Sunteți pe pagina 1din 16

Analytical Biochemistry 417 (2011) 116

Contents lists available at ScienceDirect

Analytical Biochemistry
journal homepage: www.elsevier.com/locate/yabio

Review

Gold nanoparticle-based signal amplication for biosensing


Xiaodong Cao a, Yongkang Ye b, Songqin Liu a,
a
State Key Laboratory of Bioelectronics, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
b
School of Biotechnology and Food Engineering, Hefei University of Technology, Hefei 230009, China

a r t i c l e i n f o a b s t r a c t

Article history: Colloidal gold nanoparticles (AuNPs), with unique properties such as highly resonant particle plasmons,
Received 4 March 2011 direct visualization of single nanoclusters by scattering of light, catalytic size enhancement by silver
Received in revised form 9 May 2011 deposition, conductivity, and electrochemical properties, are very attractive materials for several applica-
Accepted 17 May 2011
tions in biotechnology. Furthermore, as excellent biological tags, AuNPs can be easily conjugated with
Available online 26 May 2011
biomolecules and retain the biochemical activity of the tagged biomolecules, making AuNPs ideal trans-
ducers for several biorecognition applications. The goal of this article is to review recent advances of
Keywords:
using AuNPs as labels for signal amplication in biosensing applications. We focus on the signal ampli-
Gold nanoparticle
Amplication
cation strategies of AuNPs in biosensing/biorecognition, more specically, on the main optical and elec-
Recognition trochemical detection methods that involve AuNP-based biosensing. Particular attention is given to
Biosensing recent advances and trends in sensing applications.
2011 Elsevier Inc. All rights reserved.

Achieving high sensitivity is one of the major goals in biological ultrasensitive optical and electrical detections with polymerase
assays such as the monitoring of protein (enzymes, antigens, and chain reaction (PCR)1-like sensitivity.
antibodies) interactions and nucleic acidnucleic acid/protein Colloidal gold nanoparticles (AuNPs) have been widely used as
hybridization events. Many efforts have been made toward the labels for analytical signal amplication and biomedical purposes
exploration of novel means to enhance detection sensitivity. Suc- for many years [11,13,14]. AuNPs can be produced in an aqueous
cessful strategies include the employment of new labels such as solution with rapid stirring by reducing chloroauric acid (HAuCl4)
electroactive molecules, redox complexes, and metal ions; the inte- with reducing agent. The Au3+ ions are reduced to neutral gold
gration of enzyme-assisted signal amplication processes; and the atoms. The solution becomes supersaturated as a result of more
incorporation of nanomaterials to increase the upload of electro- gold atoms forming, and gold gradually starts to precipitate in
chemical tags and so on [16]. Among these approaches, the nano- the form of sub-nanometer particles. The rest of the gold atoms
material-based one has been widely used during recent years as that formed during reduction are absorbed on the existing parti-
versatile and sensitive tracers for the electronic, optical, and micro- cles. In addition, if the solution is stirred vigorously enough, the
gravimetric transduction of different biomolecular recognition
events [712]. The remarkable signal enhancement associated 1
Abbreviations used: PCR, polymerase chain reaction; AuNP, gold nanoparticle;
with the use of nanoparticle amplication labels and the formation HAuCl4, chloroauric acid; ECL, electrochemiluminescence; BSA, bovine serum albu-
of nanoparticlebiomolecule assemblies provides the basis for min; IgG, immunoglobulin G; PPD, poly(o-phenylenediamine); Fc, ferrocene; QD,
quantum dot; HRP, horseradish peroxidase; TMB, 3,30 ,5,50 tetramethylbenzidine; P-
gp, P-glycoprotein; ConA, concanavalin A; ssDNA, single-stranded DNA; ELISA,
enzyme-linked immunosorbent assay; CL, chemiluminescent; AFP, a-fetoprotein;
ASV, anodic stripping voltammetry; HQ, hydroquinone; SERS, surface-enhanced
Raman scattering; scFv, single-chain variable fragment; HNP, Hantaan virus nucle-
ocapsid protein; FRET, Frster resonance energy transfer; Lec-QD, lectin-conjugated
QD; PL, photoluminescence; Carbo-AuNP, carbohydrate-conjugated AuNP; PRRSV,
porcine reproductive and respiratory syndrome virus; TR-FRET, time-resolved
uorescence resonance energy transfer; LSPR, localized surface plasmon resonance;
rRNA, ribosomal RNA; SPR, surface plasmon resonance; RSD, relative standard
deviation; RCA, rolling circle amplication; LSPCF, localized surface plasmon coupled
uorescence; NIR, near infrared; HSA, human serum albumin; DLS, dynamic light
scattering; PSA, prostate-specic antigen; f-PSA, free PSA; RLS, resonance light
scattering; ApoAI, apolipoprotein AI; DPV, differential pulse voltammetry; SWV,
Corresponding author. Fax: +86 25 52090618. square wave voltammetry; EIS, electrochemical impedance spectroscopy; MEA,
E-mail address: liusq@seu.edu.cn (S. Liu). microelectrode array; ICP-MS, inductively coupled plasma mass spectrometry.

0003-2697/$ - see front matter 2011 Elsevier Inc. All rights reserved.
doi:10.1016/j.ab.2011.05.027
2 Gold nanoparticle-based signal amplication / X. Cao et al. / Anal. Biochem. 417 (2011) 116

particles will be fairly uniform in size. AuNPs have advantages such which generated electrochemiluminescence (ECL) signal on con-
as rapid and easy synthesis, narrow size distribution, efcient sur- tact with tris(2,20 -bipyridine)ruthenium solution. Because of the
face modication by thiols or other bioligands, and desirable bio- high adsorption capacity of AuNPs, 10- and 6-fold sensitivity
compatibility. Properties such as electron dense core, highly enhancement was obtained for bovine serum albumin (BSA) and
resonant particle plasmons, direct visualization of single nanoclus- immunoglobulin G (IgG), respectively, over their direct immobili-
ters by scattered light, catalytic size enhancement by silver depo- zations on the electrode [20]. This signal amplication was also
sition, conductivity, and electrochemical properties have made demonstrated with N-(aminobutyl)-N-ethylisoluminol as labels
AuNPs to be attractive materials for several applications in bio- [19]. Owing to more labeled protein being accumulated on AuNPs,
technology. Furthermore, AuNPs can be easily conjugated with bio- the sensitivity of the resulting immunosensor had 3 orders of mag-
molecules and retain the biochemical activity of the tagged nitude improvement over those without AuNPs as the immobiliza-
biomolecules, leading AuNPs to be excellent transducers for sev- tion support. Similar signal amplication was reported by Shen and
eral biorecognition applications [1518]. Here we provide a com- coworkers, where a sensitive electrochemical immunoassay, based
prehensive overview of recent advances in biosensing by using on poly(o-phenylenediamine) (PPD) lm/AuNP amplication and
AuNPs as labels for signal amplication. Various signal amplica- ferrocene (Fc) labels, was developed for the detection of immuno-
tion routes by AuNPs in biosensing/biorecognition, specically species [24]. Zhu and coworkers presented a nonlabeled ECL
where AuNPs serve as carriers for capture probe, catalyst, and plat- immunosensor, based on CdSe quantum dots (QDs), for the detec-
form for the deposition of silver to provide further signal enhance- tion of human prealbumin [22]. Two layers of AuNPs were con-
ment, are summarized. In addition, the main optical and structed on the gold electrode surface for the linkage of
electrochemical detection techniques and methods involved in prealbumin antibodies. A detection limit of 1.0  1011 g ml1 for
the use of AuNPs are discussed. prealbumin was obtained based on the inhibition of the ECL reac-
tion between CdSe QDs and K2S2O8 through the formation of
immunocomplexes. More recently, Lin and coworkers fabricated
Sensing/signal amplication strategies
a highly specic and sensitive DNA detection platform. It was
accomplished by means of more dually labeled stemloop DNA
To achieve high sensitivity for the detection of biotargets, a num-
probes, immobilized on the AuNPs to a 96-well microplate, an-
ber of sensing/signal amplication strategies have been developed
chored by a protein [21]. The hybridizations between the hairpin
through the use of various size/shape gold nanostructures (mostly
DNA probes and the target DNA caused signicant conformational
AuNPs) coupled with biorecognition steps such as nucleic acid
changes of the probes, which forced biotin away from the surface
hybridization and the antibody/antigen sandwich-type procedure.
of AuNPs. As a result, the biotin became accessible to the strepta-
These strategies include the use of AuNPs as carriers for both probe
vidinhorseradish peroxidase (HRP) and the target hybridization
and signal molecule loading, as indicators to induce the deposition
events could be sensitively detected via the HRP-catalyzed sub-
of silver on AuNPs for both optical and conductivity detection, as la-
strate 3,30 ,5,50 -tetramethylbenzidine (TMB) with the use of a spec-
bels in coupling with bio-barcode techniques, and as receptors
trophotometric method. Based on this signal amplication
when employing Frster resonance energy transfer methods.
strategy, they could detect DNA at the femtomolar level.
Zhu and coworkers designed a new strategy for the assessment of
AuNPs as carriers cell surface carbohydrates and P-glycoprotein (P-gp) expression sta-
tus by quantication of the cell numbers with an electrochemical
Because of the narrow size distribution, good biocompatibility, immunoassay [26]. Due to the good biocompatibility of AuNPs and
and ease of modication with thiol groups, AuNPs could serve as excellent conductivity of nitrogen-doped carbon nanotubes, a novel
carriers for the immobilization of captured probes. These applica- three-dimensional architecture was initially fabricated by a combi-
tions include immobilization of more biorecognition elements or nation of nitrogen-doped carbon nanotubes, thionine, and AuNPs
a large number of optical or electrochemical tags on AuNPs, which via a simple layer-by-layer method (Fig. 2). This architecture pro-
provide more binding sites or amplication of the analytical signal vided an effective matrix for concanavalin A (ConA) binding and a
in a single recognition reaction (Fig. 1) [1940], and target-induced predominant capability for cell capture. Accompanied by the enzy-
chemical deposition of metals [4173], especially silver [4167]. matic reaction of HRP toward the oxidation of thionine by H2O2, the
proposed cytosensor showed excellent analytical performance for
Immobilization of more active molecules on AuNPs the detection of HeLa cells and cell surface receptors P-gp.
Wang and coworkers fabricated an AuNP layer on the surface of Contrary to the direct immobilization of AuNPs on solid support
gold electrode for immobilization of additional primary antibodies. as an efcient matrix to increase the loading of primary analytes,
4-(Dimethylamino)butyric acid was tagged on the (anti)-analytes, the development of novel colloidal gold labels has received

Fig.1. Signal amplication strategies based on AuNPs serving as carriers. (A) AuNPs were immobilized directly onto a solid substrate to capture more capture probes.
Sandwich-type complexes could be formed after two steps: specic recognition between capture probes and targets and targets and detection probe modied with signal
inducers. The signal amplication could be achieved, although more than one specic recognition occurred on a single AuNP. (Reformed from Ref. [20].) (B) Capture probes
were rst immobilized on a solid substrate. After the rst step, specic recognition between capture probes and targets, AuNPs carrying lots of detecting probes modied with
signal inducers were introduced to the substrate through the second step specic recognition, thereby resulting in the signal amplication. (Reformed from Ref. [30].)
Gold nanoparticle-based signal amplication / X. Cao et al. / Anal. Biochem. 417 (2011) 116 3

Fig.2. Schematic representation of the fabrication of the cell-based electrochemical enzyme-linked immunoassay. ConA, concanavalin A; Ab, antibody. (From Ref. [26].)

