Вы находитесь на странице: 1из 26

+Model

NANTOD-359; No. of Pages 26 ARTICLE IN PRESS


Nano Today (2014) xxx, xxx—xxx

Available online at www.sciencedirect.com

ScienceDirect

journal homepage: www.elsevier.com/locate/nanotoday

REVIEW

Metal nanoclusters: New fluorescent probes


for sensors and bioimaging
Libing Zhang a,b, Erkang Wang a,b,∗

a
State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese
Academy of Sciences, Changchun 130022, Jilin, China
b
University of Chinese Academy of Sciences, Beijing 100049, China

Received 14 December 2013; received in revised form 11 February 2014; accepted 12 February 2014

KEYWORDS Summary Fluorescent metal nanoclusters (NCs) as a new class of fluorophores have attracted
Metal nanocluster; more and more attention due to their unique electronic structures and the subsequent unusual
Fluorescent probe; physical and chemical properties. The size of metal NCs approaches the Fermi wavelength
Biolabels; of electrons, between metal atoms and nanoparticles, resulting in molecule-like properties
Sensor; including discrete energy levels, size-dependent fluorescence, good photostability and bio-
Imaging compatibility. These excellent properties make them ideal fluorescent probes for biological
application. Up to now, significant efforts have been devoted to the synthesis, property and
application studies of gold and silver NCs. Recently, a growing number of studies on copper and
other metal clusters have also been reported. In this review article, we focus on summariz-
ing recent advances in controllable synthesis strategies, chemical and optical properties, and
sensing and imaging applications of metal NCs (mainly including Au, Ag, Cu, etc.). Finally, we
conclude with a look at the future challenges and prospects of the future development of metal
NCs.
© 2014 Elsevier Ltd. All rights reserved.

Introduction

Nanomaterials have been recognized as the most modish


research topics in the past decades [1—3]. Nobel metal

nanomaterials with interesting size-dependent electrical,
Corresponding author at: State Key Laboratory of Electroanalyt-
optical, magnetic, and chemical properties have been inten-
ical Chemistry, Changchun Institute of Applied Chemistry, Chinese
sively pursued, not only for their fundamental scientific
Academy of Sciences, Changchun 130022, Jilin, China.
Fax: +86 431 85689711. interest, but also for their many technological applications
E-mail addresses: ekwang@ciac.ac.cn, ekwang@ciac.jl.cn [4—6]. Especially, metal nanoclusters (NCs) have attracted
(E. Wang). special attention due to their attractive features and

http://dx.doi.org/10.1016/j.nantod.2014.02.010
1748-0132/© 2014 Elsevier Ltd. All rights reserved.

Please cite this article in press as: L. Zhang, E. Wang, Nano Today (2014), http://dx.doi.org/10.1016/j.nantod.2014.02.010
+Model
NANTOD-359; No. of Pages 26 ARTICLE IN PRESS
2 L. Zhang, E. Wang

molecule-like properties [7—16]. Metal NCs usually consist a sub-nanometer size and size or scaffold-dependent tun-
of a few to a hundred atoms, and the sizes are comparable able fluorescence. They have an appealing set of features
to the Ferimi wavelength of electrons [7], which endows that complements the conventional fluorophores. These
them an important role-the missing link between single properties establish them as a new class of ultra-small, bio-
metal atoms and plasmonic metal nanoparticles. In this compatible fluorophores for applications as biological labels
size regime, the continuous density of states breaks up into or optoelectronics emitters.
discrete energy levels [16,17]. Due to the electrons of metal It should be pointed out that several excellent review
atoms confined in molecular dimensions and the special dis- papers have been dedicated to the metal NCs [8—14]. How-
crete energy levels, metal NCs exhibit dramatically different ever, these previous reviews focused on either one kind of
optical, electronical and chemical properties, including metal or metal NCs with actually a wide size range and
strong photoluminescence, excellent photostability, good different focusing aspects. In this article, we will mainly
biocompatibility and sub-nanometer size. Such novel prop- summarize recent advances in the synthesis, special prop-
erties make metal NCs an ideal nanomaterial for promise erties and the applications of metal NCs (Au, Ag, Cu, etc.)
applications in biological analysis and imaging, environmen- as new fluorescent probes for analytical sensing and biologi-
tal monitoring, industrial catalysis and electronic devices. cal imaging. In the final section, we will give a brief outlook
Fluorescent metal NCs have been developed as a new on the challenges and opportunities for future metal NCs
class of fluorophores. Current fluorescence applications research.
mostly involve organic fluorophores, semiconductor quan-
tum dots (QDs) [18] or fluorescent proteins [19]. Organic
fluorophores exist in a wide range of chemical struc- Energy levels: from bulk metals to
tures and spectral properties; however, they are prone to nanoclusters
photobleaching, which may limit their many applications.
Semiconductor QDs are fluorescence-tunable and photo- The physical and chemical properties of metals depend
stable; however, their large physical size may hinder their greatly on their size. With the varying size, their behaviors
use as fluorescent reporters of binding events, which may go through several noticeable transitions (Fig. 1) [17]. Bulk
compromise their use for in vivo applications. Fluores- metals are good optical reflectors and electrical conductors.
cent proteins are genetically encodable, which can be The electronic situation in bulk metals is characterized by
produced by the cells and organisms themselves. Conse- the existence of energy bands. They result from the com-
quently, there is no need for additional labeling and/or bination of an infinite number of energetically very similar
other chemical procedures in studies of live cells and organ- orbitals. The valence band contains the relevant valence
isms. Metal NCs show strong photoluminescence, combined electrons. The conduction band of metals overlaps to some
with good photostability and high emission rates, and have extent with the valence band and so becomes partially

Figure 1 The effect of size on metals. Whereas bulk metal and metal nanoparticles have a continuous band of energy levels,
the limited number of atoms in metal nanoclusters results in discrete energy levels, allowing interaction with light by electronic
transitions between energy levels. Metal nanoclusters bridge the gap between single atoms and nanoparticles.
Reprinted from Ref. [17] with permission by Springer-Verlag.

Please cite this article in press as: L. Zhang, E. Wang, Nano Today (2014), http://dx.doi.org/10.1016/j.nantod.2014.02.010
+Model
NANTOD-359; No. of Pages 26 ARTICLE IN PRESS
Metal nanoclusters 3

occupied with electrons. These electrons are finally respon- method, Au3+ ions are reduced in the presence of a suitable
sible for the electric conductivity of metals. In contrast to ligand to form the clusters. Through this strategy, fluores-
the electrons in a filled band, those in the conduction band cence emission of formed Au NCs could be effectively tuned
are fully mobile and make conductivity possible. In bulk from blue to near-IR regimes. A group of water-soluble
metals, the energy levels of the electrons are continuous. glutathione-protected Au clusters with defined chemical
When the size of metals reduces to become metal compositions have been synthesized by reducing Au3+ ions
nanoparticles, the motion of electrons becomes limited by in the presence of glutathione [21,22,26]. In addition, the
the size of the nanoparticle and interactions are expected alkanethiol-bound Au NCs with various emission wavelengths
to be mostly with the surface. Metal nanoparticles will dis- also have been prepared in the presence of different chain
play intense colors due to surface plasmon resonance effect, lengths of alkanethiols [27—29]. In similar approaches, many
in which the optical properties are determined by the col- other thiols such as tiopronin [30], phenylethylthiolate [31],
lective oscillation of conduction electrons upon interaction and D-penicillamine [32] all have been used to prepare
with light. Usually, metal nanoparticles absorb light strongly, monolayer-protected Au NCs.
but they do not or only show weak luminescence. Jin group developed a kinetically controlled method for
Metal nanoparticles further reduced in size, they will synthesizing high purity Au25 (SR)18 (where, SR = thiolate)
change to metal NCs. At this stage, the properties of parti- NCs in high yield [33]. An interesting ‘‘size focusing’’
cles disappear and the bands turn into more or less discrete method was discovered [34], which has been demonstrated
energy levels. Metal NCs are not conductors any more, as to be versatile for different thiolate ligands, including 2-
the energy levels are too far separated. Thus, the collec- phenylethanethiol (—SC2 H4 Ph), dodecanethiol (—SC12 H25 ),
tive oscillation of electrons is obstructed and metal NCs do and glutathione (—SG) [34], as well as for the different-sized
not give rise to surface plasmon resonance effect. However, Aun (SR)m NCs [35]. The fluorescence of Au25 (SR)18 clusters
they will follow quantum mechanical rules. Through interac- (R = C6 H13 (hexanethiol), C2 H4 Ph, glutathiolate, etc.) was
tion with light and then electronic transitions between the found to be affected by the ligands and metal core charge
energy levels, they will show bright luminescence. state [36]. Bigioni et al. observed photoluminescence in the
near-infrared region (1100—1600 nm) for 1.1 and 1.7 nm par-
Synthesis of metal nanoclusters ticles [37]. Wu et al. found that the surface of Au25 (SR)18
played a major role in the fluorescence generation [36].
Specifically, the ligands with electron rich atoms (e.g., O,
The controllable synthesis of metal NCs with high quality
N) or groups (e.g., —COOH, —NH2 ) can considerably enhance
is of paramount importance and highly desirable. To date,
fluorescence. Recently, Jin group reported a one-pot synthe-
in order to obtain high-quality metal NCs, some key factors
sis of Au NCs using captopril (Capt) as protected ligand [38].
should be taken into consideration. (1) The ligand should
The obtained Au25 (Capt)18 exhibited high thermal stability,
have strong interaction with metal NCs. (2) The reducing
which should be attributed to the ligand stability of capto-
condition should be strict. Generally, strong reducing agents
pril. The chiral ligands (Capt) give rise to distinct chiroptical
or light irradiation or sonication should be employed to
features of Au NCs. Those excellent properties of Au25 (SR)18
improve the quantum yield (QY) of NCs. (3) Long aging time
NCs render this material quite promising for biological appli-
is also important for obtaining high-quality NCs. Consider-
cations.
ing these crucial factors, different approaches have been
devised to synthesize high-quality metal NCs with various
scaffolds in a controllable manner. The scaffolds typically
have multiple groups that allow strong interaction with
metal ions, which plays a crucial role in determining some
of the properties of the clusters. The scaffolds influence the
geometry of the clusters and also provide suitable conditions
to avoid aggregation of the clusters into large nanoparti-
cles [20]. They potentially alter the electronic structure
and hence significantly influence their properties. The used
scaffolds for the synthesis of clusters usually include small
thiol molecules, DNA, peptides and proteins, dendrimers and
polymers. To date, significant efforts have been devoted to
the studies of gold and silver NCs. More recently, a grow-
ing number of studies on copper and other metal NCs have
also been reported. Accordingly, we will discuss the vari-
ous synthesis strategies for Au, Ag, Cu and other metal NCs,
respectively.

Au nanoclusters
Figure 2 Excitation (dashed) and emission (solid) spectra of
In the aspects of Au NCs, small thiol molecules have firstly different gold nanoclusters. Excitation and emission maxima
been employed to synthesize Au NCs via chemical reduc- shift to longer wavelength with increasing nanocluster size.
tion of Au precursor in the presence of NaBH4 owing to Reprinted from Ref. [40] with permission by the American Phys-
the strong interaction between Au and S [21—25]. In this ical Society.

Please cite this article in press as: L. Zhang, E. Wang, Nano Today (2014), http://dx.doi.org/10.1016/j.nantod.2014.02.010
+Model
NANTOD-359; No. of Pages 26 ARTICLE IN PRESS
4 L. Zhang, E. Wang

Dendrimers are repetitively branched molecules with spectrometry, they were able to identify the sizes of differ-
well-defined molecular weights and small cavities, which ent emissive Au NCs. As shown in Fig. 2, emissions at 3.22,
have been employed to synthesize Au NCs. For example, 2.72, 2.43, 1.65, and 1.41 eV correspond to Au5 , Au8 , Au13 ,
poly(amidoamine) dendrimer (PAMAM) was used as tem- Au23 and Au31 , respectively.
plates to encapsulate few-atom Au NCs [39]. After gold ions Fluorescent Au NCs could be prepared through etch-
were introduced into the solution of PAMAM, an equivalent ing metallic nanoparticles using large amounts of suitably
of NaBH4 was added into the solution and some reduced selected ligands. Pradeep group has presented two possi-
gold atoms aggregated within the dendrimers to form very ble mechanisms regarding the formation of clusters [41].
small dendrimer-encapsulated Au NCs. Once the large NPs In the first route, Au atoms are removed from the surface
were removed through centrifugation, luminescent Au NCs of nanoparticles by excess ligands as a gold(I)—ligand com-
were obtained, which exhibited bright blue emission and plex, which may undergo strong aurophilic interaction and
two-order-of-magnitude improved fluorescence QY (42%) in then to form the clusters. In another route, ligand may
aqueous solution. Subsequently, to further understand size- etch the surface gold atoms of the nanoparticles leading to
dependent emission from Au NCs, Zheng et al. tuned molar reduction in the size of the nanoparticles in steps, resulting
ratios between dendrimer and gold ions as well as the in the formation of clusters. For example, two fluorescent
amount of NaBH4 used in the reactions, and a class of Au Au NCs have been synthesized from mercaptosuccinic acid
NCs with different emission maxima ranging from UV to IR (MSA)-protected gold nanoparticles by etching with excess
were obtained [40]. Using electrospray ionization (ESI) mass glutathione [41]. Etching at pH 3 was yielded Au25 , while

Figure 3 (a) General strategy to fabricate water-soluble fluorescent Au nanoclusters. DDAB-stabilized gold nanoparticles (Au
NP@DDAB) are etched by the addition of Au precursors (HAuCl4 or AuCl3 ) to smaller nanoclusters (Au NC@DDAB). By the addition of
reducing agent (TBAB) the Au NCs grow again reversibly into bigger Au NPs. The hydrophobic Au NC@DDAB become water soluble
upon ligand exchange with dihydrolipoic acid (Au NC@DHLA); (b) TEM images of Au NP@DDAB, Au NC@DDAB, and Au NC@DHLA; 100
particles were randomly selected for measuring the size distribution, resulting in 5.55 ± 0.68 nm, 3.17 ± 0.35 nm, and 1.56 ± 0.3 nm
in diameter, respectively. (c) Pictures of particle solutions under daylight. Contrary to Au NP@DDAB solution which features the
red color of surface plasmon absorption, Au NC@DDAB and Au NC@DHLA display a colorless and brown translucent solution without
plasmon absorption, respectively. (d) Pictures of the same particle solutions under UV excitation. The Au NC@DHLA solution shows
red photoluminescence.
Reprinted from Ref. [43] with permission by American Chemical Society.

