You are on page 1of 8

View Article Online / Journal Homepage / Table of Contents for this issue

i-SECTION: EDUCATION www.rsc.org/analyst | The Analyst

An introduction to electrochemical DNA


biosensors
Katherine J. Odenthal and J. Justin Gooding*
Published on 23 April 2007. Downloaded by Indian Institute of Technology Chennai on 23/10/2017 15:00:10.

DOI: 10.1039/b701816a

Electrochemical DNA biosensors exploit the affinity of single-stranded DNA for


complementary strands of DNA and are used in the detection of specific
sequences of DNA with a view towards developing portable analytical devices.
Great progress has been made in this field but there are still numerous challenges
to overcome. This review for researchers new to the field describes the
components of electrochemical DNA biosensors and the important issues in their
design. Methods of transducing DNA binding events are discussed along with
future directions for DNA biosensors.

Introduction
The detection of genetic disorders is
clearly of importance for preventative
health care. Many of these disorders are
caused by mutations of the double-
stranded DNA (ds-DNA) in our cells
that comprises the blueprint of our
genetic makeup. Double-stranded DNA
is formed by two single strands (ss-DNA)
bonding to each other to form the well-
known double helix. Because of the way
the two strands wind around each other,
a DNA duplex has two grooves spiralling Fig. 1 Recognition interface consisting of the immobilised DNA probe and steps in the
around the outside of the DNA duplex. detection of a specific DNA target strand.
The larger one is called the major groove
and the smaller one the minor groove, as
shown in Fig. 1. The double helix forms complementary strand of ss-DNA. DNA can be outputted to the end user (see
when all the bases on one strand match microarrays are one example of a Fig. 1). In electrochemical DNA biosen-
with their partner bases on the opposite laboratory-based DNA biosensor that sors, the transducer is an electrode onto
strand. This pairing of bases, known as has found enormous use in scientific which DNA as the biorecognition species
WatsonCrick base pairing, sees a gua- research for gene identification and gene is immobilised. It follows then that there
nine base matched with a cytosine and expression profiling.1 Ideally though, for are two aspects in developing a DNA
thymine matched with adenine. Many the detection of genetic disorders, a biosensor. Firstly the fabrication of the
disorders are caused by even a single portable device that could be used in a biorecognition layer which involves the
mismatch in this base pairing. Single point of care environment for quick immobilisation of DNA. Second is a
base-pair mismatches are also known as diagnosis is desired.2 Electrochemical method of transducing the biorecogni-
single nucleotide polymorphisms or DNA biosensors are one strategy being tion event of a single strand of DNA in
SNPs. Hence the detection of specific explored for the development of DNA solution hybridising with the immobi-
sequences of DNA and the base-pair biosensors as portable devices. This is lised strand. There have been two main
composition is important in diagnosing because electrochemical technology is approaches to the electrochemical trans-
genetic disorders. amenable to miniaturisation, can be duction of DNA hybridisation, which
DNA biosensors are commonly accurate and sensitive with simple self- can be broadly referred to as labelled
employed to detect specific sequences of contained instrumentation and easily methods and label-free methods. Labelled
DNA. They can achieve this by exploit- controlled reaction conditions.2 methods use redox active molecules that
ing the specific affinity of ss-DNA for a Biosensors consist of two main parts: a bind to DNA. They can do this in several
biorecognition interface, that enables the ways including by binding in the minor
selective detection of the analyte, and the groove (see Fig. 1), intercalating a planar
School of Chemistry, University of New South
Wales, Sydney, NSW 2052, Australia. transducer, which converts the recogni- aromatic ring between base pairs or by
E-mail: justin.gooding@unsw.edu.au tion event into an electronic signal that interaction with one of the bases. The

This journal is The Royal Society of Chemistry 2007 Analyst, 2007, 132, 603610 | 603
View Article Online

first electrochemical DNA hybridisation with a peptide-like backbone, is of the conventional double helix does not
biosensor was reported by Mikkelsen and particular interest in DNA biosensing. form but rather the target binds to the
co-workers36 using the minor groove The peptide backbone is neutral and adsorbed probe to give a highly asymme-
binding redox active label Co(Phen)33+ hence a PNA strand binds to DNA with trical unwound duplex. The other draw-
that gave different electrochemical signals a higher affinity than DNA does itself. A back is the affinity of DNA for the
depending on whether the DNA was consequence of this stronger binding is transducer surface meaning therefore the
Published on 23 April 2007. Downloaded by Indian Institute of Technology Chennai on 23/10/2017 15:00:10.

