Transcript
Page 1: Self-assembly of designer biosurfactants

Dynamic Article LinksC<Soft Matter

Cite this: Soft Matter, 2011, 7, 7150

www.rsc.org/softmatter REVIEW

Publ

ishe

d on

31

May

201

1. D

ownl

oade

d by

Uni

vers

ity o

f N

ewca

stle

on

26/0

9/20

14 1

9:00

:30.

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

Self-assembly of designer biosurfactants

Debora Berti,* Costanza Montis and Piero Baglioni*

Received 6th February 2011, Accepted 27th April 2011

DOI: 10.1039/c1sm05197k

This contribution reports on some recent advances in the field of self-assembly of biologically inspired

amphiphiles, focusing on nucleolipids. Due to their important potential for biomedical applications,

the synthesis of this class of compounds can be considered a mature field, but precise relationships

between molecular level details and self-assembly properties still have to be elucidated in detail. This

review will present the most recent progresses in the field, along with the new possibilities opened up by

DNA nanobiotechnology, where the hydrophobic functionalization is meant as a tether for

incorporation in soft matrices, such as planar or free standing lipid bilayers.

Introduction

Self-assembly is the main construction pathway for soft matter

systems.1 At the same time it is the principal strategy that Nature

uses to build-up multifunctional systems, whose efficiency,

specificity and energetics have been refined through the iterations

of evolution. Non-covalent interactions between molecular

repeat units are at the basis of self-assembly. A prominent role in

this field is played by surfactant aggregation, where identical

amphiphilic building blocks associate thanks to the solvophobic

interactions of the hydrophobic tails.2 The most obvious natural

counterparts are cell membranes, whose skeleton is formed by

lipid molecules which have an amphiphilic character.3

However, in the bio-world self-assembly is realized not only

due to hydrophobic effects, but it also often results from

a combination of the previous contribution combined with

structural complementarity and specific biological interactions,

generally termed as molecular recognition.4 The similarities

between many biological systems and amphiphilic assemblies,

and the partial conceptual and methodological superposition

between the two research fields, have naturally led to a biomi-

metic fertilization of the surfactant world, directing the evolution

of conventional surfactant molecular units towards the incor-

poration of bio-relevant functions.5 With few notable exceptions,

the chemical modifications mainly concern the polar head

portion and therefore deal with the insertion of biological water-

soluble functionalities.6

The overall aim of this research field is to provide self-

assemblies with some of the biomimetic interactions that Nature

uses in addition to hydrophobic association; in this framework

these systems can be defined as biomimetic self-assemblies.7 The

CSGI and Department of Chemistry, University of Florence, Via dellaLastruccia 3, 50019 Sesto Fiorentino, Firenze, Italy. E-mail: [email protected]; [email protected]; Fax: +39 055 4573032; Tel: +39 0554573033

7150 | Soft Matter, 2011, 7, 7150–7158

term biosurfactant is not used in this review to define surface-

active molecules synthesized by living cells, but rather represents

a contraction of the definition bio-inspired surfactants, and

clearly identifies amphiphiles of synthetic origin.

The most important and paradigmatic examples concern

peptide amphiphiles8,9 and nucleolipids.6,10 The rapid growth of

the use of nucleotides and aminoacid constituents as building

blocks for novel nanostructured materials has been made

possible by the availability of designer custom sequences offered

by the solid-phase peptide11 and oligonucleotide synthesis

methods.12

For peptide amphiphiles, the choice of the sequence includes

about 20 aminoacids with varying charge and hydrophobicity;

this allows a wide selection both for the polar head sequence and

for the hydrophobic tag. When both amphiphilic portions are

formed by aminoacid constituents, the resulting molecule is

called full-peptide amphiphile,10 to distinguish it from the case

where the apolar tail is an alkyl chain attached to the N-terminus

of the peptide sequence. Several examples and reports of self-

assembly from peptide amphiphiles have appeared in the litera-

ture; the latest advances in this field have been extensively

reviewed by Zhang and co-workers, and we refer the reader

interested in this important and emerging area, to this

contribution.11

According to the definition proposed by Rosemeyer in 2005,12

a nucleolipid is a molecule where a nucleic base, a nucleoside,

a nucleotide or an oligonucleotide (see Fig. 1) is covalently

conjugated to a lipid or more generally to a hydrophobic

molecular unit.

The choice for the nucleic portion is surely more limited (four

different kinds of bases in DNA) than for peptide amphiphiles.

The high fidelity of A–T (A–U in RNA) and G–CWatson–Crick

hydrogen-bonding pairing, provides a unique toolbox, despite

the simplicity of this simple four-letter alphabet.13

A nucleo-amphiphile can be globally neutral if a nucleoside or

a nucleic base, both uncharged (see Fig. 1), is derivatized with

This journal is ª The Royal Society of Chemistry 2011

Page 2: Self-assembly of designer biosurfactants

Fig. 1 Chemical structure of DNA chain with the four nucleobases

encircled and the nucleoside and nucleotide structures highlighted.

Publ

ishe

d on

31

May

201

1. D

ownl

oade

d by

Uni

vers

ity o

f N

ewca

stle

on

26/0

9/20

14 1

9:00

:30.

View Article Online

a single or a double hydrophobic chain. In some particular cases,

the insertion of cationic groups in the lipid architecture provides

cationic or zwitterionic nucleolipids. The self-assembly and the

applications of these nucleolipids have been reviewed

recently.6,12,14 In the vast majority of examples, when a nucleotide

or an oligonucleotide is used, the molecular unit has a single or

a multiple negative charge. The latter case is the more relevant in

the quest for biomimetic self-assembly.

Oligonucleotide tags can function as molecular zippers

between molecular units or self-assembled objects, provided the

complementarity of the single strands, in a way that is responsive

to temperature and ionic strength.

The limited nucleic bases synthetic library is enriched by

a relevant number of synthetic base analogues which possess

antiviral or antitumor therapeutic efficacy15,16 and can be

included in the ensemble of nucleic base library accessible to

chemists. This research area is also of importance in the field of

enhanced prodrugs, where the lipophilic tag induces self-

assembly that in turn modulates bioavailability.17–19

As we will see, a host of different applications can be envisaged

for the nanostructured organized assemblies arising from self-

assembly of these biosurfactants, from biomedical formulations

to building block for bionanotechnology, to biomimetic systems.

The level of specificity is high by design, therefore the

formulation of organized assemblies from biosurfactants, both

for fundamental studies and for applicative purposes, requires

This journal is ª The Royal Society of Chemistry 2011

a careful consideration of synthetic and chemical details. As the

level of chemical complexity of the molecular units increases, it is

important to maintain structural and dynamical control over the

formed assemblies. This requires an integrated approach to the

field and, considering the application in biomedical and biomi-

metic fields and the novel emerging possibilities in nanotech-

nology, a truly interdisciplinary research strategy.

The focus of this paper is to examine recent advances in the

field of self-assembly of biologically inspired synthetic

nucleolipids.

This subject has been reviewed in the past years,10,12 but it is

undoubtedly a fast growing area. In the following sections, after

an introduction of the synthetic approaches, we will provide an

outline of only the most recent advances in the field.

