Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Feature Article

Recombinant Spider Silk Proteins for


Applications in Biomaterials

Kristina Spiess, Andreas Lammel, Thomas Scheibel*

Due to their extraordinary mechanical and biochemical properties, silks have long been in
focus of research. In vivo, fibers are formed from silk proteins, in vitro, however, a variety of
materials can be produced in addition to fibers including capsules, particles, films, foams, and
gels. The versatility of silk proteins, along with their
biocompatibility, biodegradability, and potential for
processing in aqueous solution under ambient con-
ditions make silk-based materials good candidates
for biomedical applications such as drug delivery sys-
tems and scaffolds for tissue engineering. Here, we
summarize recent progress in research employing
recombinantly produced engineered spider silk
proteins with a focus on the fundamentals of silk
protein processing. We highlight recombinant spider
silk films and particles as morphologies that represent
model systems with adjustable material properties
controlled by process parameters.

Introduction prey, yielding outstanding materials with high tensile


strength combined with high extensibility.[1–3] In contrast
Silk from the silkworm Bombyx mori has been used to to silkworms, breeding spiders and harvesting their silk has
produce textiles for thousands of years due to its lustre, light been limited due to their cannibalistic nature. Through
weight, and tear resistance. During the past few decades, advances in biotechnology it is now possible to produce
spider silks have also aroused increasing interest in spider silk proteins recombinantly as well as to engineer
material science due to some extraordinary material silk genes and therefore proteins with properties tailored
properties. During 400 million years of evolution, spider for specific requirements. Among the different types of
silk has been optimized for several tasks such as catching spider silks, dragline silk, which composes the frame and
radii of the web, is the most extensively studied and the best
characterized.[4–8] In general, dragline silk is mainly
K. Spiess, T. Scheibel composed of two types of protein.[7] In the case of silk
Lehrstuhl Biomaterialien, Fakultät Angewandte from the spider Araneus diadematus the main proteins are
Naturwissenschaften, Universität Bayreuth Universitätsstr. 30,
ADF3 (A. diadematus fibroin) (which is relatively hydro-
95440 Bayreuth, Germany
philic) and ADF4 (which is relatively hydrophobic)
Fax: (þ49) 921 55 7346;
E-mail: thomas.scheibel@bm.uni-bayreuth.de
(Figure 1A).[7] The outstanding properties of spider silk
A. Lammel fibers can be explained by the hierarchical and sophisti-
Department Chemie, Technische Universität München, Lehrstuhl cated composition of structural elements of these two
Biotechnologie, Lichtenbergstraße 4, D-85747 Garching, Germany proteins with crystalline regions of different dimensions
a
These authors made equivalent contributions to this manuscript being embedded in an amorphous or pre-oriented matrix.[9]

Macromol. Biosci. 2010, 10, 998–1007


998 ß 2010 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim wileyonlinelibrary.com DOI: 10.1002/mabi.201000071
Recombinant Spider Silk Proteins for Applications in . . .

The amorphous matrix is likely to be responsible for the Kristina Spiess studied biology at the University
of Regensburg (Germany), where she received
extensibility of dragline silk, while crystalline regions are
her Diploma in 2005. After five months working
responsible for their strength.[10,11] The extent and type of
on an industrial collaboration project in the
structural features depend on the amino acid sequence of
group of Thomas Scheibel at the Technische
the underlying proteins (primary structure). Figure 1B Universität München (TUM, Germany) she
provides an overview of typical silk motifs of dragline silk started her PhD in his research group and moved
proteins, which have been identified as being responsible with him to the University of Bayreuth
for specific structure formation. Polyalanine motifs align to (Germany), where she is currently finishing her
form b-crystallites (tightly packed b-sheets) whereas PhD studies. She is an associate member of the
polyglycine motifs form primarily amorphous regions. International Graduate School of Science and
Our approach of recombinant silk protein production Engineering at TUM. Her research focuses on
(reviewed in ref.[12]) is based on multimerization of amino characterization and functionalization of recom-
binant spider silk proteins and their processing
acid consensus motifs (Modules A, Q, and C as illustrated in
into different morphologies for medical appli-
Figure 1C) derived from the repetitive part of the natural
cations.
proteins ADF3 and ADF4 leading to the engineered proteins
Andreas Lammel studied mechanical engineer-
eADF3 and eADF4.
ing in the field of medical and microtechnology
In the following sections we will outline how unraveling
at Technische Universität München (Germany)
the natural assembly process has given rise to the and chemical engineering at Rice University in
development of various new processing techniques yield- Houston (Texas). He is a fellow of the Inter-
ing novel silk morphologies. We will further describe how national Graduate School of Science and Engin-
material properties can be adjusted and controlled by eering at TUM as well as of the international
variation of process parameters using exemplarily the doctorate program Material Science of Complex
morphologies of silk particles and films. Interfaces within the Elite Network of Bavaria.
Since 2007 he has worked on his PhD under the
supervision of Thomas Scheibel. His research
focuses on the development of silk protein based
The Natural Silk Assembly Process
drug delivery systems.
Thomas Scheibel is full professor and chair of
The formation of silk fibers is a sophisticated process which
biomaterials at the Universität Bayreuth in
involves a series of tightly controlled parameters.[1] In
Germany. He received both his Diploma of bio-
nature, silk proteins are produced and excreted by
chemistry and a Dr. rer. nat. from the Universität
specialized cells lining the epithelium of a gland; in case Regensburg and his habilitation from the Tech-
of dragline silks this is the major ampullate gland (Figure 2). nische Universität München in Germany. He was
The proteins are stored as the so-called dope in the glands’ a Kemper Foundation postdoctoral fellow and a
lumen at remarkably high concentrations (up to 50 wt.-%) DFG postdoctoral fellow at the University of
without aggregation, and assemble on their way through Chicago. He gained several awards including
the spinning duct into stable and tough fibers which are the Biomimetics award of the German Bundes-
then released to the exterior (reviewed in ref.[1]). This ministerium für Bildung and Forschung (BMBF)
assembly process is characterized by a liquid-solid phase in 2006, and the ‘‘Innovation by nature award’’
of the BMBF in 2007. He is one of ten recipients
transition accompanied by structural changes of the
of the 2006 innovation tribute of the Bavarian
proteins; in the ampulla the core domains of the proteins
prime minister, received the Heinz-Maier-Leib-
are thought to possess no defined secondary structure, but
nitz Medal in 2007, and the Karl-Heinz-Beckurts
the proteins are most likely to be arranged in micelle-like Award in 2008.
assemblies, resulting from their amphiphilic nature.[13,14]
The final fiber, however, is rich in b-sheet crystallites
aligned parallel to the long axis of the fiber. Several (‘‘salting-in’’) ions sodium and chloride are exchanged by
biochemical and physical factors promote the structural the more kosmotropic (‘‘salting-out’’) ions phosphate and
transition. A slight acidification of the spinning dope potassium. In the next step, water is resorbed from the
(from pH 7.2 in the gland to pH 6.3) most likely leads to a solution at the distal part of the duct by endothelial cells,
neutralization of abundant acidic side chains, allowing increasing the protein concentration, which additionally
tighter interactions of the proteins.[15,16] Recently, the favors intermolecular interactions. Finally, the viscous
aminoterminus of the proteins has been identified as pH- solution is subjected to increasing elongational flow and
sensitive regulator of self-assembly.[17] Hydrophobic inter- shear forces, which are thought to further align the
molecular interactions are further enhanced by a change in molecules.[13,18] This finding is supported by the fact that
the ionic composition inside the duct: the more chaotropic higher flow rates lead to better aligned fibers.[19] In vitro