Please cite this article in press as: L. Zhang, E. Wang, Nano Today (2014), http://dx.doi.org/10.1016/j.nantod.2014.02.010
+Model
NANTOD-359; No. of Pages 26 ARTICLE IN PRESS
Metal nanoclusters 5

that at pH 7—8 it was Au8 . Nie group reported a green demonstrated the synthesis of AuAg NCs clusters with tune-
fluorescent Au NCs was synthesized by using multivalent able compositions in a BSA template starting from Au and
polyethylenimine (PEI) to etch colloidal gold nanoparti- Ag clusters [50]. The results revealed that the alloy cluster
cles [42]. In addition, didodecyldimethylammonium bromide core of ∼1.2 nm diameter is composed of nearly zero-valent
(DDAB) stabilized Au nanoparticle was brought to aqueous Au and Ag atoms that exhibit distinctly different steady
phase upon ligand exchange with dihydrolipoic acid (DHLA) state and time resolved excited state luminescence profiles
[43], resulting in the formation of brightly red emitting compared to the parent clusters. They believe that alloy
Au NCs (Fig. 3). The resulting Au NC@DHLA particles have clusters with the combined properties of the constituents
a QY of around 1—3%, reduced photobleaching compared in versatile protein templates would have potential appli-
to organic fluorophores, and very good colloidal stability. cations in the future. In addition, horseradish peroxidase
Interestingly, an already synthesized Au NCs also could be [51], insulin [52], pepsin [53] and transferrin protein [54,55]
converted to another Au NCs through the ligand exchange all have been used as scaffolds for the preparation of Au
reaction. A phosphine-stabilized Au11 cluster was treated NCs, respectively. These functionalized Au NCs all exhibit
with water soluble thiol glutathione, the glutathione pro- excellent properties and great potential for biological appli-
tected Au25 were obtained [44] and the subsequent research cations.
confirms that Au25 have the more extraordinary thermody- Recently, DNA has also been used as template for the
namic stability [45]. Similarly, bright-red-emitting Au23 NCs preparation of fluorescent Au NCs. DNA template synthesis
were synthesized from Au25 NCs [46] and Au75 NCs were has several advantages. First, DNA is non-toxic with good
obtained through the reaction of phosphine-protected Au55 biocompatibility. Second, DNA possesses functional roles
NCs with hexanethiol and other thiol [47]. for molecular recognition (e.g. aptamers), it is possible to
Biologically important molecules also can act as tem- directly couple analyte binding with fluorescence signal-
plates for the synthesis of highly fluorescent Au NCs. The ing through designing appropriate DNA sequences. Third,
use of bio-ligand can effectively reduce the possible toxicity chemical synthesized DNA can be easily modified, which
of Au NCs and further achieve good biological applica- facilitating fundamental studies and practical applications.
tions. An important result was given by Ying et al. [48], Liu group [56] successfully prepared blue fluorescent Au NCs
who reported a simple, one-pot and ‘‘green’’ synthetic in the presence of poly-cytosine DNAs at low pH and poly-
route for the preparation of Au NCs at the physiologi- adenine at neutral pH using citrate as the reducing agent,
cal temperature (37 ◦ C) with red emission using bovine and they explored the affecting factors for the synthesis
serum albumin (BSA) as a bio-ligand (Fig. 4). This process of Au NCs in detail. Subsequently, Shao group [57] synthe-
is similar to the bio-mineralization behavior of organisms sized fluorescent Au NCs using various DNA as template.
in nature. Upon addition of Au ions to the BSA solution, The emission behavior of the hairpin (HP)-DNA-stabilized Au
the protein molecules sequestered Au ions and entrapped NCs is dependent on the loop sequence. The cytosine loop
them. The reduction ability of BSA molecules was then acti- is the most efficient host to produce fluorescent Au NCs.
vated by adjusting the reaction pH to 12 and entrapped However, the loop composed of thymine as well as adenine
ions underwent progressive reduction to form Au NCs in creates Au NCs with a much weaker emission than the cyto-
situ. Notably, BSA functions both as a stabilizing agent sine and guanine loops. The cytosine-favored Au NC growth
and a reductant in this process. This work is remarkable was further confirmed using the sing-strand DNA (ssDNA)
not only for providing an alternative approach of syn- having an identical base composition as the HP-DNA. Addi-
thesizing highly fluorescent Au NCs, but also providing a tionally, the fully matched DNAs seem to be less efficient
successful paradigm for exploring other proteins and met- than the corresponding HP-DNA and ssDNA structures. The
als. Later, Lu’s group [49] found that lysozyme could also DNA-stabilized Au NCs can be used as probes to in situ ana-
act as a good reducing and stabilizing agent for the prepa- lyze the DNA hybridization and cytosine-related mutation.
ration of highly fluorescent Au NCs. Recently, Pradeep group This DNA sequence-dependent growth of fluorescent Au NCs

Figure 4 Schematic of the formation of Au NCs in BSA solution.


Reprinted from Ref. [48] with permission by American Chemical Society.

Please cite this article in press as: L. Zhang, E. Wang, Nano Today (2014), http://dx.doi.org/10.1016/j.nantod.2014.02.010
+Model
NANTOD-359; No. of Pages 26 ARTICLE IN PRESS
6 L. Zhang, E. Wang

will hold a great promise for developing biological labels, Luminescent Ag NCs formed at lower molar ratio of Ag:L,
biosensors, and DNA-related nanomaterials. while nonluminescent silver nanoparticles were produced
at higher molar ratio (Fig. 5). Interestingly, nonluminescent
Ag nanoclusters silver nanoparticles could be converted into luminescent Ag
NCs, via a process referred to as ‘‘size focusing’’ in the pres-
Compared with Au NCs, Ag NCs are easily synthesized via ence of added excess ligands and reducing agent. Another
designing different ligands as stabilizing agents. However, type of polyelectrolyte polyethyleneimine (PEI) was utilized
the stability of Ag NCs was more easily affected by many as scaffolds for the synthesis of fluorescent Ag NCs through
factors. Except for their vulnerability to atmospheric oxy- the strong interaction between N atom and Ag atom [66].
gen and other electron acceptors in solution, reduced silver The PEI capped Ag NCs showed a blue emission at 455 nm
atoms have a strong tendency to agglomerate, it is necessary and a high stability which made it kept at room temperature
to prevent stable NCs formation in favor of thermodynami- for at least 30 days.
cally favored large silver nanoparticle generation. Small molecules containing carboxylic groups or thiols
The pioneering reported water-soluble fluorescent Ag also can be used as templates for the synthesis of fluorescent
NCs were prepared in the presence of dendrimers, by Dick- Ag NCs. Pradeep group [67] synthesized red- and blue-green-
son and co-workers [58]. OH-terminated dendrimers PAMAM emitting clusters Ag8 (H2 MSA)8 and Ag7 (H2 MSA)7 (H2 MSA:
G4 and G2 (fourth- and second-generation) were used as mercaptosuccinic acid) through an interfacial etching reac-
scaffolds to synthesize fluorescent Ag NCs through direct tion starting from H2 MSA-protected silver nanoparticles as
photoreduntion. The Ag NCs exhibited distinct fluorescence precursor. The result of X-ray photoelectron spectroscopy
peaks ranging from 533 to 648 nm with high photostability. (XPS) showed that the Ag:S atomic ratio for each band
Latterly, Balogh and coworkers also utilized PAMAM G5 to was 1:1 and that the silver was fully reduced. The detec-
prepare dendrimer templated Ag NCs by irradiating the solu- tion of [Ag8 (H2 MSA)4 (HMSA)4 ]− and [Ag7 (H2 MSA)5 (HMSA)2 ]−
tions of silver(I)—dendrimer complexes with UV light [59]. by the mass spectrometry was further used as strong evi-
Polymers containing amino and carboxylic groups that can dence to assign the Ag8 as a red emitter and Ag7 as a
form complexes with Ag ions have been used for the prepa- blue emitter. Recently, Chen group presented a facile one-
ration of fluorescent Ag NCs. Dong’s group [60] reported a pot sonochemical approach to prepare highly blue-emitting
new approach for the synthesis of fluorescent and water- Ag NCs using glutathione as a stabilizing agent in aqueous
soluble Ag NCs, using the common polyelectrolyte PMAA solution [68]. The results reveal that it is a rapid and envi-
as the template under UV illumination, with an absorp- ronmentally friendly method for preparing fluorescent Ag
tion peak near 510 nm that overlaps with the excitation NCs. The Ag NCs consist of 12 silver atoms and the exper-
spectrum for 620 nm emission. The Ag NCs show lumines- imental conditions have been optimized to derive Ag NCs
cence lifetimes of 2.3 ns and a high QY of 18.6%. They found with high emission. The Ag NCs exhibit excellent water
that pH of solution has an important role on the fluores- solubility, high stability, and have an excellent sensitivity
cent emission of obtained Ag NCs. The maximum intensity and high selectivity toward S2− , with a detection limit of
was obtained at pH 4.5 whereas either lower or higher pH 2 nM.
value led to weaker fluorescent intensity. Instead of ultra- Proteins and peptides have been used for the synthesis
violet light, Ras’s group [61] found that visible light also of Ag NCs. Proteins possess abundant binding sites that can
could initiate the reduction of silver ion in the presence potentially bind and some of them can further reduce Ag
of PMAA and further lead to the formation of silver NCs in ion, thus offering better scaffolds for template-driven for-
an environmentally friendly manner. Their results indicate mation of fluorescent Ag NCs. Cell matrices were considered
a phenomenon that higher Ag concentration leads to the to be rich libraries of various proteins for the production
generation of Ag NCs with emission at the redder end. An of luminescent Ag NCs. The proteins of nucleolus, such as
increase in the Ag/PMAA molar ratio resulted in red-shifts nucleolin, are known to have high affinity to silver ions due
of the absorption peaks and the corresponding emission to their amino-terminal domain, was employed as the major
peaks. Later, Suslick et al. [62] presented a new and eas- scaffold for the formation of fluorescent Ag NCs by Dickson
ily controlled sonochemical strategy for the synthesis of and coworkers [69]. The fluorescent Ag NCs can be gener-
water-soluble fluorescent Ag NCs also in the presence of ated by photoactivation of cell fed with silver salt in vivo
PMAA. The evolution of absorption and fluorescence of Ag ambient-temperature condition, which opens a new path
NCs is quite similar to that of photoactivation, the obtained for the application of Ag NCs in biological systems. Besides
Ag NCs exhibit red emission with a maximum excitation the formation of Ag NCs in fixed cells, the fluorescent Ag
of 510 nm. In addition, poly(acrylic acid) derivatives [63] NCs can also be synthesized in the presence of proteins.
and poly(N-isopropylacrylamide-acrylic acid-2-hydroxyethyl Narayanan and Pal [70] reported on the use of an enzyme,
acrylate) [64] both were used for the synthesized fluorescent bovine pancreatic ␣-chymotrypsin (CHT) as scaffold through
Ag NCs under UV irradiation (365 nm). Recently, Mattoussi the chemical reduction of silver ions to produce well dis-
and coworkers [65] developed one phase growth reaction to persed and stable fluorescent Ag NCs. The protein-stabilized
prepare a series of Ag nanoparticles and luminescent Ag NCs Ag NCs show emission at 680 nm when excited at 500 nm.
using NaBH4 as a reducing agent in the presence of polyethy- Later, denatured bovine serum albumin (dBSA) also was used
lene glycol (PEG) appended with lipoic acid (LA) groups as scaffold for the synthesis of fluorescent Ag NCs [71]. The
at one end and reactive (—COOH/—NH2 ) or insert (—OCH3 ) dBSA had 35 free cysteine residues which could contribute
functional groups at the other end. The size and properties to polyvalent interactions with the Ag NCs and serve as
of Ag NCs were primarily controlled by varying the Ag-to- effective stabilizing agents for the Ag NCs. More recently,
ligand (Ag:L) molar ratios and the molar amount of NaBH4 . Chen and Yang group [72] developed a facile approach to

Please cite this article in press as: L. Zhang, E. Wang, Nano Today (2014), http://dx.doi.org/10.1016/j.nantod.2014.02.010
+Model
NANTOD-359; No. of Pages 26 ARTICLE IN PRESS
Metal nanoclusters 7

synthesize red-emitting fluorescent Ag NCs using lysozyme show blue and red emission under different alkaline condi-
as scaffold and NaBH4 as a reducing agent at room tem- tions.
perature. The water-soluble Ag NCs have a positive surface DNA is also an effective template for the preparation
charge and good stability, which is still kept through adjust- of fluorescent Ag NCs mainly because the cytosine base
ing the pH using acetic acid. This is the first report of the exhibits strong affinity for silver ions. The presence of
synthesis of fluorescent Ag NCs with positive surface charge both carbonyl oxygens and doubly bonded ring nitrogens
using a protein as scaffold. in heterocyclic bases plays an important role in determin-
Peptides contain carboxylic acids with high silver affinity ing their interaction strength with Ag ions [75]. At present,
and a variety of amino acid sequences also offer potential more investigations have focused on the use of DNA as
tenability in the local site to improve silver NCs stability. scaffolds for the synthesis of Ag NCs [76—89]. These stud-
Wright group [73] reported a new approach for the synthesis ies reveal that the generation of these novel fluorescent
of fluorescent Ag NCs using the histidine-rich AHHAHHAAD as NCs is highly DNA-sequence-dependent [77,79,80] and their
template. Later, inspired by the protein, uncleolin, Dickson photoluminescence emission band could be easily tuned
and coworkers [69] design several short peptides used for the throughout the visible and near-IR range by simply changing
synthesis of Ag NCs. KECDKKECDKKECDK (P1), incorporating the sequence of the oligonucleotide.
the specific amino acids most prevalent in nucleolin: glu- The first DNA-protected Ag NCs was reported by Dick-
tamic acid (15.6%), lysine (12.7%), and aspartic acid (8.5%), son and coworkers in 2004 [90]. They use ssDNA containing
was designed for the synthesized of Ag NCs. However, the the 12-base 5 -AGGTCGCCGCCC-3 as templates for synthe-
Ag NCs were only moderately stable at room temperature sizing Ag NCs in a phosphate buffer. Mass spectroscopy
(chemical lifetime of 3 days) due to the short length of demonstrated the fluorescent Ag NCs with 1—4 atoms were
peptide. The incorporation of several hydrophobic amino formed, which show distinct absorption and fluorescence
acids, another two peptides HDCHLHLHKCHLHLHCDH (P2) spectral features. Their results confirm that it may be fea-
and HDCNKDKHDCNKDKHDCN (P3) were designed to synthe- sible to control the formation of NCs using DNA strands with
size Ag NCs. The chemical lifetimes of P2- or P3-stabilized specific sequences, and cytosine is the most favored base
Ag NCs were extended to two weeks in deionized water and for the formation of Ag NCs. Subsequently, they use the
5 weeks in PBS, respectively. More recently, Gao group [74] 12-mer cytosine as template for the synthesis of Ag NCs.
has utilized an artificial peptide with amino acid sequence The obtained Ag NCs show red and blue/green emission.
CCYRGRKKRRQRRR to biomineralize a series of Ag NCs, which Base sequence tuning has been found to result in distinct

Figure 5 (A) Chemical structures of the ligands, (B) synthetic strategy of NPs/NCs of various core sizes. (C) Table showing the
Ag:L ratios used for the synthesis. (D, E) Photographs of aqueous dispersions of the particles synthesized at various Ag:L ratios under
room light and UV light irradiation, respectively. Images were collected from a set of freshly prepared dispersions, without filtration
or adjustment of the nanoparticle or cluster concentrations.
Reprinted from Ref. [65] with permission by American Chemical Society.