Fig.3. Schematic illustration of proposed DNA detection strategy. (From Ref. [28].)

increasing attention [2740]. Cui and coworkers reported an ECL Ju and coworkers designed a novel dual-functionalized nanop-
detection method for specic DNA sequences by the integration robe for highly sensitive and selective in situ evaluation of carbohy-
of luminol-functionalized AuNP labels, amplication of AuNPs, drates on living cells by the integration of specic carbohydrate
and the biotinstreptavidin reaction system [28]. The luminol- recognition and enzymatic signal amplication of proteins on AuN-
functionalized AuNP labels were synthesized via the direct reduc- Ps [35]. In their work, the nanoprobes were prepared by mixing HRP
tion of HAuCl4 by luminol [74]. Thousands of luminol molecules and ConA directly with AuNPs. A nanoscaffold of nanohorns func-
were coated on the AuNPs as stabilizers [75]. This kind of lumi- tionalized with arginineglycineaspartic acidserine tetrapeptide
nolAuNPs could be coupled with single-stranded DNA (ssDNA) was prepared on an electrode surface for cell capture and enhance-
without the loss of ECL activity and change in specicity of the ment of the electrical connectivity. The electrochemical signal was
hybridization reaction. Thus, the luminol-labeled DNA signal produced from the reduction of enzymatic reaction product on the
probes, which were associated with the residual base sequence modied electrode. The enhanced sensitivity could be attributed to
of the targets, were assembled on the electrode through specic the combination of dual signal amplication based on enzymatic
base pairing to form sandwich DNA conjugates on the modied catalysis, high surface-to-volume ratio of AuNPs for protein loading,
electrode (Fig. 3). As a result, an ultrasensitive DNA assay for detec- and high molar ratio of enzyme to lectin on AuNPs. With this ap-
tion of sequence-specic DNA was achieved with a detection limit proach, they could electrochemically detect the model cells with
of 0.19 fM. Such a strategy has already been applied to an enzyme- a detection limit down to 15 cells and monitor dynamic variation
linked immunosorbent assay (ELISA) immunoassay. This was the of carbohydrate expression on living cells in response to drugs.
analysis of CA15-3 antigen with AuNPs as carriers for the immobi-
lization of more HRP-labeled anti-CA15-3 to achieve an amplica- AuNP-induced chemical deposition of silver for signal amplication
tion of the optical signal [30]. Zhang and coworkers represented an The immunogold labeling technique was commonly used be-
enhanced chemiluminescent (CL) immunoassay for sensitive cause of the high electron density of AuNPs, which exhibited as a
determination of a-fetoprotein (AFP) [31,37]. The method made dark brown spot under an electron microscope and resulted in a
full use of magnetic beads as bimolecular immobilization carriers distinct image for labeling. In addition, this image response could
to separate target molecules as well as double-codied AuNP labels be further enhanced through immunogold silver staining, a tech-
modied with HRP-conjugated anti-AFP for signal amplication. nique by which silver particles were precipitated on the surface
The formation of sandwich immunocomplexes between the mag- of the AuNPs [4167]. Three strategies have been reported for
netic beads and AuNP labels allowed the monitoring of the concen- amplication of the detection signals of gold tracers (Fig. 4).
tration of AFP with high sensitivity through the HRP labels activity According to the rst strategy, after the conjugates of oligonucleo-
toward the oxidation of luminol to produce light emittance. To fur- tide/anti-protein nanoparticle and the electrode-bound targets
ther increase the loading of signal molecules, nanogold-enwrapped formed, silver was deposited on the AuNPs to obtain an enhanced
graphene nanocomposites were prepared as trace labels for clinical electrochemical signal (Fig. 4A) [4249].
immunoassays [34]. Signal amplication was achieved with enor- Inspired by the use of AuNP labeling and subsequent silver
mous loading of HRP-conjugated antibodies on the surface of nano- enhancement, Wang et al. proposed a strategy for the detection
gold-enwrapped graphene nanocomposites. of DNA hybridization by measurement of the deposited silver with
4 Gold nanoparticle-based signal amplication / X. Cao et al. / Anal. Biochem. 417 (2011) 116

Fig.4. Detection strategies based on silver enhancement on AuNPs. (A) Voltammetric stripping assay. The hybridization event occurred between DNA strand immobilized on
the solid substrate and gold-tagged DNA. The AuNPs were rst covered with silver by a deposition treatment and then detected by stripping techniques via silver-enhanced
signal. (Adapted from Ref. [18].) (B) Conductivity assay. Probe DNA immobilized in a small gap between two electrodes was hybridized with target DNA and then with gold-
modied DNA probes. Gold was accumulated in the gap. Silver enhancement was performed in the presence of hydroquinone (HQ). The silver precipitated onto the AuNPs
improved the sensitivity of the assay by lowering the resistance across the electrode gap. (C) Scanometric assay. A surface-bound capture oligonucleotide bound one-half of
the target of interest, and an oligonucleotide-functionalized AuNP probe bound the other half. Catalytic reduction of silver onto the capture/target/probe sandwich resulted in
a signal that could be recorded with a microplate reader. (Reformed from Ref. [52].)

an extremely sensitive technique of electrochemical stripping me- The second strategy was based on the specic biological interac-
tal analysis [42]. Dramatic signal amplication was achieved by tions that occurred on a supporting surface with varied resistance or
efcient magnetic removal of noncomplementary DNA. The strat- conductance of the substrate. This method could be termed as con-
egy could also be employed for the detection of proteins [4449]. ductometric assay. Mirkin and coworkers exploited the silver deposi-
An electrochemical immunosensor, based on the precipitation of tion technique to construct a sensor based on conductivity
silver on colloidal gold labels, has been developed. Silver metal measurements according to the second strategy (Fig. 4B) [14]. In
on dissolution in an acidic solution was indirectly determined by their approach, the oligonucleotide capture strands were immobi-
anodic stripping voltammetry (ASV) at a glassy carbon electrode. lized in the gap (20 lm) between two electrodes. A three-compo-
The analytical procedure consisted of the immunoreaction of the nent sandwich approach was performed by hybridized target DNA
antigen (analyte) with the primary antibody adsorbed on the walls to capture AuNP-tagged reporter probes between the electrode
of a polystyrene microwell. After that, a secondary colloidal gold- gaps. In the absence of target DNA, there was no current ow across
labeled antibody was bound. Then silver enhancement, acid disso- the electrode gap. However, in the presence of target DNA, the asso-
lution, and stripping detection of the silver occurred at a glassy car- ciated nanoparticle probes and catalytically deposited silver current
bon electrode [44]. A high sensitivity with a detection limit of could ow between the electrodes, which resulted in a sharp drop in
6  1012 M human IgG was achieved. This level of detection could the resistance of the circuit. Using this method, DNA could be de-
be attributed to the sensitive electrochemical determination of sil- tected with a detection limit of 500 fM. A similar approach was
ver ions and to the catalytic precipitation of a large number of sil- developed for immunoassay by using an electro-microchip to detect
ver ions on the colloidal gold-labeled antibody. Recently, a the immunoreaction signal [50]. The AuNPs were introduced into
sensitive magnetoimmunosensing method for antigen detection, the electro-microchip by a typical sandwiched procedure. The silver
based on the catalytic effect of AuNPs on the electroreduction of precipitation constructed a bridge between two electrodes of the
silver ions, was reported [47]. The authors employed micropara- electro-microchip, which allowed electrons to pass and led the var-
magnetic beads as primary antibody immobilization platforms iation of resistance. The identical strategy could also be applied in
and AuNPs modied with secondary antibodies as highly sensitive protein immunoassay [50] and bacterial biosensors [51] with an
electrocatalytical labels to achieve high sensitivity. A built-in mag- electro-microchip transducer. The detection limits for protein A
net carbon electrode allowed the local collection/immobilization of and Acinetobacter baumannii genomic DNA sample were 1 and
the microparamagnetic beads with the immunological sandwich 0.825 ng ml1 (1.2 fM), respectively.
and AuNP catalysts to attach onto the electrode surface. The selec- The third strategy (Fig. 4C) employed a simple commercial at-
tive electrocatalytic reduction of silver ions onto the surface of bed scanner as a detector for the colorimetric detection of protein
AuNPs led to improved sensitivity, which resulted in a lowered and DNA microarrays [5267]. Mirkin and coworkers developed a
protein detection limit to 23 fg ml1. As an alternative detection scanometric DNA array system coupled with a signal enhance-
method, the silver precipitation on colloidal gold tags could be ment method based on the precipitation of silver on AuNP tags
monitored by a CL method using the Ag+K2S2O8Mn2+H3PO4 [52]. The assay consisted of a captured DNA strand immobilized
luminol system (Fig. 5) [49]. This approach could be applied to on a glass chip that recognized the DNA of interest. Oligonucleo-
the determination of human IgG in human serum samples with tide-functionalized nanoparticle probes and oligonucleotide
high sensitivity and a detection limit of 3  1014 M. targets were then cohybridized to the substrate in buffer solution.
Gold nanoparticle-based signal amplication / X. Cao et al. / Anal. Biochem. 417 (2011) 116 5

Fig.5. Schematic representation of the proposed CL immunoassay based on silver deposition on colloidal gold labels. GIgG, goat anti-human IgG; HIgG, human IgG. (From Ref.
[49].)

After catalytic reduction of silver onto the surfaces of AuNPs to am- labeled proteins or antibodies to the array of carbohydrates immo-
plify the signal, the captured strand/target/nanoparticle sandwich bilized on glass slides and to help them encounter the carbohy-
was visualized with a atbed scanner (hence the term scanometric drates at very low concentration. The gold shell provided a
is used to describe the approach). Target concentrations as low as well-established platform for conjugation of biomolecules. When
50 fM could be detected, which was a nearly 100-fold increase in coupled with a signal amplication method that was based on
sensitivity over traditional uorescence-based assays. With similar nanoparticle-promoted reduction of silver, the sensitivity of an
principles, AuNPs have been applied in ultrasensitive detection of iron oxide/gold core/shell nanoparticle-based assay reached the
DNA hybridization, proteinprotein interactions, and carbohy- subattomole level in carbohydrate detection. This result exceeded
drateprotein interactions [5356]. Xu and coworkers used hydro- that of the analogous uorophore system by 1 order of magnitude.
quinone (HQ) as the reduction agent to facilitate the AuNP- Surface-enhanced Raman scattering (SERS) is one of the most
catalyzed reduction of silver ions to amplify signals for the detec- sensitive diagnostic approaches available to the analytical chemist.
tion of cytokines. The assay used a sandwiched format with an This approach is similar to the method that employed multiple
AuNP-modied aptamer and a biotin-modied aptamer to simul- uorophores as labels; however, the spectroscopic lines in Raman
taneously bind the target protein. The absorbance signal generated spectroscopy are not as broad as the bands in uorescence spec-
by the aptamerprotein complex was enhanced by the deposition troscopy, and the spectral window is much broader. This, in princi-
of silver on the surface of AuNPs [58]. The same amplication tech- ple, allows a greater degree of multiplexing. Furthermore, only
nique has been demonstrated in a convenient and label-free scano- single-wavelength laser radiation is needed to scan a highly multi-
metric approach. This approach integrated the bioconjugation and plex array with numerous target-specic Raman dyes. This is in
aggregation of glyconanoparticles, the specic recognition of lectin contrast to array-based detection of molecular uorophore probes,
by carbohydrate on the cell surface and glyconanoparticles, the where different excitation frequencies are needed for each uoro-
aggregation-regulated silver signal amplication, and the spot test phore. Indeed, target DNA sequences specic to multiple different
for in situ cell surface carbohydrate assay [61]. In this work, the bioterrorism agents have been identied using this approach with
glyconanoparticle was obtained by conjugation of AuNPs with spectroscopically distinguishable nanoparticle probes. Attachment
mannan. These glyconanoparticles exhibited fast aggregation of Raman dye-labeled oligonucleotides to the AuNP probes gener-
when exposed to ConA. In the presence of mannose motif-abun- ated spectroscopic codes for individual targets of interest, thereby
dant cells, the lectin-induced aggregation could be inhibited by permitting multiplex detection of analytes [63,64]. Specically, the
specic binding of lectin to the cell surface carbohydrate to de- presence of the target was conrmed by silver staining, and the
crease the concentration of free lectin in the glyconanoparticle rec- detection of the target was revealed by recording SERS signal of
ognition system. The lectin-induced aggregation could be directly the Raman dye near the nanoparticle surface (Fig. 6). The unoptim-
and quickly visualized by an AuNP-catalyzed silver enhancement ized detection limit of this method was 20 fmol [63].
process due to the fact that the aggregated AuNPs had lower silver
enhancement capability than dispersed AuNPs [62]. This provided AuNP aggregates
an easy-to-use method for in situ monitoring of cell surface
carbohydrates. The aggregation of AuNPs leads to the formation of a new
Ultrasensitive detection of carbohydrateprotein interaction absorption band at longer wavelengths. This is a direct result of
was achieved by magnetic preconcentration combined with silver electric dipoledipole interaction and coupling between the plas-
enhancement amplication technology [59]. The iron oxide/gold mons of neighboring particles in the formed aggregates. Oligonu-
core/shell nanoparticles were synthesized to visualize carbohy- cleotide bridged AuNP aggregates provoked a red-to-blue color
drateantibody or carbohydratelectin hybridization at very low change [7678]. This simple phenomenon pointed out the possibil-
concentration. In this glycan array system, the iron core made it ity of AuNP as a DNA detection agent [11]. The milestone discovery
possible for the magnetic eld to quickly bring nanoparticle- by Mirkin and Letsingers group [76] was that the color change
6 Gold nanoparticle-based signal amplication / X. Cao et al. / Anal. Biochem. 417 (2011) 116

Fig.6. Signal amplication with silver staining by using SERS approach. If Raman dyes (blue spheres) were attached to the labeling probe in the scanometric assay, the targets
could be encoded and detected via the Raman signal of their labels. (For interpretation of the reference to color in this gure legend, the reader is referred to the web version
of this article.) (From Ref. [63].)