Macromol. Biosci. 2010, 10, 998–1007


ß 2010 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.mbs-journal.de 999
K. Spiess, A. Lammel, T. Scheibel

aqueous solution is the induction of


phase separation. The two key para-
meters involved in phase separation are
the protein concentration in solution and
the method with which desolvation of
the protein is triggered. The triggers can
be chemical such as pH and the amount/
type of additives (salts, e.g., potassium
phosphate, non-solvents, e.g., methanol,
polymers, e.g., poly(ethylene glycol), etc.)
or physical conditions like mechanical
shear and temperature.[6,7,21,23–26]
To visualize the influence of chemical
and physical parameters, Figure 3A
shows a qualitative ternary phase
diagram at constant temperature and
pressure. Each corner of the triangle
represents 100% of the respective com-
Figure 1. Model of the structure of a spider dragline silk. (A) A. diadematus spider
dragline silk consists mainly of two proteins, ADF3 and ADF4. On the molecular level,
ponent: silk protein, solvent (e.g., formic
[27]
spider silk exhibits crystalline regions of different dimensions embedded in an amor- acid, hexafluoroisopropanol,[28] ionic
[29]
phous matrix (figure adapted from ref. [9]). (B) Typical silk motifs and their correspond- liquids, and water[23,30]) and non-
[11]
ing putative secondary structure (adapted from ref. ). (C) Engineered spider silk solvent (e.g., methanol,[6] toluene,[31]
modules adopted from ADF3 and ADF4 for recombinant production (figure adapted etc.). Any point along the side of the
from ref. [7]).
triangle represents the composition of
the binary mixtures constituted by the
similar results were obtained: after addition of potassium two components at the corresponding corners. Therefore, a
phosphate (500 mM) to eADF3 spherical aggregates were state inside the phase diagram can be expressed by the silk
formed.[20,21] When accompanied by a pH shift and concentration and the solvent quality expressed by the
simultaneous elongational flow—applied by the geometry ratio of solvent to non-solvent.
of the employed microfluidic setup—eADF3 formed The Spinodal represents the set of conditions denoting
fibers.[13,21,22] the boundary of absolute instability of a solution where
small fluctuations in composition lead to a spontaneous
phase separation via spinodal decomposition. The region
Processing into Various Morphologies between the Spinodal and Binodal is the metastable region
where the Binodal denotes the boundary condition at
As described above, the fundamental principal that is which two distinct phases may coexist. As can be seen in the
crucial for the processing of materials from silk proteins in phase diagram, depending on the initial protein concentra-
tion, a different decrease in solvent quality is necessary to
cross the Spinodal to obtain a new stable state. This state
can be in form of a gel or particles. A gel is formed if the
concentration is high enough to build a continuous network
by which the liquid component is immobilized.[32] If the
concentration is below a certain threshold, particles are
formed by nucleation and growth.[18,20] A film can be
obtained by simple evaporation of the solvent. If porogens
are introduced into the silk protein solution and the solvent
is evaporated, porous structures or foams can be produced.
For fiber formation, phase separation has to take place in
combination with mechanical shearing and elongation.[21]
Figure 2. Natural assembly of spider silk proteins into fibers. As outlined above, depending on the processing route,
Dragline silk proteins are stored in the spinning gland. Upon
phase transition of spider silk proteins is controlled by
passage of the spinning dope through the spinning duct, the
proteins are subjected to a change of physico-chemical chemical and physical process parameters leading to
parameters leading to a liquid-solid phase transition necessary different materials morphologies. The characteristics of
for fiber formation (figure adapted from ref. [1]). the different morphologies along with their material

Macromol. Biosci. 2010, 10, 998–1007


1000 ß 2010 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim DOI: 10.1002/mabi.201000071
Recombinant Spider Silk Proteins for Applications in . . .

