You are on page 1of 10

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/47510909

Electrical cell-substrate impedance sensing as a non-invasive tool for cancer


cell study

Article in The Analyst · October 2010


DOI: 10.1039/c0an00560f · Source: PubMed

CITATIONS READS

171 775

5 authors, including:

Jongin Hong Mohana Marimuthu


Chung-Ang University Dhanalakshmi Srinivasan University
215 PUBLICATIONS 2,983 CITATIONS 40 PUBLICATIONS 674 CITATIONS

SEE PROFILE SEE PROFILE

Cheol Soo Choi Sanghyo Kim


Gachon university college of medicine, Korea Gachon University
168 PUBLICATIONS 10,223 CITATIONS 149 PUBLICATIONS 3,462 CITATIONS

SEE PROFILE SEE PROFILE

All content following this page was uploaded by Mohana Marimuthu on 15 February 2018.

The user has requested enhancement of the downloaded file.


View Online

MINIREVIEW www.rsc.org/analyst | Analyst

Electrical cell-substrate impedance sensing as a non-invasive tool for cancer


cell study
Jongin Hong,†*a Karthikeyan Kandasamy,b Mohana Marimuthu,b Cheol Soo Choic and Sanghyo Kim*b
Received 22nd July 2010, Accepted 20th September 2010
DOI: 10.1039/c0an00560f

Cell-substrate interactions are investigated in a number of studies for drug targets including
angiogenesis, arteriosclerosis, chronic inflammatory diseases and carcinogenesis. One characteristic of
malignant cancerous cells is their ability to invade tissue. Cell adhesion and cytoskeletal activity have
served as valuable indicators for understanding the cancer cell behaviours, such as proliferation,
Published on 20 October 2010 on http://pubs.rsc.org | doi:10.1039/C0AN00560F

migration and invasion. This review focuses on bio-impedance based measurement for monitoring the
behaviours in real time and without using labels. Electric cell-substrate impedance sensing (ECIS)
provides rich information about cell-substrate interactions, cell-cell communication and cell adhesion.
Downloaded by Imperial College London on 26 October 2010

High sensitivity of the ECIS method allows for observing events down to single-cell level and achieving
nanoscale resolution of cell-substrate distances. Recently, its miniaturization and integration with
fluorescent detection techniques have been highlighted as a new tool to deliver a high-content platform
for anticancer drug development.

Introduction year of 2020. Accordingly, it has been of significant importance


to uncover the roles of various genes, protein products and
Cancer is a disease in which abnormal cells vigorously divide growth factors in the abnormal behavior of cancer cells. In
without control, invade adjacent tissues and spread through the particular, vascular endothelial growth factors (VEGFs) play
body via bloodstreams or lymphatic vessels.1,2 Last decades a major role on inducing the proliferation, migration and
cancer has been a major public concern and become the leading survival of endothelial cells and leading to the formation of new
cause of natural death. For example, breast cancer in women and blood vessels that are vital for tumour growth and metastases.3
prostate cancer in men are common malignancies in the world. Recently, special attention has been paid to test safety and effi-
New cases over ten millions are diagnosed annually worldwide, ciency of new anticancer drug candidates4,5 and thus in vitro
and the number is expected to increase to twenty millions by the cytotoxicity testing with live cells is invaluable in the assessment
of their adverse effects and environmental threats. Importantly,
a
Department of Chemistry and Institute of Biomedical Engineering, label-free and non-invasive techniques have been highly on
Imperial College London, South Kensington Campus, London, SW7 demand since conventional cell-based assays for the analysis of
2AZ, United Kingdom. E-mail: hong.jongin@gmail.com
b
College of Bionanotechnology, Kyungwon University, San-65, Bokjeong-
cell proliferation and viability are labor intensive, involve
dong, Sujeong-gu, Seongnam-si, Gyeonggi-do, 461-701, Korea. E-mail: labeling steps and provide a snapshot of the experiment. A
samkim@kyungwon.ac.kr number of label-free technologies including electrical cell-
c
Korea Mouse Metabolic Phenotyping Centre, Lee Gil Ya Cancer and substrate impedance sensing (ECIS), quartz microbalance and
Diabetes Institute, Gachon University of Medicine and Science, 7-45,
Songdo-dong, Yeonsu-gu, Incheon, 406-840, Korea
optical lightmode spectroscopy, have emerged as a means of
† Present address: Department of Material Science & Engineering, monitoring cellular processes in real-time.6–9 Among them, ECIS
KAIST, Daejeon 305-701, Korea has been recognized as a powerful tool to investigate cellular

Jongin Hong is currently a research professor in the Department of Material


Science & Engineering at KAIST and also an academic visitor in the
Department of Chemistry at Imperial College London (ICL). Before his
current position, he has worked as a postdoctoral research associate at ICL
for 3 years (2007–2010). Sanghyo Kim is an assistant professor in the
College of Bionanotechnology at Kyungwon University. Karthikeyan Kan-
dasamy and Mohana Marimuthu are studying for a Master degree at
Kyungwon University under the supervision of Jongin Hong and Sanghyo
Kim. Cheol-Soo Choi is currently an associate professor at Gachon Univer-
sity of Medicine and Science. This group is mainly engaged with the devel-
opment of high-throughput cellular assays and drug discovery for cancer.