Fig.7. Schematic for the amplied sandwich-type detection assay of the BRCA-1 gene using multifunctional crosslinked gold aggregates and magnetic particles. DNP,
detection nanoprobe; MMP, magnetic microparticles; CNP, crosslinking nanoprobe; TMB, 3,30 ,5,50 tetramethylbenzidine. (Note. The drawing is not to scale.) (From Ref. [82].)

resulting from AuNP aggregation could be applied for the quantita- using multicomponent crosslinked AuNP aggregates (Fig. 7) [82].
tive determination of DNA using colorimetric assay. Mercaptoalky- The DNA-bridged gold aggregates, which were loaded with a high-
loligonucleotide-modied AuNPs, averaging 13 nm in diameter, er amount of HRP, were employed as signal amplication labels. In
were used as reporter groups. The hybridization of the probe and the presence of target DNA, the multicomponent detection nanop-
the target sequence resulted not only in the binding of an oligonu- robes were attached to the magnetic microparticles (step I). The
cleotide probe to the target sequence but also in the formation of complexes were further hybridized with nanoprobe to form a
an extended polymeric network where the reporter units were crosslinked aggregate. Once magnetic eld was added, these sand-
interlocked by multiple short duplex segments. The signal for wich complexes were magnetically separated (step II). As a result,
hybridization was governed by the optical properties of the nano- HRP conned at the surface of gold aggregates could catalyze the
particles, which depended in part on their spacing within the poly- enzyme substrate and generate an optical signal. While in the ab-
meric aggregate. Although the detection system was not sence of target DNA, gold aggregates could not attach to magnetic
optimized, it was able to detect 10 fmol of target DNA. Since then, microparticles and, in turn, were not to be separated. The detection
intensive DNA or DNA-related detection assays based on AuNP limit was approximately 1 fmol.
aggregates have been developed [7781]. Zhao and coworkers re- Mirkins approach to DNA hybridization assays could be used to
ported a sandwich-type assay for the optical detection of DNA quantify the level of antibodies in aqueous and serum samples on
Gold nanoparticle-based signal amplication / X. Cao et al. / Anal. Biochem. 417 (2011) 116 7

the basis of the aggregation of AuNPs [16,83,84]. Thanh and Rosen- number of barcode DNA per particle and AuNP-based colorimetric
zweig used AuNPs that averaged 10 nm in diameter, coated with barcode detection. During the detection procedure, only the simple
protein A, to determine the level of anti-protein A in aqueous mixture and separation of probe solutions would result in attomo-
and serum solutions [16]. The aggregation of the AuNPs resulted lar sensitivity without using a microarrayer and complicated signal
in an absorption change at 620 nm that was monitored by an amplication steps such as enzymatic amplication and silver
absorption plate reader. A dynamic range of 2 orders of magnitude, enhancement or sophisticated signal measurement tools. The
along with a limit of detection of 1 lg ml1 of anti-protein A, was detection limit was 30 aM concentration of cytokines, which was
observed. Zeng and coworkers described the phenomena that AuN- approximately 3 orders of magnitude more sensitive than conven-
Ps were brought together to form networks at the micrometer tional nonenzymatic cytokine detection assays and 15 times more
scale after the addition of rabbit IgG, in which a large island-like sensitive than an enzyme-based amplication method.
aggregation that consisted of hundreds of AuNPs was formed. As Zhang and coworkers reported a modied barcode amplica-
is well known, each single-chain variable fragment (scFv) molecule tion assay for ultrasensitive detection of Hantaan virus nucleocap-
captured one arm of the rabbit IgG molecule and then shared the sid protein (HNP) [92]. The modied assay was based on AuNP
antigen with a neighboring scFv molecule (two binding sites per enhancement combined with the immuno-PCR technique on an
rabbit antibody). Consequently, when rabbit IgG was added into ELISA plate. During the assay, the target antigen HNP was captured
the solution of scFv-stabilized AuNPs, aggregations occurred by a polyclonal antibody coated on ELISA microplate wells, fol-
(Fig. 8). The immunosensor showed high sensitivity with a detec- lowed by the addition of AuNPs, dually modied with oligonucle-
tion limit of 1.7 nM of scFv [84]. otides and an HNP-specic monoclonal antibody L13, to form a
sandwich immunocomplex. The oligonucleotides on the AuNP con-
Bio-barcode techniques tained two strands: one as captured DNA immobilized on the sur-
face of the AuNP through an AuS bond and the other as signal
The bio-barcode assay [11,8590] is a promising new amplica- amplication DNA that was partially complementary with the cap-
tion and detection system that uses short oligonucleotides as tar- ture DNA. After the immunocomplex was formed, the signal DNA
get identication strands and surrogate amplication units in was released by heat and was consequently characterized by
both protein and nucleic acid detection. The typical assay involves PCR/gel electrophoresis and SYBR Green real-time PCR. The detec-
two types of particles. One is magnetic beads and has recognition tion limit of this method could be improved down to 10 fg ml1 for
elements for the target of interest attached to their surface, and detection of puried HNP in buffers as well as in human serum,
the other is AuNPs functionalized with both target capture and which was approximately 7 orders of magnitude more sensitive
hundreds of barcode capture oligonucleotides that are hybridized than that of conventional ELISA.
to barcode DNA. In the presence of target DNA, the magnetic
microparticles and the AuNPs form sandwich structures that are Frster resonance energy transfer method
magnetically separate from solution and washed with water to re-
move the hybridized barcode DNA. The barcodes are detected Frster resonance energy transfer (FRET), also called uores-
using either a uorescence or scanometric method [90]. cence resonance energy transfer in some cases, is a mechanism that
Most of the bio-barcode detection schemes still require micro- describes energy transfer between two chromophores. An excited
arrayer-based immobilization of oligonucleotide on a glass chip, state donor chromophore may transfer energy passed a certain dis-
surface passivation chemistry to minimize nonspecic binding, sil- tance (typically 38-nm range) to an acceptor chromophore
ver enhancement of immobilized AuNPs on a chip, light scattering through nonradiative dipoledipole coupling. Muhammed et al.
measurement, and a quantication step. Importantly, sophisticated showed an efcient FRET between the molecular gold cluster (a class
instruments such as microarrayers and chip imaging tools limit of compounds of gold called quantum clusters) Au25 and the dansyl
portability, and the assay cost is high. To obviate or minimize the chromophore. Energy transfer from the dansyl donor to the Au25
above requirements without sacricing attomolar sensitivity of core was manifested by way of the reduced lifetime of the excited
the bio-barcode assay, Groves and coworkers reported an ultrasen- state of the former and drastic quenching of its uorescence. The
sitive colorimetric bio-barcode amplication method that relied on metal (especially AuNP)-enhanced FRET technique was widely used
porous silica beads and an AuNP-based colorimetric DNA detection for studies of the structural dynamics of biomolecules due to its high
scheme for straightforward readout [91]. The assay was based on sensitivity and simplicity for detection of ligand (receptor binding
porous microparticles, which enabled the loading of a large observed merely by the enhanced signal of the acceptor) [93].

Fig.8. Scheme of the aggregation process of scFv-stabilized AuNPs after the addition of rabbit IgG. (From Ref. [84].)
8 Gold nanoparticle-based signal amplication / X. Cao et al. / Anal. Biochem. 417 (2011) 116

Fig.9. Schematic illustration of the selection of quadruplex-binding ligand-based FRET assay using AuNPs as a uorescence quencher. GNP, gold nanoparticle. (From Ref.
[96].)

Fig.10. Schematic principle for assay of the glycoprotein based on the energy transfer between Lectin-modied QDs (Lec-QDs) and carbohydrate-conjugated AuNPs (Carbo-
AuNPs) on a glass slide. (A) strong PL signal from Lec-QDs immobilized on an amine-reactive glass surface (NHS-hydrogel slide); (B) low PL signal due to the energy transfer
between Lec-QDs (as a donor) and Carbo-AuNPs (as a quencher); (C) partial recovery of the PL in presence of glycoproteins because of the reduction in energy transfer
between Lec-QDs and Carbo-AuNPs due to competitive inhibition. (From Ref. [98].)

Ray et al. reviewed the development of the nanoparticle-based uorescence probe for DNA detection based on the FRET process
FRET method for ssDNA sequence recognition and monitored the [97]. After the hybridization step, the FRET occurred because the
cleavage of DNA by single-stranded-specic nucleases [94,95]. Jin donors and acceptors were close enough to each other.
et al. designed a FRET system for selection of quadruplex-binding Kim et al. described a chip-based system for the detection of
ligands with unmodied AuNPs as a uorescence quencher. The as- protein glycosylation using FRET-based energy transfer between
say was based on the properties of AuNP that ssDNA could be ab- QDs and AuNPs. The basic principle of this method relied on mod-
sorbed onto negatively charged AuNPs at a chosen ionic strength, ulation of the energy transfer efciency between the lectin-modi-
whereas the G-quadruplex does not at the same ionic strength ed QDs and carbohydrate-conjugated AuNPs by glycoproteins
(Fig. 9). When a dye-labeled oligonucleotide single-stranded probe [98]. Lectin-conjugated QDs (Lec-QDs) were immobilized on an
was adhered to the AuNPs, the attendant proximity of the dye to amine-reactive glass surface (NHShydrogel slide). The irradiation
the gold led to uorescence quenching of the dye [96]. Zhang of light onto the backside of the glass surface resulted in a strong
and coworkers presented a simple recognition system with CdTe photoluminescence (PL) signal from the immobilized Lec-QDs
QDs as donor and AuNPs as acceptor, linked with complementary (Fig. 10A). When in close proximity to these luminescent QDs,
ssDNA to form CdTe-double-stranded DNAAuNP conjugates as the carbohydrate-conjugated AuNPs (Carbo-AuNPs) functioned as
Gold nanoparticle-based signal amplication / X. Cao et al. / Anal. Biochem. 417 (2011) 116 9

a quencher due to the binding afnity between the lectin and car- frequency is in resonance with the collective excitation of the con-
bohydrate groups (Fig. 10B). However, if glycoproteins were also ductive electrons of the particle. This effect is termed localized sur-
present in the solution that contained the Carbo-AuNPs face plasmon resonance (LSPR). AuNPs LSPRs usually occur
(Fig. 10C), the glycan moiety on the glycoproteins would cause a between 520 and 800 nm. Because of such special plasma absor-
reduction in energy transfer between the Lec-QDs and Carbo-AuN- bance, AuNPs have been extensively used in DNA [103] and anti-
Ps. This was due to competitive inhibition, which resulted in partial bodyantigen [82,8486] analyses.
recovery of the PL of the QDs. Cationic AuNPs were used for signal amplication through ionic
Kim et al. developed a multiplexed assay system for proteases interaction with 16S ribosomal RNA (rRNA) hybridized on the pep-
along with the energy transfer between QDs and AuNPs on a glass tide nucleic acid probe-immobilized surface plasmon resonance
slide. In this system, the PL of donor QDs was immobilized on sur- (SPR) sensor chip. 16S rRNA was used as a genetic marker for iden-
face and quenched due to the presence of acceptor AuNPs in close tication of organisms and could be analyzed directly without PCR
proximity. Meanwhile, the protease activity caused modulations in amplication due to the relatively high number of copies. The pep-
the efciency of energy transfer between the acceptor and donor tide nucleic acid was a neutral backbone structure; therefore,
and, thus, enabled the protease assay. In comparison with the hybridization with 16S rRNA resulted in the ionic condition being
QDdye system, the conjugates of the QDAuNP gave rise to higher changed from neutral to negative. This method resulted in a detec-
energy transfer efciency, which resulted in quantitative assay of tion limit of Escherichia coli rRNA of 58.21.37 pg ml1, which was
proteases with higher sensitivity [99]. decreased approximately 5500-fold compared with that without
Grant and coworkers designed a biosensor by a capture anti- signal amplication. In addition, this method could be used for
body labeled with a uorescent dye bound to a protein labeled Staphylococcus aureus detection without purication of rRNA
with an AuNP or a QD. Then FRET was performed with uorescent [104]. Oligonucleotide-capped AuNPs were used in a sandwich as-
dye as donor and with AuNPs or QDs as acceptor [100]. When ex- say of oligonucleotide or polynucleotide by a ow injection SPR. A
posed to porcine reproductive and respiratory syndrome virus carboxylated dextran lm was immobilized onto the SPR sensor
(PRRSV), a binding event between the virus particle and antibody surface to eliminate nonspecic adsorption of oligonucleotide-
took place, which resulted in a conformational change. This change capped AuNPs. This also facilitated the signal enhancement associ-
altered the distance between the uorescent dye and the nanopho- ated with specic molecular recognition events. The signal
tonic molecule, thereby altering the energy transfer that took place enhancement originated from the LSPR effect and the formation
between the uorescent dye and the QD or AuNP. Using this ap- of an additional layer of a high refractive index at the sensor sur-
proach, they were able to rapidly and effectively detect PRRSV in face. A remarkable low DNA detection level could be achieved with
solution, with a detection limit of 3 particles ll1. Pihlasalo et al. low sample consumption. For the analysis of 39-mer target DNA,
developed easy-to-use homogeneous method that used time-re- the detection limit was as low as 2.1  1020 mol, which corre-
solved uorescence resonance energy transfer (TR-FRET) and uo- sponded to 1.38 fM. The method was shown to be reproducible
rescence quenching for quantication of eukaryotic cells [101]. The (relative standard deviations [RSDs] < 16%) and possessed high se-
method relied on a competitive adsorption of cells and uores- quence specicity [105].
cently labeled proteins onto citrate-stabilized colloidal AuNPs. Hsu and Huang developed a sensitive technique that used the
Fewer than ve cells were detected. rolling circle amplication (RCA) postsignal processing method
Song and Fans group combined the highly specic binding abil- for analyzing signals on protein microarray through the SPR meth-
ities of aptamers with the ultra-high quenching ability of AuNPs to od [106]. They chose oligonucleotide-modied AuNPs as the detec-
develop a multicolor gold nanoprobe for the simultaneous detec- tion probe. Use of the AuNPs tags led to a greater increase in angle
tion of three analytes: adenosine, potassium ion, and cocaine. This shift, which allowed improvements in sensitivity compared with
AuNP was fabricated by self-assembly of three dye-modied se- other non-AuNP-assisted binding events. Huang et al. developed
quences on AuNPs. In their work, dyes were held in close proximity an automated microuidic integrated LSPR-based biosensor in
to the AuNP surface through hybridization of assembled ssDNA which AuNPs were synthesized in solution and immobilized on
probes and dye-labeled aptamer sequences, which resulted in ef- quartz substrates [107]. The developed microuidic integrated sys-
cient AuNP quenched uorescence. In the presence of targets, the tem was capable of transporting a specic amount of biosamples
aptamertarget binding separated the duplex, liberated the dye-la- into the sensor chambers, which achieved sensitive and specic
beled aptamer into solution, and restored the uorescence. This biosensing with less reaction time and reagent consumption.
kind of three-color gold nanoprobe could simultaneously and The spectral position of the LSPR band is strongly dependent on
selectively detect adenosine, potassium ion, and cocaine with high the particle size and shape. The anisotropic gold nanostructures
sensitivity [102]. LSPR peaks, such as gold nanorods and gold nanocages, could be
easily tuned by changes in the synthesis parameters. This allowed
the control of the aspect ratio (AR) and, therefore, showed greater
Detection methods
enhancement in biosensing as markers [108]. Drastic sensitivity
enhancement was obtained through longitudinal plasmonic reso-
Optical methods
nance of gold nanorods for ultrasensitive SPR biosensing. This pro-
cedure used functionalized gold nanorods as amplication labels
AuNPs have been widely used in optical biosensing because of
due to the electromagnetic interactions between the nanotag and
their unique optical properties, especially plasma absorbance.
the sensing lm. The detection sensitivity of the nanorod-conju-
The spectral shifts that originated from adjacent or aggregated
gated anti-rabbit antibody for rabbit IgG was estimated to be
AuNPs have attracted increased interest in the development of
40 pg ml1 [108].
optical biosensors based on biomaterialnanoparticle hybrid
systems.
Fluorescence detection
Fluorescence detection is critical in cell imaging, target recogni-
Surface plasmon resonance tion, disease diagnosis, and fundamental studies of biological pro-
The bright colors of AuNPs, which is the basis for their applica- cesses. Metallic nanoparticles have been shown to enhance the
tion as markers, are due to the presence of a plasmon absorption uorescence emission and decrease the molecular excited state
band. This absorption band is produced when the incident photon lifetimes of vicinal uorophores [87,91,93]. Such an enhancement
10 Gold nanoparticle-based signal amplication / X. Cao et al. / Anal. Biochem. 417 (2011) 116

