dc electric fields direct breast cancer cell migration, induce egfr polarization, and increase the...

11
ORIGINAL PAPER DC Electric Fields Direct Breast Cancer Cell Migration, Induce EGFR Polarization, and Increase the Intracellular Level of Calcium Ions Dan Wu Xiuli Ma Francis Lin Ó Springer Science+Business Media New York 2013 Abstract Migration of cancer cells leads to invasion of primary tumors to distant organs (i.e., metastasis). Growing number of studies have demonstrated the migration of various cancer cell types directed by applied direct current electric fields (dcEF), i.e., electrotaxis, and suggested its potential implications in metastasis. MDA-MB-231 cell, a human metastatic breast cancer cell line, has been shown to migrate toward the anode of dcEF. Further characteriza- tions of MDA-MB-231 cell electrotaxis and investigation of its underlying signaling mechanisms will lead to a better understanding of electrically guided cancer cell migration and metastasis. Therefore, we quantitatively characterized MDA-MB-231 cell electrotaxis and a few associated sig- naling events. Using a microfluidic device that can create well-controlled dcEF, we showed the anode-directing migration of MDA-MB-231 cells. In addition, surface staining of epidermal growth factor receptor (EGFR) and confocal microscopy showed the dcEF-induced anodal EGFR polarization in MDA-MB-231 cells. Furthermore, we showed an increase of intracellular calcium ions in MDA-MB-231 cells upon dcEF stimulation. Altogether, our study provided quantitative measurements of electro- tactic migration of MDA-MB-231 cells, and demonstrated the electric field-mediated EGFR and calcium signaling events, suggesting their involvement in breast cancer cell electrotaxis. Keywords Breast cancer cell Á Electrotaxis Á EGFR Á Calcium Á Microfluidic device Abbreviations dcEF Direct current electric fields ECM Extracellular matrix EGFR Epidermal growth factor receptor PDMS Polydimethylsiloxane EI Electrotactic index SEM Standard error of the mean MSD Mean square displacement Introduction Cell migration plays important roles in mediating a wide range of biological processes such as wound healing [1, 2], neuron guidance [3, 4], embryogenesis [5], and cancer metastasis [6, 7]. Among the diverse cellular guiding mechanisms, chemotaxis, a process that cells sense and migrate up a chemical concentration gradient, is well studied [8, 9]. In addition, physical parameters such as direct current electric fields (dcEF) can also direct the migration of various cell types [1013], a process termed Electronic supplementary material The online version of this article (doi:10.1007/s12013-013-9615-7) contains supplementary material, which is available to authorized users. D. Wu Á X. Ma Á F. Lin (&) Department of Physics and Astronomy, University of Manitoba, Winnipeg, MB R3T 2N2, Canada e-mail: fl[email protected] F. Lin Department of Immunology, University of Manitoba, Winnipeg, MB R3E 0T5, Canada F. Lin Department of Biological Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada F. Lin Department of Biosystems Engineering, University of Manitoba, Winnipeg, MB R3T 2N2, Canada 123 Cell Biochem Biophys DOI 10.1007/s12013-013-9615-7

Upload: francis-lin

Post on 11-Dec-2016

218 views

Category:

Documents


4 download

TRANSCRIPT

Page 1: DC Electric Fields Direct Breast Cancer Cell Migration, Induce EGFR Polarization, and Increase the Intracellular Level of Calcium Ions

ORIGINAL PAPER

DC Electric Fields Direct Breast Cancer Cell Migration, InduceEGFR Polarization, and Increase the Intracellular Levelof Calcium Ions

Dan Wu • Xiuli Ma • Francis Lin

� Springer Science+Business Media New York 2013

Abstract Migration of cancer cells leads to invasion of

primary tumors to distant organs (i.e., metastasis). Growing

number of studies have demonstrated the migration of

various cancer cell types directed by applied direct current

electric fields (dcEF), i.e., electrotaxis, and suggested its

potential implications in metastasis. MDA-MB-231 cell, a

human metastatic breast cancer cell line, has been shown to

migrate toward the anode of dcEF. Further characteriza-

tions of MDA-MB-231 cell electrotaxis and investigation

of its underlying signaling mechanisms will lead to a better

understanding of electrically guided cancer cell migration

and metastasis. Therefore, we quantitatively characterized

MDA-MB-231 cell electrotaxis and a few associated sig-

naling events. Using a microfluidic device that can create

well-controlled dcEF, we showed the anode-directing

migration of MDA-MB-231 cells. In addition, surface

staining of epidermal growth factor receptor (EGFR) and

confocal microscopy showed the dcEF-induced anodal

EGFR polarization in MDA-MB-231 cells. Furthermore,

we showed an increase of intracellular calcium ions in

MDA-MB-231 cells upon dcEF stimulation. Altogether,

our study provided quantitative measurements of electro-

tactic migration of MDA-MB-231 cells, and demonstrated

the electric field-mediated EGFR and calcium signaling

events, suggesting their involvement in breast cancer cell

electrotaxis.

Keywords Breast cancer cell � Electrotaxis � EGFR �Calcium � Microfluidic device

Abbreviations

dcEF Direct current electric fields

ECM Extracellular matrix

EGFR Epidermal growth factor receptor

PDMS Polydimethylsiloxane

EI Electrotactic index

SEM Standard error of the mean

MSD Mean square displacement

Introduction

Cell migration plays important roles in mediating a wide

range of biological processes such as wound healing [1, 2],

neuron guidance [3, 4], embryogenesis [5], and cancer

metastasis [6, 7]. Among the diverse cellular guiding

mechanisms, chemotaxis, a process that cells sense and

migrate up a chemical concentration gradient, is well

studied [8, 9]. In addition, physical parameters such as

direct current electric fields (dcEF) can also direct the

migration of various cell types [10–13], a process termed

Electronic supplementary material The online version of thisarticle (doi:10.1007/s12013-013-9615-7) contains supplementarymaterial, which is available to authorized users.

