You are on page 1of 16

Biochimica et Biophysica Acta 1760 (2006) 636 – 651

http://www.elsevier.com/locate/bba

Review
Glyconanoparticles: Types, synthesis and applications in glycoscience,
biomedicine and material science
Jesús M. de la Fuente ⁎, Soledad Penadés ⁎
Grupo de Carbohidratos, Instituto de Investigaciones Químicas, CSIC, Isla de la Cartuja, Américo Vespucio 49, 41092 Sevilla, Spain
Received 27 October 2005; received in revised form 30 November 2005; accepted 1 December 2005
Available online 28 December 2005

Abstract

Nanoparticles are the subject of numerous papers and reports and are full of promises for electronic, optical, magnetic and biomedical
applications. Although metallic nanoparticles have been functionalized with peptides, proteins and DNA during the last 20 years, carbohydrates
have not been used with this purpose until 2001. Since the first synthesis of gold nanoparticles functionalized with carbohydrates
(glyconanoparticles) was reported, the number of published articles has considerably increased. This article reviews progress in the development
of nanoparticles functionalized with biological relevant oligosaccharides. The glyconanoparticles constitute a good bio-mimetic model of
carbohydrate presentation at the cell surface, and maybe, excellent tools for Glycobiology, Biomedicine and Material Science investigations.
© 2005 Elsevier B.V. All rights reserved.

Keywords: Glyconanoparticle; Quantum dot; Magnetic nanoparticle; Carbohydrate interaction; Multivalence; Glycobiology

1. Introduction nanoparticles has revolutionized the visualization of cellular


and/or tissue components by electron microscopy. During the
During the last 20 years, there has been a great deal of last 10 years, several groups have prepared gold nanoparticles
interest in the self-assembly fabrication of hybrid materials from functionalized with proteins and DNA [1]. These nanoparticles
inorganic nanoparticles and biomolecules [1]. As nanoparticles are being used for assembling new materials, developing
and biomolecules are of a similar length scale (Fig. 1), it seems bioassays and as multivalent systems for interaction studies.
logical that the combination of biomacromolecules to nano- Although several reviews on nanomaterials functionalized
materials can provide interesting tools for mimicking the with proteins, peptides, DNA and RNA have been published in
biomolecules which are present at cellular systems, probing the the last decade [1,2,6–11], as far as we know, only one of them has
mechanisms of biological processes, as well as developing been partially reported on nanoparticles covered with carbohy-
chemical means by handling and manipulating biological drates (glyconanoparticles) [12]. For this reason, the main aim of
components [2]. At present, it is straightforward to control this manuscript is the review of the synthetic methods and
and modify the properties of nanostructures to better suit their applications of these new hybrid materials made of inorganic
integration with biological systems; for example, controlling nanoparticles and biologically relevant oligosaccharides.
their size, modifying their surface layer for enhanced aqueous Carbohydrates are, together with nucleic acids and proteins,
solubility, biocompatibility, or biorecognition [2–4]. The use of important molecules for life. Much is already known about the
gold nanoparticles in biological applications was first high- structure, interactions and function of nucleic acids and proteins,
lighted in the 1970s with the immunogold staining procedures however, the role of carbohydrates in the cell is less clear. The
[5]. Since then, the labeling of target molecules with gold surface of mammalian cells is covered by a dense coating of
carbohydrates named glycocalyx [13]. In the glycocalyx,
⁎ Corresponding authors. Tel.: +34 954489561; fax: +34 95 4460565. carbohydrates appear mainly conjugated to proteins and lipids
E-mail addresses: lafuente@iiq.csic.es (J.M. de la Fuente), (glycoproteins, glycolipids and proteoglycans) and it is as
penades@iiq.csic.es (S. Penadés). glycoconjugates that they develop their biological function.
0304-4165/$ - see front matter © 2005 Elsevier B.V. All rights reserved.
doi:10.1016/j.bbagen.2005.12.001
J.M. de la Fuente, S. Penadés / Biochimica et Biophysica Acta 1760 (2006) 636–651 637

Fig. 1. The figure depicts the sizes of nanoparticles in relation to other biological objects.

Now, it is known that these complex oligosaccharides are involved Although numerous examples of gold and magnetic
in the control of many normal and pathological processes [14,15]. nanoparticles protected and stabilized with polysaccharides as
A characteristic feature of the biological interactions where chitosan or dextran have appeared in the literature [17–22], this
carbohydrates are involved is their extreme low affinity that has to review is focused on nanoparticles functionalized with
be compensated by multivalent presentation of the ligands. For this biologically relevant oligosaccharides, where the oligosacchar-
reason, we developed a new integrated approach based on the use ides confer biological functionality to the inorganic core of the
of nanoparticles that we have named the Glyconanotechnology nanomaterial rather than playing a simple stabilizing role.
strategy [12] to study and evaluate carbohydrate–carbohydrate and
carbohydrate–protein interactions. After our first manuscript on 2. Types and synthesis of glyconanoparticles
the application of gold glyconanoparticles to study the self-
aggregation of Lex trisaccharide in the presence of calcium Three different types of nanoparticles functionalized with
appeared in 2001 [16], several research groups have developed oligosaccharides have been reported so far: gold and silver
different strategies to prepare and apply nanoparticles functiona- glyconanoparticles, semiconductor glyco-quantum dots and
lized with biologically relevant oligosaccharides to study magnetic glyconanoparticles.
carbohydrate interactions or to intervene in carbohydrate-mediated
biological processes. After 5 years, there are enough results to state 2.1. Gold and silver glyconanoparticles
that carbohydrates are joining proteins, peptides and DNA as
biological partners of inorganic nanomaterials to intervene in Gold nanoparticles are the most stable metal nanoparticles,
biological processes (Fig. 2). and present interesting properties which include a wide array
of assembling model and unexpected size-related electronic,
magnetic and optical properties (quantum size effect). They
already have a number of applications in catalysis and
biology. Therefore, their promises are in these fields as well
as in the bottom-up approach of nanotechnology, and they
will be key materials and building blocks in the 21st century.
While numerous routes exist for the production of colloidal
gold nanoparticles, the method described by Brust et al. [23]
and its variation [24,25] are the most popular synthetic
schemes in the field. As far as we know, most of the
monolayer protected glyconanoparticles have been prepared
by Brust's method. This two-phase synthesis uses a thiol
ligand that strongly binds gold due to the soft character of
both Au and S (which protect the metallic core by a covalent
Au\S bond). A gold salt (AuCl4−) is transferred from a water
solution to toluene using tetraoctylammonium bromide as the
phase-transfer reagent and then reduced by NaBH4 in the
Fig. 2. Carbohydrates join DNA, proteins and peptides as biological partner of presence of dodecanethiol (Scheme 1) [23]. The organic
inorganic nanoparticles. phase changes colour from orange to deep brown within a
638 J.M. de la Fuente, S. Penadés / Biochimica et Biophysica Acta 1760 (2006) 636–651

