You are on page 1of 18

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/305451717

Design and Applications of Protein-Cage-Based Nanomaterials

Article  in  Chemistry - An Asian Journal · July 2016


DOI: 10.1002/asia.201600769

CITATIONS READS

19 419

3 authors, including:

Yu Zhang Maziar S. Ardejani


Nanjing Forestry University The Scripps Research Institute
46 PUBLICATIONS   423 CITATIONS    16 PUBLICATIONS   119 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Enzyme View project

All content following this page was uploaded by Yu Zhang on 27 October 2017.

The user has requested enhancement of the downloaded file.


CHEMISTRY
AN ASIAN JOURNAL
www.chemasianj.org

Accepted Article

Title: Design and applications of protein cage-based nanomaterials

Authors: Yu Zhang; Maziar Soleymani Ardejani; Brendan Patrick Orner

This manuscript has been accepted after peer review and the authors have elected to
post their Accepted Article online prior to editing, proofing, and formal publication
of the final Version of Record (VoR). This work is currently citable by using the Di-
gital Object Identifier (DOI) given below. The VoR will be published online in Early
View as soon as possible and may be different to this Accepted Article as a result
of editing. Readers should obtain the VoR from the journal website shown below
when it is published to ensure accuracy of information. The authors are responsible
for the content of this Accepted Article.

To be cited as: Chem. Asian J. 10.1002/asia.201600769

Link to VoR: http://dx.doi.org/10.1002/asia.201600769

A Journal of A sister journal of Angewandte Chemie


and Chemistry – A European Journal
Chemistry - An Asian Journal 10.1002/asia.201600769

FOCUS REVIEW

Design and applications of protein cage-based nanomaterials


Yu Zhang [a]*, Maziar S. Ardejani [b] and Brendan P. Orner [c]*
Chemistry - An Asian Journal 10.1002/asia.201600769

FOCUS REVIEW

Abstract: Materials science is beginning to focus on reaction vessels with multiple potential opportunities for
biotemplation, and in support of that trend, it is realized that nanotechnology that have been extensively explored.[10] Protein
protein cages, proteins that assemble from multiple monomers cages vary in size from several to hundreds of nanometers and
into architectures with hollow interiors, can instill a number of in shape from spherical to nanotube-like structures, and these
unique advantages to nanomaterials. In addition, the structural characteristics can directly impact the properties of resulting
and functional plasticity of many protein cage systems permit templated materials.
their engineering for specific applications. In this review, the
most commonly used viral and non-viral protein cages, which
exhibit a wide diversity of size, functionality, and chemical and
thermal stabilities, are described. Moreover, how they have been
exploited for nanomaterial and nanotechnology applications is
summarized.

Introduction

The synthesis of materials with nanoscale structure has


drawn extensive attention due to these material’s emergent
properties and applications in physics and biotechnology. Taking
inspiration from naturally evolved processes, the exploitation of
biological templates is an emerging trend in the fabrication of
nanomaterials, and it has been reported that material size,
shape, and morphology can be controlled by interactions
between biomolecule and inorganic components.[1] Proteins and
Figure 1. Structural comparison of protein assemblies used to build nanoscale
peptides, due to their ready availability and large structural and
materials. All structures are shown to scale. The colors are added to highlight
functional diversity, especially have high utility for achieving
the morphological units, and are not necessarily indicative of different protein
control in the synthesis of materials.[2] Moreover, biotemplate-
sequences. All the structures are generated using UCSF Chimera.
directed syntheses have the potential to be more ‘green’ than
traditional methods due to the mild reaction conditions, such as
The shell of protein cages, usually 2 ~ 5 nm thick, separates
lower temperature, near neutral pH, and the fact that they often
the interior volume from the exterior and often contains selective
employ aqueous reaction solutions usually required for ensuring
or non-selective pores. Protein cages therefore have three
bio-molecule stability.
distinct surfaces: the inner surface that projects into the central
Many of the biotemplates that have been exploited for the
cavity, the outer surface that faces the exterior of the cage and
synthesis of nanomaterials are assemblies made up of
the inter-subunit surfaces that form the protein-protein interfaces
structurally simpler components. Nature uses self-assembly to
in the assembled cage.[11] (Figure 2) The interior cavities of
generate a wide diversity of large, complex, and often highly
these cages offer the potential to package and deliver novel
symmetric protein architectures with a minimum of synthetic
genetic material or to encapsulate other particles. Through either
remuneration,[3] and self-assembling protein systems form a
chemical modification or a genetic-based approach, each of
variety of supramolecular structures with a wide diversity of
these surfaces has potential plasticity to facilitate a protein cage
biological functions and properties. Moreover, nature provides
be the basis for the design of novel functional nanostructures.
many proteins that assemble into cage-like architectures,
In this review, we will outline the most commonly used viral
including virus,[4] bacteriophage,[5] vault,[6] heat shock,[7]
and non-viral protein cages, and how they have been recently
chaperonin[8] and ferritin proteins.[9] (Figure 1)
applied to the growing field of nanomaterials and
Protein cages are proteins that adopt structures enclosing a
nanotechnology with the focus primarily on the interior and
central volume, and they can act as robust, size-constrained
exterior surfaces.

[a] Dr. Y. Zhang


Jiangsu Key Lab of Biomass-based Green Fuels and Chemicals
College of Chemical Engineering
Nanjing Forestry University
Nanjing 210037 (P. R. China)
E-mail: yuzhang@njfu.edu.cn
[b] Dr. M. S. Ardejani
Department of Chemistry, The Scripps Research Institute,
La Jolla, CA 92037, United States
[c] Dr.B. P. Orner
Department of Chemistry, King’s College London
London SE1 1DB, United Kingdom
E-mail: brendan.orner@kcl.ac.uk
Chemistry - An Asian Journal 10.1002/asia.201600769

FOCUS REVIEW

unique, single-stranded RNA polymers inside the shell and the


highly negatively charged RNA is thought to interact strongly
with the positively charged interior capsid surface. The
generation of CCMV in vitro is straightforward, as the Pichia
pastoris heterologous expression system can be use to generate
capsomers that assemble spontaneously, and this has facilitated
the engineering of the cage. For example, complementary
interactions can be designed across the protein-protein
interfaces to generate a stabilized capsid.
(Interior) CCMV was the first icosahedral virus that was shown
to reassemble in vitro into an infectious particle.[17] The

Dr. Yu received her B.S (2004) from China


University of Mining and Technology and M.
S (2007) in Chemistry from Xiamen
University. She completed her PhD in 2012
from Nanyang Technological University under
Figure 2. Schematic representation of the three surfaces of protein cages, supervision of Dr. Brendan Orner. Now she is
each of which can be modified to impart function by design. Reproduced from an associate professor at Nanjing Forestry
reference [11]. Copyright 2007 WILEY-VCH Verlag. University. Her current research focuses on
the design and preparation of functional
nanomaterials based on protein assemblies.

1 Viruses and bacteriophage Dr. Maziar S. Ardejani graduated from Sharif


University of Technology with a B.Sc. in
Viral and bacteriophage capsids are among the major protein
chemistry in 2015. He receiced his M.Sc. and
cage classes and are the primary components of the most
Ph.D. from Nanyang Technological
abundant biological entities on earth.[12] With a variety of shapes
University. He has received postdoctoral
and sizes ranging from 20-750 nm, capsids are emerging training from King’s College London and
platforms for synthetic manipulation with a range of applications currenly is a research associate at The
from materials to medicine.[13] In this section, the viruses and Scripps Research Institute. His research
bacteriophages that are most widely used as biocontainers are revolves around the molecular self-assembly
introduced and their recent applications are described. and its applications.

1.1 Cowpea Chlorotic Mottle Virus

Icosahedral virus capsids are round proteinaceous particles Dr. Brendan Orner received his B. S. (1995)
with icosahedral symmetry in the arrangement of their from Haverford College where he conducted
assembled subunits (known as capsomers). A geometric research with Professor Terry Newirth. He
icosahedron with 532-point symmetry is comprised of twenty completed his M.S. in 1998 at the University
equilateral triangle facets fused into a closed shell around twelve of Pittsburgh and his PhD in 2001 at Yale
vertices.[14] Icosahedral capsids require at least sixty identical University both under the mentorship of ((Author Portrait))
subunits as each facet is generally composed of three Professor Andrew Hamilton. He conducted
capsomers. In 1962 Caspar and Klug proposed the concept of postdoctoral research at University of
quasi-equivalence to describe how larger viruses with Wisconsin with Professor Laura Kiessling.
icosahedral symmetry are built up with multiples of sixty He started his independent career as an
subunits.[15] Many icosahedral viruses can self-assemble into assistant professor at Nanyang Technological
non-infectious biocontainers devoid of genomic material either University in 2006 and moved his lab to the
naturally or as a consequence of genetic manipulation. UK in 2012. He currently is a senior lecturer
One icosahedral virus that has been frequently used in at King’s College London.
nanomaterials applications is cowpea chlorotic mottle virus
(CCMV) and this example will be used in this review to introduce highly positive interior surface is due to the presence of basic
common approaches that have been, or could easily be, applied residues (arginine and lysine) that interact with the negatively
across many protein cages. This virus is a member of the charged genetic RNA and these interactions are essential during
bromovirus group of the Biromoviriedae family.[16] Its capsid is the normal assembly process in vivo.
composed of 180 identical copies of the capsomer (20 kD) that Douglas and Young exploited the positively charged interior of
form an icosahedral shell with 28 nm and 24 nm outer and inner CCMV to direct the nucleation of polyoxometalate mineralization
diameters respectively. The CCMV genome contains three (tungstates H2W12O4210–, molybdates (NH4)xMoOy, and
Chemistry - An Asian Journal 10.1002/asia.201600769