was a result of interactions between uorophores and surface plas- sample solution that contained free PSA (f-PSA) antigen. The bind-
mons in metallic nanostructures. Therefore, AuNPs have been used ing of f-PSA caused nanoparticles to form dimers, oligomers, or
to enhance the sensitivity of uorescence measurement with the aggregates, depending on the concentration of the antigen. A very
SPR method. A ber-optic biosensor, based on a localized surface low concentration (0.5 ng ml1 for PSA detection) could be de-
plasmon coupled uorescence (LSPCF) system, was developed tected though DLS analysis [116]. The capability of such a method
[109]. This LSPCF biosensor was able to detect mouse IgG at a min- was later testied using mouse IgG as the target protein by the
imum concentration of 1 pg ml1 (7 fM) during the biomolecular same research group [117].
interaction of the IgG with anti-mouse IgG. The results conrmed Resonance light scattering (RLS) techniques based on self-
very high sensitivity due to the amplication of LSPCF intensity assembly of AuNPs have been exploited for small molecule
by AuNP coupling. [118,119], DNA [120], and protein [7376] detection. The large size
Fluorescent molecules emitted at wavelengths in the physiolog- of the assemblies of AuNPs and the surface plasmon coupling
ically relevant water window (700900 nm) were of particular among AuNPs can enhance RLS signals. A nano-gold-labeled
interest. This was due to the large penetration depth of near infra- immunoresonance scattering spectral method was established by
red (NIR) light in most biological media and offered the potential combining an immunoreaction and the resonance scattering ef-
for imaging at signicant depths in living tissues. However, it has fects of AuNPs [121]. This method showed high sensitivity and
been proven that it was rather difcult to achieve bright uores- good selectivity for quantitative determination of apolipoprotein
cent with photostable and biocompatible NIR uorophores. Aniso- AI (ApoAI) and ApoB in the presence of polyethylene glycol (which
tropic gold nanostructures, such as gold nanorods and gold served as stabilizer to AuNPs) in human serum. Recently, RLS has
nanoshells, have proven to be excellent materials due to longitudi- been carried out for the detection of adenosine in human urine.
nal plasmonic resonance, which could be tuned to the NIR wave- The sensitive method was based on the specic recognition and
lengths [110,111]. The uorescence enhancement of the NIR signal amplication of adenosine aptamer-modied AuNPs, which
uorophore IR800 by gold nanoshells and gold nanorods has been was fabricated by triggering a structure switch of the 30-terminus
recorded. Human serum albumin (HSA) served as a spacer layer be- G-rich sequence and aptamer duplex via G-quartet-induced nano-
tween the nanoparticle and the uorophore. The measurements re- particle assembly. RLS signal linearly correlated with the concen-
vealed that the quantum yield of IR800 was enhanced from 7%, as tration of adenosine over the range of 6115 nM. The limit of
an isolated uorophore, to 86% in nanoshellHSAIR800 complex detection for adenosine was 1.8 nM with RSDs of 2.904.80%
and 74% in nanorodHSAIR800 complex. This dramatic increase (n = 6).
in uorescence showed tremendous potential for contrast Table 1 lists some AuNP-based amplication optical detection
enhancement in uorescence-based bioimaging. methods with details.

Scattering detection Electrochemical methods


On exciting the LSPR of AuNPs or a nanostructured substrate,
the large electromagnetic elds that localized at the surface of Certainly, electrochemical techniques are the most widely
the gold nanostructure can be exploited to measure enhanced used methods in biomaterial analysis and sensing applications
vibrational molecular signatures. This occurs through the phenom- based on metal and semiconductor nanoparticles. This is due to
enon known as surface-enhanced Raman scattering. SERS is a pow- their relatively low cost and the widespread diffusion of the re-
erful spectroscopic technique that provides nondestructive and lated instruments required for such techniques in addition to en-
ultrasensitive characterization down to the single molecular level, hanced sensitivities achieved by new electrode materials during
comparable to single-molecule uorescence spectroscopy. It has recent decades [41]. AuNPs and their various nanostructures have
been used for sequence-specic DNA detection [63,112,113] and been used to achieve direct wiring of enzymes to electrode sur-
protein detection [114]. The aggregation of AuNPs caused by the face. This promoted electrochemical reaction to impose nano-bar-
specic recognition between the biomolecules carried by AuNPs code for biomaterials and to amplify signal of biorecognition
and target biomolecules or by deposition of silver on the surface event [140].
of AuNPs was responsible for the signal enhancement. Most inter-
esting and also important, the SERS could be turned on and off by Voltammetric methods
biomolecular binding and dissociation event. The mechanisms of The application of voltammetric techniques for AuNP-based sig-
such controlled metal nanoparticle-based SERS for DNA recogni- nal amplication for biosensing was based on two detection meth-
tion and signal amplication were rst demonstrated by Graham odologies. The rst one was direct methods that did not involve
et al. [115] and further conrmed by Nie and coworkers [112]. Dra- any of the prechemical dissolution of AuNPs. The second was based
matic changes occurred in both the optical absorption and surface- on previous dissolution of AuNPs and analysis of the correspondent
enhanced Raman spectra of the 60-nm SERS beacons excited at ions or catalytic methods by other species. Merkoi and coworkers
785 nm. This suggested that sequence-specic DNA hybridization developed a geomagnetic sensor for detection of DNA hybridiza-
brought two or more particles into a distance range and, thus, re- tion. They used magnetic graphiteepoxy composite electrodes to
sulted in plasmonic coupling and further enhancement of the Ra- accumulate gold tags. This involved the hybridization between a
man signals [112]. capture DNA strand, which was linked with paramagnetic beads,
Dynamic light scattering (DLS), also known as photon correla- and another DNA strand related to the BRCA1 breast cancer gene,
tion spectroscopy or quasi-elastic light scattering, is a technique which was coupled with streptavidin-coated AuNPs. The electro-
routinely used to analyze the size and size distribution of poly- chemical detection of AuNPs by differential pulse voltammetry
mers, proteins, colloids, and nanoparticles. Because of the strong (DPV) showed that the sensor provided reliable discrimination
light scattering property of AuNPs, it is natural to hypothesize that against noncomplementary DNA as well as against one- and
DLS can be a very sensitive technique for quantitative detection three-base mismatches [141]. This detection methodology was
and analysis of nanoparticle probes at low concentration. A one- also illustrated by Kerman and coworkers [142]. They employed
step, wash-free, and amplication-free assay for prostate-specic a biotinylation of the kinase substrate and a biotin-modied aden-
antigen (PSA) [116] analysis using AuNPs coupled with the DLS osine 50 -triphosphate [c]-biotinyl-3,6,9-trioxaundecanediamine as
technique was developed for the rst time in 2008 by Liu et al. the cosubstrate. After the phosphorylation and biotinylation of the
Anti-PSA-conjugated AuNPs (anti-PSA AuNPs) were mixed with a peptides, the attachment of streptavidin-coated AuNPs onto the
Gold nanoparticle-based signal amplication / X. Cao et al. / Anal. Biochem. 417 (2011) 116 11

Table 1
Some AuNP-based amplication optical detection methods.

Detection method AuNP Amplication times Target(s) Detection Linearity range(s) Reference
diameter limit(s)
(nm)
SPR detection
Surface-enhanced plasmon resonance 15 DNA [103]
SPR 30 10 times bare chip Salmonella 104105 cells ml1 [122]
typhimurium cells
SPR AChEACL1; ACL2 7 pM; 12 pM 0.0110 nM; 0.02 [123]
1.2 nM
LSPR-coupled uorescence 20 12-fold IgG 1 pg ml1, 7 fM 01 ng ml1 [109]
LSPR 13.5 BiotinBSA DNA [124]
1
SPR 25 12.5-fold Testosterone 3.7 pg ml [125]
Luminescence detection
ECL 16 Human IgG 1.68 ng ml1 5100 ng ml1 [19]
ECL 12 BSA: 10; IgG: 6 BSA; IgG 1 lg ml1; 10120 lg ml1; [20]
5 lg ml1 5100 lg ml1
ECL 12 QDs provide 1.7 order of Human prealbumin 1  1011 g ml1 5  1010 to [22]
magnitude on AuNPAu 1  106 g ml1
electrode
CL immunoassay AFP 0.01 ng ml1 0.15.0 ng ml1 [31]
ECL 20 Human IgG 1 pg ml1 7.5100 pg ml1 [39]
CL 16 Human IgG 5 pg ml1 0.0250 ng ml1 [49]
Photoelectrochemical 15 DNA 1 nM 1500 nM [126]
Light scattering detection
Optical signal 15 1000 Cytokine 83 aM 1 fM100 pM [58]
Light scattering spectra 10-fold DNA: BRCA-1 gene 1 fM [82]
SERS-plasmonic coupling 40/60 40200 DNA [112]
Enhanced RLS Urinary adenosine 1.8 nM 6115 nM [118]
Resonance Rayleigh scattering 20 1000 Transferrin 85 pM 85 pM3.4 nM [127]
50
Resonance scattering 9 ApoAI; ApoB 2.04 lg L1; 0.0083 0.333 mg L1; [121]
0.96 lg L1 0.00200.197 mg L1
Light scattering detection 13 1000-fold Human IgG 10 ng ml1 0.0510 lg ml1 [83]
Hyper-Rayleigh scattering 10 10 times colorimetric by 10 lg ml1 [128]
UVvis
DLS 40 [129]
SERS 46 4; 1010; 250-fold 1,5-Naphthalenedithiol zmol [130]
41 8
SERS Glucose [131]
Colorimetric/scanometric detection
Charge-coupled device image 8.512 Human IgG 0.0510 ng ml1 [57]
Scanometric image Carbohydrateprotein 100 fM 1091016 M [59]
interaction
Scanometric image 15; 20 1000 times ELISA PSA 1 fg ml1 104 range [89]
Scanometric image 5 IgG 1 ng ml1 [60]
Colorimetric immunoassay 15 IgG 1.7 nM 1.7170 nM [84]
Scanometric image Comparable to PCR HeLa cell 10 cells [132]
Scanometric 13 10,000 PSA, AFP, human 300 aM in buffer [133]
chorionic gonadotropin 2 fM in serum
Scanometric image 40 8 DNA 2 fmol 0500 fmol [134]
Camera image 10; 40 100 Troponin 0.01 ng ml1 0.1014.27 ng ml1 [135]
Colorimetry 13 10,000 Single-nucleotide 70 fM [136]
polymorphism
genotyping
ELISA 2 CA15-3 060 U ml1 [30]
Colorimetric image 30 107 times ELISA Hantaan virus 10 fg ml1 [92]
Colorimetric image >14 1000-fold DNA; DNA cleavage 0.5 fmol [137]
Colorimetric image 13.7 0.8 DNA 1 pmol [138]
Colorimetric image 9 Blood group [139]

Note: AChE, acetylcholinesterase, ALC1 and ALC2, AuNPs labeled carbamate inhibitors.

surface of surface plasmon-coupled emission via biotinstreptavi- phenol [32] using gold as catalyst [145]. Much attention has been
din afnity was performed. Direct DPV analysis of the captured given to the latter strategies, which yielded high sensitivities. The
AuNPs enabled the monitoring of the activity of the kinase and enhancement in sensitivity for an electrochemical immunoassay,
its substrate as well as the quantitative information about the ex- by the catalytic deposition of Au3+ onto AuNPs in the presence of
tent of the phosphorylation and biotinylation reactions. reduction agent, has been studied [145]. The chemical deposition
High sensitivity could be obtained when the indirect detection of Au3+ on the AuNP-labeled proteins enlarged the square wave
methods were performed. As mentioned before, such methods voltammetry (SWV) signals. This allowed the author to sensitively
were either based on predissolution of AuNPs and analysis of the detect analyte with a detection limit of 1.6 fM protein. This limit
corresponding ions [143,144] or based on catalytic deposition of was 3 orders of magnitude lower than that obtained by a conven-
other species such as silver [42,44,47], copper [68,69], and p-nitro- tional immunoassay that used the same AuNP label.
12 Gold nanoparticle-based signal amplication / X. Cao et al. / Anal. Biochem. 417 (2011) 116

Fig.11. Schematic representation of successive fabrication, human IgG detection (5 ng ml1), and regeneration of an MEA platform-based immunosensor. EDC/NHS, 1-ethyl-
3-(diethylaminopropyl)carbodiimide/N-hydroxysuccinimide. (From Ref. [148].)