D. Wu � X. Ma � F. Lin (&)

Department of Physics and Astronomy, University of Manitoba,

Winnipeg, MB R3T 2N2, Canada

e-mail: [email protected]

F. Lin

Department of Immunology, University of Manitoba, Winnipeg,

MB R3E 0T5, Canada

F. Lin

Department of Biological Sciences, University of Manitoba,

Winnipeg, MB R3T 2N2, Canada

F. Lin

Department of Biosystems Engineering, University of Manitoba,

Winnipeg, MB R3T 2N2, Canada

123

Cell Biochem Biophys

DOI 10.1007/s12013-013-9615-7

Page 2: DC Electric Fields Direct Breast Cancer Cell Migration, Induce EGFR Polarization, and Increase the Intracellular Level of Calcium Ions

electrotaxis or galvanotaxis. Endogenous dcEF widely

exist in biological systems, and electrotaxis participates in

relevant biological processes such as tissue repair and

embryogenesis [12–15]. Several previous studies demon-

strated electrotaxis of different cancer cells in vitro

including prostate cancer cells [16], lung cancer cells [17],

and breast cancer cells [18], and in most cases the elec-

trotactic migration is correlated with metastatic potential of

the cancer cells, suggesting the possible involvement of

electrotaxis in cancer metastasis. Interestingly, unlike

prostate or lung cancer cells that migrate cathodally, MDA-

MB-231 cells, a human metastatic breast cancer cell line,

move toward the anode of dcEF [18], suggesting that

electrotaxis of different cancer cells can be characteristi-

cally different in specific physiological environments and

disease models. In breast cancers, the most common cancer

in women, the metastatic phase of the disease is the major

cause of death [19, 20]. Therefore, it is important to better

understand the mechanisms of breast cancer cell migration

including the electrical guiding mechanism. Toward this

direction, we in the present study performed quantitative

measurements of breast cancer cell electrotaxis using

MDA-MB-231 cells as a model cell system. Our results

confirmed and quantitatively characterized the anodal

electrotaxis of MDA-MB-231 cells using microfluidic

devices. In addition, we showed that dcEF-induced polar-

ization of epidermal growth factor receptor (EGFR) and an

increase of intracellular calcium ions in MDA-MB-231

cells.

Various environmental factors are involved in mediating

cancer metastasis including extracellular matrix (ECM),

cytokines, growth factors, and their interacting receptors

expressed in cancer cells [21, 22]. Among them, EGFR was

shown to be an essential player in regulating breast cancer

metastasis [23, 24]. Relevantly, electric field can effec-

tively redistribute charged mobile entities in the plane of

cell membrane and EGFR were shown to polarize to the

cathode-facing side of the cell in several cathodally elec-

trotaxing cell types such as bovine corneal epithelial cells

[12, 25, 26], human keratinocytes [27], and lung cancer

cells [17]. Earlier studies also reported similar dcEF-

induced cell surface receptor polarization for concanavalin

A receptor in Xenopus laevis embryonic muscle cells [28]

and murine fibroblastic cells [29]. The re-distribution of

relevant cell surface receptors by dcEF results in more

receptors on the cathode-facing side of the cell, which is

proposed to mediate cell sensing of dcEF and the cathode-

directing electrotactic cell migration [12, 17, 25, 27–29].

Our previous modeling studies support such a receptor

electromigration-based cellular sensing mechanism [30]

and indeed previous studies have shown that EGFR sig-

naling is required for electrotaxis of breast cancer cells

[18]. On the other hand, because EGFR polarizes

cathodally in cathode-directing electrotactic cells while

MDA-MB-231 cells migrate toward the anode of dcEF in

conventional electrotaxis assays, it is interesting to test

whether and how dcEF can polarize EGFR in MDA-MB-

231 cells.

Additionally, both calcium ion release and calcium ion

influx are generally associated with cell migration [31, 32],

and calcium signaling is involved in the migration, inva-

sion, and metastasis of different cancer cells [33]. The

intracellular calcium ion level ([Ca2?]i) in polarized

migrating cells forms a concentration gradient across the

cell body and the elevated [Ca2?]i in the back of the cell

regulates the rear-end retraction [34, 35]. Chemical stim-

ulus such as chemokines can trigger robust calcium flux

through their cell surface G-protein coupled chemokine

receptors [34, 36]. In MDA-MB-231 cells, EGF stimulates

similar [Ca2?]i flux [37]. In electrotaxis, dcEF induces a

transient increase of [Ca2?]i in difference cell types such as

mouse embryo fibroblasts [38] and murine adipose-derived

stromal cells [39], whereas dcEF stimulates a more sus-

tained increase of [Ca2?]i in Dictyostelium cells [40], and

calcium signaling is required for electrotaxis of these cells.

For cancer cells, voltage-gated ion channels such as sodium

channels are shown to mediate electrotactic responses of

prostate cancer cells [16]. Those voltage-gated ion chan-

nels are also expressed in MDA-MB-231 cells, suggesting

the possible involvement of cellular ion signaling in elec-

trotaxis of breast cancer cells.

Taking together, dcEF is a physiologically relevant

guiding mechanism for breast cancer cell migration and

metastasis, and thus there is a great need to better under-

stand the process of electrotactic migration. In the current

study, we performed quantitative measurements of breast

cancer cell (MDA-MB-231 cells) electrotaxis. Further-

more, we focused on EGFR polarization and [Ca2?]i

dynamics to identify these signaling events and their

characteristics in MDA-MB-231 cell electrotaxis. After

initial testing with a simple well plate-based assay, we

further employed microfluidic electrotaxis devices [41] to

test electrotaxis and electrotactic signaling in MDA-MB-

231 cells under better controlled experimental conditions.

Experimental Methods

Cell Culture

MDA-MB-231 cells, a human breast adenocarcinoma cell

line, were purchased from ATCC (Manassas, VA, USA)

with verified lineage and purity. Cells were cultured in

Leibovitz’s L-15 medium supplemented with 10 % FBS

and 1 % penicillin/streptomycin in a 37 �C incubator.

MDA-MB-231 cells were passaged regularly for the use of

Cell Biochem Biophys

123

Page 3: DC Electric Fields Direct Breast Cancer Cell Migration, Induce EGFR Polarization, and Increase the Intracellular Level of Calcium Ions

specific experiments throughout this study. Trypsin–EDTA

was purchased from ATCC (Manassas, VA, USA) as the

cell dissociation buffer.

Electrotaxis Experiments Using Well Plates

A piece of coverslip (VWR micro cover glass No. 1,

thickness is 0.13–0.17 mm) was placed in the well plate

(COSTAR 24 well plate, 15.6 mm well diameter, Corning,

NY, USA). The coverslip was coated with 2 lg/mL of

Collagen Type IV (Sigma-Aldrich, Inc., MO, USA) for

30 min at room temperature followed by 2 % BSA

blocking for another hour at 37 �C before the experiment,

providing a substrate for cell adhesion and migration. The

coating concentration of Collagen Type IV was selected

according to the previous literature [42]. For each experi-

ment, cells were seeded on the coverslip and the well was

filled with 1 mL culture medium. Then a pair of platinum

electrodes connecting to a DC power supply was placed in

the well plate with the separation distance similar to the

diameter of the well to apply the electric field. After cali-

brating the electrical current, the well plate was placed

under a microscope (Nikon Ti-U) and cell migration in the

well was recorded by time-lapse microscopy at 1 frame/

min for 1–2 h using a CCD camera. The image acquisition

was controlled by the NIS Elements software.