Penades' method also provides the possibility to prepare in a


single step, hybrid nanoparticles or nanoparticles with different
density of carbohydrates (Fig. 4). Fluorescence-labeled glyco-
nanoparticles have been prepared by mixing in a determined
ratio a thiol functionalized fluorescein and the neoglycoconju-
gate prior to nanoparticle formation (Scheme 3). Similarly, the
control of the carbohydrate density on the surface of the
nanoparticles, from 5% to 30% in lactose, can be achieved by
Scheme 1. Synthesis of dodecanethiol gold nanoparticles by Brust's
methodology. (a) HAuCl4, NaBH4, toluene/water, tetraoctylammonium mixing different linkers and neoglycoconjugates before nano-
bromide. particle formation [3].
Lactose functionalized glyconanoparticles have been also
few seconds upon addition of NaBH4. This method allows the produced in a three-step procedure by Kataoka et al. [36] First,
facile synthesis of thermally and air-stable gold nanoparticles gold nanoparticles protected with a poly(ethylene glycol) (PEG)
of reduced dispersion and controlled size. Indeed, these derivative containing both mercapto and acetal groups was
nanoparticles can be repeatedly isolated and re-dissolved in prepared by Brust's method. The α-acetal-PEG layer formed on
water without irreversible aggregation or decomposition. The gold nanoparticles was converted to aldehyde by acid treatment
method can also be carried out in methanol–water solution by and, in a third step, a reductive amination with p-aminophenyl
using water soluble thiolates [26] and following this glycosides in the presence of (CH3)2NHBH3 was carried out
procedure Murray et al. reported the synthesis of water (Scheme 4). To regulate lactose density on the nanoparticles, this
soluble gold nanoparticles with the non-natural amino acid group has recently [37] used the approach previously reported by
tiopronin as a stabilizer [27]. us [3] for the preparation of hybrid glyconanoparticles.
These nanoparticles protected with alkanethiolates have A three-step procedure has also been used by Lakowicz et al.
received considerable attention due to their stability, suspensi- [38] to prepare hybrid silver glyconanoparticles. They prepared
bility in different solvents, and facile characterization by tiopronin protected nanoparticles which afterwards underwent a
standard analytical techniques. Since Brust reported his ligand exchange with thiolate boronic acids. The nanoclusters
methodology in 1994, several groups have prepared gold capped with boronic acids were coupled to polysaccharides
nanoparticles functionalized with peptides, proteins and DNA (dextran 3000) and monosaccharides (glucose). However, in
[1,2]. this case, the carbohydrates are not covalently attached to the
Based on this method, we reported the first synthesis of gold nanoparticle.
glyconanoparticles by adding a methanolic solution of a Another example of non-covalently sugar protected glyco-
neoglycoconjugate functionalized with a thiol group, to an nanoparticles was reported in 2004 by Yang et al. [39] for the
aqueous solution of tetrachloroauric acid (HAuCl4) and synthesis of gold and silver nanoparticles of chitosan and
subsequent reduction with NaBH4 (Fig. 3) [16]. Following heparin. This approach uses these polysaccharides as reducing/
this method, the synthesis of different glyconanoparticles
functionalized with the tetrasaccharide Le y ([Fucα1–2]
Galβ1–4[Fucα1–2]GlcNAcβ1) [28], the trisaccharide Lex
(Galβ1–4[Fucα1–2]GlcNAcβ1) [16], the disaccharides lactose
(Galβ1–4Glcβ1) and maltose (Glcα1–4Glcβ1) or the mono-
saccharide glucose were reported (Scheme 2) [3]. Different
linkers of hydrophobic (alkanes) and hydrophilic (polyethylene
glycol derivatives) nature were used to bind the carbohydrate to
the gold core [3]. The glyconanoparticles prepared in this way
are water soluble, stable in solution for years, non-cytotoxic and
exceptionally small (less 2 nm). This small size confers them
unusual properties as a permanent magnetism even at room
temperature [29,30].
The same synthetic method has been used by different
groups. Lin et al. [31] have prepared gold nanoparticles
functionalized with glucose, mannose, galactose and manno-
sides; Gervay-Hague et al. [32] with glucose and galactose;
Barchi et al. [33] with Thomsen-Friedenreich disaccharide; and
Kamerling et al. with oligosaccharides involved in the self
recognition process of the marine sponge Microciona prolifera
[34]. The last authors have also prepared a series of gluco- and
manno-oligosaccharide containing glyconanoparticles from free Fig. 3. A cartoon showing the mechanism of nanoparticle formation. The
oligosaccharides by a reductive amination of the saccharides neoglycoconjugates protect the clusters, control their size, and provide
with trityl-protected cysteamine and further detritylation [35]. bioactivity.
J.M. de la Fuente, S. Penadés / Biochimica et Biophysica Acta 1760 (2006) 636–651 639

Scheme 2. Synthesis of Lex, Ley, glucose, lactose and maltose gold glyconanoparticles by Penades' methodology.

stabilizing agents. In both cases the metal nanoparticles were 2.2. Glyco-quantum dots
stabilized by electrostatic interactions between the surface of the
nanoparticle and the polysaccharides. This so-called green Nanocrystals of semiconducting materials, otherwise includ-
method provides a simple and environmental friendly strategy ed in the term quantum dots (QDs), have fascinated physicists,
to prepare glyconanoparticles without any additional chemical chemists, and electronic engineers since the 1970s. The most
reducing agent and in aqueous solution. striking feature of these materials is that their chemical and
All the above-mentioned methods provide water-soluble physical properties differ markedly from those of the bulk solid
nanoparticles of reduced dispersion and controlled size. These [40]. Since their quantum size effects are understood,
nanoparticles are stable, and self-aggregation has not been fundamental and applied research on these systems has become
observed in any case. increasingly popular. One of the most interesting applications is
640 J.M. de la Fuente, S. Penadés / Biochimica et Biophysica Acta 1760 (2006) 636–651

the use of nanocrystals as luminescent labels for biological


systems [41–45]. QDs have several advantages over conven-
tional fluorescent dyes: they emit light at a variety of precise
wavelengths depending on their size and have long fluorescent
lifetimes. There are several approaches for the coupling of
biomolecules to semiconductor QDs [46]. Most commonly,
QDs are first prepared at high temperature in the presence of
protective additives to prevent crystal aggregation and to
regulate the growing rate. Alternatively, the protection process
can be performed in solution using thiols with hydrophilic end-
groups to confer stability and water solubility to the QDs. In a
second step, the biomolecules are attached to the protected
Fig. 4. The cartoon shows some examples of hybrid glyconanoparticles where nanocrystals for cell labeling. QDs bio-conjugated to peptides
DNA, RNA, lipids, peptides, or biotin coexist with antigenic oligosaccharides. and proteins [42,43,45,47–52], antibodies [43,50–54], DNA

Scheme 3. Preparation of hybrid glyconanoparticles.


J.M. de la Fuente, S. Penadés / Biochimica et Biophysica Acta 1760 (2006) 636–651 641

Scheme 4. Preparation of functionalized gold glyconanoparticles by Kataoka's methodology. (a) HAuCl4, NaBH4; (b) H+; (c) NH2-Ph-Lactose, NH2-Ph-Mannose,
(CH3)2NHBH3.

[55–58], and other molecules [59–61] have recently been maltose protected CdS and Zns nanocrystals were carried out.
prepared mainly by coupling the biomolecules to the thiol The procedure was also used for the synthesis of tiopronin and
protected QDs and tested as biological markers. However, there tiopronin-Tat functionalized QDs which where able to label the
are only few examples of QDs conjugated to carbohydrate nucleus of human fibroblasts [45].
antigens for specific cell targeting. Fang et al. have recently reported the functionalization with
The first example of QDs protected with polysaccharides was β-N-acetylglucosamine of CdSe/ZnS core-shell QDs previously
reported in 2003 by Rosenzweig et al. [62]. This group prepared encapsulated with pyridine. The pyridine-encapsulated QDs
CdSe-ZnS quantum dots protected with carboxymethyldextran were treated with a disulphide derivative of N-acetylglucosa-
and polylysine, and they proved the high affinity of the QDs mine and NaBH4 in aqueous solution to give water-soluble
toward the glucose binding protein-Concanavalin A (Con A). glyco-QDs [65].
Chaikof et al. reported in 2004 the coupling of commercially
available QDs-streptavidin with a biotin end-terminated lactose 2.3. Magnetic glyconanoparticles
glycopolymer [63]. Confocal microscopy confirmed fluorescent
staining of Ricinus communis agglutinin (RCA120)-immobilized Magnetic nanoparticles offer exciting new opportunities
agarose beads due to the glycopolymer–lectin interaction. including the improvement of the quality of magnetic resonance
We have reported the preparation of water soluble nanoclus- imaging (MRI), hyperthermic treatment for malignant cells,
ters of cadmium sulphide and zinc sulphide covalently bound to site-specific drug delivery and also the recent research interest
biologically significant neoglycoconjugates by a single step of manipulating cell membranes (Fig. 5) [11,21,22,66,67].
solution procedure (Scheme 5) [64]. The synthesis of Lex and Previously reported iron oxide superparamagnetic nanoparticles
prepared by different surface chemistry have been widely used
for numerous in vivo applications. The biological applications
of these nanomaterials require these nanoparticles to have high
magnetization values, size smaller than 20 nm, narrow particle

Scheme 5. Preparation of glyco-QDs, (a) Cd(NO3)2·4H2O, Na2S, pH 10; (b) Zn


(NO3)2 6H2O, Na2S, pH 10. Fig. 5. Potential applications of magnetic glyconanoparticles.
642 J.M. de la Fuente, S. Penadés / Biochimica et Biophysica Acta 1760 (2006) 636–651