Nucleolipid synthesis

Synthetic nucleolipid chemistry can be considered a mature field,

whose development has been promoted by the use of such

molecules as drugs or prodrugs, consisting in cytotoxic nucleic

base analogues functionalized with hydrophobic tags. These

molecules are efficient chemotherapeutics, with improved phar-

macokinetic, pharmacodynamic and toxicological properties

thanks to the enhanced uptake from cells and an increased

therapeutic efficacy (mainly antitumoral), associated to lower

adverse effects.

Examples of natural nucleolipids involved in important biolog-

ical processes aremolecules characterizedby antibiotic activity,20as

Tunicamycin,21 which works as an inhibitor of oligosaccharide

synthesis in eukaryotic cells and has demonstrated therapeutic

properties as antimicrobial, antifungal, antiviral and antitumor

agent12,22 or cytidinediphosphatediacylglycerol coenzyme (CDP-

DAG), a natural nucleolipid present both in mammalian and

prokaryotic cells involved in the biosynthesis of glycer-

ophospholipids and in the formation of some membrane compo-

nents like phosphatidylinosides and cardiolipins.23

The important role played by these molecules in biological

systems has inspired the synthesis and investigation of new

nucleolipids characterized by therapeutic activities, and at the

same time has prompted the development of novel synthetic

routes for the derivatization of nucleic acid functions.

The synthetic routes to achieve hybrid lipid–nucleoside/

nucleotide structures can target different atoms for the covalent

attachment of the lipid moiety as shown in Fig. 2.

50-Ribose or deoxyribose functionalization

The first possibility is to bind the lipid moiety to the 50 carbonatom of the sugar ring to obtain nucleoside alkyl esters or

nucleotide phosphate alkyl esters. Examples of this route are the

MacCoss synthesis of 1-b-D-arabinofuranosylcytosine (ara-C)24

and 1-b-D-arabinofuranosyladenine (ara-A)25,26 lipid derivatives,

obtained through the reaction of the nucleoside-50-mono-

phosphate converted to the respective 50-phosphomorpholidate

with the pyridinium salt of L-a-dipalmitoylphosphatidic acid.

The presence of the lipid chain determined for this nucleolipid an

enhancement of the antitumoral effect with respect to naked ara-

A and ara-C, attributed to the enhanced cell uptake and cata-

bolic stability of the lipid derivatives.

Soft Matter, 2011, 7, 7150–7158 | 7151

Page 3: Self-assembly of designer biosurfactants

Fig. 2 Scheme illustrating the different derivatization strategies in the

synthesis of nucleolipids.

Publ

ishe

d on

31

May

201

1. D

ownl

oade

d by

Uni

vers

ity o

f N

ewca

stle

on

26/0

9/20

14 1

9:00

:30.

View Article Online

Ara-C analogues have been also studied by Turcotte et al.,27,28

who synthesized compounds derived by either cytosine-50-diphosphate and the corresponding ara-C nucleotide as well as

by Hong et al.,29 who reported the synthesis of new ara-CDP-

steroids and ara-CMP- and ara-CDP-alkyl esters, as reported in

an exhaustive review by Rosemeyer.12

In 2003 Kim and co-workers30 proposed the synthesis of

thymidine-based nucleolipids obtained by using four different

linking units (urea, amide, carbamate and ester) for the attach-

ment of a lipid chain in 50 position. The properties of these

molecules as organogelators were evaluated: fibrous structures

were obtained in urea-linked gelator/cyclohexane systems, woven

structures in ester-linked gelator/toluene and lamellar structures

in amide-linked gelator/toluene. The differences in the organo-

gelators’ gelation ability was attributed to the formation of

mutually exclusive gel networks caused by intermolecular

hydrogen-bonding interactions.

In 1987 Shuto and co-workers31 proposed an enzymatically-

catalyzed reaction for the formation of hybrid lipid–nucleoside

structures. The synthesis of nucleolipids derives from phos-

phocholines and is based on transphosphatidylation reaction

catalyzed by phospholipase D from Streptomyces sp. AA 586

(PLDP) with different alkanols as acceptors. This synthetic route

is particularly relevant for therapeutic applications, due to the

catalytic activity of phospholipases in vivo that provides a meta-

bolic pathway for the degradation of the drug and a decrease of

possible toxic effects. A two-phase CHCl3/acetate buffer reaction

was used for the synthesis of various 50-(3-sn-phosphatidyl)nucleosides carrying pharmacologically active nucleoside head-

groups, such as 5-fluorouridine and arabinocytidine, from the

corresponding phosphatidylcholine and nucleoside PLDP

substrate.32,33 A modification of this synthetic route was carried

out by Berti et al.34–36 for the synthesis of phosphatidylnucleo-

sides characterized by different chains and polar headgroups.

The self-assembly properties of nucleolipids have been investi-

gated highlighting the role of nucleolipid polar headgroups in the

structure of nucleolipid self-assemblies, which will be illustrated

in the following section.

20–30-Ribose or 30-deoxyribose functionalization

Other reactive positions on nucleosides and nucleotides, useful

for the functionalization of nucleic acids and the formation of

7152 | Soft Matter, 2011, 7, 7150–7158

hybrid lipid–nucleoside/nucleotide structures, are ribose 20 anddeoxyribose 30 hydroxy groups.

Schwendener et al.37 reported the synthesis of 30-O-palmitoyl-

nucleolipids of 5-fluoro-20-deoxyuridine by direct acylation with

the corresponding acylchloride. The lipid derivatives showed

a stronger efficacy as anticancer drugs with respect to the naked

fluorinated deoxyuridine; this was attributed to the incorpora-

tion of 30-O-palmitoyl-nucleolipids within liposomes that

provided a protection from degradation inside living organisms.

Barth�el�emy and co-workers presented the synthesis of phos-

phocholineuridineamphiphiles functionalized with two identical

fatty esters on the 20 and 30 positions.38 Dimirystoyl, dipalmitoyl,

distearoyl, diarachidoyl and dioleoyl derivatives were synthe-

sized and their self-assembly properties in aqueous solvent were

investigated. They formed fibers or bilayer supramolecular

structures and evidenced a thermoreversible transformation

from one structure to the other, depending on the melting

temperature of the amphiphiles. A similar fluorinated nucleo-

side-based amphiphile (20,30-O-di-2H,2H,3H,3H-perfluoro-

undecanoyl-uridine-50-phosphocholine) was synthesized with the

substitution of the 20 and 30 nucleoside ribose positions by fluo-

rocarbon chains and its self-assembly properties were investi-

gated.39 Shimizu and co-workers40 presented an example of

nucleotide bola-amphiphiles with a 30-phosphorylated thymidine

moiety at each end of oligomethylene spacers. The structure of

these nucleotide lipid conjugates is compatible with the structural

prerequisites of a good hydrogelator: the sugar and phospho-

diester moieties play an important role in imparting the addi-

tional hydrophilic functionality; the sugar moiety can provide

multiple chiral centers and the thymidine moiety is connected to

both ends of long oligomethylene chains. The oligonucleotide-

templated self-assembly of the thymidine bola-amphiphile with

a series of oligoadenylic acids was also investigated and the

formation of DNA-like nanofibers was obtained thanks to the

Watson–Crick interaction between adenine and thymine bases.41

Amphiphilic cationic uridine derivatives were synthesized42,43

as possible DNA vectors characterized by hydrogen bonding and

p-stacking capabilities, hence capable of summing up to elec-

trostatic compensation between DNA and the synthetic vector.