FOCUS REVIEW

vanadates V10O286–) inside the empty cage.[18] They also Figure 3. The pH-dependent gating of CCMV and a capsid reassembly
reported that by altering the charge of the interior surface from strategy to afford guest cargo encapsulation. (A) Unswollen and swollen forms
positive to negative (by glutamic acid substitution of the of cowpea chloriotic mosaic virus (CCMV). The swollen form has sixty, 2 nm
internally projecting basic residues), the mineralization of metal pores that are pH-gated. (B) The capsid of CCMV can be engineered to
oxide particles (Fe2O3, Fe3O4, Co2O3) can be achieved without assemble around EGFP. Reprinted from reference [21,23]. Copyright 2009
disrupting cage assembly.[19] Furthermore, after some additional American Chemical Society.
protein engineering, CCMV has also been used as a biotemplate
for the potentiated reduction and directed synthesis of gold (Exterior) The exterior surface of icosahedral virus capsids
nanoparticles.[20] provides the opportunity for multivalent display of ligands in
A remarkable and highly useful property of CCMV is that its defined orientation with respect to each other and CCMV has
capsid can undergo pH and metal ion-dependent structural been employed as a robust platform in this direction. There are a
“gating”, which can be exploited to control the acquisition and total of 540 lysines and 560 carboxylates in wild type CCMV that
release of guest cargos.[16, 21] This gating involves an increase in have been demonstrated to be reactive with isothiocyanate or
size of up to 10% and the formation of sixty, 2 nm pores in the malonate presenting dyes respectively.[24] Although the native
capsid at pH above 6.5 or with the forced removal of divalent cysteines are not reactive toward chemical modification, reactive
metal ions.[21] (Figure 3A) The gating allows the protein to be thiol groups can be introduced through mutagenesis. These
loaded for some delivery applications without requiring conjugation approaches have been used to attach a wide variety
reassembly. of ligands to the exterior surface of CCMV, including small
In vitro reassembly can also be used to encapsulate larger peptides, biotin, fluorophores, photosensitizers, and even intact
cargo inside the CCMV capsid. For example, this approach was IgG antibodies.[24-25] In fact, Comellas-Aragones et al. were able
used to package the 40-kDa horseradish peroxidase (HRP) to incorporate two different synthetic polymers onto two different
enzyme within CCMV.[22] Once reassembled, pH dependent surfaces of the capsid. First polyethylene glycol (PEG) was
capsid gating influences access of substrate to the active, conjugated through an NHS ester and lysines on the virus
encapsulated enzyme and subsequent release of product. exterior. After disassembly and removal of viral RNA,
Engineered nanoreactors like these, especially if designed to reassembly of the conjugate was inhibited probably due to steric
incorporate multiple enzymes in a multi-step metabolic pathway, interactions between the PEG chains. However, addition of
could have great utility in small molecule synthesis and in aiding anonic polystyrene sulfonate (PSS) resulted in assembly
in the fundamental understanding of interactions between effectively generating a CCMV capsid encapsualting PSS and
enzymes and metabolites on single enzyme scales. presenting PEG on its exterior.[26] (Figure 4) It is conceivable that
Using a similar capsid reassembly strategy, Cornelissen et al. along with enhancing the generation of materials, multivalently
generated a construct consisting of multiple enhanced green presented ligands can be exploited for the directed delivery of
fluorescent proteins (EGFP) encapsulated within a CCMV encapsulated materials as part of biomedical applications or for
capsid by employing heterodimeric coiled-coil peptide fusions.[23] providing the geometric and chemically specific interactions that
Through genetic modification, the complementary coild-coil pair, would be required in the design of higher order nanostructure.
K-coil (KIAALKE)3 and E-coil (EIAALEK)3, were fused to the N-
terminus of the CCMV capsid protein and the C-terminus of
EGFP, respectively. Noncovalent heterodimerization of the two
coiled-coil elements brings the unassembled capsid proteins
together with the EGFP and adjusting the pH assembles the
capsid. Depending on the concentration and ratio of EGFP-
capsid protein conjugate and the wild type capsid protein, up to
fifteen EGFP proteins can be encapsulated per assembled
capsid. (Figure 3B) The two strategies of pore gatting and
reassembly have been implemented in other protein cage
systems for nanomaterials applications.

Figure 4. Schematic pathway toward a PSS-CCMV-PEG particle. The


polyethylene glycol (PEG) chains were attached through lysine residues on
the outer surface of the CCMV capsid. The fluorescein-functionalized PEG
chains induced irreversible dissociation of the virus due to steric interference
between the protein subunits and subsequent release of the viral RNA.
Incubation of the PEG–CCMV conjugate with the anionic polystyrene sulfonate
(PSS) resulted in formation of stability-enhanced reassembled PSS-CCMV-
PEG particles. Reprinted from reference [26]. Copyright 2009 American
Chemical Society.
Chemistry - An Asian Journal 10.1002/asia.201600769

FOCUS REVIEW

1.2 Cowpea Mosaic Virus extensively in the CPMV system. The Huisgen reaction (also
known as “click chemistry”), is a highly specific orthogonal
Cowpea mosaic virus (CPMV) is a plant virus of the method that allows single-site conjugation in large
comovirus group. CPMV capsids are icosahedral, 30 nm in biomacromolecules.[34] In this reaction, alkynes and azides react
diameter, and assembled from sixty large and sixty small subunit in a [3+2] cycloaddition to form a triazol with little cross reactivity
proteins. The small capsid proteins form the twelve, pentameric to other functional groups found in proteins and nucleic acids.
caps at the 5-fold symmetry axes, and the large capsid proteins This reaction has been developed to operate catalytically,[35] with
trimerize at the twenty, 3-fold axes of symmetry.[10a] The highly photoactivation[36] and with strained and electron-poor
symmetric and heteromeric nature of the capsid provides a alkynes.[37] To effectuate the Huisgen reaction on the CPMV
remarkable opportunity to differentially introduce multiple types exterior, lysine and cysteine residues have been modified into
of functionality with control over both the spatial distribution and either alkynes or azides via conventional conjugation strategies,
the degree of multivalency. With discrete shape and size, and a and various challenging substrates including complex sugars,
high degree of symmetry and polyvalency, plant viruses such as peptides, PEG and the iron carrier protein transferrin have been
CPMV, can provide versatility to a wide range of conjugated to the capsid surface.[31] Even hydrophobic fullerenes
bionanomaterials. Moreover, CPMV can remain intact at have been attached to capsids enhancing the fullerene water
temperatures up to 60 °C for an hour and at pH 4.0 or 9.0 at solubility and bioavailability in HeLa cells.[38] (Figure 5) Possible
room temperature for at least two days. With a variety of developments of these strategies for anti-tumor therapeutic
appealing properties, CPMV has been extensively investigated devices will be discussed below.
as a robust, monodisperse nano-building block has been
extensively investigated.
(Interior) Nucleic acid-free CCMV capsids can be obtained by
in vitro assembly after isolation from infected plant material,
however this method can be impeded by the fact that the
disassembled protein monomers are only soluble in the
presence of denaturants. To overcome this problem, Saunders Figure 5. The mutilvalent derivatization of CPMV and bacteriophage Qβ
and coworkers managed to obtain empty CPMV capsids in capsids with multiple fullerenes. The well characterized fullerene, PCBA ([6,6]-
abundance by co-expressing in plants the coat protein precursor phenyl-C61-butyric acid) was activated as the N-hydroxysuccinimate ester
along with the 24K proteinase. In this method, the proteinase (NHS) and coupled to solvent-exposed Lys residues on exterior of a virus
processes the coat protein precursor into large and small capsid (in this figure depicted as an icosahedron viewed down a C2 symmetry
proteins which can efficiently co-assemble into empty capsid.[27] axis). Reprinted from reference [38]. Copyright 2009 American Chemical
Such empty capsids can be used to encapsulate a wide range of Society.
cargo, such as metals, fluorescent dyes or drugs.[28] For
example, without forming protein conjugates, Aljabali et al. CPMV capsids have also been used as polyvalent platforms
succefully loaded cobalt metal and iron oxide using sodium for the presentation of full-length proteins. One strategy for doing
borohydide as the reductant within the interior of empty CPMV this began by genetically converting four of the five outer surface
capsid obtained through Sauders’ methodology.[28a] Steinmetz lysine residues in the monomer to arginine and introducing a
and coworkers systematically examined the capacity of empty cysteine residue on the surface of CPMV capsid. The remaining
CPMV particles to encapsulate a wide variety of molecules. lysine or the introduced cysteine was conjugated to various
Through targeting reactive cysteines, fluorophores, biotin affinity proteins, including T4 lysozyme, the LRR domain of internalin B
tags, poly(ethylene glycol) (PEG) and various peptides were and the Intron 8 gene product of the HER2 tyrosine kinase
selectively displayed on the interior surface.[29] The in vitro and receptor, activated as NHS esters.[39] In every example, the
in vivo properties of many of these protein conjugates were protein structure and biological activity was preserved, opening
investigated in tissue culture and a preclinical mouse model as the door for biomedical applications including vaccines and
the foundation for their developmemt in biomedical applications. antiviral therapeutics.
(Exterior) Through the genetic or synthetic introduction of (Applications) CPMV has been extensively investigated as a
chemical functional groups onto the protein subunits, the template for new materials, expecially in drug delivery and
derivatization of the CPMV capsid exterior has been adopted as medical imageing applications. For example, to overcome the
a strategy to utilize the virus capsid as a nanoscale building dose-limiting non-specific toxicities of Cr3+, which can selectively
block as part of larger, designed architectures.[30] By controlling inhibit the proliferation of high glucose-induced human aortic
the degree of multivalency and spatial distribution of conjugation smooth muscle cells (HASMC) in vitro, chromium chloride was
through carboxylate and ammonium groups in glutamate, loaded into the CPMV interior cavity, and the resulting particles
aspartate, and lysine side chains, well-established chemistry has significantly reduced glucose-induced HASMC proliferation and
been used to attach various dyes,[30a] polymers,[31] and presented anti-atherogenic effects under hyperglycemic
antigens,[32] and to crosslink the exterior of CPMV for various conditions.[40]
applications.[33] A number of approaches have been explored to direct CPMV
One capsid exterior conjugation strategy, due to its ubiquity toward in vivo applications. To understand the fate of CPMV
and power, is worth specific mention, and it has been adopted after intravenous injection, Manchester and coworkers tracked
Chemistry - An Asian Journal 10.1002/asia.201600769