Electrotaxis Experiments Using Microfluidic Devices

A previously developed polydimethylsiloxane (PDMS)

microfluidic electrotaxis device was modified for the

present study [41] (Fig. S1). In brief, the microfluidic

device with a simple straight channel was designed in

Freehand 9.0 (Macromedia) and the design was printed to a

transparency mask by a high resolution printer. The mas-

ters were fabricated at The Nano Systems Fabrication

Laboratory (NSFL) at the University of Manitoba. The

design was patterned on a silicon wafer by contact photo-

lithography with SU-8 photoresist (Micro Chem, MA)

through the transparency mask and the SU-8 pattern yields

*100 lm thickness. The PDMS replicas were then fabri-

cated by molding PDMS (Sylgard 184 silicon elastomer,

Dow Corning, MI, USA) against the master. Two 4 mm

diameter medium reservoirs/electrode wells at the two ends

of a 350 lm (W) 9 1 cm (L) channel were punched out

using sharpened needles (Fig. S1). In addition, a 1-mm

diameter hole for the fluidic inlet was punched out in the

PDMS device for infusing medium (Fig. S1). The PDMS

replica was then plasma bonded to a glass slide using a

plasma cleaner (Harrick Plasma, Ithaca, NY, USA). Poly-

ethylene tubing (PE-20, Becton–Dickinson, MD, USA)

was inserted into the fluidic inlet to connect the microflu-

idic device to a syringe pump (KD Scientific, Holliston,

MA, USA) with a 250-lL syringe containing medium for

fluidic infusion. The medium was continuously infused into

the device by the syringe pump through the inlet at the flow

rate of 0.2 lL/min (i.e., linear speed of 5.7 mm/min).

External platinum electrodes (SPPL-010, Omega Engi-

neering, Inc.) that were attached to conducting wires were

inserted into two medium reservoirs and the wires were

connected to a DC power supply to apply electric field to

the microchannel. The microfluidic channel was coated

with Collagen Type IV for 30 min at room temperature

followed by blocking with 2 % BSA for 1 h at 37 �C to

provide a substrate for cell adhesion and migration. A new

microfluidic device was used for each experiment and the

time-lapse imaging of cell migration follows the same

method as in the well plate-based experiments.

The actual potential difference across the well-plate or

microfluidic channel is generally lower than the total

voltage applied from the power supply due to the electrical

potential drop at the electrodes. For simplicity, in this

article, we calculated the dcEF in our experiments using

the total applied voltage from the power supply and pro-

vide a calibration table in the Supplementary Information

(Table S1).

Analysis of Cell Migration Data

Movement of individual cells in the well-plate or micro-

fluidic device was tracked using MetaMorph (Molecular

Devices Inc., Offline Premier Version, v7.7.3). The images

were calibrated to distance and only the cells that migrated

within the field of view were selected for tracking. The

tracking data were exported to Excel and MATLAB for

analysis. Following previously established analysis meth-

ods [43, 44] (Fig. S1), the movement of cells was quanti-

tatively evaluated by (a) the percentage of cells that

migrated toward the anode of the electric field; (b) elec-

trotactic index (EI), which is the ratio of the displacement

of cells toward the anode of the electric field to the total

migration distance, presented as the average value ±

standard error of the mean (SEM) (Fig. S1); (c) the speed

of cells, which is the total migration distance divided by the

migration time, presented as the average value ± SEM

(Fig. S1); (d) statistical analysis of migration angles per-

formed using MATLAB to examine the directionality of

cell movement (Fig. S1). Specifically, migration angles

(calculated from x–y coordinates at the beginning and the

end of the cell tracks) were summarized in a direction plot,

which is a rose diagram showing the distribution of angles

grouped in defined intervals (18�), with the radius of each

wedge indicating the cell number; (e) to quantify the

motility of cells along the direction of electric field, the

mean square displacement (MSD) of cells in the x-direction

(i.e., the dcEF direction) was calculated as a function of

Cell Biochem Biophys

123

Page 4: DC Electric Fields Direct Breast Cancer Cell Migration, Induce EGFR Polarization, and Increase the Intracellular Level of Calcium Ions

time,\x2(t)[. The parameters between different conditions

were compared by the 2-sample t test. 38–142 cells were

analyzed for each experiment. Multiple independent

experiments were performed for each condition with sim-

ilar results and data from representative experiments were

shown in the figures.

Imaging of EGFR Staining

After electrotaxis or control experiments, cells on the

coverslip in either the well-plate or the microfluidic device

were washed quickly with staining buffer (PBS with 0.5 %

BSA), then immediately fixed with 4 % formaldehyde. For

better staining and imaging results of cells in microfluidic

devices, PDMS was reversibly bonded to the coverslip

without plasma treatment. The PDMS top was removed

from the coverslip after cell fixing and then the cell-con-

taining coverslips were used for staining and imaging in

this set of experiment. Alexa 488 tagged monoclonal anti-

human EGFR antibody (Santa Cruz Biotechnology, Inc.,

CA, USA) was diluted in the staining buffer at 1:50 ratio

and was then used to stain cells at 4 �C for 30 min. Cells

were washed three times with the staining buffer and fur-

ther treated by DAPI for nucleus staining followed by

additional washing steps before confocal imaging. The

coverslips were mounted in mountant permafluor (Lab

Vision Corporation, CA, USA) and viewed with a confocal

microscope (Nikon Ti-U microscope equipped with a

C1-Plus confocal system). The image acquisition was

controlled by the EZ-C1 software (ver3.80).

[Ca2?]i Measurement

[Ca2?]i in MDA-MB-231 cells was measured by flow cyto-

metric analysis (FACS analysis) and confocal microscopy.

Cells were loaded with Fluo-4 AM (Molecular Probes, Inc.,

Eugene, Oregon, USA) in IMDM medium (Hyclone Labo-

ratories, Inc., Utah, USA) with 1 % FBS for 30–45 min in a

37 �C incubator in dark. After Fluo-4 loading, the cells were

washed twice with the same medium and recovered in a

37 �C incubator in dark for 30 min before the calcium

experiments. For the FACS experiment, cells were stimu-

lated in the well plate by dcEF of different strength over

defined period of time (as detailed in the ‘‘Results’’ section)

or EGF (recombinant human EGF, Sigma-Aldrich Co. LLC,

MO, USA) of different concentrations (as detailed in the

‘‘Results’’ section; in this case, cells were mixed with EGF

immediately followed by FACS monitoring), and then the

cells were transferred to a FACS tube for FACS analysis

using a flow cytometer (BD FACSCalibur, Becton–Dickin-

son). The Fluo-4 signal was monitored by FACS at 3 s time

resolution. The FACS data was further analyzed using Flow

Jo (Tree Star, Inc., OR). For the confocal experiment, cells

were stimulated in the well-plate or the microfluidic device

by dcEF or EGF and Fluo-4 intensity was monitored in real-

time by time-lapse confocal microscopy at 6 frames/min.

Fig. 1 Electrotaxis of MDA-

MB-231 cells in well plates.

a Angular histogram shows

random cell migration in the

control condition without dcEF.

b Angular histogram shows cell

migration toward the anode of

the applied 1.5 V/cm electric

filed. c Comparison of

electrotactic index (EI),

percentage of electrotaxing

cells, and the speed of cells in

the absence or in the presence of

dcEF. d Comparison of mean

square displacement of cells

along the x-direction as a

function of time \x(t)2[ in the

control experiment or with dcEF

application. The error barsrepresent the standard error of

the mean (SEM). The p values

for each comparison from

2-sample t test are shown.