Figure 3. Phase behavior and morphologies of spider silk assemblies. (A) Qualitative ternary phase diagram of silk protein-solvent-non-
solvent system (figure adapted from ref. [94]). (B) Morphologies useful for potential applications as drug carriers. (C) Morphologies useful for
potential applications as scaffolds in tissue engineering.

properties show potential for different biomedical applica- of either eADF4(C16) or potassium phosphate
tions. For example, silk particles and microcapsules are (Figure 4A).[46] A detailed investigation regarding the
envisioned as drug carriers (Figure 3B), whereas silk films, applicability of eADF4(C16) particles employing 12 differ-
foams, and non-wovens could be used as scaffolds in tissue ent model drugs revealed that spider silk particle loading
engineering (Figure 3C). and release follows the mechanism depicted in Figure 4D
In the following, we will use spider silk particles as an (unpublished results). In the first step, drug molecules are
example of potential drug carriers, outlining their specific attracted to the particle by electrostatic forces (1). After
properties. Furthermore, using spider silk films as an particle surface saturation, low molecular weight drugs
example, we will illustrate how material properties can be start to diffuse into the biopolymer matrix (2). Drug
controlled and designed according to the desired needs for a molecules interact with the matrix via attractive hydro-
specific application. phobic and electrostatic interactions (3). Figure 4B shows
the obtained loading efficiencies of eADF4(C16) particles
with methyl violet (MV) as a function of molar ratio. Up to a
Drug Carriers molar ratio of MV: eADF4(C16)  10 the loading increases
linearly with the amount of MV added. Above a molar ratio
In order to achieve constant drug levels in plasma during
of 10 the loading reaches a plateau, leading to a decrease of
therapy, colloidal micro- and nanoparticulate carriers have
loading efficiency. Upon incubation of loaded particles in
been developed on the basis of various synthetic and
release media, drug molecules are transported to the
natural polymers.[33–40] These systems enable a reduction
particle surface due to concentration gradient driven
of toxic side effects and the number of doses, while
transport processes (4). With time, drug molecules are
improving cellular uptake and bioavailability.[41–45] Stable
slowly released from the surface (in an equilibrium
silk protein particles can be obtained in a simple all-
process), leading to constant release rates for > 30 d at
aqueous process important for encapsulation of sensitive
physiologic conditions (37 8C, pH 7.4) (5) (shown for day 0 to
compounds. Detailed studies of the thermodynamic
5 in Figure 4C). The release can be triggered by pH enabling
assembly process of eADF4(C16) (which is composed of
accelerated release behavior at lower pH (Figure 4C)
16 repeats of module C, Figure 1C) into microparticles
(unpublished results).
revealed that solid particles with high b-sheet content and
smooth surfaces are formed upon addition of kosmotropic
salts such as potassium phosphate.[20,21,46] The conversion
of monomeric soluble spider silk proteins to solid silk Silk Films—Controlled Processing Allowing
particles can be solely triggered by potassium phos- Different Applications
phate[7,20,21,23,25,46,47]; a fact that is very important in terms
of biocompatibility of the drug-delivery system considering In contrast to particles, which can be used as mobile carriers,
both the material and its processing. films are more suitable for applications in which a
The particle size (250 nm to 3 mm) and size distribution stationary phase is needed. It has been shown previously
can be controlled by mixing intensity and the concentration that recombinantly produced engineered spider silk

Macromol. Biosci. 2010, 10, 998–1007


ß 2010 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.mbs-journal.de 1001
K. Spiess, A. Lammel, T. Scheibel

Figure 4. Spider silk based drug carrier. (A) Influence of protein concentration and mixing intensity on spider silk particle size. Particles are
produced by salting out with potassium phosphate (figure adapted from ref.[46]). (B) Loading and loading efficiency of eADF4(C16) with
methyl violet particles as a function of molar ratio. (C) In vitro release kinetics of methyl violet over a period of 5 d at different pH as
indicated. (D) Drug loading and release mechanism of spider silk particles. Constant release is driven by transport processes influenced by
electrostatic and hydrophobic interactions as well as concentration gradients: (1) attraction, (2) saturation and diffusion, (3) binding, (4)
transport to the surface (5) release (unpublished results).

proteins can be assembled into transparent and stable been carried out with silk fibroins of the silkworm B. mori,
films.[48–50] however, similar applications can be envisioned for
recombinant spider silk proteins due to the similarity in
their physico-chemical properties. Controlled release has
Biomedical Applications of Silk Films
been demonstrated for substances which were either
Possible applications studied so far include the use of silk directly integrated into the silk film during layer-by-layer
films as devices for controlled substance (e.g., drugs) release, deposition,[51] or loaded onto microparticles, which were
as biochemical sensors and mainly as cell supporting embedded in or coated by a silk film, resulting in retarded
scaffolds for tissue engineering. Most of these studies have release kinetics.[52,53] Preparation of transparent films with

Macromol. Biosci. 2010, 10, 998–1007


1002 ß 2010 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim DOI: 10.1002/mabi.201000071
Recombinant Spider Silk Proteins for Applications in . . .