Please cite this article in press as: L. Zhang, E. Wang, Nano Today (2014), http://dx.doi.org/10.1016/j.nantod.2014.02.010
+Model
NANTOD-359; No. of Pages 26 ARTICLE IN PRESS
8 L. Zhang, E. Wang

emission properties. Richards et al. [79] reported on five dis- scaffolds was highly sequence-dependent, which could iden-
tinct Ag NCs obtained based on five different sequences DNA tify a typical single-nucleotide mutation, the sickle cell
as scaffolds with tunable fluorescence emission throughout mutation (Fig. 6). Afterwards, Li and coworkers further
the visible and near IR. Blue and green emitting species investigated the effect of DNA templates with different
were only created in unbuffered solutions, and yellow, secondary structures, such as i-motif, G-quadruplex and
red and near-IR emitting species formed best in appropri- Watson—Crick duplex [93]. Polymorphic DNA templated Ag
ately buffered solutions. Their results demonstrate that the NCs show distinct fluorescent properties. With DNA template
photostability of Ag NCs was drastically increased with these adopting i-motif or duplex structure, silver atoms tend to
longer wavelength emitters. aggregate inside the encapsulated spaces of nucleobases,
The sequence and secondary structure of DNA both affect and the formed Ag NCs are positively charged with high flu-
the fluorescence properties of DNA-encapsulated Ag NCs. orescent spectral features. While with G-quadruplex DNA
Gwinn and coworkers [80] used six 19-mer DNA oligomers as template, silver atoms favor to aggregate outside of the
as templates for the synthesis of Ag NCs. They found that G-tetrad, which results in the formation of larger silver crys-
both C-rich and G-rich single-strand DNA (ssDNA) produced tals without fluorescence property. More recently, Qu and
brightly fluorescent Ag NCs, while the duplex formed by coworkers have demonstrated site-specific, homogeneous,
C-rich and G-rich DNA was an invalid template for the forma- and bright Ag NCs with triplex DNA as template [94]. By
tion of fluorescent Ag NCs, accompanied by negligible visible reasonable design of DNA sequence, homogeneous Ag2 clus-
fluorescence. Thus, the spectral properties of DNA-attached ter was obtained in the predefined position of CG.C+ site of
few-atom Ag clusters are sensitive to the secondary struc- triplex DNA. This strategy was also explored for controlled
ture of DNA. Furthermore, a set of hairpin structures named alignment of Ag NCs on the DNA nanoscaffold.
C-loop, G-loop, A-loop and T-loop were designed to compare
the relative capacities of C, G, A, and T homopolymers to Cu nanoclusters
host fluorescent species. C-loop and G-loop solutions exhibit
fluorescence of similar brightness, while A-loop solutions Compared with Au and Ag NCs, reports on the synthesis of Cu
fluoresce weakly. T-loop solutions even produce no fluores- NCs are still scarce primarily because of their susceptibility
cence for excitation at visible wavelengths. Later, Petty and to oxidation and the difficulty in preparing extremely tiny
coworkers [91] first explored pH-dependent i-motif DNAs as particles. Up to now, several methods have been success-
templates for the synthesis of Ag NCs. Two C-rich sequences fully developed to synthesize Cu NCs with unique optical
(dTA2 C4 )4 and (dC4 A2 )3 C4 with a common C4 i-motif core are and catalytic properties. Crooks and coworkers [95] used
chosen as model systems. They obtained red and green emit- OH-terminated dendrimers PAMAM G4 (fourth-generation)
ting fluorescent Ag NCs. Their results indicate that protons as scaffolds to synthesize fluorescent Cu NCs. The PAMAM
dominate DNA folding for the red emissive species, while the dendrimers have an ethylenediamine core (G4-OH) contain-
green emissive clusters themselves determine the shape of ing interior tertiary amines that can strongly interact with
their DNA matrix. These studies provide the basis for under- Cu ions, after the reduction by sodium borohydride to form
standing how specific base arrangements and environmental Cu NCs. Almost at the same time, Balogh and Tomalia [96]
factors influence the formation of fluorescent NCs. also successfully synthesized stable Cu NCs using PAMAM den-
Recently, our group is amazing to find that hybridized drimers as scaffolds.
DNA duplexes could also be developed as capping scaffolds Later, Vazquez-Vazquez et al. [97] reported the prepa-
for the generation of fluorescent Ag NCs [92]. And, the ration of a series of small atomic copper clusters by the
formation of fluorescent Ag NCs in hybridized DNA duplex microemulsion technique. Their size can be controlled by

Figure 6 Use of two different DNA duplexes with inserted cytosine loops working as synthetic scaffolds to generate fluorescent
silver NCs for the identification of the sickle cell anemia gene mutation (black dots represent hydrogen bonds formed in base pairing
and black dashed lines the sugar-phosphate backbone).
Reprinted from Ref. [92] with permission by American Chemical Society.

Please cite this article in press as: L. Zhang, E. Wang, Nano Today (2014), http://dx.doi.org/10.1016/j.nantod.2014.02.010
+Model
NANTOD-359; No. of Pages 26 ARTICLE IN PRESS
Metal nanoclusters 9

role in producing red-emissive fluorescent Cu NCs on ssDNA


templates. The thymine-dependent growth of the fluores-
cent Cu NCs is confirmed by Hg2+ mediated T—T base pair
in comparison with the other non-specific metal ions, which
could be developed into a practical sensor for turn-on fluo-
rescence detection of Hg2+ with a high selectivity.

Other metal nanoclusters

Except for Au, Ag and Cu NCs, other metal materials such as


Pt [104—106] and Pd [107], could also been made into fluo-
Figure 7 Schematic representation of detection strategy on rescent NCs. For instance, Kawasaki group [105] and Obora
the basis of the formation of fluorescent Cu NCs (Y: SNP site). group [107] demonstrated simple, one-pot strategies for the
Reprinted from Ref. [102] with permission by American Chemi- synthesis of Pt and Pd NCs in N, N-dimethylformamide (DMF)
cal Society. solution in the absence of any capping agents such as surfac-
tant, polymer, or thiolate-organic compounds, respectively.
The synthesis process is similar except for the different pre-
adjusting the percentage of the reducing agent. Photolu- cursors (H2 PtCl6 for Pt clusters and PdCl2 for Pd clusters)
minescent copper clusters (Cun , n ≤ 13) can be obtained and modified reaction time (8 h for Pt and 6 h for Pd). The
using very low percentages of the reducing agent (<10% of as-prepared Pt and Pd NCs are highly stable in organic, aque-
the stochiometric amount), while photoluminescent clusters ous, or salt solutions and they were readily dispersible in a
disappear for larger percentages of reducing agent. Vilar- variety of solvents. The property will greatly facilitate their
Vidal and coworkers [98] reported a simple electrochemical use in metal catalysis and bioimaging applications.
approach for the synthesis of fluorescent Cun (n ≤ 14) NCs Recently, Inouye and Jin group [106] reported a simple
stabilized by tetrabutylammonium nitrate. Copper ions pro- method for the synthesis of water soluble and fluorescent
duced during the electrolysis from the Cu anode were Pt NCs using the fourth-generation PAMAM (G4-OH) den-
reduced to NCs on the cathode. The prepared Cu NCs are drimer as the template by reducing chloroplatinic acid with
very stable over years. They show photoluminescence in the NaBH4 . The synthesized Pt NCs emitted a strong blue fluores-
visible range with unusual high QY of 13% and exhibit an cence under UV light (365 nm) irradiation. However, simple
amphiphilic character and can be dispersed in both polar oxidation of PAMAM (G4-OH) with (NH4 )2 S2 O8 can also pro-
and apolar solvents, which are suitable for applications in duce species that emit blue fluorescence [108]. In order to
biological system. avoid the possible fluorescence interference from oxidation
Recently, Kawasaki et al. [99] reported the synthesis of species of the dendrimer, they applied mercaptoacetic acid
fluorescent Cu NCs via a microwave-assisted polyol method (MAA) to replace PAMAM (G4-OH) as the ligand for the Pt NCs
without using additional protective and reducing agents. through a ligand exchange reaction. Atomically monodis-
Their results demonstrated that the synthesized Cu NCs persed Pt5 (MAA)8 NCs were obtained by using size-exclusion
were highly stable and exhibited strong blue fluorescence. high performance liquid chromatography (HPLC) and the Pt5
Chen group [100] reported a simple on-pot wet chemical NCs show blue photoluminescence with an 18% quantum
reduction method for the synthesis of fluorescent Cu NCs. yield in water.
The synthesized stable Cun (n ≤ 8) NCs use 2-mercapto-
5-n-propylpyrimidine (MPP) as protecting ligands, which
exhibited interesting photoluminescence with dual emis- Properties of metal nanoclusters
sions at 423 and 593 nm, respectively.
DNA can also be used as an effective template for Absorption properties
the preparation of fluorescent Cu NCs. Mokhir group [101]
reported a method for the selective formation of fluorescent The absorption properties of metal nanoparticles are mainly
Cu NCs on double-stranded DNA in solution. The synthe- dependent on the surface plasmon resonance of conduc-
sized Cu NCs show emission range 587—600 nm when excited tion electrons. Generally, metal nanoparticles show intense
at 340 nm. The size of copper NCs can be controlled by colors due to the collective oscillation of conduction elec-
the use of a double-stranded DNA (dsDNA) template of a trons upon interaction with visible light. While, as for metal
selected length. The metallization is highly selective for NCs, the continuous density of states breaks up into dis-
dsDNA compared to ssDNA, which can be used for the detec- crete energy levels [16], they no longer exhibit plasmonic
tion of dsDNA by fluorescence. Subsequently, our group [102] properties, but they still interact with light through elec-
reported that the fluorescence of DNA-hosted Cu NCs is tronic transitions between energy levels, leading to sharp
very sensitive to base type located in the major groove of absorption. For instance, Collings et al. [109] reported that
dsDNA (Fig. 7). The research results have shed some light the optical absorption spectra of a series of Aun (n = 7, 9,
on the luminescent mechanism of Cu NCs, owing to its high 11, 13) and their cations located between 1.9 and 5.6 eV.
specificity and easy operation without rigorously controlled Recently, Jin group [110] found that glutathione-protected
temperature and arduous probe DNA design. Recently, Liu Au25 showed characteristic absorption features in the range
et al. [103] successfully synthesized fluorescent Cu NCs 400—1000 nm, which are believed to arise from intraband
using ssDNA as templates and to identify the critical DNA (sp←sp) or interband (sp←d) transitions (Fig. 8). Tsukuda
sequences. They found DNA thymine base plays a dominant group [21] found that the spacing between the discrete

Please cite this article in press as: L. Zhang, E. Wang, Nano Today (2014), http://dx.doi.org/10.1016/j.nantod.2014.02.010
+Model
NANTOD-359; No. of Pages 26 ARTICLE IN PRESS
10 L. Zhang, E. Wang

nanometer level, the efficiency of luminescence obviously


enhances. In particular, the sizes approach to the Ferimi
wavelength of electrons [7], metal NCs will show strong
luminescence properties. The luminescence of metal nan-
oclusters is generally assigned to the electronic transitions
between occupied d bands and states above the Fermi level
(ca. sp bands) or the electronic transitions between the
highest occupied orbital and the lowest unoccupied orbital
(HOMO—LUMO) [12]. With the development of the synthesis
methods for metal NCs, their fluorescence properties
get more attention and more in-depth development. For
instance, Dickson group synthesized a series of Ag NCs with
tunable fluorescence emission throughout the visible and
near IR [79]. They also reported DNA-encapsulated Ag nan-
oclusters yield highly stable and bright fluorescence in the
near IR [78]. The very high emission rates without blinking
on experimentally relevant time scales (0.1 to > 1000 ms).
Ying group [48] reported the synthesis of water-soluble and
fluorescent Au25 NCs with good biocompatibility and the Au25
NCs show emission peak at 640 nm. Recently, Jin group [36]
reported that the surface ligand play a major role in the
fluorescent of thiol-protected Au NCs. These studies suggest
that there are two major sources for explaining the emission
properties of metal NCs, the fluorescence may arise from (i)
the metal core through a manifestation of intrinsic quanti-
zation effects and (ii) the particle surface due to interaction
of the metal core and surface ligands.

Two-photon absorption

Two-photon absorption (TPA) is the simultaneous absorp-


tion of two photons of identical or different frequencies
Figure 8 (A) Kohn—Sham orbital energy level diagram for a in order to excite a molecule from one state (usually
model compound Au25 (SH)18 − . Each KS orbital is drawn to indi- the ground state) to a higher energy electronic state.
cate the relative contributions (line length with color labels) Compared with one-photon excitation, two-photon excita-
of the atomic orbitals of Au (6sp) in green, Au (5d) in blue, tion in the near-infrared region increases the penetration
S (3p) in orange, and others in gray (those unspecified atomic depth, spatial resolution due to lower scattering, and min-
orbitals, each with a <1% contribution). The left column of the imizes autofluorescence [112], which is suitable for in vivo
KS orbitals shows the orbital symmetry (g, u) and degeneracy (in imaging and photodynamic therapy. Goodson group [113]
parentheses); the right column shows the HOMO and LUMO sets. reported that two-photon emission of Au25 NCs is observed
(B) The theoretical absorption spectrum of Au25 (SH)18 − . Peak at 830 nm by exciting at 1290 nm, and the two-photon
assignments: peak a corresponds to 1.8 eV (observed), peak b absorption cross-section was measured to be 42,700 GM
corresponds to 2.75 eV (observed), and peak c corresponds to (Göpert-Mayer unit, 10−50 cm4 s), which is superior to the
3.1 eV (observed). TPA cross-sections of many organic chromophores. Recently,
Reprinted from Ref. [110] with permission by American Chemi- glutathione-protected Au NCs was also observed strong two-
cal Society. photon emission, and the TPA cross-section was determined
to be 189,740 GM [114]. In addition, DNA-protected Ag NCs
also show two-photon emission and possess high TPA cross-
states in each band increased with the size decrease of sections, the cross section of the 660 nm emitter peaks is
glutathione-protected Au NCs, leading a blue shift in the 35,000 GM, while the 680 nm emitter peaks is 34,000 GM, and
absorption peaks. Dickson group [90] reported the absorp- the 710 nm emitter reaches 50 000 GM [115]. More recently,
tion peaks of fluorescent Ag NCs with 1—4 atoms at 357 nm Goodson group [84] reported that two-photon excited fluo-
and 440 nm. In recent work, DHLA-protected Ag4 and Ag5 rescence was detected for Ag NC on dsDNA at 630 nm when
show absorption peaks at 435 nm and 335 nm, respectively, excited at 800 nm. The two-photon absorption cross-section
with another shoulder peak at 500 nm [111]. was calculated to be ∼3000 GM.