Sanghyo Kim (Left), Mohana Marimuthu, Jongin Hong and


Karthikeyan Kandasamy (Right)

This journal is ª The Royal Society of Chemistry 2010 Analyst


View Online

properties and physiological functions of cells after Giaever and considered as dielectric particles due to the insulating properties
Keese firstly invented it in 1984.10,11 To date, ECIS measurements of their membranes and thus, when they attach and spread on the
have been successfully established to monitor motion, attach- small electrode, interfere the current flow between the two elec-
ment and spreading of cultured cells, quantify effects of trodes. This leads to large and measurable changes in impedance
biochemical compounds including cytotoxicity and assess cell and the impedance increases until they are fully spread
migration in wound healing assays.11–21 (a confluent layer of the cells is established). After reducing the
Typically, an ECIS system consists of gold electrodes of area available for the current flow, the measured impedance
a small sensing electrode and a large counter electrode in the fluctuates with time. If the change in cell shape or motion or both
bottom of cell culture dishes, a lock-in amplifier with an internal occurs, this results in the corresponding change in the current
oscillator and a personal computer that controls the measure- flow between the ventral surface (i.e. the side closest to the
ment and stores the data. The system is based on measuring the ground) of the cells and the substratum (i.e. a solid surface over
impedance change with weak and non-invasive AC signals that which a cell moves or upon which a cell grows) and the resistance
can theoretically be modeled as a RC circuit. The oscillator that between the cells. The degree of the changes is determined by the
Published on 20 October 2010 on http://pubs.rsc.org | doi:10.1039/C0AN00560F

relays to switch between the different wells applies an AC signal number of cells seeded on the sensing electrode, cell-cell inter-
of 1 mA (usually in the frequency range from 1 to 40 kHz) action, cell morphological changes and cell motility. In addition,
through a series of a 1 MU resistor between the relatively small the impedance fluctuation provides information about cellular
microelectrode (103 cm2) and the larger counter electrode. The dynamics depending on different cell types. Accordingly, time-
Downloaded by Imperial College London on 26 October 2010

working electrode deposited on a cell culture dish should be 103 resolved impedance measurements provide real-time fingerprints
cm2 or even smaller since a resistance component from a bulk of the cells under a variety of culture conditions. To explain the
electrolyte (normally cell culture medium) dominates the resulting impedance, mathematical models have been developed
measured impedance when the larger electrode is used. The to calculate cell morphological parameters, such as the narrow
amplifier collects the in-phase and out-of-phase voltages, which spaces between cells (barrier resistance, Rb), the narrow spaces
are converted to time series of resistance and capacitance, across between the ventral surface of the cells and the electrode (h), and
the system. When cells adhere and spread on the electrode the cell membrane capacitance (Cm).22–25 Such parameters can be
surface, the impedance is altered because the cells restrict the determined by fitting the theoretical model to the experimental
current flow. Accordingly, this change can be utilized to analyze data. Recently, Lovelady et al. have demonstrated that the
dynamic information about cell shape and locomotion. The signatures of electrical noise in ECIS measurements can be useful
measured impedance is also susceptible of external conditions, to distinguish different cell types and their dynamics.26 There-
such as pH, temperature and an addition of biological and fore, the ECIS has been considered as a non-invasive tool to
chemical compounds. Different types of configurations including quantitatively study cell behaviours, such as cell spreading,
eight wells with one electrode (8W1E), eight wells with ten elec- attachment, proliferation, differentiation and migration, and
trodes (8W10E) and 96 wells with integrated electrodes (96WE) cytoxicity of compounds or drugs. ECIS measurements can also
are to monitor the cell behavior in real-time and without the use give information about electrochemical processes in the tissues
of labels. A schematic diagram of an ECIS system and its and be used to monitor their physiological changes against
working principle are shown in Fig. 1. Basically, the cells are environmental conditions. In addition, the utilization of

Fig. 1 (a) A schematic diagram of the ECIS measurement system and (b) current flows without cells (top) and with cells (bottom) from the surface of Au
sensing electrodes.

Analyst This journal is ª The Royal Society of Chemistry 2010


View Online

impedance as a cellular readout allows for the preclusion of


conventional labels. This can circumvent their negative impact
on cellular processes and place cells in their native and physio-
logically relevant status. Importantly, the ECIS system provides
kinetic data of initial cell attachment and prolonged steady-state
adhesion in real time and the process is fully automated with
conventional attachment assays. It also detects vertical motions
of the cells as they move on or off the working electrode with the
spatial resolution less than 1 nm and thus this technique is
a unique measurement of cell motility. In this mini-review, we
will evaluate ‘Electrical Cell-substrate Impedance Sensing
(ECIS)’ as a label-free and non-invasive tool for cancer biology.
Published on 20 October 2010 on http://pubs.rsc.org | doi:10.1039/C0AN00560F

Cell adhesion, migration and proliferation


When normal cells are grown up in a culture vessel (test tube,
bottle, flask or dish), they tend to divide until its surface is
Downloaded by Imperial College London on 26 October 2010