distinct patterned surfaces offers potential applications for requirements for tissue engineering devices for skin
use as biochemical sensors. This could be accomplished by substitutes, soft tissues, bone, or ligaments), degradation
incorporation of biologically active components[54] or an behavior and chemical stability, surface topography and
additional responsiveness to refractive indices of the porosity, and additional functionalities. The versatility of
surrounding medium.[55] However, due to its biomedical silk proteins provides the option of varying and controlling
relevance, most effort has been put into the development several of these properties in the final assembly, mainly
and investigation of novel scaffolds suitable for tissue through the processing conditions.
engineering. The culturing of a fibroblast cell line (L929) and Silk films are generally produced by casting, dip- or spray-
human bone marrow stem cells on regenerated B. mori silk coating of protein solutions onto a substrate. Evaporation of
films revealed good cell adhesion, growth, and differentia- the solvent yields a silk film that is stabilized via (non-
tion[56] and allowed these films to be classified as ‘‘non- covalent) intermolecular interactions, and if required, the
cytotoxic’’.[57] Silk films have either been employed directly, films can be peeled off the surface. Silk films may be further
e.g., as wound dressings,[58] or as a coating of commercially structurally or chemically modified by subsequent post-
available porous membranes, where fibroblast adhesion treatments and/or subsequent modifications (mediated for
and proliferation, scaffold strength, and hydrophilicity example through effector molecules). There are two main
were increased.[59] By demonstrating the cultivation and components determining the overall properties of the
differentiation of different cell types (like osteoblast-like resulting film: the structure on the molecular level (secon-
cells,[60,61] mesenchymal stem cells,[62] (corneal) fibro- dary structure of the underlying proteins and intermolecular
blasts,[63–65] or HepG2 cells[66]) on silk substrates, it has interactions[48,74]) and the macroscopic structure, which
been suggested that silk films are suitable for regeneration provides the materials’ interface with its environment.
of corneal tissue or skin or bone. Induction of bone tissue
growth could be further enhanced by addition of cell
Control of Material Properties through the Molecular
adhesive peptides like RGD.[60,61,67] Biomineralization of
Structure of the Silk Proteins
silk films (with, e.g., hydroxyapaptite) was also exploited to
improve bone regeneration.[68] Other attempts to improve The secondary structure is determined and influenced by
the biocompatibility or specifity of silk matrices for tissue the primary structure of the employed protein, the solvent,
engineering include the formation of blends with for and post-treatment conditions. We have shown that films
example chitosan,[69] collagen,[70] keratin,[71] PLA,[72] cast from different recombinant spider silk proteins
PEG,[73] or hyaluronan[62] in order to modify mechanical (mimicking the dragline proteins ADF3 and ADF4 from
or degradation behavior or to prevent cell adhesion for the garden spider A. diadematus) show a secondary
antithrombotic effects. structure composition depending on the used silk pro-
teins/blends (Table 1).[48] For a given protein, the solvent
used (most often water, hexafluoro-2-propanol (HFIP) or
Processing Parameters
formic acid) will determine the predominant secondary
Various material properties are important when preparing structure in the as-cast films. Silk films prepared from
silk-based materials for specific applications, including the aqueous solution mainly consist of random coil struc-
mechanical properties of the material (e.g., different ture,[71,75,76] whereas films prepared using HFIP as the

Table 1. Distribution of secondary structure elements of films of eADF4(C16), eADF3((AQ)12) and blends of both as determined by
deconvolution of the amide I bands after FTIR-spectroscopy.

Protein Post-treatment a-helix b-sheet b-turn Random coil

eADF4(C16) – 19.0 35.0 32.2 13.8


MeOH 12.9 45.2 23.8 18.0
KxH3xPO4 13.2 52.1 25.5 9.2
eADF3((AQ)12) – 23.7 27.7 30.1 18.5
MeOH 18.4 34.8 29.2 17.6
KxH3xPO4 9.9 36.5 32.0 21.6
eADF4/eADF3 – 20.5 20.5 35.7 23.3
MeOH 8.2 46.4 19.9 25.5

Films were analysed either as-cast or after treatment with methanol (MeOH) or potassium phosphate (KxH3xPO4) (adapted from ref. [48]).

Macromol. Biosci. 2010, 10, 998–1007


ß 2010 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.mbs-journal.de 1003
K. Spiess, A. Lammel, T. Scheibel