3. Application of glyconanoparticles

The glyconanoparticles constitute a good bio-mimetic model


of carbohydrate presentation at the cell surface, opening new
avenues in the field of chemical Glycobiology [12] and
Biomedicine (Fig. 6). In spite of the short history of these
new glyconanomaterials, some applications have already been
reported, focused mainly on the study and evaluation of
carbohydrate interactions. Furthermore, few reports have
appeared on glyconanoparticles as biolabels or biosensors and
some applications in biomedicine or material science have also
been described.
Fig. 6. Applications of glyconanoparticles.
3.1. Glyconanoparticles for the evaluation of carbohydrate
interactions
size distribution and a special surface coating for both avoiding
toxicity and allowing the coupling of biomolecules [21]. Most Three general aspects of glyconanoparticles make them
commonly, iron oxide nanoparticles are prepared by co- suitable model systems for the study and evaluation of
precipitation of ferrous and ferric salts solution and stabilized interactions where sugars are involved. Firstly, glyconano-
using biocompatible molecules as dextran or oleic acid. In a particles are similar in size range to many common biomole-
second step, the biomolecules are attached by covalent or cules, and for instance they can imitate carbohydrate
electrostatic coupling to the protected nanoparticle. Bio- presentation in glycoproteins or in glycosphingolipids patches.
conjugated magnetic nanoparticles to peptides, proteins and Secondly, glyconanoparticles provide a glycocalyx-like surface,
antibodies have been developed and tested as biological presenting carbohydrates in a globular and polyvalent config-
markers or in hyperthermia treatments. However, there was uration on their surface. Thirdly, glyconanoparticles have
not one example of magnetic glyconanoparticles. Following the unusual physical properties due to the quantum size effect
same procedure as for the gold glyconanoparticles, we have [6,29] that can be used for the detection and evaluation of the
prepared gold–iron nanoparticles functionalized with glucose, interactions.
maltose and lactose by adding an iron salt to the aqueous AuCl4− So far, glyconanoparticles have been used to study both
solution [68]. The so produced nanoparticles are water soluble, carbohydrate–carbohydrate (carb–carb) interactions and carbo-
stable and exceptionally small. The properties of these hydrate–protein interactions.
nanoparticles are particularly interesting as a result of their
quantum size. They show a complex magnetic behaviour with 3.1.1. Glyconanoparticles in carbohydrate–carbohydrate
both paramagnetic and ferromagnetic components. This interactions
anomalous behaviour may be attributed to the gold atoms that In 1989, Hakomori proposed that the first phase of cell
predominate in the metallic cluster with an atomic Au/Fe ratio adhesion initiates through a carb–carb interaction between
from 50:1 to 5:1. Previously, we demonstrated that gold multivalent glycosphingolipid-domains. This initial step is
glyconanoparticles present a permanent magnetism even at followed by protein–protein interactions between adhesion
room temperature [29,30]. Work directed to achieve a better receptors and protein–carbohydrate interactions between
understanding of this phenomenon is currently underway. lectins and glycoconjugates. Embryogenesis, metastasis and
The effect of these nanoparticles on human dermal other cellular proliferation processes are according to
fibroblasts in vitro has been assessed in terms of cytotoxicity, Hakomori, mediated by carb–carb interactions [70–75].
light microscopy and scanning electron microscopy [69]. Several glycosphingolipids involved in carb–carb interactions

Scheme 6. Structure of GM3, Gg3, lactose and KDNGM3 glycosphingolipids.


J.M. de la Fuente, S. Penadés / Biochimica et Biophysica Acta 1760 (2006) 636–651 643

have already been identified. For example, the antigen carbohydrate presentations at the cell surface. Gold glycona-
determinant Lex seems to mediate morula compaction in noparticles functionalized with the trisaccharide Lex (Lex–Au)
mouse via a carbohydrate self-interaction [70], and the have been used to investigate the selective self-recognition of
interaction between the glycosphingolipids GM3 and Gg3 the Lex antigen via carb–carb interaction (Fig. 7) [12].
has been proposed to be involved in metastasis of melanoma Transmission electron microscopy (TEM) has allowed to
cells in mouse [72–74]. It has been demonstrated recently that observe directly the self-aggregation of the Lex nanoparticles
the interaction between KDNGM3 and Gg3 is involved in in 10 mM Ca2+ solution [16]. The TEM micrographs obtained
the binding of sperm to egg membrane in rainbow trout revealed clear differences between lactose and Lex nanoparti-
fertilization (Scheme 6) [75]. cles. The Lex nanoparticles in Ca2+ solution showed large three-
Outside from the eukaryotic world, another example where dimensional aggregates at all concentrations, while the lacto
carb–carb interactions are involved is the species-specific cell particles remained dispersed as in the water solution. Removal
aggregation of marine sponges [76–78]. It seems that a large of the Ca2+ from the solution by addition of EDTA resulted in
extra cellular acidic proteoglycan (g200) is responsible, via a the dispersion of the Lex aggregates. Isothermal titration
Ca2+-dependent, homophilic and polyvalent carb–carb interac- calorimetry (ITC) has confirmed that this process in solution
tion, for this aggregation. A sulfated disaccharide and a is a slow process that takes place with a decrease in enthalpy of
pyruvated trisaccharide have been identified as the repetitive 160± 30 kcal mol−1, while the heat evolved in the case of
epitopes of the glycan responsible for aggregation [79,80]. lactose and maltose glyconanoparticles was very low and
Characteristic features of these interactions are their thermal equilibrium was quickly achieved [86].
specificity, their strong dependency on divalent cations, and A similar approach has been followed by Kamerling et al. to
their extreme low affinity that has to be compensated by explore the carbohydrate-mediated self-recognition of marine
multivalent presentation of the ligands. The study of carb–carb sponge cells. This group has used water-soluble gold glyco-
interactions faces the important challenge of this extreme low nanoparticles coated with a synthetic sulfated disaccharide
affinity and attempts to characterize and quantify this fragment as multivalent systems to investigate the g-200
interaction in solution with monomer ligands have usually glycan–glycan interaction by TEM [87].
failed [81–83]. Multivalent presentation also has been proven to The first kinetics data of Lex–Lex interaction have been
be important in the study of carbohydrate–protein interactions obtained by us using a combination of self-assembled
[84,85]. However, polyvalence seems even more mandatory in monolayer (SAMs) of a Lex neoglycoconjugate on a biosensor
the case of carbohydrate–carbohydrate interactions. gold surface as substrate, Lex gold glyconanoparticles as the
It was searching for appropriate multivalent systems with analyte and surface plasmon resonance (SPR) detection [88].
well-defined chemical composition, that we have developed The sensorgrams obtained for the binding of Lex glyconano-
first gold glyconanoparticles as a tool to imitate multivalent particles to the Lex-SAMs indicated a slow association phase

Fig. 7. Glyconanoparticles for studying carbohydrate–carbohydrate interactions.


644 J.M. de la Fuente, S. Penadés / Biochimica et Biophysica Acta 1760 (2006) 636–651

(kon 2.3 ×103 M−1 s−1) and a gradual dissociation phase (koff
1.5 ×10−3 s−1) in the presence of calcium cations. The binding
was of high affinity with dissociation constant in the
micromolar range (Kd, 5.4 ×10−7 M).

3.1.2. Glyconanoparticles in carbohydrate–protein


interactions
Characteristic features of protein–carbohydrate interactions Fig. 9. Schematic illustration of the interactions of glyconanoparticles and ConA
are high specificity and low affinity. Nature overcomes this low on the biosensor chip in the competition assay. Reprinted with permission from
affinity by clustering of ligands and receptors at cell surfaces ref 31 (Lin's group). Copyright 2003 The Royal Society of Chemistry.
[85], the concept of multivalence becoming an important issue in
glycobiology. For understanding the mechanism of these density of lactose [37]. This lectin specifically recognizes the β-
interactions, many researchers have developed a variety of D-galactose residue, inducing changes in the absorption
model systems bearing spherical or linear arrays of carbohy- spectrum. The aggregation of the lactose glyconanoparticles
drates [84,89,90], including glycodendrimers [91–93], glyco- originated a visible colour change from pinkish-red to purple.
polymers [94–97], glycopeptides and glycoproteins [98], and This aggregation was reversible and the degree of aggregation
self-assembled systems, e.g., liposomes [74,99], micelles was proportional to lectin concentration. More recently, Chen et
[100,101] and rotaxane [102]. Some of these multivalent al. have applied the visible colour change for the detection of
systems have been shown to be superior ligands for the protein–protein interactions [112]. Thyroglobulin, a binding
binding to endogenous lectins [103,104] and bacterial toxins protein for ConA, was added over the mannose-encapsulated
such as Shiga-like toxin [105] or cholera toxin [106], drawing glyconanoparticles and ConA complex. The change from
attention to their potential clinical applications. The presenta- purple to pinkish-red, due to the dispersion of glyconano-
tion and density of the ligand in these recognition processes particles, allowed the qualitatively and quantitatively evalu-
have been demonstrated to critically influence the binding ation of the thyroglobulin–ConA interaction.
[107–111]. Such an influence is intimately related to the The merge between gold glyconanoparticles and SPR
particular arrangement and architecture of carbohydrate- technique have demonstrated to be a powerful tool to evaluate
binding sites in the receptor. carbohydrate interactions. Since we reported this combination to
In the search for new multivalent systems with well-defined obtain kinetic data for Lex–Lex [88], several groups have used
chemical composition, several groups have used gold glycona- this approach to quantify carbohydrate–protein interactions. For
noparticles. The first group using gold glyconanoparticles to example, Lin et al. have used SPR technique to quantitatively
study protein–carbohydrate interactions was Kataoka et al. They analyze the interaction between mannose gold glyconanoparti-
observed the selective aggregation of lactose gold nanoparticles cles and the Concanavalin A lectin (Con A) [31]. They have
exposed to RCA120 (Fig. 8) [36] and the ligand density effects on found that the binding has a strong multivalent effect and that the
the aggregation through the evaluations of the interaction affinity of mannose glyconanoparticles for Con A could be
between the lectin and gold glyconanoparticles with regulated adjusted by altering the nanoparticle size or the sugar moiety

Fig. 8. Schematic representation of the reversible aggregation–dispersion behaviour of lactose gold nanoparticles by sequential addition of RCA120 lectin and
galactose with actual concomitant change in colour from pinkish-red to purple to pinkish-red. Reprinted with permission from ref 36 (Kataoka's group). Copyright
2001 American Chemical Society.
J.M. de la Fuente, S. Penadés / Biochimica et Biophysica Acta 1760 (2006) 636–651 645

(Fig. 9). Recently, Kamerling et al. have evaluated the


interaction between different mannosides and glucosides
prepared from free oligosaccharides and the Con A lectin [35],
demonstrating the multivalent binding character of this
interaction.
Gervay-Hague et al. have reported investigations of
multivalent interactions of galactose and glucose gold
glyconanoparticles with recombinant gp120 [32]. A biotin
NeutrAvidin adhesion assay was used to evaluate the relative
ability of gold glyconanoparticles to displace rgp120 from
plate-bound GalCer. This group found that the gold glycona-
noparticles were more than 300 times more active than the
corresponding dimer.
QDs have been used to evaluate carbohydrate–protein
interactions as well. Fang et al. have recently used the optical
properties of these fluorescent materials to demonstrate the
affinity of β-N-acetylglucosamine-encapsulated QDs with the
wheat germ agglutinin (WGA) by fluorimetry, transmission
electron microscopy, dynamic light scattering microscopy and
flow cytometry [65].