double- or single-stranded. There are also that the higher selectivity as a PNA- target DNA will also have an affinity for
label-free methods which rely on either modified transducer surface can the surface, leading to non-specific
changes to the electrical characteristics of differentiate between the perfect comple- adsorption of DNA. This issue is exacer-
the DNA-modified interface upon hybri- mentary strand of target DNA and one bated when the transduction technique
disation or on the natural electroactivity with a single base-pair mismatch.15 used is indicating the amount of DNA
of DNA. Both the label-free and labelled However, the majority of research has hybridisation occurring by the amount of
approaches will be discussed below. Work been conducted using DNA, partly DNA at a surface (see later).
in this area has been looking to achieve because of the issues related to the The alternative to adsorption of the
both sensitivity and selectivity, and these availability and cost of PNA, but reports DNA onto a surface is to chemically
are the driving forces behind much of the involving PNA are becoming increas- bond the DNA to the surface, preferably
research to date.710 ingly more frequent. from one end only. The emphasis on end-
The purpose of this paper is to Once the type of probe nucleic acids point attachment again relates to the
introduce electrochemical DNA biosen- has been selected, there are number of issue of hybridisation efficiency and
sors and to outline the important factors other important factors associated with ensuring maximum configurational free-
in designing the biorecognition interface immobilising the DNA probe onto the dom for the probe strand. There are
and different approaches to determining electrode surface, regardless of the type several methods published in achieving
transduction. It is important to note that of electrode. How the DNA is immobi- end-point attachment of DNA, which
the DNA biosensor is really the end lised, including how the probe DNA typically relate to having a reactive group
point of an entire analytical device with interacts with the surface to which it is at the end of the DNA, which is used to
steps related to removing the DNA immobilised, needs to be considered. covalently attach the probe DNA to an
from cells, purifying it, cutting it into Much of the early work on electroche- electrode surface. The most popular
usable lengths and amplifying it, all mical DNA biosensors, and many of the method for achieving end-point attach-
preceding the actual detection by the DNA microarrays, involve immobilising ment of probe DNA is via the synthesis
DNA biosensor. DNA to the transducer surface via of mercaptoalkyl linkers on one end of
electrostatic adsorption. The merit of the DNA (thiolated-DNA). We describe
How to design the such a strategy is its simplicity: no this strategy here in detail to illustrate
complicated surface chemistry is required some of the important aspects that need
biorecognition interface
and it is highly compatible with spotting to be considered in designing DNA
For a biosensor to detect DNA, it is techniques. The drawbacks come from recognition interfaces and want to
necessary to have a biorecognition ele- the fact that for the strategy to work remind the reader that this is not the
ment which is selective for the sequence DNA must have a strong affinity for the only viable strategy for the fabrication of
of DNA to be detected. It is important surface. This strong affinity results in the DNA interfaces. This chemistry exploits
that this biorecognition element is probe DNA being fixed onto the surface the affinity of thiols for gold, forming a
integrated with the signal transducer. by multiple points. Considering that self-assembled monolayer, but can also
Integration is most commonly achieved hybridisation requires the two strands be used to attach DNA to maleimide-
by immobilising ss-DNA on the electrode of DNA to wind around each other to terminated surfaces17 and epoxide-
surface. The immobilised DNA is fre- form the double helix, multipoint attach- terminated surfaces.18 The mercaptoalkyl
quently referred to as the probe strand ment typically results in low hybridisa- linker on the end of the DNA ensures a
and the sequence to be detected is called tion efficiency as the configurational strong bond between the DNA and the
the target strand. To fabricate a DNA freedom of the probe strand is signifi- electrode surface, but end-point attach-
biorecognition interface that can selec- cantly restricted by the immobilisation ment alone is not necessarily sufficient to
tively detect target DNA with a high method. In fact, multipoint attachment ensure immobilised DNA with a high
degree of efficiency, the immobilisation of probe DNA raises the question of how degree of configurational freedom.
of the probe DNA and the entire hybridisation is even possible. The nave Tarlov and co-workers19,20 have ele-
interfacial design needs to be carefully explanation is that as the DNA duplex is gantly illustrated the impact of the
thought through. being formed, the probe DNA partially affinity of DNA for the surface on
First of all, the type of probe needs to desorbs from the surface, winds around hybridisation efficiency. Assembling
be considered. Apart from conventional the target strand to form the duplex and thiolated-DNA alone on the surface
DNA there are other variants on the re-adsorbs. Subsequently another por- resulted in a low hybridisation efficiency
nucleic acid theme that can be used as tion of the probe desorbs to allow the with target DNA (between 0 and 5%)
probes, most notably PNA (peptide hybridisation to proceed. However, which was attributed to the affinity of the
nucleic acid). PNA,1114 where the Lemeshko et al.16 have provided some DNA bases for gold surfaces. However,
sugar-phosphate backbone is replaced evidence on charged surfaces that in fact if the DNA-modified surface is incubated

604 | Analyst, 2007, 132, 603610 This journal is The Royal Society of Chemistry 2007
View Article Online

in a solution of mercaptohexanol (MCH) significant steric or electrostatic interfer- changes to the electrical properties of
as a diluent layer, the affinity of the thiol ence between chains would occur. an interface,7 the change in flexibility
moiety on the mercaptohexanol diluent A recent strategy that uses end-point from ss-DNA to the rigid ds-DNA29,30 or
lifts the DNA up off the gold surface and attachment to form SAMs that could the electrochemical oxidation of guanine
the negative dipole of the alcohol on the provide exquisite control of the spacing bases.31,32 The latter is far and away the
distal end of the diluent repels the between probes on graphite electrodes has most popular label-free method, partly
Published on 23 April 2007. Downloaded by Indian Institute of Technology Chennai on 23/10/2017 15:00:10.