stability of the respective film. Those rich


in random coil or helical conformation
are water soluble, while b-sheet rich films
are insoluble in water. As for most
applications stability in aqueous solution
is necessary, a subsequent treatment of
water soluble films is applied in order to
render them stable, which is typically
achieved using dehydrating agents like
alcohols[49,50] (usually methanol) or kos-
motropic ions[48] (like potassium phos-
phate, which is known to influence silk
protein structure during the natural
spinning process) and water vapor[80] or
high temperature.[81] All treatments lead
to a structural transition from random
coil/a-helical to a higher content of b-
sheet structure (Table 1). The portion of
induced b-sheet structure can be regu-
lated by the incubation time or by
treatment of the film with solutions of
Figure 5. Processing parameters to control material properties of silk films. (A) Schematic methanol/water mixed at various ratios
presentation of individual steps necessary for film formation and influencing factors. (Figure 5C) (unpublished results).
The material properties can be controlled by variation and combination of distinct The content of b-sheet structure and
parameters at each step. (B) and (C) FTIR-spectra illustrating the influence of different
crystallinity not only determines the
solvents (B) or post-treatments (C) on the secondary structure of eADF4(C16) films
(unpublished results). (D) Functionalization of eADF4(C16) films by covalent coupling of stability of silk films, but also influences
b-galactosidase (þ). Blue precipitate of X-gal (seen as dark color in black and white their mechanical characteristics. Gener-
reprint) indicates enzyme activity. Non-coupled b-galactosidase does not adhere to the ally, an increase in b-sheet structure
silk surface ().[49] (E) Control of porosity of eADF4(C16) films generated by blending increases the elastic modulus and
with different amounts of PEO. Upper panel: 20% (wt.-%) PEO, lower panel: 333% (wt.-%)
strength of the film, but reduces its
PEO. Scale bar: 5 mm.
elasticity[82] (Table 2, films as-cast and
treated with 50% methanol). Not only the
solvent typically yield a-helix-rich structures[48,49,75,77] amount of b-sheets, but also their molecular arrangement is
(Figure 5B). In contrast, films prepared from formic crucial to the final properties.[74] A high amount of b-sheet
acid solutions are rich in b-sheet structure polyalanine regions and long-range order crystals have
(Figure 5B).[71,76,78,79] Interestingly, these different struc- been correlated with stiffness and brittleness in silk
tural conformations of the proteins determine the chemical films.[50,76] Relative humidity of the environment as well

Table 2. Mechanical properties of eADF4(C16) films cast from HFIP and of eADF4(C16) films made in presence of 0, 10, 20, and 40% glycerol
with and without 50% methanol (MeOH) post-treatment (PT).

Material Glycerol smax (Pa) e at sma (%)

% no PT 50% MeOH no PT 50% MeOH

eADF4(C16) – 1.5  107 3.9  107 11 12


7
eADF4(C16) 10 2.3  10 2.6  107 47 21
6 7
eADF4(C16) 20 8.3  10 2.4  10 89 56
eADF4(C16) 40 9.9  106 1.2  107 129 100

The mechanical characteristics (tensile strength and elongation) were assessed in a custom built vertical tensile test setup. Briefly, the top
end of the samples was fixed to a force transducer (FORT25, WPI, Sarosota, USA) and the bottom end to a linear motor (Linmot PS01-23  80
motor, Linmot AG, Switzerland). The pulling velocity for each measurement was 1 mm  s1 and the data was recorded at a rate of 500
datapoints s1.

Macromol. Biosci. 2010, 10, 998–1007


1004 ß 2010 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim DOI: 10.1002/mabi.201000071
Recombinant Spider Silk Proteins for Applications in . . .

as water content of the film also influence the mechanical removal by a washing step after film formation yields pores
properties, with higher water content rendering the film (Figure 5E). The size of the pores as well as their
more elastic, as demonstrated on B. mori fibroin films.[83–85] interconnectivity can be varied by concentration and size
Accordingly, addition of hygroscopic plasticizers like of the employed salts or the concentration of PEG, as shown
glycerol, which alter intermolecular interactions, renders for porous films made of eADF4(C16) (Figure 5E).
spider silk films more elastic (Table 2) and can therefore be Taken together, the combination of processing para-
used to further adjust the mechanical properties.[84] As an meters allows the generation of tailored silk films.
example, a content of 40% (v/w) glycerol increases the However, for specific applications, additional features
elasticity of a eADF4(C16) film about tenfold, accompanied might be necessary; a task which can be met by combining
by a tenfold decrease in elastic modulus and a slight silk films with additional functional groups such as
decrease in strength (Table 2). components of the extracellular matrix for better cell
Another important topic for the suitability of silk films in adhesion or differentiation. Modification of surface hydro-
biomedical applications is the degradation of the respective philicity or coupling of (poly-)peptides is also feasible via
material. For example, in functional tissue design, the chemical coupling through reactive side chains of amino
replacement of an implant by the body’s own tissue is acids (Figure 5D).[67,93] Our established recombinant
desired.[86] Silk proteins are degraded by distinct proteases production technology of spider silk proteins offers various
in vivo, mediated by the foreign body response[87] in case of possibilities to introduce new/additional amino acid side
implantation, or by enzymes of the intestinal tract in case of chains by genetic engineering, which can be utilized for
oral administration. In vitro analysis, for instance, revealed further functionalization.
the degradation of eADF4(C16) particles by artificial
intestinal fluid, but not by artificial gastric fluid.[88] Studies
on B. mori fibroin revealed that besides the morphology of Summary
the material (including porositiy) and the presence of
recognition sites for proteolytic enzymes, the secondary Recombinantly produced spider silk proteins possess a high
structure and degree of crystallinity also influence the rate potential for biomedical applications. Here, we highlighted
of degradation,[86,87,89] with the crystalline regions being the production of silk particles and films showing that the
the most stable structures. molecular and macroscopic material properties can be
easily controlled in vitro.
In case of eADF4(C16) particles, one major advantage of
Material Function Controlled by its Macroscopic the system is the production and loading within an all-
Structure aqueous process under ambient conditions, which is
Besides the silk properties determined by the molecular extremely important considering encapsulation of labile
structure of the underlying proteins, the materials’ macro- compounds and the biocompatibility of the product. In case
scopic structure has a direct impact on biomedical of silk films, we showed that molecular structure can be
applications. Silk fibroin films, for instance, displaying a influenced by solvents and postcasting treatments and that
distinct surface morphology can be prepared by casting the macroscopic properties like porosity and mechanical
films on grooved or patterned substrates,[54,65] with the stability can be influenced by additives like PEO and
structure of the substrate being reflected in the films’ glycerol. This level of control allows the adaptation of such
topography. Such patterned film surfaces can be used to films for a large variety of biomedical applications. The
induce aligned cell growth.[65] Another option of generating diversity of spider silk proteins and the processing control
altered surface topography on silk films is to apply different along with their biocompatibility and biodegradability
post-treatment conditions. Applying diverse methanol renders them valuable tools for future applications in novel
treatments led to varying roughness of the individual biomaterials.
films and resulted in different growth behavior of
fibroblasts.[90] In general, the topography of a cell support- Acknowledgements: This work was supported by the Interna-
tional Graduate School of Science and Engineering (IGSSE to AL)
ing scaffold has an impact on cell morphology, adhesion,
within the Elite Network of Bavaria and funding by the Army
alignment, proliferation, and differentiation.[90,91] In order Research Office (grant W911NF-0810284). The authors wish to
to obtain interfaces for applications ranging from tissue thank Eileen Lintz and Dr. John Hardy for critical reading the
engineering to controlling diffusion of molecules, porous manuscript.
films have been prepared by salt leaching, gas foaming, or
freeze-drying.[92] In salt leaching, salt crystals are Received: February 15, 2010; Revised: April 1, 2010; Published
embedded during film formation and washed out after online: July 5, 2010; DOI: 10.1002/mabi.201000071
stabilizing the film. Similar to the salt leaching process, Keywords: biopolymers; drug delivery systems; process para-
addition of PEG to the protein solution and its subsequent meters; recombinant spider silks; tissue engineering