3.2. Glyconanoparticles as biolabels

Gold nanoparticles that are functionalized with proteins


have long been used as tools in biosciences [1,2]. For
example, gold nanoparticles were initially applied in biolog-
ical applications in 1971, when Faulk and Taylor devised the
immunogold staining procedure. The labeling of targeting Fig. 10. Typical TEM images of sectioned areas of (A) pili of the E. coli
molecules with nanoparticles has revolutionized the visuali- ORN178 strain bound with m-AuNP, (B) the E. coli ORN208 strain deficient of
zation of cellular or tissue components by electron microscopy the fimH gene without m-AuNP binding. Scale bar = 100 nm. Reprinted with
permission from ref 120 (Lin's group). Copyright 2002 American Chemical
[5]. In fact, metal, semiconductor, polymers and magnetic
Society.
particles functionalized with proteins, peptides, antibodies or
nucleic acids, have been extensively used to target cells concentrated at the sperm heads, while mannose-encapsulat-
[41,113–119]. However, there are only few examples of ed QDs tended to spread over the whole sperm body, due to
glyconanoparticles used as labels. the different distribution of the GlcNAc and Man receptors
Mannose gold nanoparticles have been used to observe the on the sperm surface.
specific binding to FimH adhesin of bacterial type 1 pili by
TEM [120]. Lin et al. confirmed the specific binding of 3.3. Glyconanoparticles for bio-amplification strategies
mannose nanoparticles to FimH, incubating the nanoparticles
with ORN178 (strain expresses wild-type type I pili) and with The emergence of nanotechnology is broadening new
ORN208 (strain is deficient on the fimH gene) and the horizons for highly sensitive bioaffinity assays. Metal or
binding of mannose nanoparticles to bacterial pili in each semiconductor nanoparticles functionalized with nucleic acids
condition was examined by TEM (Fig. 10). The selective and antibodies have been previously used as amplifier for the
binding of these glyconanoparticles to bacterial type I pili detection of DNA and proteins [10]. This use of metal
presented a novel method of labeling specific receptors for glyconanoparticles is one of the most promising applications
carbohydrates on the cell surface using carbohydrate conju- for this type of materials. As far as we know, there are only
gated nanoparticles. two examples of this application.
Due to the optical properties of semiconductor nanocrys- Geddes et al. have developed a method to quantify glucose
tals, glyco-QDs are potential candidates to be used as cell monitoring the shift and broadening of the plasmon absorption
labels. The used of QDs for cell labeling has been growing of gold glyconanoparticles functionalized with a high-molec-
in the last decade, and it is expected that in the following ular-weight dextran [121,122]. The method is based on the
years glyco-QDs will be applied in cell biology as a routine aggregation of the gold nanoparticles with Con A, which results
tool to analyze carbohydrate receptors. In fact, Fang et al. in a significant shift and broadening of the gold plasmon
have recently reported the labeling of mice, pigs and sea- absorption. The addition of glucose competitively binds to Con
urchin live sperm with CdSe/ZnS core shells QDs A, reducing gold nanoparticles aggregation and therefore
functionalized with β-N-acetylglucosamine or mannose decreasing the plasmon absorption when monitored at a near-
(Fig. 11) [65]. Acetylglucosamine-encapsulated QDs were red arbitrary wavelength.
646 J.M. de la Fuente, S. Penadés / Biochimica et Biophysica Acta 1760 (2006) 636–651

Fig. 11. Confocal microscope imaging for staining of sperm with glyco-QDs: (A) selective β-N-acetylglucosamine-encapsulated quantum dots labeling on the heads of
sea-urchin sperm (Scale bar = 20 μm), (B) close-up of β-N-acetylglucosamine-encapsulated quantum dots-labeled sea-urchin sperm, and (C) close-up of mannose-
encapsulated quantum dots-labeled mouse sperm. Reprinted with permission from ref 65 (Fang's group). Copyright 2005 Wiley-VCH.

The second example has been showed by Lin et al. [123]. cell selectins promote tumour cell metastasis. In addition to
This group has used glucose and Pk (Galα1 →4Galβ1 → 4Glc) this mechanism, carb–carb interactions between glycosphin-
gold glyconanoparticles for the separation and enrichment of golipids expressed on the tumour and endothelial cell surfaces
target proteins by incubation with a mixture of proteins (Fig. also seem to be involved in the critical adhesion step. A
12). Using this approach a lectin (PA-IL) was isolated from a carb–carb interaction between GM3 expressed in a murine
mixture of proteins and detected at femtomole concentrations melanoma cell line (B16) and Gg3 or lactosylceramide
using glucose nanoparticles and MALDI-TOF MS analysis. (Scheme 6) of endothelium cells has been proposed to be
The methodology allows the identification of unknown target involved in the first adhesion step of tumour cells to
proteins by in situ digestion with chymotrypsin and the analysis endothelium before transmigration [72].
peptide mass fingerprinting map of chymotryptic peptides. Therefore, inhibition of this step by glyconanoparticles that
present carbohydrate antigens expressed either in the tumour
3.4. Glyconanoparticles in biomedicine or the endothelium cells might provide effective anti-adhesion
therapy (Fig. 13). Based on the involvement in cell adhesion
Glyconanoparticles constitute a good biomimetic model to of the antigen lactosylceramide, lactose glyconanoparticles
intervene in carbohydrate-mediated biological processes. For were tested as a potential inhibitor of the binding of
this reason, glyconanoparticles have been produced and applied melanoma cells to endothelium. An ex vivo experiment was
in biomedical applications. designed for the evaluation of the anti-metastasis potential of
We have recently shown that lactose gold glyconanopar- the glyconanoparticles. Mice were injected with melanoma
ticles maybe potential tools in anti-adhesive therapy [124]. cells pre-incubated with lactose gold glyconanoparticles, and
One of the critical steps in metastasis is the adhesion of after 3 weeks, the animals were sacrificed and both lungs
tumour cells to the vascular endothelium. After adhesion, evaluated under the microscope for analysis of tumour foci. A
tumour cells transmigrate and create new tumour foci. 70% of tumour inhibition was reported as compared with the
Interactions between tumour-associated antigens and epithelial group inoculated only with melanoma cells [124].

Fig. 12. The analytical scheme of the Lin's technique for the specific capture of target proteins and the rapid mapping of binding-epitope-containing peptides.
Reprinted with permission from ref 123 (Lin's group). Copyright 2005 Wiley-VCH.
J.M. de la Fuente, S. Penadés / Biochimica et Biophysica Acta 1760 (2006) 636–651 647

Fig. 13. Possible action mechanism of lactose glyconanoparticles in anti-adhesive therapy. Reprinted with permission from ref 12 (Penades's group). Copyright 2004
Kluwer.