negatively charged probe backbone caus- recently been described.25 This method because of its simplicity and partly
ing the ss-DNA probe to project out in exploits the interaction between the elec- because of its versatility, having also
solution. The result is an increase in the trode surface, highly orientated pyrolytic been used to explore small molecules
hybridisation efficiency to almost 100%.7 graphite, and pyrenes that are attached to binding with DNA.33,34 Guanine is the
A further issue to consider is the the end of the probe forming self- most easily oxidisable base with redox
number of base pairs the probe has. assembled monolayers. By altering the peaks being observed at around +1 V (vs.
The probe length is frequently around size of the pyrene moiety, the change in Ag/AgCl) on most electrode materials
1550 bases long with a linker to the thiol spacing between probes could be altered. (more anodic peak potentials have been
of 36 carbons. This linker length can There are other factors that affect the observed on bamboo-like carbon nano-
play an important part in the sensitivity hybridisation efficiency such as salt con- tubes).35 This strategy is exemplified by
of the sensor along with the length of centration and temperature.8 An increase the pioneering work of Wang et al.36,37
the diluent layer. Recent work by the in salt concentration gives an increase in where the probe strand has inosine bases
Gooding group21 illustrated that the DNA hybrid stability26 and the rate of substituted for guanine; inosine also
optimal arrangement is to have the linker hybridisation;27 however, if the detection binds preferentially to cytosine bases
longer than the diluent as the further the method (see below) uses an intercalator but its oxidation signal is well separated
DNA probe is from the surface (which such as Co(phen)33+ then lower salt from the guanine peak. Thus, prior to
really means the top of the diluent layer), concentrations need to be considered.28 hybridisation with the target sequence no
the greater the configurational freedom To summarise this section on the guanine signal is observed. Once the
of the probe and the greater the hybridi- biorecognition interface, when develop- target strand is hybridised to the immo-
sation efficiency. The same conclusion is ing an electrochemical DNA biosensor bilised inosine-containing probe, the
drawn by Watterson et al.22 in that the the researcher must consider not only guanine peaks from the target DNA
efficiency of hybridisation is reduced what type of probe nucleic acid to were detected using chronopotentiome-
when the target region of probe strand employ and how to immobilise it but try. However, owing to the affinity of the
is close to the surface and increased when also how the DNA will interact with the surface for DNA, non-specific binding of
this target region was further away from surface, the length of the linker used to the target DNA is an issue, as is the
the surface. However, for electrochemical couple the DNA to the surface, the configurational freedom of the probe
biosensors there may be a cost in surface coverage of probe DNA and DNA. The increase in background signal
increasing the length of the linker. The the solution environment in which the due to non-specific adsorption inevitably
longer the linker, the harder it is for biosensor will be placed. increases the detection limit. Non-speci-
electron transfer to occur between the fic binding is a significant problem as the
electrode and the DNA. This phenom- technique measures the amount of gua-
enon is important for several electroche- Approaches to achieving nine base present on the electrode surface
mical DNA biosensors which use redox transduction regardless of whether it has hybridised
reporters as is discussed below. Research into transducing DNA hybri- with a probe strand or has non-specifi-
The surface coverage of the ss-DNA disation has been exceedingly active;7,8 cally adsorbed onto the electrode. Wang
probe, which with thiolated-DNA can be transduction strategies can be subdivided et al. have explored strategies to resol-
controlled using the diluent, also has an ving the issue of non-specific binding by
into two main categories, label-free and
effect on the hybridisation efficiency of using magnetic beads which separate and
labelled approaches. Primary considera-
the biosensor. It would seem that the remove the non-specifically bound DNA
tions for DNA biosensors are the detec-
more probes attached to the surface the from hybridised DNA using a biomag-
tion limits (as this will have a big bearing
more sensitive the biosensor will be, but netic processing technology.38 Detection
on the extent of amplification of the
it is also necessary to maintain sufficient limits from these types of devices are
target DNA) and the selectivity. In the
space between the probes to limit repul- below 100 nM,7 and when a non-
ideal case a DNA biosensor would be
sion of the incoming targets and steric complementary strand of DNA was
able to discriminate complementary
effects between the probe strands. introduced a substantially smaller gua-
target DNA from DNA with a single
Peterlinz and Georgiadis23 suggest that nine signal was observed than for a
base-pair mismatch without requiring
a surface coverage of around 5 6 1012 complementary strand.
amplification of the sample. This is a
molecules cm22 is ideal. Repulsion of the Label-free methods can be extended to
goal that has yet to be achieved
target also depends on the ionic strength using electrochemical impedance spectro-
of the buffer solution. For a 1 M scopy (EIS).7,3950 This method measures
Label-free approaches
phosphate buffer with a surface coverage the change in the faradaic impedance, in
of 3 6 1012 molecules cm22 or less, Label-free methods for the detection of the presence of a redox species such as
Levicky et al.24 have determined that no DNA hybridisation either rely on ferricyanide, from an ss-DNA-modified

This journal is The Royal Society of Chemistry 2007 Analyst, 2007, 132, 603610 | 605
View Article Online

electrode to its hybridised version. Owing developed examples of the preferential gold electrode and giving a good
to the repulsion of ferricyanide by the binding strategy. The important differ- electrochemical signal. Once hybridisa-
negatively charged duplex, an increase in ence from the Mikkelsen approach is tion occurs, the DNA again projects
charge transfer resistance is observed. that the redox marker has a higher normal to the surface, distancing the
Work in this area first concentrated on affinity for the ss-DNA as distinct from ferrocene from the electrode surface and
the amplification of the signal by exploit- the ds-DNA. Methylene Blue (MB) can hence switching off the electrochemical
Published on 23 April 2007. Downloaded by Indian Institute of Technology Chennai on 23/10/2017 15:00:10.