** p \ 0.01

Cell Biochem Biophys

123

Page 5: DC Electric Fields Direct Breast Cancer Cell Migration, Induce EGFR Polarization, and Increase the Intracellular Level of Calcium Ions

Multiple independent experiments were performed for each

condition with similar results.

Results

Anodal Electrotaxis of MDA-MB-231 Cells

In a previous study, MDA-MB-231 cells were shown to

migrate toward the anode of dcEF using a conventional

dish-based electrotaxis assay [18]. Here, we want to con-

firm this result in our cell migration system. In the first set

of experiment, we applied a 1.5-V/cm of dcEF to MDA-

MB-231 cells seeded in a well plate and measured cell

migration by time-lapse microscopy. As shown by the

migration angle histograms (Fig. 1a, b), cells showed clear

migration toward the anode of the applied dcEF. By con-

trast, cells migrated randomly in the absence of dcEF. This

result was also supported by the higher percentage of

electrotaxing cells to the anode of dcEF and the higher EI

of cells in dcEF compared with it in the absence of dcEF

(Fig. 1c). Furthermore, cell speed (Fig. 1c) and the MSD

(\x2(t)[) analysis showed higher total cell motility on the

2D substrate (i.e., speed) and higher motility along the

dcEF direction (i.e., increase of MSD over time) respec-

tively for cells in dcEF than it in the absence of dcEF

(Fig. 1d). The main limitation of the well-plate assay is the

lack of control of electric field. In particular, the current

set-up of this assay does not allow higher dcEF application,

which produces bubbles and causes significant pH change

due to electrolysis.

Thus, we next performed the electrotaxis experiments

using a microfluidic device. Compared to conventional dish-

based electrotaxis assays, microfluidics devices have

advantages in control of dcEF application and cell migration

environments [45]. This device is in principle similar to our

previously developed microfluidic device for studying

electrotaxis of T cells [41] with the addition of a fluidic inlet

for continuous perfusion of medium into the channel (Fig.

S1). This new flow feature of the device provides continuous

supply of fresh medium and removes waste or other cell

products in the channel. In addition, the medium flow is

configured along the same direction of the dcEF such that the

true anodally electrotaxing cells will need to migrate against

the flow. No bubbles are produced in the channel even at

higher dcEF. It is worth pointing out that there is a significant

change of pH in the two reservoirs, where electrodes were

inserted, before and after applying dcEF. However, the pH in

the channel is not expected to change because fresh medium

from syringe pump is continuously infused into the channel

throughout the experiment (Fig. S1).

Thus, this microfluidic system allowed us to measure

electrotaxis of MDA-MB-231 cells in better controlled dcEF

with higher magnitude comparing to the well plate. In the

Fig. 2 Electrotaxis of MDA-

MB-231 cells in microfluidic

devices. a Angular histogram

shows random cell migration

with a bias along the medium

flow direction in the absence of

dcEF. b Angular histogram

shows cell migration toward the

anode of 5 V/cm dcEF.

c Comparison of electrotactic

index (EI) and speed of cells in

the absence or in the presence of

dcEF. d Comparison of mean

square displacement of cells

along the x-direction as a

function of time \x(t)2[ in the

control experiment or with dcEF

application. The error barsrepresent the standard error of

the mean (SEM). The p values

for each comparison from

2-sample t test are shown.

** p \ 0.01, and * p \ 0.05

Cell Biochem Biophys

123

Page 6: DC Electric Fields Direct Breast Cancer Cell Migration, Induce EGFR Polarization, and Increase the Intracellular Level of Calcium Ions

control experiment without dcEF, cells migrated randomly

with a detectable bias along the medium flow direction as

expected (Fig. 2a, c). By contrast, cells overcame the med-

ium flow and migrated toward the anode of the applied dcEF

(Fig. 2b–d). In this fluidic system, the optimal cell speed is in

the 4 V/cm dcEF (i.e., 33.12 lm/h). This speed is compa-

rable to the previously reported migration speed of MDA-

MB-231 cells in EGF gradients [42]. MSD analysis more

clearly showed the higher motility of cells along the

x-direction in the dcEF experiments comparing to it in the

control experiment (Fig. 2d).

Taking together, the results of electrotaxis experiments

in both the well-plate and microfluidic devices confirmed

the anodal electrotaxis of MDA-MB-231 cells over the

similar range of dcEF as in the previous study [18], pro-

viding the quantitative electrotactic migration characteris-

tics of MDA-MB-231 cells for further electrotactic

signaling-related analysis in this study.

EGFR Polarization in MDA-MB-231 Cells Toward

the Anode of the dcEF

Based on the demonstrated anodal electrotaxis of MDA-

MB-231 cells, we want to next examine the relevant cel-

lular biophysics events in MDA-MB-231 cells upon dcEF

stimulation to identify possible electrotactic signaling. One

candidate event is dcEF-induced polarization of EGFR.

EGFR is expressed in various electrotaxing cells and sev-

eral previous studies have demonstrated polarization of cell

surface EGFR that coincides with the direction of elec-

trotactic cell migration [25–27]. Therefore, in MDA-MB-

231 cells, we expect EGFR will polarize toward the anode-

facing side of the cells in dcEF.

To test this hypothesis, we employed immunofluores-

cence staining against cell surface EGFR to measure EGFR

distribution after electrotaxis experiments either in the

well-plate or microfluidic device. Our results showed that

before dcEF was applied cells typically form round shape

morphology or orient randomly (Fig. 3a). Upon exposure

to 4 V/cm dcEF, many cells formed clear leading edges

and among them most cells oriented toward the anode of

the dcEF (Fig. 3b). Immunofluorescence staining by anti-

EGFR antibody showed that EGFR polarized toward the

anode-facing side of the electrotaxing cells (Fig. 3c), i.e.,

our analysis revealed the detectable more EGFR distribu-

tion on the anode-facing side of more than 50 % of total

cells in confocal images and the remaining cells showed no

clear EGFR polarization at the time of fixation. Quantita-

tive analysis provides complementary indication of EGFR

polarization toward the anode-facing side of the cell

(Supplementary Information; Fig. S2). In the control

experiment without dcEF, EGFR uniformly distributed in

all cells.

dcEF Induce [Ca2?]i Elevation in MDA-MB-231 Cells

Motivated by previous studies showing calcium signaling

in electrotaxing cells [38–40], we in this study analyzed the

dcEF-induced intracellular calcium ion ([Ca2?]i) to test the

possible involvement of calcium signaling in breast cancer

cell electrotaxis.