Fluorescence properties Electrochemiluminescence

Bulk metals normally show very weak luminescence owning Electrochemiluminescence (ECL) is the process whereby
to the efficient nonradiative decay and the absence of an species generated at electrodes undergo high-energy
energy gap. As the sizes of metals gradually decrease to the electron-transfer reactions to form excited states that emit

Please cite this article in press as: L. Zhang, E. Wang, Nano Today (2014), http://dx.doi.org/10.1016/j.nantod.2014.02.010
+Model
NANTOD-359; No. of Pages 26 ARTICLE IN PRESS
Metal nanoclusters 11

light [116], has received considerable attention in the ana- some reported metal NCs. Such metal NCs with lifetimes
lytical field due to its high sensitivity, low background noise from sub-microsecond to microsecond are ideal for the
and cost. In the ECL system of metal NCs, the NCs usu- utilization in fluorescence detection and imaging, which
ally show strong ECL in the presence of co-reactant. A greatly eliminates autofluorescence from biological tis-
co-reactant is a species that produces a highly reactive sue. For example, DHLA- and lipoic acid-protected Au
intermediate upon oxidation or reduction, which reacts with NCs both exhibit sub-microsecond fluorescence lifetimes
a luminescent species to either produce an excited state [29,124]. While, lipoic acid-stabilized Ag NCs show a life-
or commence an excitation pathway by a one-electron oxi- time of 37 ␮s, with 652 nm emission at 425 nm excitation
dation or reduction. The most common excitation pathway [111]. Multiple lifetime components longer than 20 ␮s were
in ECL is the ox-red excitation pathway, in which the lumi- observed in PMAA-encapsulated Ag NCs as well [61]. These
nescent species is first oxidized by a cathodically produced metal NCs have great potential applications in biological
oxidizing radical and then reduced by one energetic elec- imaging.
tron to the excited form of its original oxidation state. Ras
group [61] first reported that PMAA-protected Ag NCs exhib-
ited ECL. Recently, Chen group [117] found BSA-stabilized
Polarized emmission
Au NCs also showed ECL using triethylamine as the co-
reactant on the Pt electrode surface. At the same time, Recently, Raut and coworkers [125] found BSA-protected
Zhu group [118] observed ECL from BSA-stabilized Au NCs Au25 clusters exhibited polarization properties. They studied
on a modified indium tin oxide electrode surface, where steady state and time resolved fluorescence and polariza-
K2 S2 O8 was used as the co-reactant. More recently, our group tion behavior in water, propylene glycol and glycerol. The
[119] reported that organic-soluble fluorescent Au8 NCs major observation is the presence of polarization behavior
synthesized through a unique heterophase ligand-exchange in these long fluorescence lifetime clusters. We expect that
induced etching of gold NPs, and the fluorescent Au8 NCs the polarization properties of metal NCs can be utilized in
exhibited both the annihilation ECL in organic solution and polarization based sensing applications and other exciting
the co-reactant ECL in aqueous solution. applications such as the study of large molecular weight
protein complexes.
Solvatochromic properties
Applications of metal nanoclusters
As we all know, metal NPs exhibit obvious solvatochromic
effect due to the surface plasmon phenomenon [120]. The unique physicochemical properties of metal NCs make
Recent studies demonstrated that metal NCs also dis- them attractive for use in various fields including both fun-
play similar solvatochromic properties. Chen group [121] damental science studies and technological exploration. In
observed solvent-dependent photoluminescence properties this section, we summarize recent advances in the applica-
of Au8 NCs when exposed to different organic solvents. tion of metal NCs as new fluorescent probes for analytical
They also investigated the solvatochromic effect of Au8 sensors and biological imaging.
NCs protected by different ligands, and they attributed the
solvatochromic effect of Au NCs to the rich surface prop-
erties of the clusters. In addition, variously capped organic Metal ion sensors
soluble and aqueous Ag NCs also exhibit spectral shifts as sol-
vent polarity is changed [61,122]. DNA-encapsulated Ag NCs The determination of heavy metal ions in biological sys-
were found to exhibit solvatochromic properties, absorption tems and environment is of tremendous significance due to
and emission bands red-shift as the content of methanol in their hazardous effects on the ecosystem and the human
aqueous solution increased. This may result from structural health. In recent years, numerous of types of fluorescent
changes of the cluster—ligand system or from high frequency metal NCs have been applied to detect heavy metal ions. Our
dielectric response in response to ground vs. excited state group [126] has developed a new strategy for the selective
dipole moment changes, but the reversible solvatochromism detection of Hg2+ using DNA-stabilized Ag NCs as fluorescent
suggests that the optical shifts are not from cluster size probes. The DNA-stabilized Ag NCs were found to be good
changes. Those studies indicate that the change in chem- indicators for the selective identification of Hg2+ based on
ical environment may induce electronic energy splitting and quenching the fluorescence of Ag NCs by Hg2+ (Fig. 9). The
electron redistribution on the cluster surface, leading to a unchanged fluorescence lifetime implies that the quenching
variation of the optical properties of the metal NCs. of the Ag NCs by Hg2+ ions obeyed a simple static quenching
mechanism. Thus, the Hg2+ ions and DNA-stabilized Ag NCs
Fluorescence lifetimes may form a kind of non-fluorescent complex, which results
in quenching of the fluorescence. The proposed detection
As for the fluorescence lifetime of metal NCs, most metal protocol is environmentally-friendly, sensitive and selective,
NCs exhibit monoexponential nanosecond fluorescence life- and a detection limit of 5 nM was obtained. Subsequently,
times. Some polymer- and peptide-protected metal NCs Dong group [127] developed a label-free method for ‘‘turn-
show biexponential lifetime decays, with a hundreds on’’ detection of Hg2+ using DNA duplex stabilized Ag NCs as
of picoseconds component and the other 2—3 nanosec- fluorescent probes. The strategy utilizes the Hg2+ -mediated
onds component [10]. However, thiol-protected metal NCs T-T formation to strengthen the DNA duplexes and influ-
present extremely short picoseconds fluorescence lifetimes ence the formation of fluorescent Ag NCs, which shows high
[67,123]. Microsecond lifetimes were also detected from sensitivity and selectivity. In another study, Wei et al. [49]

Please cite this article in press as: L. Zhang, E. Wang, Nano Today (2014), http://dx.doi.org/10.1016/j.nantod.2014.02.010
+Model
NANTOD-359; No. of Pages 26 ARTICLE IN PRESS
12 L. Zhang, E. Wang

of Cu2+ induced a quite significant fluorescence enhance-


ment, which could be developed for selective and sensitive
detection of Cu2+ . The limit of detection (LOD) for Cu2+
was estimated to be 8 nM. In another protocol, they devel-
oped DNA-Cu/Ag NCs-based fluorescent assay for ‘‘turn-on’’
detection of Cu2+ [132]. The fluorescence of DNA-Cu/Ag NCs
was quenched by 3-mercaptopropionic acid (MPA), which
was recovered after the addition of Cu2+ . The LOD reached as
low as 2.7 nM for Cu2+ . The practicality of the present strat-
egy for the detection of Cu2+ in environmental samples was
validated through analysis of soil and pond water samples.
Ag(I) ions are reported to be greatly toxic to a lot of
bacteria, viruses, algae and fungi, and such antibacterial
activity of Ag(I) ions has been employed into many appli-
cation fields (such as toiletry, timbering, clinical material)
[133]. However, once the concentration of Ag(I) ions is high
enough, they can still bring harmful side-effects to the envi-
ronments and the human health. Jin [134] group developed
Figure 9 The ‘‘turn-off’’ fluorescent mechanism for Hg2+ a novel type of nanosensor for silver ion detection utiliz-
sensing with high selectivity. ing glutathionate (SG)-capped Au25 NCs as fluorescent probe
Reprinted from Ref. [126] with permission by Royal Society of based on the fluorescence enhancement. The Ag+ detection
Chemistry. limit is approximately 200 nM in combination with the good
selectivity against other cations. Further experiments reveal
synthesized lysozyme-stabilized fluorescent Au NCs, which three important factors responsible for the unique fluores-
can be used as a sensor for sensitive and selective Hg2+ cence enhancement caused by silver ions. Among them, the
detection with a detection limit of 10 nM. Inspired by this oxidation (leading to a higher charge state of Au25 ) con-
finding, Zhou et al. [72] synthesized positively charged and tributes most, with the other two contributions coming from
red-emitting lysozyme-stabilized Ag NCs, which also could the interactions of Ag+ with Au25 and Ag0 with Au25 . This work
be applied in the highly selective determination of Hg2+ indicates another potential application of Au NCs, offers
based on their fluorescence quenching due to the 5d10 (Hg2+ )- new strategies for nanocluster-based chemical sensing, and
4d10 (Ag+ ) metallophilic interaction. Recently, Shang et al. reveals a new way to influence nanocluster chemistry for
[128] reported a rapid synthesis of fluorescent DHLA-capped potential applications.
Au NCs through a microwave-assisted strategy. This method In aqueous solutions, chromium mainly exists in two
could enhance the fluorescence QY of Au NCs by about 5- valence states: Cr(III) and Cr(VI). Cr(III) in trace amounts is
fold and shorten the reaction time from hours to several an essential nutrient required for sugar and fat metabolism,
minutes. The DHLA-Au NCs were capable of sensing Hg2+ its high concentration is believed to induce oxidative DNA
through quenching the fluorescence of the Au NCs. The fluo- damage, causing cancer. While Cr(VI) is approximately 100
rescence decay of Au NCs became faster in the presence times more toxic and is considered a human carcinogen with
of Hg2+ , with the average lifetime decrease. This result adverse impact on human skin, stomachs, lung, liver and
implies that fluorescence quenching results from interac- kidneys [135]. Zhu group [136] prepared highly fluorescent
tions between Hg2+ and Au+ on the nanoparticle surface due Ag NCs via a rapid microwave-assisted green approach and
to the specific and strong d10 —d10 metallophilic interaction used them as novel fluorescence probes for the sensitive
(e.g., 5d10 (Hg2+ )—5d10 (Au+ )). And the limit of detection for and selective determination of Cr3+ ions based on quench-
Hg2+ was 0.5 nM. Moreover, they have successfully demon- ing the fluorescence of Ag NCs. The detection limit is found
strated the utilization of DHLA-Au NCs for monitoring Hg2+ to be 28 nM. Recently, Liu group [137] developed a novel
in living cells. method for selective determination of Cr(III) and Cr(VI) in
Copper is essential to all living systems for its key role environmental water samples based on target-induced fluo-
in cooperating with certain proteins to produce numer- rescence quenching of glutathione-stabilized gold NCs. The
ous enzymes. However, Cu2+ ions are toxic when its level obtained detection limits are found to be 2.5 ␮g/L for Cr(III)
exceeds cellular needs, which will cause serious diseases and 0.5 ␮g/L for Cr(VI), respectively.
[129]. Considerable efforts have been devoted to detecting In addition, Banerjee [138] developed Au cluster-based
Cu2+ using fluorescent metal NCs as probes. Dong group [130] fluorescent sensor for ‘‘turn-on’’ detection of AsIII with high
has developed a simple and sensitive fluorescent sensor for sensitivity and selectivity. The Au NCs synthesized through
Cu2+ detection based on fluorescent PMAA protected Ag NCs. a wet-chemical approach using a dipeptide L-cysteinyl-L-
A detection limit of 8 nM was obtained, which is much lower cysteine, which show the emission maximum at 410 nm with
than the U.S. Environmental Protection Agency (EPA) limit a QY of 41.3%. The addition of AsIII induced a quite signifi-
for Cu2+ in drinking water (20 ␮M). Chang and coworkers cant fluorescence enhancement of Au NCs, which should be
have developed a novel, simple and turn-on method for the attributed to the fact that positive charged AsIII ions interact
detection of Cu2+ using water-soluble DNA/Ag NCs [131]. with the negatively charged gold clusters and the electrons
The fluorescent Ag NCs were prepared with a reported DNA can flow from the electron rich gold clusters system to the
sequence as synthesis template. The obtained DNA/Ag NCs electron deficient AsIII ion, resulting in an increase in the
display yellow emission with a QY of 11.5%. The introduction radiative decay rate Au NCs. The LOD reached as low as

Please cite this article in press as: L. Zhang, E. Wang, Nano Today (2014), http://dx.doi.org/10.1016/j.nantod.2014.02.010
+Model
NANTOD-359; No. of Pages 26 ARTICLE IN PRESS
Metal nanoclusters 13

on the signal-to-noise ratio and exhibited excellent selec-


tivity for ATP detection over other ATP analogs GTP, CTP,
and UTP. More recently, Willner and coworkers [144] have
reported a hybrid system consisting of nucleic-acid func-
tionalized Ag NCs and grapheme oxide (GO) are used for
the development of fluorescent ATP sensors. The designed
nucleic acid consisting of the anti-ATP aptamer sequence
and conjugated to the nucleic-acid-protected Ag NCs acted
as functional probe. The Ag NCs were adsorbed onto GO
to yield the quenched Ag NCs. In the presence of ATP, the
resulting ATP-aptamer complex is desorbed from GO, lead-
ing to the fluorescence of the Ag NCs. This strategy shows
high sensitivity and selectivity for ATP detection.
Cocaine is a powerfully addictive stimulant drug that
increases the level of dopamine, abusing cocaine has a vari-
ety of adverse effects on the body. Simple and sensitive
detection of cocaine is very important for law enforcement
and clinical diagnostics. Dong group [140] designed a sim-
ple and convenient method for turn-on detection of cocaine
Figure 10 Schematic illustrations of the analysis of ATP using using DNA protected Ag NCs as fluorescence probe. The
DNA/Ag NCs-G-quadruplex/Hemin conjugates. design contains two DNA strands which are both chimeric
Reprinted from Ref. [143] with permission by American Chemi- conjugates of the aptamer sequence fragment and G-rich
cal Society. sequence fragment. The formed Ag NCs show weak fluores-
cence emission. In the presence of cocaine, however, the
two aptamer fragments bind cocaine, which in turn puts the
53.7 nM, which is far below the permissible limit (133 nM) of
two G-rich sequence fragments in proximity and the fluores-
AsIII in drinking water permitted by U.S. EPA.
cent intensity of DNA/Ag NCs enhances greatly. The strategy
shows high sensitivity and selectivity, and a detection limit
Small molecule sensors of 0.1 ␮M was obtained. In another study, they use dsDNA-
templated Cu NCs as fluorescent probe for cocaine detection
Adenosine triphosphate (ATP) is generally acknowledged as based on quenching the fluorescence of Cu NCs [141], and a
‘‘energy currency’’ in most animate beings, and plays an detection limit of 0.1 ␮M was obtained.
important role in most enzymatic activities [139]. The con- Dopamine (DA), as an important catecholamine neu-
centration and dissipative rate of ATP have been found to be rotransmitter, plays a vital role in the function of the
closely related to many diseases. Dong group [140] designed central nervous, renal, hormonal, and cardiovascular sys-
a simple and convenient method for turn-on detection of ATP tems. Abnormal DA concentrations in the brain may result
using DNA protected Ag NCs as fluorescence probe, which in serious diseases. Qu group [145] developed an easy pre-
shows high sensitivity and selectivity, and a detection limit pared fluorometric and colorimetric dual channel probe for
of 0.2 ␮M was obtained. In another study, they use dsDNA- DA detection with high sensitivity and selectivity by use
templated Cu NCs as fluorescent probe for ATP detection of BSA-stabilized Au NCs (Fig. 11). The BSA-Au NCs exhibit
based on quenching the fluorescence of Cu NCs [141], and strong fluorescence emission, while upon addition of DA,
a detection limit of 28 nM was obtained. Subsequently, Ye the Au NCs show a dramatic decrease of the fluorescence
group [142] demonstrated that the DNA/Ag NCs can be used intensity as a result of the photoinduced electron transfer
as a fluorescent molecular beacon for simple and selec- process from the electrostatically attached DA to the BSA-
tive detection of ATP using the guanine-rich (G-rich) DNA Au NCs. The detection limit of DA can be as low as 10 nM.
sequence as a signal transducer, which can induce the flu- In addition, the assay for DA can also be easy to implement
orescent enhancement of Ag NCs enabling the detection of for visual detection due to the observed inhibition of the
ATP. Recently, our group [143] has reported a novel DNA/Ag peroxidase-like activity of Au NCs in the presence of DA,
NCs-based photoinduced electron transfer (PET) system that with a detection limit of 10 nM. Both fluorometric and col-
enables the specific detection of ATP with high sensitivity. As orimetric methods exhibit excellent selectivity toward DA
shown in Fig. 10, the designed A-G4-ATP DNA assembles into over interfering substances.
a hairpin structure consisting of the G-quadruplex sequence In addition, other important small molecules, such as
(black) and the aptamer part against ATP (blue). This hairpin biothiols [146,147] including cysteine, homocysteine and
composition ensures that parts of the aptamer and G- glutathione, and hydrogen peroxide [148,149], can also be
quadruplex sequences are caged in the stem of the hairpin. detected by fluorescent metal NCs, which has been reviewed
Formation of the ATP-aptamer complex opens the hair- in detail by shang et al. [11].
pin structure and deprotects the G-quadruplex sequence,
accompanied by self-assembling of the G-quadruplex/hemin Protein sensors
complex. The photoinduced electrons of DNA/Ag NCs were
transferred to the G-quadruplex/hemin complex, leading The selective detection of proteins with fluorescent metal
to the quenching of the fluorescence of DNA/Ag NCs. This NCs as probes often needs to combine selective recogni-
approach allowed the detection limit of 8.0 nM for ATP based tion molecules (e.g. antibodies and aptamers). Considerable

Please cite this article in press as: L. Zhang, E. Wang, Nano Today (2014), http://dx.doi.org/10.1016/j.nantod.2014.02.010
+Model
NANTOD-359; No. of Pages 26 ARTICLE IN PRESS
14 L. Zhang, E. Wang