Moreover, recently the same authors presented the synthesis of

anionic nucleotide based lipids44 featuring thymidine-30-mono-

phosphate as the nucleotide and 1,2-diacyl-sn-glycerol as the

lipid moiety for in vitro delivery of nucleic acids.

Purinic or pyrimidinic heterocycle functionalization

Few examples in the literature report the direct functionalization

of heterocycles’ nucleobases with a lipid moiety. Purine’s N9 and

pyrimidine’s N1, chemically bound to the sugar moiety in

nucleosides and nucleotides, are possible reactive positions for

the attachment of a lipid chain, an example45 is the synthesis of

an acycloguanosine (Acyclovir) lipid derivative with therapeutic

properties for the treatment of Herpes simplex (Type I). An

acylguanosine nucleolipid, effective in the treatment of the same

disease, has been reported by Welch and co-workers.46 Other N9

purine nucleobases–lipid and N1 pyrimidine nucleobases–lipid

hybrid systems were synthesized by Rosemeyer and co-workers

and investigated in monolayers at the air–water interface.47

This journal is ª The Royal Society of Chemistry 2011

Page 4: Self-assembly of designer biosurfactants

Publ

ishe

d on

31

May

201

1. D

ownl

oade

d by

Uni

vers

ity o

f N

ewca

stle

on

26/0

9/20

14 1

9:00

:30.

View Article Online

Recently Hocek and co-workers48 presented the synthesis of

bile acid-functionalized deoxyribonucleosides (dN) and deoxy-

ribonucleoside triphosphates (dNTPs) with the attachment of the

bile acid moiety in position 5 of pyrimidine and in position 7 of

the 7-deazaadenine nucleosides, obtained by single step aqueous-

phase Sonogashira cross-coupling reactions. The bile acid

modified dNTPs were successfully incorporated in DNA

notwithstanding the presence of a very bulky and hydrophobic

steroid moiety in position 5 of pyrimidine or position 7 of

7-deazaadenine dNTPs.

Oligonucleotide–lipid conjugates

The possibility of synthetic oligonucleotides to interfere with the

regulation of genetic information in biological systems49–51 has

opened the way to their use in gene therapy, and determined

a growing interest also in the development of oligonucleotide–

lipid conjugates. In analogy with nucleoside lipids, the presence

of a hydrophobic segment attached to the oligonucleotide was

thought to promote its interaction with cell membrane and,

hence, to enhance cell uptake. Moreover, many lipophilic

moieties, like cholesterol, can target lipoproteins: therefore

cholesterol–oligonucleotide conjugates can be recognized by

hepatic cells and internalized by lipoprotein-mediated

endocytosis.52,53

The reader is addressed to the articles by Rosemeyer12 and

Gissot et al.17 for oligonucleotide–lipid conjugates, and to the very

recent review byDefrancq et al.54 for oligonucleotide conjugation

methodologies. From a synthetic point of view, it is possible to

insert the lipid moieties during the solid-phase synthesis of the

oligonucleotide sequence, or to modify oligonucleotides with the

hydrophobic moieties once the solid-phase synthesis is accom-

plished and oligonucleotides have been cleaved from the solid

support anddeprotected.Very few examples of the latter synthetic

route are reported in the literature, and the most common and

feasible approach is the first one. The conjugation of oligonucle-

otides is mostly at 50- or 30-termini because of their easy accessi-

bility. Moreover, due to the elongation of the oligonucleotide

chain in the 30–50 direction during the solid phase synthesis, the

modification ismore easily attached to the 50-end, compared to the

30-end; however, it has been suggested that conjugation through

the 30-end enhances the exonuclease resistance. A synthetic route

to achieve 30-end oligonucleotide conjugation was developed by

Letsinger et al.55 Liebscher and co-workers56 also presented the

synthesis of lipophilic uridines containing a lipid moiety into the

20-position. The importance of these 20-lipid functionalized

nucleosides is related to the possibility of introducing themodified

nucleosides into any position of an oligonucleotide strand. The

uridine derivatives containing one or two lipophilic groups in the

20-position, were studied for their incorporation within lipid

membranes and the effect of the exposure of nucleobases to an

aqueous environment for possible base-pairing.

Recently, Tan and co-workers57 proposed the solid-phase

synthesis of DNA–diacyl-lipid conjugate amphiphiles containing

a first segment of single-stranded DNA, highly hydrophilic,

a second segment of a pyrene molecule used as a fluorescence

reporter and a highly hydrophobic third segment composed of

two C18 hydrocarbon tails. When dispersed in aqueous solution,

such amphiphilic DNA spontaneously self-assembled into three-

This journal is ª The Royal Society of Chemistry 2011

dimensional spherical micelles with a DNA corona and a lipid

core. Moreover, micelles disintegrate when incubated with bio-

logical cells through permeation of the cell membrane by endo-

cytosis, opening perspectives for medical applications of these

systems in gene therapy.

Nucleolipid self-assembly: from fundamentals to bio-relevant applications

The self-assembly of amphiphilic molecules produces aggregates

characterized by a large structural diversity, in terms of size,

shape, and interfacial flexibility. The amphiphilic phase diagram

depends on parameters such as surfactant kind, volume fraction,

temperature, and ionic strength.

The main structural features of self-assemblies are related, in

the simplest approximation, to the relative cross-sectional areas

of the hydrophilic and hydrophobic parts, encoded in the

optimal packing parameter of the amphiphile.58 Subtle structural

variations of molecular details can lead to cascade effects with

dramatic morphological transitions on the mesoscale.

The possibility to integrate nucleic motifs into amphiphilic

molecules and use the know-how of soft matter to engineer nano-

objects with nucleic decoration can ultimately lead to extremely

complex and fascinating structures. As the molecular ‘‘bricks’’

become more and more sophisticated and additional control

parameters are chemically introduced, it becomes central to

assess how these added functionalities affect self-assembly and

respond to variation of external conditions. Simple consider-

ations on the relative steric hindrances of the molecular portions

(i.e. the packing parameter) are usually not sufficient, as polar

heads do not merely interact through excluded volume and

electrostatic repulsions.59,60 Such structural complexity requires

an in-depth structural characterization, necessary to highlight

the contribution from base–base interactions to the overall

properties of the assemblies.

An ideal system to test such fundamental ideas is represented

by very simple nucleolipids, i.e. phospholipid-based anionic

nucleolipids, PLN (phosphatidyl nucleosides),31–36 where one

single RNA nucleoside is enzymatically attached to the polar

head of a lecithin substituting a choline headgroup, as already

mentioned in the previous paragraph. These derivatives have

been studied in detail in the last decade. Both the hydrophobic

moiety and the nucleic headgroup have been systematically

varied to modulate the packing parameter and trigger/tune base–

base interactions at the interface of the self-assembled structures.