MDA-MB-231 cells were loaded with Fluo-4 and

stimulated in a well plate by dcEF for 10 min, and Fluo-4

intensity was measured by FACS (before and after 10 min

of dcEF stimulation) or by confocal microscopy (continu-

ous monitoring) to indicate the dynamic changes of

[Ca2?]i. In the FACS analysis, 1.5 V/cm dcEF induced a

very small but more sustained increase of [Ca2?]i (Fig. 4e).

By contrast, the higher magnitude of dcEF (3 V/cm)

induced a clear increase of [Ca2?]i, which decreased over

time and stabilized at a level higher than the baseline

(Fig. 4e). Consistent with the previous research that

showed a transient calcium spike lasting 2 min in MDA-

MB-231 cells in response to EGF stimulation [37], treating

cells with a range of EGF doses in our study caused a

Fig. 3 dcEF-induced morphological change and EGFR polarization

in MDA-MB-231 cells in microfluidic device. a Phase contrast

images of three representative cells before the dcEF exposure.

b Phase contrast images of the same three cells in a after 1.5 h

exposure to a 4-V/cm dcEF. The white arrows point to the leading

edge of the migrating cells. c Confocal images of the same three cells

by anti-EGFR antibody (green) staining and nucleus staining (blue)

showing polarization of EGFR to the anode-facing side of the cell

(Color figure online)

Cell Biochem Biophys

123

Page 7: DC Electric Fields Direct Breast Cancer Cell Migration, Induce EGFR Polarization, and Increase the Intracellular Level of Calcium Ions

transient increase of [Ca2?]i in cells and the duration of

[Ca2?]i spikes also generally lasted *2 min before com-

pletely adapt to the baseline level (Fig. 4f). Unlike the

FACS analysis, which measures [Ca2?]i without concurrent

dcEF stimulation, time-lapse confocal microscopy allows

us to further monitor [Ca2?]i in cells at the population level

(i.e., measuring Fluo-4 intensity of the entire image) in

real-time over the time course of dcEF stimulation. Our

results confirmed the increase of [Ca2?]i by dcEF stimu-

lation (Fig. 4a–d). Consistent with the FACS analysis

results, this increase was only detectable for 3 V/cm but

not 1.5 V/cm dcEF stimulation, and it occurred toward the

end of the 10 min stimulation and lasted even after the

dcEF was turned off. For both the FACS and confocal

measurements, Fluo-4 intensity (F) was normalized to the

initial intensity (F0), and this ratio (F/F0) was used to

indicate the relative [Ca2?]i in cells. Notably, some bubbles

were typically produced around the electrodes in the well

plate in 3 V/cm or higher magnitude of dcEF, which

complicates the interpretation of the observed increase of

[Ca2?]i. Although trypan blue staining showed that 95 %

cells were viable in 3 V/cm of dcEF, we want to further

test the calcium response under better controlled experi-

mental conditions.

To further analyze [Ca2?]i dynamics in a better con-

trolled cellular environment, we used microfluidic devices

as an experimental platform to monitor [Ca2?]i in MDA-

MB-231 cells at the single cell level in dcEF or by EGF

stimulation using time-lapse confocal microscopy. In the

control experiments, Fluo-4 intensity decreased over time

due to photobleaching by confocal scanning and thus set

the baseline curve for comparison. In 3 V/cm dcEF,

[Ca2?]i exhibited a small but long lasting increase that is

more clear a few minutes after the dcEF was applied

(Fig. 5a). In a higher dcEF (4 V/cm), [Ca2?]i significantly

increased (Fig. 5a, b). Similar to the results in well plates,

the increase of [Ca2?]i maintained after dcEF was turned

off, particularly for 4 V/cm. In addition, consistent with the

EGF stimulation results in FACS (Fig. 4f), [Ca2?]i tran-

siently increased upon EGF stimulation in microfluidic

devices (Fig. 5a, c), again suggesting the distinct charac-

teristics of calcium responses to dcEF and EGF stimula-

tions. In addition to analyzing the average [Ca2?]i to

compare the mean difference among conditions, we further

looked at [Ca2?]i in individual cells. dcEF or EGF stimu-

lations-induced [Ca2?]i oscillations in many individual

cells, suggesting similar [Ca2?]i responses as observed in

other electrotaxing cells [40].

Fig. 4 [Ca2?]i elevation in MDA-MB-231 cells in well plates

induced by dcEF or EGF stimulation. a–c Time-lapse confocal

images of cells. 3 V/cm dcEF was applied between the 5th and the

15th min. d Analysis of Fluo-4 intensity of the time-lapse confocal

images. F/F0 is the relative Fluo-4 intensity normalized to the initial

baseline level. e Fluo-4 intensity measured by FACS. Fluo-4 intensity

was measured by FACS before and after 10 min dcEF treatment in

the well plate, i.e., 0–1 min for the baseline before dcEF treatment;

1–5 min after 10 min dcEF treatment in the plot. f Fluo-4 intensity

measured by FACS in cells stimulated by EGF at the indicated

concentrations from the 2nd min

Cell Biochem Biophys

123

Page 8: DC Electric Fields Direct Breast Cancer Cell Migration, Induce EGFR Polarization, and Increase the Intracellular Level of Calcium Ions

It is worth pointing out the different effective dcEF for

triggering significant [Ca2?]i increase in different experi-

mental systems in this study. dcEF below 4 V/cm in

microfluidic device or below 3 V/cm in well plates or

FACS could not elicit significant calcium responses in

cells, likely resulting from the differences of experimental

systems (microfluidic device vs. well plate or FACS) and

analysis method (single cell vs. cell population) that set

different thresholds. This general threshold phenomenon

further suggests the possible regulations of voltage-gated

calcium channels for dcEF-induced calcium responses in

MDA-MB-231 cells. On the other hand, results in all three

experimental systems consistently showed calcium eleva-

tion in MDA-MB-231 cells upon dcEF stimulations, sug-

gesting the possible involvement of calcium signaling in

breast cancer cell electrotaxis.

Conclusion and Discussion

In this study, we quantitatively characterized the migration

of MDA-MB-231 cells in dcEF and tested relevant signal-

ing events with the focus on EGFR polarization and intra-

cellular calcium ion dynamics. Our results confirmed the

anodal electrotaxis of MDA-MB-231 cells. As discussed in

the previous study [18], an estimated dcEF of 6 V/cm

across the epithelial layer exists with lumen side as the

anode resulting from the transepithelial electrical potential

(TEP) difference in breast epithelium. Such physiological

dcEF is comparable to the effective dcEF strength in our

experiments using microfluidic devices. On the other hand,

electrotaxis of MDA-MB-231 cells was observed in the

well-plate assay under much lower dcEF, which is close to

the lower limit of effective dcEF reported in the previous

study using a dish-based electrotaxis assay [18].

