You are on page 1of 16

Acta Biomaterialia 31 (2016) 17–32

Contents lists available at ScienceDirect

Acta Biomaterialia
journal homepage: www.elsevier.com/locate/actabiomat

Review article

Silk protein-based hydrogels: Promising advanced materials


for biomedical applications
Sonia Kapoor a,b, Subhas C. Kundu a,⇑
a
Department of Biotechnology, Indian Institute of Technology Kharagpur, West Bengal 721302, India
b
University Institute of Engineering and Technology, Panjab University, Chandigarh 160014, India

a r t i c l e i n f o a b s t r a c t

Article history: Hydrogels are a class of advanced material forms that closely mimic properties of the soft biological tis-
Received 10 March 2015 sues. Several polymers have been explored for preparing hydrogels with structural and functional fea-
Received in revised form 8 November 2015 tures resembling that of the extracellular matrix. Favourable material properties, biocompatibility and
Accepted 17 November 2015
easy processing of silk protein fibers into several forms make it a suitable material for biomedical appli-
Available online 18 November 2015
cations. Hydrogels made from silk proteins have shown a potential in overcoming limitations of hydro-
gels prepared from conventional polymers. A great deal of effort has been made to control the properties
Keywords:
and to integrate novel topographical and functional characteristics in the hydrogel composed from silk
Silk proteins
Fibroin
proteins. This review provides overview of the advances in silk protein-based hydrogels with a primary
Hydrogel emphasis on hydrogels of fibroin. It describes the approaches used to fabricate fibroin hydrogels.
Biomaterial Attempts to improve the existing properties or to incorporate new features in the hydrogels by making
Biomedical applications composites and by improving fibroin properties by genetic engineering approaches are also described.
Applications of the fibroin hydrogels in the realms of tissue engineering and controlled release are
reviewed and their future potentials are discussed.

Statement of Significance

This review describes the potentiality of silk fibroin hydrogel. Silk Fibroin has been widely recognized as
an interesting biomaterial. Due to its properties including high mechanical strength and excellent bio-
compatibility, it has gained wide attention. Several groups are exploring silk-based materials including
films, hydrogels, nanofibers and nanoparticles for different biomedical applications. Although there is a
good amount of literature available on general properties and applications of silk based biomaterials,
there is an inadequacy of extensive review articles that specifically focus on silk based hydrogels. Silk-
based hydrogels have a strong potential to be utilized in biomedical applications. Our work is an effort
to highlight the research that has been done in the area of silk-based hydrogels. It aims to provide an
overview of the advances that have been made and the future course available. It will provide an over-
view of the silk-based hydrogels as well as may direct the readers to the specific areas of application.
Ó 2015 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
2. Structure and properties of silk. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
3. Silk-based hydrogels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
3.1. Fibroin hydrogels. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
3.1.1. Preparation and characterization. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
3.1.2. Sol–gel transition mechanism of fibroin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21
3.2. Silk fibroin composite hydrogels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22

⇑ Corresponding author.
E-mail address: kundu@hijli.iitkgp.ernet.in (S.C. Kundu).

http://dx.doi.org/10.1016/j.actbio.2015.11.034
1742-7061/Ó 2015 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.
18 S. Kapoor, S.C. Kundu / Acta Biomaterialia 31 (2016) 17–32

3.3. Hydrogel fabrication from genetically engineered silk . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24


3.3.1. Silk-elastin like hydrogel . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
4. Biomedical applications of silk-based hydrogels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
4.1. Tissue engineering. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
4.1.1. Bone tissue engineering . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
4.1.2. Cartilage tissue engineering . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26
4.2. Controlled release . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26
4.2.1. Controlled drug delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26
4.2.2. Controlled release of DNA for gene therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
5. Challenges and future prospects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29

1. Introduction Among the naturally occurring fibers, silk occupies a special


position because of its properties. Silk refers to protein fibers pro-
Biomaterials play a significant role in regenerative medicine as duced by several species of phylum Arthropoda. Several insects and
implants, sutures, contact lenses, hip joints, vascular grafts, wound spiders are able to spin silk fibers; the silks from the domesticated
dressing materials, drug releasing stents and several other biomed- mulberry silkworm, Bombyx mori, and spiders, Nephila clavipes and
ical devices [1]. Recent developments that have offered the ability Araneus diadematus have been extensively characterized [55–60].
to control material properties have led to tremendous success in Of late, various forms of silk-based materials including films, scaf-
the field of tissue engineering and therapeutic medicine [2]. folds, sponges, tubes, electrospun fibers and hydrogels have been
Hydrogels have come to the forefronts as an important form of evaluated for several applications [61,62]. This review focuses on
materials for biomedical applications [3–7]. Hydrogels are three- the advances achieved in making hydrogels from silk proteins. It
dimensional polymeric networks that are capable of absorbing describes several methods that have been used for making silk pro-
large amounts of water while maintaining their structural integ- tein hydrogels, structural and functional characteristics and appli-
rity. They are usually classified into two categories namely physical cations of these hydrogels.
gels (where the polymeric network is held together by secondary
forces that include hydrogen bonds, ionic as well as hydrophobic
interactions) and chemical hydrogels (where covalent linkages 2. Structure and properties of silk
form the network). Since the introduction of hydrogels as biomate-
rials in the 1960s [8]), they have gained considerable attention and Silk proteins from different silkworm species exhibit variations
have found numerous applications in tissue engineering, drug in their structure and properties [55,59,63]. Silk proteins can be
delivery, implantable devices, biosensors and bio- isolated from the cocoons or the silk glands of silkworms [64,65].
nanotechnology. The primary reason of the widespread applicabil- Silk obtained from silkworms is mainly composed of two classes
ity of hydrogels for biomedical applications is the close resem- of proteins, fibroin and sericin. B. mori fibroin protein consists of
blance of their physical and mechanical properties to that of a glycoprotein named P25 and a light (26 kDa) and a heavy chain
biological tissues [7]. (325 kDa), linked by a disulphide bond [60]. On the other hand,
Several natural and synthetic polymeric materials have been fibroin protein of Antheraea mylitta silkworm is composed of two
explored to prepare hydrogels. Proteins, the fundamentally similar-sized polypeptides with an estimated molecular weight
important macromolecules of living systems, have evolved to per- of 197 kDa each which are linked together by a disulfide bond
form very specific biochemical, mechanical and structural roles. [66]. Sericin proteins, having a molecular weight ranging between
The increased understanding about the structural and functional 10 and 300 kDa, act as glue like coating to keep fibroin chains
features of a variety of proteins and advancement in techniques together. Bundles of nanofibrils form fibroin filaments that consti-
to manipulate them has opened avenues to utilize them for tute 70–75% of weight of silk fiber; the remaining weight is sericin.
unconventional purposes [9–11]. Due to their inherent advan- In general, the primary sequence of fibroin consists of highly repet-
tages including biocompatibility, ease of large-scale production itive motifs [55,59,67]. For B. mori, the primary repetitive sequence
via recombinant DNA technology and facile manipulation by is the hexapeptide GAGAGS [55,59,67]. These predominant
chemical or enzymatic means, several proteins have been evalu- hydrophobic blocks lead to extensive hydrogen and hydrophobic
ated for their performance as biomaterials (Table 1) [9–11]. Pro- interactions throughout the protein chains resulting in homoge-
tein polymers display a variety of properties, which prove neous secondary structure. These interactions impart crystallinity
especially useful for biomedical applications [9–11]. They to the protein which, in turn, enhances environmental stability of
undergo hierarchical self-organization that can be mimicked out- fibroin [68]. These highly crystalline regions (containing repetitive
side cellular environment, thus, offering a bottom-up approach alanine or glycine rich sequences) associate with less organized
for assembling biomedical devices [48]. Proteins often contain regions of 34–40 amino acids domains which are interspersed
several domains that assist in cell signalling through their inter- between the crystalline regions [67] and result in the remarkable
action with other proteins or ligands [49,50]. The biomaterials strength and elasticity of silk protein fibers [69]. Not only do
prepared from them can, therefore, be anticipated to retain this fibroin fibers have good mechanical strength, they also possess
function. Proteins can be integrated with synthetic polymers excellent process ability and have been processed into several
easily to further enhance their material properties or can be forms including films, sponges, mats, gels, scaffolds and tubes
made to adhere on synthetic surfaces, providing a hybrid inter- [70]. Silk-based biomaterials have been found to be highly biocom-
face [10,51,52]. Moreover, rational genetic engineering patible with various cell types. For example, adhesion and prolifer-
approaches provide us with a possibility to introduce novel fea- ation studies of human and animal cell lines on films formed from
tures such as self-assembly, sensitivity to specific stimuli, native silkworm fibroin and regenerated silk fibroin [71–73] have
biorecognition, and controlled degradation in proteins [53,54]. suggested it to be a suitable matrix for cell growth. Although, the
S. Kapoor, S.C. Kundu / Acta Biomaterialia 31 (2016) 17–32 19

Table 1
Proteins that have been used to prepare hydrogels.

Proteins Functions in nature Typical Advantages Dis- Applications Reference


Mechanical advantages (s)
Properties of
protein fiber
Collagen Major protein of the 1.2 GPaa Good biocompatibility Mechanical Tissue regeneration, Corneal [12–16]
extracellular matrix weakness and replacement, Cartilage repair,
and provides rapid gene therapy, growth factor
structural support degradation delivery
Gelatin Degraded form of NA Extremely biocompatible, lower Mechanical Differentiation of pluripotent [17–22]
collagen immunogenicity as compared to weakness cells; Growth factor delivery,
collagen, forms gels by temperature drug delivery, tissue
change, Pharmaceutically approved as a engineering, wound dressing
coating material
Fibrin/fibronectin Component of the 1–10 MPab Very closely mimics the properties of the Limited Wound healing, Angiogenesis, [23–29]
extracellular matrix soft tissues, promotes cell adherence mechanical Tissue engineering
strength
Elastin Provides elasticity to 1.1 MPac Self assembles under physiological Calcification of Tissue engineering [30–32]
the tissues conditions elastin
implants,
Rapid
degradation
Fibroin Basic material to 7 GPad High mechanical strength Slow gelation Tissue repair, repair of bone [33–38]
form cocoons, nets, rate defects, delivery of therapeutic
traps by insects and molecules
silkworms
Whey proteins Protein components 0.2 kPae Cheap availability, by-product of milk Mechanical Controlled drug release [39–41]
of milk apart from industry, preparation of gel does not weakness
casein and provide need chemical cross-linkers
nutrition
Sericin Acts as a glue to bind 0.9 MPaf Easily available as by-product of silk May result in Wound dressing material [42–44]
fibroin fibrils industry immunogenic
reactions
a
Young’s modulus, E, of collagen from mammalian tendon [45].
b
Young’s modulus, E, of uncross-linked fibrin fibers [26].
c
Young’s modulus, E, of elastin fiber from bovine ligament [46].
d
Bombyx mori cocoon silk [47].
e
Equilibrium modulus of a typical hydrogel made from 12% whey protein solution [40].
f
Maximum stress of a typical hydrogel in wet state at failure [42].

adherence of cells on silk surfaces has been found to vary depend- [79]. Mechanical strength, biocompatibility, stability to heat and
ing on the source of protein and the processing conditions, never- humidity, high permeability to oxygen and other molecules along
theless, silk-based biomaterials provide good support for cell with the opportunity to control the material characteristics
adhesion and proliferation and do not cause any major cell toxicity through methanol treatment and genetic engineering has made
[73]. It is well emerging that fibroin does not cause activation of silk protein-based materials much sought after for biomedical pur-
immune response. In fact, it evokes a minimum foreign body poses [61,80–82].
response [68,74,75] and has been used as a suture material for
many centuries.
Silk proteins degrade via enzymatic and non-enzymatic means 3. Silk-based hydrogels
[76]. The degradation of silk biopolymer has been found to be
slower as compared to several natural polymers [68,74,76]. Due In vivo, the cells grow, divide, perform their functions, commu-
to its high crystallinity, it may take several days or up to weeks nicate with each other and migrate. All these functions, in one way
to degrade in vivo with the rate of absorption depending on factors or the other, are supported by a matrix – the extracellular matrix
like implantation site, the mechanical environment, the type of silk (ECM) that provides mechanical support as well as physicochemi-
(virgin or processed), the diameter of the silk fiber and the sec- cal cues to cells to perform these functions [83]. Extracellular
ondary structure [68,74]. Due to the slow degradation rate of matrix is composed of fibrous proteins (laminin, collagen and
fibroin, silk-based devices provide a suitable material that can sup- fibronectin) and proteoglycans. The protein chains form a mesh
port the neo-forming tissues for long duration. Moreover, the like network and provide the mechanical support while the proteo-
degradation products of silk fibroin materials have been shown glycans occupy the interstitial sites of this polymeric network [83].
to be harmless to the human body [76]. Dynamic microrestructuring of ECM occurs continuously and
In addition to tailoring the properties of silk proteins by chem- essentially maintains the tissue homeostasis [84]. Hydrogels have
ical and physical modifications, their properties can be modulated been widely accepted as near prototypes of the ECM and have been
according to the requirements through genetic engineering. Bio- found to be suitable 3-dimensional matrices for cell growth [7].
compatibility and cell adhesion of silk proteins has been improved The hydrogels prepared earlier have been more of a passive 3D
by integrating sequences from other proteins like collagen and platform for cell growth. The significance of properties like
fibronectin [77,78]. For instance, Morgan group have prepared mechanical stiffness of hydrogels in mechanotransduction and of
scaffolds by blending fibroin and synthetic spidroin, which con- biochemical and topological cues in promoting physiological func-
tains RGD sequences, thus, leading to easy and inexpensive incor- tions of cells has been identified lately [85]. Thus, the emphasis has
poration of sequences to promote cell adhesion and differentiation now shifted to fabricating the hydrogels that reflect the essence of
20 S. Kapoor, S.C. Kundu / Acta Biomaterialia 31 (2016) 17–32