covered by a single layer of cells and then further cell division is


inhibited (called density-dependent inhibition). In contrast,
cancer cells tend to proliferate and gradually pile up on top of
Fig. 2 A typical result of the ECIS measurement during cell culture. At
another, forming multilayered aggregates. Another difference
time zero, human fibroblastic cells were inoculated in an ECIS well giving
between normal and cancer cells involves the requirement of a final cell density of 105 per cm2.16
anchorage. Most normal cells only grow well when they are
provided with an appropriate solid surface to which they can
adhere (called anchorage-dependent). If the anchorage is pre- marrow macrophage (BMDM) attachment on protein-coated
vented, normal cells cannot be divided and may commit suicide surfaces and spreading in response to activation agents, such as
by apoptosis (i.e. an orchestrated programme for cell death). interferon-g (IFN-g), lipopolysaccharide (LPS) and heat-killed
However, cancer cells are not subject to this normal safeguard. Listeria monocytogenes (HKLM).13 The ECIS measurements
Accordingly, the continual monitoring of cell attachment, detected that the degree of spreading induced by either LPS or
spreading and proliferation are crucial for understanding cellular HKLM (i.e. second signal agents related to cytolytic activity of
behaviors of both normal and cancer cells. Traditional methods macrophages and protein synthesis) was different from that
using an optical microscope are tiring and tedious because of induced by IFN-g (i.e. directly bind to its receptor and increase
many manipulations to maintain the cells in a healthy state while protein kinase C activity). Furthermore, this simple and accurate
they are on the microscope state. The quantification of the cell AC method can be useful for studying epithelial transport and
adhesion and spreading requires a sophisticated live imaging morphology. For example, Lo et al. investigated transepithelial
system and further data processing. When compared to the live impedance of a confluent layer of Madin-Darby canine kidney
cell imaging, the ECIS system offers distinguishing features. For (MDCK) cells and developed a new theoretical model applicable
example, this can quantitatively and continuously monitor cell to the epithelial cells because their apical cell membranes were
growth and dynamic behaviour over days with a temporal connected and sealed with tight junctions.17 Wegener et al.
resolution of minutes. The impedance data represent a statisti- examined the kinetics of MDCK cell spreading on the electrodes
cally relevant ensemble and thus no further data processing is coated with different proteins and the influence of divalent
needed. Moreover, this label-free and non-invasive approach can cations on its kinetics.20 They also quantified the inhibitory effect
be combined with conventional fluorescent assays to obtain an of soluble Arg-Gly-Asp-Ser (RGDS) peptides, which could
additional information about the cellular behaviours. compete with extracellular matrix proteins for the integrin
Giaver and Keese have firstly monitored the behavior of binding sites, on cell attachment. Similarly, the impedance
mammalian fibroblasts lines, such as WI-38 and WI-38/VA-13, measurements can monitor cellular transformation (i.e. the first
by measuring the impedance for long periods of time.11 Fig. 2 step in cancer development) and assess the difference between
shows a typical result of the in- and out-of-phase voltages as normal and transformed cells. For example, Park et al. quanti-
a function of time. The cells act as insulating particles and thus tatively examined the proliferation of NIH3T3 mouse fibroblasts
play a passive role by blocking the current. After they are fully and the morphological changes associated with cellular trans-
spread, the measured impedance fluctuates with time. These formation.27 D143V_CXCR2 overexpression caused the contin-
fluctuations result from the constant locomotion of the cells. uous activation of signaling pathways leading to the
They also observed that normal and transformed cells showed transformation and this was reflected in the great increase of the
different signatures of impedance fluctuations. Subsequently, impedance.
they reported how normal and transformed cells interact with Cell proliferation is an increase in cell populations as a result
four different protein layers (bovine serum albumin, gelatin, of cell development and cell division (reproduction). In cells that
bovine fetuin and human plasma fibronectin) coated on the are dividing, the nuclear DNA should be replicated and then
microelectrodes (i.e. fibronectin and gelatin were good distributed in a way that the two new cells receive a complete set
substrates).12 In addition, Kowolenko et al. monitored bone- of genetic information each other. To prepare and accomplish

This journal is ª The Royal Society of Chemistry 2010 Analyst


View Online

these tasks, an orderly sequence of events (G1 phase, S phase, G2 live Human cervical carcinoma cells (HeLa) by the ECIS
phase and M phase) takes place in a cell leading its division and measurements.30 To monitor the progression of the cell cycle,
duplication and the cell cycle is an essential mechanism in which HeLa cells were synchronized with double thymidine block and
all living things reproduce. Obviously, normal cells have mech- then the time-course of the cellular impedance was obtained over
anisms for regulating progression through the cell cycle, such as 5 days (Fig. 3). The impedance increased in G1 phase and S phase
restriction point, DNA replication checkpoint and spindle (i.e. Cells reattach and adhere to the electrode in G1 phase and
checkpoint. Contrary to normal cells, cancer cells continue to they grow and spread out in S phase), whilst it decreased in G2
grow and divide without halting at the restriction point of the cell phase and M phase (i.e. Cells round up and detach partially from
cycle even if under extremely adverse conditions. The most the electrode in G2 phase and divide into two daughter cells in M
common method for distinguish the different cell cycle stages is phase). Notably, time-course impedance curves mirror the real-
to use cell cycle markers with fluorescence.28,29 However, these time progression of the cell cycle in mammalian cells and the
markers can undergo dynamic changes and interactions shape of the curves is related to the synchronicity of the cell
throughout the cell cycle. In addition, the cell cycle may be population. We expect that this time-resolved cell cycle moni-
Published on 20 October 2010 on http://pubs.rsc.org | doi:10.1039/C0AN00560F

perturbed by the markers. Recently, Wang et al. successfully toring has great potentials in biomedical applications including
demonstrated the real-time dynamic analysis of the cell cycle in pharmacology, toxicology and high-throughput screening.
Downloaded by Imperial College London on 26 October 2010