Furthermore, our results demonstrated the dcEF-induced

anodal polarization of EGFR and intracellular calcium ion

increase. These results provide further experimental evi-

dence of breast cancer cell electrotaxis with quantitative

details and suggest the possible cellular mechanisms that

involve EGFR and calcium signaling. In particular, previ-

ous study has already shown that EGFR signaling is

required for breast cancer cell electrotaxis [18]. Our results

further suggest the possible involvement of cell surface

EGFR polarization for enabling electric field sensing and

migration of MDA-MB-231 cells. Cathodal EGFR polari-

zation has been reported for several cathodally electro-

taxing cell types such as bovine corneal epithelial cells [12,

25, 26], human keratinocytes [27], and lung cancer cells

[17]. Our results showed that EGFR can also polarize to the

anode-facing side of the cell for anodally electrotaxing

MDA-MB-231 cells, and therefore the same receptor can

be differentially polarized in different cell types depending

on the direction of electrotactic migration.

Despite the different threshold dcEF for triggering cal-

cium response and the detailed [Ca2?]i dynamics in

Fig. 5 [Ca2?]i responses in

MDA-MB-231 cells in

microfluidic devices induced by

electric field or EGF. a [Ca2?]i

over time without stimulation,

or stimulated by dcEF or EGF

(100 ng/mL). The data (F/F0) is

represented by the average

Fluo-4 intensity (F) of all

individual cells analyzed and is

normalized to the initial Fluo-4

intensity (F0). dcEF was applied

to the cells till the 10th min.

EGF-containing medium was

infused to the cells in the

microfluidic channel from the

5th min. b–d The same analysis

was shown for individual cells

for the dcEF (b), EGF (c), and

control experiments (d)

Cell Biochem Biophys

123

Page 9: DC Electric Fields Direct Breast Cancer Cell Migration, Induce EGFR Polarization, and Increase the Intracellular Level of Calcium Ions

different experimental systems used in this study, our

results showed clear elevation of [Ca2?]i in MDA-MB-231

cells upon effective dcEF stimulation. On the other hand,

the results of electrotaxis and calcium elevation derived

from the well-plate assay are not always consistent. We

believe it is due to the limitation of the well-plate assay in

control of dcEF application and cell migration environ-

ments. Although it is a simple assay to test if there is any

response at all to dcEF, the calcium elevation in well-plate

assay may be caused by other factors such as pH change of

the medium or cell death. Therefore, microfluidic devices,

which can better control dcEF application and cell migra-

tion environments, were used to further examine calcium

response of cells to dcEF and EGF stimulations.

Previous studies showed that dcEF induce [Ca2?]i influx

of Dictyostelium cells during electrotaxis [40]. In addition,

embryo fibroblasts stimulated by dcEF showed an increase

of [Ca2?]i. Both studies demonstrated that the cathodal

electrotaxis is calcium-dependent [38, 40]. Our results

showed consistent calcium responses to dcEF for MDA-

MB-231 cells. Importantly, the [Ca2?]i increase induced by

dcEF in MDA-MB-231 cells is more sustained in clear

comparison with the EGF stimulation-induced transient

[Ca2?]i elevation. Previous studies showed that EGF pre-

treatment of cells promotes calcium oscillations in

response to glutamate, ATP, or thimerosal (which directly

activates the inositol-1,4,5-triphosphate receptor) in astro-

cytes [46, 47] and EGF can directly induce transient

[Ca2?]i increase in MDA-MB-468 cells [48]. In the elec-

trotactic Dictyostelium cells, cAMP induces transient cal-

cium flux whereas the [Ca2?]i increase to dcEF is more

sustained [40]. Similar transient calcium response was also

recently reported for MDA-MB-231 cells in direct response

to EGF stimulation [37]. Thus, the characteristics of cal-

cium response to dcEF in electrotaxing cells are different

from it induced by chemoattractants or growth factors.

While some possible factors that may cause the different

calcium response to dcEF and chemical stimulations were

previously discussed such as the delayed calcium spikes

upon dcEF stimulation and passive calcium influx [40], the

underlying mechanisms remain to be determined.

Metastasis is the major cause of death for cancer patients,

which requires the migratory ability of cancer cells. Exis-

tence of physiological electric field between the tumor and

normal tissues during metastasis of breast cancers and the

potential role of such electric field in mediating the invasion

of electrotaxing breast cancer cells into the surrounding

tissues for metastasis have been suggested [16, 18, 49].

Thus, understanding the mechanisms for electrotactic

migration of breast cancer cells will have important impact

on more effectively managing breast cancer metastasis.

Together with other studies [18], our results suggest the

possible involvement of EGFR and calcium signaling in

breast cancer cell electrotaxis. An interesting aspect is the

experimental evidence for EGFR-mediated chemotactic

migration of breast cancer cells to EGF gradients [42] and

the requirement of EGFR signaling for breast cancer cell

electrotaxis [18] as reported in previous studies and the

anodal polarization of EGFR induced by dcEF in MDA-

MB-231 cells as reported in this study that collectively

suggests the comprehensive roles played by EGFR for

mediating breast cancer cell chemotaxis and electrotaxis

during metastasis. However, the EGFR-mediated signaling

mechanisms for chemotaxis and electrotaxis may be char-

acteristically different as evidenced by the different calcium

responses to EGF and dcEF in MDA-MB-231 cells. Further

testing MDA-MB-231 cells in different combinations of

dcEF and EGF fields will provide interesting insight into the

competing or collaborative guidance by chemical and

electrical cues for breast cancer cell migration. In this

regard, our recent study reported a novel microfluidic

device that can better control superimposed chemical and

electric fields and this device was used for studying T cell

migration in co-existing chemokine gradients and dcEF

[50]. Similar approach can be applied to MDA-MB-231 cell

migration and clarify the relative potency of EGF and dcEF

in attracting cells with relevance to developing potential

clinical applications by electrically manipulating cancer

cell trafficking in tissues. More generally, microfluidic

devices have been increasingly developed and applied to

electrotaxis studies [44, 45, 51, 52], providing useful

experimental tools for probing this important cell migration

process. Our study for the first time demonstrated the

effective use of microfluidic device for studying MDA-MB-

231 cell electrotaxis in 2D. However, the process and

mechanism of cell migration on 2D substrates can be lar-

gely different from it in 3D ECM. Although the comparison

of cell migration in 2D and 3D ECM depends on the type of

cells and ECM, cell migration generally replies more on

focal adhesion in 2D than in 3D, and the stiffness of 3D

ECM can significantly affects the morphology and direc-

tional migration of cells [53]. In addition, dcEF is inevitably

more complex when applied to 3D ECM comparing to it in

2D systems. Indeed, a recent study has shown differences of

lung cancer cell electrotaxis between 2D and 3D systems

[54]. In addition, the migratory behaviors of cells are

expected to be different when cells move collectively at

high density [55]. This is clearly the case in epithelial cell

electrotaxis that collective migration of epithelial mono-

layer is more efficient than isolated cells or smaller cell

clusters [56] and thus collective electrotaxis of tumor cells

in 3D tissues can also be sensitive to a lower effective dcEF

as compared with in vitro experiments. Therefore, we are

interested in furthering our studies in the future to test breast

cancer cell electrotaxis in 3D scaffold at the single cell level

or as cell groups.