Fig. 1. Schematic representation of hydrogel fabrication from silk fibroin protein: Cocoons and the silk glands are main sources of silk fibroin proteins. Larvae and cocoons of
domesticated mulberry silkworm, Bombyx mori and non-mulberry tropical tasar silkworm, Antheraea mylitta are shown. Silk fibroin protein is isolated from middle silk glands
and cocoons of different silkworms and is regenerated. The regenerated silk fibroin protein is processed through various treatments to get hydrogel. A typical image of fibroin
hydrogel fabricated in our laboratory is shown.

cellular microenvironment and can mimic the key aspects of ECM. The major properties that need to be characterized for a hydrogel
As noted above, fibroin-based material promotes cell attachment include its porosity, water content and swelling behaviour, size
and proliferation [73]. In particular, the silk from wild silkworm and shape, mechanical and rheological properties, biocompatibility
like Antheraea pernyi contains RGD sequences which are known and biodegradation. Not only is it essential to determine the initial
to be the recognition sequences for integrin receptors that mediate properties of hydrogels, but equally important is to consider how
the interaction between cells and ECM [86,87]. In addition, the these properties change with time while the hydrogel is still in
mechanical properties including extensibility and toughness of use. These properties and their dynamics influence the spatiotem-
fibroin fibers, particularly that from wild silkworm fiber are even poral behaviour of the hydrogel, thus governing its utility for a
higher than polymers like elastin and Kevlar [68,75]. The superior specific application. Since the processing conditions greatly influ-
mechanical strength of hydrogels prepared from silk fibers may ence the characteristics of the hydrogels, it is desirable to control
thus provide opportunities to overcome the limitations that are the processing of hydrogels to exert a strict control over the resul-
often faced while using the hydrogels obtained from other natural tant properties. As discussed below, it is becoming increasingly
polymers [62]. possible to fine tune the properties of silk hydrogels.
Fig.1 schematically outlines the general steps to fabricate silk
3.1. Fibroin hydrogels hydrogels. Table 2 lists the methods that have been used to pro-
duce silk protein fibroin hydrogels. Predominance of hydrophobic
3.1.1. Preparation and characterization amino acid groups like glycine, serine and alanine [100] in fibroin
In general, hydrogels are obtained by physical or chemical makes gelation possible without addition of any gelling agent. For
cross-linking of the polymeric chains. Since the performance of example, Ayub et al. prepared fibroin gel of mechanical strength
hydrogels in the in vivo setting primarily depends on their proper- ranging from 1 kg/cm2 to 30 kg/cm2 by keeping fibroin solution
ties, it is essential to determine the fundamental characteristics of at 20 °C, at a relative humidity of 56–64% [101]. Slow rate of gela-
a hydrogel before it can be explored for its biomedical potential. tion of fibroin has been one of the major challenges in preparing
S. Kapoor, S.C. Kundu / Acta Biomaterialia 31 (2016) 17–32 21

Table 2
Methods for preparation of silk fibroin protein hydrogels.

Method Mechanical propertiesa Specific conditions Comments Reference


(s)
Compressive % strain
modulus at
(MPa) failure
Temperature changes 1.2b 50% 4, 25, 37, 60 °C have generally Higher temperature improved the mechanical [88–90]
been used strength, Method not suitable for direct
encapsulation of cells and temperature sensitive
peptides and bioactive proteins
Lowering of pH towards isoelectric NA NA Citric acid, Citrate–phosphate Neutralization of acid is needed, method is not [36,89]
point of fibroin buffer and HCl are mainly used suitable for direct encapsulation of pH sensitive
molecules and cells
Shearing forces induced by 1.6c NA Power output = 20–50% Method found suitable for making injectable gels [91–95]
sonication and vortexing of amplitude, time = 5–30 s, and for encapsulating cells prior to final setting of
fibroin solution 3200 rpm using vortexer gel
CO2 acidification 64.0 kPad 2 wt% fibroin solution, pressure Ease of fabrication; hydrogels free of mineral acids [96,97]
CO2 = 60 bar or chemical cross-linkers
Phase separation induced by NA 16% Water soluble organic solvent Generally leads to formation of hydrogels with [98,99]
freezing and freeze thawing heterogeneous porosity
a
Typical mechanical properties of the hydrogels prepared by different methods as reported in literature.
b
Gel prepared from 8% fibroin solution at 37 °C.
c
Gel prepared from 8% fibroin solution at power output = 30% amplitude.
d
Gel prepared from 4% fibroin solution processed for 8 h.

silk gels. For example, 2% fibroin solution (pH 6.4–6.8) kept at 37 °C homogeneous distribution of the cells/bioactive molecules in the
has been reported to form gel after about 30 days [88]. This limita- matrix. Lately, vortexing and electric current have also been used
tion has been overcome by stimulating gel formation in fibroin by to fabricate silk hydrogels [93,108]. The treatments including son-
changing temperature, pH or ionic concentration by addition of ication and vortexing promote b-sheet transformation in silk,
salts like CaCl2 or KCl [88,89,98,102] and by using several methods which ultimately result in gel formation [92–94].
like shearing, sonication, removal of bulk water by osmotic stres-
ses, vortexing, heating and exposure to solvents, gases and surfac- 3.1.2. Sol–gel transition mechanism of fibroin
tants like sodium dodecyl sulfate and sodium N-lauroyl sarcosinate Various groups have worked to understand the molecular
[90,96,97,103–106]. Silk fibroin solution treated at higher temper- mechanisms involved in the gelation of fibroin. Initially, it was pro-
ature (60 °C) gels faster than the solution kept at 37 °C [84,85,101]. posed that b-sheet transformation is responsible for gelation of silk
Similarly, an increase in fibroin concentration as well as a decrease solution [109,110] and it has later been shown that there is higher
in pH towards its isoelectric point (3.8) also results in faster gela- b-sheet content in silk gels as compared to silk solution and silk
tion. Not only the gelation rate, but the intrinsic hydrogel proper- films [101]. Matsumoto et al. have provided a detailed insight into
ties like mechanical properties and pore-size of silk-gels can be the gelation mechanism of silk fibroin [103]. FTIR and CD analysis
easily manipulated by controlling external conditions. It has been have shown that regenerated silk solution obtained after dialysis
seen that the breaking stress of gels prepared at 55 °C increased contains about 10–20% b-sheet fractions (depending on the con-
to about 19 N/m2 as compared to about 8 N/m2 for the gels pre- centration of protein). During initial gelation, only weak interac-
pared at 15 °C suggesting that the fibroin gels prepared at higher tions take place between the protein chains without any
temperature have better mechanical properties [102]. Similar secondary structure changes. Thus, there is no significant increase
impact of temperature on mechanical strength of gels were in the b-sheet fraction. Until this stage, silk gelation is a reversible
showed by Kim et al. who found that 16% fibroin solution, which process [111]. However, as soon as about 15% gelation is complete,
undergoes gelation at 60 °C, has double the compressive strength strong interactions follow, resulting in a continuous increase in b-
(3 MPa) than a solution of same concentration that undergoes gela- sheet content [111]. These thermodynamically stable cross-links
tion at 37 °C (1.5 MPa) [88]. This may be due to increased crystal- ultimately result in gelation of the fibroin solution. At this stage,
lization at higher temperatures. The gel strength has also been b-sheet content reaches as high as 50% [103]. Non-linear micro-
shown to vary with pH of the solution; the gels prepared at pH scopic techniques, namely photon excited fluorescence (TPEF)
7.0 have showed highest strength. Non-porous fibroin hydrogels and second harmonic generation imaging have also showed that
have been prepared by adding glycerol (30% (v/v)) to silk solution fibroin undergoes structural transformation to b-sheet as hydrogel
[107]. The gels containing glycerol are slow in losing moisture formation takes place [112].
[107], suggesting that certain agents like glycerol can modulate Factors like pH, temperature and ionic strength have been used
the moisture retention capacity of silk hydrogels. to control the gelation rate of fibroin. The ultrasensitivity of fibroin
When hydrogels are used as matrices for encapsulating cells or gelation to pH arises because of its structure [103]. Silk is an
other bioactive molecules, extremely mild methods of preparation amphiphilic protein consisting of hydrophilic N- and C-termini
are used. Any reagent or intermittent process should not be detri- and large hydrophobic domains interspersed with very short
mental to cell viability or to the activity of entrapped bioactive hydrophilic stretches. The heavy chain N-terminus, rich in acidic
agent. Sonication has been used to fabricate fibroin hydrogels amino acids is expected to have a pI of about 4.59 while C-
encapsulating cells [91,92,95] or bioactive molecules such as terminus is rich in basic amino acids and has a pI of about 10.53.
growth factors [94]. The concentration of the protein solution is On the other hand, C-terminus of the light chain is rich in acidic
crucial for such a process. Viscosity of the solution should allow groups and has a pI of about 5.06. As pH is lowered, more and more
uniform propagations of waves throughout the solution or else it carboxyl groups get protonated, reducing charge-charge repulsion
may trigger heterogeneous gelation. Uniform mixing ensures a and thus resulting in faster gelation [103]. Thus, a decrease in neg-
22 S. Kapoor, S.C. Kundu / Acta Biomaterialia 31 (2016) 17–32

Fig. 2. Approaches to improve the properties of silk hydrogels: (a) Properties of fibroin hydrogels can be modulated by varying the processing conditions, (b) Fibroin-
composite hydrogels can be prepared by combining fibroin with synthetic or natural polymers and (c) silk proteins can be genetically engineered to introduce ‘designer’
features into them.

ative charge by protonation of acidic amino acid side chains pro- improve the mechanical properties of silk hydrogels, our labora-
motes refolding of protein chains to a more ordered state that is tory combined polyacrylamide with silk to obtain hydrogels
accompanied by an exclusion of water. A similar increase in whose properties can be manipulated by changing the ratios of
hydrophobic interaction is observed when temperature and ionic the two components [115]. Our data suggested that the gels pos-
strength of fibroin solution are varied. The profound effect of tem- sessed high mechanical strength and were cytocompatible. In
perature on gelation rate of fibroin has been attributed to the other studies, Young’s modulus of polyvinyl alcohol-silk fibroin
reduction of local dielectric properties caused by temperature- 50:50 blend gels (146.7 MPa) increased three fold as compared
induced dehydration [103]. Two-Dimensional Raman Correlation to that of fibroin only gel (50 MPa) [117]. Additionally, the poros-
Spectroscopy and 13C Solid-State NMR have shown the occurrence ity of the hydrogels increased on increasing the freeze–thaw
of transition of secondary structure of silk from random coils to b- cycles, corroborating that in addition to the polymer, method of
sheet in presence of calcium ions [113]. Thus, b-sheet transition blending affects the ultimate properties of the hydrogels and thus
mainly triggers the gelation of fibroin and the conditions that have needs to be optimized [117,118]. In addition to improving the
been used to prepare fibroin hydrogels promote this transition in physical properties of the hydrogels, novel features including
silk, thus, facilitating gel formation. Lately, it has been shown that stimuli-sensitivity and reversibility of sol–gel transition have
the sol–gel transition of fibroin can be reversibly controlled even in been introduced by fabricating silk composite hydrogels. Kang
b-sheet-rich silk by regulating its self-assembly process in aqueous et al. first reported that poloxamer 407, a type of pluronic, can
solution [114]. Such reversible gelation may impart an ability to influence gelation of fibroin [119]. Pluronics are a class of non-
exert a temporal control in encapsulating molecules in hydrogels ionic tri-block copolymers consisting of a central hydrophobic
by assembly and subsequently controlling their release by disas- chain of polyoxypropylene and two terminal chains of hydrophi-
sembly of hydrogels. lic polyoxyethylene. Varying the length of chains can vary their
properties. Gelation of fibroin poloxamer system containing 3%
3.2. Silk fibroin composite hydrogels fibroin was found to be reversible as poloxamers have reversible
sol–gel properties at different temperatures due to variation in
In addition to modulating the properties of silk hydrogels by interactions between the hydrophobic and hydrophilic chains
varying external conditions as described above, other approaches [119]. Thus, incorporation of poloxamer resulted in tempe
are also used to fine tune their material properties (Fig. 2). These rature-sensitive protein hydrogels.
include producing silk fibroin hybrid hydrogels and fabricating Silk has been combined with natural polymers as well to fabri-
hydrogels using genetically engineered silk. Table 3 lists the poly- cate composite hydrogels. Mixed protein hydrogels comprising of
mers that have been used to fabricate fibroin-composite hydro- fibroin and collagen or gelatin have been prepared [127–131].
gels and the advantages of the resulting matrices. Regenerated Since gelatin hydrogels as such are not stable due to dissolution
silk fibroin has been blended or chemically cross-linked with sev- of gelatin at 37 °C, mixing of silk fibroin with gelatin proved advan-
eral synthetic polymers. For instance, in a continued effort to tageous, as the presence of silk fibroin b-sheets provided stability
S. Kapoor, S.C. Kundu / Acta Biomaterialia 31 (2016) 17–32 23

Table 3
The polymers that have been used in combination with silk fibroin protein to obtain composite hydrogels with improved properties.