FOCUS REVIEW

the fluorescently labeled virus in a murine model,[41] and the displaying specific biomineralization peptides, both the anatase
intact capsids were detected in a number of organs suggesting and rutile mineral forms of TiO2 could be generated.[54]
that CPMV could serve as a multi-tissue delivery vehicle. In this Furthermore, by utilizing metal-ligand coordination chemistry,
direction, some work has been undertaken to target CPMV various guest molecules such as diphan can be encapsulated to
specifically to tumor cells, and both the Finn and Manchester the interior of P22 capsid.[55] In this approach, cysteine residues
groups reported the chemical attachment of folic acid to the were genetically introduced onto the capsid interior surface,
capsid using the copper-catalyzed Huisgen reaction.[42] Not only followed by chemical modification with metal ligand-presenting
was folic acid-conjugated CPMV able to efficiently target the 5-iodoacetamido-1,10-phenanthroline, the coordination by
cell-surface folate receptors on cancer cells, the study also various metal complexes. By attaching small molecules to
showed that higher density loading of targeting ligands on exogenously introduced ligands for the metal complex, the
CPMV might not be necessary for efficient delivery. loading of cargo inside the P22 cage could be achieved. This
Imaging is another in vivo application toward which CPMV approach, performed under relatively mild conditions, could be
capsids have been steered. For example, fluorescent dyes were used as a versatile and general strategy for introducing a wide
displayed in a multivalent fashion on the outer surface of the variety of molecules into protein cage nanoparticles.
CPMV capsid, and these conjugates were used to image Taking advantage of the interaction between the coat and
vasculature and blood flow to a depth of up to 500 um in living scaffolding proteins, Douglas and coworkers packaged an
mouse and chick embryos.[43] Moreover, CPMV capsids have exogenous protein, alcohol dehydrogenase D, efficiently into the
also been used as magnetic resonance imaging (MRI) contrast P22 interior space.[56] (Figure 6) This research showed that
agents. Both the Finn and Manchester groups modified azide- encapsulation of AdhD inside P22 affected the enzyme’s kinetic
displaying viruses with an alkyne-modified DOTA parameters in comparison with the free enzyme, thereby
(tetraazacyclododecane tetraacetic acid) ligand via the Huisgen underlining the considerations involved in using modified protein
reaction.[44] After loading with gadolinium, the agent shows cages as catalytic reaction vessels. Similarly, cytochrome P450
increased T1 relaxivity relative to free Gd (DOTA) complexes in (CYP) was encapsulated inside the capsid and the enzyme’s
solution. These reagents, which were determined to be nontoxic stability towards protease degradation and acidic pH was
in vivo, were subsequently used for the MRI imaging of live mice. enhanced.[57] This work suggests the potential of these
[45]
biocatalytic virus-like particles as delivery vehicles for clinically
relvant enzymatic activity in biomedical applications.
1.3 Bacteriophage MS2 and P22

In analogy to the work done with eukaryotic viruses, phage


(viruses that infect bacteria) capsids have also been employed
as the basis for nanomaterials.[46] Both bacteriophage MS2,
which encapsulates single stranded RNA, and P22, which is a
double stranded DNA phage, have icosahedral capsids that
have been used for a variety of applications. Bacteriophage MS2
which infects E. coli, self-assembles from 180 copies of identical
monomers into a capsid that is 27 nm in diameter with thirty-two,
2 nm wide pores that provide access to the capsid’s interior Figure 6. Encapsulation of active alcohol dehydrogenase D (AdhD) inside
space.[47] It exhibits high stability in a wide range of temperatures, assembled P22 capsids. The co-assembly of bacteriophage coat protein (blue)
pHs, and organic solvents, suggesting that it could be easily and AdhD (red) is enabled through the fusion of a capsid scaffold protein
applied to materials and biomedical applications. [48] The (yellow) to the AdhD. Reprinted from reference [56]. Copyright 2012 American
Salmonella bacteriophage P22 assembles from 420 copies of a Chemical Society.
coat protein with the assistance from an approximately 300
amino acid residue scaffolding protein[49] which can be removed (Exterior) Modification of the exterior surface of
after assembly by heating to 65 °C and subsequent treatment bacteriophage particles makes it possible to control how these
with the protease thrombin.[50] Moreover, the fact that the protein assemblies interact with their environment. Francis and
structure of the P22 capsid is reversibly responsive to coworkers used periodate chemistry to attach sixty copies of
temperature,[51] suggests that it could act as a dynamic platform phenylene diamine-substituted nucleic acid aptamers, selective
for a number of applications. for cell-specific surface receptors, to the outside of genome-free
(Interior) P22 possesses a relatively large inner cavity that can MS2 capsids that were modified with aniline functional
be loaded with a wide variety of cargo directed to a diverse set groups.[52b] The cell delivery of proof of principle fluorescent dye
of applications.[52] For example, Douglas and co-workers was enhanced due to the multivalent effect. (Figure 7) Using a
employed P22 for the size constrained synthesis of Fe 2O3 similar concept, Ashley et al. reported the selective delivery of
nanoparticles by fusing a polyanionic peptide to the scaffolding chemically diverse therapeutic and imaging agents to human
protein to thereby conceptually mimic the charged interior cavity hepatocellular carcinoma (HCC) by using MS2 modified with a
of ferritin.[53] A similar strategy was also used to synthesize targeting peptide that binds to HCC cell surface receptors. [58]
photoactive, small diameter, TiO2 nanoparticles. By using fusions
Chemistry - An Asian Journal 10.1002/asia.201600769

FOCUS REVIEW

Decoration protein (Dec) has been used to demonstrate the was observed. [63] This work demonstrates the utility of viral
feasibility of presenting large proteins on the surface of P22 capsids as essential components of complex systems that have
capsid in a modular, fully folded, and functional manner.[59] The the potential to be used to understand fundamental physics.
successful assembly of the fusion resulted in a displayed protein
that maintained its function and a P22 capsid that was minimally 1.4 Tobacco Mosaic Virus
altered, highlighting the broad potential for similar presentation
strategies. Tobacco mosaic virus (TMV), also known as tobamovirus,
was the first virus particle to be isolated and has been studied
intensively as a model system for diverse investigations in the
materials and analytical sciences.[64] The TMV capsid is a 300
nm long rod with an 18 nm outer diameter. Its rod shape results
from a regular helical array of 2130 identical protein subunits
which encloses the single stranded RNA genome. [65] Due to its
Figure 7. Covalent loading of functional cargo at the interior surface of the unique anisotropic shape and remarkable stability, TMV has
MS2 bacteriophage capsid and exterior surface modification with cell-specific been used as the template of choice for protein-based rod
nucleic acid aptamers (depicted as red sqiggles). Reprinted from reference shaped materials synthesis.
[52b]. Copyright 2009 American Chemical Society. The capsid of TMV undergoes a reversible state change from
micron-length rods at pH 5.5 to double disc morphologies in
(Applications) Bacteriophage P22 capsids can also serve as phosphate buffer at pH 7.0.[66] Through chemical and genetic
nanocontainers for therapeutic applications. Kelly et al. modification of the inner and outer surfaces, the assembled
developed a peptide drug delivery system using P22 capsids have been used as templates to grow metal and metal
bacteriophage which is modified to serve as a tunable oxide nanowires, liquid crystals, and coated conductive
nanocontainer for the packaging and applied ring opening nanowires.
metathesis polymerization (ROMP) reaction to trigger (Interior) A variety of metals including Cu, Pt, Ag, Au, Ni, and
disassembly of the capsid and release of bioactive peptides Co have been deposited in the narrow cavity of the rod-shaped
under physiological conditions.[60] TMV particle.[67] In addition, metal alloy nanowires with lengths
Gd3+-based MR contrast agents have drawn strong interest in up to 100 nm and 4 nm diameter composed of CoPt, CoPt 3 and
recent years. Site selective initiation of atom transfer radical FePt3 have been synthesized inside TMV capsids. [68] However,
polymerization (ATRP) reactions has been recently established metal deposition on the inner surface of TMV can be difficult due
for high-density fabrication and delivery of Gd. The Douglas to weak initiation. To enhance nucleation, Kobayashi et al.
research group reported the use of ATRP to make addressable genetically modified the capsid to introduce the positively
polymer networks within the interior cavity of P22 capsid. A 2- charged amino acid lysine on the inner surface and were able to
aminoethyl methacrylate (AEMA) network was used as a generate 3 nm aligned Pt and Co nanoparticles inside the
scaffold for the attachment of Gd-based MRI contrast agents, virus.[69] Interestingly, control experiments with unmodified TMV
through reaction with the primary amine groups of the resulted in internalized nanowires instead of discrete
poly(AEMA). Substantial increase in the degree of labeling per nanoparticles, emphasizing the importance of appropriately
capsid was achieved compared to those of other virus-like presented interior surface charge.
particles.[61] Similarly, MS2 has also been applied as a scaffold In a similar vein, Wang et al. achieved site-specific nucleation
for the generation of MR contrast agents.[62] In this case, the of gold nanoparticles at sulfhydryl groups arrayed on the inner
protein cage was conjugated with Gd chelates on either its surface of a TMV nanotube generated by point mutation of a key
internal or external surfaces. Datta et al. labeled the MS2 with threonine to cysteine.[70] (Figure 8) The strong interaction
TREN-bis(HOPO)-terephthalamide ligand, either at the interior between the sulfhydryl group and gold secures the site-selective
of the capsid through tyrosine residues or on the exterior chemisorption, nucleation, and growth of Au seeds within the
through lysine residues, by functionalization with an aldehyde, genetically modified TMV nanotubes.
followed by conjugation with the ligand via oxime condensation.
The externally modified MS2 was measured to have an iron
relaxivity of 307 mM-1s-1, while that of the internally modified
MS2 was 416 mM-1s-1, a 5-fold increase compared to small
molecule Gd chelates. This result suggests that there are
significant advantages to using the internal surface capsids for
contrast agent attachment, which fortuitously leaves the exterior
surface available for tissue-targeting group modifications.
Recently, MS2 cages encapsulating a Au nanoparticle (AuNP) Figure 8. Schematic illustration of the site-specific biomineralization of gold
inside and bearing fluorescent dyes on the outer surface have nanostructures inside TMV protein nanotubes. The mutated cysteines arrayed
been fabricated for energy transfer studies. The dye-AuNP on the interior surface of T103C-TMV can specifically bind gold ions which
distance could be tuned with DNA linkers of different lengths, initiate growth of Au nanoparticle chains or Au nanorods in the confined cavity
and distance-dependent fluorescence intensity enhancement
Chemistry - An Asian Journal 10.1002/asia.201600769