ECIS-based cancer research


To understand the framework in regard to the ECIS-based
cancer research, we intend to outline ‘signal transduction’ and
‘cell migration’. The cell (i.e. the functional basic unit of our life)
receives and acts on signals from beyond the plasma membrane.
Each information is converted into a specific cellular response
and this is a universal property of living cells (called ‘signal
transduction’). Cell behaviour is regulated by a complex network
of intracellular and extracellular signal transduction pathways.
Extracellular signaling molecules, such as cytokines or growth
factors or extracellular matrix proteins, bind to cell-surface
receptors and then the receptor-ligand interactions trigger events
inside the cell. The intracellular signaling cascades recruit the
signaling molecules including GTPases, Ca2+-regulated proteins
and protein kinases. Fig. 4 shows the major signal pathways
relevant to cancer in human cells.31 In particular, cancerous cells
possess mutated versions of oncogenes and tumor suppressors.
Such mutations alter signal pathway components in a way that
causes them to inappropriately deliver proliferative signals,
switching on cell growth, DNA replication and cell division.
Cancer cells ignore the signals from their environment that
normally keep cell proliferation under tight control. To date, two
types of genes, oncogenes and tumor suppressor genes, have been
recognized as playing a big role in cancer. Their discovery and
understanding have facilitated the development of new kinds of
cancer therapies. The ECIS-based assessment has been one of
important tools for these studies. Recently, Zudaire et al.
quantitatively assessed that knockdown of the aryl hydrocarbon
receptor repressor (AHRR), a bHLH/Per-ARNT-Sim tran-
scription factor located in a region of chromosome 5 (5p15.3), in
human lung cancers significantly enhanced in vitro anchorage-
dependent and –independent cell growth.32 Importantly, the
AHRR was suggested as a putative new tumor suppressor gene
Fig. 3 (a) A schematic diagram of cell cycle. Under normal conditions, in multiply type of human cancers. Transforming growth factors
the ability to pass through the restriction point is mainly governed by
(TGFs) are known as proteins to induce oncogenic trans-
external conditions, such as the presence of growth factors. On the other
formation in a specific cell culture system. Sawhney et al. applied
hand, checkpoint pathways monitor conditions inside the cell and tran-
siently halt the cell cycle at various points if conditions are not appro- quantitative cell micromotion experiments to studying multiple
priate for continuing. (b) normalized impedance as a function of time of cell signaling mechanisms, such as extracellular signal-regulated
HeLa cells measured at 60 kHz. The cells on CIS1 and CIS2 were kinase/mitogen-activated protein kinase (ERK/MAPK), regu-
synchronized by double thymidine block, whilst those on CIS3 were not lating cell adhesion functions in human colon cancer cells.33 They
synchronized. No cell on CIS4 was control. The six numbered black demonstrated that autocrine transforming growth factor-
arrows indicate the exchange of culture medium.30 a (TGF-a) controlled biological functions via activation of

Analyst This journal is ª The Royal Society of Chemistry 2010


View Online
Published on 20 October 2010 on http://pubs.rsc.org | doi:10.1039/C0AN00560F
Downloaded by Imperial College London on 26 October 2010

Fig. 4 A schematic representation of the seven signaling pathways: JAK/STAT (orange), Notch, MAPK/ERK (blue), PI3K/AKT (green), NFkB
(brown), Wnt (yellow) and TGF-b (purple). All ligands are colored in red and transcription factors in white. Factors that exert an inhibitory effect onto
a particular pathway are framed with a black line. Cellular membrane and nuclear membrane are drawn in green and grey, respectively.31

p70S6K enzyme at specific phosphorylation sites. The prostate for targeting specific signal transduction molecules. Importantly,
transglutaminase (TGase-4) regulates the interaction between the ECIS measurements provide several advantages and/or
prostate cancer and vascular endothelial cells. Jiang et al. found unique features for drug discovery when compared to other
that overexpression of prostate transglutaminase (TGase-4) chemotaxis assays including Boyden chamber assays: better
facilitated the adhesion of prostate cancer cells onto endothelial mimic of in vivo events, high sensitivity and real-time monitoring.
cells and its action was independent of the Rho-associated kinase Malignant tumors undergo a series of changes when pro-
(ROCK) pathway.34 Placenta growth factor (PlGF) is a key gressing the formation of metastasis. Many genes and factors
molecule in angiogenesis and vasculogenesis and a member of the have been involved in the tumor invasion including adhesion
VEGF family. Chen et al. investigated the direct biological molecules, cytoskeletal proteins and proteases.36 To investigate
function of endogenous PlGF in human lung cancer cell line the function of such factors in physiological and pathological cell
A549.35 They used the ECIS system to determine the migration of invasion, a quantitative invasion assay in real-time is of signifi-
A549 cells and the knockdown of PlGF resulted in remarkably cant importance. Ren et al. conducted invasion assays of ovarian
reduced migrating capacity. cancer cells (SKOV3 and HEY) using the ECIS system.37 They
On the other hand, metastasis is the most feared aspect of assessed the ability of SKOV3 cells to invade into a confluent
cancer. An ability to invade neighboring tissues is an unique monolayer of peritoneal mesothelial cells producing lysophos-
feature of malignant tumors. To date, although such invasiveness phatidic acid (LPA) since LPA is a potential diagnostic marker
is not thoroughly understood, it is of significant importance to and a therapeutic target for ovarian cancer. Cell invasion
understand mechanisms of the adhesion and migration of the through the mesothelial cell layer resulted in a dramatic drop in
malignant cancers. In general, cell migration has been well resistance, whilst other cell types, such as NIH3T3 and MCF7
studied in non-neoplastic cells, such as fibroblasts and epithelial cells, didn’t cause a significant drop in resistance (Fig. 5a).
cells. It is a highly complex integrated process depending on the Through the transfection with siRNAs against LPA1–3 as well as
actin-rich cortex beneath the plasma membrane. Three distinct control siRNAs against glyceraldehyde-3-phosphate dehydro-
activities involved in cell crawling are as follows. Actin-rich genase (GAPDH) or green fluorescent protein (GFP), they
structures are pushed out at the front of cell (protrusion). The determined whether the receptors for LPA were involved in cell
actin skeleton connects across the plasma membrane to the invasion (Fig. 5b). siRNAs against LPA2 and LPA3 significantly
substratum (attachment) and the bulk of the trailing cytoplasm is inhibited the invasion process, whilst those against LPA1 and the
drawn forward (traction). The cycle is repeated and this results in controls had no influence on cell invasion. They suggested that
moving the cell forward in a stepwise function. It should be noted LPA2 and LPA3 were strongly related to the cellular functions of
that cell migration involves multiple processes regulated by SKOV3 cells. In addition, they pretreated the mesothelial cells
various signaling molecules. For example, the actin cytoskeleton with HELSS (an inhibitor of the calcium-independent phos-
and its regulatory proteins are crucial for the migration in most of pholipase A2, iPLA2) and AACOCF3 (an inhibitor of both
cells. Integrins also mediate the signaling between the cell interior cytosolic PLA2 (cPLA2) and iPLA2). They showed both inhibi-
and the extracellular matrix (ECM). These interactions serve not tors delayed invasion time and reduced invasion extent (Fig. 5c).
only to anchor cells to the ECM but also to provide means of cell CD44 is a cell-surface glycoprotein associated with various
migration. Importantly, cancer cells move within tissues during adhesion-dependent cellular processes including cell migration,
invasion and metastasis by their own motility. Recently, new tumor cell metastasis and invasion. Kuo et al. studied the inva-
cancer treatments, such as anticancer drugs, have been developed sive behavior of human breast carcinoma cell lines by the time-