3.5. Glyconanoparticles in material science The inclusion of a fluorescein molecule, attached to the gold
nanoparticle by a thiol chain, gives a hydrophobic character
The design, synthesis and characterization of surface- to the gold glyconanoparticles and also introduces the
modified nanoparticles are of fundamental importance in possibility of aromatic stacking interactions. The so-prepared
controlling the mesoscopic properties of new materials and in glyconanoparticles formed a superstructure of holes sur-
developing new tools for nanofabrication [125–128]. Self- rounded by nanoparticles as observed by TEM. In the regions
assembled monolayers of thiolates on colloidal gold clusters covered by nanoparticles a two-dimensional hexagonal
are therefore ideal substrates for investigating nanoparticle structure defined by gold dots is observed (Fig. 14) [132].
microfabrication techniques and the effect of surface-bound A centre to centre distance of 6 nm was found, which
reagents on structure–properties relationships [129–131]. corresponds to the length of two consecutive neoglycoconju-
We have prepared hybrid gold glyconanoparticles with gate molecules. This result suggested that the manipulation of
fluorescein molecules, with the aim of having a fluorescence the molecule attached to the gold glyconanoparticles can
probe for labeling the nanoparticles for biological tests [3]. control the organization of the gold clusters opening new

Fig. 14. TEM images of (a) lactose-fluorescein gold nanoparticles and (b) Lex-fluorescein gold nanoparticles, and the proposed model to explain the hexagonal
package. Inset: High-magnification images of the indicated areas.
648 J.M. de la Fuente, S. Penadés / Biochimica et Biophysica Acta 1760 (2006) 636–651

Fig. 15. The glyconanoparticle concept and its potential applications.

possibilities for size-selection devices at the nanoscale by a Furthermore, the control of glyconanoparticle composition
“bottom-up” strategy. using different carbohydrate and non-carbohydrate ligands
In general, metallic nanoparticles are extensively studied would allow to design and to prepare a great variety of hybrid
since they exhibit novel electronic, optical, and magnetic glyconanoparticles with increasing utility in drug delivery to
properties. Most of these new properties arise from the so-called generate “multi-action” drugs or as a new vaccine platform,
“size effect” which affects the electronic structure, as well as going from glyconanoparticles to “artificial nanocells”.
from the increase of the ratio of atoms located at the surface with
respect to the total number of atoms of the nanoparticle Acknowledgements
[133,134]. Glyconanoparticles are not an exception and they
show very interesting physical properties. In fact, we have We thank the colleagues in the Carbohydrate Group who
reported that 1.4–1.8 nm gold glyconanoparticles functionalized have decisively contributed to the glyconanoparticles develop-
with thiol derivatives exhibit a localized permanent magnetism ment. They are cited in the corresponding references. We thank
in contrast to the metallic diamagnetism characteristic of bulk also Prof. M. Martin-Lomas for his support and fruitful
Au or gold nanoparticles functionalized with amminated discussions. This work was supported by MEC (Grant
derivatives [29,30]. BQU2002-03734) and CSIC (I3P contract).

4. Conclusions and perspectives References

In this review, we have presented methods to prepare, in an [1] C.M. Niemeyer, Nanoparticles, proteins, and nucleic acids: biotechnol-
ogy meets materials science, Angew. Chem., Int. Ed. 40 (2001)
easy way, a variety of water-soluble and stable glyconanopar-
4128–4158.
ticles. All these nanomaterials are good biomimetic models of [2] E. Katz, I. Willner, Integrated nanoparticle-biomolecule hybrid systems:
carbohydrate presentation at the cell surface. synthesis, properties, and applications, Angew. Chem. Int. Ed. 33 (2004)
As mentioned above, the main application of these new 6042–6108.
multivalent systems is the study of carbohydrate-mediated [3] A.G. Barrientos, J.M de la Fuente, T.C. Rojas, A. Fernández, S. Penadés,
Gold glyconanoparticles: synthetic polyvalent ligands mimicking
interactions opening new avenues in Glycobiology. Though
glycocalyx-like surfaces as tools for glycobiological studies, Chem.-
the simple and flexible preparation of these glyconanoclusters Eur. J. 9 (2003) 1909–1921.
and their interesting physical, chemical and biological [4] J.M. Perez, L. Josephson, R. Weissleder, Use of magnetic nanoparticles
properties allow them to extend their utility in Biomedicine, as nanosensors to probe for molecular interactions, ChemBioChem 5
Biotechnology and Material Science, their use as biolabels or (2004) 261–264.
[5] J.M. Polak, I.M. Vamdell, Immunolabeling for Electron Microscopy,
biosensing has not been extensively developed. We expect that
Elsevier Science Publishers, Amsterdam, 1984.
a variety of applications of glyco-quantum dots, gold [6] C.B. Murray, C.R. Kagan, M.G. Bawendi, Synthesis and characterization
glyconanoparticles and magnetic glyconanoparticles will be of monodisperse nanocrystals and close-packed nanocrystals assemblies,
developed in the near future (Fig. 15). Annu. Rev. Mater. Sci. 30 (2000) 545–610.
J.M. de la Fuente, S. Penadés / Biochimica et Biophysica Acta 1760 (2006) 636–651 649