Macromol. Biosci. 2010, 10, 998–1007


ß 2010 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.mbs-journal.de 1005
K. Spiess, A. Lammel, T. Scheibel

[1] M. Heim, D. Keerl, T. Scheibel, Angew. Chem. Int. Ed. 2009, 48, [37] Y.-W. Won, Y.-H. Kim, J. Controlled Release 2008, 127, 154.
3584. [38] T. M. Allen, P. R. Cullis, Science 2004, 303, 1818.
[2] F. Vollrath, Int. J. Biol. Macromol. 1999, 24, 81. [39] U. Edlund, A. Albertsson, in: Degradable Aliphatic Polyesters,
[3] F. Vollrath, Rev. Mol. Biotechnol. 2000, 74, 67. A. C. Albertsson, Ed., Springer, Berlin 2002, pp. 67.
[4] S. Winkler, D. L. Kaplan, Rev. Mol. Biotechnol. 2000, 74, 85. [40] R. Arshady, J. Bioact. Compatible Polym. 1990, 5, 315.
[5] J. D. van Beek, S. Hess, F. Vollrath, B. H. Meier, Proc. Natl. Acad. [41] N. Yasutomo, N. Yoshiharu, W. Masahisa, K. Shigenori, M. Jun,
Sci. USA 2002, 99, 10266. J. Appl. Polym. Sci. 2002, 86, 3425.
[6] D. Huemmerich, T. Scheibel, F. Vollrath, S. Cohen, U. Gat, [42] J. K. Vasir, K. Tambwekar, S. Garg, Int. J. Pharm. 2003, 255, 13.
S. Ittah, Curr. Biol. 2004, 14, 2070. [43] M. Roser, D. Fischer, T. Kissel, Eur. J. Pharm. Biopharm. 1998,
[7] D. Huemmerich, C. W. Helsen, S. Quedzuweit, J. Oschmann, 46, 255.
R. Rudolph, T. Scheibel, Biochemistry 2004, 43, 13604. [44] K. E. Uhrich, S. M. Cannizzaro, R. S. Langer, K. M. Shakesheff,
[8] B. O. Swanson, T. A. Blackledge, J. Beltrán, C. Y. Hayashi, Appl. Chem. Rev. 1999, 99, 3181.
Phys. A: Mater. Sci. Process. 2006, 82, 213. [45] S. K. Daniel, Biotechnol. Bioeng. 2007, 96, 203.
[9] A. M. Smith, T. Scheibel, Macromol. Chem. Phys. 2010, 211, [46] A. Lammel, M. Schwab, U. Slotta, G. Winter, T. Scheibel, Chem.
127. Sus. Chem. 2008, 1, 413.
[10] J. M. Gosline, P. A. Guerette, C. S. Ortlepp, K. N. Savage, J. Exp. [47] J. H. Exler, D. Hummerich, T. Scheibel, Angew. Chem. Int. Ed.
Biol. 1999, 202. 3295. Engl. 2007, 46, 3559.
[11] C. Y. Hayashi, N. H. Shipley, R. V. Lewis, Int. J. Biol. Macromol. [48] U. Slotta, M. Tammer, F. Kremer, P. Koelsch, T. Scheibel,
1999, 24, 271. Supramol. Chem. 2006, 18, 465.
[12] C. Vendrely, T. Scheibel, Macromol. Biosci. 2007, 7, 401. [49] D. Huemmerich, U. Slotta, T. Scheibel, Appl. Phys. A 2006, 82,
[13] F. Hagn, L. Eisoldt, J. G. Hardy, C. Vendrely, C. Murray, 219.
T. Scheibel, H. Kessler, Nature 2010, 465, 239. [50] K. Spiess, S. Wohlrab, T. Scheibel, 2010, In revision.
[14] H. J. Jin, D. L. Kaplan, Nature 2003, 424, 1057. [51] X. Y. Wang, X. Hu, A. Daley, O. Rabotyagova, P. Cebe, D. L.
[15] F. Vollrath, D. P. Knight, X. W. Hu, Proc. R Soc. London Ser. B- Kaplan, J. Controlled Release 2007, 121, 190.
Biol. Sci. 1998, 265, 817. [52] X. Wang, E. Wenk, X. Hu, G. R. Castro, L. Meinel, X. Wang, C. Li,
[16] C. Dicko, J. M. Kenney, D. Knight, F. Vollrath, Biochemistry H. Merkle, D. L. Kaplan, Biomaterials 2007, 28, 4161.
2004, 43, 14080. [53] E. Wenk, A. J. Meinel, S. Wildy, H. P. Merkle, L. Meinel,
[17] G. Askarieh, M. Hedhammar, K. Nordling, A. Saenz, C. Casals, Biomaterials 2009, 30, 2571.
A. Rising, J. Johansson, S. D. Knight, Nature 2010, 465, 236. [54] B. D. Lawrence, M. Cronin-Golomb, I. Georgakoudi, D. L.
[18] J. G. Hardy, L. M. Roemer, T. R. Scheibel, Polymer 2008, 49, Kaplan, F. G. Omenetto, Biomacromolecules 2008, 9, 1214.
4309. [55] J. J. Amsden, H. Perry, S. V. Boriskina, A. Gopinath, D. L. Kaplan,