Table 2
Some AuNP-based amplication electrochemical detection methods.

Detection method AuNP diameter (nm) Amplication times Target(s) Detection Linearity range(s) Reference
limit(s)
Voltammetric detection
ASV 20 4.7 IgG 1 ng ml1 1.6627.25 ng ml1 [44]
ASV 13 Human IgG 0.5 ng ml1 2250 ng ml1 [68]
ASV 15 Salmonella typhi 98.9 cfu ml1 1.30  102 to [69]
2.6  103 cfu ml1
ASV 16; 5 200 DNA 0.5 fM 1 pM1 nM [149]
AC voltammetry 13 300% IgG 10 pg ml1 251000 pg ml1 [24]
CV Comparable to the AFP 0.6 ng ml1 155 ng ml1 [23]
immunoradiometric assay
CV DNA 1 fM 1 fM0.1 lM [32]
CV 15 3 orders of magnitude Human IgG 23 fg ml1 5  108 to [47]
7.5  107 lg ml1
CV CA15-3 0.5 U ml1 2.5120 U ml1 [150]
CV 16 2.28 HBsAg 0.1 ng ml1 0.5650 ng ml1 [151]
CV AuNP seeds: 13; Au- 50 CEA; AFP 4 pg ml1; 0.0480 ng ml1; [152]
PB-Fe3O4: 80 7 pg ml1 0.074142 ng ml1
CV 10 Comparable to bio-barcode Protein, mouse IgG 1 fg ml1, 7 aM 1 fg ml1 to [153]
assay 10 lg ml1
CV AuNW (200 nm Label free without Testosterone 0.1 ng ml1 1.283.5 ng ml1 [154]
diameter, 10 lm length) mediator
1 1
CV 16 AFP 0.23 ng ml 1.25200 ng ml [155]
CV 16 CEA 0.06 ng ml1 0.2120 ng ml1 [156]
CV 10 Mouse IgG 1 fg ml1 (CV) 1 fg ml1 to [157]
100 lg ml1
CV 16 Human IgG 8  107 M 5.0  106 to [158]
9.6  104 M
DPV 13 AFP 1.0500 ng ml1; [27]
0.8 ng ml1
1
DPV 20 Human IgG 40 pg ml 0.12580 ng ml1 [33]
DPV 10 Mouse IgG 100 ag ml1 1 fg ml1 to [157]
100 lg ml1
DPV 20; 41; 71.5 HBsAg 2360 ng ml1 [159]
DPV 10 Thrombin 7.82 aM 74.7 aM1.5 fM [160]
Amperometric detection
it Plot 20200 3-fold Estradiol 6 pg ml1 1200 pg ml1 [161]
it Curve 5; 10; 15 Ascorbic acid, dopamine, and 1.5  107 M 5.1  107 to [162]
hydrogen peroxide (ascorbic acid) 6.7  104 M
Electrochemical impedance spectroscopy
EIS 5 Protein A 1 ng ml1 [50]
EIS 15 1000 times lower IgG 36 pg ml1 0.05100 ng ml1 [148]
(human IgG)
EIS 25 Salmonella spp. 102 cfu ml1 102105 cfu ml1 [163]
EIS 2.56 2 orders of magnitude ApoAI 50 pg ml1 0.110 ng ml1 [164]
EIS 10 Fluorescein 15 ng ml1 [165]

Note: CV, cyclic voltammetry; it, currenttime; PB, Prussian blue; AuNW, gold nano-wire; HbSAb, Hepatitis B surface antigen; CEA, Carcinoembryonic antigen; CA 15-3,
cancer 15-3; ApoAI, apolipoprotein A-I.

Electrochemical impedance spectroscopy resistance occurring at conductive or semiconductive surfaces.


Electrochemical impedance spectroscopy (EIS) is a very power- Electrochemical transformations that occurred at the electrode/
ful tool for the analysis of changes in interfacial capacitance or electrolyte interface can be modeled by extraction of components
Gold nanoparticle-based signal amplication / X. Cao et al. / Anal. Biochem. 417 (2011) 116 13

in the electronic equivalent circuits that corresponded to the acid-digested and quantied, through the determination of gold,
experimental impedance spectra [146]. AuNPs have recently been using ICP-MS. A rapid and sensitive assay for the E. coli O157:H7
used in impedimetry-based biosensing applications with the main bacteria could be completed with antibody afnity binding, AuNP
goal of amplifying electrical signals generated by biorecognition labeling, and ICP-MS detection. The assay was able to detect as
events that occurred at the electrode surface. Bonannia et al. few as 500 E. coli O157:H7 cells in 1 ml of sample (500 cfu ml1).
[147] recently used AuNPs for impedimetric signal amplication Tests with nonpathogenic E. coli (DH5a, ATCC35218, and
in a DNA sensing device. In their work, the probe oligomer was ad- ATCC25922) showed high specicity of the assay for E. coli
sorbed onto a glassy carbon electrode surface and the impedance O157:H7. Each assay was completed within 40 min. Demonstra-
measurement was performed in a solution containing the redox tion of this assay for E. coli O157:H7 suggested potential for detec-
marker ferrocyanide/ferricyanide. They used streptavidin-coated tion of a variety of bacterial pathogens.
AuNPs for specic binding of the target biotinylated oligomer after
the hybridization event. A silver enhancement treatment was per-
Conclusion and outlook
formed at the end to visualize the hybridization extension on the
electrode surface. This technique offered further signal amplica-
The studies described above demonstrated the potential of bio-
tion. Yu and coworkers introduced a renewable, site-selective
conjugated nanoparticles for amplication transduction of biorec-
immobilization platform of microelectrode array (MEA) for multi-
ognition events. Understanding of AuNPs nanostructures and their
plexed immunoassays by using pencil graphite particles coated
role in the catalytic events was a critical step forward for biosen-
with gold layers as microelectrodes (Fig. 11) [148]. The results
sing applications. Given the enormous amplication afforded by
showed that the developed MEA sensor allowed detection of hu-
AuNPs, remarkable sensitivity for detection of small molecules,
man IgG with a wide linear range (0.05100 ng ml1) and more
disease markers, biothreat agents, infectious agents, and cells
than three times higher sensitivity than that of the conventional
was achieved.
gold electrode. The primary antibodies were site selective and
Successful realization of the new signal amplication strategies
immobilized on this platform to selectively capture their antigens
required proper attention to nonspecic adsorption issues that
from the sample media. The subsequent introduction of AuNP-la-
commonly controlled the detectability of bioafnity assays. Thus,
beled antibodies resulted in a signicant decrease in the interfacial
proper washing and surface blocking steps should be employed
electron transfer resistance. In addition, this site-selective immobi-
to avoid amplication of background signals (associated with non-
lization platform could be easily regenerated by a simple electro-
specic adsorption of the nanoparticle ampliers). These strategies
chemical treatment.
included the use of AuNPs as carriers for both probe and signal
Table 2 lists some AuNP-based amplication electrochemical
molecule loading, as indicators to induce the deposition of silver
detection methods with details. Although the specic electrochem-
on AuNPs for both optical and conductivity detection, as labels in
ical methods listed were used for quantitative measurement of the
coupling with bio-barcode techniques, and as receptors when
substrates in each reference, other electrochemical or optical
employing FRET methods. Recent work, based on FRET for the
methods were also employed for characterization of the sensing
investigation of metal and semiconductor nanoparticles and QDs,
interface/interphase conditions. This gave more information such
illustrated the extraordinary sensitivity achievable by this tech-
as the inuence of different nanoparticles size or type, electro-
nique. In conclusion, the extraordinary physical and chemical
chemical behaviors, and performances of the sensor.
properties of structures available for AuNPs, along with their appli-
cations, will promote fundamental studies in connection with
Inductively coupled plasma mass spectrometry
those of other inorganic/organic molecules. This potential enables
biomaterials in multi-/interdisciplinary research involving various
Inductively coupled plasma mass spectrometry (ICP-MS) has
sciences (e.g., physics, chemistry, biology, food, health care), agri-
proved to be one of the most sensitive techniques for trace element
culture, and industry.
analysis, with upsides including large dynamic range, low detec-
tion limits, and multielement and rapid analysis capability [166].
By taking advantage of its high sensitivity and wide linear dynamic Acknowledgments
range, ICP-MS was used as a powerful detection method in biosen-
sing with the signal amplications of AuNPs [166,167]. Recently The project was supported by the Key Program (21035002)
Zhao et al. demonstrated a novel sandwich assay for human a- from the National Natural Science Foundation of China, the Na-
thrombin that used two afnity aptamers for increasing specicity, tional Natural Science Foundation of China (20875013), the Key
AuNPs for signal amplication, and ICP-MS for highly sensitive Program (BK2010059) from the Natural Science Foundation of
detection [168]. The rst aptamer, Apt29, was conjugated to AuNPs Jiangsu Province, the Technology Support Program of Jiangsu Prov-
(10 nm). The second aptamer, Apt15, was attached to streptavidin- ince (SBE200900289), and the Open Research Fund of State Key
coated magnetic beads (1 lm). The binding of the two aptamers to Laboratory of Bioelectronics, Southeast University. We gratefully
the same thrombin molecule resulted in a sandwich complex acknowledge D. Shen (Canada) for polishing the English.
(Apt29thrombinApt15). The use of magnetic beads facilitated
the capture of targets in homogeneous solution and provided fast References
and efcient magnetic separation of the afnity complexes. While
the magnetic beads were removed, the AuNPs in the solution were [1] J. Wang, Nanomaterial-based electrochemical biosensors, Analyst 130 (2005)
421.
analyzed by ICP-MS. The detection limit of human a-thrombin was [2] A. Warsinke, A. Benkert, F.W. Scheller, Electrochemical immunoassays,
as low as 0.5 fmol. The dynamic range covered approximately 3 or- Fresenius J. Anal. Chem. 366 (2000) 622634.
ders of magnitude. [3] N.J. Wittenberg, C.L. Haynes, Using nanoparticles to push the limits of
detection, Wiley Interdiscipl. Rev. Nanomed. Nanobiotechnol. 1 (2009) 237
An afnity assay for bacterial cells using AuNP labeling and ICP-
254.
MS detection was described for the rst time by Li et al. [169]. The [4] J. Wang, Nanoparticle-based electrochemical bioassays of proteins,
target bacterial cells bound with the antibody to form afnity com- Electroanalysis 19 (2007) 769776.
plexes. This linked the cell, antibody, and AuNPs. The mixture was [5] M. Seydack, Nanoparticle labels in immunosensing using optical detection
methods, Biosens. Bioelectron. 20 (2005) 24542469.
centrifuged to separate the AuNPs bound to the cells from the [6] J. Wang, Nanomaterial-based amplied transduction of biomolecular
unbound AuNPs. The fraction of the cell-bound AuNPs was then interactions, Small 1 (2005) 10361043.
14 Gold nanoparticle-based signal amplication / X. Cao et al. / Anal. Biochem. 417 (2011) 116