FOCUS REVIEW

of the nanotubes. Reprinted from reference [70b]. Copyright 2009 American In the realm of analytical chemistry, Wang and coworkers
Chemical Society. used TMV as a template to develop a thin film sensor for the
detection of volatile organic compounds. [80] Oligoaniline, a small
(Exterior) The exterior surface of TMV has also been exploited molecule that is electroactive, has found application in
for the generation of nanomaterials. The structure of the coat electronics and sensing, and in this example, oligoaniline motifs
protein monomer reveals three potential regions for chemical were chemically conjugated to the outer surface of TMV capsids.
modification on the exterior surface of the assembled capsid, the Due to the high-density distribution of oligoaniline on the TMV
N-terminus, the C-terminus and the 63–66 loop region.[71] Harris surface and good processing ability of modified TMV, the virus
and coworkers inserted two cysteine residues at the TMV coat was readily fabricated into a thin film by directly drop coating
protein N-terminus and the resulting assembled capsid onto a glass substrate. Upon integration of the glass substrate
facilitated the ready deposition of Au, Ag, Pd, and Au-Pd into a prototypical device, the virus-based thin film exhibited
clusters onto the cysteine-modified template with enhanced good sensitivity and selectivity toward ethanol and methanol
density and stability in comparison to the wild-type TMV vapor.
template.[72] Similarly, the exterior exposed C-terminus has also
been utilized, by introducing His-tag metal-binding peptide, to
create protein nanorods that could bind gold nanoparticles 2. Non-viral protein cages
uniformly.[73] Demir et al. introduced a single lysine by point
mutation at the C-terminus, which can be selectively coupled to 2.1 Ferritins
small moieties such as FITC and biotin using NHS chemistry for
the production of a variety of chemospecific nano-tubular Ferritins, one of the most widely studied and utilized family of
materials.[74] cage proteins play an in vivo role in iron homeostasis and ferric
Chemical modification can enhance the performance of the oxide storage[81] and have highly conserved biochemical and
exterior of TMV to act as a template. Culver and coworkers structural properties. Structurally, mini-ferritins are formed from
investigated the coating of TMV with silica to both enhance twelve identical monomer subunits which assemble into ~ 9 nm
template stability and increase its affinity for metal ions, and outer diameter tetrahedrally symmetric cages with 5 nm inner
various metal nanoparticles including Au, Ag, Pt and Pd were diameter cavities that can store around 500 Fe atoms.
successfully deposited on the silica coated exterior surface of Conversely, maxi-ferritins are made up of twenty-four identical or
the TMV capsid.[75] similar monomers that form an octahedrally symmetric cage with
Other nanomaterials including silicon nanotubes, TiO2 and ~ 12 and 8 nm outer and inner diameters, respectively, and the
Al2O3 nanotubes, and Cu nanorods and nanowires have been maxi-ferritin cavity can typically accommodate up to ~ 4500 Fe
generated at different lengths ranging from 300 nm to microns atoms.
on the outer surface of wild type or modified TMV. [76] Such (Interior) Due to their small cavity and distinct symmetries,
biological-based materials may find applications in ferritins have been utilized as the basis of a wide variety of
nanoelectronics, sensing, or cancer therapy. inorganic materials with unique electronic, magnetic and optical
As a large array of unnatural amino acids can be incorporated, properties. In addition, these protein cages have been exploited
site specifically, into proteins by using a nonsense codon and as size-constrained reaction vessels to synthesize various
orthogonal tRNA-synthetase pairs, this methodology should nanoparticles including metals, oxides, hydroxides, carbonates
prove powerful when applied to TMV. Wu et al. developed an and semi-conductors[12, 82]. Therefore, multiple methods have
effective method for site-specific and high yield modification of been developed to mineralize nanoparticles using ferritins [83].
TMV coat protein using amber codon suppression and Huisgen Many of these strategies involved the engineering of the interior
reaction.[77] Biotin molecules were successfully conjugated on of the ferritin cavity with functionality that facilitates nanoparticle
the coat protein and the resulting TMV nanoparticles self- formation. For example, Kramer et al. fused a peptide sequence
assembled into large disk-like and normal rod-like structures to the internally projecting C-terminus of human L-chain ferritin.
under different conditions. This peptide has been shown to bind specifically to silver
(Applications) Through precise genetic and chemical nanoparticles.[84] Thus, the assembled protein cage was used for
modification at defined positions on its capsid, TMV can be the generation of silver nanocrystals with tight
directed toward multiple applications, most notably those polydispersities.[85] Embracing an alternative approach, Fan et al.
involving cell-targeted imaging, and the generation of reagents developed a two-step process to mineralize gold nanoparticles
for influencing immunology.[78] For example, Yin et al. produced inside native horse spleen apoferritin. [86] This strategy bypassed
TMV capsids conjugated to tumor-associated carbohydrate the necessity of engineering additional functional groups onto
antigens (TACAs) through Huisgen chemistry. [79] These particles the ferritin interior surface either through chemical or molecular
were able to elicit an immune response and the resulting high biology means and thus should be prove to be a strategy
titer IgGs were strongly reactive to the antigen when presented applicable to protein cages in general. [87]
on cancer cells. Interestingly, it was found that the position and (Exterior) The exterior surface of ferritins can be engineered
presentation of the immobilized antigen on the capsid had great to project functionality by modifying positions near the N-
influence on the strength of the resulting immune responses. terminus.[88] For example, Wang and coworkers
chemoselectively modified horse spleen apoferritin via acylation
Chemistry - An Asian Journal 10.1002/asia.201600769

FOCUS REVIEW

of N-terminal lysine residues followed by a series of other they could be used for specific imaging of cancer cells. The
conjugation reactions, including the Huisgen reaction. The ferritin itself provides selective targeting for cancer cells that
resulting conjugate was used to template ATRP to generate overexpress transferrin receptors and the magnetoferritin
ferritin shells grafted to oligo ethylene glycol methacrylate- nanoparticles catalyze the oxidation of peroxidase substrates in
polymers, a material that is soluble in dichloromethane. [89] the presence of H2O2 to produce a colored response that is used
The highly symmetric nature of protein cages is to visualize the cancer tissue.[98]
advantageous for the formation of isotropic hierarchal structures,
however, breaking this functional symmetry is essential for the 2.2 Small heat shock proteins
topological control required for applications relying on
anisotropic assembly or polarized multicomponent presentation. Small heat shock protein (sHsp), originally isolated from the
Using a masking/unmasking strategy on a solid support hyperthermophilic archeaon M. jannaschii, assembles into a
presenting two differentially activated thiol groups, Kang et al. protein cage with symmetry similar to that of maxiferritin. The
generated Janus-like miniferritins dual functionalized with both a cage of sHsp is made up of 24 monomer subunits and has a 12
fluorescent and an affinity label.[90] This process was initiated nm exterior diameter and 6.5 nm interior cavity[10c]. However,
through the genetic modification of exterior surface residues of diverging from the structure of maxiferritin, sHsp has large pores
Listeria innocua DNA binding protein from starved cells (LiDPS). (3 nm diameter), which facilitate exchange between inside and
Functional groups were differentiated spatially on the protein outside of the cage.[99]
cage exterior, and hierarchical toposelective assembly was Similar to other nanocage proteins, sHsp has been used as a
achieved through a layer-by-layer strategy. delivery vehicle for imaging and therapeutic agents. [100] For
(Applications) Because of ferritin’s native iron mineralization example, Uchida et al. engineered LyP-1, a nine-residue peptide
activity, these proteins have drawn considerable interest in which targets tumor-associated macrophages, onto the exterior
magnetic imaging applications. For example, human ferritins surface of sHsp, and this cage was used to deliver the imaging
have been employed as diagnostic MRI agents to detect agent Cy5.5 encapsulated inside the central cavity. The resulting
vascular macrophages and Uchida et al. engineered dual conjugate allowed visualization of macrophage-rich murine
functional human H-chain with genetically modified cell-specific carotid lesions by fluorescence imaging. [101] In a related example,
targeting peptides on the exterior surface and a synthetic iron an sHsp cage, genetically modified through the C-terminus to
oxide (magnetite) nanoparticle within the interior cavity. [91] display externally the neuropilin 1-
 binding peptide (iRGD),
Ferritin has also been well utilized for drug delivery. For which is known to interact with integrin and neuropilin-1
example, the anticancer drug cisplatin was encapsulated into the receptors on pancreatic cancer cells, and loaded internally with
horse spleen apoferritin cage and the cellular uptake of this the cytotoxic drug OSU03012, was shown to be selectively
species was found to be increased by four-fold over that of free cytoxic to pancreatic cancer cells.[102]
cisplatin presumably a result of enhanced endocytosis.[92] In The protein cage sHsp has also been directed toward hybrid
addition, the anticancer drug doxorubicin and a zinc- materials. For example, to generate artificial hydrogenase
phthalocyanine-based photosensitizer have both been delivered activity, Pt clusters were introduced into the interior cavity to act
from the interior of apoferritin cages with high success.[93] The as sites for the reduction of H+ to H2.[103] In another example
Yan group demonstrated that native H-ferritin nanocages could Douglas, Young, and coworkers synthesized a cross-linked
tumor-specifically deliver high doses of doxorubicin (DOX) branched dendritic polymer using the Huisgen reaction and
without the conjugation of a targeting ligand. Presumably, the initiation by cysteine residues within the interior cavity of
enhanced delivery was facilitated by the overexpression of sHsp.[104] Protein cages encapsulating the branched polymer
transferrin receptor 1 in tumor cells. maintain their native shape and size distribution but are
Because of their unique small size compared to other protein remarkably more stable. (Figure 9)
cages, the metallic nanoparticles generated inside ferritins are
expected to have a higher surface area to mass ratio and thus
there is much interest in applying them to catalysis. For example
Pd nanoclusters synthesized inside an apoferritin cage were
used as hydrogenation catalysts.[94] Building upon this, Au/Pd
core-shell and alloy nanoparticles were also prepared inside
ferritin resulting in material that delivered a two- to four-fold
increase in catalytic activity.[95]
Metal nanoparticles prepared in ferritin cages can mimic the
activities of enzymes, such as catalases and peroxidases, that
control in vivo oxidative stress.[96] Exhibiting higher peroxidase
activity than monometallic particles, bimetallic Fe-Pt
nanoparticles have been constructed, and the improved catalytic
activity may result from a synergic effect between Fe and Pt. [97]
Capitalizing on this concept, Yan and coworkers synthesized Figure 9. Fabrication of a dendritic polymer inside small heat shock protein
iron oxide nanoparticles inside ferritin and demonstrated that (sHsp). (A) A cartoon of a protein cage filled with a branched polymer that can
Chemistry - An Asian Journal 10.1002/asia.201600769

FOCUS REVIEW

be used to load drugs, imaging agents, or other functional components. (B) A


cutaway view of the HspG41C interior cavity highlighting exposed cysteine
residues (red). (C) Scheme for sequential synthesis of the dendritic polymer.
Generation numbers are indicated at the bottom. Reprinted from reference
[104]. Copyright 2009 American Chemical Society.