For a given lipid derivatization, the microstructure of self-

assemblies depends markedly on the kind of nucleic base due to

interpolar-head attractive interactions, notwithstanding the

negative charge, which would rather predict the prevalence of

unspecific electrostatic repulsion.61–63

An illuminating example is provided by dilauroyl-phospha-

tidyl-N (DLPN), with N being either adenosine or uridine, whose

aggregation properties have been studied in detail,64–71 to better

highlight the contribution of the functional polar head to local

and mesoscopic properties of the aggregates.

Freshly prepared DLPA solutions are mostly composed of

threadlike micelles whose cross-section size is about 5 nm. The

observed time evolution of the aggregate morphology results in

the twisted superstructures reported in Fig. 3 and accompanied by

Soft Matter, 2011, 7, 7150–7158 | 7153

Page 5: Self-assembly of designer biosurfactants

Fig. 4 Illustrative cartoon for a possible polyU arrangement in

a nucleolipoplex. For the sake of clarity polyU is here represented as

a rigid rod. (Reprinted with permission from ref. 81.)

Publ

ishe

d on

31

May

201

1. D

ownl

oade

d by

Uni

vers

ity o

f N

ewca

stle

on

26/0

9/20

14 1

9:00

:30.

View Article Online

the appearance of an ultraslow relaxation in quasielastic light

scattering, arisen from a hierarchical aggregation of wormlike

structures identical to DLPU micelles. The higher stacking

constants of the former nucleotide are responsible for the different

self-assembly behavior, which is strictly dependent on the thermal

history of the samples. This behavior has been proven to be

dependent on the thermally-activated redistribution of syn–anti

conformations of adenosine headgroups, which is then collectively

frozen thanks to molecular constraints present in the structure.71

Noticeably, mixed complementary phosphatidyl-nucleoside

self-assemblies (micelles, bilayers, etc.) bearing complementary

bases on the polar heads exhibit the signatures of interfacial

interactions both through stacking and specific hydrogen bonds,

according to a Watson–Crick pattern as in nucleic acids.35,36,72–78

Molecular recognition is triggered by self-assembly and affects

the structural features of the aggregates.

These assemblies provide negatively charged interfaces deco-

rated with nucleic motifs that can complex complementary

nucleic acid strands using, as the driving force, molecular

recognition instead of electrostatic interactions. Globular

micelles composed of 1,2-dioctanoyl-phosphatidyl-adenosine

interact with complementary RNA single strands (ss-poly-

uridylic acid), forming a new hexagonal phase where the nucleic

acids are confined among cylindrical micelles.79 This provides

a proof-of-principle that complexation is possible notwith-

standing the same charge of both partners (i.e., lipid assembly

and nucleic acid). The investigation has also been extended to

zero-curvature assemblies (i.e., nucleolipid membranes) formed

by POPNs.80–82 Nucleolipid bilayers of POPA, when swollen with

aqueous solutions containing single-strand complementary

polynucleotides (polyuridylic acid) show an increase in the

smectic period with respect to binary POPA/water systems and

the appearance, upon annealing, of an additional SAXS peak

ascribable to 1D ordering of the biopolymer between the lamellar

stacks, as sketched in the cartoon reported in Fig. 4.81

None of the observed effects could be detected for identically

charged membranes formed by POPG, thus ruling out any

nonspecific effects due to the presence of biopolymer in the

swelling solution.

The extra-peak position scales in the SAXS spectrum with

biopolymer concentration similar to the scaling of lipoplexes.

Such novel structures, sketched in Fig. 4, have been termed

nucleolipoplexes, in analogy to classical lipoplexes.

Fig. 3 Cryo-TEM pictures of micellar aggregates formed by DLPU and

DLPA(1� 10�2M)Thebar is 100nm(Reprintedwithpermissionfromref. 68.)

7154 | Soft Matter, 2011, 7, 7150–7158

The above outlined approach represents a strategic shift from

a specific to tailored vectors for DNA delivery to modulate gene

function in vitro or in vivo. The use of anionic species such as

nucleotide lipids in formulations can optimize transfection effi-

cacy and offer an alternative to cationic species, which can be, in

some instances, cytotoxic.83 This strategy has been successfully

implemented with a similar anionic nucleolipid, diC16-30-dT, byBarth�el�emy and co-workers, who have demonstrated that

complexes with DNA are formed in the presence of calcium ions

and that the biomolecule is internalized into mammalian HEK

cells with enhanced transfection efficacy with respect to the

natural anionic DPPA and DPPG.44

Recently, Drummond and collaborators84 have reported on

the self-assembly of a novel amphiphile pro-drug 5-deoxy-5-flu-

oro-N4-(cis-9-octadecenyloxycarbonyl) cytidine. Similar to

many other nucleosidic chemotherapeutics, the insertion of

a specific lipid carrier linked to the nucleoside-analogue can

reduce the usually severe side effects, improve pharmacokinetic

behavior and overcome or decrease the development of resis-

tance against the drug, considerably improving the quality of life

for the patients. On the other side, the self-assembly pattern

modulates the bioavailability and the bioactivity profile.

The relevance of this study went, however, beyond these

important applicative aspects, to cover fundamental aspects of

self-assembly, yet to be addressed. The authors have observed for

the first time the sequence of structural intermediates, shown in

Fig. 5, in a temperature induced La to QIID transition, following

the formation of interbilayer contacts, the development of

membrane pores, the evolution into a hexagonal structure that

eventually evolves to a bicontinuous cubic structure. These

observations provide mechanistic insights into the formation of

highly ordered cubic membrane structures, exhibited by many

cellular organelles.

Recently, the synthesis of a novel family of glycosyl-nucleoside

lipids with a ‘‘double click’’ chemistry route was reported and

their characterization in terms of self-assembly properties,

described.85 These amphiphiles spontaneously assemble into

This journal is ª The Royal Society of Chemistry 2011

Page 6: Self-assembly of designer biosurfactants

Fig. 5 Schematic representation of the major transformation stages

involved in the creation of a bicontinuous cubic structure from a planar

lamellar structure. (Reprinted with permission from ref. 84.)

Fig. 6 DNA-programmed lipid assembles to form spherical lamellar

vesicles capable of switching phase to form small spherical micelles via

DNA hybridization (+DNA2) and strand invasion (+DNA3) cycles.

(Reprinted with permission from ref. 87.)

Publ

ishe

d on

31

May

201

1. D

ownl

oade

d by

Uni

vers

ity o

f N

ewca

stle

on

26/0

9/20

14 1

9:00

:30.

View Article Online

supramolecular structures, including fibers, vesicles, hydrogels,

and organogels, and are able to induce gelation both in water and

chloroform. The delivery of oligonucleotides into human hepatic

cells has also been reported by the same authors.86

The structure-encoded response to external stimuli is one of the

most interesting prerogatives of nucleolipid self-assemblies. The

design of molecular components for a tailored aggregation

behavior can concern the hydrophilic assembler, as the assembly

modulator, or the polar head. In the case of oligonucleotide-

conjugates, the design of the nucleic sequence represents probably

the most powerful and sensitive tool to control self-assembly.