Nature of Polymer Method of preparation Characterization Properties/Advantages of the resulting Reference


polymer hybrid hydrogel (s)
Synthetic Polyacrylamide Chemical cross-linking of silk and Swelling, morphological, rheological, Improved mechanical strength [115,116]
acrylamide solution mechanical properties and
biocompatibility
Polyethylene Mixing powdered PEO in silk solution Gelation kinetics, morphological and Enhanced gelation rate of silk solution [88]
oxide followed by heat treatment mechanical properties
Polyvinyl Freezing and Freeze thawing Mechanical properties and porosity Improved mechanical properties [117,118]
alcohol
Poloxamer Photocross-linking of silk and Gelation kinetics, thermal stability, Reversible sol–gel transition of silk [119,120]
poloxamer solution, Physical mechanical and swelling properties solution
blending of silk and poloxamer
solutions
Polyurethane Chemical crosslinking Mechanical, rheological properties and Injectable; improved mechanical [121]
biocompatibility properties, suitability for application in
nucleus pulposus replacement
NIPAAma Physical blending of silk and NIPAAm Swelling and rheological properties Increased deswelling kinetics of [122,123]
solution composite hydrogels as compared to
NIPAAm hydrogels
Polyethylene Physical blending of silk and PEG 300 Gelation kinetics, rheological Injectable, In situ gelling, low initial cell [124]
glycol or PEG 400 solution properties, mechanical properties, attachment, thus suitable as, anti-
degradation kinetics and cell fouling and anti-adhesion surface
attachment
Natural Gelatin Physical blending of silk and gelatin Thermal stability, viscoelastic, swelling Temperature sensitive conformational [125–130]
protein solutions, Crosslinking using and morphological properties transition of gelatin from helix to coil,
Genipin formation of stable gels
Collagen Chemical cross-linking of Thermal, viscoelastic, swelling, Improved mechanical strength, thermal [131]
fibroin/collagen solution morphological properties and stability and substrate stiffness
biocompatibility
Chitosan Chemical/physical cross-linking of Swelling properties and ion and pH pH and ion sensitive hydrogel, [132,133]
silk and chitosan solution sensitivity of gels improvement in release kinetics of
drugs
Hyaluronan Cross-linking of HA in presence of an Gelation kinetics, thermal stability, Improved mechanical properties and [134,135]
electrospinned silk mat, Physical morphology, mechanical and swelling controlled degradation
blending using ultrasonication properties, enzymatic degradation
Methylcellulose Physical cross-linking of silk and MC Morphology,drug release kinetics Control of sol–gel transition of fibroin [136]
solution at 50 °C and drug release properties
Pectin Dialysis against methanol Mechanical Properties, swelling Increased stiffness of hydrogel [137]
properties biodegradability,
biocompatibility
Alginate Calcium ion-induced gelation Porosity, intravital imaging, Mechanical properties of the hydrogel [138,139]
Rheological properties, Immune facilitated stem cell differentiation
response Supports biomimetic crystallization of
hydroxyapatite
a
NIPAAm: N-isopropylacrylamide.

to the resulting silk fibroin/gelatin (SF/G) blend hydrogels [128]. Different variations of fibroin-based covalent or non covalent
Differential scanning calorimetry and dynamic rheological analysis interpenetrating networks (IPNs) and semi-IPNs (SIPNs) have also
indicated that the presence of silk fibroin b-sheets increased the been synthesized. Interpenetrating networks are materials com-
dynamic elastic modulus of the G/SF hydrogels, which in turn sta- posed of two polymers, each existing as an independent network
bilized them at higher temperatures [128]. Interestingly, blended [140]. The two networks are polymerized or cross-linked such that
hydrogels have been found to be stimuli-sensitive with the swel- the resultant networks are interlocking. Combining two networks
ling of hydrogel and dissolution of gelatin from hydrogels being provides an opportunity to create composite materials that other-
higher at 37 °C compared to that at 20 °C [128]. Collagen-fibroin wise cannot be obtained by mixing polymers due to phase separa-
composite hydrogels have been prepared by cross-linking collagen tion. IPNs result in unique properties, which are not present in the
and fibroin by using optimized amounts of 1-ethyl-3-(3-dimethyla individual polymer networks [141,142]. Semi IPNs of silk fibroin
minopropyl)carbodiimide hydrochloride (EDC) [131]. In addition and modified poloxamer 407 having an acrylate-terminated poly-
to improving the mechanical strength and thermal stability, the ethylene oxide (PEO) derivative, modified polyethylene glycol
gel exhibited gel-to-sol transition in medium of pH 4.0. The rever- (PEG) macromer and PEG have also been prepared [120,143,144].
sible gelation characteristic can be attributed to an increase in The SF (silk fibroin)/PEG SIPN has been prepared using a pho-
electrostatic repulsion between protein chains with decrease in topolymerization method. The SIPN hydrogels have higher com-
pH value [131]. Hyaluronan (HA), a major glycosaminoglycan of pression resistance and mechanical strength as compared to SF
the extracellular matrix, has also been used together with fibroin or polymer (PEG/poloxamer) hydrogels. The increase in strength
to fabricate a composite hydrogel [134,135]. HA solution has makes SIPN hydrogels desirable for biomedical applications, espe-
chemically been cross-linked using poly(ethylene glycol)- cially skin grafting and wound dressing [143]. Our lab has synthe-
diacrylate in presence of a prefabricated electrospun mesh of sized photo-crosslinked PVA/SF hydrogels. PVA methacrylate,
fibroin [134] or has physically been blended with fibroin through obtained by reacting PVA with 2-isocyanatoethyl methacrylate,
ultrasonication [135]. The presence of fibroin acts as mechanical was freeze dried. The resuspended solution was blended with
reinforcement and slows degradation of the otherwise mechani- fibroin and blend solutions were photopolymerized giving rise to
cally weaker, fast degrading HA hydrogel. semi-IPNs [117]. Using a similar approach, Xiao et al. fabricated
24 S. Kapoor, S.C. Kundu / Acta Biomaterialia 31 (2016) 17–32

protein IPNs where gelatin methacrylate and SF have been inde- Not only the proteins have been obtained in larger quantities
pendently crosslinked in the interpenetrating network [130]. Gela- but also, new features, which are not found in native proteins, have
tin methacrylate has been polymerized by UV-irradiation in the been incorporated [149]. The main functional modalities that have
presence of Irgacure 2959, a photoinitiator and the SIPN has subse- been incorporated in these genetically-engineered proteins are ori-
quently been exposed to methanol to introduce cross-links in SF. ented towards making the proteins self assembling, stimuli-
Introducing SF improved the mechanical properties of hydrogels sensitive, mechanically stable and information rich so that they
and it has been seen that exposing SF to methanol further contain specific epitopes and domains that assist in cell adhesion
enhanced the strength of hydrogels [125]. In yet another study, and signalling. Incorporation of these functionalities ultimately
Gil et al. have made SIPN consisting of poly(N-isop aims to make the genetically engineered protein-based hydrogels
ropylacrylamide) (PNIPAAm) and fibroin [122]. PNIPAAm is one more suitable for tissue engineering and drug delivery purposes
of the most thoroughly investigated thermoresponsive polymers. [165,166].
It shows a sharp phase transition when the temperature is
increased above or decreased below its lower critical solution tem- 3.3.1. Silk-elastin like hydrogel
perature i.e. 32 °C [145]. This type of stimuli-sensitive polymer has Elastin-like proteins have gained considerable attention due to
many applications in devices that require an on–off mechanism. their striking self-assembling properties, attributed mainly to the
The major drawback of this system is its low de-swelling rate transitions that take place in the polymer backbone in the presence
due to formation of a skin layer [146]. Various strategies have been of water. Thorough details about elastin-like proteins are provided
suggested for improving the deswelling kinetics of PNIPAAm [147]. elsewhere [167–169]. Silk-elastin like polymers (SELPs), developed
Introduction of fibroin in PNIPAAm has been shown to alter its by Cappello and colleagues, constitute an important class of genet-
microstructure, leading to an increase in its de-swelling rate that ically engineered silk proteins [170]. These synthetic proteins con-
results from reduced phenomena of skin layer formation in the sist of (a) repetitive silk peptide sequence of B. mori (Gly-Ala-Gly-
gels, while maintaining the swelling rate [122]. To further improve Ala-Gly-Ser) that provides mechanical strength to block copolymer
the swelling/deswelling rates of these composite hydrogels, a protein and (b) elastin peptide sequence (Gly-Val-Gly-Val-Pro) that
novel strategy has been used [123]. The composite hydrogels have provides flexibility to the protein, improves its solubility and
been lyophilized and the lyophilized matrix has been exposed to determines the cross-link density within the hydrogels [170,171].
methanol. Freeze-drying introduces temporary macropores in the Varying the composition of the two constituent peptide blocks pro-
hydrogels, while exposing the matrix to methanol introduces b- vides a means to regulate the properties including strength,
sheets in SF that assists in maintaining the macroporous structure immunogenicity, solubility and degradation rate of the resulting
of hydrogels even after swelling [123]. These examples clearly sug- biomaterial [172]. The silk-elastin like polymers have been well
gest that several of the limitations associated with parent polymers characterized to determine their properties and their utility for
can be overcome and novel properties, which do not exist in the various applications. The general techniques for making and char-
individual parent polymers, can be introduced by preparing fibroin acterizing these polymers have been reviewed in detail [170–174].
hybrid hydrogels. It should be noted, however, that when combin- Certain members of this family of proteins, called Prolastins, exhi-
ing silk with other polymers and varying the relative amount of the bit sol-to-gel transition and hydrogel forming capabilities [175].
two polymers in the combination, method of blending and cross- They mainly consist of two or more silk-like sequences per mono-
linking density may simultaneously affect many properties includ- mer. Hydrogel formation is primarily regulated by temperature
ing strength, porosity, swelling, biocompatibility and morphologi- (due to dependence of assembly of elastin-like blocks and hydro-
cal properties [120,122,139]. Therefore, while fabricating a hybrid gen bonding of silk-like blocks on temperature). Under physiolog-
hydrogel, it is essential to ensure that an attempt to improve a par- ical conditions, the hydrogen bond formation between silk-like
ticular aspect of hydrogel does not affect other physical or biolog- units causes protein solution to undergo crystallization, which
ical properties adversely. results in gelation [175]. The major advantage is that no chemical
cross-linking or solvent treatment is needed to prepare SELP
hydrogels. Thus, SELP hydrogels present novel opportunities to
3.3. Hydrogel fabrication from genetically engineered silk encapsulate bioactive molecules or cells under ambient conditions
and form in situ gelling hydrogels for tissue engineering and ther-
Advancement in biochemistry and molecular biology of silk at apeutic purposes.
genetic and protein levels have provided a strong platform for SELP-47K and SELP-415K, consisting of four silk-like and seven
genetic engineering of silk proteins (Fig. 2). One of the unparalleled and fifteen elastin-like blocks, respectively, are main examples of
strengths of genetic engineering of proteins is the ability to pre- gel-forming proteins. SELP-47K, which undergoes irreversible sol-
cisely customize the structure and function according to specific to-gel transition at body temperature, is the most extensively
needs. Thus, we can have biomaterials with desirable properties investigated gel-forming Prolastin polymer [173–178]. Swelling
by utilizing tremendous chemical diversity available in the amino characteristics and equilibrium water content are important
acid building blocks [148]. Another reason for cloning silk genes is parameters of the matrix and primarily govern the release proper-
to obtain larger quantities of protein in a cost effective way. ties of the molecules from hydrogels. Hydrogel swelling character-
Although protein can be obtained in larg amounts from silkworms istics are, in general, controlled by factors like polymer
like B. mori and spiders, the total availability of protein is not suf- concentration and cross-link density as well as environmental
ficient considering the amounts required for various applications. stimuli such as pH, temperature, and ionic strength of solvent.
Silk-like proteins have been obtained by creating variants of the However, in case of SELP-47K, the swelling of the gels is relatively
repeat sequences of N. clavipes and B. mori. Continuous efforts in independent of temperature, pH and ionic strength, suggesting
this direction have resulted in successful production of recombi- that this polymer can be utilized to make hydrogels whose release
nant silk proteins [149–156]. Various strategies and host systems properties remain constant in spite of external variations [35].
including insect cells [157], E. coli [158], microbes [159,160], mam- Although hydrogels which maintain similar swelling properties
malian cells [161], goats [162] and potato and tobacco plants under different conditions are robust, there may be conditions
[163,164] have been tried to optimize the yield of protein, to where it is desirable to have hydrogels with stimuli-sensitive swel-
improve the solubility of protein and to allow proper folding and ling and release properties. Swelling properties of SELP-415K have
post translational modification of protein. been found to be sensitive to temperature and ionic strength [179].
S. Kapoor, S.C. Kundu / Acta Biomaterialia 31 (2016) 17–32 25