2.3 Chaperonins

Chaperonins are a class of hollow, cylindrical protein


assemblies that catalyze the native folding of other proteins.
Many chaperonins are double-ring structures with 14-18
subunits, depending on the organism of origin. Based on
sequence comparisons, chaperonins can be organized into two
groups with canonical representatives. Group I assembled from
14 subunits has structural homology to Escherichia coli GroEL,
whereas group II has 16 or 18 subunits and the widely Figure 10. Light- and magnetic field-switchable 1-D arrays of
considered examples are thermosomes and rosettasomes, superparamagnetic nanoparticles encased in chaperonin protein cages.
respectively, both of archaeal origin.[105] Composed of two Superparamagnetic nanoparticles (SNPs) were encapsulated by GroEL
stacked supramolecular protein rings, chaperonin proteins open chaperonins which are stitched together with Mg2+ cations bound through
and close through the binding and hydrolysis of ATP. As a merocynanine (MC) which is conjugated to the rim of the GroEL. Light
protein cage that operates uniquely as a conformational switch, photoisomerizes the MC so it changes conformation, releasing the cations and
the ATP-driven chaperonins have been utilized to conditionally dispersing the arrays. The arrays can also be reversibly formed into bundles
encapsulate and release drugs and semiconducting through the application of a magnetic field. Reprinted from reference [111].
nanoparticles.[106] Copyright 2015 American Chemical Society.
There has been much recent work to manipulate the
conditionality of chaperonins. For example, Kortemme and
colleagues demonstrated that group II chaperonins could be 3 Two- and three-dimentional supra-
designed to be light activated through the placement of assemblies of protein cages
photoswitchable azobenzene-based functionality at a key hinge
region. Photoisomerization of the azobenzene drives large-scale It is widely recognized that the next generation of materials
conformational changes of the protein assembly. [107] Recently, engineering will focus on the simultaneous manipulation of
derivatives of azobenzene that use different colors of light to structure across multiple length scales with a high degree of
achieve enhanced conversion have been developed, and these, specificity. For example, the two and three-dimensional
could provide improved photocontrol in similar chaperonin-based assembly of nanoparticles has drawn significant interest
systems.[108] In principle, the coupling of photoisomerization to because these higher order structures could exhibit collective
other cooperatively self-assembling proteins could lead to a wide behaviors and properties beyond those of the individual
variety of novel photocontrolled devices.[109] particles.[112] With regard to the use of biomolecules, much of the
The chaperonin GroEL has been recently developed into early focus in building up two- and three-dimensional structures
photoconfigurable nanotubes. First, six cysteine residues were has relied on DNA and peptides as building blocks and affinity
genetically inserted onto the rim of the barrel-shaped protein, elements.[113] However, folded proteins have been under-
and these were chemically conjugated with the protochromic exploited.[114]
ionic molecule merocyanin which can bind Mg2+ and Protein cages, as discussed in the earlier sections of this
photoisomerize into a non-polar spiropyran that does not bind review, have been employed in the generation of various
divalent cations. Thus, in the presence of Mg2+, the GroEL nanomaterials as size constrained reaction vessels and as
assembles into nanotubes, however, upon photoexcitation, platforms for functional design. However, the fabrication of
these tubes disassemble.[110] Placing superparamagnetic iron higher-order hierarchical structures from protein cages has
oxide nanoparticles (SNPs) inside the cavity of this engineered become an active research area only recently. Protein cages
chaperonin provides an additional level of structural control. possess many key features that make them desirable building
When the SNP-containing chaperonin nanotubes are subjected blocks for the engineering of hierarchical suprastructure. Among
to a 0.5 T magnetic field, they assemble laterally into thick these are: 1) Their size and structure are homogeneous; 2)
bundles. These bundles disperse into nanotubes upon removal Their central cavities provide the ability to transport and
of the field.[111] (Figure 10) sequester various cargo, and the variety in volume and shape of
this cavity across the family add to their attractiveness.
Moreover the cavities can act as excellent templates for the in
situ synthesis of nanoparticles or quantum dots; 3) A variety of
functionality can be incorporated onto the various surfaces of the
individual cages, either through bioconjugate chemistry or
Chemistry - An Asian Journal 10.1002/asia.201600769

FOCUS REVIEW

molecular biology, thereby enhancing their properties. Moreover, nanoscopic channels between the virus particles. The catalytic
protein cage-guided formation of nanoparticle supralattices precipitation of palladium can be achieved within these channels
provides a biocompatible platform that will facilitate the by soaking CPMV crystals in buffers containing Pd (II) and Pt (II)
development of advanced delivery and sensing applications in and relying on specific binding of the cations to basic residues
biological systems.[115] on the virus exterior surface. [118]
To construct two- and three-dimensional architectures from While a strategy to generate three-dimensional supra-
protein cage building blocks, two general strategies have been assembled lattices exploiting protein cage surface charge
developed. The first utilizes direct physical interaction, such as involves relatively straightforward design principles and provides
external surface electrostatics, between the protein cages, and a surprising degree of morphological control, a linker-focused
the second employs either covalent or non-covalent linker strategy contributes opportunities to optimize and tune additional
functionality.[112, 116] parameters, such as interaction strength, distance between
The Kostianien group has extensively utilized a supra- cages, specificity, and conformational flexibility.
assembly strategy based on surface electrostatics. For example,
lattices were assembled between positively charged gold
nanoparticles and protein cages that displayed surface negative
charge. It is possible to generate face centered cubic lattices if
the protein cage is CCMV with encapsulated RNA or simple
cubic lattices if the cage is ferritin filled with magnetic iron oxide
nanoparticles. It is thought that these materials will have
applications in delivery or imaging, respectively.[116a] (Figure 11)
In another example, the same lab explored lattices formed
between apoferritin protein cages displaying negative surface
charges and poly (amidoamine) dendrimers (PAMAM) which
present positively charged surfaces. It was found that the
morphology of the resulting lattices could be rationally tuned by
either altering the concentration of salt which shields the
Coulomibic attraction between the particles or the effective size
of the dendrimer with respect to that of the ferritin. [116b] In a third
example, body centered cubic lattices were assembled between Figure 12. Pre-and post-functionalization approaches of CCMV-avidin
negative-presenting CCMV cages and positively charged avidin crystals. Electrostatically assembled CCMV (acidic)/avidin (basic) co-crystals
proteins. Because CCMV presents its acidic residues in can be functionalized selectively with biotin-tagged functional units. Method1:
icosahedrally symmetric patches and avidin its basic patches Pre-functionalization of avidin with the biotinylated functional unit followed by
with tetrahedral symmetry, it was thought that the lattice addition of the virus particles. Method 2: Assembly of the crystals followed by
morphology is controlled by the geometric display of the charge, post-functionalization with the biotinylated agent. Reprinted from reference
thus adding an additional layer of control. Moreover, the [117]. Copyright 2014 American Chemical Society.
resulting material could be functionalized with compounds
conjugated with biotin, a specific and high affinity ligand for One linker-based strategy involves the “capsid decoration
avidin.[117] (Figure 12) protein” (Dec) from phage L which is know to specifically bind to
sites of three-fold symmetry on the P22 capsid. Using this
knowledge, Douglas and coworkers generated linkers of
dimerized Dec or of ferritin displaying multiple copies of Dec on
its external surface. These linkers were not only used to stich
together lattices of the P22 protein cage, but, along with wild-
type Dec, were employed as part of a strategy to build up
hierarchical protein cage materials layer-by-layer.[112]
Nucleic acids can introduce the robust logic of Watson-Crick
Figure 11. Cryo-TEM images of superlattices of CCMV with encapsulated interactions to highly specific linkers for the supra-assembly of
gold nanoparticles viewed along [100], [110] and [111] zone axes (scale bar protein cages. For example CPMV cages were first chemically
50 nm, inset is image Fourier transform). Reprinted from reference [116a]. conjugated to DNA through a lysine or cysteine. These
Copyright 2012 American Chemical Society. conjugates could assemble with cages displaying complimentary
DNA strands into supra-assembles whose stability was
Virus capsid-based, three-dimensional crystals provide a dependent on the degree of strand overlap and that could be
porous, highly organized scaffold around which an inorganic broken up through the addition of unconjugated, competing
matrix can be assembled. Work in this direction has been DNA.[119] Using a similar strategy, Fin, Park, and coworkers
performed in the solid phase to generate hybrid materials on a created a non-compact, diamond-type lattice made up of
larger scale. For example, it is known that the capsid of CPMV alternating DNA-presenting gold nanoparticles and
crystallizes into a body centered cubic morphology with large complementary DNA-functionalized Qβ phage capsids.
Chemistry - An Asian Journal 10.1002/asia.201600769