This journal is ª The Royal Society of Chemistry 2010 Analyst


View Online
Published on 20 October 2010 on http://pubs.rsc.org | doi:10.1039/C0AN00560F
Downloaded by Imperial College London on 26 October 2010

Fig. 5 (a) Invasion of ovarian cancer cells (SKOV3) into peritoneal mesothelial cells (LP9). SKOV3, NIH3T3 or MCF7 cells (1  105 cells in 300 ml
medium) were added on top of the LP9 cell layer. 300 ml-medium without cells was added as control. (b) the inhibitory effect of siRNAs against LPA1–3,
GAPDH and GFP on cell invasion. (c) the effect of inhibitors (HELSS and AACOCF3)on the invasion of SKOV3 cells. LP9 cell monolayers were
pretreated with HELSS (1 mmol/l, 30 min) or AACOCF3 (100 mmol/l, 30 min) before adding SKOV3 cells.37

course impedance measurements.38 They monitored the real-time Cytotoxicity


invasion of human breast carcinoma cell lines (MDA-MB-435
cells) to human umbilical vein endothelial cells (HUVECs). In Cytotoxicity is the cell-killing property of chemical compounds
Fig. 6a, MDA-MB-435 cells were pretreated with TGF-b for 12 including food, cosmetics and drugs or a mediated cell such as
or 24 h and then they added to the HUVEC monolayer after 1 h a cytotoxic T cell. This is independent from the mechanisms of
recording. TGF-b treated cells substantially invaded the typical death; necrosis (accidental cell death) and apoptosis (pro-
endothelial cells when compared to untreated cells. The break- grammed cell death). Cell-based assays can provide essential
down in the cell-cell contact between HUVEC cells resulted the information about the potential effects of chemical compounds
rapid decrease in resistance after 4 h. They elucidated that TGF- on specific cell properties. It should be noted that cells are finely
b would induce CD44 cleavage on the cell surface and this balanced homeostatic machines that respond to external stimuli
enhanced the migration and invasiveness of the MDA-MB-435 through complex pathways. Accordingly, toxicity could be the
cells. Similarly, Saxena et al. quantitatively investigated the effect result of a multitude of cellular events including the changes in
of leptin, a key in the regulation of energy balance and body cell morphology, differentiation, proliferation, function and/or
weight control, on hepatocellular carcinogenesis.39 After communication. Cytotoxicity assays are widely used to screen
HUVECs were fully spread, the confluent HUVEC layers were cytotoxic compounds in the pharmaceutical industry or ‘hits’
challenged with hepatocellular carcinoma cell lines, such as without unwanted cytotoxic effects. For example, vital dyes,
HepG2 and Huh 7 cells. The reduction of impedance indicated such as trypan blue or propidium iodide, are utilized to assess cell
that the invasion resulted from retraction of the endothelial cell membrane integrity. Biochemical methods, such as the MTT or
junctions and extravasation of the two cells on the substratum MTS assay,40,41 the neutral red uptake assay42 and ATP-based
(Fig. 6b). Interestingly, leptin-treated cells showed more steeper assays, have been commonly used to assess viable (living) cells.
decrease in impedance when compared to controls. In addition, Other cytotoxicity tests are based on the pH changes induced by
they evaluated the effect of pharmacologic inhibitors on the acidic metabolites in the neighborhood of cultured cells.43 In
signaling pathways regulated by the leptin. The leptin was related addition, intracellular motion of small vesicles or organelles
to activation of signal transducers and activators of transcription responded to toxicants are characterized by image analysis and
3 (STAT3), AKT, and extracellular signal-regulated kinase video-enhanced microscopy. Although each of these techniques
(ERK) signaling pathways and leptin-induced phosphorylation has its own advantages and disadvantages, these traditional
of AKT and ERK was dependent on Janus-activated kinase methods are time consuming and require complex steps with
(JAK)/STAT activation. The inhibitors of JAK/STAT effectively multiple reagents. Accordingly, it is highly on demand to
blocked leptin-induced invasion. continuously monitor cell behavior against toxic effects and to