Physiologically relevant sensitivity to pH- and temperature has The real challenge yet lies in replicating their structural and func-
been introduced in these polymer gels by strategic replacement tional properties according to the desired tissue type where the
of valine in elastin chain with glutamic acid (a charged amino acid) hydrogel is to be implanted and in creating the dynamic chemo-
through genetic engineering techniques [180]. Dandu et al. have mechanical environment that exists in vivo.
prepared hydrogels from another silk-elastin like protein (SELP-
815K) and have shown that the silk sequences provide mechanical 4.1.1. Bone tissue engineering
stiffness to the hydrogels whereas the elastin sequences determine Bone, which mainly consists of collagen protein and hydroxya-
the cross-link density within the hydrogels [171]. Recently Silk- patite, provides support and strength for the movements. Critical
Elastin like proteins with matrix metalloproteinase (MMP) degrad- bone defects or severe bone injuries, which cannot be regenerated
able sequences inserted in between the polymer backbone have naturally, may need external interventions in the form of implants.
been expressed. The degradation behavior of the hydrogels fabri- Implants provide mechanical strength to bear stresses at the site of
cated from this recombinant protein can evidently be finely con- injury and milieu that accelerate natural tissue growth. Bone tissue
trolled by environmental cues like the expression of MMPs, engineering is an approach, which differs from permanent bone
which have been found to be overexpressed in certain tumors implants in a sense that the tissue engineered bone is designed
[150,181]. Recently, in situ gelling injectable SELP-47K and SELP- to integrate and absorb in the environment where it is implanted.
815K based-hydrogels have been investigated for formulating For this, the implant should be biodegradable and bioresorbable
transarterial embolics to selectively prevent tumoral blood supply [188,189]. The major challenge for successful bone tissue engineer-
as a novel strategy for chemotherapeutics [178]. A 12% w/w solu- ing lies in understanding the spatial and temporal distribution of
tion of SELP-815K fabricated into hydrogel by shearing has been cells and growth factors and their interaction with extra cellular
found to exhibit acceptable rheological properties and embolic matrix for osteogenesis in diseased conditions. In addition, the
capability [178]. Although, the research about SELP hydrogels is implant material should provide mechanical, chemical and struc-
still in a budding stage, it is clear that hydrogels from silk- tural signals for osteoconduction and osteoinduction, essential
copolymers can be made with varied mechanical properties, degra- for in vivo tissue growth. Several materials have been investigated
dation signals, assembly characteristics and bioactive signals, mak- for bone tissue engineering [188,189]. Metals have been the tradi-
ing them useful for future biomedical applications. tional choice as implant material due to their load bearing capabil-
ity. However, they suffer from several disadvantages like the stress
shielding effects, poor biodegradability, and lack of integration
4. Biomedical applications of silk-based hydrogels with the surrounding tissue [190]. Ceramics, though resemble to
the mineral phase of bone, are severely limited in application by
4.1. Tissue engineering their brittle nature and poor biodegradation properties [191].
Amongst natural polymers, collagen-based materials offer oppor-
Engineering the polymeric materials to replace/repair a mal- tunity to influence cellular responses but often exhibit poor
functioning or damaged tissue or organ is one of the most impor- mechanical strength and undesirable immune responses [192].
tant advances in the recent decades [182]. The success of Biomaterials fabricated from alginate have adequate mechanical
hydrogels as tissue engineering matrices lie in the fact that their properties, however, they have limitation for different cell-based
biochemical composition and properties such as water content, applications due to lack of cell-specific bioactivities [193]. To
viscoelasticity and mechanical strength can be made to mimic dif- address some of these challenges, silk implant material has been
ferent types of natural tissues. Controlling the factors like mono- evaluated for bone regeneration.
mer concentration, cross-linker concentration and nature of Silk proteins can be tailored to make a bioactive material for
functional groups during processing can optimise the properties bone implants to induce direct bone formation and ultimately
of hydrogels. Similarity of hydrogels to natural tissues and their result in osteointegration, although much work still needs to be
ability to support functions like transport of bioactive molecules done in this direction. Silk fibroin hydrogels have shown superior
such as hormones, growth factors and peptide sequences while ability to promote cell metabolism as well as bone remodelling
maintaining structural integrity make them attractive for tissue as compared to poly(lactide-co-glycolic acid) (PLGA), an FDA
engineering applications [183]. Another advantage of using hydro- approved polymer, which is a popular choice for comparing the
gels for tissue engineering applications is the ability to entrap cells characteristics of a material for tissue engineering applications
within hydrogels during fabrication, provided that mild conditions because of its long clinical standing, favorable biodegradation
are used for preparation so that the cell viability is not affected properties, biocompatibility and a minimal systemic toxicity
[184]. Cell entrapment during gelation results in a more uniform [193,194]. The bone-healing rate, proliferation and differentiation
distribution of cells, as compared to populating the matrix after of the osteoblasts in presence of silk hydrogels have also been bet-
gelation, which results in higher cell populations near the surface ter than the control [37]. In another study, porous silk matrix (pre-
of the gel [185], though considerable success has been achieved pared by using hexafluoroisopropanol (HFIP) as solvent) was
with the latter strategy as well [186]. Thus, the aqueous environ- seeded with stem cells derived from human bone marrow and
ment, ease of transportation of nutrients and entrapped molecules, the cells have been allowed to grow under osteogenic conditions
ease of modification and utility as in situ forming matrices repre- with medium being supplemented with ascorbic acid-2-
sent important advantages of hydrogels as tissue engineering phosphate, dexamethasone, b-glycerolphosphate, and BMP-2
matrices. The disadvantages may include difficulty in handling, [195]. Various approaches including (1) tissue engineered bone
sterilization and lack of mechanical strength [187]. Three main (cells seeded on matrix grown in bioreactor for 5 weeks), (2) scaf-
design approaches to use hydrogels as functional matrices include folds seeded with cells at the day of surgery, (3) scaffold alone, or
preparing acellular hydrogels, cell-laden hydrogels and tissue- (4) no implant (unfilled) were evaluated by implanting in mouse.
engineered hydrogels, which, in addition to the polymeric scaffolds Clearly visible trabecular structures could be observed through
and cells, contain several other factors like chemical moieties and/ micro-computed tomography after about 5 weeks of implantation
or bioactive molecules such as growth factors and peptide in case of the defects treated with the engineered bone, suggesting
sequences to modulate degradation/remodelling of matrix, cell that silk based implants can be utilised for effective bone regener-
adhesion, cell motility, cell growth and differentiation [187]. Silk ation. It is worth noting that the scaffolds prepared from protein
hydrogels based on all these approaches have been produced. obtained from the same source but differing in the preparation
26 S. Kapoor, S.C. Kundu / Acta Biomaterialia 31 (2016) 17–32

method exhibit different osteogenic responses [196]. Differences in Bioreactors can be used to stimulate functional regeneration of
bone related outcomes such as mineralization and expression of tissues and to continuously supply the nutrients and growth fac-
collagen type I, alkaline phosphate, osteopontin and matrix metal- tors to the cultured cells. This represents an advanced approach
loproteinase 13, which regulate the remodelling of ECM have also holding immense potential for improving cartilage tissue engineer-
been observed [196]. The differences in the processing of protein ing. This approach has been used with fibroin hydrogel [201]. The
lead to subtle changes in structure and properties of resulting cells grown in matrix homed within a stirring chamber have been
matrix, which ultimately lead to different cell response [197]. found to exhibit higher DNA and glycosaminoglycan content as
Recently, fibroin/sodium alginate hydrogels have been used as compared to the cells not grown in the reactor. Positive histological
template for promoting controlled biomineralization of hydroxya- staining of proteoglycans and collagen in tissue grown in this sys-
patite crystals for bone repair [139]. The preliminary success of tem vis-à-vis the one in a system lacking stirring chamber sug-
hydrogels derived from silk fibroin suggests that further studies gested that stirrer facilitated the maturity of cartilage matrix by
are warranted. Incorporation of additional biological signals as providing mechanical stimulation to the cellular microenviron-
well as utilization of polymer blends, interpenetrating networks ment. This system could support chondrogenesis in vivo and
and silk copolymers may provide ideal scaffolds for bone repair. resulted in cartilage regeneration in the rabbits corroborating its
clinical potential [201].
Primary cell lines and chondrocytes are mainly used as cell
4.1.2. Cartilage tissue engineering source for cartilage tissue regeneration. Since they are available
Cartilage, a connective tissue, is mainly composed of type II col- in limited supply, scientists are now focusing on the use of stem
lagen and is rich in proteoglycans, particularly chondroitin sul- cells that show unlimited potential to differentiate in several tissue
phate while the primary cell type is chondrocytes [198]. This types including cartilage [202,203]. Human mesenchymal stem
tissue contains almost 65–70% water. The clinical impact of defects cells (hMSC) encapsulated in SELP-47K hydrogel and cultured in
in cartilage tissue due to congenital abnormalities or trauma is of chondrogenic medium in the presence of TGF-b3 exhibited differ-
particular concern due to the low regenerative capability of this entiation and chondrogenesis [204]. The cells remained metaboli-
tissue, resulting from an absence of blood vessels, nerve tissue, cally active even after 28 days of culture and histological
and lymphocytes and the low cell density, slow cell proliferation analyses have revealed the formation of extracellular matrix along
and slow matrix turnover rate. Therefore, cell-based therapies, in with the expression of SOX9, and matrix proteins aggrecan and col-
which 3D scaffolds carrying stem cells or differentiated chondro- lagen. The studies indicate that fibroin hydrogels have potential for
cytes can be implanted at the site of injury, are of significant inter- mimicking the extracellular matrix and promoting cartilage
est. Due to their close resemblance to cartilage tissue, hydrogels regeneration.
have been considered as a suitable matrix for encapsulating cells
and the growth factors (e.g. TGF-b superfamily, IGF, FGF, BMP, 4.2. Controlled release
PDGF, and EGF). Natural materials like collagen and synthetic
material like poly(lactic acid) (PLA) and PLGA have been evaluated 4.2.1. Controlled drug delivery
for this purpose. Though agarose hydrogels have particularly Controlled release implies that the delivery of the active agent
shown utility as scaffolds for cartilage tissue engineering because should follow a predetermined course of release. It may consist
of their high mechanical strength, their success is limited by poor of both sustained (for prolonged duration) and targeted (localized)
biocompatibility, lack of biodegradability and poor host tissue inte- release. Supplying essential amounts of active molecule at a partic-
gration [91]. The mechanical strength, frictional properties and the ular area for required duration is a major challenge in the field of
uncertainty of interaction between the matrix and the cells have drug release. This is more so in the case of bioactive agents like
remained the major issues with most of the scaffolding materials hormones and peptides, which are highly selective in their action
[91]. Aoki and colleagues have prepared fibroin hydrogel and com- and are sensitive to processing treatments. Silk based hydrogels
pared its cartilage regeneration performance with collagen gels have been found to be suitable for controlled release. One of the
[33]. They inoculated the chondrocytes isolated from the proximal first studies investigating silk fibroin for controlled release has
humerus, distal femur and proximal tibia of 4-week-old Japanese not used a hydrogel but a membrane. In this early study, Chen
white rabbits into the fibroin-hydrogel sponge, formed by phase et al. examined the permeation of pharmaceuticals like 5-
separation of freezed fibroin solution and collagen gels [33]. The fluorouracil, L-(+)-ascorbic acid, resorcinol, sodium phenolsul-
cells were then cultured for 4 weeks and were found growing in fonate and benzyltrimethylammonium chloride through the
the pores and the outer surface of the fibroin hydrogel and the cell fibroin membrane [205]. They proposed that since fibroin mem-
density increased with incubation times. On the other hand, the brane consists of weak basic and acidic groups, it can act as an
cell density in the collagen gels was seen to be higher initially, amphoteric ion-exchanger and controlling the pH may control
but it did not increase with time. Similarly, the rate of increase the passage of molecules through it. Since then fibroin has been
of chondroitin sulfate in fibroin hydrogel was higher than that in considered as pH responsive drug delivery material. Hanawa
collagen gels [33]. Positive histological staining for key cartilage et al. have showed that the release behaviour of benfotiamine, a
extracellular matrix components (chondroitin sulfate and type II synthetic derivative of thiamine (vitamin B-1), in the glycerol con-
collagen) indicated formation of hyaline cartilage-like tissue in taining fibroin hydrogels is inversely related to the fibroin content
the pores of the fibroin hydrogel. Chondrocytes maintain round in the matrix [206]. This is likely due to the increased number of
morphology and retain their differentiated phenotype within the interchain interactions at higher protein concentration, which
fibroin hydrogel [33]. Cell microaggregates seeded in fibroin resulted in smaller pore diameters and hence slower release. Not
hydrogel closely mimic the initial stages of tissue formation and only the protein concentration but molecular weight of silk protein
have been found to be very efficient in forming extracellular matrix affects the release rate as well [36]. High molecular weight pro-
for cartilage tissue regeneration [199]. To improve the mechanical teins (76 kDa) have been shown to effectively slow down the
properties of fibroin hydrogels, recently a novel approach has been release of buprenorphine in comparison to low molecular weight
used, wherein silk hydrogel has been reinforced with silk microfi- silk protein (18 kDa) when used as matrix [36]. Our laboratory
bers. The composite hydrogel has exhibited mechanical properties has evaluated fibroin/polyacrylamide SIPN hydrogels for drug
comparable to that of the agarose hydrogels with proven mechan- release [115]. Two model compounds namely trypan-blue and
ical robustness [200]. FITC-inulin have been used to determine the effect of physico-
S. Kapoor, S.C. Kundu / Acta Biomaterialia 31 (2016) 17–32 27