[7] C.M. Niemeyer, Nanoparticles, proteins, and nucleic acids: biotechnology [34] Z. Zhong, W. Wu, D. Wang, D. Wang, J. Shan, Y. Qing, Z. Zhang, Nanogold-
meets materials science, Angew. Chem. Int. Ed. Engl. 40 (2001) 4128 enwrapped graphene nanocomposites as trace labels for sensitivity
4158. enhancement of electrochemical immunosensors in clinical immunoassays:
[8] P. Alivisatos, The use of nanocrystals in biological detection, Nat. Biotechnol. carcinoembryonic antigen as a model, Biosens. Bioelectron. 25 (2010) 2379
22 (2003) 4752. 2383.
[9] E. Katz, I. Willner, Integrated nanoparticlebiomolecule hybrid systems: [35] L. Ding, Q. Ji, R. Qian, W. Cheng, H. Ju, Lectin-based nanoprobes functionalized
synthesis, properties, and applications, Angew. Chem. Int. Ed. Engl. 43 (2004) with enzyme for highly sensitive electrochemical monitoring of dynamic
60426108. carbohydrate expression on living cells, Anal. Chem. 82 (2010) 12921298.
[10] E. Katz, I. Willner, J. Wang, Electroanalytical and bioelectroanalytical systems [36] Y. Li, H. Qi, Q. Gao, J. Yang, C. Zhang, Nanomaterial-amplied signal off/on
based on metal and semiconductor nanoparticles, Electroanalysis 16 (2004) electrogenerated chemiluminescence aptasensors for the detection of
1944. thrombin, Biosens. Bioelectron. 26 (2010) 754759.
[11] N.L. Rosi, C.A. Mirkin, Nanostructures in biodiagnostics, Chem. Rev. 105 [37] X.-Y. Yang, Y.-S. Guo, S. Bi, S.-S. Zhang, Ultrasensitive enhanced
(2005) 15471562. chemiluminescence enzyme immunoassay for the determination of a-
[12] C.A. Mirkin, R.L. Letsinger, R.C. Mucic, J.J. Storhoff, A DNA-based method for fetoprotein amplied by double-codied gold nanoparticles labels, Biosens.
rationally assembling nanoparticles into macroscopic materials, Nature 382 Bioelectron. 24 (2009) 27072711.
(1996) 607609. [38] M. Liu, C. Jia, Q. Jin, X. Lou, S. Yao, J. Xiang, J. Zhao, Novel colorimetric enzyme
[13] M.-C. Daniel, D. Astruc, Gold nanoparticles: assembly, supramolecular immunoassay for the detection of carcinoembryonic antigen, Talanta 81
chemistry, quantum-size-related properties, and applications toward (2010) 16251629.
biology, catalysis, and nanotechnology, Chem. Rev. 104 (2004) 293346. [39] D. Tian, C. Duan, W. Wang, H. Cui, Ultrasensitive electrochemiluminescence
[14] S.-J. Park, T.A. Taton, C.A. Mirkin, Array-based electrical detection of DNA with immunosensor based on luminol functionalized gold nanoparticle labeling,
nanoparticle probes, Science 295 (2002) 15031506. Biosens. Bioelectron. 25 (2010) 22902295.
[15] B. Tang, L. Cao, K. Xu, L. Zhuo, J. Ge, Q. Li, L. Yu, A new nanobiosensor for [40] D.-J. Kim, N.-E. Lee, J.-S. Park, I.-J. Park, J.-G. Kim, H.J. Cho, Organic
glucose with high sensitivity and selectivity in serum based on uorescence electrochemical transistor based immunosensor for prostate specic
resonance energy transfer (FRET) between CdTe quantum dots and Au antigen (PSA) detection using gold nanoparticles for signal amplication,
nanoparticles, Chemistry 14 (2008) 36373644. Biosens. Bioelectron. 25 (2010) 24772482.
[16] N.T.K. Thanh, Z. Rosenzweig, Development of an aggregation-based [41] A. de la Escosura-Muiz, A. Ambrosi, A. Merkoi, Electrochemical analysis
immunoassay for anti-protein A using gold nanoparticles, Anal. Chem. 74 with nanoparticle-based biosystems, Trends Anal. Chem. 27 (2008) 568584.
(2002) 16241628. [42] J. Wang, R. Polsky, D. Xu, Silver-enhanced colloidal gold electrochemical
[17] C.J. Ackerson, P.D. Jadzinsky, G.J. Jensen, R.D. Kornberg, Rigid, specic, and stripping detection of DNA hybridization, Langmuir 17 (2001) 57395741.
discrete gold nanoparticle/antibody conjugates, J. Am. Chem. Soc. 128 (2006) [43] J. Wang, D. Xu, R. Polsky, Magnetically-induced solid-state electrochemical
26352640. detection of DNA hybridization, J. Am. Chem. Soc. 124 (2002) 42084209.
[18] A. Merkoci, M. Aldavert, S. Marin, S. Alegret, New materials for [44] X. Chu, X. Fu, K. Chen, G. Shen, R. Yu, An electrochemical stripping
electrochemical sensing: V. Nanoparticles for DNA labeling, Trends Anal. metalloimmunoassay based on silver-enhanced gold nanoparticle label,
Chem. 24 (2005) 341349. Biosens. Bioelectron. 20 (2005) 18051812.
[19] D. Tian, C. Duan, W. Wang, N. Li, H. Zhang, H. Cui, Y. Lu, Sandwich-type [45] H. Guo, N. He, S. Ge, D. Yang, J. Zhang, MCM-41 mesoporous material
electrochemiluminescence immunosensor based on N-(aminobutyl)-N- modied carbon paste electrode for the determination of cardiac troponin I
ethylisoluminol labeling and gold nanoparticle amplication, Talanta 78 by anodic stripping voltammetry, Talanta 68 (2005) 6166.
(2009) 399404. [46] K.Y. Chumbimuni-Torres, Z. Dai, N. Rubinova, Y. Xiang, E. Pretsch, J. Wang, E.
[20] X.-B. Yin, B. Qi, X. Sun, X. Yang, E. Wang, 4-Dimethylamino butyric acid Bakker, Potentiometric biosensing of proteins with ultrasensitive ion-
labeling for electrochemiluminescence detection of biological substances by selective microelectrodes and nanoparticle labels, J. Am. Chem. Soc. 128
increasing sensitivity with gold nanoparticle amplication, Anal. Chem. 77 (2006) 1367613677.
(2005) 35253530. [47] A. de la Escosura-Muiz, M. Maltez-da Costa, A. Merkoi, Controlling the
[21] Y. Zhang, Z. Tang, J. Wang, H. Wu, A. Maham, Y. Lin, Hairpin DNA switch for electrochemical deposition of silver onto gold nanoparticles: reducing
ultrasensitive spectrophotometric detection of DNA hybridization based on interferences and increasing the sensitivity of magnetoimmuno assays,
gold nanoparticles and enzyme signal amplication, Anal. Chem. 82 (2010) Biosens. Bioelectron. 24 (2009) 24752482.
64406446. [48] I.-H. Min, L. Choi, K.-S. Ahn, B.K. Kim, B.Y. Lee, K.S. Kim, H.N. Choi, W.-Y. Lee,
[22] G. Jie, H. Huang, X. Sun, J.-J. Zhu, Electrochemiluminescence of CdSe quantum Electrochemical determination of carbohydrate-binding proteins using
dots for immunosensing of human prealbumin, Biosens. Bioelectron. 23 carbohydrate-stabilized gold nanoparticles and silver enhancement,
(2008) 18961899. Biosens. Bioelectron. 26 (2010) 13261331.
[23] J. Lin, C. He, L. Zhang, S. Zhang, Sensitive amperometric immunosensor for a- [49] Z. Li, C. Liu, Y. Fan, Y. Wang, X. Duan, A chemiluminescent
fetoprotein based on carbon nanotube/gold nanoparticle doped chitosan lm, metalloimmunoassay based on silver deposition on colloidal gold labels,
Anal. Biochem. 384 (2009) 130135. Anal. Biochem. 359 (2006) 247252.
[24] S. Zhang, F. Zheng, Z. Wu, G. Shen, R. Yu, Highly sensitive electrochemical [50] C.-H. Yeh, H.-H. Huang, T.-C. Chang, H.-P. Lin, Y.-C. Lin, Using an electro-
detection of immunospecies based on combination of Fc label and PPD microchip, a nanogold probe, and silver enhancement in an immunoassay,
lm/gold nanoparticle amplication, Biosens. Bioelectron. 24 (2008) 129 Biosens. Bioelectron. 24 (2009) 16611666.
135. [51] C.H. Yeh, Y.H. Chang, T.C. Chang, H.P. Lin, Y.C. Lin, Electro-microchip DNA-
[25] C. Xiang, Y. Zou, L. Sun, F. Xu, Direct electrochemistry and electrocatalysis of biosensor for bacteria detection, Analyst 135 (2010) 27172722.
cytochrome c immobilized on gold nanoparticleschitosancarbon [52] T.A. Taton, C.A. Mirkin, R.L. Lesinger, Scanometric DNA array detection with
nanotubes-modied electrode, Talanta 74 (2007) 206211. nanoparticle probes, Science 289 (2000) 17571760.
[26] J.-J. Zhang, F.-F. Cheng, T.-T. Zheng, J.-J. Zhu, Design and implementation of [53] Z. Wang, J. Lee, A.R. Cossins, M. Brust, Microarray-based detection of protein
electrochemical cytosensor for evaluation of cell surface carbohydrate and binding and functionality by gold nanoparticle probes, Anal. Chem. 77 (2005)
glycoprotein, Anal. Chem. 82 (2010) 35473555. 57705774.
[27] X. Liu, H. Wu, Y. Zheng, Z. Wu, J. Jiang, G. Shen, R. Yu, A sensitive [54] S.-Y. Hou, H.-K. Chen, H.-C. Cheng, C.-Y. Huang, Development of zeptomole
electrochemical immunosensor for a-fetoprotein detection with colloidal and attomolar detection sensitivity of biotinpeptide using a dotblot gold
gold-based dentritical enzyme complex amplication, Electroanalysis 22 nanoparticle immunoassay, Anal. Chem. 79 (2007) 980985.
(2010) 244250. [55] J. Gao, D. Liu, Z. Wang, Microarray-based study of carbohydrateprotein
[28] Y. Chai, D. Tian, W. Wang, H. Cui, A novel electrochemiluminescence strategy binding by gold nanoparticle probes, Anal. Chem. 80 (2008) 88228827.
for ultrasensitive DNA assay using luminol functionalized gold nanoparticles [56] C. Guarise, Gold nanoparticles-based protease assay, Proc. Natl. Acad. Sci. USA
multi-labeling and amplication of gold nanoparticles and biotin 103 (2006) 39783982.
streptavidin system, Chem. Commun. 46 (2010) 75607562. [57] M. Yang, C. Wang, Label-free immunosensor based on gold nanoparticle silver
[29] G. Li, X. Li, J. Wan, S. Zhang, Dendrimers-based DNA biosensors for highly enhancement, Anal. Biochem. 385 (2009) 128131.
sensitive electrochemical detection of DNA hybridization using reporter [58] Y.Y. Li, C. Zhang, B.S. Li, L.F. Zhao, X.B. Li, W.J. Yang, S.Q. Xu, Ultrasensitive
probe DNA modied with Au nanoparticles, Biosens. Bioelectron. 24 (2009) densitometry detection of cytokines with nanoparticle-modied aptamers,
32813287. Clin. Chem. 53 (2007) 10611066.
[30] A. Ambrosi, F. Air, A. Merkoi, Enhanced gold nanoparticle based ELISA for a [59] C.-H. Liang, C.-C. Wang, Y.-C. Lin, C.-H. Chen, C.-H. Wong, C.-Y. Wu, Iron
breast cancer biomarker, Anal. Chem. 82 (2010) 11511156. oxidegold coreshell nanoparticles for ultrasensitive detection of
[31] S. Bi, Y. Yan, X. Yang, S. Zhang, Gold nanolabels for new enhanced carbohydrateprotein interactions, Anal. Chem. 81 (2009) 77507756.
chemiluminescence immunoassay of a-fetoprotein based on magnetic [60] C.-H. Yeh, C.-Y. Hung, T.C. Chang, H.-P. Lin, Y.-C. Lin, An immunoassay using
beads, Chemistry 15 (2009) 47044709. antibodygold nanoparticle conjugate, silver enhancement, and atbed
[32] T. Selvaraju, J. Das, K. Jo, K. Kwon, C.-H. Huh, T.K. Kim, H. Yang, Nanocatalyst- scanner, Microuid. Nanouid. 6 (2008) 8591.
based assay using DNA-conjugated Au nanoparticles for electrochemical DNA [61] L. Ding, R. Qian, Y. Xue, W. Cheng, H. Ju, In situ scanometric assay of cell
detection, Langmuir 24 (2008) 98839888. surface carbohydrate by glyconanoparticle-aggregation-regulated silver
[33] R. Cui, H. Huang, Z. Yin, D. Gao, J.-J. Zhu, Horseradish peroxidase- enhancement, Anal. Chem. 82 (2010) 58045809.
functionalized gold nanoparticle label for amplied immunoanalysis based [62] Z. Zhang, C. Chen, X. Sheng Zhao, A simple and sensitive biosensor based on
on gold nanoparticles/carbon nanotubes hybrids modied biosensor, Biosens. silver enhancement of aptamergold nanoparticle aggregation,
Bioelectron. 23 (2008) 16661673. Electroanalysis 21 (2009) 13161320.
Gold nanoparticle-based signal amplication / X. Cao et al. / Anal. Biochem. 417 (2011) 116 15