Analyst This journal is ª The Royal Society of Chemistry 2010


View Online
Published on 20 October 2010 on http://pubs.rsc.org | doi:10.1039/C0AN00560F
Downloaded by Imperial College London on 26 October 2010

Fig. 6 (a) the effect of TGF-b treatment on transendothelial migration.38 Resistance changes in impedance are challenged with MDA-MB-435 cells.
The cancer cells with or without TGF-b (10 ng/ml) were added to the HUVECs monolayer after 1 h of recording. (b) that of leptin treatment on cell
invasion potential of hepatocellular carcinoma cells.39 HepG2 and Huh7 cells were cultured in Matrigel invasion chambers in serum-free media con-
taining 100 ng/ml leptin (L) for 24 h. For combined treatment, cells were pretreated with 100 mmol/l AG490 (L + AG), 10 mmol/l PD98059 (L + PD) or
10 mmol/l LY294002 (L + LY) for 45 min followed by leptin treatment. Resistance changes in the impedance at 4 kHz as confluent layers of HUVECs
were challenged with HepG2 and Huh7 cell suspensions. The control was the addition of media without HepG2 or Huh7 cells.

accurately analyze the change in cell viability for a long term. a multiarrayed electrode system has been developed to achieve
Recently, ECIS has been considered as a tool for in vitro toxicity high-throughput cytotoxicity testing.47–50 Yeon et al. demon-
testing.44–50 Xiao et al. continuously monitored the impedance of strated a microfabricated cell chip to monitor cell growth and
attached cells exposed to toxicants, such as cadmium chloride, cytotoxic effects of human hepatocellular carcinoma cells
sodium arsenate and benzalkonium chloride.44–46 Fig. 7 shows (HepG2) after treatments with several toxicants, such as
the cytotoxicity effect as a function of concentration of all three tamoxifen and menadione.47 Opp et al. evaluated dose-depen-
compounds tested. Importantly, the length course of toxicant dent responses of HUVEC in response to cytochalasin B known
exposure made the attached and infected cells die or detach from to disrupt actin filaments and rapidly affect cellular
the working electrode and this corresponded to a gradual morphology.48 Interestingly, Curtis et al. showed that four of 10
decrease in capacitance. In addition, the inhibition assays cell lines were appropriate for use in the ECIS system and two of
provided half-inhibition concentration in real time that agreed these, bovine pulmonary artery endothelial cells (BLMVECs)
with that of the standard neutral red assay. They also assessed and iguana heart (IgH-2) cells, responded well with improved
the cytotoxicity of mercury chloride as a model of acute toxicants sensitivity to waterborne industrial chemicals.49 Very recently,
and 1,3,5-trinitrobenzene (TNB) as that for studying long-term Yun et al. determined the effect of magnesium ions on osteoblast
cytotoxicity effects using the ECIS measurements. Recently, (U2-OS) cell activity using an eight-well ECIS electrode.50

This journal is ª The Royal Society of Chemistry 2010 Analyst


View Online

the cells. This healing can take from several hours to over a day
and this depends on cell types, culture conditions and the extent of
the wound region. Instead of mechanical damage to the cells, an
electrical means to wound confluent cell layers can be imple-
mented to the ECIS measurements. Noiri et al. firstly used a DC
pulse to kill the cells and then monitored the repopulation of the
wounded area.51 However, DC signals were not suitable to con-
trollably damage the cells because of the electrochemical genera-
tion of toxic species or the damage of the electrode area depending
on the polarity of the wounding current. Recently, they circum-
vented the problems by using milliampere-level AC wounding
currents at relatively high frequency in the ECIS system.21
Accordingly, coupling invasive and non-invasive features would
Published on 20 October 2010 on http://pubs.rsc.org | doi:10.1039/C0AN00560F

enable the ECIS system to comprise an entirely automated means


to carry out wound-healing assays. When the wounding current
was applied to the cell-covered electrode, the plasma membranes
were electroporated and this resulted in cell killing. Fig. 8a shows
Downloaded by Imperial College London on 26 October 2010

the response of cells to different time exposures of the wounding


current and their recovery. In the next few hours depending on
Fig. 7 (Left) Time-course responses of fibroblast V79 cells exposed to a degree of killed cells, the resistance was increased back to the
cytotoxic chemicals (mM): (top) cadmium chloride, (a) 2.9, (b) 4.6, (c) 6.2 levels of the cell-covered electrode because of the migration of cells
and (d) 8.1; (middle) benzalkonium chloride, (a) 15.2, (b) 18.3, (c) 21.3
from its perimeter inward to replaced the dead cells. In addition,
and (d) 30.4; sodium arsenate, (a) 45, (b) 60, (c) 140 and (d) 200. and
they also investigated the effects of electrode sizes and cell types on
(Right) inhibition curves derived from the time-course responses at t0 ¼
16 h. Half-inhibition concentrations of CdCl2, BAK and NaHAsO4 were the wound-healing assays (Fig. 8b) and their reproducibility after
4.0, 14.0 and 50.1 mM, respectively.44 multiple wounding (Fig. 8c). The ECIS-based wound healing
assays have been successfully demonstrated with different cell
Wound healing lines including BS-C-1 (African green monkey kidney cells; CCL-
26), NRK (normal rat kidney cells; CRL-6509), MDCK (Madin-
Wound healing assays allow for studying cell migration, cell-cell Darby canine kidney cells; CLL-34) and endothelial cells.
interactions and proliferation rates under a variety of culture
conditions. In general, a small area of a cell monolayer is disrupted
Conclusions
or displaced (creating a ‘wound’), the images at the beginning and
at regular intervals are captured and then the images are In this Minireview, we have described that impedance-based
compared to quantify both migration and proliferation rates of cellular assays have distinct advantages for cancer biology and