Figure 11 (A) Schematic illustration of the fluorescence response of the BSA-stabilized Au nanoclusters to dopamine. (B) Schematic
representation of peroxidase-like catalytic color reaction for sensitive sensing of dopamine.
Reprinted from Ref. [145] with permission by Elsevier.

effort has been devoted to constructing metal NCs-based form G-quadruplex structure, and released from the duplex
fluorescent protein sensors. Thrombin is a coagulation pro- structure. Then, G-rich DNA could enhance the fluorescence
tein in the bloodstream, which converts soluble fibrinogen intensity of DNA/Ag NCs, realizing the detection of throm-
into insoluble strands of fibrin as well as catalyzing many bin, and a detection limit of 10 nM was obtained. More
other coagulation-related reactions. Martinez and cowork- recently, Willner and coworkers [144] reported a hybrid sys-
ers [150] developed a new strategy for the detection of tem consisting of DNA functionalized Ag NCs and GO are
thrombin that combines the bright DNA-templated Ag NCs used for the development of fluorescent thrombin sensors.
with the specificity and strong binding affinity of DNA The designed nucleic acid consisting of the anti-thrombin
aptamers for thrombin. The used DNA sequence with two aptamer sequence and conjugated to the DNA-protected Ag
active domains: one domain was an aptamer for throm-
bin, and the other was a sequence able to generate Ag
NCs. The fluorescence of Ag NCs was quenched considerably
after the binding of thrombin with its aptamer. Through this
approach, nanomolar concentrations of thrombin could be
detected. Subsequently, Zhu and coworkers [151] presented
a binding-induced fluorescence turn-on assay of thrombin
using DNA/Ag NCs as probes. Key features of this assay
include aptamer binding-induced DNA hybridization and flu-
orescent enhancement of Ag NCs with G-rich DNA sequences.
When thrombin binds with its two aptamer, hybridization
process would be initiated between the complementary
sequences attached to each aptamer, thereby making the
G-rich overhang in proximity with Ag NCs and resulting in a
significant fluorescence enhancement. With this approach,
a detection limit of 1 nM was obtained for thrombin detec-
tion. Recently, our group [152] developed a new DNA/Ag
NCs-based beacon for turn-on detection of thrombin through
specific G-rich DNA sequences enhance the fluorescence
intensity of DNA/Ag NCs. This process could be modu- Figure 12 Schematic analysis of thrombin by the light-up
lated by the binding-induced strand displacement reactions DNA/Ag NCs system through binding-induced strand displace-
(Fig. 12). Initially, aptamer of thrombin inhibit the G-rich ment reactions.
DNA to enhance the fluorescence. However, in the presence Reprinted from Ref. [152] with permission by Royal Society of
of thrombin, thrombin could combine with its aptamer to Chemistry.

Please cite this article in press as: L. Zhang, E. Wang, Nano Today (2014), http://dx.doi.org/10.1016/j.nantod.2014.02.010
+Model
NANTOD-359; No. of Pages 26 ARTICLE IN PRESS
Metal nanoclusters 15

NCs acted as functional probe. The Ag NCs were adsorbed relies on the BSA scaffold degradation caused by the cys-
onto GO to yield the quenched Ag NCs. Desorption of the teine protease activity of papain and the specific inhibition
probe upon formation of the probe-thrombin aptamer com- of papain activity by Cys C. The fluorescence of BSA-Au NCs
plex leads to the recovery of fluorescence, achieving the can be effectively quenched by papain, and restored by the
detection of thrombin. coexistence of Cys C. This method enables sensitive and
Proteases are a major class of enzymes that catalyze selective measurement of Cys C with the detection limit of
the hydrolysis of peptide bonds to break down proteins into 4.0 ng/mL.
smaller pieces in a process known as proteolysis [153]. Ubiq-
uitous in nature, proteases are present in all living cells
and organisms, and are known to play a pivotal role in the DNA and miRNA sensors
development and control of many biological processes. Xia
group [154] demonstrated a new nanoscale platform based The detection and quantification of DNA are important
on protein-protected Au NCs for the fluorescence detection for in vivo real-time monitoring of cellular processes and
of proteases with high sensitivity and selectivity. The detec- for in vitro biosensing and clinical diagnosis. By virtue of
tion is based on the concept that the protein shell can serve the highly sequence-dependent generation of fluorescent
as a protective layer to prevent the fluorescent Au NCs from DNA/Ag NCs, our group [92] explored Ag NCs-based assay
being exposed to the O2 from ambient air. When the pro- for the detection of single nucleotide mutation, in partic-
tein shell is degraded by a protease, the O2 molecules can ular the mutation associated with sickle cell anemia. In
quickly access the Au NCs and thus quench the fluorescence. this study, DNA duplexes with an inserted cytosine loop
The advantages of this new detection system lies in its sim- have been developed as capping scaffolds for the gener-
plicity, low cost and ‘‘one-step’’ of preparation, as well as ation of fluorescent Ag NCs. The obtained results showed
the potential use with a wide range of protein substrates. that even a single-nucleotide mismatch located two bases
Plateletderived growth factor B-chain homodimer (PDGF- away from the nanocluster formation site would prohibit
BB) is a protein growth factor that is concerned with the generation of fluorescent Ag NCs. Subsequently, Ren and
controlling the proliferation or differentiation of cells coworkers [159] reported the site-specific growth of flu-
including fibroblasts, smooth muscle cells and glial cells. Its orescent Ag NCs by using a mismatched dsDNA template.
overexpression has been reported in some human tumors. Few-atom, molecular-scale Ag NCs are found to localize at
As a potential protein marker for cancer diagnosis, sensitive the mismatched site and this formed Ag NCs can be utilized
and rapid detection of PDGF-BB could help early diagnosis, as functional biological probes to identify single-nucleotide
treatment, and prognosis of cancers. Yang and coworkers polymorphisms. In addition, our group [102] also utilized
[155] reported a turn-on and homogeneous aptasensor for DNA-hosted Cu NCs for fluorescent identification of single
PDGF-BB detection, which relies on target induced forma- nucleotide polymorphisms. The fluorescence of Cu NCs is
tion of Ag NCs. The aptasensor contains two hairpin DNA very sensitive to base type located in the major groove of
probes, one consists of the aptamer sequence of PDGF-BB dsDNA. This intriguing finding provides a sensitive fluori-
and the other contains the Ag NCs nucleation sequence, metric diagnostic of the mismatch type in a specific DNA
which is blocked by the hairpin stem region. They can co- sequence.
exist metastably in the absence of PDGF-BB and maintain Based on the ‘‘light up’’ of DNA/Ag NCs in proximity to
hairpin structure. However, in the presence of PDGF-BB, the G-rich DNA sequences, DNA/Ag NCs have been designed for
binding of PDGF-BB with aptamer will result in the hybridiza- the analysis of DNA. Yeh et al. [160] designed a nanoclus-
tion of the two hairpin DNA structures, and release the ter beacon (NCB) consisting consists of two short linear DNA
Ag NCs nucleation sequence. In this case, Ag NCs can be probes (a NC probe and a G-rich probe). They were brought
formed via the reduction of Ag+ by NaBH4 . By monitoring into proximity through hybridization with the target DNA.
the increase in fluorescence intensity, a detection limit of The turn-on of red emission indicated the presence of tar-
0.37 nM was obtained for PDGF-BB detection. get DNA, a sequence from influenza A virus was detected
SsDNA-binding protein (SSB) is an important protein in with a signal/background (S/B) ratio of 175. Similar detec-
cells of all living organisms. It binds selectively and cooper- tion based on the moleclular beacon yields a S/B ratio of
atively to single-strand region of DNA to prevent premature 32, illustrating the advantages of this NCB. Subsequently,
annealing that is essential for DNA replication, recombi- they reported a chameleon NCB that lights up into different
nation, and repair [156]. Chang and coworkers [157] have colors upon binding single-nucleotide polymorphisms (SNP)
reported a DNA-Cu/Ag NCs-based approach for the highly targets [161]. They found the fluorescence emission color of
sensitive and selective detection of SSB.110 SSB can strongly a NCB can change substantially depending upon the align-
and specifically interact with ssDNA template, and thus ment between the Ag NCs and the DNA enhancer sequence.
change the conformation of the template, leading to the Chameleon NCBs exploit this color shift to directly detect
decreased fluorescence of ssDNA-stabilized Cu/Ag NCs. The SNPs, and this method has been validated on all single-
approach provided a limit of detection of 0.2 nM for SSB nucleotide substitution scenarios in three synthetic DNA
detection. targets, in six disease-related SNP targets (Fig. 13), and
Cystatin C (Cys C) is a significant cysteine protease in two clinical samples taken from patients with ovarian
inhibitor in human bodies, and is proposed as a fascinating serous borderline tumors. Samples with single-nucleotide
novel marker of glomerular filtration rate for kidney injury variations can be easily identified by the naked eye under
detection. Huang group [158] report a simple, immune- UV excitation, making this method a reliable and low-cost
independent and label-free method for Cys C detection using assay with a simple readout format. Recently, our group
BSA-stabilized Au NCs as fluorescent probes. This method [152] achieved to modulate the fluorescent enhancement

Please cite this article in press as: L. Zhang, E. Wang, Nano Today (2014), http://dx.doi.org/10.1016/j.nantod.2014.02.010
+Model
NANTOD-359; No. of Pages 26 ARTICLE IN PRESS
16 L. Zhang, E. Wang

Figure 13 Chameleon NCBs (cNCBs) was applied to detect a wide variety of disease-related SNPs, which covered all 6 types of
single-nucleotide substitution scenarios (C→G: Set 1, G→A: Set 2, C→T: Set 3, A→C: Set 4, T→A: Set 5, and G→T: Set 6). (A)
Design of cNCBs and nomenclature of the samples. (B) Detection results of the 6 cNCBs. As expected, all SNP targets were clearly
differentiated based on their emission spectra (for position +1 samples always being blue-shifted), proving the general use of cNCBs.
The integrated emission spectrum from 420 to 800 nm (normalized to unity) was divided into three elements: blue, 420—510 nm;
green, 510—640 nm; and red, 640—800 nm The percentiles of the 3 color elements in each detection sample are shown in the stacked
columns on the right, serving as a simple criterion for SNP scoring.
Reprinted from Ref. [161] with permission by American Chemical Society.

of DNA/Ag NCs through toehold-mediated strand displace- DNA/Ag NCs and G-quadruplex/hemin complexes, accompa-
ment reactions to place DNA/Ag NCs in proximity to G-rich nied by a decrease in the fluorescence of the DNA/Ag NCs.
DNA sequences. This approach could be used to develop a Based on this mechanism, we developed a new nanocluster-
new fluorescent sensing platform for the detection of tar- based molecular beacon for the detection of target DNA
get DNA with high selectivity and sensitivity. Furthermore, with high sensitivity and selectivity. In this PET process, a
to improve the sensitivity, exonuclease III-catalyzed ampli- parallel Gquadruplex and the sensing sequences are blocked
fication was applied in the present DNA detection system, by a duplex. The specific combination of target DNA with the
a very low detection limit of 0.1 nM was obtained, which is sensing sequence triggers the release of the G-quadruplex
much lower than that of the traditional unamplified strategy. and allows it to fold properly and bind hemin to form a stable
In addition, our group [143] also found a new phe- G-quadruplex/hemin complex, which promotes the electron
nomenon of photoinduced electron transfer (PET) between transfer from the DNA/Ag NCs to the hemin FeIII center, thus

Please cite this article in press as: L. Zhang, E. Wang, Nano Today (2014), http://dx.doi.org/10.1016/j.nantod.2014.02.010
+Model
NANTOD-359; No. of Pages 26 ARTICLE IN PRESS
Metal nanoclusters 17

Figure 14 Multiplexed analysis of the HBV gene (5) and of the HIV gene (8) using the near-infrared- and red-emitting Ag NCs/GO
hybrid system.
Reprinted from Ref. [144] with permission by American Chemical Society.

resulting in a decrease in the fluorescence intensity of the DNA-12nt-RED-172 probe to increase formation of secondary
DNA/Ag NCs. structure. The redesigned DNA-GG172-12nt-RED showed a
More recently, Willner group [144] reported on the inte- dramatic increase in red emission, and the presence of tar-
gration of DNA/Ag NCs with GO, and the implementation of get RNA-miR172 could efficiently quench its fluorescence,
these hybrid materials for the development of DNA sensors. providing an approach for RNA-miR172 detection.
As shown in Fig. 14, the nucleic acid sequence protecting the In addition, Zhang and coworkers [164] developed a
Ag NCs is elongated with the nucleic acid probe sequence simple, sensitive, and label-free method for miRNA detec-
that is complementary to the target DNA. The adsorption of tion using DNA/Ag NCs as effective electrochemical probes
the Ag NCs-functionalized DNA to GO induced the quenching (Fig. 15). The functional DNA probe integrates both recog-
of the fluorescence of Ag NCs. In the presence of target DNA, nition sequence for hybridization and template sequence
the formation of the duplex between Ag NCs-functionalized
DNA and target DNA triggers the desorption from the sur-
face of GO, leading to the regeneration of fluorescence.
More important, they implemented the sensing platform to
analyze the hepatitis B virus (HBV) gene, the human immun-
odeficiency virus (HIV) gene, and the syphilis (Treponema
pallidum) gene.
Detection of microRNAs (miRNAs) is also significant
because they play important roles in the regulation of gene
expression. The individual levels of miRNAs can be useful
biomarkers for cellular events or disease diagnosis. Yang and
Vosch [162] have designed a DNA/Ag NC probe that can mon-
itor the presence of target miRNA. The red fluorescence of
the DNA/Ag NC probe is diminished in the presence of target
miRNA, and high specificity toward detecting specific miRNA
sequences is performed. Subsequently, they investigated the
influence of nucleic acid secondary structure on the fast for-
mation of bright red emissive Ag NCs in a DNA sequence
(DNA-12nt-RED-160), and designed for the detection of a
microRNA sequence (RNA-miR160) [163]. Their results indi- Figure 15 Illustration of electrochemical detection of MiRNA
cate that the reduction in secondary structures led to the using oligonucleotide encapsulated Ag-NCs
decreased amount of red emissive Ag NCs. On the basis of the Reprinted from Ref. [164] with permission by American Chemi-
finding, they rearranged the sequence of the low-emissive cal Society.