ing the extra length of the target strand act as a DNA intercalator, but in this signal. Although this method gives a
beyond the probe.40 This extra length can work it shows an affinity for exposed signal off device, which is prone to false
be hybridised to a third strand with guanine bases.63 Therefore, upon positives, its detection limit of 10 pM 66 is
biotin attached to one end. Avidin can hybridisation, there is a decrease in the encouraging. Recently, the Heeger group
then bind to this strand, causing a greater MB signal as the access of MB to the has further improved the idea to give a
increase in the charge transfer resistance. guanine bases is restricted. There is a signal on device by using target-induced
The attachment of further biotins via linear relationship between the number strand displacement.67
liposomes gives even greater amplifica- of guanine bases in the immobilised A particularly powerful strategy to
tion.49 More recently, the use of EIS has probe and the amount of charge passed transduce DNA hybridisation is by the
been developed with PNA/DNA biosen- which allows the determination of the change in charge transfer properties
sors,46 conducting polymers47 and gold number of probes immobilised if the between a single strand and a double
nanoparticle-based sensors.45 The latter number of guanines per probe is strand of DNA. Long-range charge
method involves attachment of gold known.63 transfer through DNA can be used not
nanoparticles to a bilayer of a thiol- All the methods of detecting DNA only to transduce that a DNA duplex has
containing silane immobilised on a gold hybridisation discussed thus far indicate formed but also to provide information
electrode. The ss-DNA can then be hybridisation due to a difference in on the quality of the duplex formed. The
attached to the nanoparticle without electrochemical signal based on a change result is a DNA biosensor with superior
losing its ability to bind the target strand, in the amount of DNA at an electrode selectivity and, in some cases, simplicity
giving a detection limit of 5 nM. surface. More recently, electrochemical of operation. This strategy was pioneered
biosensors have been developed where by Barton and co-workers6870 using
Labelled approaches the electrochemical signal is actually intercalators such as Methylene Blue or
dependent on the formation of a DNA Daunomycin. The strategy is reliant on
Labelled methods are significantly more duplex. This can be achieved either by the ability of ds-DNA to act as a conduit
popular than label-free approaches using electrochemistry to monitor the for charge transfer over distances of 40 A
because there are many more ways in change in flexibility of DNA upon or more,71 an ability which ss-DNA does
which transduction can be configured hybridisation or by probing the change not possess. Thus, when a DNA duplex is
with high sensitivity and selectivity. In in electron transfer properties. formed, the MB or daunomycin inter-
electrochemical DNA biosensors the Physical changes in DNA from the calate into the DNA duplex and can be
labels are typically redox active mole- flexible ss-DNA to the rigid rod-like oxidised via charge transfer through the
cules36,5153 which either intercalate ds-DNA can be used for detection in DNA. If the probe DNA has not
between the WatsonCrick base pairs of labelled methods, as well as the label-free hybridised with the target then a greatly
a DNA duplex, and hence do not interact methods mentioned above, by attaching reduced current was observed. Single
significantly with ss-DNA, or they are a ferrocene tag to the end of the base-pair mismatches were detected using
molecules that bind preferentially to immobilised probe.30,64 The flexibility of this strategy without the need for any of
either ss-DNA or ds-DNA. There have ss-DNA enables the ferrocene tag to the stringency washings, (stringency
also been a number of reported strategies come within close proximity of the washings are required to differentiate
where the labels are enzymes,54,55 nano- electrode surface such that it can be between perfectly complementary strands
particles38,5661 or liposomes.49,50 oxidised and reduced. Upon hybridisa- and mismatch targets with most other
The first electrochemical DNA hybri- tion, however, the rigid DNA duplex systems). The absence of the need for
disation biosensor by Mikkelsen and co- stands normal to the electrode surface. stringency washings greatly simplifies the
workers,36 where Co(Phen)33+ and This increases the electron transfer dis- operation of this electrochemical DNA
Co(bpy)33+ bound to the minor groove tance from the electrode to the ferrocene biosensor.
of ds-DNA is an example of where the tag to well beyond where any appreciable Related work by Wong and Gooding72
label has a higher affinity for ds-DNA electron transfer can be observed. Heeger has improved both the robustness and the
over ss-DNA. Recent developments in and co-workers65,66 improved these ideas simplicity of the charge transfer approach
this area include the synthesis of a new to ensure that the ferrocene was near the using the negatively charged inter-
cobalt groove binder62 for the detection electrode surface before hybridisation. A calators anthraquinonemonosulfonic acid
of sequences from the HIV DNA show- ferrocene tag is attached to a DNA stem- (AQMS; anthraquinone-2-sulfonic acid)73
ing a detection limit of 27 pM. loop structure immobilised on gold or anthraquinone-2,6-disulfonic acid
The work of Ozsoz and co-workers (Fig. 2); before hybridisation, the loop (ADQS).21,72 Fig. 3 depicts how the
using the redox marker Methylene Blue is arranged such that a section of it DNA biosensor is operated. A DNA
in the detection of sequences related to hybridises to itself, bringing the recognition interface is prepared using
the hepatitis B virus51 is one of the best ferrocene tag into close contact with the the method of Tarlov and co-workers,19

606 | Analyst, 2007, 132, 603610 This journal is The Royal Society of Chemistry 2007
View Article Online

nanoparticles.38,5661 This method is


based on the hybridisation of biotiny-
lated target DNA to detect DNA immo-
bilised onto a magnetic bead.
Streptavidin-modified gold nanoparticles
were then added, which bound the
Published on 23 April 2007. Downloaded by Indian Institute of Technology Chennai on 23/10/2017 15:00:10.

biotinylated target DNA. Hybridisation


is then detected by dissolving the gold
nanoparticles with acid and carrying out
stripping potentiometric analysis at car-
bon electrodes56 (see Fig. 4). Other
similar methods of carrying out the
detection have also been developed.58,60
A label-free method of detecting hybri-
disation has also been developed based
on the inosine methods mentioned ear-
lier. First, the non-hybridised targets are
magnetically removed before denaturing
the hybrid and detecting guanine oxida-
tion56 which is amplified by the addition
of copper.61 These multi-step techniques
give detection limits with the copper
amplification of 0.8 nM.