chemical properties of hydrogels on release of molecules. Our viral vectors from MMP responsive-SELPs has been evaluated in a
results have indicated that these composite hydrogels were suit- mice model of head and neck squamous cell carcinoma for tumor
able for sustained release of low and high molecular weight mole- therapy [181]. The rate of release of viral vectors has been found
cules alike. Recently, silk hydrogels have shown potential for to be dependent on the expression of MMPs, frequently found to
sustained release of bevacizumab, a clinically used anti-VEGF ther- be overexpressed in several tumors [181]. Such a system may
apeutic for certain metastatic cancers [207]. The controlled release enable localized delivery of bioactive therapeutics to tumors
of a chemotherapeutic over a long period may eventually reduce specifically, thus limiting systemic host toxicity.
the dosing frequency, which increases patient compliance.
As noted above, specific Prolastins are important particularly as
in situ gel forming delivery systems [169,175,178]. They sponta- 5. Challenges and future prospects
neously form a gel over a period of time after being injected via fine
gauge hypodermic needles [175]. Cappello et al. have characterized Silk proteins represent interesting polymeric biomaterial
the release of many fluorescently labelled model compounds of dif- because of their mechanical properties, thermal stability, biocom-
ferent molecular weights and a model pharmaceutical protein pan- patibility, and possibility of control via genetic engineering. Several
tarin from Prolastins of different peptide sequence compositions examples mentioned in this work elucidate the potential of using
[175]. The rate of release of entrapped molecules has been fibroin for producing hydrogels. A wide range of applications
observed to depend on the composition as well as the concentra- including controlled release of active molecules and tissue engi-
tion of the recombinant Prolastin. Moreover, the entrapped thera- neering have also been described (Fig. 3). Advances in fabrication
peutic protein retained the bioactivity after being released from methods, ability to process and sterilize, control of properties like
gels [175]. The interactions between the entrapped molecule and degradation and strength, and possibility of encapsulating cells
protein matrix might affect the solute partitioning and hence the have made silk hydrogels suitable constructs for complex tissue
release profiles of molecules. This has been shown in the case of regeneration.
cytochrome c, Vitamin B12 and theophylline release from SELP Nevertheless, there are limitations that need to be overcome to
hydrogels [35]. explore complete potential of silk hydrogels. A greater understand-
ing of the fundamentals underlying structure–function relation-
4.2.2. Controlled release of DNA for gene therapy ship of these proteins is needed to control the properties
There has been a tremendous increase in understanding the according to the specific requirements of design criteria. As more
implications of the diseases caused due to genetic mutations. specialized hydrogels are made for advanced applications, rigorous
Hence, a lot of effort is being placed on replacing abnormal genes and accurate mathematical modelling will be required to describe
with the correct ones. Successful gene delivery remains the key the systems and the mechanisms associated with them. Fig. 4 out-
for gene therapy. Targeting the cells, controlled activation and lays the novel possibilities with silk hydrogels. In situ gelling
expression of the genes and getting rid of the side effects remain hydrogels can assist in overcoming the invasive implantation of
major challenges for success in this endeavour. Safe and effective hydrogels for tissue reconstruction, thus, increasing the patient
delivery systems are thus the main clinical concerns. SELP poly- compliance. Self-assembling silk-like and silk-elastin like proteins
mers have recently been evaluated for DNA delivery for tumor provide huge opportunities to design in situ forming gels under
therapy [179,181,208–211]. The ease of sol-to-gel transition of mild conditions but we need to incorporate simultaneously
these block copolymers enables the loading of DNA or viral parti- ‘‘smartness (stimuli-sensitive response character)” in these pro-
cles into an aqueous solution, which may be injected by minimal teins. The ‘decision making’ silk hydrogels that can be programmed
invasive procedures to form solid matrix in and around the tumour to respond to physical, biochemical and mechanical cues will
at physiological temperature. Megeed et al. have studied the enable temporal control over several functions including the
release of plasmid DNA from SELP-47K hydrogels [208]. Lysine release of the encapsulated bioactive agents.
(thirteen) and arginine (five) residues present in SELP-47K are pro- Consistent with several other natural and synthetic polymers
tonated at pH 7.4, and can thus interact with the negatively and as described herein, silk hydrogels support encapsulation of
charged phosphate backbone of DNA and affect its release profile. cells and therapeutic molecules. They are porous to allow diffusion
Ionic strength, concentration of the SELP polymer and time for cur- of nutrients and wastes as well as to allow intercellular communi-
ing the gels have also been found to influence the release rates cation with secreted molecules. However, they lack many of the
[208]. In addition, the conformation (supercoiled, open circular or signals of the native extracellular matrix (ECM). Included in these
linear) and molecular weight of the DNA significantly influence signals are insoluble bioactive cues stemming from the proteins,
the release profiles while the concentration of DNA (within a range glycoproteins, and proteoglycans and soluble signals such as
of 50–250 mg/ml) has no impact [209]. Finally, gene expression cytokines and growth factors that are secreted by the ECM. Future
analysis showed that the released DNA retains its bioactivity even studies will need to carefully consider incorporation of biological
after 28 days of release [209]. Further, SELP matrices have been signals to improve tissue regeneration. We should gain guidance
found to be suitable for localizing and prolonging the release of from the increasing knowledge regarding the importance of gly-
adenovirus and viral transduction in murine models of breast cosaminoglycans and proteoglycans in tissue homeostasis and dis-
and head and neck tumour xenografts [210]. Haider et al. have cre- ease. Studies investigating blends of silk fibroins and other
ated a new matrix, SELP-415K, having more elastin units for gene polymers in the form of interpenetrating networks and semi-
delivery purpose. The release rate of DNA from this polymer matrix interpenetrating networks and even as block-co-polymers begin
increased with an increase in the number of elastin units [179]. to move toward materials that better mimic the native ECM
Recently, the release behaviour of incorporated plasmid has been [95,105,139,212]. These blends show improved mechanical charac-
studied as a function of biodegradation of the SELP-47 and SELP- teristics and in some cases include cell adhesion domains. Through
415K hydrogels in presence of elastase enzyme [211]. In case of incorporation of key features of proteoglycans, the ‘information
both the hydrogels, the release of DNA is much faster when the rich’ silk hydrogels can better mimic the native ECM and will also
release medium contains elastase as compared to controls. More- contain natural binding sites for cytokines and growth factors that
over, higher number of elastin blocks in protein resulted in a faster are secreted by the regenerating engineered tissue.
elastase-induced degradation of the matrix, and loss of integrity of An interesting feature that has not yet been thoroughly
hydrogel, thus, resulting in faster DNA release [211]. The release of explored in case of the silk hydrogels is the multilayered hydrogels.
28 S. Kapoor, S.C. Kundu / Acta Biomaterialia 31 (2016) 17–32

Fig. 3. Applications of silk hydrogels: Silk fibroin based hydrogels are currently being explored for tissue engineering and controlled release of therapeutic molecules.

Fig. 4. Some future directions for silk hydrogels: (a) Silk hydrogels that undergo spontaneous gelation in physiological conditions at the site of injection will pave way for
their non-invasive clinical applications in tissue engineering and drug release, (b) Multilayered hydrogels consisting of separate silk gel layers with distinct properties will
enable ‘true’ compartmentalization of the matrix. Such compartmentalized multilayered hydrogel can be used for carrying different ‘payloads’ to achieve spatiotemporally
complex release profiles, (c) Integration of biomimetic signals on silk hydrogel surface will enable the ‘information rich’ hydrogels to be targeted to specified regions where
they can replicate the cellular microenvironment and facilitate signal dependent cell functions, (d) Multifunctional hydrogels whose properties are optimized for performing
different functions simultaneously (for instance, one that supports cell growth and release of growth factors) can play better role in regenerative medicine, (e) Stimuli-
sensitive silk hydrogels that respond to physiologically relevant environmental signals will be used as ‘Smart’ decision making biomaterials.
S. Kapoor, S.C. Kundu / Acta Biomaterialia 31 (2016) 17–32 29

Instead of hydrogels with bulk properties, novel hydrogels that [9] J.E. Gagner, W. Kim, E.L. Chaikof, Designing protein-based biomaterials for
medical applications, Acta Biomater. 10 (2014) 1542–1557.
contain multiple layers can be prepared from silk proteins in near
[10] O.D. Krishna, K.L. Kiick, Protein- and peptide-modified synthetic polymeric
future. Layer by layer assembling techniques can be utilized for biomaterials, Biopolymers 94 (2010) 32–48.
this purpose. The properties of each layer can be tailored to provide [11] D. Sengupta, S.C. Heilshorn, Protein-engineered biomaterials: highly tunable
a hierarchical organization suitable for distinct functions. Such tissue engineering scaffolds, Tissue Eng. Part B Rev. 16 (2010) 285–293.
[12] A. Heymer, D. Haddad, M. Weber, U. Gbureck, P.M. Jakob, J. Eulert, U. Notoh,
hydrogels will help realizing true compartmentalization in bioma- Iron oxide labelling of human mesenchymal stem cells in collagen hydrogels
terials that is needed to achieve biologically relevant spatio- for articular cartilage repair, Biomaterials 29 (2008) 1473–1483.
temporal complexity. Different cell types or different molecules [13] J.B. Phillips, Building stable anisotropic tissues using cellular collagen gels,
Organogenesis 10 (2014) 6–8.
can be encapsulated in distinct layers to enable complex release [14] M. Placzek, Tissue recombinations in collagen gels, Methods Mol. Biol. 461
patterns. In addition, gradients of the release molecules can be (2008) 325–335.
achieved. Several multilayered hydrogel systems have been [15] A.L. Plant, K. Bhadriraju, T.A. Spurlin, J.T. Elliott, Cell response to matrix
mechanics: focus on collagen, Biochim. Biophys. Acta 1793 (2009) 893–902.
described recently with natural and synthetic polymers [213– [16] Y. Tabata, M. Miyao, M. Ozeki, Y. Ikada, Controlled release of vascular
215]. Recently, hydroxyapatite nanoparticle containing silk hydro- endothelial growth factor by use of collagen hydrogels, J. Biomater. Sci.
gels with superior bone regenerating potential have been fabri- Polym. Ed. 11 (2000) 915–930.
[17] Y. Kimura, Y. Tabata, Controlled release of stromal-cell-derived factor-1 from
cated [216]. The challenge in getting multilayered silk hydrogels gelatin hydrogels enhances angiogenesis, J. Biomater. Sci. Polym. Ed. 21
will be to find novel preparation methods that allow the genera- (2010) 37–51.
tion of multiple layers. It is needed to make sure that the distinct [18] M. Konishi et al., In vivo anti-tumor effect through the controlled release of
cisplatin from biodegradable gelatin hydrogel, J. Control. Release 92 (2003)
layers remain separate. Another challenge will be to better under-
301–313.
stand the properties of boundaries and the interface between each [19] J.Y. Lai, Y.T. Li, Functional assessment of cross-linked porous gelatin hydrogels
layer. for bioengineered cell sheet carriers, Biomacromolecules 11 (2010) 1387–1397.
The utilization of silk proteins for making hydrogels for novel [20] V. Rattanaruengsrikul, N. Pimpha, P. Supaphol, Development of gelatin
hydrogel pads as antibacterial wound dressings, Macromol. Biosci. 9 (2009)
applications in diagnostics including synthesis of microdevices, 1004–1015.
microchannels and micropatterning for guided cell proliferation [21] K.R. Stevens, N.J. Einerson, J.A. Burmania, W.J. Kao, In vivo biocompatibility of
and differentiation as well as for targeted release of molecule gelatin-based hydrogels and interpenetrating networks, J. Biomater. Sci.
Polym. Ed. 13 (2002) 1353–1366.
remains to be explored. In addition to fabricating hydrogels from [22] M. Yamamoto, Y. Ikada, Y. Tabata, Controlled release of growth factors based
alternative sources of silk proteins from non-mulberry silkworms on biodegradation of gelatin hydrogel, J. Biomater. Sci. Polym. Ed. 12 (2001)
including A. mylitta [217], A. assamensies, A. pernyi, Samia recini 77–88.
[23] M.N. Asmani et al., Three-dimensional culture of differentiated endometrial
and spider silks. The efforts are being made to improve the quality stromal cells to oligodendrocyte progenitor cells (OPCs) in fibrin hydrogel,
of the silk proteins by genetic engineering. As we progress towards Cell Biol. Int. 37 (2013) 1340–1349.
finding new techniques to observe as well as control microscopic [24] A. des Rieux, A. Shikanov, L.D. Shea, Fibrin hydrogels for non-viral vector
delivery in vitro, J. Control. Release 136 (2009) 148–154.
morphological features, it can be anticipated that silk-based hydro- [25] T.C. Gamboa-Martínez, V. Luque-Guillén, C. González-García, J.L. Gómez
gels will enable the development of new therapeutic approaches Ribelles, G. Gallego Ferrer, Crosslinked fibrin gels for tissue engineering: two
and help us to meet the demands in regenerative medicine. approaches to improve their properties, J. Biomed. Mater. Res. A 103 (2015)
614–621.
[26] M. Guthold et al., Comparison of the mechanical and structural properties of
fibrin fibers with other protein fibers, Cell Biochem. Biophys. 49 (2007) 165–
Acknowledgements 181.
[27] H. Hall, Modified fibrin hydrogel matrices: both, 3D-scaffolds and local and
This work is supported by start-up grant from University Grant controlled release systems to stimulate angiogenesis, Curr. Pharm. Des. 13
(2007) 3597–3607.
Commission, Government of India to SK. We appreciate Ms Sarani [28] P. Lei, R.M. Padmashali, S.T. Andreadis, Cell-controlled and spatially arrayed
Ghoshal for taking interest during the initial stages of this write up. gene delivery from fibrin hydrogels, Biomaterials 30 (2009) 3790–3799.
We are greatly indebted to Dr. Alyssa Panitch, Weldon School of [29] G.M. Peretti, J.W. Xu, L.J. Bonassar, C.H. Kirchhoff, M.J. Yaremchuk, M.A.
Randolph, Review of injectable cartilage engineering using fibrin gel in mice
Biomedical Engineering of Purdue University for critical reading, and swine models, Tissue Eng. 12 (2006) 1151–1168.
excellent scientific inputs and suggestions for the improvement [30] A. Fathi, S.M. Mithieux, H. Wei, W. Chrzanowski, P. Valtchev, A.S. Weiss, F.
of the manuscript. SCK’s laboratory has financially been supported Dehghani, Elastin based cell-laden injectable hydrogels with tunable gelation,
mechanical and biodegradation properties, Biomaterials 35 (2014) 5425–
by Indo Australia Biotechnology Fund, Department of Biotechnol-
5435.
ogy and its Bioinformatics facility, Indo-Russia Biotechnology Pro- [31] D.W. Lim, D.L. Nettles, L.A. Setton, A. Chilkoti, In situ cross-linking of elastin-
gramme, Department of Science and Technology, Indian Council of like polypeptide block copolymers for tissue repair, Biomacromolecules 9
Medical Research, Govt of India, and Indo US Science and Technol- (2008) 222–230.
[32] M.K. McHale, L.A. Setton, A. Chilkoti, Synthesis and in vitro evaluation of
ogy Forum, New Delhi. enzymatically cross-linked elastin-like polypeptide gels for cartilaginous
tissue repair, Tissue Eng. 11 (2005) 1768–1779.
[33] H. Aoki, N. Tomita, Y. Morita, K. Hattori, Y. Harada, M. Sonobe, Culture of
References chondrocytes in fibroin–hydrogel sponge, Biomed. Mater. Eng. 13 (2003)
309–316.
[34] T. Diab, E.M. Pritchard, B.A. Uhrig, J.D. Boerckel, D.L. Kaplan, R.E. Guldberg, A
[1] N. Huebsch, D.J. Mooney, Inspiration and application in the evolution of
silk hydrogel-based delivery system of bone morphogenetic protein for the
biomaterials, Nature 462 (2009) 426–432.
treatment of large bone defects, J. Mech. Behav. Biomed. Mater. 11 (2012)
[2] C. Helary, M.F. Desimone, Recent advances in biomaterials for tissue
123–131.
engineering and controlled drug delivery, Curr. Pharm. Biotechnol. 16
[35] A.A. Dinerman, J. Cappello, H. Ghandehari, S.W. Hoag, Solute diffusion in
(2015) 635–645.
genetically engineered silk–elastinlike protein polymer hydrogels, J. Control.
[3] A. Khademhosseini, R. Langer, Microengineered hydrogels for tissue
Release 82 (2002) 277–287.
engineering, Biomaterials 28 (2007) 5087–5092.
[36] J.Y. Fang, J.P. Chen, Y.L. Leu, H. Wang, Characterization and evaluation of silk
[4] A.M. Lowman, N.A. Peppas, Hydrogels, in: E. Mathiowitz (Ed.), Encyclopedia
protein hydrogels for drug delivery, Chem. Pharm. Bull. 54 (2006) 156–162.
of Controlled Drug Delivery, Wiley, New York, 1999, pp. 397–418.
[37] M. Fini et al., The healing of confined critical size cancellous defects in the
[5] N.A. Peppas, J.Z. Hilt, A. Khademhosseini, R. Langer, Hydrogels in biology and
presence of silk fibroin hydrogel, Biomaterials 26 (2005) 3527–3536.
medicine: from molecular principles to bionanotechnology, Adv. Mater. 18
[38] B. Kundu, S.C. Kundu, Bio-inspired fabrication of fibroin cryogels from the
(2006) 1345–1360.
muga silkworm Antheraea assamensis for liver tissue engineering, Biomed.
[6] B.V. Slaughter, S.S. Khurshid, O.Z. Fisher, A. Khademhosseini, N.A. Peppas,
Mater. 8 (2013) 055003.
Hydrogels in regenerative medicine, Adv. Mater. 21 (2009) 3307–3329.
[39] A.S. Eissa, S.A. Khan, Acid-induced gelation of enzymatically modified,
[7] M.W. Tibbitt, K.S. Anseth, Hydrogels as extracellular matrix mimics for 3D cell
preheated whey proteins, J. Agric. Food Chem. 53 (2005) 5010–5017.
culture, Biotechnol. Bioeng. 103 (2009) 655–663.
[40] S. Gunasekaran, S. Ko, L. Xiao, Use of whey proteins for encapsulation and
[8] O. Wichterle, D. Lim, Hydrophilic gels for biological use, Nature 185 (1960)
controlled delivery applications, J. Food Eng. 83 (2007) 31–40.
117–118.
30 S. Kapoor, S.C. Kundu / Acta Biomaterialia 31 (2016) 17–32