FOCUS REVIEW

Interestingly, the size of the nanoparticles could be varied Programs Project of Jiangsu Higher Education Institutions
without changing the lattice morphology if the DNA linker length (TAPP), a Marie Curie CIG (PCIG13-GA-2013-618538), and
was changed accordingly.[120] The use of nucleic acids as linkers BPO’s personal salary.
to build up hierarchical structures based on proteins cages has
the additional advantage that it can be coupled to DNA origami Keywords: protein cages • viruses • protein design •
structures which can then be employed to scaffold the cages nanoparticles • nanomaterials
with precise nanoscale positioning.[121] For example, origami tiles
presenting oligonucleotide strands have been used to arrange
References
MS2 phage capsids chemically conjugated with complimentary
single stranded DNA. These tiles can also be stapled together [1] M. B. Dickerson, K. H. Sandhage, R. R. Naik, Chem. Rev. 2008, 108,
into arrays with nanoscale structural control.[122] 4935-4978.
Another linker strategy to generate protein cage arrays has [2] M. S. Ardejani, B. P. Orner, Science 2013, 340, 561-562.
[3] Y. Zhang, B. P. Orner, Int. J. Mol. Sci. 2011, 12, 5406-5421.
relied on metal coordination complexes. For example a [4] a) A. Zlotnick, J. Mol. Biol. 1994, 241, 59-67; b) F. H. Crick, J. D.
thermophillic mini-ferritin that was decorated with phenathroline Watson, Nature 1956, 177, 473-475.
[5] R. Golmohammadi, K. Valegård, K. Fridborg, L. Liljas, J. Mol. Biol. 1993,
ligands could form amorphous assemblies in the presence of 234, 620-639.
iron (II). [123] [6] W. Berger, E. Steiner, M. Grusch, L. Elbling, M. Micksche, Cell. Mol.
Branched dendric polymers[124] can act as multivalent linkers Life Sci. 2009, 66, 43-61.
[7] J. D. Trent, FEMS Microbiol. Rev. 1996, 18, 249-258.
to stitch together assembles of protein cages. For example [8] M. J. Ellis, S. Knapp, P. J. B. Koeck, Z. Fakoor-Biniaz, R. Ladenstein, H.
polycationic dendrimers were used to “glue” together CCMV Hebert, J. Struct. Biol. 1998, 123, 30-36.
[9] a) R. A. Grant, D. J. Filman, S. E. Finkel, R. Kolter, J. M. Hogle, Nat.
capsids presenting that present anionic surface charges. Struct. Biol. 1998, 5, 294-303; b) G. Zhao, J. Biol. Chem. 2002, 277,
Because photolytically degradable dendrons were employed, the 27689-27696.
virus particles could be released from the arrays by treatment [10] a) T. Lin, Z. Chen, R. Usha, C. V. Stauffacher, J.-B. Dai, T. Schmidt, J.
E. Johnson, Virology 1999, 265, 20-34; b) R. W. Lucas, S. B. Larson, A.
with light. A very similar strategy generated light dependent, face McPherson, J. Mol. Biol. 2002, 317, 95-108; c) K. K. Kim, R. Kim, S.-H.
centered cubic lattices of ferritin. Interestingly if the ferritin Kim, Nature 1998, 394, 595-599; d) D. M. Lawson, P. J. Artymiuk, S. J.
Yewdall, J. M. Smith, J. C. Livingstone, A. Treffry, A. Luzzago, S. Levi,
cages were loaded with iron oxide magnetic nanoparticles, the P. Arosio, G. Cesareni, C. D. Thomas,W. V. Shaw,P. M. Harrison,
magnetic properties of the material could be influenced by the Nature 1991, 349, 541-544.
supra-assembly state of the ferritin cages.[125] The ability to tune [11] M. Uchida, M. T. Klem, M. Allen, P. Suci, M. Flenniken, E. Gillitzer, Z.
Varpness, L. O. Liepold, M. Young, T. Douglas, Adv. Mater. 2007, 19,
magnetic properties of nanoparticles by assembling them into 1025-1042.
ordered superstructure and to reverse the assembly process by [12] N. L. Rosi, C. A. Mirkin, Chem. Rev. 2005, 105, 1547-1562.
external stimuli will surely facilitate the development of a variety [13] a) K. J. Koudelka, A. S. Pitek, M. Manchester, N. F. Steinmetz, Annu.
Rev. Virol. 2015, 2, 379-401; b) A. M. Wen, N. F. Steinmetz, Chem. Soc.
of novel practical applications. Rev. 2016; c) B. Cao, M. Yang, C. Mao, Acc. Chem. Res. 2016, 49,
1111-1120.
[14] Y. Wu, H. Yang, H.-J. Shin, Korean J. Chem. Eng. 2013, 30, 1359-1367.
[15] D. L. Caspar, A. Klug, Cold Spring Harb. Symp. Quant. Biol. 1962, 27,
Conclusion 1-24.
[16] J. A. Speir, S. Munshi, G. Wang, T. S. Baker, J. E. Johnson, Structure
1995, 3, 63-78.
This review concisely summarized the viral and non-viral [17] a) J. M. Fox, G. Wang, J. A. Speir, N. H. Olson, J. E. Johnson, T. S.
protein cages commonly employed as templates for the Baker, M. J. Young, Virology 1998, 244, 212-218; b) S. Brumfield, D.
Willits, L. Tang, J. E. Johnson, T. Douglas, M. Young, J. Gen. Virology
generation of nanomaterials and described how these materials 2004, 85, 1049-1053.
have been directed toward multiple applications. The [18] T. Douglas, M. Young, Nature 1998, 393, 152-155.
[19] T. Douglas, E. Strable, D. Willits, A. Aitouchen, M. Libera, M. Young,
exploitation of new bio-based templates and novel pathways for Adv. Mater. 2002, 14, 415-418.
nanomaterial synthesis has drawn tremendous attention over [20] J. M. Slocik, R. R. Naik, M. O. Stone, D. W. Wright, J. Mater. Chem.
the past decades. The use of protein cage architectures of viral 2005, 15, 749-753.
[21] F. Tama, C. L. Brooks, 3rd, J. Mol. Biol. 2002, 318, 733-747.
and non-viral origin provides a number of unique advantages. [22] M. Comellas-Aragones, H. Engelkamp, V. I. Claessen, N. A.
They are biological in origin and thus amenable to genetic Sommerdijk, A. E. Rowan, P. C. Christianen, J. C. Maan, B. J. Verduin,
J. J. Cornelissen, R. J. Nolte, Nat. Nanotechnol. 2007, 2, 635-639.
manipulation and large-scale production. Furthermore, they can [23] I. J. Minten, L. J. Hendriks, R. J. Nolte, J. J. Cornelissen, J. Am. Chem.
be incorporated into nanostructured architectures with functions Soc. 2009, 131, 17771-17773.
not available by conventional material synthesis. Thus bio- [24] E. Gillitzer, D. Willits, M. Young, T. Douglas, Chem. Commun. 2002,
2390-2391.
conjugated protein cage architectures can facilitate various [25] a) P. A. Suci, Z. Varpness, E. Gillitzer, T. Douglas, M. Young, Langmuir
biomedical applications such as imaging and the delivery of bio- 2007, 23, 12280-12286; b) P. A. Suci, D. L. Berglund, L. Liepold, S.
Brumfield, B. Pitts, W. Davison, L. Oltrogge, K. O. Hoyt, S. Codd, P. S.
active molecules. Stewart, M. Young, T. Douglas, Chem. Biol. 2007, 14, 387-398.
[26] M. Comellas-Aragones, A. de la Escosura, A. J. Dirks, A. van der Ham,
A. Fuste-Cune, J. J. Cornelissen, R. J. Nolte, Biomacromolecules 2009,
10, 3141-3147.
Acknowledgements [27] K. Saunders, F. Sainsbury, G. P. Lomonossoff, Virology 2009, 393,
329-337.
[28] a) A. A. Aljabali, F. Sainsbury, G. P. Lomonossoff, D. J. Evans, Small
Financial support from National Natural Science Fundation of 2010, 6, 818-821; b) R. N. Seetharam, A. S. Blum, C. M. Soto, J. L.
China (No. 31200564), Natural Science Foundation of Jiangsu Whitley, K. E. Sapsford, A. Chatterji, T. Lin, J. E. Johnson, C. Guerra, P.
Satir, Nanotechnology 2008, 19, 105504.
Higher Education of China (16KJB180009), Top-notch Academic
Chemistry - An Asian Journal 10.1002/asia.201600769