Please cite this article in press as: L. Zhang, E. Wang, Nano Today (2014), http://dx.doi.org/10.1016/j.nantod.2014.02.010
+Model
NANTOD-359; No. of Pages 26 ARTICLE IN PRESS
18 L. Zhang, E. Wang

for in situ synthesis of Ag NCs, which appears to possess imaging (Fig. 16). Importantly, this does not occur in non-
exceptional metal mimic enzyme properties for catalyzing cancerous cells, as evidenced with human embryo liver cells
H2 O2 reduction. The miRNA assay employs gold electrodes (L02) used as controls. This dichotomy was exploited for a
to immobilize the molecular beacon (MB) probe. After the new strategy for specific fluorescent self-bio-marking of the
MB probe subsequently hybridizes with the target and func- tumors, which opens up promising opportunities for biomed-
tional probe, the DNA-encapsulated Ag NCs are brought to ical applications requiring specific and sensitive imaging of
the electrode surface and produce a detection signal, in tumors without direct injection of vectorized nanoparticles
response to H2 O2 reduction. A detection limit of 67 fM was or molecular fluorescent probes. Recently, Qu and Ren group
obtained for target miRNA with high selectivity. This is the [169] report the synthesis of the polyethyleneimine (PEI)-
first application of the electrocatalytic activity of Ag NCs in templated Au NCs (PEI-Au NCs) as an efficient carrier for
bioanalysis, which would be attractive for genetic analysis gene delivery. The PEI-Au NCs integrate the advantages of
and clinic biomedical application. PEI and Au NCs: the presence of Au NCs can effectively
decrease the cytotoxicity of PEI, making it possible to apply
them in biological systems, while the cationic polymer layer
Biological labeling and imaging PEI with positive charges is essential for enhanced gene
transfection efficiency. In addition, with excellent photo-
Metal NCs possess an attractive set of features, such luminescent properties, the Au NCs also endow the system
as ultrasmall size, good biocompatibility, brightness and with the versatility of fluorescent imaging, indicating a great
photostability, which renders them attractive alternatives potential as an ideal fluorescent probe to track the trans-
as fluorescent probes for biological labeling and imaging fection behavior.
[165,166]. Moreover, large Stoke shifts of metal NCs can pre- In addition, multifunctional nano-conjugates based on
vent spectral cross-talk and, thus, enhance the detection Au NCs have been applied in tumor imaging. Gu and
signal. In recent years, a number of works have reported coworkers [170] investigated the utilization of BSA pro-
biological labeling and imaging applications based on flu- tected Au NCs as fluorescent probes for tumor imaging.
orescent metal NCs. Lin group [167] reported a galvanic Au NCs were conjugated with methionine (Met) and MPA,
replacement route to prepare fluorescent Au NCs using a near-infrared fluorescent dye, giving a probe, Au-Met-
presynthesized and size-controlled Ag NCs as templates. The MPA. They investigated the internalization of fluorescent
obtained Au NCs show strong fluorescence and favorable bio- Au-BSA and Au-Met-MPA by L02 normal human cells and
compatibility. They used the Au NCs as fluorescent probes MCF-7 tumor cells. The fluorescence microscopy images
for cellular marking in CAL-27 cancer cells and MC3T3-E1 were shown in Fig. 17. With regard to the images of
normal cells, respectively. The results indicated that the flu- L02 cells, only weak red fluorescence emitted by the Au
orescence signals were not only distributed in the cytoplasm NCs in cells could be observed indicating that little Au-
but most came into the cellular nucleus for both CAL-27 and BSA or Au-Met-MPA had entered normal cells. By contrast,
MC3T3-E1. Later, Wang group [168] reported a novel strat- Au NCs in MCF-7 tumor cells showed strong red fluores-
egy for synthesis of Au NCs and applied them in fluorescence cence. The fluorescence of cells incubated with Au-Met-MPA
imaging in vivo. The fluorescent Au NCs were spontaneously which was found to have entered cytoplasm and nucleus
biosynthesized by cancerous cell (i.e., HepG2, human hep- of MCF-7 tumor cells could obviously eclipse that of the
atocarcinoma cell line; K562, leukemia cell line) through cells treated with Au-BSA. This clearly demonstrated that
Au(III) reduction inside cells cytoplasms, and ultimately con- the presence of Met on the surface of Au NCs endowed
centrated around their nucleoli, thus affording precise cell the tumor targeting capability, which increased its cellular

Figure 16 Schematic illustration of in situ biosynthesis of gold nanoclusters in cells and tumor imaging. HAuCl4 solution was
incubated in vitro with target cells or subcutaneously injected in vivo near a tumor. The sequestration and reduction of AuCl4 − anions
inside cells give rise to the progressive formation of gold nanoclusters. After incubation for 24—48 h, fluorescent gold nanoclusters
were observed inside the tumor cells or accumulated around the tumor.
Reprinted from Ref. [168] with permission by Nature Publishing Group.

Please cite this article in press as: L. Zhang, E. Wang, Nano Today (2014), http://dx.doi.org/10.1016/j.nantod.2014.02.010
+Model
NANTOD-359; No. of Pages 26 ARTICLE IN PRESS
Metal nanoclusters 19

Figure 17 The fluorescence microscopy images of L02 normal human cells and MCF-7 tumor cells after incubated with Au-BSA
and Au-Met-MPA.
Reprinted from Ref. [170] with permission by Elsevier.

uptake by MCF-7 tumor cells. Chen et al. [171] synthesized the specificity to target cancer cells due to the enhanced cell
multifunctional no-conjugated amphiphilic copolymers (Au uptake mediated by FA moiety. Furthermore, the nanocar-
NCs-PLAGPPS-FA) base on BSA protected Au NCS for tumor- riers could be tracked at the cellular level for advance
targeted drug delivery, which consisted by Au NCs as core therapy.
and folate (FA)-conjugated amphiphilic hyperbranched block Two-photo excitation confocal microscopy was used
copolymer as shell based on poly(L-lactide) (PLA) inner arm to observe biological imaging, which can afford some
and FA-conjugated sulfated polysaccharide (GPPS—FA) outer advantages, such as the ability of deeper imaging inside
arm. The nanocarriers displayed strong red emission at tissues and the reduced phototoxicity of NIR light. Shang
620 nm when excited at 480 nm and good biocompatibility. et al. [32] investigated the internalization of fluorescent
They used the nanocarriers as fluorescent probes for cel- D-penicillamine conjugated Au NCs (DPA-Au NCs) in the endo-
lular imaging in HeLa cells. The results indicated that the somal vesicles of human cervical cancer (HeLa) cells via
nanocarriers provided high anticancer activity and showed two-photo fluorescence imaging. They could easily observe

Please cite this article in press as: L. Zhang, E. Wang, Nano Today (2014), http://dx.doi.org/10.1016/j.nantod.2014.02.010
+Model
NANTOD-359; No. of Pages 26 ARTICLE IN PRESS
20 L. Zhang, E. Wang

Figure 18 Microscopic observation of internalization of the insulin-Au nanoclusters (NCs). Differentiated C2C12 myoblasts were
treated with insulin-Au NCs for 2 h. (a) Cell nucleus stained with 4 ,6-diamidino-2-phenylindole (DAPI, blue). (b) Actin fiber stained
with Alexa Fluor 488 phalloidin to confirm the cell boundary (green). (c) Insulin-Au NCs exhibit red luminescence. (d) Fluorescence
image overlay of the three images.
Reprinted from ref. [52] with permission by Wiley-VCH.

the bright fluorescence of Au NCs inside the cells. The DHLA. The Au NCs shows many attractive features includ-
images of 3D reconstruction further indicate that DPA-Au ing ultrasmall size, bright near-infrared luminescence, high
NCs inside the cells as well as attached to the outside of the colloidal stability, and good biocompatibility. Moreover,
plasma membrane. Recently, Liu et al. [52] reported the their fluorescence lifetime is longer than 100 ns. These
synthesis of fluorescent Au NCs by using insulin as protected properties make them attractive as labels in FLIM appli-
ligand. The as-prepared insulin-Au NCs show excellent bio- cations. They investigated the internalization of DHLA-Au
compatibility and retain the insulin bioactivity in reducing NCs by live HeLa cells. The results indicated that the
blood glucose. The uptake efficiency of insulin-Au NCs for distributions of Au NCs was in the intracellular and mem-
C2C12 cells may serve as a biomarker to distinguish the dif- brane regions and the Au NCs exhibited bright fluorescence
ferentiated versus undifferentiated C2C12 myoblasts. The luminescent with long fluorescence lifetimes (500—800 ns),
confocal images depicted in Fig. 18 clearly show that the red which could avoid significant autofluorescence emitted by
fluorescence of Au NCs is dispersed in the fully differentiated the cells. FLIM images not only reveal the uptake of Au
C2C12 mouse myoblasts. A further detailed two-photon z- NCs by the cells but also provide information on their
stacking study confirms that insulin-Au NCs entered into the changed local environment. Subsequently, they synthesized
cell and were distributed in the cytoplasm but not simply lipoic acid-protected Au NCs, which emit bright fluores-
adhered on the surface of the membranes. Moreover, they cence in the near-IR region and show the pronounced
also presented that the fluorescent Au NCs could be used in temperature dependence of the steady-state fluorescence
X-ray computed tomography (CT) imaging. emission spectra, the fluorescence intensity decreases up
Fluorescence lifetime imaging (FLIM) is also an alter- raising the temperature [124]. Moreover, the Au NCs exhibit
native technique for cell imaging that takes advantages excellent stability in biological media and display a char-
of the longer lifetime (>100 ns) of metal NCs than that acteristic, nonexponential fluorescence decay that can be
of auto-fluorescence of cellular organelles. Shang et al. fitted with a sum of three exponentials, with a short
[29] reported a facile strategy to synthesize water-soluble ( 1 ≈ 24 ns), a medium ( 2 ≈ 130 ns), and a long lifetime
fluorescent Au NCs stabilized with the bidentate ligand (600 ns <  3 < 750 ns). Those properties endow the Au NCs

Please cite this article in press as: L. Zhang, E. Wang, Nano Today (2014), http://dx.doi.org/10.1016/j.nantod.2014.02.010
+Model
NANTOD-359; No. of Pages 26 ARTICLE IN PRESS
Metal nanoclusters 21

Figure 19 Typical FLIM images of HeLa cells with internalized Au NCs at four different temperatures.
Reprinted from Ref. [124] with permission by Wiley-VCH.

with the capability for temperature sensing in HeLa human commercially available transfection reagent Lipofectamine,
cancer cells using FLIM. Lifetime maps were calculated on a and the internalization of C24 /Ag NCs was followed with
pixel-by-pixel basis by fitting the fluorescence decay of each confocal fluorescence microscopy. Bright near-infrared flu-
pixel with a tri-exponential function. As shown in Fig. 19, orescence was observed from inside the transfected HeLa
these images provide clear evidence that the long fluo- cells, which opens up the possibility for the utilization of
rescence lifetime components in the range of 600—1000 ns Ag NCs for biological labeling and imaging of living cells and
arise from internalized Au NCs rather than cellular autoflu- for monitoring the transfection process with limited harm to
orescence. With increasing temperature, the fluorescence the living cells.
lifetime decreased markedly from 970 ns at 14 ◦ C to 670 ns Combining the specific recognition of aptamer toward
at 43 ◦ C. CCRF-CEM live cells, Gao and coworkers have successfully
In addition to Au NCs, functional Ag NCs have been achieved the specifically marking the nucleus of live cells
employed for biological imaging. Dickson and coworkers [82] by DNA/Ag NCs [173]. Sgc8c aptamer, selected by cell-
synthesized Ag NCs with the linear poly(-acrylic acid) (PA) SELEX, has been shown to be a specific aptamer for the
as stabilizer. They found that the formed PA-Ag NCs readily endosomes of CCRF-CEM cells [174]. Eight cytosine bases
transfer NCs to high-affinity ssDNA sequences, which results were inserted at the 5 end of the sgc8c aptamer to form
in the loss of PA-Ag NCs fluorescence and generation of char- Msgc8, which was used for the synthesis of Ag NCs. The
acteristic DNA-encapsulated Ag NCs emission. Concomitant formed cluster—aptamer hybrids could specially target the
with the spectral shifts, cluster transfer from low-quantum- nucleus of CCRF-CEM cells after incubation with the cells for
yield PA-Ag NCs to high-quantum-yield ssDNA/Ag NCs results 2 hours at room temperature. Confocal microscope observa-
in a 10-fold increase in cluster brightness. Using this flu- tion showed that the cluster—aptamer hybrids were mainly
orogenic NCs shuttle, they fluorescently labeled cellular located in the nucleus of the live cells.
components by first conjugating C12 DNA to antibodies, stain- AS1411 is an antiproliferative G-rich phosphodiester DNA
ing with these nonfluorescent DNA-conjugated antibodies, and currently an anticancer agent under phase II clinical
and transferring NCs as the final step. The results demon- trials. Its function as an aptamer to nucleolin has led to
strate that the emission image from the NCs (Fig. 20A) the identification of nucleolin as a new molecular target for
colocalized well with that of the fluorescein isothiocyanate cancer therapy [175]. Zhu and coworkers [176] presented a
(FITC) (Fig. 20B), thus revealing antibody location and strategy to synthesize AS1411-functionalized Ag NCs through
indicating actin staining with NCs through NCs transfer a facile one-pot process. The obtained Ag NCs emitted
(Fig. 20C). Furthermore, they have successfully stained red color and the fluorescence quantum yield could reach
microtubules and labeled live cell with the same manner. 40.1%, which was beneficial to the applications of biological
Vosch and coworkers used C24 -encapsulated fluorescent Ag imaging. AS1411 retained its anticancer nature within the
NCs to monitor the transfection of DNA/Ag NCs inside liv- complex, and unexpectedly, functionalized Ag NCs demon-
ing HeLa cells [172]. The C24 /Ag NCs were complexed with a strated enhanced efficiency of growth inhibition compared

Please cite this article in press as: L. Zhang, E. Wang, Nano Today (2014), http://dx.doi.org/10.1016/j.nantod.2014.02.010
+Model
NANTOD-359; No. of Pages 26 ARTICLE IN PRESS
22 L. Zhang, E. Wang

Figure 20 (A) Fluorescence images of BPAEC from Ag NCs and (B) FITC tagged to actin C12 to indicate the location of actin, with
colocalization in (C).
Reprinted from Ref. [82] with permission by Wiley-VCH.

Figure 21 Schematic illustration and confocal fluorescence images of HeLa cells upon incubation with DHLA-AgNCs (25 ␮g/mL)
for 2 h in DMEM medium without (A—C) and with 100 ␮mol/L HSA (D—F). Cell membranes were stained with CellMaskTM DeepRed (B
and E).
Reprinted from Ref. [177] with permission by Springer-Verlag.

with naked AS1411. In addition, such functionalized Ag NCs reduced in the presence of HSA, as seen from the much lower
could be internalized into MCF-7 human breast cancer cells fluorescence intensity (Fig. 21D). The results indicate that
and stain the nucleus via receptor-mediated endocytosis. protein adsorption has a significant effect on the biological
The obtained results could improve the permeability of DNA response to Ag NCs exposure.
protection groups for live cell imaging and help understand
the therapeutic mechanism of AS1411. Conclusion and perspective
Recently, Shang et al. [177] rationally connected DHLA-
Ag NCs with a model protein, human serum albumin
In summary, we provide recent advances of the research
(HSA), which has been have been systematically investi-
progress on metal NCs, from the challenging synthesis,
gated by using a variety of techniques. The results show
unique properties to the promising application in analyt-
that the physicochemical properties of both proteins and
ical sensors and biological imaging. Now, metal NCs have
Ag NCs undergo changes upon their interactions. However,
been developed as a new class of fluorophores, their excel-
it appears that the overall conformation of HSA remains
lent properties endow them attractive fluorescent probes
essentially unaffected in the complex. Furthermore, they
for biological applications. These research results reveal
investigated the biological implications of protein adsorp-
that metal NCs can open many good opportunities in an
tion by evaluating responses of HeLa cells to Ag NC exposure
extremely multidisciplinary environment for promoting the
through live-cell fluorescence microscopy. As shown in
rapid developments of different research fields. Despite
Fig. 21, intense fluorescence emission from Ag NCs was
substantial progress in metal NCs, a variety of challenges
observed from inside the cells, suggesting an efficient inter-
remains and needs to be addressed.
nalization of these luminescent particles by the cells. In
At present, one of the major challenges for the deeper
contrast to the pronounced uptake of bare Ag NCs, the
development of metal NCs has been lack of an efficient
amount of Ag NCs internalized by the cells is substantially
route for preparing size precisely controlled metal NCs