[41] G.E. Remondetto, E. Beyssac, M. Subirade, Iron availability from whey protein [75] O. Hakimi, D.P. Knight, F. Vollrath, P. Vadgama, Spider and mulberry silkworm
hydrogels: an in vitro study, J. Agric. Food Chem. 52 (2004) 8137–8143. silks as compatible biomaterials, Compos. B 38 (2007) 324–337.
[42] H. Teramoto, T. Kameda, Y. Tamada, Preparation of gel film from Bombyx [76] Y. Cao, B. Wang, Biodegradation of Silk Biomaterials, Int. J. Mol. Sci. 10 (2009)
mori silk sericin and its characterization as a wound dressing, Biosci. 1514–1524.
Biotechnol. Biochem. 72 (2008) 3189–3196. [77] K. Hu, F. Cui, Q. Lv, J. Ma, Q. Feng, L. Xu, D. Fan, Preparation of fibroin/
[43] H. Teramoto, K. Nakajima, C. Takabayashi, Preparation of elastic silk sericin recombinant human-like collagen scaffold to promote fibroblasts
hydrogel, Biosci. Biotechnol. Biochem. 69 (2005) 845–847. compatibility, J. Biomed. Mater. Res. A 84 (2008) 483–490.
[44] Z. Wang et al., Exploring natural silk protein sericin for regenerative [78] S. Yanagisawa, Z. Zhu, I. Kobayashi, K. Uchino, Y. Tamada, T. Tamura, T.
medicine: an injectable, photoluminescent, cell-adhesive 3D hydrogel, Sci. Asakura, Improving cell-adhesive properties of recombinant Bombyx mori
Rep. 4 (2014) 7064. silk by incorporation of collagen or fibronectin derived peptides produced by
[45] C.M. Pollock, R.E. Shadwick, Relationship between body mass and transgenic silkworms, Biomacromolecules 8 (2007) 3487–3492.
biomechanical properties of limb tendons of adult mammals, Am. J. Physiol. [79] A.W. Morgan, K.E. Roskov, S. Lin-Gibson, D.L. Kaplan, M.L. Becker, C.G. Simon
35 (1994) R1016–R1021. Jr., Characterization and optimization of RGD-containing silk blends to
[46] B.B. Aaron, J.M. Gosline, Elastin as a random-network elastomer: a support osteoblastic differentiation, Biomaterials 29 (2008) 2556–2563.
mechanical and optical analysis of single elastin fibers, Biopolymers 20 [80] N. Kasoju, U. Bora, Silk fibroin in tissue engineering, Adv. Healthc. Mater. 1
(1981) 1247–1260. (2012) 393–412.
[47] J.M. Gosline, P.A. Guerette, C.S. Ortlepp, K.N. Savage, The mechanical design of [81] B. Kundu, R. Rajkhowa, S.C. Kundu, X. Wang, Silk fibroin biomaterials for
spider silks: from fibroin sequence to mechanical function, J. Exp. Biol. 202 tissue regenerations, Adv. Drug Deliv. Rev. 65 (2013) 457–470.
(1999) 3295–3303. [82] H. Tao, D.L. Kaplan, F.G. Omenetto, Silk materials–a road to sustainable high
[48] H. Hosseinkhani, P.D. Hong, D.S. Yu, Self-assembled proteins and peptides for technology, Adv. Mater. 24 (2012) 2824–2837.
regenerative medicine, Chem. Rev. 113 (2013) 4837–4861. [83] A. Naba, K.R. Clauser, H. Ding, C.A. Whittaker, S.A. Carr, R.O. Hynes, The
[49] L.K. Langeberg, J.D. Scott, Signalling scaffolds and local organization of extracellular matrix: tools and insights for the ‘‘omics” era, Matrix Biol.
cellular behaviour, Nat. Rev. Mol. Cell Biol. 16 (2015) 232–244. (2015). pii: S0945-053X(15)00121-3.
[50] C.E. Chua, B.L. Tang, Role of Rab GTPases and their interacting proteins in [84] W.P. Daley, S.B. Peters, M. Larsen, Extracellular matrix dynamics in
mediating metabolic signalling and regulation, Cell. Mol. Life Sci. 72 (2015) development and regenerative medicine, J. Cell Sci. 121 (2008) 255–264.
2289–2304. [85] D.E. Discher, P. Janmey, Y.L. Wang, Tissue cells feel and respond to the
[51] C. Wang, R.J. Stewart, J. Kopeek, Hybrid hydrogels assembled from synthetic stiffness of their substrate, Science 310 (2005) 1139–1143.
polymers and coiled-coil protein domains, Nature 397 (1999) 417–420. [86] N. Minoura, S. Aiba, M. Higuchi, Y. Gotoh, M. Tsukada, Y. Imai, Attachment
[52] B. Jung, P. Theato, Chemical strategies for the synthesis of protein-polymer and growth of fibroblast cells on silk fibroin, Biochem. Biophys. Res. Commun.
conjugates, Adv. Polym. Sci. 253 (2013) 37–70. 208 (1995) 511–516.
[53] S. Gomes, I.B. Leonor, J.F. Mano, R.L. Reis, D.L. Kaplan, Natural and genetically [87] D. Harjanto, M.H. Zaman, Matrix mechanics and receptor-ligand interactions
engineered proteins for tissue engineering, Prog. Polym. Sci. 37 (2012) 1–17. in cell adhesion, Org. Biomol. Chem. 8 (2010) 299–304.
[54] M. Haider, Z. Megeed, H. Ghandehari, Genetically engineered polymers: [88] U.J. Kim, J. Park, C. Li, H.J. Jin, R. Valluzzi, D.L. Kaplan, Structure and properties
status and prospects for controlled release, J. Control. Release 20 (2004) 1–26. of silk hydrogels, Biomacromolecules 5 (2004) 786–792.
[55] C. Fu, Z. Shao, V. Fritz, Animal silks: their structures, properties and artificial [89] A. Motta, C. Migliaresi, F. Faccioni, P. Torricelli, M. Fini, R. Giardino, Fibroin
production, Chem. Commun. (Camb.) 21 (2009) 6515–6529. hydrogels for biomedical applications: preparation, characterization and
[56] X. Hu, K. Vasanthavada, K. Kohler, S. McNary, A.M. Moore, C.A. Vierra, in vitro cell culture studies, J. Biomater. Sci. Polym. Ed. 15 (2004) 851–864.
Molecular mechanisms of spider silk, Cell. Mol. Life Sci. 63 (2006) 1986–1999. [90] M. Ribeiro, M.A. de Moraes, M.M. Beppu, F.J. Monteiro, M.P. Ferraz, The role of
[57] B. Kłudkiewicz, K. Grzelak, Silk proteins of B. mori–characteristics, dialysis and freezing on structural conformation, thermal properties and
biosynthesis and regulation of gene expression, Postepy Biochem. 39 morphology of silk fibroin hydrogels, Biomatter 4 (2014) e28536.
(1993) 105–111. [91] P.H. Chao, S. Yodmuang, X. Wang, L. Sun, D.L. Kaplan, G. Vunjak-Novakovic,
[58] A. Rising et al., Spider silk proteins–mechanical property and gene sequence, Silk hydrogel for cartilage tissue engineering, J. Biomed. Mater. Res. B Appl.
Zool. Sci. 22 (2005) 273–281. Biomater. 95 (2010) 84–90.
[59] T.D. Sutherland, J.H. Young, S. Weisman, C.Y. Hayashi, D.J. Merritt, Insect silk: [92] X. Wang, J.A. Kluge, G.G. Leisk, D.L. Kaplan, Sonication-induced gelation of silk
one name, many materials, Annu. Rev. Entomol. 55 (2010) 171–188. fibroin for cell encapsulation, Biomaterials 29 (2008) 1054–1064.
[60] R. Valluzzi, S. Winkler, D. Wilson, D.L. Kaplan, Silk: molecular organization [93] T. Yucel, P. Cebe, D.L. Kaplan, Vortex-induced injectable silk fibroin hydrogels,
and control of assembly, Philos. Trans. R. Soc. Lond. B Biol. Sci. 28 (2002) 165– Biophys. J. 97 (2009) 2044–2050.
167. [94] W. Zhang et al., The use of injectable sonication-induced silk hydrogel for
[61] C. Vepari, D.L. Kaplan, Silk as a Biomaterial, Prog. Polym. Sci. 32 (2007) 991– VEGF(165) and BMP-2 delivery for elevation of the maxillary sinus floor,
1007. Biomaterials 32 (2011) 9415–9424.
[62] H.Y. Wang, Y.Q. Zhang, Processing silk hydrogel and its applications in [95] W. Sun, T. Incitti, C. Migliaresi, A. Quattrone, S. Casarosa, A. Motta, Viability
biomedical materials, Biotechnol. Prog. 31 (2015) 630–640. and neuronal differentiation of neural stem cells encapsulated in silk fibroin
[63] C.L. Craig, M. Hsu, D. Kaplan, N.E. Pierce, A comparison of the composition of hydrogel functionalized with an IKVAV peptide, J. Tissue Eng. Regen. Med.
silk proteins produced by spiders and insects, Int. J. Biol. Macromol. 24 (1999) (2015), http://dx.doi.org/10.1002/term.2053.
109–118. [96] M.L. Floren, S. Spilimbergo, A. Motta, C. Migliaresi, Carbon dioxide induced
[64] B. Kundu, N.E. Kurland, V.K. Yadavalli, S.C. Kundu, Isolation and processing of silk protein gelation for biomedical applications, Biomacromolecules 13
silk proteins for biomedical applications, Int. J. Biol. Macromol. 70 (2014) 70– (2012) 2060–2072.
77. [97] R.R. Mallepally, M.A. Marin, M.A. McHugh, CO2-assisted synthesis of silk
[65] D.N. Rockwood, R.C. Preda, T. Yücel, X. Wang, M.L. Lovett, D.L. Kaplan, fibroin hydrogels and aerogels, Acta Biomater. (2014). pii: S1742-7061(14)
Materials fabrication from Bombyx mori silk fibroin, Nat. Protoc. 6 (2011) 00254-2.
1612–1631. [98] S. Min, T. Nakamura, A. Teramoto, K. Abe, Preparation and characterization of
[66] A. Datta, A.K. Ghosh, S.C. Kundu, Purification and characterization of fibroin crosslinked porous silk fibroin gel, Sekiyu Gakkaishi 54 (1998) 85–92.
from the tropical Saturniid silkworm, Antheraea mylitta, Insect Biochem. Mol. [99] Y. Morita, N. Tomita, H. Aoki, S. Wakitani, Y. Tamada, T. Suguro, K. Ikeuchi,
Biol. 31 (2001) 1013–1018. Visco-elastic properties of cartilage tissue regenerated with fibroin sponge,
[67] C.L. Craig, C. Riekel, Comparative architecture of silks, fibrous proteins and Biomed. Mater. Eng. 12 (2002) 291–298.
their encoding genes in insects and spiders, Comp. Biochem. Physiol. B: [100] A. Fournier, Quantitative data on the Bombyx mori L. silkworm: a review,
Biochem. Mol. Biol. 133 (2002) 493–507. Biochimie 19 (1979) 283–320.
[68] G.H. Altman, F. Diaz, C. Jakuba, T. Calabro, R.L. Horan, J. Chen, H. Lu, J. [101] Z.H. Ayub, M. Arai, K. Hirabayashi, Mechanism of the gelation of fibroin
Richmond, D.L. Kaplan, Silk-based biomaterials, Biomaterials 24 (2003) 401– solution, Biosci. Biotechnol. Biochem. 57 (1993) 1910–1912.
416. [102] Y. Miyaguchi, J. Hu, Physicochemical properties of silk fibroin after
[69] Z. Shao, F. Vollrath, Surprising strength of silkworm silk, Nature 418 (2002) solubilization using calcium chloride with or without ethanol, Food Sci.
741. Technol. Res. 11 (2005) 37–42.
[70] F.G. Omenetto, D.L. Kaplan, New opportunities for an ancient material, [103] A. Matsumoto, J. Chen, A.L. Collette, U.J. Kim, G.H. Altman, P. Cebe, D. Kaplan,
Science 329 (2010) 528–531. Mechanisms of silk fibroin sol-gel transitions, J. Phys. Chem. B 110 (2006)
[71] Y. Gotoh, M. Tsukada, N. Minoura, Effect of the chemical modification of the 21630–21638.
arginyl residue in Bombyx mori silk fibroin on the attachment and growth of [104] X. Wu, J. Hou, M. Li, J. Wang, D.L. Kaplan, S. Lu, Sodium dodecyl sulfate-
fibroblast cells, J. Biomed. Mater. Res. 39 (1998) 351–357. induced rapid gelation of silk fibroin, Acta Biomater. 8 (2012) 2185–2192.
[72] K. Inouye, M. Kurokawa, S. Nishikawa, M. Tsukada, Use of Bombyx mori silk [105] F. Zhang, J. Li, T. Zhu, S. Zhang, S.C. Kundu, S. Lu, Potential of biocompatible
fibroin as a substratum for cultivation of animal cells, J. Biochem. Biophys. regenerated silk fibroin/sodium N-lauroyl sarcosinate hydrogels, J. Biomater.
Methods 37 (1998) 159–164. Sci. Polym. Ed. 8 (2015) 1–20.
[73] N. Minoura, S. Aiba, Y. Gotoh, M. Tsukada, Y. Imai, Attachment and growth of [106] N. Guziewicz, A. Best, B. Perez-Ramirez, D.L. Kaplan, Lyophilized silk fibroin
cultured fibroblast cells on silk protein matrices, J. Biomed. Mater. Res. 29 hydrogels for the sustained local delivery of therapeutic monoclonal
(1995) 1215–1221. antibodies, Biomaterials 32 (2011) 2642–2650.
[74] A. Leal-Egaña, T. Scheibel, Silk-based materials for biomedical applications, [107] T. Hanawa, A. Watanabe, T. Tsuchiya, R. Ikoma, M. Hidaka, M. Sugihara, New
Biotechnol. Appl. Biochem. 55 (2010) 155–167. oral dosage form for elderly patients: preparation and characterization of silk
fibroin gel, Chem. Pharm. Bull. (Tokyo) 43 (1995) 284–288.
S. Kapoor, S.C. Kundu / Acta Biomaterialia 31 (2016) 17–32 31