Fig. 2 (A) Schematic of the DNA stem loop strategy for detecting DNA hybridisation by Future directions
Heeger and co-workers.65,66 A ferrocene tag attached to the end of the DNA stem loop is
Enormous progress has been made in the
electrochemically accessible. Upon hybridisation the ferrocene is removed from the
electrode surface and the electrochemical signal is turned off. (B) Voltammograms development of electrochemical DNA
(background-subtracted) of the sensor containing a range of complementary DNA biosensors but there are still many
concentrations; from top to bottom: 0 M, 30 pM, 500 pM, 30 nM, 800 nM, 5 mM. Inset hurdles to be overcome for portable
shows a calibration curve of peak current with complementary DNA concentration. (Fig. 2 electrochemical DNA biosensors to
reproduced with permission from ref. 66. Copyright 2003, National Academy of Sciences, become widely commercialised. The first
USA.) challenge relates to the fact that DNA
biosensors require some sample prepara-
tion because, unlike many other types of
where thiolated-DNA is assembled onto be differentiated from the current due biosensors, the sample to be analysed is
a gold electrode surface followed by to AQMS in solution, accessing the itself not readily available. All the
MCH as the diluent. The sensor is electrode directly. The final DNA bio- systems discussed above only focus on
exposed to a sample of DNA and to sensor gives a response within one the analysis of a prepared sample; how-
the AQMS intercalator simultaneously hour, can differentiate between comple- ever, much work has been done on
and after a defined time period (one mentary sequences of DNA and both developing miniaturised methods of sam-
hour for the most favourable interface CA and GA mismatches and has a ple preparation and amplification.74,75
for DNA hybridisation) long-range detection limit of 0.5 nM. Furthermore, Even then, the majority of biosensors
charge transfer through ds-DNA allows this DNA biosensor allows DNA hybri- developed thus far possess either good
hybridisation to be transduced. This is disation to be monitored in real time.21 sensitivity or selectivity, and so, in the
possibly the simplest DNA biosensor to The long-range charge transfer strat- future, work needs to be directed towards
operate yet developed. The simplicity egy is one way of improving the selectiv- combining these issues. Sensitivity is
arises from the target DNA and the ity and simplicity of electrochemical an issue since amplification using the
label all being present in the same DNA hybridisation biosensors but per- polymerase chain reaction (PCR) is
solution so there is only one step to haps at the cost of sensitivity. Other usually required to increase copy num-
operate, exposing the biosensor to the methods have made a conscious decision bers, but the amplification process can
sample. This simplicity arises from an not to aim for a one-step procedure, also cause amplification of contami-
anodic shift in the oxidation potential considering any DNA assay will require nants. Reducing the detection limit
of AQMS upon intercalation compared sample preparation to get the DNA out decreases the number of amplification
with when in bulk solution (Fig. 3). of the cell and ready for detection, and cycles and hence alleviates this problem.
This potential shift, attributed to the hence concentrate on sensitivity and Work is required on detecting target
change in environment of the AQMS selectivity instead. A nice example of DNA from a mixture of multiple
upon intercalation, means that the this approach is by Wang et al. who sequences and is something that needs
current due to intercalated AQMS can developed multi-step arrays using to be developed. In some respects this

This journal is The Royal Society of Chemistry 2007 Analyst, 2007, 132, 603610 | 607
View Article Online

Developing this further, certain proteins,


such as MutY, contain a redox centre
that is capable of binding to the DNA
duplex allowing electron transfer to the
electrode and an electrochemical signal
to be detected.79 Other work on protein
Published on 23 April 2007. Downloaded by Indian Institute of Technology Chennai on 23/10/2017 15:00:10.

detection by Wangs group80 uses apta-


mers. Aptamers are sequences of nucleic
acids that selectively bind to proteins. A
biotinylated aptamer is attached to mag-
netic beads that have been coated with
streptavidin. The attached aptamers then
bind the target protein. This is followed
by magnetic separation of the beads to
which the aptamer and protein are
attached. The protein is then released
using NaOH and is detected via a
chronopotentiometric stripping method.
This method of protein detection is
related to other work by Wang et al. on
nanoparticles, discussed above, and gives
detection limits of around 7 nM. Kraatz
et al. have also worked in this field and
used the protein MutS to determine
single base mismatch as MutS only binds
to duplexes containing a mismatch.81 For
all these detection methods, surface
Fig. 3 (A) Electrochemical DNA biosensor based on long-range charge transfer by Wong control is important to allow room for
et al.73 where the hybridisation of the target, intercalation of the redox molecule AQMS and proteins to bind.
the electrochemical measurement were carried out simultaneously. (B) Osteryoung square
Anti-cancer drugs, such as cis-platin,
wave voltammetry results showing the appearance of the shoulder peak due to the
target genetic diseases by binding to
intercalation of AQMS into the DNA duplex. Inset shows background-subtracted shoulder
peak. (Fig. 3B reprinted with permission from ref. 73. Copyright 2006, American Chemical DNA through intercalation or binding
Society.) to the DNA grooves, and information on
the efficiency of these drugs and
improvements in the drug design can be
work has already started.21,76 For proteinDNA interactions have in fun- investigated using electrochemical DNA
example, a long-range charge transfer damental cellular processes, such as biosensors.82 The binding of the small
method using AQMS has already been transcription. Studies of DNAprotein molecules to DNA causes a decrease in
shown to be able to selectively detect interactions are commonly carried out by the guanine signal when the drug binds
target DNA without the loss of signal, techniques such as gel retardation.78,90 to ds-DNA.83 These biosensors have also
even in a solution containing a mixture However, Barton and co-workers77,78 been used in drug activity studies, such as
of excess salmon testes, salmon sperm, have employed their long-range charge the work carried out by Ozsoz and co-
calf thymus or non-complementary transfer biosensor to explore the interac- workers84 in the development of novel
oligonucleotides.76 tions of proteins with ds-DNA: the redox anti-cancer drugs related to cis-platin
What is the future of electrochemical label, daunomycin, is covalently linked such as cis-bis(3-aminoflavone)dichloro-
DNA biosensors? Recently, work in this to the DNA at the top of the film and platinum(II) which exploits the decrease
field appears to be moving beyond immobilised onto a gold surface with a in adenine oxidation signals on the
hybridisation. There are analytes other diluent such as MCH. In one example, binding of these drugs.
than DNA that these biosensors can be the binding of the protein methyltrans- DNA damage and interaction of drugs
employed to detect. A few examples are ferase Hhal to a specific sequence of ds- such as the anti-tumour antibiotic mitox-
proteins,7781 which can be at abnormal DNA is monitored by the decrease in the antrone with DNA duplexes has been
levels in the presence of cancer, charge transfer through the DNA, and investigated also by Oliveira-Brett
drugs such as the anti-cancer drug hence the electrochemical signal, due to et al.9193 This work explores the
cis-platin,8284 and phenotyping, which the protein catalysing the methylation of decrease of guanine and adenine signals
still detects DNA damage but not via cytosine bases. The methylation of cyto- on the binding of these drugs to the bases
hybridisation.79,8589 sine hence disrupts the base-pair stack- to investigate possible DNA damage that
Investigating the interaction of DNA ing. With a protein that does not bind to might be caused by such drugs.
binding proteins with DNA is important ds-DNA such as bovine serum albumin The other trend that seems to be
due to the significant role that these (BSA), no such current drop is observed. emerging is the enzymatic modification