Please cite this article in press as: L. Zhang, E. Wang, Nano Today (2014), http://dx.doi.org/10.1016/j.nantod.2014.02.010
+Model
NANTOD-359; No. of Pages 26 ARTICLE IN PRESS
Metal nanoclusters 23

with high quality and high yield due to the properties of [10] S. Choi, R.M. Dickson, J. Yu, Chem. Soc. Rev. 41 (2012)
metal NCs are strongly dependent on the size and the cur- 1867—1891.
rently reported metal NCs possess relatively low QY. In [11] L. Shang, S. Dong, G.U. Nienhaus, Nano Today 6 (2011)
addition, the high monodispersity and purity are also the 401—418.
critical points of the synthesis of metal NCs. Thus, how to [12] Y. Lu, W. Chen, Chem. Soc. Rev. 41 (2012) 3594—3623.
synthesize size-controlled metal NCs and how to purify the [13] H. Qian, M. Zhu, Z. Wu, R. Jin, Acc. Chem. Res. 45 (2012)
1470—1479.
synthesized metal NCs will be challenging work. Up to now,
[14] Y.-C. Shiang, C.-C. Huang, W.-Y. Chen, P.-C. Chen, H.-T.
many researches have been concentrated on Au and Ag NCs. Chang, J. Mater. Chem. 22 (2012) 12972—12982.
Only in recent years, the studies on Cu and other metal NCs [15] G. Li, R. Jin, Acc. Chem. Res. 46 (2013) 1749—1758.
emerged gradually, and the emerging metal NCS also show [16] I. Diez, R.H.A. Ras, Nanoscale 3 (2011) 1963—1970.
unique and excellent properties. Those properties make [17] I. Díez, R. Ras, Advanced Fluorescence Reporters in Chemistry
them potentially useful in a wide variety of applications in and Biology II, Springer, 2010, pp. 307—332.
fluorescent detection and other fields. Therefore, it is nec- [18] N. de Souza, Nat. Methods 4 (2007) 540.
essary to develop methods for the synthesis of other new [19] K. Nienhaus, G. Ulrich Nienhaus, Chem. Soc. Rev. 43 (2014)
metal NCs, including the size-controlled synthesis, structure 1088—1106.
characterization and their property studies. The new metal [20] M.A.H. Muhammed, T. Pradeep, in: A.P. Demchenko (Ed.),
Advanced Fluorescence Reporters in Chemistry and Biology
NCs may exhibit surprising properties, which will broaden
II, Springer, Berlin, Heidelberg, 2010, pp. 333—353.
the applications of metal NCs. [21] Y. Negishi, K. Nobusada, T. Tsukuda, J. Am. Chem. Soc. 127
Many reports have suggested that these fluorescent metal (2005) 5261—5270.
NCs are promising tools for sensing and intracellular imag- [22] Y. Negishi, Y. Takasugi, S. Sato, H. Yao, K. Kimura, T. Tsukuda,
ing. It could be useful to simultaneously endow metal NCs J. Am. Chem. Soc. 126 (2004) 6518—6519.
with other properties, such bi- or multifunctional probes [23] J.-i. Nishigaki, R. Tsunoyama, H. Tsunoyama, N. Ichikuni, S.
bear a lot of promise for applications. Although many unique Yamazoe, Y. Negishi, M. Ito, T. Matsuo, K. Tamao, T. Tsukuda,
optical and electronic properties have been discovered, the J. Am. Chem. Soc. 134 (2012) 14295—14297.
structures and fundamental understanding of fluorescent [24] P. Maity, S. Xie, M. Yamauchi, T. Tsukuda, Nanoscale 4 (2012)
4027—4037.
metal NCs are still lacking. The sensitivity and stability of
[25] D.-e. Jiang, Nanoscale 5 (2013) 7149—7160.
metal NCs to their environments offer the opportunity for
[26] A. Sanchez-Castillo, C. Noguez, I.L. Garzon, J. Am. Chem.
the applications in sensors and imaging. However, the coor- Soc. 132 (2010) 1504—1505.
dination nature of the NCs-protection group complex implies [27] O. Varnavski, G. Ramakrishna, J. Kim, D. Lee, T. Goodson, J.
that the stability of metal NCs is affected by the presence Am. Chem. Soc. 132 (2010) 16—17.
of other strong competing ligands in aqueous solution. The [28] M. Yu, C. Zhou, J. Liu, J.D. Hankins, J. Zheng, J. Am. Chem.
intracellular stability of metal NCs is also a challenging issue. Soc. 133 (2011) 11014—11017.
Until the issues are fully resolved, metal NCs will be more [29] L. Shang, N. Azadfar, F. Stockmar, W. Send, V. Trouil-
useful in vitro/in vivo imaging under one-photon or two- let, M. Bruns, D. Gerthsen, G.U. Nienhaus, Small 7 (2011)
photon excitation. 2614—2620.
[30] T. Huang, R.W. Murray, J. Phys. Chem. B 105 (2001)
Overall, significant progress has been achieved in the
12498—12502.
field of metal NCs in the past decades. Many challenges have
[31] D. Lee, R.L. Donkers, G. Wang, A.S. Harper, R.W. Murray, J.
to be faced and resolved in the future work. With contin- Am. Chem. Soc. 126 (2004) 6193—6199.
ued development, we believe that metal NCs will find wide [32] L. Shang, R.M. Doerlich, S. Brandholt, R. Schneider, V. Trouil-
applications in addressing important issues related to more let, M. Bruns, D. Gerthsen, G.U. Nienhaus, Nanoscale 3 (2011)
different fields. 2009—2014.
[33] M. Zhu, E. Lanni, N. Garg, M.E. Bier, R. Jin, J. Am. Chem. Soc.
130 (2008) 1138—1139.
Acknowledgments
[34] Z. Wu, J. Suhan, R. Jin, J. Mater. Chem. 19 (2009) 622—626.
[35] R. Jin, H. Qian, Z. Wu, Y. Zhu, M. Zhu, A. Mohanty, N. Garg,
This work was supported by the National Natural Science J. Phys. Chem. Lett. 1 (2010) 2903—2910.
Foundation of China (Nos. 21190040 and 91227114) and 973 [36] Z. Wu, R. Jin, Nano Lett. 10 (2010) 2568—2573.
Project (Nos. 2010CB33600 and 2011CB911000). [37] T.P. Bigioni, R.L. Whetten, O. Dag, J. Phys. Chem. B 104 (2000)
6983—6986.
[38] S. Kumar, R. Jin, Nanoscale 4 (2012) 4222—4227.
References [39] J. Zheng, J.T. Petty, R.M. Dickson, J. Am. Chem. Soc. 125
(2003) 7780—7781.
[1] G. Aragay, F. Pino, A. Merkoçi, Chem. Rev. 112 (2012) [40] J. Zheng, C.W. Zhang, R.M. Dickson, Phys. Rev. Lett. 93 (2004)
5317—5338. 077402.
[2] S. Guo, S. Dong, Chem. Soc. Rev. 40 (2011) 2644—2672. [41] M.A.H. Muhammed, S. Ramesh, S.S. Sinha, S.K. Pal, T.
[3] S. Guo, E. Wang, Acc. Chem. Res. 44 (2011) 491—500. Pradeep, Nano Res. 1 (2008) 333—340.
[4] T.K. Sau, A.L. Rogach, Adv. Mater. 22 (2010) 1781—1804. [42] H. Duan, S. Nie, J. Am. Chem. Soc. 129 (2007) 2412—2413.
[5] C.M. Cobley, J. Chen, E.C. Cho, L.V. Wang, Y. Xia, Chem. Soc. [43] C.-A.J. Lin, T.-Y. Yang, C.-H. Lee, S.H. Huang, R.A. Sperling,
Rev. 40 (2011) 44—56. M. Zanella, J.K. Li, J.-L. Shen, H.-H. Wang, H.-I. Yeh, W.J.
[6] S. Guo, E. Wang, Nano Today 6 (2011) 240—264. Parak, W.H. Chang, ACS Nano 3 (2009) 395—401.
[7] J. Zheng, P.R. Nicovich, R.M. Dickson, Annu. Rev. Phys. Chem. [44] Y. Shichibu, Y. Negishi, T. Tsukuda, T. Teranishi, J. Am. Chem.
(2007) 409—431. Soc. 127 (2005) 13464—13465.
[8] B. Han, E. Wang, Anal. Bioanal. Chem. 402 (2012) 129—138. [45] Y. Shichibu, Y. Negishi, H. Tsunoyama, M. Kanehara, T. Teran-
[9] A. Latorre, A. Somoza, Chembiochem 13 (2012) 951—958. ishi, T. Tsukuda, Small 3 (2007) 835—839.

Please cite this article in press as: L. Zhang, E. Wang, Nano Today (2014), http://dx.doi.org/10.1016/j.nantod.2014.02.010
+Model
NANTOD-359; No. of Pages 26 ARTICLE IN PRESS
24 L. Zhang, E. Wang

[46] M.A.H. Muhammed, P.K. Verma, S.K. Pal, R.C.A. Kumar, S. [78] T. Vosch, Y. Antoku, J.-C. Hsiang, C.I. Richards, J.I. Gon-
Paul, R.V. Omkumar, T. Pradeep, Chem. Eur. J. 15 (2009) zalez, R.M. Dickson, Proc. Natl. Acad. Sci. USA 104 (2007)
10110—10120. 12616—12621.
[47] R. Balasubramanian, R. Guo, A.J. Mills, R.W. Murray, J. Am. [79] C.I. Richards, S. Choi, J.-C. Hsiang, Y. Antoku, T. Vosch, A.
Chem. Soc. 127 (2005) 8126—8132. Bongiorno, Y.-L. Tzeng, R.M. Dickson, J. Am. Chem. Soc. 130
[48] J. Xie, Y. Zheng, J.Y. Ying, J. Am. Chem. Soc. 131 (2009) (2008) 5038—5039.
888—889. [80] E.G. Gwinn, P. O’Neill, A.J. Guerrero, D. Bouwmeester, D.K.
[49] H. Wei, Z. Wang, L. Yang, S. Tian, C. Hou, Y. Lu, Analyst 135 Fygenson, Adv. Mater. 20 (2008) 279—283.
(2010) 1406—1410. [81] C.I. Richards, J.-C. Hsiang, D. Senapati, S. Patel, J. Yu,
[50] J.S. Mohanty, P.L. Xavier, K. Chaudhari, M.S. Bootharaju, T. Vosch, R.M. Dickson, J. Am. Chem. Soc. 131 (2009)
N. Goswami, S.K. Pal, T. Pradeep, Nanoscale 4 (2012) 4619—4621.
4255—4262. [82] J. Yu, S. Choi, R.M. Dickson, Angew. Chem. Int. Ed. 48 (2009)
[51] F. Wen, Y. Dong, L. Feng, S. Wang, S. Zhang, X. Zhang, Anal. 318—320.
Chem. 83 (2011) 1193—1196. [83] J. Sharma, R.C. Rocha, M.L. Phipps, H.-C. Yeh, K.A. Balatsky,
[52] C.-L. Liu, H.-T. Wu, Y.-H. Hsiao, C.-W. Lai, C.-W. Shih, Y.- D.M. Vu, A.P. Shreve, J.H. Werner, J.S. Martinez, Nanoscale 4
K. Peng, K.-C. Tang, H.-W. Chang, Y.-C. Chien, J.-K. Hsiao, (2012) 4107—4110.
J.-T. Cheng, P.-T. Chou, Angew. Chem. Int. Ed. 50 (2011) [84] S.H. Yau, N. Abeyasinghe, M. Orr, L. Upton, O. Varnavski, J.H.
7056—7060. Werner, H.-C. Yeh, J. Sharma, A.P. Shreve, J.S. Martinez, T.
[53] H. Kawasaki, K. Hamaguchi, I. Osaka, R. Arakawa, Adv. Funct. Goodson III, Nanoscale 4 (2012) 4247—4254.
Mater. 21 (2011) 3508—3515. [85] W. Guo, R. Orbach, I. Mironi-Harpaz, D. Seliktar, I. Willner,
[54] P.L. Xavier, K. Chaudhari, P.K. Verma, S.K. Pal, T. Pradeep, Small 9 (2013) 3748—3752.
Nanoscale 2 (2010) 2769—2776. [86] T. Li, L. Zhang, J. Ai, S. Dong, E. Wang, Acs Nano 5 (2011)
[55] C. Sun, H. Yang, Y. Yuan, X. Tian, L. Wang, Y. Guo, L. Xu, J. 6334—6338.
Lei, N. Gao, G.J. Anderson, X.-J. Liang, C. Chen, Y. Zhao, G. [87] J. Yuan, W. Guo, E. Wang, Anal. Chim. Acta 706 (2011)
Nie, J. Am. Chem. Soc. 133 (2011) 8617—8624. 338—342.
[56] T.A.C. Kennedy, J.L. MacLean, J. Liu, Chem. Commun. 48 [88] W. Guo, J. Yuan, E. Wang, Chem. Commun. 47 (2011)
(2012) 6845—6847. 10930—10932.
[57] G. Liu, Y. Shao, F. Wu, S. Xu, J. Peng, L. Liu, Nanotechnology [89] J.T. Petty, O.O. Sergev, D.A. Nicholson, P.M. Goodwin, B. Giri,
24 (2013) 015503. D.R. McMullan, Anal. Chem. 85 (2013) 9868—9876.
[58] J. Zheng, R.M. Dickson, J. Am. Chem. Soc. 124 (2002) [90] J.T. Petty, J. Zheng, N.V. Hud, R.M. Dickson, J. Am. Chem.
13982—13983. Soc. 126 (2004) 5207—5212.
[59] W. Lesniak, A.U. Bielinska, K. Sun, K.W. Janczak, X. Shi, J.R. [91] B. Sengupta, K. Springer, J.G. Buckman, S.P. Story, O.H.
Baker, L.P. Balogh, Nano Lett. 5 (2005) 2123—2130. Abe, Z.W. Hasan, Z.D. Prudowsky, S.E. Rudisill, N.N.
[60] L. Shang, S. Dong, Chem. Commun. (2008) 1088—1090. Degtyareva, J.T. Petty, J. Phys. Chem. C 113 (2009)
[61] I. Diez, M. Pusa, S. Kulmala, H. Jiang, A. Walther, A.S. Gold- 19518—19524.
mann, A.H.E. Muller, O. Ikkala, R.H.A. Ras, Angew. Chem. Int. [92] W. Guo, J. Yuan, Q. Dong, E. Wang, J. Am. Chem. Soc. 132
Ed. 48 (2009) 2122—2125. (2010) 932—933.
[62] H. Xu, K.S. Suslick, ACS Nano 4 (2010) 3209—3214. [93] J. Wu, Y. Fu, Z. He, Y. Han, L. Zheng, J. Zhang, W. Li, J. Phys.
[63] Z. Shen, H. Duan, H. Frey, Adv. Mater. 19 (2007) 349—352. Chem. B 116 (2012) 1655—1665.
[64] J.G. Zhang, S.Q. Xu, E. Kumacheva, Adv. Mater. 17 (2005) [94] L. Feng, Z. Huang, J. Ren, X. Qu, Nucleic Acids Res. 40 (2012)
2336—2340. e122.
[65] M.A.H. Muhammed, F. Aldeek, G. Palui, L. Trapiella-Alfonso, [95] M. Zhao, L. Sun, R.M. Crooks, J. Am. Chem. Soc. 120 (1998)
H. Mattoussi, ACS Nano 6 (2012) 8950—8961. 4877—4878.
[66] T. Wen, F. Qu, N.B. Li, H.Q. Luo, Anal. Chim. Acta 749 (2012) [96] L. Balogh, D.A. Tomalia, J. Am. Chem. Soc. 120 (1998)
56—62. 7355—7356.
[67] T.U.B. Rao, T. Pradeep, Angew. Chem. Int. Ed. 49 (2010) [97] C. Vazquez-Vazquez, M. Banobre-Lopez, A. Mitra, M.
3925—3929. Arturo Lopez-Quintela, J. Rivas, Langmuir 25 (2009)
[68] T. Zhou, M. Rong, Z. Cai, C.J. Yang, X. Chen, Nanoscale 4 8208—8216.
(2012) 4103—4106. [98] N. Vilar-Vidal, M.C. Blanco, M.A. Lopez-Quintela, J. Rivas, C.
[69] J. Yu, S.A. Patel, R.M. Dickson, Angew. Chem. Int. Ed. 46 Serra, J. Phys. Chem. C 114 (2010) 15924—15930.
(2007) 2028—2030. [99] H. Kawasaki, Y. Kosaka, Y. Myoujin, T. Narushima, T.
[70] S.S. Narayanan, S.K. Pal, J. Phys. Chem. C 112 (2008) Yonezawa, R. Arakawa, Chem. Commun. 47 (2011)
4874—4879. 7740—7742.
[71] C. Guo, J. Irudayaraj, Anal. Chem. 83 (2011) 2883—2889. [100] W. Wei, Y. Lu, W. Chen, S. Chen, J. Am. Chem. Soc. 133 (2011)
[72] T. Zhou, Y. Huang, W. Li, Z. Cai, F. Luo, C.J. Yang, X. Chen, 2060—2063.
Nanoscale 4 (2012) 5312—5315. [101] A. Rotaru, S. Dutta, E. Jentzsch, K. Gothelf, A. Mokhir, Angew.
[73] T.K. Mandal, M.S. Fleming, D.R. Walt, Nano Lett. 2 (2002) Chem. Int. Ed. 49 (2010) 5665—5667.
3—7. [102] X. Jia, J. Li, L. Han, J. Ren, X. Yang, E. Wang, Acs Nano 6
[74] Y. Cui, Y. Wang, R. Liu, Z. Sun, Y. Wei, Y. Zhao, X. Gao, ACS (2012) 3311—3317.
Nano 5 (2011) 8684—8689. [103] G. Liu, Y. Shao, J. Peng, W. Dai, L. Liu, S. Xu, F. Wu, X. Wu,
[75] C.M. Ritchie, K.R. Johnsen, J.R. Kiser, Y. Antoku, R.M. Dick- Nanotechnology 24 (2013) 345502.
son, J.T. Petty, J. Phys. Chem. C 111 (2007) 175—181. [104] K. Yamamoto, T. Imaoka, W.-J. Chun, O. Enoki, H. Katoh, M.
[76] G. Shemer, O. Krichevski, G. Markovich, T. Molotsky, Takenaga, A. Sonoi, Nat. Chem. 1 (2009) 397—402.
I. Lubitz, A.B. Kotlyar, J. Am. Chem. Soc. 128 (2006) [105] H. Kawasaki, H. Yamamoto, H. Fujimori, R. Arakawa,
11006—11007. M. Inada, Y. Iwasaki, Chem. Commun. 46 (2010)
[77] B. Sengupta, C.M. Ritchie, J.G. Buckman, K.R. Johnsen, 3759—3761.
P.M. Goodwin, J.T. Petty, J. Phys. Chem. C 112 (2008) [106] S.-I. Tanaka, J. Miyazaki, D.K. Tiwari, T. Jin, Y. Inouye, Angew.
18776—18782. Chem. Int. Ed. 50 (2011) 431—435.