[19] J. Perez-Riguero, M. Elices, J. Llorca, C. Viney, J. Appl. Polym. Sci. L. Dal Negro, F. G. Omenetto, Opt. Express 2009, 17, 21271.
2001, 82, 2245. [56] X. Y. Wang, H. J. Kim, P. Xu, A. Matsumoto, D. L. Kaplan,
[20] U. K. Slotta, S. Rammensee, S. Gorb, T. Scheibel, Angew. Chem. Langmuir 2005, 21, 11335.
Int. Ed. Engl. 2008, 47, 4592. [57] T. L. Liu, J. C. Miao, W. H. Sheng, Y. F. Xie, Q. Huang, Y. B. Shan,
[21] S. Rammensee, U. Slotta, T. Scheibel, A. R. Bausch, Proc. Natl. J. C. Yang, J. Zhejiang Univ. Sci. B 2010, 11, 10.
Acad. Sci. USA 2008, 105, 6590. [58] A. Sugihara, K. Sugiura, H. Morita, T. Ninagawa, K. Tubouchi,
[22] L. Eisoldt, J. G. Hardy, M. Heim, T. R. Scheibel, J. Struct. Biol. R. Tobe, M. Izumiya, T. Horio, N. G. Abraham, S. Ikehara, Proc.
2010, 170, 413. Soc. Exp. Biol. Med. 2000, 225, 58.
[23] T. Scheibel, Microb. Cell Fact. 2004, 3, 14. [59] C. Cassinelli, G. Cascardo, M. Morra, L. Draghi, A. Motta,
[24] C. W. P. Foo, E. Bini, J. Hensman, D. P. Knight, R. V. Lewis, D. L. G. Catapano, Int. J. Artif. Organs 2006, 29, 881.
Kaplan, Appl. Phys. A 2006, 82, 223. [60] E. Bini, C. W. P. Foo, J. Huang, V. Karageorgiou, B. Kitchel, D. L.
[25] U. Slotta, S. Hess, K. Spiess, T. Stromer, L. Serpell, T. Scheibel, Kaplan, Biomacromolecules 2006, 7, 3139.
Macromol. Biosci. 2007, 7, 183. [61] A. W. Morgan, K. E. Roskov, S. Lin-Gibson, D. L. Kaplan, M. L.
[26] A. Matsumoto, A. Lindsay, B. Abedian, D. L. Kaplan, Macromol. Becker, C. G. Simon, Biomaterials 2008, 29, 2556.
Biosci. 2008, 8, 1006. [62] M. Garcia-Fuentes, A. J. Meinel, M. Hilbe, L. Meinel, H. P.
[27] I. C. Um, H. Kweon, Y. H. Park, S. Hudson, Int. J. Biol. Macromol. Merkle, Biomaterials 2009, 30, 5068.
2001, 29, 91. [63] J. S. Chen, G. H. Altman, V. Karageorgiou, R. Horan, A. Collette,
[28] Z. Chenhua, Y. Juming, M. Hiromi, K. Raghuvansh, A. Tetsuo, V. Volloch, T. Colabro, D. L. Kaplan, J. Biomed. Mater. Res.
Biopolymers 2003, 69, 253. Part A 2003, 67A, 559.
[29] D. M. Phillips, L. F. Drummy, D. G. Conrady, D. M. Fox, R. R. [64] A. Chiarini, P. Petrini, S. Bozzini, I. Dal Pra, U. Armato, Bio-
Naik, M. O. Stone, P. C. Trulove, H. C. De Long, R. A. Mantz, materials 2003, 24, 789.
J. Am. Chem. Soc. 2004, 126, 14350. [65] B. D. Lawrence, J. K. Marchant, M. A. Pindrus, F. G. Omenetto,
[30] B. Lawrence, F. Omenetto, K. Chui, D. Kaplan, J. Mater. Sci. D. L. Kaplan, Biomaterials 2009, 30, 1299.
2008, 43, 6967. [66] Q. Lv, K. Hu, Q. L. Feng, F. Z. Cui, J. Appl. Polym. Sci. 2008, 109,
[31] K. D. Hermanson, D. Huemmerich, T. Scheibel, A. R. Bausch, 1577.
Adv. Mater. 2007, 19, 1810. [67] S. Sofia, M. B. McCarthy, G. Gronowicz, D. L. Kaplan, J. Biomed.
[32] S. Rammensee, D. Huemmerich, K. D. Hermanson, T. Scheibel, Mater. Res. 2001, 54, 139.
A. R. Bausch, Appl. Phys. A 2006, 82, 261. [68] Y. Tamada, T. Furuzono, T. Taguchi, A. Kishida, M. Akashi,
[33] S. Freiberg, X. X. Zhu, Int. J. Pharm. 2004, 282, 1. J. Biomater. Sci. Polym. Ed. 1999, 10, 787.
[34] L. Robert, A. P. Nicholas, AIChE J. 2003, 49, 2990. [69] K. Cai, Y. Hu, K. D. Jandt, J. Biomed. Mater. Res. Part A 2007,
[35] M. George, T. E. Abraham, J. Controlled Release 2006, 114, 1. 82A, 927.
[36] X. Liu, Q. Sun, H. Wang, L. Zhang, J.-Y. Wang, Biomaterials [70] T. Asakura, J. Yao, T. Yamane, K. Umemura, A. S. Ulrich, J. Am.
2005, 26, 109. Chem. Soc. 2002, 124, 8794.