608 | Analyst, 2007, 132, 603610 This journal is The Royal Society of Chemistry 2007
View Article Online

9 T. G. Drummond, M. G. Hill and


J. K. Barton, Nat. Biotechnol., 2003, 21,
11921199.
10 E. Palecek, Talanta, 2002, 56, 809819.
11 P. E. Nielsen, M. Egholm, R. H. Berg and
O. Buchardt, Science, 1991, 254,
14971500.
12 M. Egholm, O. Buchardt, P. E. Nielsen
Published on 23 April 2007. Downloaded by Indian Institute of Technology Chennai on 23/10/2017 15:00:10.

and R. H. Berg, J. Am. Chem. Soc., 1992,


114, 18951897.
13 K. Kerman, M. Vestergaard, N. Nagatani,
Y. Takamura and E. Tamiya, Anal. Chem.,
2006, 78, 21822189.
14 C. Briones, E. Mateo-Marti, C. Gomez-
Navarro, V. Parro, E. Roman and
J. A. Martin-Gago, J. Mol. Catal. A:
Chem., 2005, 228, 131136.
15 J. Wang, Biosens. Bioelectron., 1998, 13,
757762.
16 S. V. Lemeshko, T. Powdrill, Y. Y.
Belosludtsev and M. Hogan, Nucleic
Acids Res., 2001, 29, 30513058.
17 A. A. Vaidya and M. L. Norton,
Langmuir, 2004, 20, 11 10011 107.
18 T. Bocking, K. A. Killan, K. Gaus and
J. J. Gooding, Langmuir, 2006, 22,
Fig. 4 Steps involved in nanoparticle-based detection developed by Wang et al.:56 (a) 34943496.
streptavidin-coated bead; (b) biotinylated probe immobilised onto the magnetic bead; (c) the 19 T. M. Herne and M. J. Tarlov, J. Am.
biotinylated target is hybridised to the probe; (d) capture of streptavidin-coated gold Chem. Soc., 1997, 119, 89168920.
nanoparticles by the hybridised target; (e) the gold tag is dissolved and detection by 20 A. B. Steel, T. M. Herne and M. J. Tarlov,
potentiometric stripping analysis occurs. (Fig. 4 reprinted with permission from ref. 56. Anal. Chem., 1998, 70, 46704677.
Copyright 2001, American Chemical Society.) 21 E. L. S. Wong, E. Chow and J. J. Gooding,
Langmuir, 2005, 21, 69576965.
22 J. Watterson, P. A. E. Piunno and
U. J. Krull, Anal. Chim. Acta, 2002, 469,
115127.
of DNA with labelled bases for pheno- The message from this review we hope 23 K. A. Peterlinz and R. M. Georgiadis,
J. Am. Chem. Soc., 1997, 119, 34013402.
typing of SNPs by the extension of a the reader has absorbed is that tremen-
24 R. Levicky, T. M. Herne, M. J. Tarlov and
DNA template. Generally this requires dous progress has been made into devel- S. K. Satija, J. Am. Chem. Soc., 1998, 120,
enhancing the redox properties of DNA oping electrochemical DNA biosensors, 97879792.
by developing electroactive nucleotides but equally well huge opportunities exist 25 A. A. Gorodetsky and J. K. Barton,
Langmuir, 2006, 22, 79177922.
and incorporating these into DNA for new workers in the field. The field is 26 K. Rose, J. O. Mason and R. Lathe,
strands. King and co-workers79,85,86 have sufficiently mature that many concepts BioTechniques, 2002, 33, 5458.
developed ferrocene- and anthraquinone- have been developed and many of the 27 M. Tsuruoka, K. Yano, K. Ikebukuro,
labelled nucleotides that have been hurdles that must be overcome have been H. Nakayama, Y. Masuda and I. Karube,
J. Biotechnol., 1996, 48, 201208.
incorporated in DNA and RNA. These identified. So we approach an exciting 28 A. Erdem, B. Meric, K. Kerman,
labelled nucleotides were incorporated time where solutions to these hurdles are T. Dalbasti and M. Ozsoz, Electro-
using single base extension for the detec- being found and commercial devices may analysis, 1999, 11, 13721376.
be part of the near future. 29 J. J. Gooding, A. Chou, F. J. Mearns,
tion of SNPs: the different ferrocene and
E. Wong and K. L. Jericho, Chem.
anthraquinone labels gave distinguish- Commun., 2003, 19381939.
able electrochemical signals, which References 30 F. J. Mearns, E. L. S. Wong, K. Short,
would allow different bases to be D. B. Hibbert and J. J. Gooding,
1 J. Wang, Nucleic Acids Res., 2000, 28, Electroanalysis, 2006, 18, 19711981.
detected. Work carried out by Brazill 30113016. 31 E. Palecek, Nature, 1960, 188, 656657.
et al.87,88 uses single base extension with 2 J. Wang, Biosens. Bioelectron., 2006, 21, 32 E. Palecek, Electroanalysis, 1996, 8, 714.
capillary gel electrophoresis and end 18871892. 33 M. Ozsoz, A. Erdem, P. Kara, K. Kerman
column electrochemical detection, again 3 K. M. Millan, A. Saraullo and S. R. and D. Ozkan, Electroanalysis, 2003, 15,
Mikkelsen, Anal. Chem., 1994, 66, 613619.
for SNP detection. Patolsky et al.89 29432948. 34 A. Erdem and M. Ozsoz, Electroanalysis,
extend viral DNA hybridised to a probe 4 K. M. Millan and S. R. Mikkelsen, Anal. 2002, 14, 965974.
immobilised on a gold electrode surface Chem., 1993, 65, 23172323. 35 L. Y. Heng, A. Chou, J. Yu, Y. Chen and
with dNTPs and ferrocene-labelled 5 K. M. Millan, A. J. Spurmanis and J. J. Gooding, Electrochem. Commun.,
S. R. Mikkelsen, Electroanalysis, 1992, 4, 2005, 7, 14571462.
dUTP then use GOx to catalyse trans- 929932. 36 J. Wang and A. N. Kawde, Anal. Chim.
formation of ferrocene to give an ampli- 6 S. R. Mikkelsen, Electroanalysis, 1996, 8, Acta, 2001, 431, 219224.
fied signal. This allows the detection of 1519. 37 J. Wang, G. Rivas, J. R. Fernandes,
7 J. J. Gooding, Electroanalysis, 2002, 14, J. L. L. Paz, M. Jiang and R. Waymire,
the viral DNA as the extension can only
11491156. Anal. Chim. Acta, 1998, 375, 197203.
occur with the template (the viral DNA) 8 J. Wang, Anal. Chim. Acta, 2002, 469, 38 J. Wang, A. N. Kawde, A. Erdem and
hybridised to the probe. 6371. M. Salazar, Analyst, 2001, 126, 20202024.