Please cite this article in press as: L. Zhang, E. Wang, Nano Today (2014), http://dx.doi.org/10.1016/j.nantod.2014.02.010
+Model
NANTOD-359; No. of Pages 26 ARTICLE IN PRESS
Metal nanoclusters 25

[107] M. Hyotanishi, Y. Isomura, H. Yamamoto, H. Kawasaki, Y. [142] M. Zhang, S.-M. Guo, Y.-R. Li, P. Zuo, B.-C. Ye, Chem. Com-
Obora, Chem. Commun. 47 (2011) 5750—5752. mun. 48 (2012) 5488—5490.
[108] W.I. Lee, Y. Bae, A.J. Bard, J. Am. Chem. Soc. 126 (2004) [143] L. Zhang, J. Zhu, S. Guo, T. Li, J. Li, E. Wang, J. Am. Chem.
8358—8359. Soc. 135 (2013) 2403—2406.
[109] B.A. Collings, K. Athanassenas, D. Lacombe, D.M. Rayner, P.A. [144] X. Liu, F. Wang, R. Aizen, O. Yehezkeli, I. Willner, J. Am.
Hackett, J. Chem. Phys. 101 (1994) 3506—3513. Chem. Soc. 135 (2013) 11832—11839.
[110] M. Zhu, C.M. Aikens, F.J. Hollander, G.C. Schatz, R. Jin, J. [145] Y. Tao, Y. Lin, J. Ren, X. Qu, Biosens. Bioelectron. 42 (2013)
Am. Chem. Soc. 130 (2008) 5883—5885. 41—46.
[111] B. Adhikari, A. Banerjee, Chem. Mater. 22 (2010) 4364—4371. [146] B. Han, E. Wang, Biosens. Bioelectron. 26 (2011) 2585—2589.
[112] P.T.C. So, C.Y. Dong, B.R. Masters, K.M. Berland, Annu. Rev. [147] L. Shang, S. Dong, Biosens. Bioelectron. 24 (2009)
Biomed. Eng. 2 (2000) 399—429. 1569—1573.
[113] G. Ramakrishna, O. Varnavski, J. Kim, D. Lee, T. Goodson, J. [148] L. Shang, S. Dong, Anal. Chem. 81 (2009) 1465—1470.
Am. Chem. Soc. 130 (2008) 5032—5033. [149] L. Shang, H. Chen, L. Deng, S. Dong, Biosens. Bioelectron. 23
[114] L. Polavarapu, M. Manna, Q.-H. Xu, Nanoscale 3 (2011) (2008) 1180—1184.
429—434. [150] J. Sharma, H.-C. Yeh, H. Yoo, J.H. Werner, J.S. Martinez,
[115] S.A. Patel, C.I. Richards, J.-C. Hsiang, R.M. Dickson, J. Am. Chem. Commun. 47 (2011) 2294—2296.
Chem. Soc. 130 (2008) 11602—11603. [151] J. Li, X. Zhong, H. Zhang, X.C. Le, J.-J. Zhu, Anal. Chem. 84
[116] L. Zhang, J. Li, Y. Xu, Y. Zhai, Y. Li, E. Wang, Talanta 79 (2009) (2012) 5170—5174.
454—459. [152] L. Zhang, J. Zhu, Z. Zhou, S. Guo, J. Li, S. Dong, E. Wang,
[117] Y.-M. Fang, J. Song, J. Li, Y.-W. Wang, H.-H. Yang, J.-J. Sun, Chem. Sci. 4 (2013) 4004—4010.
G.-N. Chen, Chem. Commun. 47 (2011) 2369—2371. [153] M.S. Wolfe, Chem. Rev. 109 (2009) 1599—1612.
[118] L. Li, H. Liu, Y. Shen, J. Zhang, J.-J. Zhu, Anal. Chem. 83 [154] Y. Wang, Y. Wang, F. Zhou, P. Kim, Y. Xia, Small 8 (2012)
(2011) 661—665. 3769—3773.
[119] W. Guo, J. Yuan, E. Wang, Chem. Commun. 48 (2012) [155] J.-J. Liu, X.-R. Song, Y.-W. Wang, A.-X. Zheng, G.-N. Chen,
3076—3078. H.-H. Yang, Anal. Chim. Acta 749 (2012) 70—74.
[120] H. Chen, X. Kou, Z. Yang, W. Ni, J. Wang, Langmuir 24 (2008) [156] H. Kuhn, V.V. Demidov, J.M. Coull, M.J. Fiandaca, B.D. Gildea,
5233—5237. M.D. Frank-Kamenetskii, J. Am. Chem. Soc. 124 (2002)
[121] R. Zhou, M. Shi, X. Chen, M. Wang, H. Chen, Chem. Eur. J. 15 1097—1103.
(2009) 4944—4951. [157] G.-Y. Lan, W.-Y. Chen, H.-T. Chang, Analyst 136 (2011)
[122] L. Maretti, P.S. Billone, Y. Liu, J.C. Scaiano, J. Am. Chem. 3623—3628.
Soc. 131 (2009) 13972—13980. [158] H. Lin, L. Li, C. Lei, X. Xu, Z. Nie, M. Guo, Y. Huang, S. Yao,
[123] H. Qian, M.Y. Sfeir, R. Jin, J. Phys. Chem. C 114 (2010) Biosens. Bioelectron. 41 (2013) 256—261.
19935—19940. [159] Z. Huang, F. Pu, D. Hu, C. Wang, J. Ren, X. Qu, Chem. Eur. J.
[124] L. Shang, F. Stockmar, N. Azadfar, G.U. Nienhaus, Angew. 17 (2011) 3774—3780.
Chem. Int. Ed. 52 (2013) 11154—11157. [160] H.-C. Yeh, J. Sharma, J.J. Han, J.S. Martinez, J.H. Werner,
[125] S. Raut, R. Chib, R. Rich, D. Shumilov, Z. Gryczynski, I. Nano Lett. 10 (2010) 3106—3110.
Gryczynski, Nanoscale 5 (2013) 3441—3446. [161] H.-C. Yeh, J. Sharma, I.-M. Shih, D.M. Vu, J.S. Martinez, J.H.
[126] W. Guo, J. Yuan, E. Wang, Chem. Commun. (2009) 3395—3397. Werner, J. Am. Chem. Soc. 134 (2012) 11550—11558.
[127] L. Deng, Z. Zhou, J. Li, T. Li, S. Dong, Chem. Commun. 47 [162] S.W. Yang, T. Vosch, Anal. Chem. 83 (2011) 6935—6939.
(2011) 11065—11067. [163] P. Shah, A. Rørvig-Lund, S.B. Chaabane, P.W. Thulstrup, H.G.
[128] L. Shang, L. Yang, F. Stockmar, R. Popescu, V. Trouillet, Kjaergaard, E. Fron, J. Hofkens, S.W. Yang, T. Vosch, ACS Nano
M. Bruns, D. Gerthsen, G.U. Nienhaus, Nanoscale 4 (2012) 6 (2012) 8803—8814.
4155—4160. [164] H. Dong, S. Jin, H. Ju, K. Hao, L.-P. Xu, H. Lu, X. Zhang, Anal.
[129] L. Zhang, J. Zhu, J. Ai, Z. Zhou, X. Jia, E. Wang, Biosens. Chem. 84 (2012) 8670—8674.
Bioelectron. 39 (2013) 268—273. [165] W.J. Stark, Angew. Chem. Int. Ed. 50 (2011) 1242—1258.
[130] L. Shang, S. Dong, J. Mater. Chem. 18 (2008) 4636—4640. [166] L. Shang, G.U. Nienhaus, Mater. Today 16 (2013) 58—66.
[131] G.-Y. Lan, C.-C. Huang, H.-T. Chang, Chem. Commun. 46 [167] C. Wang, Y. Wang, L. Xu, X. Shi, X. Li, X. Xu, H. Sun, B. Yang,
(2010) 1257—1259. Q. Lin, Small 9 (2013) 413—420.
[132] Y.-T. Su, G.-Y. Lan, W.-Y. Chen, H.-T. Chang, Anal. Chem. 82 [168] J. Wang, G. Zhang, Q. Li, H. Jiang, C. Liu, C. Amatore, X.
(2010) 8566—8572. Wang, Sci. Rep. 3 (2013) 1157.
[133] B. Li, Y. Du, S. Dong, Anal. Chim. Acta 644 (2009) 78—82. [169] Y. Tao, Z. Li, E. Ju, J. Ren, X. Qu, Nanoscale 5 (2013)
[134] Z. Wu, M. Wang, J. Yang, X. Zheng, W. Cai, G. Meng, H. Qian, 6154—6160.
H. Wang, R. Jin, Small 8 (2012) 2028—2035. [170] H. Chen, B. Li, X. Ren, S. Li, Y. Ma, S. Cui, Y. Gu, Biomaterials
[135] D. Li, J. Li, X. Jia, Y. Xia, X. Zhang, E. Wang, Anal. Chim. Acta 33 (2012) 8461—8476.
804 (2013) 98—103. [171] T. Chen, S. Xu, T. Zhao, L. Zhu, D. Wei, Y. Li, H. Zhang, C.
[136] S. Liu, F. Lu, J.-J. Zhu, Chem. Commun. 47 (2011) Zhao, ACS Appl. Mater. Interfaces 4 (2012) 5766—5774.
2661—2663. [172] Y. Antoku, J.-i. Hotta, H. Mizuno, R.M. Dickson, J. Hofkens,
[137] H. Zhang, Q. Liu, W. Thanh, Z. Yun, G. Li, J. Liu, G. Jiang, T. Vosch, Photochem. Photobiol. Sci. 9 (2010) 716—721.
Anal. Chim. Acta 770 (2013) 140—146. [173] Z. Sun, Y. Wang, Y. Wei, R. Liu, H. Zhu, Y. Cui, Y. Zhao, X. Gao,
[138] S. Roy, G. Palui, A. Banerjee, Nanoscale 4 (2012) Chem. Commun. 47 (2011) 11960—11962.
2734—2740. [174] D. Shangguan, Z. Tang, P. Mallikaratchy, Z. Xiao, W. Tan,
[139] L. Zhang, H. Wei, J. Li, T. Li, D. Li, Y. Li, E. Wang, Biosens. Chembiochem 8 (2007) 603—606.
Bioelectron. 25 (2010) 1897—1901. [175] P.J. Bates, D.A. Laber, D.M. Miller, S.D. Thomas, J.O. Trent,
[140] Z. Zhou, Y. Du, S. Dong, Biosens. Bioelectron. 28 (2011) Exp. Mol. Pathol. 86 (2009) 151—164.
33—37. [176] J. Li, X. Zhong, F. Cheng, J.-R. Zhang, L.-P. Jiang, J.-J. Zhu,
[141] Z. Zhou, Y. Du, S. Dong, Anal. Chem. 83 (2011) 5122—5127. Anal. Chem. 84 (2012) 4140—4146.

Please cite this article in press as: L. Zhang, E. Wang, Nano Today (2014), http://dx.doi.org/10.1016/j.nantod.2014.02.010
+Model
NANTOD-359; No. of Pages 26 ARTICLE IN PRESS
26 L. Zhang, E. Wang

[177] L. Shang, R.M. Doerlich, V. Trouillet, M. Bruns, G.U. Nienhaus,


Erkang Wang, Professor of Chemistry at
Nano Res. 5 (2012) 531—542.
Changchun Institute of Applied Chemistry,
Chinese Academy of Sciences (CAS), and an
Libing Zhang was born in Shandong Province, advisor in the State Key Laboratory of Elec-
China. He received his B.S. degree from troanalytical Chemistry. He received his Ph.D.
Liaocheng University in 2007 and M.S. degree degree in 1959 from Czechoslovak Academy
from Changchun University of Science and of Sciences directly under Professor J. Hey-
Technology in 2010. Then, he moved to rovsky, the Nobel Prize Laureate. He is
Changchun Institute of Applied Chemistry as a academician of both the CAS and the Academy
Ph.D. student under the direction of Professor of Sciences for the Developing World. He has
Erkang Wang, and received his Ph.D. degree in been on the Editorial and Advisory Board of
2013. He is currently a Postdoctoral Research nine international journals. His research interests lie in the fields of
Associate in Biodesign Institute at Arizona nanomaterials/nanotechnology, biosensors, electrochemistry and
State University. His scientific interests focus electrochemiluminescence. He has published over 900 papers and
on functional nucleic acids and nanomaterials for analytical and monographs in international journals with the SCI over 17,000 times
biological applications. cited.

Please cite this article in press as: L. Zhang, E. Wang, Nano Today (2014), http://dx.doi.org/10.1016/j.nantod.2014.02.010

Вам также может понравиться