FOCUS REVIEW

[29] A. M. Wen, S. Shukla, P. Saxena, A. A. Aljabali, I. Yildiz, S. Dey, J. E. [61] J. Lucon, S. Qazi, M. Uchida, G. J. Bedwell, B. LaFrance, P. E.
Mealy, A. C. Yang, D. J. Evans, G. P. Lomonossoff, Prevelige Jr, T. Douglas, Nat. Chem. 2012, 4, 781-788.
Biomacromolecules 2012, 13, 3990-4001. [62] A. Datta, J. M. Hooker, M. Botta, M. B. Francis, S. Aime, K. N.
[30] a) N. F. Steinmetz, G. P. Lomonossoff, D. J. Evans, Langmuir 2006, 22, Raymond, J. Am. Chem. Soc. 2008, 130, 2546-2552.
3488-3490; b) Q. Wang, T. Lin, L. Tang, J. E. Johnson, M. Finn, Angew. [63] S. L. Capehart, M. P. Coyle, J. E. Glasgow, M. B. Francis, J. Am. Chem.
Chem. Int. Ed. Engl. 2002, 41, 459-462; c) Q. Wang, E. Kaltgrad, T. Lin, Soc. 2013, 135, 3011-3016.
J. E. Johnson, M. Finn, Chem. Biol. 2002, 9, 805-811. [64] a) W. M. Stanley, Science 1935, 81, 644-645; b) N. C. Santos, M. A.
[31] S. S. Gupta, J. Kuzelka, P. Singh, W. G. Lewis, M. Manchester, M. Finn, Castanho, Biophys. J. 1996, 71, 1641-1650; c) K.-B. G. Scholthof, J. G.
Bioconjugate Chem. 2005, 16, 1572-1579. Shaw, M. Zaitlin, APS Press, 1999.
[32] A. Miermont, H. Barnhill, E. Strable, X. Lu, K. A. Wall, Q. Wang, M. e. G. [65] A. Klug, Philos. Trans. R. Soc., B 1999, 354, 531-535.
Finn, X. Huang, Chem. -Eur. J. 2008, 14, 4939-4947. [66] P. J. Butler, A. C. Bloomer, J. T. Finch, J. Mol. Biol. 1992, 224, 381-394.
[33] N. F. Steinmetz, D. J. Evans, Org. Biomol. Chem. 2007, 5, 2891-2902. [67] a) S. Balci, A. Bittner, K. Hahn, C. Scheu, M. Knez, A. Kadri, C. Wege,
[34] a) . K. ha, K. L. ooley, . . ystr m, D. . Burke, . . Kade, C. . H. Jeske, K. Kern, Electrochim. Acta 2006, 51, 6251-6257; b) E.
Hawker, Chem. Rev. 2009, 109, 5620-5686; b) M. D. Best, Dujardin, C. Peet, G. Stubbs, J. N. Culver, S. Mann, Nano Lett. 2003, 3,
Biochemistry 2009, 48, 6571-6584. 413-417; c) M. Knez, A. M. Bittner, F. Boes, C. Wege, H. Jeske, E.
[35] H. C. Kolb, M. Finn, K. B. Sharpless, Angew. Chem. Int. Ed. Engl. 2001, aiβ, K. Kern, Nano Lett. 2003, 3, 1079-1082; d) N. Poudyal, G. S.
40, 2004-2021. Chaubey, V. Nandwana, C.-b. Rong, K. Yano, J. P. Liu,
[36] J. Wang, W. Zhang, W. Song, Y. Wang, Z. Yu, J. Li, M. Wu, L. Wang, J. Nanotechnology 2008, 19, 355601.
Zang, Q. Lin, J. Am. Chem. Soc. 2010, 132, 14812-14818. [68] R. Tsukamoto, M. Muraoka, M. Seki, H. Tabata, I. Yamashita, Chem.
[37] a) N. J. Agard, J. A. Prescher, C. R. Bertozzi, J. Am. Chem. Soc. 2004, Mater. 2007, 19, 2389-2391.
126, 15046-15047; b) J. Dommerholt, O. van Rooijen, A. Borrmann, C. [69] M. Kobayashi, M. Seki, H. Tabata, Y. Watanabe, I. Yamashita, Nano
F. Guerra, F. M. Bickelhaupt, F. L. van Delft, Nat. Commun. 2014, 5, Lett. 2010, 10, 773-776.
5378. [70] a) K. Zhou, F. Li, G. Dai, C. Meng, Q. Wang, Biomacromolecules 2013,
[38] N. F. Steinmetz, V. Hong, E. D. Spoerke, P. Lu, K. Breitenkamp, M. 14, 2593-2600; b) K. Zhou, J. Zhang, Q. Wang, Small 2015, 11, 2505-
Finn, M. Manchester, J. Am. Chem. Soc. 2009, 131, 17093-17095. 2509.
[39] A. Chatterji, W. Ochoa, L. Shamieh, S. P. Salakian, S. M. Wong, G. [71] K. Namba, R. Pattanayek, G. Stubbs, J. Mol. Biol. 1989, 208, 307-325.
Clinton, P. Ghosh, T. Lin, J. E. Johnson, Bioconjugate Chem. 2004, 15, [72] S. Y. Lee, E. Royston, J. N. Culver, M. T. Harris, Nanotechnology 2005,
807-813. 16, S435.
[40] R. Ganguly, A. M. Wen, A. B. Myer, T. Czech, S. Sahu, N. F. Steinmetz, [73] . nęk, . Ł. Górzny, . ard, C. älti, . Davies, . Brydson, S.
P. Raman, Nanoscale 2016, 8, 6542-6554. Evans, P. Stockley, Nanotechnology 2013, 24, 025605.
[41] C. S. Rae, W. Khor, Q. Wang, G. Destito, M. J. Gonzalez, P. Singh, D. [74] M. Demir, M. H. Stowell, Nanotechnology 2002, 13, 541.
M. Thomas, M. N. Estrada, E. Powell, M. Finn, Virology 2005, 343, 224- [75] E. S. Royston, A. D. Brown, M. T. Harris, J. N. Culver, J. Colloid. Interf.
235. Sci. 2009, 332, 402-407.
[42] G. Destito, R. Yeh, C. S. Rae, M. Finn, M. Manchester, Chem. Biol. [76] a) V. A. Fonoberov, A. A. Balandin, Nano Lett. 2005, 5, 1920-1923; b)
2007, 14, 1152-1162. M. Knez, A. Kadri, C. Wege, U. Gösele, H. Jeske, K. Nielsch, Nano Lett.
[43] J. D. Lewis, G. Destito, A. Zijlstra, M. J. Gonzalez, J. P. Quigley, M. 2006, 6, 1172-1177; c) J. C. Zhou, C. M. Soto, M.-S. Chen, M. A.
Manchester, H. Stuhlmann, Nat. Med. 2006, 12, 354-360. Bruckman, M. H. Moore, E. Barry, B. R. Ratna, P. E. Pehrsson, B. R.
[44] D. E. Prasuhn Jr, R. M. Yeh, A. Obenaus, M. Manchester, M. Finn, Spies, T. S. Confer, J. Nanobiotech. 2012, 10, 1-12.
Chem. Commun. 2007, 12, 1269-1271. [77] F. Wu, H. Zhang, Q. Zhou, M. Wu, Z. Ballard, Y. Tian, J. Wang, Z. Niu,
[45] P. Singh, D. Prasuhn, R. M. Yeh, G. Destito, C. S. Rae, K. Osborn, M. Y. Huang, Chem. Commun. 2014, 50, 4007-4009.
Finn, M. Manchester, J. Control. Release 2007, 120, 41-50. [78] M. A. Bruckman, K. Jiang, E. J. Simpson, L. N. Randolph, L. G. Luyt, X.
[46] J. W. Oh, W. J. Chung, K. Heo, H. E. Jin, B. Y. Lee, E. Wang, C. Yu, N. F. Steinmetz, Nano Lett. 2014, 14, 1551-1558.
Zueger, W. Wong, J. Meyer, C. Kim, Nat. Commun. 2014, 5: 3043. [79] Z. Yin, H. G. Nguyen, S. Chowdhury, P. Bentley, M. A. Bruckman, A.
[47] K. Valegård, L. Liljas, K. Fridborg, T. Unge, Nature 1990, 345, 36-41. Miermont, J. C. Gildersleeve, Q. Wang, X. Huang, Bioconjugate Chem.
[48] J. E. Glasgow, S. L. Capehart, M. B. Francis, D. Tullman-Ercek, ACS 2012, 23, 1694-1703.
Nano 2012, 6, 8658-8664. [80] M. A. Bruckman, J. Liu, G. Koley, Y. Li, B. Benicewicz, Z. Niu, Q. Wang,
[49] M. H. Parker, S. Casjens, P. E. Prevelige, J. Mol. Biol. 1998, 281, 69-79. J. Mater. Chem. 2010, 20, 5715-5719.
[50] A. O'Neil, C. Reichhardt, B. Johnson, P. E. Prevelige, T. Douglas, [81] K. Honarmand Ebrahimi, P.-L. Hagedoorn, W. R. Hagen, Chem. Rev.
Angew. Chem. Int. Ed. Engl. 2011, 50, 7425-7428. 2014, 115, 295-326.
[51] C. M. Teschke, A. McGough, P. A. Thuman-Commike, Biophys. J. 2003, [82] a) K. Iwahori, I. Yamashita, Nanotechnology 2008, 19, 495601; b) R.
84, 2585-2592. Tsukamoto, K. Iwahori, M. Muraoka, I. Yamashita, Bull. Chem. Soc. Jpn.
[52] a) J. M. Hooker, A. Datta, M. Botta, K. N. Raymond, M. B. Francis, 2005, 78, 2075-2081; c) R. Tsukamoto, M. Muraoka, Y. Fukushige, H.
Nano Lett. 2007, 7, 2207-2210; b) G. J. Tong, S. C. Hsiao, Z. M. Nakagawa, T. Kawaguchi, Y. Nakatsuji, I. Yamashita, Bull. Chem. Soc.
Carrico, M. B. Francis, J. Am. Chem. Soc. 2009, 131, 11174-11178; c) Jpn. 2008, 81, 1669-1674; d) T. Douglas, V. T. Stark, Inorg. Chem.
W. Wu, S. C. Hsiao, Z. M. Carrico, M. B. Francis, Angew. Chem. Int. Ed. 2000, 39, 1828-1830; e) M. T. Klem, J. Mosolf, M. Young, T. Douglas,
Engl. 2009, 48, 9493-9497. Inorg. Chem. 2008, 47, 2237-2239; f) M. Li, C. Viravaidya, S. Mann,
[53] C. Reichhardt, M. Uchida, A. O'Neil, R. Li, P. E. Prevelige, T. Douglas, Small 2007, 3, 1477-1481; g) B. Hennequin, L. Turyanska, T. Ben, A. M.
Chem. Commun. 2011, 47, 6326-6328. Beltran, S. I. Molina, M. Li, S. Mann, A. Patane, N. R. Thomas, Adv.
[54] G. J. Bedwell, Z. Zhou, M. Uchida, T. Douglas, A. Gupta, P. E. Mater. 2008, 20, 3592-3596.
Prevelige, Biomacromolecules 2014, 16, 214-218. [83] J. C. Cheung-Lau, D. Liu, K. W. Pulsipher, W. Liu, I. J. Dmochowski, J.
[55] M. Uchida, D. S. Morris, S. Kang, C. C. Jolley, J. Lucon, L. O. Liepold, Inorg. Biochem. 2014, 130, 59-68.
B. LaFrance, P. E. Prevelige Jr, T. Douglas, Langmuir 2011, 28, 1998- [84] R. R. Naik, S. J. Stringer, G. Agarwal, S. E. Jones, M. O. Stone, Nat.
2006. Mater. 2002, 1, 169-172.
[56] a) D. P. Patterson, P. E. Prevelige, T. Douglas, ACS Nano 2012, 6, [85] R. M. Kramer, C. Li, D. C. Carter, M. O. Stone, R. R. Naik, J. Am. Chem.
5000-5009; b) D. P. Patterson, B. Schwarz, R. S. Waters, T. Gedeon, T. Soc. 2004, 126, 13282-13286.
Douglas, ACS Chem. Biol. 2013, 9, 359-365; c) D. P. Patterson, B. [86] R. Fan, S. W. Chew, V. V. Cheong, B. P. Orner, Small 2010, 6, 1483-
Schwarz, K. El-Boubbou, J. van der Oost, P. E. Prevelige, T. Douglas, 1487.
Soft Matter 2012, 8, 10158-10166. [87] C. A. Butts, J. Swift, S.-g. Kang, L. Di Costanzo, D. W. Christianson, J.
[57] L. Sánchez-Sánchez, A. Tapia-Moreno, K. Juarez-Moreno, D. P. G. Saven, I. J. Dmochowski, Biochemistry 2008, 47, 12729-12739.
Patterson, R. D. Cadena-Nava, T. Douglas, R. Vazquez-Duhalt, J. [88] a) K. I. Sano, K. Ajima, K. Iwahori, M. Yudasaka, S. Iijima, I. Yamashita,
Nanobiotech. 2015, 13, 66. K. Shiba, Small 2005, 1, 826-832; b) A. Jääskeläinen, R.-R. Harinen, T.
[58] C. E. Ashley, E. C. Carnes, G. K. Phillips, P. N. Durfee, M. D. Buley, C. Soukka, U. Lamminmäki, T. Korpimäki, M. Virta, Anal. Chem. 2008, 80,
A. Lino, D. P. Padilla, B. Phillips, M. B. Carter, C. L. Willman, ACS 583-587.
Nano 2011, 5, 5729-5745. [89] Q. Zeng, T. Li, B. Cash, S. Li, F. Xie, Q. Wang, Chem. Commun. 2007,
[59] B. Schwarz, P. Madden, J. Avera, B. Gordon, K. Larson, H. M. 1453-1455.
Miettinen, M. Uchida, B. LaFrance, G. Basu, A. Rynda-Apple, ACS [90] S. Kang, P. A. Suci, C. C. Broomell, K. Iwahori, M. Kobayashi, I.
Nano 2015, 9, 9134-9147. Yamashita, M. Young, T. Douglas, Nano Lett. 2009, 9, 2360-2366.
[60] P. Kelly, P. Anand, A. Uvaydov, S. Chakravartula, C. Sherpa, E. Pires, [91] M. Uchida, M. L. Flenniken, M. Allen, D. A. Willits, B. E. Crowley, S.
. O’ eil, T. Douglas, . Holford, Int. J. Environ. Res. Public Health. Brumfield, A. F. Willis, L. Jackiw, M. Jutila, M. J. Young, J. Am. Chem.
2015, 12, 12543-12555. Soc. 2006, 128, 16626-16633.
Chemistry - An Asian Journal 10.1002/asia.201600769

FOCUS REVIEW

[92] a) Z. Yang, X. Wang, H. Diao, J. Zhang, H. Li, H. Sun, Z. Guo, Chem.