One recent example concerns a very interesting change in

morphology, exhibited by a double-chained oligonucleotide

lipid.87 The design scheme, reported in Fig. 6, is conceived to

enable a programmable soft, discrete nanoscale morphology,

using DNA pairing to encode the amphiphilic building block

with information.

The derivative assembles spontaneously into bilayered vesi-

cles, but, upon pairing with a complementary oligonucleotide,

evolves into a micellar structure, in view of the increase of the

cross-sectional area of the hydrophilic section. If the pairing

oligonucleotide is designed with a dangling end, the subsequent

addition of a third oligonucleotide of identical contour length

and fully complementary to the second one (strand invasion),

reverts the self-assembly from micelles to vesicles, via strand

disassembly at the micellar surface.

Recently Arbuzova and co-workers have reported that

mixtures of simple synthetic phospholipids, like DOPC, with

a novel lipophilic nucleoside 20-N-(2-(cholesteryl)-succinyl)-20-deoxy-20-aminouridin self-assemble in microtubular morphol-

ogies. These authors observed the formation of open-ended

cylindrical structures with outer diameters between 2 and 3 mm

and lengths between 20 and 40 mm after hydration of mixed dry

lipid films, followed by heating to 70 �C and slow cooling to

room temperature. The composition of the initial lipid films and

the conditions of preparation allow some degree of control over

the final morphology of the tubes.88

Nucleolipids for nanobiotechnology

In this last paragraph we will provide some recent advances of

the use of nucleolipid in nanobiotechnology oriented

This journal is ª The Royal Society of Chemistry 2011

applications. The distinction from the cases treated in the

previous paragraph is somewhat arbitrary, as the boundaries

between these areas are actually blurred.

However, in these cases we will refer invariably to oligonu-

cleotide amphiphiles, where the design is operated on the oligo-

nucleotide portion and enables adsorption and orientation of

DNA chains at fluid surfaces including the air–water and oil–

water interfaces. The resulting assemblies are provided with

chemical information that can be translated into further hierar-

chical aggregation in functional arrays of nanounits.

Non-covalent DNA immobilization strategies can be realized

inserting the desired single-strand sequence bearing a hydro-

phobic anchor into a bilayer membrane; in this case the fluid two-

dimensional membrane allows for in-plane free diffusion and

rotation of lipid-associated molecules, thus promoting the

hybridization process at the surface. Therefore the insertion of

a lipophilic tag into an oligonucleotide sequence is solely inten-

ded as the hydrophobic anchor that provides tethering at a lipid/

water interface. It is then clear that oligonucleotide amphiphiles

are ‘‘functional dopants’’ into fluid matrices, typically bilayers;

they are used in very small quantities and their self-assembly is

generally not a concern, while it should be at least considered as

a competitive event with respect to adsorption at fluid interfaces.

Since their introduction89 cholesteryl-TEG conjugates are

generally considered good anchoring candidates since they show

low tendency to self-aggregation and induce a stabilizing effect

on the lipid bilayer. H€o€ok and Benkoski and Boxer et al. pio-

neered the concept that DNA hybridization can be used to tether

vesicles to planar lipid bilayers, at the same time allowing them to

freely diffuse in a plane parallel to the supported bilayer.90 Such

an approach can be envisioned as a useful method for arraying

Soft Matter, 2011, 7, 7150–7158 | 7155

Page 7: Self-assembly of designer biosurfactants

Fig. 7 Mixing/demixing of DNA_tocopherol*/DNAc2* (a–d; FITC-

labeled) and PNA_C16/DNAc1 (e–h; rhodamine-labeled) hybrids

incorporated in POPC/SSM/Chol GUVs due to temperature increase and

decrease. Below the phase transition temperature, the two lipids

DNA_tocopherol*/DNAc2* (a) and PNA_C16/DNAc1 (e) are located to

different domains (�5 �C). Above the phase transition temperature,

domains disappeared, and both constructs mix (b and f) (�55 �C). After

cooling (�46 �C), new smaller domains are formed (c and g) that merge to

larger domains separating again both types of constructs (d and h).

Bars ¼ 10 mm. (Reprinted with permission from ref. 101.)

Fig. 8 Incorporation of cholesteryl-TEG-functionalized oligonucleo-

tides within lipid membranes covering Layer-by-Layer SiO2 nano-

particles. (Reprinted with permission from ref. 99.)

Publ

ishe

d on

31

May

201

1. D

ownl

oade

d by

Uni

vers

ity o

f N

ewca

stle

on

26/0

9/20

14 1

9:00

:30.

View Article Online

integral membrane proteins in vesicles and as a platform for

studying their interactions with other proteins, small molecules,

and/or vesicles. Many collisions between individual tethered

vesicles have been observed with fluorescence microscopy, and

the vesicles show no tendency to stick to each other or fuse, while

docking between vesicles occurs if a second complementary

sequence, introduced through 50 lipophilic oligonucleotides, is

present. If vesicle coupling is accomplished through pairing of

50-lipophilic oligonucleotides with the complementary 30-conju-gate, the anti-parallel hybridization mediates vesicle fusion,

revealed by quick lipid mixing and slow content mixing.91–93 This

system represents a mimic of the complex events leading to

protein-mediated membrane fusion in synaptic vesicles, and, by

changing the length of the oligonucleotide sequence, allows

probing the effects of the distance between bilayers in the

process. The incorporation of cholesteryl-tagged oligonucleo-

tides in phospholipid vesicles has been also studied in solution,

establishing that the oligonucleotide conformation and hybrid-

ization kinetics with the complementary strand in solution

strongly depend on the macromolecular density at the liposomal

surface, and above a critical grafting density connected to the

lipophilic oligonucleotide excluded area, coupling kinetics are

slowed down relative to strand pairing in solution.94

Novel synthetic strategies for ss-DNA carrying lipophilic tags

have been recently presented, which allow for the introduction of

lipophilic groups into any position of the oligonucleotide

sequence applying phosphoramidite methodology.95,96 In the

case of multiple cholesteryl derivatives, the effects of competitive

aggregation in solution on adsorption have been elucidated by

comparing single cholesteryl with multicholesterol derivatives in

vesicles and in supported lipid bilayers.97

The construction of a lipid membrane/pseudohexagonal DNA

hybrid, anchored on the membrane through the same choles-

teryl-functionalization has also been reported. The effects of

grafting density, lipid/DNA ratio, liposome number density and

preparation procedure on the final structure and yield of the

resulting hybrid nanomaterial were demonstrated. This work

was the first example of the realization of nonlinear DNA

constructs onto fluid membranes.98 Remarkably, closely match-

ing results for the thickness of the DNA layer have been obtained

for DNA adsorption on liposomes and on SLB at the same

grafting density (Fig. 8).