[63] Y.C. Cao, R. Jin, C.A. Mirkin, Nanoparticles with Raman spectroscopic [92] L. Chen, H. Wei, Y. Guo, Z. Cui, Z. Zhang, X.-E. Zhang, Gold nanoparticle
ngerprints for DNA and RNA detection, Science 297 (2002) 15361540. enhanced immuno-PCR for ultrasensitive detection of Hantaan virus
[64] K. Faulds, W.E. Smith, D. Graham, Evaluation of surface-enhanced resonance nucleocapsid protein, J. Immunol. Methods 346 (2009) 6470.
Raman scattering for quantitative DNA analysis, Anal. Chem. 76 (2004) 412 [93] M.A.H. Muhammed, A.K. Shaw, S.K. Pal, T. Pradeep, Quantum clusters of gold
417. exhibiting FRET, J. Phys. Chem. C 112 (2008) 1432414330.
[65] X. Xu, D.G. Georganopoulou, H.D. Hill, C.A. Mirkin, Homogeneous detection of [94] P.C. Ray, G.K. Darbha, A. Ray, J. Walker, W. Hardy, Gold nanoparticle based
nucleic acids based upon the light scattering properties of silver-coated FRET for DNA detection, Plasmonics 2 (2007) 173183.
nanoparticle probes, Anal. Chem. 79 (2007) 66506654. [95] P.C. Ray, A. Fortner, G.K. Darbha, Gold nanoparticle based FRET assay for the
[66] D. Chiao, R. Shyu, C. Hu, H. Chiang, S. Tang, Colloidal gold-based detection of DNA cleavage, J. Phys. Chem. B 110 (2006) 2074520748.
immunochromatographic assay for detection of botulinum neurotoxin type [96] Y. Jin, H. Li, J. Bai, Homogeneous selecting of a quadruplex-binding ligand-
B, J. Chromatogr. B 809 (2004) 3741. based gold nanoparticle uorescence resonance energy transfer assay, Anal.
[67] J. Storhoff, Gold nanoparticle-based detection of genomic DNA targets on Chem. 81 (2009) 57095715.
microarrays using a novel optical detection system, Biosens. Bioelectron. 19 [97] Z. Dai, J. Zhang, Q. Dong, N. Guo, S. Xu, B. Sun, Y. Bu, Adaption of Au
(2004) 875883. nanoparticles and CdTe quantum dots in DNA detection, Chin. J. Chem. Eng.
[68] X. Mao, J. Jiang, Y. Luo, G. Shen, R. Yu, Copper-enhanced gold nanoparticle tags 15 (2007) 791794.
for electrochemical stripping detection of human IgG, Talanta 73 (2007) 420 [98] Y. Kim, S. Park, E. Oh, Y. Oh, H. Kim, On-chip detection of protein glycosylation
424. based on energy transfer between nanoparticles, Biosens. Bioelectron. 24
[69] W. Dungchai, W. Siangproh, W. Chaicumpa, P. Tongtawe, O. Chailapakul, (2009) 11891194.
Salmonella typhi determination using voltammetric amplication of [99] Y.-P. Kim, Y.-H. Oh, E. Oh, S. Ko, M.-K. Han, H.-S. Kim, Energy transfer-based
nanoparticles: a highly sensitive strategy for metalloimmunoassay based on multiplexed assay of proteases by using gold nanoparticle and quantum dot
a copper-enhanced gold label, Talanta 77 (2008) 727732. conjugates on a surface, Anal. Chem. 80 (2008) 46344641.
[70] X. Wei, A. Liang, S. Zhang, Z. Jiang, A selective resonance scattering assay for [100] R.C. Stringer, S. Schommer, D. Hoehn, S.A. Grant, Development of an optical
immunoglobulin G using Cu(II)ascorbic acidimmunonanogold reaction, biosensor using gold nanoparticles and quantum dots for the detection of
Anal. Biochem. 380 (2008) 223228. porcine reproductive and respiratory syndrome virus, Sens. Actuat. B 134
[71] G. Shen, Y. Zhang, Highly sensitive electrochemical stripping detection of (2008) 427431.
hepatitis B surface antigen based on copper-enhanced gold nanoparticle tags [101] S. Pihlasalo, L. Pellonper, E. Martikkala, P. Hnninen, H. Hrm, Sensitive
and magnetic nanoparticles, Anal. Chim. Acta 674 (2010) 2731. uorometric nanoparticle assays for cell counting and viability, Anal. Chem.
[72] B. Shlyahovsky, E. Katz, Y. Xiao, V. Pavlov, I. Willner, Optical and 82 (2010) 92829288.
electrochemical detection of NADH and of NAD+-dependent biocatalyzed [102] J. Zhang, L. Wang, H. Zhang, F. Boey, S. Song, C. Fan, Aptamer-based multicolor
processes by the catalytic deposition of copper on gold nanoparticles, Small 1 uorescent gold nanoprobes for multiplex detection in homogeneous
(2005) 213216. solution, Small 6 (2010) 201204.
[73] G. Xu, H. Li, X. Ma, X. Jia, J. Dong, W. Qian, A cuttlebone-derived matrix [103] S. Moon, D.J. Kim, K. Kim, D. Kim, H. Lee, K. Lee, S. Haam, Surface-enhanced
substrate for hydrogen peroxide/glucose detection, Biosens. Bioelectron. 25 plasmon resonance detection of nanoparticle-conjugated DNA hybridization,
(2009) 362367. Appl. Opt. 49 (2010) 484491.
[74] H. Cui, W. Wang, C.-F. Duan, Y.-P. Dong, J.-Z. Guo, Synthesis, characterization, [104] H. Joung, N. Lee, S. Lee, J. Ahn, Y. Shin, H. Choi, C. Lee, S. Kim, M. Kim, High
and electrochemiluminescence of luminol-reduced gold nanoparticles and sensitivity detection of 16S rRNA using peptide nucleic acid probes and a
their application in a hydrogen peroxide sensor, Chemistry 13 (2007) 6975 surface plasmon resonance biosensor, Anal. Chim. Acta 630 (2008) 168173.
6984. [105] S. Kinge, M. Crego-Calama, D.N. Reinhoudt, Self-assembling nanoparticles at
[75] W. Wang, T. Xiong, H. Cui, Fluorescence and electrochemiluminescence of surfaces and interfaces, ChemPhysChem 9 (2008) 2042.
luminol-reduced gold nanoparticles: photostability and platform effect, [106] H.-Y. Hsu, Y.-Y. Huang, RCA combined nanoparticle-based optical detection
Langmuir 24 (2008) 28262833. technique for protein microarray: a novel approach, Biosens. Bioelectron. 20
[76] R. Elghanian, J.J. Storhoff, R.C. Mucic, R.L. Letsinger, C.A. Mirkin, Selective (2004) 123126.
colorimetric detection of polynucleotides based on the distance-dependent [107] C. Huang, K. Bonroy, G. Reekman, K. Verstreken, L. Lagae, G. Borghs, An on-
optical properties of gold nanoparticles, Science 277 (1997) 10781081. chip localized surface plasmon resonance-based biosensor for label-free
[77] R.C. Mucic, J.J. Storhoff, C.A. Mirkin, R.L. Letsinger, DNA-directed synthesis of monitoring of antigenantibody reaction, Microelectron. Eng. 86 (2009)
binary nanoparticle network materials, J. Am. Chem. Soc. 120 (1998) 12674 24372441.
12675. [108] W.-C. Law, K.-T. Yong, A. Baev, R. Hu, P.N. Prasad, Nanoparticle enhanced
[78] J.J. Storhoff, A.A. Lazarides, R.C. Mucic, C.A. Mirkin, R.L. Letsinger, G.C. Schatz, surface plasmon resonance biosensing: application of gold nanorods, Opt.
What controls the optical properties of DNA-linked gold nanoparticle Express 17 (2009) 1904119046.
assemblies, J. Am. Chem. Soc. 122 (2000) 46404650. [109] B.-Y. Hsieh, Y.-F. Chang, M.-Y. Ng, W.-C. Liu, C.-H. Lin, H.-T. Wu, C. Chou,
[79] R.A.I. Reynolds, C.A. Mirkin, R.L. Letsinger, Homogeneous nanoparticle-based Localized surface plasmon coupled uorescence ber-optic biosensor with
quantitative colorimetric detection of oligonucleotides, J. Am. Chem. Soc. 122 gold nanoparticles, Anal. Chem. 79 (2007) 34873493.
(2000) 37953796. [110] N.J. Durr, T. Larson, D.K. Smith, B.A. Korgel, K. Sokolov, A. Ben-Yakar, Two-
[80] S.A. Claridge, A.J. Mastroianni, Y.B. Au, H.W. Liang, C.M. Micheel, J.M.J. Frchet, photon luminescence imaging of cancer cells using molecularly targeted gold
A.P. Alivisatos, Enzymatic ligation creates discrete multinanoparticle building nanorods, Nano Lett. 7 (2007) 941945.
blocks for self-assembly, J. Am. Chem. Soc. 130 (2008) 95989605. [111] S.-H. Guo, S.-J. Tsai, H.-C. Kan, D.-H. Tsai, M.R. Zachariah, R.J. Phaneuf, The
[81] C. Fang, Y. Fan, J. Kong, Z. Gao, N. Balasubramanian, Electrical detection of effect of an active substrate on nanoparticle-enhanced uorescence, Adv.
oligonucleotide using an aggregate of gold nanoparticles as a conductive tag, Mater. 20 (2008) 14241428.
Anal. Chem. 80 (2008) 93879394. [112] X. Qian, X. Zhou, S. Nie, Surface-enhanced Raman nanoparticle beacons based
[82] J. Li, S. Song, D. Li, Y. Su, Q. Huang, Y. Zhao, C. Fan, Multi-functional crosslinked on bioconjugated gold nanocrystals and long range plasmonic coupling, J.
Au nanoaggregates for the amplied optical DNA detection, Biosens. Am. Chem. Soc. 130 (2008) 1493414935.
Bioelectron. 24 (2009) 33113315. [113] D.G. Thompson, R.J. Stokes, R.W. Martin, P.J. Lundahl, K. Faulds, D. Graham,
[83] B. Du, Z. Li, Y. Cheng, Homogeneous immunoassay based on aggregation of Synthesis of unique nanostructures with novel optical properties using
antibody-functionalized gold nanoparticles coupled with light scattering oligonucleotide mixed-metal nanoparticle conjugates, Small 4 (2008) 1054
detection, Talanta 75 (2008) 959964. 1057.
[84] Y. Liu, Y. Liu, R.L. Mernaugh, X. Zeng, Single chain fragment variable [114] D.S. Grubisha, R.J. Lipert, H.-Y. Park, J. Driskell, M.D. Porter, Femtomolar
recombinant antibody functionalized gold nanoparticles for a highly detection of prostate-specic antigen: an immunoassay based on surface
sensitive colorimetric immunoassay, Biosens. Bioelectron. 24 (2009) 2853 enhanced Raman scattering and immunogold labels, Anal. Chem. 75 (2003)
2857. 59365943.
[85] J.M. Nam, Nanoparticle-based bio-bar codes for the ultrasensitive detection of [115] D. Graham, D.G. Thompson, W.E. Smith, K. Faulds, Control of enhanced
proteins, Science 301 (2003) 18841886. Raman scattering using a DNA-based assembly process of dye-coded
[86] J.-M. Nam, S.I. Stoeva, C.A. Mirkin, Bio-bar-code-based DNA detection with nanoparticles, Nat. Nanotechnol. 3 (2008) 548551.
PCR-like sensitivity, J. Am. Chem. Soc. 126 (2004) 59325933. [116] X. Liu, Q. Dai, L. Austin, J. Coutts, G. Knowles, J. Zou, H. Chen, Q. Huo, A one-
[87] D.G. Georganopoulou, Nanoparticle-based detection in cerebral spinal uid of step homogeneous immunoassay for cancer biomarker detection using gold
a soluble pathogenic biomarker for Alzheimers disease, Proc. Natl. Acad. Sci. nanoparticle probes coupled with dynamic light scattering, J. Am. Chem. Soc.
USA 102 (2005) 22732276. 130 (2008) 27802782.
[88] J.-M. Nam, S.-J. Park, C.A. Mirkin, Bio-barcodes based on oligonucleotide- [117] X. Liu, Q. Huo, A washing-free and amplication-free one-step homogeneous
modied nanoparticles, J. Am. Chem. Soc. 124 (2002) 38203821. assay for protein detection using gold nanoparticle probes and dynamic light
[89] Y.P. Bao, T.-F. Wei, P.A. Lefebvre, H. An, L. He, G.T. Kunkel, U.R. Mller, scattering, J. Immunol. Methods 349 (2009) 3844.
Detection of protein analytes via nanoparticle-based bio bar code technology, [118] J.-Q. Zhang, Y.-S. Wang, Y. He, T. Jiang, H.-M. Yang, X. Tan, R.-H. Kang, Y.-K.
Anal. Chem. 78 (2006) 20552059. Yuan, L.-F. Shi, Determination of urinary adenosine using resonance light
[90] C.S. Thaxton, H.D. Hill, D.G. Georganopoulou, S.I. Stoeva, C.A. Mirkin, A bio- scattering of gold nanoparticles modied structure-switching aptamer, Anal.
bar-code assay based upon dithiothreitol-induced oligonucleotide release, Biochem. 397 (2010) 212217.
Anal. Chem. 77 (2005) 81748178. [119] Z. Li, X. Duan, C. Liu, B. Du, Selective determination of cysteine by resonance
[91] J.-M. Nam, A.R. Wise, J.T. Groves, Colorimetric bio-barcode amplication light scattering technique based on self-assembly of gold nanoparticles, Anal.
assay for cytokines, Anal. Chem. 77 (2005) 69856988. Biochem. 351 (2006) 1825.
16 Gold nanoparticle-based signal amplication / X. Cao et al. / Anal. Biochem. 417 (2011) 116