Commun. 2007, 3453-3455; b) R. Xing, X. Wang, C. Zhang, Y. Zhang, [107] D. Hoersch, S.-H. Roh, W. Chiu, T. Kortemme, Nat. Nanotech. 2013, 8,
Q. Wang, Z. Yang, Z. Guo, J. Inorg. Biochem. 2009, 103, 1039-1044; c) 928-932.
E. Fantechi, C. Innocenti, M. Zanardelli, M. Fittipaldi, E. Falvo, M. [108] S. Samanta, A. A. Beharry, O. Sadovski, T. M. McCormick, A.
Carbo, V. Shullani, L. Di Cesare Mannelli, C. Ghelardini, A. M. Ferretti, Babalhavaeji, V. Tropepe, G. A. Woolley, J. Am. Chem. Soc. 2013, 135,
ACS Nano 2014, 8, 4705-4719; d) E. Falvo, E. Tremante, R. Fraioli, C. 9777-9784.
Leonetti, C. Zamparelli, A. Boffi, V. Morea, P. Ceci, P. Giacomini, [109] G. A. Woolley, Nat. Nanotech. 2013, 8, 892-893.
Nanoscale 2013, 5, 12278-12285. [110] a) S. Biswas, K. Kinbara, N. Oya, N. Ishii, H. Taguchi, T. Aida, J. Am.
[93] a) Z. Zhen, W. Tang, C. Guo, H. Chen, X. Lin, G. Liu, B. Fei, X. Chen, B. Chem. Soc. 2009, 131, 7556-7557; b) T. Sendai, S. Biswas, T. Aida, J.
Xu, J. Xie, ACS Nano 2013, 7, 6988-6996; b) M. Liang, K. Fan, M. Zhou, Am. Chem. Soc. 2013, 135, 11509-11512.
D. Duan, J. Zheng, D. Yang, J. Feng, X. Yan, Proc. Natl. Acad. Sci. U. [111] S. Sim, D. Miyajima, T. Niwa, H. Taguchi, T. Aida, J. Am. Chem. Soc.
S. A. 2014, 111, 14900-14905. 2015, 137, 4658-4661.
[94] T. Ueno, M. Suzuki, T. Goto, T. Matsumoto, K. Nagayama, Y. [112] M. Uchida, B. LaFrance, C. C. Broomell, P. E. Prevelige, T. Douglas,
Watanabe, Angew. Chem., Int. Ed. 2004, 116, 2581-2584. Small 2015, 11, 1562-1570.
[95] M. Suzuki, M. Abe, T. Ueno, S. Abe, T. Goto, Y. Toda, T. Akita, Y. [113] a) S. M. Douglas, H. Dietz, T. Liedl, B. Högberg, F. Graf, W. M. Shih,
Yamada, Y. Watanabe, Chem. Commun. 2009, 4871-4873. Nature 2009, 459, 414-418; b) R. J. Macfarlane, B. Lee, M. R. Jones, N.
[96] a) B. Maity, K. Fujita, T. Ueno, Curr. Opin. Chem. Biol. 2015, 25, 88-97; Harris, G. C. Schatz, C. A. Mirkin, Science 2011, 334, 204-208; c) G.
b) L. hang, L. Laug, . unchgesang, . ippel, . G sele, . Colombo, P. Soto, E. Gazit, Trends Biotechnol. 2007, 25, 211-218.
Brandsch, M. Knez, Nano Lett. 2009, 10, 219-223. [114] J. C. Sinclair, K. M. Davies, C. Vénien-Bryan, M. E. Noble, Nat.
[97] W. Zhang, X. Liu, D. Walsh, S. Yao, Y. Kou, D. Ma, Small 2012, 8, Nanotech. 2011, 6, 558-562.
2948-2953. [115] R. A. McMillan, C. D. Paavola, J. Howard, S. L. Chan, N. J. Zaluzec, J.
[98] K. Fan, C. Cao, Y. Pan, D. Lu, D. Yang, J. Feng, L. Song, M. Liang, X. D. Trent, Nat. Mater. 2002, 1, 247-252.
Yan, Nat. Nanotechnol. 2012, 7, 459-464. [116] a) M. A. Kostiainen, P. Hiekkataipale, A. Laiho, V. Lemieux, J.
[99] a) J. M. Galloway, S. S. Staniland, J. Mater. Chem. 2012, 22, 12423- Seitsonen, J. Ruokolainen, P. Ceci, Nat. Nanotech. 2013, 8, 52-56; b) V.
12434; b) M. T. Klem, D. Willits, D. J. Solis, A. M. Belcher, M. Young, T. Liljeström, J. Seitsonen, M. A. Kostiainen, ACS Nano 2015; c) J. D.
Douglas, Adv. Funct. Mater. 2005, 15, 1489-1494. Brodin, X. Ambroggio, C. Tang, K. N. Parent, T. S. Baker, F. A. Tezcan,
[100] a) M. Murata, S. Narahara, K. Umezaki, R. Toita, S. Tabata, J. S. Piao, Nat. Chem. 2012, 4, 375-382.
K. Abe, J.-H. Kang, K. Ohuchida, L. Cui, Int. J. Nanomed. 2012, 7, [117] V. Liljeström, J. Mikkilä, M. A. Kostiainen, Nat. Commun. 2014, 5, 4445.
4353; b) J. ShuáPiao, Chem. Commun. 2013, 49, 7442-7444; c) R. [118] J. C. Falkner, M. E. Turner, J. K. Bosworth, T. J. Trentler, J. E. Johnson,
Toita, M. Murata, K. Abe, S. Narahara, J. S. Piao, J.-H. Kang, K. T. Lin, V. L. Colvin, J. Am. Chem. Soc. 2005, 127, 5274-5275.
Ohuchida, M. Hashizume, Int. J. Nanomed. 2013, 8, 1989; d) R. Toita, [119] E. Strable, J. E. Johnson, M. Finn, Nano Lett. 2004, 4, 1385-1389.
M. Murata, S. Tabata, K. Abe, S. Narahara, J. S. Piao, J.-H. Kang, M. [120] P. Cigler, A. K. Lytton-Jean, D. G. Anderson, M. Finn, S. Y. Park, Nat.
Hashizume, Bioconjugate Chem. 2012, 23, 1494-1501. Mater. 2010, 9, 918-922.
[101] M. Uchida, H. Kosuge, M. Terashima, D. A. Willits, L. O. Liepold, M. J. [121] P. W. Rothemund, Nature 2006, 440, 297-302.
Young, M. V. McConnell, T. Douglas, ACS Nano 2011, 5, 2493-2502. [122] N. Stephanopoulos, M. Liu, G. J. Tong, Z. Li, Y. Liu, H. Yan, M. B.
[102] M. Murata, S. Narahara, T. Kawano, N. Hamano, J. S. Piao, J.-H. Kang, Francis, Nano Lett. 2010, 10, 2714-2720.
K. Ohuchida, T. Murakami, M. Hashizume, Mol. Pharmaceutics 2015, [123] L. Cristiano, J. SkováPedersen, Soft Matter 2010, 6, 3167-3171.
12, 1422-1430. [124] C. C. Lee, J. A. MacKay, J. M. Fréchet, F. C. Szoka, Nat. Biotech. 2005,
[103] Z. Varpness, J. Peters, M. Young, T. Douglas, Nano Lett. 2005, 5, 23, 1517-1526.
2306-2309. [125] a) M. A. Kostiainen, O. Kasyutich, J. J. Cornelissen, R. J. Nolte, Nat.
[104] M. J. Abedin, L. Liepold, P. Suci, M. Young, T. Douglas, J. Am. Chem. Chem. 2010, 2, 394-399; b) M. A. Kostiainen, P. Ceci, M. Fornara, P.
Soc. 2009, 131, 4346-4354. Hiekkataipale, O. Kasyutich, R. J. Nolte, J. J. Cornelissen, R. D.
[105] J. D. Trent, H. K. Kagawa, T. Yaoi, E. Olle, N. J. Zaluzec, Proc. Natl. Desautels, J. van Lierop, ACS Nano 2011, 5, 6394-6402.
Acad. Sci. U. S. A. 1997, 94, 5383-5388.
[106] D. Ishii, K. Kinbara, Y. Ishida, N. Ishii, M. Okochi, M. Yohda, T. Aida,
Nature 2003, 423, 628-632.
Chemistry - An Asian Journal 10.1002/asia.201600769

FOCUS REVIEW
Entry for the Table of Contents

FOCUS REVIEW
Protein cages, assembling from Yu Zhang*, Maziar S. Ardejani and
multiple monomers into architechtures Brendan P. Orner*
with hollow interiors, can instill a
number of unique advantages to Page No. – Page No.
nanomaterials. In this review, the most
Design and Applications of Protein
commonly used viral and non-viral
Cage-Based Nanomaterials
protein cages that exhibit a wide
diversity of size, functionality, and
chemical and thermal properties are
described. Moreover, how they have
been expoited for nanomaterial and
nanotechnology applications is
summarized.
Chemistry - An Asian Journal 10.1002/asia.201600769

FOCUS REVIEW

View publication stats

You might also like