Very recently, the possibility of creating Janus-like vesicles

with coexisting lipid domains where amphiphilic DNAmolecules

can preferentially partition has been shown. Lipophilic oligo-

nucleotide partition was obtained using tocopherol and choles-

terol-modified DNA.98–102 Upon heating the lateral domains

merge and reform, by cooling these sticky patches would break

the spherical symmetry of inter-vesicle interactions, opening up

the possibility of directional vesicle adhesion. However, exclusive

partitioning of a conjugate into the Lo phase, shown in Fig. 7,

has not been reported until recently.101 Again, inspiration from

Nature has been the elegant guiding principle to design this latter

nucleolipid. To force partitioning into the liquid-ordered phase,

palmitoylation of a DNA-recognizing PNA (peptide-nucleic

acid) has been accomplished in GUV (Giant Unilamellar Vesi-

cles). Palmitoylation of membrane proteins plays in fact in

Nature a decisive strategy to target them to raft domain. The

lateral domains are thermoreversible, as shown in Fig. 7, and

7156 | Soft Matter, 2011, 7, 7150–7158

partition can be thus thermally cycled. This approach offers

a reversible tool for triggering interactions among the structures

and for the arrangement of reactions and signalling cascades on

biomimetic surfaces. This powerful bio-inspired strategy shows

how it is possible to realize synthetic rafts with structural control,

possessing distinct oligonucleotide functionalization and there-

fore distinct and extremely selective coupling functions, and will

presumably have an important impact both in nano-

biotechnology and as a biomimetic system.

An interesting example of the use of oligonucleotide-decorated

vesicles in the hierarchical assembly of simpler units has been

reported recently. Layer-by-layer (LbL) particles were prepared

by the adsorption of polyelectrolyte layers on silica beads and the

outermost polyelectrolyte layer has been modified via covalent

coupling of dA21 oligonucleotides with an amino-linker.102

The outer polyelectrolyte layer provides a template for vesicle

binding via DNA hybridization, resulting in the coating with

a tunable arrangement of several layers of intact liposomes,

functionalized with thymine-based DNA oligonucleotides,

conjugated with two tocopherol moieties and incorporated into

lipid membranes.

This journal is ª The Royal Society of Chemistry 2011

Page 8: Self-assembly of designer biosurfactants

Publ

ishe

d on

31

May

201

1. D

ownl

oade

d by

Uni

vers

ity o

f N

ewca

stle

on

26/0

9/20

14 1

9:00

:30.

View Article Online

The disintegration of distinct vesicle layers can be achieved, for

example, by sequence-specific melting of nucleotide bases.

Further applications that can derive from this concept include

the entrapment of different reagents into different layers and

their regulated release on demand.

Conclusions and future perspectives

In this review we have examined the latest progresses in the field

of self-assembly of designer nucleolipids. Nucleolipids join the

self-assembly properties due to the presence of a hydrophobic

assembler with some biospecific functionalities, as those dis-

played by nucleic acids. These properties enrich both the self-

assembly scenario and the range of applications. Innovative

design principles can be applied both to the hydrophobic tail

architecture and to the structure, length and sequence of the

polar head.

From what is reported, it is clear that the field is truly inter-

disciplinary, as many other areas in soft matter. While novel

synthetic strategies enable more and more sophisticated molec-

ular repeat units to be prepared, the challenge will be the control

over the structural and dynamical properties of the self-assem-

blies, and therefore on their possible use to several new

biotechnological applications.

Acknowledgements

This work has been funded by CSGI and the Italian Ministry of

Research (PRIN 20087K9A2J001 and FIRB RBPR05JH2P

Italnanonet).

Notes and references

1 I. W. Hamley, Introduction to Soft Matter: Synthetic and BiologicalSelf-Assembling Materials, John Wiley & Sons, Chichester, UK,2007.

2 B. W. Ninham and P. Lo Nostro, Molecular Forces and SelfAssembly in Colloid, Nano Sciences and Biology, CambridgeUniversity Press, New York, 2010.

3 Y. S. Lee, Self-Assembly and Nanotechnology: a Force BalanceApproach, John Wiley & Sons, Hoboken, New Jersey, 2008.

4 O. G. Mouritsen, Life–As a Matter of Fat: the Emerging Science ofLipidomics, Springer-Verlag, Berlin, 200f5.

5 M. Ryadnov, Bionanodesign: Following Natures Touch, RSCPublishing, Cambridge, 2009.

6 P. Baglioni and D. Berti, Curr. Opin. Colloid Interface Sci., 2003, 8,55, and references therein.

7 D. Berti, Curr. Opin. Colloid Interface Sci., 2006, 11, 74.8 E. Kokkoli, A. Mardilovich, A. Wedekind, E. L. Rexeisen, A. Gargand J. A. Craig, Soft Matter, 2006, 2, 1015.

9 F. Peypoux, J. M. Bonmatin and J. Wallach, Appl. Microbiol.Biotechnol., 1999, 51, 553.

10 J. R. Lu, X. B. Zhao and M. Yaseen, Curr. Opin. Colloid InterfaceSci., 2007, 12, 60.

11 X. Zhao, F. Pan, H. Xu, M. Yaseen, H. Shan, C. A. E. Hauser,S. Zhang and J. R. Lu, Chem. Soc. Rev., 2010, 39, 3480.

12 H. Rosemeyer, Chem. Biodiversity, 2005, 2, 977.13 P. Barth�el�emy, C. R. Chim., 2009, 12, 17.14 C. B. Reese, Org. Biomol. Chem., 2005, 3, 3851.15 W. Saenger, Principles of Nucleic Acid Structure, Springer-Verlag

Inc, Boston, 1984.16 G. I. Neil, P. F. Wiley, R. C.Manak and T. E.Moxeley,Cancer Res.,

1970, 30, 1047.17 A. Gissot, M. Camplo, M. W. Grinstaff and P. Barth�el�emy, Org.

Biomol. Chem., 2008, 6, 1324.18 P. Poijarvi-Virta and H. Lonnberg, Curr. Med. Chem., 2006, 13,

3441.

This journal is ª The Royal Society of Chemistry 2011

19 L. W. Peterson and C. E. McKenna, Expert Opin. Drug Delivery,2009, 6, 405.

20 K. Isono, M. Uramoto, H. Kusakabe, K. Kimura, K. Isaki,C. C. Nelson and J. A. McCloskey, J. Antibiot., 1985, 38, 1617.

21 A. Takatsuki, K. Arima and G. Tamura, J. Antibiot., 1971, 24, 215.22 D. C. Hiss, G. A. Gabriels and P. I. Folb,Cancer Cell Int., 2007, 7, 5.23 A. M. Heacock and B. W. Agranoff, Biochim. Biophys. Acta, 1997,

1348, 166.24 M. MacCoss, E. K. Ryu and T. Matsushita, Biochem. Biophys. Res.