[108] H.Y. Wang, Y.Q. Zhang, Processing and characterisation of a novel [136] C.H. Park, L. Jeong, D. Cho, O.H. Kwon, W.H. Park, Effect of methylcellulose on
electropolymerized silk fibroin hydrogel membrane, Sci. Rep. 4 (2014) 6182. the formation and drug release behavior of silk fibroin hydrogel, Carbohydr.
[109] K. Hirabayashi, Z.H. Ayub, Y. Kume, Gelation of silk fibroin, Sekiyu Gakkaishi Polym. 98 (2013) 1179–1185.
46 (1990) 521–524. [137] K. Numata, S. Yamazaki, T. Katashima, J.A. Chuah, N. Naga, T. Sakai, Silk-
[110] A.E. Terry, D.P. Knight, D. Porter, F. Vollrath, PH induced changes in the pectin hydrogel with superior mechanical properties, biodegradability, and
rheology of silk fibroin solution from the middle division of Bombyx mori biocompatibility, Macromol. Biosci. 14 (2014) 799–806.
silkworm, Biomacromolecules 5 (2004) 768–772. [138] K. Ziv et al., A tunable silk-alginate hydrogel scaffold for stem cell culture and
[111] S. Nagarkar, A. Patil, A. Lele, S. Bhat, J. Bellare, R.A. Mashelkar, Some transplantation, Biomaterials 35 (2014) 3736–3743.
mechanistic insights into the gelation of regenerated silk fibroin sol, Ind. Eng. [139] J. Ming, Z. Jiang, P. Wang, S. Bie, B. Zuo, Silk fibroin/sodium alginate fibrous
Chem. Res. 48 (2009) 8014–8023. hydrogels regulated hydroxyapatite crystal growth, Mater. Sci. Eng. C 51
[112] W.L. Rice et al., Non-invasive characterization of structure and morphology of (2015) 287–293.
silk fibroin biomaterials using non-linear microscopy, Biomaterials 29 (2008) [140] L.H. Sperling, Interpenetrating polymer networks: an overview, in: D.
2015–2024. Klempner, L.H. Sperling, L. Utracki (Eds.), Interpenetrating Polymer
[113] P. Zhou, X. Xie, D.P. Knight, X.H. Zong, F. Deng, W.H. Yao, Effects of pH and Networks, ACS publications, Washington, DC, 1994, pp. 3–38.
calcium ions on the conformational transitions in silk fibroin using 2D raman [141] D. Myung et al., Biomimetic strain hardening in interpenetrating polymer
correlation spectroscopy and 13C solid-state NMR, Biochemistry 43 (2004) network hydrogels, Polymer 48 (2007) 5376–5387.
11302–11311. [142] I.R. Schmolka, A comparison of block copolymer surfactant gels, JAOCS 68
[114] S. Bai, X. Zhang, Q. Lu, W. Sheng, L. Liu, B. Dong, D.L. Kaplan, H. Zhu, Reversible (1991) 206–209.
hydrogel-solution system of silk with high Beta-sheet content, [143] H.Y. Kweon, S.H. Park, J.H. Yeo, Y.W. Lee, C.S. Cho, Preparation of semi-
Biomacromolecules 15 (2014) 3044–3051. interpenetrating polymer networks composed of silk fibroin and poly
[115] B.B. Mandal, S. Kapoor, S.C. Kundu, Silk fibroin/polyacrylamide semi- (ethylene glycol) macromer, J. Appl. Polym. Sci. 80 (2001) 1848–1853.
interpenetrating network hydrogels for controlled drug release, [144] H. Kweon, J.H. Yeo, K.G. Lee, H.C. Lee, H.S. Na, Y. Won, C.S. Cho, Semi-
Biomaterials 30 (2009) 2826–2836. interpenetrating polymer networks composed of silk fibroin and poly
[116] G. Li, Y. Kong, Y. Zhao, Y. Zhao, L. Zhang, Y. Yang, Fabrication and (ethylene glycol) for wound dressing, Biomed. Mater. 3 (2008) 034115.
characterization of polyacrylamide/silk fibroin hydrogels for peripheral [145] N.A. Peppas, P. Bures, W. Leobandung, H. Ichikawa, Hydrogels in
nerve regeneration, J. Biomater. Sci. Polym. Ed. 26 (2015) 1–34. pharmaceutical formulations, Eur. J. Pharm. Biopharm. 50 (2000) 27–46.
[117] J. Kundu, L.A. Poole-Warren, P. Martens, S.C. Kundu, Silk fibroin/poly(vinyl [146] E. Sato Matsuo, T. Toyoichi, Kinetics of discontinuous volume–phase
alcohol) photocrosslinked hydrogels for delivery of macromolecular drugs, transition of gels, J. Chem. Phys. 89 (1988) 1695–1703.
Acta Biomater. 8 (2012) 1720–1729. [147] T. Okano, Y.H. Bae, H. Jacobs, S.W. Kim, Thermally on-off switching polymers
[118] M. Li, S. Lu, Z. Wu, K. Tan, N. Minoura, S. Kuga, Structure and properties of silk for drug permeation and release, J. Control. Release 11 (1990) 255–265.
fibroin-poly(vinyl alcohol) gel, Int. J. Biol. Macromol. 30 (2002) 89–94. [148] D. Chow, M.L. Nunalee, D.W. Lim, A.J. Simnick, A. Chilkoti, Peptide-based
[119] G.D. Kang, J.H. Nahm, J.S. Park, J.Y. Moon, C.S. Cho, J.H. Yeo, Effects of biopolymers in biomedicine and biotechnology, Mater. Sci. Eng. R Rep. 62
poloxamer on the gelation of silk fibroin, Macromol. Rapid Commun. 21 (2008) 125–155.
(2000) 788–791. [149] J.G. Hardy, T.R. Scheibel, Silk-inspired polymers and proteins, Biochem. Soc.
[120] M.K. Yoo, H.Y. Kweon, K.G. Lee, H.C. Lee, C.S. Cho, Preparation of semi- Trans. 37 (2009) 677–681.
interpenetrating polymer networks composed of silk fibroin and poloxamer [150] R. Price, A. Poursaid, J. Cappello, H. Ghandehari, Effect of shear on
macromer, Int. J. Biol. Macromol. 34 (2004) 263–270. physicochemical properties of matrix metalloproteinase responsive silk-
[121] J. Hu, B. Chen, F. Guo, J. Du, P. Gu, X. Lin, et al., Injectable silk fibroin/ elastinlike hydrogels, J. Control. Release (2014). pii: S0168-3659(14)00525-2.
polyurethane composite hydrogel for nucleus pulposus replacement, J. [151] T. Scheibel, Spider silks: recombinant synthesis, assembly, spinning, and
Mater. Sci. Mater. Med. 23 (2012) 711–722. engineering of synthetic proteins, Microb. Cell Fact. 3 (2004) 14.
[122] E.S. Gil, S.M. Hudson, Effect of silk fibroin interpenetrating networks on [152] M. Stark, S. Grip, A. Rising, M. Hedhammar, W. Engström, G. Hjälm, J.
swelling/deswelling kinetics and rheological properties of poly (N- Johansson, Macroscopic fibers self-assembled from recombinant miniature
isopropylacrylamide) hydrogels, Biomacromolecules 1 (2007) 258–264. spider silk proteins, Biomacromolecules 8 (2007) 1695–1701.
[123] E.S. Gil, S.H. Park, L.W. Tien, B. Trimmer, S.M. Hudson, D.L. Kaplan, [153] F. Teulé, A.R. Cooper, W.A. Furin, D. Bittencourt, E.L. Rech, A. Brooks, R.V.
Mechanically robust, rapidly actuating, and biologically functionalized Lewis, A protocol for the production of recombinant spider silk-like proteins
macroporous poly(N-isopropylacrylamide)/silk hybrid hydrogels, Langmuir for artificial fiber spinning, Nat. Protoc. 4 (2009) 341–355.
26 (2010) 15614–15624. [154] C. Vendrely, T. Scheibel, Biotechnological production of spider-silk proteins
[124] X. Wang, B. Partlow, J. Liu, Z. Zheng, B. Su, Y. Wang, D.L. Kaplan, Injectable enables new applications, Macromol. Biosci. 7 (2007) 401–409.
silk-polyethylene glycol hydrogels, Acta Biomater. 12 (2015) 51–61. [155] C. Vendrely, C. Ackerschott, L. Römer, T. Scheibel, Molecular design of
[125] P. Thitiwuthikiat, M. Ii, T. Saito, M. Asahi, S. Kanokpanont, Y. Tabata, A performance proteins with repetitive sequences: recombinant flagelliform
vascular patch prepared from Thai silk fibroin and gelatin hydrogel spider silk as basis for biomaterials, Methods Mol. Biol. 474 (2008) 3–14.
incorporating simvastatin-micelles to recruit endothelial progenitor cells, [156] C. Wong Po Foo, D.L. Kaplan, Genetic engineering of fibrous proteins: spider
Tissue Eng. Part A 21 (2015) 1309–1319. dragline silk and collagen, Adv. Drug Deliv. Rev. 54 (2002) 1131–1143.
[126] S. Das et al., Bioprintable, cell-laden silk fibroin-gelatin hydrogel supporting [157] D. Huemmerich, T. Scheibel, F. Vollrath, S. Cohen, U. Gat, S. Ittah, Novel
multilineage differentiation of stem cells for fabrication of three-dimensional assembly properties of recombinant spider dragline silk proteins, Curr. Biol.
tissue constructs, Acta Biomater. 11 (2015) 233–246. 14 (2004) 2070–2074.
[127] E.S. Gil, R.J. Spontak, S.M. Hudson, Effect of beta-sheet crystals on the thermal [158] S.R. Fahnestock, S.L. Irwin, Synthetic spider dragline silk proteins and their
and rheological behavior of protein-based hydrogels derived from gelatin and production in Escherichia coli, Appl. Microbiol. Biotechnol. 47 (1997) 23–32.
silk fibroin, Macromol. Biosci. 5 (2005) 702–709. [159] S.R. Fahnestock, L.A. Bedzyk, Production of synthetic spider dragline silk
[128] E.S. Gil, D.J. Frankowski, R.J. Spontak, S.M. Hudson, Swelling behavior and protein in Pichia pastoris, Appl. Microbiol. Biotechnol. 47 (1997) 33–39.
morphological evolution of mixed gelatin/silk fibroin hydrogels, [160] S.R. Fahnestock, Z. Yao, L.A. Bedzyk, Microbial production of spider silk
Biomacromolecules 6 (2005) 3079–3087. proteins, J. Biotechnol. 74 (2000) 105–119.
[129] W. Sun, T. Incitti, C. Migliaresi, A. Quattrone, S. Casarosa, A. Motta, Genipin- [161] A. Lazaris, S. Arcidiacono, Y. Huang, J.F. Zhou, F. Duguay, N. Chretien, E.A.
crosslinked gelatin-silk fibroin hydrogels for modulating the behaviour of Welsh, J.W. Soares, C.N. Karatzas, Spider silk fibers spun from soluble
pluripotent cells, J. Tissue Eng. Regen. Med. (2014), http://dx.doi.org/10.1002/ recombinant silk produced in mammalian cells, Science 295 (2002) 472–476.
term.1868. [162] D. Williams, Sows’ ears, silk purses and goats’ milk: new production methods
[130] W. Xiao et al., Synthesis and characterization of photocrosslinkable gelatin and medical applications for silk, Med. Device Technol. 14 (2003) 9–11.
and silk fibroin interpenetrating polymer network hydrogels, Acta Biomater. [163] A. Pandey, Plants to make silk, Trends Genet. 17 (2001) 442.
7 (2011) 2384–2393. [164] J. Scheller, K.H. Gührs, F. Grosse, U. Conrad, Production of spider silk proteins
[131] Q. Lv, K. Hu, Q.L. Feng, F. Cui, Fibroin/collagen hybrid hydrogels with in tobacco and potato, Nat. Biotechnol. 19 (2001) 573–577.
crosslinking method: preparation, properties, and cytocompatibility, J. [165] S. Rammensee, U. Slotta, T. Scheibel, A.R. Bausch, Assembly mechanism of
Biomed. Mater. Res., Part A 1 (2008) 198–207. recombinant spider silk proteins, Proc. Natl. Acad. Sci. U.S.A. 105 (2008)
[132] X. Chen, W.J. Li, W. Zhong, Y.H. Lu, T.Y. Yu, PH sensitivity and ion sensitivity of 6590–6595.
hydrogels based on complex-forming chitosan/silk fibroin interpenetrating [166] K. Schacht, T. Scheibel, Controlled hydrogel formation of a recombinant
polymer network, J. Appl. Polym. Sci. 65 (1997) 2257–2262. spider silk protein, Biomacromolecules 12 (2011) 2488–2495.
[133] F. Mirahmadi, M. Tafazzoli-Shadpour, M.A. Shokrgozar, S. Bonakdar, [167] J.C.R. Cabello, J. Reguera, A. Girotti, M. Alonso, A.M. Testera, Developing
Enhanced mechanical properties of thermosensitive chitosan hydrogel by functionality in elastin-like polymers by increasing their molecular
silk fibers for cartilage tissue engineering, Mater. Sci. Eng. C 33 (2013) 4786– complexity: the power of the genetic engineering approach, Prog. Polym.
4794. Sci. 30 (2005) 1119–1145.
[134] R. Elia et al., Silk-hyaluronan-based composite hydrogels: A novel, securable [168] W.F. Daamen, J.H. Veerkamp, J.C.M. van Hest, T.H. van Kuppevelt, Elastin as a
vehicle for drug delivery, J. Biomater. Appl. 27 (2013) 749–762. biomaterial for tissue engineering, Biomaterials 28 (2007) 4378–4398.
[135] X. Hu, Q. Lu, L. Sun, P. Cebe, X. Wang, X. Zhang, D.L. Kaplan, Biomaterials from [169] W. Huang, A. Rollett, D.L. Kaplan, Silk-elastin-like protein biomaterials for the
ultrasonication-induced silk fibroin-hyaluronic acid hydrogels, controlled delivery of therapeutics, Expert Opin. Drug Deliv. 12 (2015) 779–
Biomacromolecules 11 (2010) 3178–3188. 791.
32 S. Kapoor, S.C. Kundu / Acta Biomaterialia 31 (2016) 17–32