[7] M.-C. Daniel, D. Astruc, Gold nanoparticles: assembly, supramolecular [30] P. Crespo, R. Litrán, T.C. Rojas, M. Multigner, J.M. de la Fuente, J.C.
chemistry, quantum-sized-related properties, and applications towards Sanchez-Lopez, M.A. García, A. Hernando, S. Penadés, A. Fernández,
biology, catalysis and nanotechnology, Chem. Rev. 104 (2004) Permanent magnetism, magnetic anisotropy, and hysteresis of thiol-
293–346. capped gold nanoparticles, Phys. Rev. Lett. 93 (2004) 087204.
[8] U. Drechesler, B. Erdogan, V.M. Rotello, Nanoparticles: scaffolds for [31] C.-C. Lin, Y.-C. Yeh, C.-Y. Yang, G.-F. Chen, Y.-C. Chen, Y.-C. Wu,
molecular recognition, Chem.-Eur. J. 10 (2004) 5570–5579. C.-C. Chen, Quantitative analysis of multivalent interactions of
[9] A. Verma, V.M. Rotello, Surface recognition of biomacromolecules using carbohydrate-encapsulated gold nanoparticles with concanavalin A,
nanoparticle receptors, Chem. Comm. 3 (2005) 303–312. Chem. Commun. (2003) 2920–2921.
[10] J. Wang, Nanomaterial-based amplified transduction of biomolecular [32] B. Nolting, J.-J. Yu, G.-Y. Liu, S.-J. Cho, S. Kauzlarich, J. Gervay-Hague,
interactions, Small 1 (2005) 1036–1043. Synthesis of gold glyconanoparticles and biological evaluation of
[11] D.L. Huber, Synthesis, properties, and applications of iron nanoparticles, recombinant Gp120 interactions, Langmuir 19 (2003) 6465–6473.
Small 1 (2005) 482–501. [33] S.A. Svarovsky, Z. Szekely, J.J. Barchi, Synthesis of gold nanoparticles
[12] J.M. de la Fuente, S. Penadés, Understanding carbohydrate–carbohydrate bearing the Thomsen–Friedenreich disaccharide: a new multivalent
interactions by means of glyconanotechnology, Glycoconj. J. 21 (2004) presentation of an important tumor antigen, Tetrahedron: Asymmetry 16
149–163. (2005) 587–598.
[13] A. Frey, K.T. Giannasca, R. Weltzin, P.J. Giannasca, H. Reggio, W.I. [34] A.C. de Souza, K.M. Halkes, J.D. Meeldijk, A.J. Verkleij, J.F.G.
Lencer, M.R. Neutra, Role of glycocalyx in regulating access of Vliegenthart, J.P. Kamerling, Synthesis of gold glyconanoparticles:
microparticles to apical plasma membranes of intestine epithelial cells: possible probes for the exploration of carbohydrate-mediated self-
implications for microbial attachment and oral vaccine targeting, J. Exp. recognition of marine sponge cells, Eur. J. Org. Chem. (2004) 4323–4339.
Med. 184 (1996) 1045–1059. [35] K.M. Halkes, A.C. de Souza, C.E.P. Maljaars, G.J. Gerwig, J.P.
[14] A. Varki, Biological roles of oligosaccharides: all of the theories are Kamerling, A facile method for the preparation of gold glyconanoparticle
correct, Glycobiology 3 (1993) 97–130. from free oligosaccharides and their applicability in carbohydrate–
[15] R.A. Dwek, Glycobiology: toward understanding the function of sugars, protein interaction studies, Eur. J. Org. Chem. (2005) 3650–3659.
Chem. Rev. 96 (1996) 683–720. [36] H. Otsuka, Y. Akiyama, Y. Nagasaki, K. Kataoka, Quantitative and
[16] J.M. de la Fuente, A.G. Barrientos, T.C. Rojas, J. Rojo, J. Cañada, A. reversible lectin-induced association of gold nanoparticles modified with
Fernández, S. Penadés, Gold glyconanoparticles as water-soluble α-Lactosyl-ω-mercapto-poly(ethylene glycol), J. Am. Chem. Soc. 123
polyvalent models to study carbohydrate interactions, Angew. Chem. (2001) 8226–8230.
Int. Ed. 40 (2001) 2257–2261. [37] S. Takae, Y. Akiyama, H. Otsuka, T. Nakamura, Y. Nagasaki, K. Kataoka,
[17] E.A. Schellenberger, F. Reynolds, R. Weissleder, L. Josephson, Surface- Ligand desity effect on biorecognition by pegylated gold nanoparticles:
functionalized nanoparticle library yields probes for apoptotic cells, regulated interaction of RCA120 lectin with lactose installed to the distal
ChemBioChem 5 (2004) 275–279. end of tethered PEG strands on gold surface, Biomacromolecules 6
[18] C.C. Berry, S. Wells, S. Charles, G. Aitchison, A.S.G. Curtis, Cell (2005) 818–824.
response to dextran-derivatised iron oxide nanoparticles post internalisa- [38] J. Zhang, C.D. Geddes, J.R. Lakowicz, Complexation of polysaccharide
tion, Biomaterials 25 (2004) 5405–5413. and monosaccharide with thiolate boronic acid capped on silver
[19] L. Qi, Z. Xu, X. Jiang, Y. Li, M. Wang, Cytotoxic activities of chitosan nanoparticle, Anal. Biochem. 332 (2004) 253–260.
nanoparticles an copper-loaded nanoparticles, Bioorg. Med. Chem. Lett. [39] H. Huang, Q. Yuan, X. Yang, Preparation and characterization of metal-
15 (2005) 1397–1399. chitosan nanocomposites, Colloids Surf., B 39 (2004) 31–37.
[20] T.H. Kim, I.K. Park, J.W. Nah, Y.J. Choi, C.S. Cho, Galactosylated [40] H. Gleiter, Nanostructured materials, Adv. Mater. 4 (1992) 474–481.
chitosan/DNA nanoparticles prepared using water-soluble chitosan as a [41] M. Bruchez, M. Moronne, P. Gin, S. Weiss, A.P. Alivisatos,
gene carrier, Biomaterials 25 (2004) 3783–3792. Semiconductor nanocrystals as fluorescent biological labels, Science
[21] P. Tartaj, M.P. Morales, S. Veintemillas-Verdaguer, T. Gonzalez-Carreño, 281 (1998) 2013–2016.
C.J. Serna, The preparation of magnetic nanoparticles for applications in [42] W.C.W. Chan, S. Nie, Quantum dot bioconjugates for ultrasensitive
biomedicine, J. Phys., D, Appl. Phys. 36 (2003) R182–R197. nonisotopic detection, Science 281 (1998) 2016–2018.
[22] S. Mornet, S. Vasseur, F. Grasset, E. Duguet, Magnetic nanoparticles [43] H. Mattousi, J.M. Mauro, E.R. Goldmann, G.P. Anderson, V.C. Sundar,
design for medical diagnosis and therapy, J. Mater. Chem. 14 (2004) F.V. Mikulec, M.G. Bawendi, Self-assembly of CdSe-ZnS quantum dot
2161–2175. bioconjugates using an engineered recombinant protein, J. Am. Chem.
[23] M. Brust, M. Walker, D. Bethell, D.J. Schiffrin, R. Whyman, Synthesis of Soc. 122 (2000) 12142–12150.
thiol-derivatized gold nanoparticles in a 2-phase liquid–liquid system, [44] A.P. Alivisatos, Colloidal quantum dots. From scaling laws to biological
J. Chem. Soc. Chem. Commun. (1994) 801–802. applications, Pure Appl. Chem. 72 (2000) 3–9.
[24] C.P. Collier, R.J. Saykally, J.J. Shiang, S.E. Henrichs, J.R. Heath, [45] J.M. de la Fuente, M. Fandel, C.C. Berry, M. Riehle, L. Cronin, G.
Reversible tuning of silver quantum dots monolayers through the metal- Aitchison, A.S.G. Curtis, Quantum dots protected with tiopronin: a new
insulator transition, Science 277 (1997) 1978–1981. fluorescent system for cell-biology studies, ChemBioChem 6 (2005)
[25] D.E. Cliffel, F.P. Zamborini, S.M. Gross, R.W. Murray, Mercaptoammo- 989–991.
nium-monolayer-protected, water-soluble gold, silver, and palladium [46] A.J. Sutherland, Quantum dots as luminescent probes in biological
clusters, Langmuir 16 (2000) 9699–9702. systems, Curr. Opin. Solid State Mater. Sci. 6 (2002) 365–370.
[26] M. Brust, J. Fink, D. Bethell, D.J. Schiffrin, C.J. Kiely, Synthesis and [47] J.O. Winter, T.Y. Liu, B.A. Korgel, C.E. Schmidt, Recognition molecule
reactions of functionalised gold nanoparticles, J. Chem. Soc. Chem. directed interfacing between semiconductor quantum dots and nerve
Commun. (1995) 1655–1656. cells, Adv. Mater. 13 (2001) 1673–1677.
[27] A.C. Templeton, S. Chen, S.M. Gross, R.W. Murray, Water-soluble, [48] M.E. Akerman, W.C.W. Chan, P. Laakkonen, S.N. Bhatia, E. Ruoslahti,
isolable gold clusters protected by tiopronin and coenzyme A Nanocrystals targeting in vivo, Proc. Natl. Acad. Sci. U. S. A. 99 (2002)
monolayers, Langmuir 15 (1999) 66–76. 12617–12621.
[28] J.L. de Paz, R. Ojeda, A.G. Barrientos, S. Penadés, M. Martín-Lomas, [49] K. Hanaki, A. Momo, T. Oku, A. Komoto, S. Maensono, Y. Yamaguchi,
Synthesis of a Ley neoglycoconjugate and Ley-functionalized gold K. Yamamoto, Semiconductor quantum dot/albumin complex is a long-
glyconanoparticles, Tetrahedron: Asymmetry 16 (2005) 149–158. life and highly photostable endosome marker, Biochem. Biophys. Res.
[29] C. Lopez-Cartes, T.C. Rojas, R. Litran, D. Martinez-Martinez, J.M. de la Commun. 302 (2003) 496–501.
Fuente, S. Penades, A. Fernandez, Gold nanoparticles with different [50] G. Spreitzer, J.M. Whitling, J.D. Madura, D.W. Wright, Peptide-
capping systems: an electronic and structural XAS analysis, J. Phys. encapsulated CdS nanoclusters from a combinatorial ligand library,
Chem., B 109 (2005) 8761–8766. Chem. Commun. (2000) 209–210.
650 J.M. de la Fuente, S. Penadés / Biochimica et Biophysica Acta 1760 (2006) 636–651