Cell Biochem Biophys

123

Page 10: DC Electric Fields Direct Breast Cancer Cell Migration, Induce EGFR Polarization, and Increase the Intracellular Level of Calcium Ions

Acknowledgments This Research is supported by Grants from the

Natural Sciences and Engineering Research Council of Canada

(NSERC), the Canada Foundation for Innovation (CFI), the Manitoba

Health Research Council (MHRC), and the University of Manitoba.

We thank The Nano Systems Fabrication Laboratory (NSFL) at the

University of Manitoba, and the Manitoba Centre for Proteomics and

Systems Biology for research support. We thank Saravanan Nan-

dagopal for helping collect chemical reagents, Jing Li and Jiandong

Wu for helping with microfluidic device preparation. D.W. thanks

MHRC for a postdoctoral fellowship.

Conflict of interest The authors declare no conflict of interest.

References

1. Matsubayashi, Y., Ebisuya, M., Honjoh, S., & Nishida, E. (2004).

ERK activation propagates in epithelial cell sheets and regulates

their migration during wound healing. Current Biology, 14,

731–735.

2. McDougall, S., Dallon, J., Sherratt, J., & Maini, P. (2006).

Fibroblast migration and collagen deposition during dermal

wound healing: Mathematical modelling and clinical implica-

tions. Philosophical Transactions Series A, Mathematical,Physical, and Engineering Sciences, 364, 1385–1405.

3. Ayala, R., Shu, T., & Tsai, L. H. (2007). Trekking across the

brain: The journey of neuronal migration. Cell, 128, 29–43.

4. Hatten, M. E. (2002). New directions in neuronal migration.

Science, 297, 1660–1663.

5. Keller, R. (2005). Cell migration during gastrulation. CurrentOpinion in Cell Biology, 17, 533–541.

6. Friedl, P., & Wolf, K. (2003). Tumour-cell invasion and migra-

tion: Diversity and escape mechanisms. Nature Reviews Cancer,3, 362–374.

7. Yamaguchi, H., Wyckoff, J., & Condeelis, J. (2005). Cell

migration in tumors. Current Opinion in Cell Biology, 17,

559–564.

8. Kunkel, E. J., & Butcher, E. C. (2003). Plasma-cell homing.

Nature Reviews Immunology, 3, 822–829.

9. Lin, F., Nguyen, C., Wang, S., Saadi, W., Gross, S., & Jeon, N.

(2005). Neutrophil migration in opposing chemoattractant gra-

dients using microfluidic chemotaxis devices. Annals of Bio-medical Engineering, 33, 475–482.

10. Cooper, M., & Keller, R. (1984). Perpendicular orientation and

directional migration of amphibian neural crest cells in DC

electrical fields. Proceedings of the National Academy of Sci-ences USA, 81, 160–164.

11. Tai, G., Reid, B., Cao, L., & Zhao, M. (2009). Electrotaxis and

wound healing: Experimental methods to study electric fields as a

directional signal for cell migration. Methods in MolecularBiology, 571, 77–97.

12. McCaig, C., Rajnicek, A., Song, B., & Zhao, M. (2005). Con-

trolling cell behavior electrically: Current views and future

potential. Physiological Reviews, 85, 943–978.

13. Mycielska, M., & Djamgoz, M. (2004). Cellular mechanisms of

direct-current electric field effects: Galvanotaxis and metastatic

disease. Journal of Cell Science, 117, 1631–1639.

14. Robinson, K., & Messerli, M. (2003). Left/right, up/down: The

role of endogenous electrical fields as directional signals in

development, repair and invasion. Bioessays, 25, 759–766.

15. Nuccitelli, R. (1988). Physiological electric fields can influence

cell motility, growth and polarity. Advanced Cell Biology, 2, 21.

16. Djamgoz, M. B. A., Mycielska, M., Madeja, Z., Fraser, S., &

Korohoda, W. (2001). Directional movement of rat prostate

cancer cells in direct-current electric field: Involvement of volt-

agegated Na? channel activity. Journal of Cell Science, 114,

2697–2705.

17. Yan, X., Han, J., Zhang, Z., Wang, J., Cheng, Q., Gao, K., et al.

(2009). Lung cancer A549 cells migrate directionally in DC

electric fields with polarized and activated EGFRs. Bioelectro-magnetics, 30, 29–35.

18. Pu, J., McCaig, C. D., Cao, L., Zhao, Z., Segall, J. E., & Zhao, M.

(2007). EGF receptor signalling is essential for electric-field-

directed migration of breast cancer cells. Journal of Cell Science,120, 3395–3403.

19. Price, J. E. (1996). Metastasis from human breast cancer cell

lines. Breast Cancer Research and Treatment, 39, 93–102.

20. Christofori, G. (2003). Changing neighbours, changing behav-

iour: Cell adhesion molecule-mediated signalling during tumour

progression. EMBO Journal, 22, 2318–2323.

21. Segall, J. E., Tyerech, S., Boselli, L., Masseling, S., Helft, J.,

Chan, A., et al. (1996). EGF stimulates lamellipod extension in

metastatic mammary adenocarcinoma cells by an actin-dependent

mechanism. Clinical & Experimental Metastasis, 14, 61–72.

22. Levine, M. D., Liotta, L. A., & Stracke, M. L. (1995). Stimulationand regulation of tumor cell motility in invasion and metastasis.

EXS, 74, 157–179.

23. Xue, C., Wyckoff, J., Liang, F., Sidani, M., Violini, S., Tsai, K.

L., et al. (2006). Epidermal growth factor receptor overexpression

results in increased tumor cell motility in vivo coordinately with

enhanced intravasation and metastasis. Cancer Research, 66,

192–197.

24. Hirsch, D. S., Shen, Y., & Wu, W. J. (2006). Growth and motility

inhibition of breast cancer cells by epidermal growth factor

receptor degradation is correlated with inactivation of Cdc42.

Cancer Research, 66, 3523–3530.

25. Zhao, M., Dick, A., Forrester, J., & McCaig, C. (1999). Electric

field-directed cell motility involves up-regulated expression and

asymmetric redistribution of the epidermal growth factor recep-

tors and is enhanced by fibronectin and laminin. MolecularBiology of the Cell, 10, 1259–1276.

26. Zhao, M., Pu, J., Forrester, J. V., & McCaig, C. D. (2002).

Membrane lipids, EGF receptors, and intracellular signals colo-

calize and are polarized in epithelial cells moving directionally in

a physiological electric field. FASEB J, 16, 857–859.

27. Fang, K. S., Ionides, E., Oster, G., Nuccitelli, R., & Isseroff, R. R.

(1999). Epidermal growth factor receptor relocalization and

kinase activity are necessary for directional migration of kerati-

nocytes in DC electric fields. Journal of Cell Science, 112(Pt 12),

1967–1978.