Fig. 8 Electrical wound-healing assays: (a) different time exposures of an elevated field, (b) different size electrodes and (c) multiple wounding and
recovery.21 BS-C-1 cells were grown to confluence in cell culture wells before measurements started. The impedance measured at 4 kHz is plotted as
a function of time.

Analyst This journal is ª The Royal Society of Chemistry 2010


View Online

drug discovery. Fortunately, entire ECIS systems, such as 17 C.-M. Lo, C. R. Keese and I. Giaever, Biophys. J., 1995, 69, 2800.
ECIS from Applied Biophysics, xCELLigence from ACEA 18 L. Reddy, H.-S. Wang, C. R. Keese, I. Giaever and T. J. Smith, Exp.
Cell Res., 1998, 245, 360.
Biosciences and Roche and Cell Key from MDS Sciex, are now 19 E. Noiri, E. Lee, J. Testa, J. Quigley, D. Colflesh, C. R. Keese,
commercially available and thus they have allowed for better I. Giaever and M. S. Goligorsky, Am. J. Physiol., 1998, 274, C236.
approaches for cancer diagnosis, treatment and prevention. 20 J. Wegener, C. R. Keese and I. Giaever, Exp. Cell Res., 2000, 259, 158.
Accordingly, we believe that these label-free and non-invasive 21 C. R. Keese, J. Wegener, S. R. Walker and I. Giaever, Proc. Natl.
Acad. Sci. U. S. A., 2004, 101, 1554.
measurements in real-time are undoubtedly crucial to understand 22 I. Giaever and C. R. Keese, Proc. Natl. Acad. Sci. U. S. A., 1991, 88,
the behaviours of cancer cells and eventually to design new 7896.
anticancer drugs. In addition, we expect that the ECIS 23 C.-M. Lo and J. Ferrier, Eur. Biophys. J., 1999, 28, 112.
24 B. Burns, D. Walker, E. Brown, L. Thurmon, R. Bowden,
measurements will be readily integrated with standard end-point C. R. Keese, S. Simon, M. Entman and C. Smith, J. Immunol, 1997,
assays to improve the quality and reproducibility of measured 159, 2893.
data. In particular, it is desirable to simultaneously perform both 25 A. R. Burns, R. A. Bowden, S. D. MacDonell, D. C. Walker,
optical and electrochemical investigations. For this purpose, T. O. Odebunmi, E. M. Donnachie, S. I. Simon, M. L. Entman and
Published on 20 October 2010 on http://pubs.rsc.org | doi:10.1039/C0AN00560F

C. W. Smith, J. Cell. Sci, 2000, 113, 45.


transparent electrode materials, such as indium tin oxide (ITO) 26 D. C. Lovelady, T. C. Richmond, A. N. Maggi, C.-M. Lo and
and Poly(3,4-ethylenedioxythiophene) poly(styrenesulfonate) D. A. Rabson, Phys. Rev. E: Stat., Nonlinear, Soft Matter Phys.,
(PEDOT:PSS), can receive much attention if ECIS is combined 2007, 76, 041908.
27 G. Park, C. K. Choi, A. E. English and T. E. Sparer, Cell Biol. Int.,
with inverted phase-contrast and fluorescence microscopes.
Downloaded by Imperial College London on 26 October 2010

2009, 33, 429.


