;80:
79 – 84.
34. Izumi H, Torigoe T, Ishiguchi H, et al. Cellular pH regulators:
potentially promising molecular targets for cancer chemotherapy. Cancer
Treat Rev 2003;29:541 – 9.
19. Kulke MH, Mayer RJ. Carcinoid tumors. N Engl J Med 1999;340:
858 – 68.
35. Piwon N, Gunther W, Schwake M, Bosl MR, Jentsch TJ. ClC-5 Cl
channel disruption impairs endocytosis in a mouse model for Dent’s
disease. Nature 2000;408:369 – 73.
20. Jain N, Lam YM, Pym J, Campling BG. Mechanisms of resistance of
human small cell lung cancer lines selected in VP-16 and cisplatin. Cancer
1996;77:1797 – 808.
21. O’Neil MJ, editor. The Merck index: an encyclopedia of chemicals,
drugs, and biologicals. 13th ed. Whitehouse Station (NJ): Merck and Co.;
2001. p. 687.
22. von Wichert G, Jehle PM, Hoeflich A, et al. Insulin-like growth factor-I
is an autocrine regulator of chromogranin A secretion and growth in human
neuroendocrine tumor cells. Cancer Res 2000;60:4573 – 81.
36. Gunther W, Luchow A, Cluzeaud F, Vandewalle A, Jentsch TJ. ClC-5,
the chloride channel mutated in Dent’s disease, colocalizes with the proton
pump in endocytotically active kidney cells. Proc Natl Acad Sci U S A
1998;95:8075 – 80.
37. Hara-Chikuma M, Wang Y, Guggino SE, Guggino WB, Verkman AS.
Impaired acidification in early endosomes of ClC-5 deficient proximal
tubule. Biochem Biophys Res Commun 2005;329:941 – 6.
23. Vitale G, de Herder WW, van Koetsveld PM, et al. IFN-h is a highly
potent inhibitor of gastroenteropancreatic neuroendocrine tumor cell
growth in vitro . Cancer Res 2006;66:554 – 62.
38. Nadler G, Morvan M, Delimoge I, et al. (2Z ,4E )-5-(5,6-dichloro-2indolyl)-2-methoxy-N-(1,2,2,6,6-pentamethylpiperidin-4-yl)-2,4-pentadienamide, a novel, potent and selective inhibitor of the osteoclast V-ATPase.
Bioorg Med Chem Lett 1998;8:3621 – 6.
24. Lundqvist M, Mark J, Funa K, et al. Characterisation of a cell line
(LCC-18) from a cultured human neuroendocrine-differentiated colonic
carcinoma. Eur J Cancer 1991;27:1663 – 8.
39. Visentin L, Dodds RA, Valente M, et al. A selective inhibitor of the
osteoclastic V-H(+)-ATPase prevents bone loss in both thyroparathyroidectomized and ovariectomized rats. J Clin Invest 2000;106:309 – 18.
Mol Cancer Ther 2007;6(3). March 2007
Downloaded from mct.aacrjournals.org on July 8, 2017. © 2007 American Association for Cancer Research.
ClC-3 expression enhances etoposide resistance by
increasing acidification of the late endocytic compartment
Karsten H. Weylandt, Maxim Nebrig, Nils Jansen-Rosseck, et al.
Mol Cancer Ther 2007;6:979-986.
Updated version
Supplementary
Material
Cited articles
Citing articles
E-mail alerts
Reprints and
Subscriptions
Permissions
Access the most recent version of this article at:
http://mct.aacrjournals.org/content/6/3/979
Access the most recent supplemental material at:
http://mct.aacrjournals.org/content/suppl/2007/03/20/6.3.979.DC1
This article cites 38 articles, 17 of which you can access for free at:
http://mct.aacrjournals.org/content/6/3/979.full#ref-list-1
This article has been cited by 5 HighWire-hosted articles. Access the articles at:
http://mct.aacrjournals.org/content/6/3/979.full#related-urls
Sign up to receive free email-alerts related to this article or journal.
To order reprints of this article or to subscribe to the journal, contact the AACR Publications
Department at [email protected].
To request permission to re-use all or part of this article, contact the AACR Publications
Department at [email protected].
Downloaded from mct.aacrjournals.org on July 8, 2017. © 2007 American Association for Cancer Research.
Téléchargement