28. Poo, M., & Robinson, K. (1977). Electrophoresis of concanavalin

A receptors along embryonic muscle cell membrane. Nature, 265,

602–605.

29. Brown, M. J., & Loew, L. M. (1994). Electric field-directed

fibroblast locomotion involves cell surface molecular reorgani-

zation and is calcium independent. Journal of Cell Biology, 127,

117–128.

30. Wu, D., & Lin, F. (2011). A receptor-electromigration-based

model for cellular electrotactic sensing and migration. Bio-chemical and Biophysical Research Communications, 411,

695–701.

31. Yang, S., & Huang, X. Y. (2005). Ca2? influx through L-type

Ca2? channels controls the trailing tail contraction in growth

factor-induced fibroblast cell migration. Journal of BiologicalChemistry, 280, 27130–27137.

32. Agle, K. A., Vongsa, R. A., & Dwinell, M. B. (2010). Calcium

mobilization triggered by the chemokine CXCL12 regulates

Cell Biochem Biophys

123

Page 11: DC Electric Fields Direct Breast Cancer Cell Migration, Induce EGFR Polarization, and Increase the Intracellular Level of Calcium Ions

migration in wounded intestinal epithelial monolayers. Journal ofBiological Chemistry, 285, 16066–16075.

33. Prevarskaya, N., Skryma, R., & Shuba, Y. (2011). Calcium in

tumour metastasis: New roles for known actors. Nature ReviewsCancer, 11, 609–618.

34. Brundage, R. A., Fogarty, K. E., Tuft, R. A., & Fay, F. S. (1991).

Calcium gradients underlying polarization and chemotaxis of

eosinophils. Science, 254, 703–706.

35. Hahn, K., DeBiasio, R., & Taylor, D. L. (1992). Patterns of

elevated free calcium and calmodulin activation in living cells.

Nature, 359, 736–738.

36. Davis, F. M., Kenny, P. A., Soo, E. T., van Denderen, B. J.,

Thompson, E. W., Cabot, P. J., et al. (2011). Remodeling of

purinergic receptor-mediated Ca2? signaling as a consequence of

EGF-induced epithelial–mesenchymal transition in breast cancer

cells. PLoS ONE, 6, e23464.

37. Raimondi, C., Chikh, A., Maffucci, T., & Falasca, M. (2012). A

novel regulatory mechanism links PLCc1 to PDK1. Journal ofCell Science, 125(Pt 13), 3153–3163.

38. Onuma, E., & Hui, S. (1985). A calcium requirement for electric

field-induced cell shape changes and preferential orientation. CellCalcium, 6, 281–292.

39. Hammerick, K. E., Longaker, M. T., & Prinz, F. B. (2010).

In vitro effects of direct current electric fields on adipose-derived

stromal cells. Biochemical and Biophysical Research Communi-cations, 397, 12–17.

40. Shanley, L., Walczysko, P., Bain, M., MacEwan, D., & Zhao, M.

(2006). Influx of extracellular Ca2? is necessary for electrotaxis

in Dictyostelium. Journal of Cell Science, 119, 4741–4748.

41. Li, J., Nandagopal, S., Wu, D., Romanuik, S. F., Paul, K.,

Thomson, D. J., et al. (2011). Activated T lymphocytes migrate

toward the cathode of DC electric fields in microfluidic devices.

Lab on a Chip, 11, 1298–1304.

42. Wang, S. J., Saadi, W., Lin, F., Minh-Canh Nguyen, C., & Li

Jeon, N. (2004). Differential effects of EGF gradient profiles on

MDA-MB-231 breast cancer cell chemotaxis. Experimental CellResearch, 300, 180–189.

43. Lin, F., & Butcher, E. (2006). T cell chemotaxis in a simple

microfluidic device. Lab on a Chip, 6, 1462–1469.

44. Lin, F., Baldessari, F., Gyenge, C. C., Sato, T., Chambers, R. D.,

Santiago, J. G., et al. (2008). Lymphocyte electrotaxis in vitro

and in vivo. The Journal of Immunology, 181, 2465–2471.

45. Li, J., & Lin, F. (2011). Microfluidic devices for studying che-

motaxis and electrotaxis. Trends in Cell Biology, 21, 489–497.

46. Morita, M., Higuchi, C., Moto, T., Kozuka, N., Susuki, J., Itofusa,

R., et al. (2003). Dual regulation of calcium oscillation in

astrocytes by growth factors and pro-inflammatory cytokines via

the mitogen-activated protein kinase cascade. Journal of Neuro-science, 23, 10944–10952.

47. Morita, M., & Kudo, Y. (2010). Growth factors upregulate

astrocyte [Ca2?]i oscillation by increasing SERCA2b expression.

Glia, 58, 1988–1995.

48. Mishra, S., & Hamburger, A. W. (1993). Role of intracellular

Ca2? in the epidermal growth factor induced inhibition of protein

tyrosine phosphatase activity in a breast cancer cell line. Bio-chemical and Biophysical Research Communications, 191,

1066–1072.

49. Faupel, M., Vanel, D., Barth, V., Davies, R., Fentiman, I. S.,

Holland, R., et al. (1997). Electropotential evaluation as a new

technique for diagnosing breast lesions. European Journal ofRadiology, 24, 33–38.

50. Li, J., Zhu, L., Zhang, M., & Lin, F. (2012). Microfluidic device

for studying cell migration in single or co-existing chemical

gradients and electric fields. Biomicrofluidics, 6, 24121–2412113.

51. Minc, N., & Chang, F. (2010). Electrical control of cell polari-

zation in the fission yeast Schizosaccharomyces pombe. CurrentBiology, 20, 710–716.

52. Rezai, P., Siddiqui, A., Selvaganapathy, P. R., & Gupta, B. P.

(2010). Electrotaxis of Caenorhabditis elegans in a microfluidic

environment. Lab on a Chip, 10, 220–226.

53. Even-Ram, S., & Yamada, K. M. (2005). Cell migration in 3D

matrix. Current Opinion in Cell Biology, 17, 524–532.

54. Sun, Y. S., Peng, S. W., Lin, K. H., & Cheng, J. Y. (2012).

Electrotaxis of lung cancer cells in ordered three-dimensional

scaffolds. Biomicrofluidics, 6, 14102–1410214.

55. Nabeshima, K., Inoue, T., Shimao, Y., Kataoka, H., & Koono, M.

(1999). Cohort migration of carcinoma cells: Differentiated

colorectal carcinoma cells move as coherent cell clusters or

sheets. Histology and Histopathology, 14, 1183–1197.

56. Li, L., Hartley, R., Reiss, B., Sun, Y., Pu, J., Wu, D., et al. (2012).

E-cadherin plays an essential role in collective directional

migration of large epithelial sheets. Cellular and Molecular LifeSciences, 69, 2779–2789.

Cell Biochem Biophys

123