Commun., 1978, 85, 714.25 T.Matsushita, E. K. Ryu, C. I. Hong andM.MacCoss,Cancer Res.,

1981, 41, 2707.26 E. K. Ryu, R. J. Ross, T. Matsushita, M. MacCoss, C. I. Hong and

C. R. West, J. Med. Chem., 1982, 25, 1322.27 J. G. Turcotte, S. P. Srivastava, J. M. Steim, P. Calabresi,

L.M.Tibbetts andM.Y.Chu,Biochim.Biophys.Acta, 1980,619, 619.28 J. G. Turcotte, S. P. Srivastava, W. A. Meresak, B. A. Rizkalla,

F. Louzon and T. P. Wunz, Biochim. Biophys. Acta, 1980, 619, 604.29 C. I. Hong, A. J. Kirisits, A. Nechaev, D. J. Buchheit and

C. R. West, J. Med. Chem., 1985, 28, 171.30 Y. Ji Yun, S. M. Park and B. H. Kim, Chem. Commun., 2003, 254.31 S. Shuto, S. Ueda, S. Imamura, K. Fukukawa, A. Matsuda and

T. Ueda, Tetrahedron Lett., 1987, 28, 199.32 S. Shuto, S. Imamura, K. Fukukawa and T. Ueda, Chem. Pharm.

Bull., 1988, 36, 5020.33 S. Shuto, H. Ito, S. Ueda, S. Imamura, K. Fukukawa, M. Tsujino,

A. Matsuda and T. Ueda, Chem. Pharm. Bull., 1988, 36, 209.34 D. Berti, L. Franchi, P. Baglioni and P. L. L. Luisi, Langmuir, 1997,

13, 3441.35 D. Berti, P. Baglioni, G. Barsacchi-Bo, S. Bonaccio and

P. L. L. Luisi, J. Phys. Chem. B, 1998, 102, 303.36 D. Berti, F. Pini, P. Baglioni and J. Teixeira, J. Phys. Chem. B, 1999,

103, 1738.37 R. A. Schwendener, A. Supersaxo, W. Rubas, H. G. Weder,

H. R. Hartmann, H. Schott, A. Ziegler and H. Hengartner,Biochem. Biophys. Res. Commun., 1985, 126, 660.

38 L. Moreau, P. Barth�el�emy, M. El Maataoui and M. W. Grinstaff, J.Am. Chem. Soc., 2004, 126, 7533.

39 L. Moreau, N. Campins, M. W. Grinstaff and P. Barth�el�emy,Tetrahedron Lett., 2006, 47, 7117.

40 R. Iwaura, K. Yoshida, M. Masuda, K. Yase and T. Shimizu, Chem.Mater., 2002, 14, 3047.

41 R. Iwaura, K. Yoshida, M. Masuda, M. Ohnishi-Kameyama,M. Yoshida and T. Shimizu, Angew. Chem., 2003, 115, 1039.

42 P.Chabaud,M.Camplo,D. Payet,G. Serin,L.Moreau, P.Barth�el�emyandM. W. Grinstaff, Bioconjugate Chem., 2006, 17, 466.

43 L. Moreau, P. Barth�el�emy, Y. Li, D. Luo, C. A. Prata andM. W. Grinstaff, Mol. BioSyst., 2005, 1, 260.

44 S. Khiati, N. Pierre, S. Andriamanarivo, M. W. Grinstaff,N. Arazam, F. Nallet, L. Navailles and P. Barth�el�emy,Bioconjugate Chem., 2009, 20, 1765.

45 H. Rosemeyer, M. Ahlers, B. Schmidt and F. Seela, Angew. Chem.,Int. Ed. Engl., 1985, 24, 501.

46 C. J. Welch, A. Larsson, A. C. Ericson, B. Oberg, R. Datema andJ. Chattopadhyaya, Acta Chem. Scand., Ser. B, 1985, 39, 47.

47 M. Ahlers, H. Ringsdorf, H. Rosemeyer and F. Seela,Colloid Polym.Sci., 1990, 268, 132.

48 S. Ikonen, H. Macickova-Cahova, R. Pohl, M. Sanda andM. Hocek, Org. Biomol. Chem., 2010, 8, 1194.

49 B. M. Paterson, B. E. Roberts and E. L. Kuff, Proc. Natl. Acad. Sci.U. S. A., 1977, 74, 4370.

50 M. L. Stephenson and P. C. Zamecnik, Proc. Natl. Acad. Sci. U. S.A., 1978, 75, 285.

51 J. B. Opalinska and A. M. Gewirtz, Nat. Rev. Drug Discovery, 2002,1, 503.

52 C. Wolfrum, S. Shi, K. N. Jayaprakash, M. Jayaraman, G. Wang,R. K. Pandey, K. G. Rajeev, T. Nakayama, K. Charrise,E. M. Ndungo, T. Zimmermann, V. Koteliansky, M. Manoharanand M. Stoffel, Nat. Biotechnol., 2007, 25, 1149.

53 J. Soutschek, A. Akinc, B. Bramlage, K. Charisse, R. Constien,M. Donoghue, S. Elbashir, A. Geick, P. Hadwiger, J. Harborth,M. John, V. Kesavan, G. Lavine, R. K. Pandey, T. Racie,K. G. Rajeev, I. R€ohl, I. Toudjarska, G. Wang, S. Wuschko,D. Bumcrot, V. Koteliansky, S. Limmer, M. Manoharan andH. P. Vornlocher, Nature, 2004, 432, 173.

Soft Matter, 2011, 7, 7150–7158 | 7157

Page 9: Self-assembly of designer biosurfactants

Publ

ishe

d on

31

May

201

1. D

ownl

oade

d by

Uni

vers

ity o

f N

ewca

stle

on

26/0

9/20

14 1

9:00

:30.

View Article Online

54 Y. Singh, P. Murat and E. Defrancq, Chem. Soc. Rev., 2010, 39,2054.

55 R. L. Letsinger, G. Zhang, D. K. Sun, T. Ikeuchi and P. S. Sarin,Proc. Natl. Acad. Sci. U. S. A., 1989, 86, 6553.

56 O. Kaczmarek, N. Brodersen, A. Bunge, L. Loser, D. Huster,A. Herrmann, A. Arbuzova and J. Liebscher, Eur. J. Org. Chem.,2008, 1917.

57 H. Liu, Z. Zhu, H. Kang, Y. Wu, K. Sefan and W. Tan, Chem.–Eur.J., 2010, 16, 3791.

58 J. Israelachvili, Intermolecular and Surface Forces, Academic Press,London, 2nd edn, 1991.

59 D. F. Evans and H. Wennerstr€om, The Colloidal Domain: WherePhysics, Chemistry and Biology, and Technology Meet, Wiley-VCH, New York, 2nd edn, 1999.

60 K. Holmberg, B. J€onsson, B. Kronberg and B. Lindman, Surfactantsand Polymers in Aqueous Solution, John Wiley & Sons, Ltd, WestSussex, 2nd edn, 2003.