Macromol. Biosci. 2010, 10, 998–1007


1006 ß 2010 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim DOI: 10.1002/mabi.201000071
Recombinant Spider Silk Proteins for Applications in . . .

[71] A. Vasconcelos, G. Freddi, A. Cavaco-Paulo, Biomacromole- [84] S. Lu, X. Wang, Q. Lu, X. Zhang, J. A. Kluge, N. Uppal,
cules 2008, 9, 1299. F. Omenetto, D. L. Kaplan, Biomacromolecules 2010, 11,
[72] H. L. Zhu, X. X. Feng, H. P. Zhang, Y. H. Guo, J. Z. Zhang, J. Y. 143.
Chen, J. Biomater. Sci. Polym. Ed. 2009, 20, 1259. [85] B. D. Lawrence, S. Wharram, J. A. Kluge, G. G. Leisk, F. G.
[73] C. Vepari, D. Matheson, L. Drummy, R. Naik, D. L. Kaplan, Omenetto, M. I. Rosenblatt, D. L. Kaplan, Macromol. Biosci.
J. Biomed. Mater. Res. Part A 2009, 93A, 595. 2010, 10, 393.
[74] E. Metwalli, U. Slotta, C. Darko, S. V. Roth, T. Scheibel, C. M. [86] C. Vepari, D. L. Kaplan, Prog. Polym. Sci. 2007, 32, 991.
Papadakis, Appl. Phys. A 2007, 89, 655. [87] G. H. Altman, F. Diaz, C. Jakuba, T. Calabro, R. L. Horan, J. S.
[75] C. Zhao, J. Yao, H. Masuda, R. Kishore, T. Asakura, Biopolymers Chen, H. Lu, J. Richmond, D. L. Kaplan, Biomaterials 2003, 24,
2003, 69, 253. 401.
[76] I. C. Um, H. Y. Kweon, Y. H. Park, S. Hudson, Int. J. Biol. [88] B. Liebmann, D. Hümmerich, T. Scheibel, M. Fehr, Colloids
Macromol. 2001, 29, 91. Surf. A 2008, 331, 126.
[77] J. S. Stephens, S. R. Fahnestock, R. S. Farmer, K. L. Kiick, D. B. [89] A. Leal-Egana, T. Scheibel, Biotechnol. Appl. Biochem. 2010, 55,
Chase, J. F. Rabolt, Biomacromolecules 2005, 6, 1405. 155.
[78] S. W. Ha, A. E. Tonelli, S. M. Hudson, Biomacromolecules 2005, [90] E. Servoli, D. Maniglio, A. Motta, R. Predazzer, C. Migliaresi,
6, 1722. Macromol. Biosci. 2005, 5, 1175.
[79] B. M. Min, L. Jeong, Y. S. Nam, J. M. Kim, J. Y. Kim, W. H. Park, [91] C. J. Bettinger, R. Langer, J. T. Borenstein, Angew. Chem. Int. Ed.
Int. J. Biol. Macromol. 2004, 34, 281. 2009, 48, 5406.
[80] H. J. Jin, S. V. Fridrikh, G. C. Rutledge, D. L. Kaplan, Biomacro- [92] Y. Z. Wang, H. J. Kim, G. Vunjak-Novakovic, D. L. Kaplan,
molecules 2002, 3, 1233. Biomaterials 2006, 27, 6064.
[81] J. Magoshi, S. Nakamura, J. Appl. Polym. Sci. 1975, 19, 1013. [93] A. R. Murphy, P. S. John, D. L. Kaplan, Biomaterials 2008, 29,
[82] N. Minoura, M. Tsukada, M. Nagura, Biomaterials 1990, 11, 430. 4260.
[83] Q. Lu, X. Hu, X. Wang, J. A. Kluge, S. Lu, P. Cebe, D. L. Kaplan, [94] J. P. Anderson, Protein Based Materials, K. McGrath, D. L.
Acta Biomater. 2009, 6, 1380. Kaplan, Eds., Birkhäuser, Boston 1996, p. 371.

Macromol. Biosci. 2010, 10, 998–1007


ß 2010 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.mbs-journal.de 1007

You might also like