This journal is The Royal Society of Chemistry 2007 Analyst, 2007, 132, 603610 | 609
View Article Online

39 A. Bardea, A. Dagan and I. Willner, Anal. 59 J. Wang, D. K. Xu, A. Erdem, R. Polsky J. K. Barton, Proc. Natl. Acad. Sci.
Chim. Acta, 1999, 385, 3343. and M. A. Salazar, Talanta, 2002, 56, U. S. A., 2003, 100, 12 54312 547.
40 A. Bardea, F. Patolsky, A. Dagan and 931938. 78 E. M. Boon, J. E. Salas and J. K.
I. Willner, Chem. Commun., 1999, 2122. 60 J. Wang, R. Polsky and D. K. Xu, Barton, Nat. Biotechnol., 2002, 20,
41 F. Patolsky, E. Katz, A. Bardea and Langmuir, 2001, 17, 57395741. 282286.
I. Willner, Langmuir, 1999, 15, 37033706. 61 J. Wang and A. B. Kawde, Analyst, 2002, 79 J. J. Gooding and G. C. King, J. Mater.
42 Y. Xu, Y. Jiang, H. Cai, P. G. He and 127, 383386. Chem., 2005, 15, 48764880.
Y. Z. Fang, Anal. Chim. Acta, 2004, 516, 62 S. Y. Niu, S. S. Zhang, L. Wang and 80 A. N. Kawde, M. C. Rodriguez,
Published on 23 April 2007. Downloaded by Indian Institute of Technology Chennai on 23/10/2017 15:00:10.