61 S. Bonaccio, M. Wessicken, D. Berti, P. Walde and P. L. L. Luisi,Langmuir, 1996, 12, 4976.

62 D. Berti, U. Keiderling and P. Baglioni, Prog. Colloid Polym. Sci.,2002, 120, 64.

63 M. Fortini, D. Berti, P. Baglioni and B. W. Ninham, Curr. Opin.Colloid Interface Sci., 2004, 9, 168.

64 F. Baldelli Bombelli, D. Berti, U. Keiderling and P. Baglioni, Appl.Phys. A: Mater. Sci. Process., 2002, 74, s1270.

65 F. Baldelli Bombelli, D. Berti, U. Keiderling and P. Baglioni, J.Phys. Chem. B, 2002, 106, 1161.

66 S. Murgia, S. Lampis, R. Angius, D. Berti and M. Monduzzi, J.Phys. Chem. B, 2009, 113, 9205.

67 F. Baldelli Bombelli, D. Berti, F. Pini, U. Keiderling and P. Baglioni,J. Phys. Chem. B, 2004, 108, 16427.

68 F. Baldelli Bombelli, D. Berti, M. Almgren, G. Karlsson andP. Baglioni, J. Phys. Chem. B, 2006, 110(35), 17627.

69 F. Betti, F. Baldelli Bombelli, D. Berti, M. Bonini, A. Brandt andP. Baglioni, J. Appl. Crystallogr., 2007, 40, s240.

70 F. Baldelli Bombelli, F. Betti, D. Berti, F. Pini, M. Heinrich andP. Baglioni, J. Phys.: Condens. Matter, 2008, 20, 104213.

71 F. Baldelli Bombelli, D. Berti, S. Milani, M. Lagi, P. Barbaro,G. Karlsson, A. Brandt and P. Baglioni, Soft Matter, 2008, 4, 1102.

72 D. Berti, L. Franchi, P. Baglioni and P. L. L. Luisi, Langmuir, 1997,13, 3438.

73 D. Berti, P. Barbaro, I. Bucci and P. Baglioni, J. Phys. Chem. B,1999, 103, 4916.

74 D. Berti, P. L. Luisi and P. Baglioni, Colloids Surf., A, 2000, 167, 95.75 D. Berti, F. Pini, P. Baglioni and J. Teixeira, Physica B

(Amsterdam), 2000, 276, 334.76 D. Berti, E. Fratini, S. Dante, T. Hauss and P. Baglioni, Appl. Phys.

A: Mater. Sci. Process., 2002, 74, s522.77 D. Berti, F. Baldelli Bombelli, M. Fortini and P. Baglioni, J. Phys.

Chem. B, 2007, 111(40), 11734.78 S. Milani, F. Baldelli Bombelli, D. Berti, S. Dante, T. Haub and

P. Baglioni, J. Phys.: Condens. Matter, 2008, 20, 104212.79 M. Banchelli, D. Berti and P. Baglioni,Angew. Chem., Int. Ed., 2007,

46, 3070–3073.

7158 | Soft Matter, 2011, 7, 7150–7158

80 S. Milani, F. Baldelli Bombelli, D. Berti, T. Hauß, S. Dante andP. Baglioni, Biophys. J., 2006, 90, 1260.

81 S. Milani, F. Baldelli Bombelli, D. Berti and P. Baglioni, J. Am.Chem. Soc., 2007, 129, 11664.

82 S. Milani, D. Berti, S. Dante, T. Haub and P. Baglioni, Langmuir,2009, 25(7), 4084.

83 A. Masotti, G. Mossa, C. Cametti, G. Ortaggi, A. Bianco,N. D. Grosso, D. Malizia and C. Esposito, Colloids Surf., B, 2008,68, 136.

84 X. Mulet, X. Gong, L. J. Waddington and C. J. Drummond, ACSNano, 2009, 3, 2789.

85 G. Godeau, J. Bernard, C. Staedel and P. Barth�el�emy, Chem.Commun., 2009, 5127.

86 G. Godeau and P. Barth�el�emy, Langmuir, 2009, 25(15), 8447.87 M. P. Thompson, M.-P. Chien, Ti-H. Ku, A. M. Rush and

N. C. Gianneschi, Nano Lett., 2010, 10, 2690.88 P. Pescador, N. Brodersen, H. A. Scheidt, M. Loew, G. Holland,

N. Bannert, J. Liebscher, A. Herrmann, D. Huster andA. Arbuzova, Chem. Commun., 2010, 46, 5358.

89 I. Pfeiffer and F. H€o€ok, J. Am. Chem. Soc., 2004, 126, 10224.90 C. Yoshina-Ishii, G. P. Miller, M. L. Kraft, E. T. Kool and

S. G. Boxer, J. Am. Chem. Soc., 2005, 127, 1356; J. J. Benkoskiand F. H€o€ok, J. Phys. Chem. B, 2005, 109, 9773; C. Yoshina-Ishii,Y.-H. M. Chan, J. M. Johnson, L. A. Kung, P. Lenz andS. G. Boxer, Langmuir, 2006, 22, 5682.

91 G. Stengel, R. Zahn and F. H€o€ok, J. Am. Chem. Soc., 2007, 129,9584.

92 G. Stengel, L. Simonsson, R. A. Campbell and F. H€o€ok, J. Phys.Chem. B, 2008, 112, 8264.

93 Y.-H. M. Chan, B. van Lengerich and S. G. Boxer, Proc. Natl. Acad.Sci. U. S. A., 2009, 106, 979.

94 M. Banchelli, F. Betti, D. Berti, G. Caminati, F. Baldelli Bombelli,T. Brown, L. M. Wilhelmsson, B. Nord�en and P. Baglioni, J.Phys. Chem. B, 2008, 112, 10942.

95 F. Gambinossi, M. Banchelli, A. Durand, D. Berti, T. Brown,G. Caminati and P. Baglioni, J. Phys. Chem. B, 2010, 114, 7338.

96 A. Kurz, A. Bunge, A. K. Windeck, M. Rost, W. Flasche,A. Arbuzova, D. Strohbach, S. M€uller, J. Liebscher, D. Husterand A. Herrmann, Angew. Chem., Int. Ed., 2006, 45, 4440.

97 M. Banchelli, F. Gambinossi, A. Durand, G. Caminati, T. Brown,D. Berti and P. Baglioni, J. Phys. Chem. B, 2010, 114, 7348.

98 F. Betti, F. Baldelli Bombelli, F. Gambinossi, G. Caminati,T. Brown, P. Baglioni and D. Berti, Soft Matter, 2009, 5, 1639.

99 A. Bunge, M. Loew, P. Pescador, A. Arbuzova, N. Brodersen,J. Kang, L. D€ahne, J. Liebscher, A. Herrmann, G. Stengel andD. Huster, J. Phys. Chem. B, 2009, 113, 16425.

100 P. A. Beales and T. K. Vanderlick, J. Phys. Chem. B, 2009, 113,13678.

101 M. Loew, R. Springer, S. Scolari, F. Altenbrunn, O. Seitz,J. Liebscher, D. Huster, A. Herrmann and A. Arbuzova, J. Am.Chem. Soc., 2010, 132, 16066.

102 M. Loew, J. Kang, L. Dahne, R. Hendus-Altenburger,O. Kaczmarek, J. Liebscher, D. Huster, K. Ludwig, C. Bottcher,A. Herrmann and A. Arbuzova, Small, 2009, 5, 320.

This journal is ª The Royal Society of Chemistry 2011


Recommended