[120] X. Long, Q. Miao, S. Bi, D. Li, C. Zhang, H. Zhao, Resonance Rayleigh scattering [146] A.J. Bard, L.R. Faulkner (Eds.), Electrochemical Methods: Fundamentals and
method for the recognition and determination of double-stranded DNA using Applications, John Wiley, New York, 1980.
amikacin, Talanta 64 (2004) 366372. [147] A. Bonannia, M.J. Esplandiua, M. del Valle, Signal amplication for
[121] Z. Jiang, S. Sun, A. Liang, W. Huang, A. Qin, Gold-labeled nanoparticle-based impedimetric genosensing using goldstreptavidin nanoparticles,
immunoresonance scattering spectral assay for trace apolipoprotein AI and Electrochim. Acta 53 (2008) 40224029.
apolipoprotein B, Clin. Chem. 52 (2006) 13891394. [148] Y. Zhang, H. Wang, J. Nie, Y. Zhang, G. Shen, R. Yu, Individually addressable
[122] S. Ko, T.J. Park, H.-S. Kim, J.-H. Kim, Y.-J. Cho, Directed self-assembly of gold microelectrode arrays fabricated with gold-coated pencil graphite particles
binding polypeptideprotein A fusion proteins for development of gold for multiplexed and high sensitive impedance immunoassays, Biosens.
nanoparticle-based SPR immunosensors, Biosens. Bioelectron. 24 (2009) Bioelectron. 25 (2009) 3440.
25922597. [149] S. Pinijsuwan, P. Rijiravanich, M. Somasundrum, W. Surareungchai, Sub-
[123] X. Huang, H. Tu, D. Zhu, D. Du, A. Zhang, A gold nanoparticle labeling strategy femtomolar electrochemical detection of DNA hybridization based on latex
for the sensitive kinetic assay of the carbamateacetylcholinesterase gold nanoparticle-assisted signal amplication, Anal. Chem. 80 (2008) 6779
interaction by surface plasmon resonance, Talanta 78 (2009) 10361042. 6784.
[124] L. Olofsson, T. Rindzevicius, I. Pfeiffer, M. Kll, F. Hk, Surface-based gold- [150] D. Tang, R. Yuan, Y. Chai, Biochemical and immunochemical characterization
nanoparticle sensor for specic and quantitative DNA hybridization of the antigenantibody reaction on a non-toxic biomimetic interface
detection, Langmuir 19 (2003) 1041410419. immobilized red blood cells of crucian carp and gold nanoparticles,
[125] J.S. Mitchell, T.E. Lowe, Ultrasensitive detection of testosterone using Biosens. Bioelectron. 22 (2007) 11161120.
conjugate linker technology in a nanoparticle-enhanced surface plasmon [151] S. Wu, Z. Zhong, D. Wang, M. Li, Y. Qing, N. Dai, Z. Li, Gold nanoparticle-
resonance biosensor, Biosens. Bioelectron. 24 (2009) 21772183. labeled detection antibodies for use in an enhanced electrochemical
[126] W. Lu, Y. Jin, G. Wang, D. Chen, J. Li, Enhanced photoelectrochemical method immunoassay of hepatitis B surface antigen in human serum, Microchim.
for linear DNA hybridization detection using Au-nanoparticle labeled DNA as Acta 166 (2009) 269275.
probe onto titanium dioxide electrode, Biosens. Bioelectron. 23 (2008) 1534 [152] Y. Zhuo, P.-X. Yuan, R. Yuan, Y.-Q. Chai, C.-L. Hong, Bienzyme functionalized
1539. three-layer composite magnetic nanoparticles for electrochemical
[127] H.-H. Cai, P.-H. Yang, J. Feng, J. Cai, Immunoassay detection using immunosensors, Biomaterials 30 (2009) 22842290.
functionalized gold nanoparticle probes coupled with resonance Rayleigh [153] J. Das, M.A. Aziz, H. Yang, A nanocatalyst-based assay for proteins: DNA-free
scattering, Sens. Actuat. B 135 (2009) 603609. ultrasensitive electrochemical detection using catalytic reduction of p-
[128] C.X. Zhang, Y. Zhang, X. Wang, Z.M. Tang, Z.H. Lu, Hyper-Rayleigh scattering nitrophenol by gold-nanoparticle labels, J. Am. Chem. Soc. 128 (2006)
of protein-modied gold nanoparticles, Anal. Biochem. 320 (2003) 136140. 1602216023.
[129] S. Thobhani, S. Attree, R. Boyd, N. Kumarswami, J. Noble, M. Szymanski, R.A. [154] K.-Z. Liang, J.-S. Qi, W.-J. Mu, Z.-G. Chen, Biomolecules/gold nanowires-doped
Porter, Bioconjugation and characterisation of gold colloid-labelled proteins, solgel lm for label-free electrochemical immunoassay of testosterone, J.
J. Immunol. Methods 356 (2010) 6069. Biochem. Biophys. Methods 70 (2008) 11561162.
[130] L. Rodriguez-Lorenzo, R.A. Alvarez-Puebla, I. Pastoriza-Santos, S. Mazzucco, O. [155] W. Liang, W. Yi, S. Li, R. Yuan, A. Chen, S. Chen, G. Xiang, C. Hu, A novel, label-
Stephan, M. Kociak, L.M. Liz-Marzan, F.J.G. de Abajo, Zeptomol detection free immunosensor for the detection of a-fetoprotein using functionalised
through controlled ultrasensitive surface-enhanced Raman scattering, J. Am. gold nanoparticles, Clin. Biochem. 42 (2009) 15241530.
Chem. Soc. 131 (2009) 46164618. [156] X. He, R. Yuan, Y. Chai, Y. Shi, A sensitive amperometric immunosensor for
[131] P. Scodeller, V. Flexer, R. Szamocki, E.J. Calvo, N. Tognalli, H. Troiani, A. carcinoembryonic antigen detection with porous nanogold lm and nano-Au/
Fainstein, Wired-enzyme coreshell Au nanoparticle biosensor, J. Am. Chem. chitosan composite as immobilization matrix, J. Biochem. Biophys. Methods
Soc. 130 (2008) 1269012697. 70 (2008) 823829.
[132] G. Zheng, W.L. Daniel, C.A. Mirkin, A new approach to amplied telomerase [157] T. Selvaraju, J. Das, S.W. Han, H. Yang, Ultrasensitive electrochemical
detection with polyvalent oligonucleotide nanoparticle conjugates, J. Am. immunosensing using magnetic beads and gold nanocatalysts, Biosens.
Chem. Soc. 130 (2008) 96449645. Bioelectron. 23 (2008) 932938.
[133] D. Kim, W.L. Daniel, C.A. Mirkin, Microarray-based multiplexed scanometric [158] D. Tang, R. Niessner, D. Knopp, Flow-injection electrochemical immunosensor
immunoassay for protein cancer markers using gold nanoparticle probes, for the detection of human IgG based on glucose oxidase-derivated
Anal. Chem. 81 (2009) 91839187. biomimetic interface, Biosens. Bioelectron. 24 (2009) 21252130.
[134] K. Glynou, P.C. Ioannou, T.K. Christopoulos, V. Syriopoulou, Oligonucleotide- [159] R. Liang, J. Qiu, P. Cai, A novel amperometric immunosensor based on three-
functionalized gold nanoparticles as probes in a dry-reagent strip biosensor dimensional solgel network and nanoparticle self-assemble technique, Anal.
for DNA analysis by hybridization, Anal. Chem. 75 (2003) 41554160. Chim. Acta 534 (2005) 223229.
[135] D.H. Choi, S.K. Lee, Y.K. Oh, B.W. Bae, S.D. Lee, S. Kim, Y.-B. Shin, M.-G. Kim, A [160] J. Zheng, W. Feng, L. Lin, F. Zhang, G. Cheng, P. He, Y. Fang, A new
dual gold nanoparticle conjugate-based lateral ow assay (LFA) method for amplication strategy for ultrasensitive electrochemical aptasensor with
the analysis of troponin I, Biosens. Bioelectron. 25 (2010) 19992002. network-like thiocyanuric acidgold nanoparticles, Biosens. Bioelectron. 23
[136] J. Li, T. Deng, X. Chu, R. Yang, J. Jiang, G. Shen, R. Yu, Rolling circle (2007) 341347.
amplication combined with gold nanoparticle aggregates for highly [161] X. Liu, D. Wong, Picogram-detection of estradiol at an electrochemical
sensitive identication of single-nucleotide polymorphisms, Anal. Chem. 82 immunosensor with a gold nanoparticle|protein G(LCSPDP)-scaffold,
(2010) 28112816. Talanta 77 (2009) 14371443.
[137] W. Xu, X. Xue, T. Li, H. Zeng, X. Liu, Ultrasensitive and selective colorimetric [162] L. Zhang, R. Yuan, Y. Chai, X. Li, Investigation of the electrochemical and
DNA detection by nicking endonuclease assisted nanoparticle amplication, electrocatalytic behavior of positively charged gold nanoparticle and L-
Angew. Chem. Int. Ed. Engl. 48 (2009) 68496852. cysteine lm on an Au electrode, Anal. Chim. Acta 596 (2007) 99105.
[138] P.-C. Soo, Y.-T. Horng, K.-C. Chang, J.-Y. Wang, P.-R. Hsueh, C.-Y. Chuang, C.-C. [163] G.-J. Yang, J.-L. Huang, W.-J. Meng, M. Shen, X.-A. Jiao, A reusable capacitive
Lu, H.-C. Lai, A simple gold nanoparticle probes assay for identication of immunosensor for detection of Salmonella spp. based on grafted ethylene
Mycobacterium tuberculosis and Mycobacterium tuberculosis complex from diamine and self-assembled gold nanoparticle monolayers, Anal. Chim. Acta
clinical specimens, Mol. Cell. Probes 23 (2009) 240246. 647 (2009) 159166.
[139] V. Wiwanitkit, A. Seereemaspun, R. Rojanathanes, Increasing the [164] S. Zhang, F. Huang, B. Liu, J. Ding, X. Xu, J. Kong, A sensitive impedance
agglutination reaction in slide test for weak B blood group by gold immunosensor based on functionalized gold nanoparticleprotein composite
nanoparticle solution: the rst world report, J. Immunol. Methods 328 lms for probing apolipoprotein A-I, Talanta 71 (2007) 874881.
(2007) 201203. [165] J. Wang, J.A. Prott, M.J. Pugia, I.I. Suni, Au nanoparticle conjugation for
[140] M. Pumera, S. Snchez, I. Ichinose, J. Tang, Electrochemical nanobiosensors, impedance and capacitance signal amplication in biosensors, Anal. Chem.
Sens. Actuat. B 123 (2007) 11951205. 78 (2006) 17691773.
[141] M.T. Castaeda, A. Merkoi, M. Pumera, S. Alegret, Electrochemical [166] V.I. Baranov, Z. Quinn, D.R. Bandura, S.D. Tanner, A sensitive and quantitative
genosensors for biomedical applications based on gold nanoparticles, element-tagged immunoassay with ICPMS detection, Anal. Chem. 74 (2002)
Biosens. Bioelectron. 22 (2007) 19611967. 16201636.
[142] K. Kerman, M. Chikae, S. Yamamura, E. Tamiya, Gold nanoparticle-based [167] C. Zhang, Z. Zhang, B. Yu, J. Shi, X. Zhang, Application of the biological
electrochemical detection of protein phosphorylation, Anal. Chim. Acta 588 conjugate between antibody and colloid Au nanoparticles as analyte to
(2007) 2633. inductively coupled plasma mass spectrometry, Anal. Chem. 74 (2002) 96
[143] M. Dequaire, C. Degrand, B. Limoges, An electrochemical metalloimmunoassay 99.
based on a colloidal gold label, Anal. Chem. 72 (2000) 55215528. [168] Q. Zhao, X. Lu, C.-G. Yuan, X.-F. Li, X.C. Le, Aptamer-linked assay for thrombin
[144] L. Authier, C. Grossiord, P. Brossier, Gold nanoparticle-based quantitative using goldmass spectrometry detection, Anal. Chem. 81 (2009) 74847489.
electrochemical detection of amplied human cytomegalovirus DNA using [169] F. Li, Q. Zhao, C. Wang, X. Lu, X.-F. Li, X.C. Le, Detection of Escherichia coli
disposable microband electrodes, Anal. Chem. 73 (2001) 44504456. O157H7 using gold nanoparticle labeling and inductively coupled plasma
[145] K. Liao, H. Huang, Femtomolar immunoassay based on coupling gold mass spectrometry, Anal. Chem. 82 (2010) 33993403.
nanoparticle enlargement with square wave stripping voltammetry, Anal.
Chim. Acta 538 (2005) 159164.

S-ar putea să vă placă și