[51] S. Sapra, J. Nanda, D.D. Sarma, F. Abed El-Al, G. Hodes, Blue emission transcription factors AP1 and CREB, J. Biol. Chem. 273 (1998)
from cysteines ester passivated cadmium sulfide nanoclusters, Chem. 2517–2525.
Commun. (2001) 2188–2189. [74] E. Otsuji, Y.S. Park, K. Tashiro, N. Kojima, T. Toyokuni, S.-I. Hakomori,
[52] E.R. Goldman, E.O. Balghian, H. Mattousi, M.K. Kuno, J.M. Mauro, P.T. Inhibition of B16 melanoma metastasis by administration of GM3- or
Tron, G.P. Anderson, Avidin: a natural bridge for quantum dot-antibody Gg3-liposomes-blocking adhesion of melanoma-cells to endothelial-cells
conjugates, J. Am. Chem. Soc. 124 (2002) 6378–6382. (antiadhesion therapy) via inhibition of GM3–Gg3Cer or GM3–LacCer
[53] X. Wu, H. Liu, J. Liu, K.N. Haley, J.A. Treadway, J.P. Larson, N. Ge, F. interaction, Int. J. Oncol. 6 (1995) 319–327.
Peale, M.P. Bruchez, Immunofluorescent labeling of cancer marker Her2 [75] S. Yu, N. Kojima, S.-I. Hakomori, S. Kudo, S. Inoue, Y. Inoue, Binding of
and other cellular targets with semiconductor quantum dots, Nat. rainbow trout sperm to eggs is mediated by strong carbohydrate-to-
Biotechnol. 21 (2003) 41–46. carbohydrate interaction between (KDN)GM3 (deaminated neuraminyl
[54] J.K. Jaiswal, H. Mattousi, J.M. Mauro, S.M. Simon, Long-term multiple ganglioside) and Gg3-like epitope, Proc. Natl. Acad. Sci. U. S. A. 99
color imaging of live cells using quantum dots, Nat. Biotechnol. 21 (2002) 2854–2859.
(2003) 47–51. [76] D. Spillman, M.M. Burger, in: B. Ernst, G.W. Hart, P. Sinay (Eds.),
[55] B. Dubertret, P. Skourides, D.J. Norris, V. Noireaux, A.H. Brivanlou, A. Carbohydrate–Carbohydrate Interactions, Wiley-VCH, Weinheim, 2000,
Libchaber, In vivo imaging of quantum dots encapsulated in phospolipid pp. 1061–1091.
micelles, Science 298 (2002) 1759–1762. [77] G.N. Misevic, M.M. Burger, Carbohydrate–carbohydrate interactions of
[56] R. Mahtab, J.P. Rogers, C.J. Murphy, Protein-sized quantum dot a novel acidic glycan can mediate sponge cell adhesion, J. Biol. Chem.
luminescence can distinguish between “straight”, “bent”, and “kinked” 268 (1993) 4922–4929.
oligonucleotides, J. Am. Chem. Soc. 117 (1995) 9099–9100. [78] W. Burkat, M.M. Burger, Reconstitution of high cell binding affinity of a
[57] D. Gerion, W.J. Parak, S.C. Williams, D. Zanchet, C.M. Micheel, A.P. marine sponge aggregation factor by cross-linking of small low affinity
Alivisatos, Sorting fluorescent nanocrystals with DNA, J. Am. Chem. fragments into a large polyvalent polymer, J. Biol. Chem. 261 (1986)
Soc. 124 (2002) 7070–7074. 2853–2859.
[58] D.J. Maxwell, J.R. Taylor, S.J. Nie, Self-assembled nanoparticle probes [79] D. Spillmann, K. Hard, J.E. Thomas-Oates, J.F.G. Vliegenthart, G.N.
for recognition and detection of biomolecules, J. Am. Chem. Soc. 124 Misevic, M.M. Burger, J. Finne, Characterization of a novel pyruvylated
(2002) 9609–9612. carbohydrate unit implicated in the cell aggregation of the marine sponge
[59] S.J. Rosenthal, I. Tomlinson, E.M. Adkins, S. Schroeter, S. Adams, L. Microciona prolifera, J. Biol. Chem. 268 (1993) 13378–13387.
Swafford, J. McBride, Y. Wang, L.J. DeFelice, R.D. Blakely, Targeting [80] D. Spillmann, J.E. Thomas-Oates, J.A. Van Kuik, J.F.G. Vliegenthart, G.
cell surface receptors with ligand-conjugated nanocrystals, J. Am. Chem. Misevic, M.M. Burger, J. Finne, Characterization of a novel sulfated
Soc. 124 (2002) 4586–4594. carbohydrate unit implicated in the carbohydrate–carbohydrate aggrega-
[60] S. Westenhoff, N.A. Kotou, Quantum dot on a rope, J. Am. Chem. Soc. tion of the marine sponge Microciona prolifera, J. Biol. Chem. 270
124 (2002) 2448–2449. (1995) 5089–5097.
[61] I. Potapova, R. Mruk, S. Preht, R. Zentel, T. Basché, A. Mews, [81] M.R. Wormald, C.J. Edge, R.A. Dwek, The solution conformation of the
Semiconductor nanocrystals with multifunctional polymer ligands, Lex group, Biochem. Biophys. Res. Commun. 180 (1991) 1214–1221.
J. Am. Chem. Soc. 125 (2003) 320–321. [82] H. Benoit, D. Herve, M. Pristchepa, P. Berthault, Y.-M. Zhang, J.M.
[62] Y. Chen, T. Ji, Z. Rosenzweig, Synthesis of glyconanospheres containing Mallet, J. Esnault, P. Sinay, NMR study of a Lewis X pentasaccharide
luminescent CdSe-ZnS quantum dots, Nano Lett. 3 (2003) 581–584. derivative: solution structure and interaction with cations, Carbohydrate
[63] X.-L. Sun, W. Cui, C. Haller, E.L. Chaikof, Site-specific multivalent Res. 315 (1999) 48–62.
carbohydrate labeling of quantum dots and magnetic beads, ChemBio- [83] N. Kojima, B.A. Fenderson, M.R. Stroud, I.R. Goldberg, R. Habermann,
Chem 5 (2004) 1593–1596. T. Toyokuni, S.-I. Hakomori, Further-studies on cell-adhesion based on
[64] J.M. de la Fuente, S. Penadés, Glyco-quantum dots: a new luminescent Le(X)–Le(X) interaction, with new approaches—embryoglycan aggre-
system with multivalent carbohydrate display, Tetrahedron: Asymmetry gation of F9 teratrocarcinoma cells, and adhesion of various tumor-cells
16 (2005) 387–391. based on Le(X) expression, Glycoconj. J. 11 (1994) 238–248.
[65] A. Robinson, J.-M. Fang, P.T. Chou, K.W. Liao, R.M. Chu, S.-J. Lee, [84] B.T. Houseman, M. Mrksich, Model systems for studying polyvalent
Probing lectin and sperm with carbohydrate-modified quantum dots, carbohydrate binding interactions, Top. Curr. Chem. 218 (2002) 1–44.
ChemBioChem 6 (2005) 1899–1905. [85] M. Mammen, S.K. Choi, G.M. Whitesides, Polyvalent interactions in
[66] C.C. Berry, A.S.G Curtis, Functionalisation of magnetic nanoparticles for biological systems: implications for design and use of multivalent ligands
applications in biomedicine, J. Phys., D, Appl. Phys. 36 (2003) and inhibitors, Angew. Chem. Int. Ed. 37 (1998) 2754–2794.
R198–R206. [86] J.M. de la Fuente, P. Eaton, A.G. Barrientos, M. Menedez, S. Penades,
[67] Q.A. Pankhurst, J. Connolly, S.K. Jones, J. Dobson, Applications of Thermodynamic evidence for Ca2+-mediated self-aggregation of Lewis X
magnetic nanoparticles in biomedicine, J. Phys., D, Appl. Phys. 36 (2003) gold glyconanoparticles. A model for cell adhesion via carbohydrate–
R167–R181. carbohydrate interaction, J. Am. Chem. Soc. 127 (2005) 6192–6197.
[68] S. Penades, M. Martin-Lomas, J.M. de la Fuente, T.W. Rademacher, [87] A.C. de Souza, K.M. Halkes, J.D. Meeldijk, A.J. Verkleij, J.F.C.
Magnetic Nanoparticles, WO 2004/108165 A2 Vliegenthart, J.P. Kamerling, Gold glyconanoparticles as probes to
[69] J.M. de la Fuente, D. Alcántara, S. Penadés, unpublished results. explore the carbohydrate-mediated self-recognition of marine sponge
[70] I. Eggens, B.A. Fenderson, T. Toyokuni, B. Dean, M.R. Stroud, S. cells, ChemBioChem 6 (2005) 828–831.
Hakomori, Specific interaction between Lex and Lex determinants: a [88] M.J. Hernáiz, J.M. de la Fuente, A.G. Barrientos, S. Penadés, A model
possible basis for cell recognition in preimplantation embryos and system mimicking glycosphingolipid clusters to quantify carbohydrate
in embryonal carcinoma cells, J. Biol. Chem. 264 (1989) self-interactions by surface plasmon resonance, Angew. Chem. Int. Ed.
9476–9484. 41 (2002) 1554–1557.
[71] S.-I. Hakomori, Carbohydrate–carbohydrate interactions as an initial step [89] S.-K. Choi, Synthetic Multivalent Molecules, Wiley-Interscience, USA,
in cell recognition, Pure Appl. Chem. 63 (1991) 473–482. 2004.
[72] N. Kojima, S.-I. Hakomori, Specific interaction between gangliotriao- [90] J.J. Lundquist, E.J. Toone, The cluster glycoside effect, Chem. Rev. 102
sylceramide (Gg3) and sialosyllactosylceramide (GM3) as a basis for (2002) 555–578.
specific cellular recognition between lymphoma and melanoma cells, [91] R. Roy, M.G. Baek, Glycodendrimers: novel glycotope isosteres
J. Biol. Chem. 264 (1989) 20159–20162. unmasking sugar coding, case with T-antigen markers from breast cancer
[73] Y. Song, D.A. Withers, S.-I. Hakomori, Globoside-dependent adhesion of MUC1 glycoprotein, J. Biotechnol. 90 (2002) 291–309.
human embryonal carcinoma cells, based on carbohydrate–carbohydrate [92] W.B. Turnbull, J.F. Stoddart, Design and synthesis of glycodendrimers,
interaction, initiates signal transduction and induces enhanced activity of Rev. Mol. Biotechnol. 90 (2002) 231–255.
J.M. de la Fuente, S. Penadés / Biochimica et Biophysica Acta 1760 (2006) 636–651 651