Furthermore, the addition of nano- and microfluidic systems will 28 N. Thomas and I. D. Goodyer, Targets, 2003, 2, 26.
enable us to perform complex processing and analysis operations 29 H. P. Easwaran, H. Leinhardt and M. C. Cardoso, Cell Cycle, 2005, 4,
in the ECIS format.52,53 453.
30 L. Wang, L. Wang, H. Yin, W. Xing, Z. Yu, M. Guo and J. Cheng,
Biosens. Bioelectron., 2010, 25, 990.
Acknowledgements 31 O. Dreesen and A. H. Brivanlou, Stem Cell Reviews and Reports,
2007, 3, 7.
J. Hong acknowledges the Brain Korea 21 Project in 2010. This 32 E. Zudaire, N. Cuesta, V. Murty, K. Woodson, L. Adams,
research was supported by the Kyungwon University Research N. Gonzalez, A. Martınez, G. Narayan, I. Kirsch, W. Franklin,
F. Hirsch, M. Birrer and F. Cuttitta, J. Clin. Invest., 2008, 118, 640.
Fund in 2010. This work was also supported by the WCU 33 R. S. Sawhney, M. M. Cookson, B. Sharma, J. Hauser and
Program (R33-2008-000-10067-0) of Korean Ministry of M. G. Brattain, J. Biol. Chem., 2004, 279, 47379.
Education & Science Technology and by the grant (Grant No: 34 W. G. Jiang, R. J. Ablin, H. G. Kynaston and M. D. Mason,
2010-B04) from Gyeonggi Regional Research Centre (GRRC) Microvasc. Res., 2009, 77, 150.
35 J. Chen, L. Ye, L. Zhang and W. G. Jiang, J. Cell. Biochem., 2008,
Program, Republic of Korea. 105, 313.
36 G. K. Wang and W. Zhang, Histol Histopathol, 2005, 20, 593.
Notes and references 37 J. Ren, Y. J. Xiao, L. S. Singh, X. Zhao, Z. Zhao, L. Feng,
T. M. Rose, G. D. Prestwich and Y. Xu, Cancer Res., 2006, 66, 3006.
1 B. Alberts, A. Johnson, J. Lewis, M. Raff, K. Roberts, P. Walter, 38 Y.-C. Kuo, C.-H. Su, C.-Y. Liu, T.-H. Chen, Chie-Pein Chen and H.-
‘‘Molecular Biology of the Cell’’, 2002, 4th ed., Garland Science. S. Wang, Int. J. Cancer, 2009, 124, 2568.
2 L. J. Kleinsmith, ‘‘Principles of Cancer Biology’’, 2006, Pearson 39 N. K. Saxena, D. Sharma, X. Ding, S. Lin, F. Marra, D. Merlin and
International Edition. F. A. Anania, Cancer Res., 2007, 67, 2497.
3 D. Nahari, R. Satchi-Fainaro, M. Chen, I. Mitchell, L. B. Task, 40 R. Ni, M. A. Leo, J. Zhao and C. S. Lieber, Alcohol and Alcoholism,
Z. Liu, J. Kihneman, A. B. Carroll, L. S. Terada and 2001, 36, 281.
F. E. Nwariaku, Mol. Cancer Ther., 2007, 6, 1329. 41 A. P. Li, C. Lu, J. A. Brent, C. Pham, A. Facket, C. E. Ruegg and
4 American Cancer Society, Cancer Facts and Figures, American P. M. Siber, Chem.-Biol. Interact., 1999, 121, 17.
Cancer Society, Inc., 2003. 42 G. Repetto, A. del Peso and J. L. Zurita, Nat. Protoc., 2008, 3, 1125.
5 R. E. White, Annu. Rev. Pharmacol. Toxicol., 2000, 40, 133. 43 J. W. Parce, J. C. Owicke, K. M. Kercso, G. B. Sigal, H. G. Wada,
6 J. Voros, R. Graf, G. L. Kenausis, A. Bruinink, J. Mayer, M. Textor, V. C. Muir, L. J. Bousse, K. L. Ross, B. I. Sikic and
E. Wintermantel and N. D. Spencer, Biosens. Bioelectron., 2000, 15, H. M. McConnell, Science, 1989, 246, 243.
423. 44 C. Xiao, B. Lachance, G. Sunahara and J. H. T. Luong, Anal. Chem.,
7 T. S. Hug, Assay Drug Dev. Technol., 2003, 1, 479. 2002, 74, 5748.
8 J. M. Atienza, N. Yu, S. L. Kirstein, B. Xi, X. Wang, X. Xu and 45 C. Xiao and J. H. T. Luong, Biotechnol. Prog., 2003, 19, 1000.
Y. A. Abassi, Assay Drug Dev. Technol., 2006, 4, 597. 46 C. Xiao and J. H. T. Luong, Toxicol. Appl. Pharmacol., 2005, 206,
9 B. Xi, N. Yu, X. Wang, X. Xu and Y. A. Abassi, Biotechnol. J., 2008, 102.
3, 484. 47 J. H. Yeon and J.-K. Park, Anal. Biochem., 2005, 341, 308.
10 M. Tarantola, A.-K. Marel, E. Sunnick, H. Adam, J. Wegner and 48 D. Opp, B. Wafula, J. Lim, E. Huang, J.-C. Lo and C.-M. Lo,
A. Janshoff, Integr. Biol., 2010, 2, 139. Biosens. Bioelectron., 2009, 24, 2625.
11 I. Giaever and C. R. Keese, Proc. Natl. Acad. Sci. U. S. A., 1984, 81, 49 T. M. Curtis, J. Tabb, L. Romeo, S. J. Schwager, M. W. Widder and
3761. W. H. van der Schalie, J. Appl. Toxicol., 2009, 29, 374.
12 I. Giaever and C. R. Keese, IEEE Trans. Biomed. Eng., 1986, 33, 242. 50 Y. H. Yun, Z. Dong, Z. Tan and M. J. Schulz, Anal. Bioanal. Chem.,
13 M. Kowolenko, C. R. Keese, D. A. Lawrence and I. Giaever, J. 2010, 396, 3009.
Immunol. Methods, 1990, 127, 71. 51 E. Noiri, Y. Hu, W. F. Bahou, C. R. Keese, I. Giaever and
14 I. Giaever and C. R. Keese, Proc. Natl. Acad. Sci. U. S. A., 1991, 88, M. S. Goligorsky, J. Biol. Chem., 1997, 272, 1747.
7896. 52 L. Kang, B. G. Chung, R. Langer and A. Khademhosseini, Drug
15 C. Tiruppathi, A. B. Malik, P. J. D. Vecchio, C. R. Keese and Discovery Today, 2008, 13, 1.
I. Giaever, Proc. Natl. Acad. Sci. USA, 1992, 89, 7919. 53 J. Hong, J. B. Edel and A. J. deMello, Drug Discovery Today, 2009,
16 I. Giaever and C. R. Keese, Nature, 1993, 366, 591. 14, 134.

This journal is ª The Royal Society of Chemistry 2010 Analyst

View publication stats

You might also like