1927. X. M. Li, J. Electroanal. Chem., 2006, 597, T. M. H. Lee and J. Wang, Electrochem.
43 Y. Z. Fu, R. Yuan, Y. Q. Chai, L. Zhou 111118. Commun., 2005, 7, 537540.
and Y. Zhang, Anal. Lett., 2006, 39, 63 W. R. Yang, M. Ozsoz, D. B. Hibbert and 81 C. Z. Li, Y. T. Long, J. S. Lee and
467482. J. J. Gooding, Electroanalysis, 2002, 14, H. B. Kraatz, Chem. Commun., 2004,
44 B. Pejcic and R. De Marco, Electrochim. 12991302. 574575.
Acta, 2006, 51, 62176229. 64 A. Anne and C. Demaille, J. Am. Chem. 82 S. Rauf, J. J. Gooding, K. Akhtar,
45 Y. Z. Fu, R. Yuan, L. Xu, Y. Q. Chai, Soc., 2006, 128, 542557. M. A. Ghauri, M. Rahman, M. A. Anwar
X. Zhong and D. P. Tang, Biochem. Eng. 65 R. Y. Lai, E. T. Lagally, S. H. Lee, and A. M. Khalid, J. Pharmaceut. Biomed.
J., 2005, 23, 3744. H. T. Soh, K. W. Plaxco and A. J. Anal., 2005, 37, 205217.
46 J. Y. Liu, S. J. Tian, P. E. Nielsen and Heeger, Proc. Natl. Acad. Sci. U. S. A., 83 G. Bagni, D. Osella, E. Sturchio and
W. Knoll, Chem. Commun., 2005, 2006, 103, 40174021. M. Mascini, Anal. Chim. Acta, 2006, 573,
29692971. 66 C. H. Fan, K. W. Plaxco and A. J. Heeger, 8189.
47 H. Peng, C. Soeller and J. Travas-Sejdic, Proc. Natl. Acad. Sci. U. S. A., 2003, 100, 84 A. Erdem, B. Kosmider, R. Osiecka,
Macromolecules, 2007, 40, 909914. 91349137. E. Zyner, J. Ochocki and M. Ozsoz,
67 Y. Xiao, A. A. Lubin, B. R. Baker,
48 F. Patolsky, K. T. Ranjit, A. Lichtenstein J. Pharmaceut. Biomed. Anal., 2005, 38,
K. W. Plaxco and A. J. Heeger, Proc.
and I. Willner, Chem. Commun., 2000, 645652.
Natl. Acad. Sci. U. S. A., 2006, 103,
10251026. 85 D. A. Di Giusto, W. A. Wlassoff,
16 67716 680.
49 F. Patolsky, A. Lichtenstein and I. Willner, S. Giesebrecht, J. J. Gooding and
68 E. M. Boon, D. M. Ceres, T. G.
Angew. Chem., Int. Ed., 2000, 39, G. C. King, J. Am. Chem. Soc., 2004,
Drummond, M. G. Hill and J. K. Barton,
940943. 126, 41204121.
Nat. Biotechnol., 2000, 18, 10961100.
50 F. Patolsky, A. Lichtenstein and I. Willner, 69 S. O. Kelley, J. K. Barton, N. M. Jackson 86 D. A. Di Giusto, W. A. Wlassoff,
J. Am. Chem. Soc., 2001, 123, 51945205. and M. G. Hill, Bioconjugate Chem., 1997, S. Giesebrecht, J. J. Gooding and
51 A. Erdem, K. Kerman, B. Meric, 8, 3137. G. C. King, Angew. Chem., Int. Ed.,
U. S. Akarca and M. Ozsoz, Anal. Chim. 70 S. O. Kelley, E. M. Boon, J. K. Barton, 2004, 43, 28092812.
Acta, 2000, 422, 139149. N. M. Jackson and M. G. Hill, Nucleic 87 S. A. Brazill, P. H. Kim and W. G. Kuhr,
52 D. H. Johnston, K. C. Glasgow and Acids Res., 1999, 27, 48304837. Anal. Chem., 2001, 73, 48824890.
H. H. Thorp, J. Am. Chem. Soc., 1995, 71 R. E. Holmlin, P. J. Dandliker and 88 S. Brazill, N. E. Hebert and W. G. Kuhr,
117, 89338938. J. K. Barton, Angew. Chem., Int. Ed. Electrophoresis, 2003, 24, 27492757.
53 P. M. Armistead and H. H. Thorp, Anal. Engl., 1997, 36, 27152730. 89 F. Patolsky, Y. Weizmann and
Chem., 2000, 72, 37643770. 72 E. L. S. Wong and J. J. Gooding, Anal. I. Willner, J. Am. Chem. Soc., 2002, 124,
54 Y. C. Zhang, H. H. Kim and A. Heller, Chem., 2003, 75, 38453852. 770772.
Anal. Chem., 2003, 75, 32673269. 73 E. L. S. Wong and J. J. Gooding, Anal. 90 D. Lane, P. Prentki and M. Chandler,
55 Y. C. Zhang, H. H. Kim, N. Mano, Chem., 2006, 78, 21382144. Microbiol. Rev., 1992, 56, 509528.
M. Dequaire and A. Heller, Anal. 74 T. M. H. Lee and I. M. Hsing, Anal. Chim. 91 A. M. Oliveira-Brett, A. M. C. Paquim,
Bioanal. Chem., 2002, 374, 10501055. Acta, 2006, 556, 2637. V. C. Diculescu and J. A. P. Piedade, Med.
56 J. Wang, D. K. Xu, A. N. Kawde and 75 S. W. Yeung, T. M. H. Lee, H. Cai and Eng. Phys., 2006, 28, 963970.
R. Polsky, Anal. Chem., 2001, 73, I. M. Hsing, Nucleic Acids Res., 2006, 34, 92 A. M. Oliveira-Brett, M. Vivan,
55765581. e118. I. R. Fernandes and J. A. P. Piedade,
57 J. Wang, Anal. Chim. Acta, 2003, 500, 76 E. L. S. Wong, F. J. Mearns and Talanta, 2002, 56, 959970.
247257. J. J. Gooding, Sens. Actuators, B, 2005, 93 A. M. O. Brett, T. R. A. Macedo,
58 H. Cai, Y. Q. Wang, P. G. He and 111, 515521. D. Raimundo, M. H. Marques and
Y. H. Fang, Anal. Chim. Acta, 2002, 469, 77 E. M. Boon, A. L. Livingston, S. H. P. Serrano, Biosens. Bioelectron.,
165172. N. H. Chmiel, S. S. David and 1998, 13, 861867.

610 | Analyst, 2007, 132, 603610 This journal is The Royal Society of Chemistry 2007

You might also like