[93] M.J. Cloninger, Biological applications of dendrimers, Curr. Opin. Chem. [113] J.M. de la Fuente, C.C. Berry, Tat peptide as an efficient molecule to
Biol. 6 (2002) 742–748. translocate gold nanoparticles into the cell nucleus, Bioconj. Chem. 16
[94] S.K. Choi, M. Mammen, G.M. Whitesides, Generation and in situ (2005) 1176–1180.
evaluation of libraries of poly(acrylic acid) presenting sialosides as side [114] J. West, N. Halas, Applications of nanotechnology to biotechnology:
chains as polyvalents inhibitors of influenza-mediated hemagglutination, commentary, Curr. Opin. Biotechnol. 11 (2000) 215–217.
J. Am. Chem. Soc. 119 (1997) 4103–4111. [115] D. Hogemann, V. Ntziachristos, L. Josephnon, R. Weissleder, High
[95] K.H. Mortell, R.V. Weatherman, L.L. Kiessling, Recognition specificity throughput magnetic resonance imaging for eveluating targeted nano-
of neoglycopolymers prepared by ring-opening metathesis, J. Am. Chem. particles probes, Bioconj. Chem. 13 (2002) 116–121.
Soc. 118 (1996) 2297–2298. [116] J. Liu, Q. Zhang, E. Remsen, K. Wooley, Nanostructured materials
[96] E. Arranz-Plaza, A.S. Tracy, A. Siriwardena, J.M. Pierce, G.-J. Boons, High- designed for cell binding and transduction, Biomacromolecules 2 (2001)
avidity, low-affinity multivalent interactions and the block to polyspermy in 362–368.
Xenopus laevis, J. Am. Chem. Soc. 124 (2002) 13035–13046. [117] S.M. Marikanos, M.F. Anderson, J. Ryan, L.D. Martin, P.L. Feldheim,
[97] S. Thobhani, B. Ember, A. Siriwardena, G.-J. Boons, Multivalency and Encapsulation, permeability, and cellular uptake characteristics of hollow
the mode of action of bacterial sialidases, J. Am. Chem. Soc. 125 (2003) nanometer-sized conductive polymer capsules, J. Phys. Chem., B 105
7154–7155. (2001) 8872–8876.
[98] Y.C. Lee, R.T. Lee, Carbohydrate–protein interactions: basis of [118] C. Feldherr, R. Lanford, D. Akin, Signal-mediated nuclear transport in
glycobiology, Acc. Chem. Res. 28 (1995) 321–327. simian virus 40-transformed cells is regulated by large tumor antigen,
[99] J.E. Kingery-Wood, K.W. Williams, G.B. Sigal, G.W. Whitesides, The Proc. Natl. Acad. Sci. U. S. A. 89 (1992) 11002–11005.
agglutination of erythrocytes by influenza virus is strongly inhibited by [119] A.G. Tkachenko, H. Xie, D. Coleman, W. Glomm, J. Ryan, M.F.
liposomes incorporating an analog of sialyl gangliosides, J. Am. Chem. Anderson, S. Frazen, D.L. Feldheim, Multifunctional gold nanoparticle–
Soc. 114 (1992) 7303–7305. peptide complexes for nuclear targeting, J. Am. Chem. Soc. 125 (2003)
[100] G. Thoma, A.G. Katopodis, N. Voelcker, R.O. Duthaler, M.B. Streiff, 4700–4701.
Novel glycodendrimers self-assemble to nanoparticles which function as [120] C.-C. Lin, Y.-C. Yeh, C.-Y. Yang, C.-L. Chen, G.-F. Chen, C.-C. Chen,
polyvalent ligands in vitro and in vivo, Angew. Chem. Int. Ed. 41 (2002) Y.-C. Wu, Selective binding of mannose-encapsulated gold nanoparticles
3195–3198. to type 1 pili in Escherichia coli, J. Am. Chem. Soc. 124 (2002)
[101] E. Jule, Y. Nagasaki, K. Kataoka, Lactose-installed poly(ethylene glycol)- 3508–3509.
poly(D,L-lactide) block copolymer micelles exhibit fast-rate binding and [121] K. Aslan, J.R. Lakowicz, C.D. Geddes, Nanogold-plasmon-resonance-
high affinity toward a protein bed simulating a cell surface. A surface based glucose sensing, Anal. Biochem. 330 (2004) 145–155.
plasmon resonance study, Bioconj. Chem. 14 (2003) 177–186. [122] K. Aslan, J. Zhang, J.R. Lakowicz, C.D. Geddes, Saccharide sensing
[102] A. Nelson, J.M. Belitsky, S. Vidal, S.C. Joiner, L.G. Baum, J.F. Stoddart, using gold and silver nanoparticles—A review, J. Fluoresc. 14 (2004)
A self-assembled multivalent pseudopolyrotaxane for binding galectin-1, 391–400.
J. Am. Chem. Soc. 126 (2004) 11914–11922. [123] Y.-J. Chen, S.-H. Chen, Y.-Y. Chien, Y.-W. Chang, H.-K. Liao, C.-Y.
[103] S. André, R.J. Pieters, I. Vrasidas, H. Kaltner, I. Kuwabara, F.-T. Liu, Chang, M.-D. Jan, K.-T. Wang, C.-C. Lin, Carbohydrate-encapsulated
R.M.J. Liskamp, H.-J. Gabius, Wedgelike glycodendrimers as inhibi- gold nanoparticles for rapid target-protein indentification and binding-
tors of binding of mammalian galectins to glycoproteins, lactose epitope mapping, ChemBioChem 6 (2005) 1169–1173.
maxiclusters, and cell surface glycoconjugates, ChemBioChem 2 [124] J. Rojo, V. Diaz, J.M. de la Fuente, I. Segura, A.G. Barrientos, H.H.
(2001) 822–830. Riese, A. Bernad, S. Penades, Gold glyconanoparticles as new tools in
[104] S. André, P.J. Ortega, M.A. Pérez, R. Roy, H.-J. Gabius, Lactose- antiadhesive therapy, ChemBioChem 5 (2004) 291–297.
containing starburst dendrimers: influence of dendrimer generation and [125] J.H. Fendler, Self-assembled nanostructured materials, Chem. Mater.
binding-site orientation of receptors (plant/animal lectins and immuno- 8 (1996) 1616–1624.
globulins) on binding properties, Glycobiology 9 (1999) 1253–1261. [126] D. Quin, Y. Xia, B. Xu, H. Yang, C. Zhu, G.M. Whitesides, Fabrication of
[105] P.I. Kitov, J.M. Sadowska, G. Mulvey, G.D. Armstrong, H. Ling, N.S. ordered two-dimensional arrays of micro- and nanoparticles using
Pannu, R.J. Read, D.R. Bundle, Shiga-like toxins are neutralized by patterned self-assembled monolayers as templates, Adv. Mater. 11
tailored multivalent carbohydrate ligands, Nature 403 (2000) 669–672. (1999) 1433–1437.
[106] E.K. Fan, Z.S. Zhang, W.E. Minke, Z. Hou, C.L.M.J. Verlinde, W.G.J. [127] R.L. Letsinger, R.V.G. Elghanian, C.A. Mirkin, Use of a steroid cyclic
Hol, High-affinity pentavalent ligands of Escherichia coli heat-labile disulfide anchor in constructing gold nanoparticle-oligonucleotide
enterotoxin by modular structure-based design, J. Am. Chem. Soc. 122 conjugates, Bioconj. Chem. 11 (2000) 289–291.
(2000) 2663–2664. [128] K.J. Watson, J. Zhu, S.-B.T. Nguyen, C.A. Mirkin, Hybrid nanoparticles
[107] N. Stromberg, P.-G. Nyholm, I. Pascher, S. Normark, Saccharide with block copolymer shell structures, J. Am. Chem. Soc. 121 (1999)
orientation at the cell-surface affects glycolipid receptor function, 462–463.
Proc. Natl. Acad. Sci. U. S. A. 88 (1991) 9340–9344. [129] L.C. Brousseau, Q. Zhao, D.A. Shultz, D.L. Feldheim, pH-gated single-
[108] P.H. Weigel, R.L. Schnaar, M.S. Kuhlenschmidt, E. Schmell, R.T. Lee, electron tunneling in chemically modified gold nanoclusters, J. Am.
Y.C. Lee, Adhesion of hepatocytes to immobilized sugars. A threshold Chem. Soc. 120 (1998) 7645–7646.
phenomenon, J. Biol. Chem. 254 (1979) 10830–10838. [130] K.C. Grabar, R.G. Freeman, M.B. Hommer, M.J. Natan, Preparation and
[109] C.W. Cairo, J.E. Gestwicki, M. Kanai, L.L. Kiessling, Control of characterization of Au colloid monolayers, J. Am. Chem. Soc. 67 (1995)
multivalent interactions by binding epitope density, J. Am. Chem. Soc. 735–743.
124 (2002) 1615–1619. [131] J. Hu, J. Zhang, F. Liu, K. Kitteredge, J.K. Whiteshell, M.A. Fox,
[110] N. Horan, L. Yan, H. Isobe, G.M. Whitesides, D. Kahne, Nonstatistical Competitive photochemical reactivity in a self-assembled monolayer on a
binding of a protein to clustered carbohydrates, Proc. Nat. Acad. Sci. colloidal gold cluster, J. Am. Chem. Soc. 123 (2001) 1464–1470.
U. S. A. 96 (1999) 11782–11786. [132] T.C. Rojas, J.M. de la Fuente, A.G. Barrientos, S. Penades, L. Ponsonnet,
[111] E.K. Woller, E.D. Walter, J.R. Morgan, D.J. Singel, M.J. Cloninger, A. Fernandez, Gold glyconanoparticles as building blocks for nanoma-
Altering the strength of lectin binding interactions and controlling the terials design, Adv. Mater. 14 (2002) 585–588.
amount of lectin clustering using mannose/hydroxyl-functionalized [133] A.P. Alivisatos, Semiconductor clusters, nanocrystals, and quantum dots,
dendrimers, J. Am. Chem Soc. 125 (2003) 8820–8826. Science 27 (1996) 933–937.
[112] C.-S. Tsai, T.-B. Yu, C.-T. Chen, Gold nanoparticle-based competitive [134] I. Coulthard, S. Degen, Y.-J. Zhu, T.K. Sham, Gold nanoclusters
colorimetric assay for detection of protein–protein interactions, Chem. reductively deposited on porous silicon: morphology and electronic
Commun. (2005) 4273–4275. structures, Can. J. Phys. 76 (1998) 1707–1716.

You might also like