[170] J. Cappello, J. Crissman, M. Dorman, M. Mikolajczak, G. Textor, M. Marquet, [195] L. Meinel et al., Silk implants for the healing of critical size bone defects, Bone
Genetic engineering of structural protein polymers, Biotechnol. Prog. 6 37 (2005) 688–698.
(1990) 198–202. [196] H.J. Kim, U.J. Kim, G.V. Novakovic, B.H. Min, D.L. Kaplan, Influence of
[171] R. Dandu, A.V. Cresce, R. Briber, P. Dowell, J. Cappello, H. Ghandehari, Silk– macroporous protein scaffolds on bone tissue engineering from bone marrow
elastinlike protein polymer hydrogels: Influence of monomer sequence on stem cells, Biomaterials 26 (2005) 4442–4452.
physicochemical properties, Polymer 50 (2009) 366–374. [197] L. Wang, G.L. Ning, M. Senn, Microstructure and gelation behavior of
[172] J. Cappello, Synthetically designed protein polymer biomaterials, in: K. Park hydroxyapatite based nanocomposite sol containing chemically modified
(Ed.), Controlled Drug Delivery: Strategies and Challenges, American silk fibroin, Colloids Surf., A 254 (2005) 159–164.
Chemical Society, Washington, DC, 1997, pp. 439–453. [198] T. Aigner, J. Stove, Collagens—major component of the physiological cartilage
[173] R. Dandu, H. Ghandehari, Delivery of bioactive agents from recombinant matrix, major target of cartilage degeneration, major tool in cartilage repair,
polymers, Prog. Polymer Sci. 32 (2007) 1008–1030. Adv. Drug Deliv. Rev. 55 (2003) 1569–1593.
[174] Z. Megeed, J. Cappello, H. Ghandehari, Genetically engineered silk-elastinlike [199] R. Takaya, N. Kachi, N. Tomita, Micro-folding culture: a new method of
protein polymers for controlled drug delivery, Adv. Drug Deliv. Rev. 54 (2002) making multicellular aggregates, Eur. Cells Mater. 12 (2006) 40.
1075–1091. [200] S. Yodmuang et al., Silk microfiber-reinforced silk hydrogel composites for
[175] J. Cappello, J.W. Crissman, M. Crissman, F.A. Ferrari, G. Textor, O. Wallis, J.R. functional cartilage tissue repair, Acta Biomater. 11 (2015) 27–36.
Whitledge, X. Zhou, D. Burman, L. Aukerman, E.R. Stedronsky, In situ self [201] C. Shangkai, T. Naohide, Y. Koji, H. Yasuji, N. Masaaki, T. Tomohiro, T. Yasushi,
assembling protein polymer gel systems for administration, delivery, and Transplantation of allogeneic chondrocytes cultured in fibroin sponge and
release of drugs, J. Control. Release 53 (1998) 105–117. stirring chamber to promote cartilage regeneration, Tissue Eng. 13 (2007)
[176] A.A. Dinerman, J. Cappello, H. Ghandehari, S.W. Hoag, Swelling behavior of a 483–492.
genetically engineered silk-elastinlike protein polymer hydrogel, [202] N. Nakayama, D. Duryea, R. Manoukian, G. Chow, C.Y. Han, Macroscopic
Biomaterials 23 (2002) 4203–4210. cartilage formation with embryonic stem-cell-derived mesodermal
[177] Z. Megeed, J. Cappello, H. Ghandehari, Thermal analysis of water in silk- progenitor cells, J. Cell Sci. 116 (2003) 2015–2028.
elastinlike hydrogels by differential scanning calorimetry, [203] L. Song, D. Baksh, R.S. Tuan, Mesenchymal stem cell-based cartilage tissue
Biomacromolecules 5 (2004) 793–797. engineering: cells, scaffold and biology, Cytotherapy 6 (2004) 596–601.
[178] A. Poursaid et al., In situ gelling silk-elastinlike protein polymer for [204] M. Haider, J. Cappello, H. Ghandehari, K.W. Leong, In vitro chondrogenesis of
transarterial chemoembolization, Biomaterials 57 (2015) 142–152. mesenchymal stem cells in recombinant silkelastinlike hydrogels, Pharm.
[179] M. Haider, V. Leung, F. Ferrari, J. Crissman, J. Powell, J. Cappello, H. Res. 25 (2008) 692–699.
Ghandehari, Molecular engineering of silk-elastinlike polymers for matrix- [205] J. Chen, N. Minoura, A. Tanioka, Transport of pharmaceuticals through silk
mediated gene delivery: biosynthesis and characterization, Mol. Pharm. 2 fibroin membrane, Polymer 35 (1994) 2853–2856.
(2005) 139–150. [206] T. Hanawa, A. Watanabe, T. Tsuchiya, R. Ikoma, M. Hidaka, M. Sugihara, New
[180] A. Nagarsekar, J. Crissman, M. Crissman, F. Ferrari, J. Cappello, H. Ghandehari, oral dosage form for elderly patients. II. Release behavior of benfotiamine
Genetic synthesis and characterization of pH- and temperature-sensitive from silk fibroin gel, Chem. Pharm. Bull. (Tokyo) 43 (1995) 872–876.
silk-elastinlike protein block copolymers, J. Biomed. Mater. Res. 62 (2002) [207] M.L. Lovett, X. Wang, T. Yucel, L. York, M. Keirstead, L. Haggerty, D.L. Kaplan,
195–203. Silk hydrogels for sustained ocular delivery of anti-vascular endothelial
[181] R. Price, A. Poursaid, J. Cappello, H. Ghandehari, In vivo evaluation of matrix growth factor (anti-VEGF) therapeutics, Eur. J. Pharm. Biopharm. (2015). pii:
metalloproteinase responsive silk-elastinlike protein polymers for cancer S0939-6411(15)00004-1.
gene therapy, J. Control. Release (2015). pii: S0168-3659(15)00629-X. [208] Z. Megeed, J. Cappello, H. Ghandehari, Controlled release of plasmid DNA
[182] J. Langer, P. Vacanti, Tissue engineering, Science 260 (1993) 920–926. from a genetically engineered silk-elastinlike hydrogel, Pharm. Res. 19 (2002)
[183] S. Varghese, J.H. Elisseeff, Hydrogels for musculoskeletal tissue engineering, 954–959.
Adv. Polym. Sci. 203 (2006) 95–144. [209] Z. Megeed, M. Haider, D. Li, B.W. O’Malley Jr, J. Cappello, H. Ghandehari, In
[184] V.L. Tsang, S.N. Bhatia, Three-dimensional tissue fabrication, Adv. Drug Deliv. vitro and in vivo evaluation of recombinant silk-elastinlike hydrogels for
Rev. 56 (2004) 1635–1647. cancer gene therapy, J. Control. Release 94 (2004) 433–445.
[185] B.P. Chan, K.W. Leong, Scaffolding in tissue engineering: general approaches [210] A. Hatefi, J. Cappello, H. Ghandehari, Adenoviral gene delivery to solid tumors
and tissue-specific considerations, Eur. Spine J. 17 (2008) 467–479. by recombinant silk-elastinlike protein polymers, Pharm. Res. 24 (2007) 773–
[186] S.A. Bencherif, R.W. Sands, D. Bhatta, P. Arany, C.S. Verbeke, D.A. Edwards, D.J. 779.
Mooney, Injectable preformed scaffolds with shape-memory properties, Proc. [211] D. Hwang, V. Moolchandani, R. Dandu, M. Haider, J. Cappello, H. Ghandehari,
Natl. Acad. Sci. U.S.A. 109 (2012) 19590–19595. Influence of polymer structure and biodegradation on DNA release from silk–
[187] A.S. Hoffman, Hydrogels for biomedical applications, Adv. Drug Deliv. Rev. 43 elastinlike protein polymer hydrogels, Int. J. Pharm. 368 (2009) 215–219.
(2002) 3–12. [212] W.H. Elliott, W. Bonani, D. Maniglio, A. Motta, W. Tan, C. Migliaresi, Silk
[188] T. Cordonnier, J. Sohier, P. Rosset, P. Ayrolle, Biomimetic materials for bone hydrogels of tunable structure and viscoelastic properties using different
tissue engineering—State of the art and future trends, Adv. Eng. Mater. 13 chronological orders of genipin and physical cross-linking, ACS Appl. Mater.
(2011) B135–B150. Interfaces 7 (2015) 12099–12108.
[189] S. Bose, M. Roy, A. Bandyopadhyay, Recent advances in bone tissue [213] J. Choi, T. Konno, M. Takai, K. Ishihara, Smart controlled preparation of
engineering scaffolds, Trends Biotechnol. 30 (2012) 546–554. multilayered hydrogel for releasing bioactive molecules, Curr. Appl. Phys. 9
[190] M.T. Andani, N. Shayesteh Moghaddam, C. Haberland, D. Dean, M.J. Miller, M. (2009) e259–e262.
Elahinia, Metals for bone implants. Part 1. Powder metallurgy and implant [214] O.V. Khutoryanskaya, M. Potgieter, V.V. Khutoryanskiy, Multilayered
rendering, Acta Biomater. 10 (2014) 4058–4070. hydrogel coatings covalently-linked to glass surfaces showing a potential to
[191] C. Gao et al., Current progress in bioactive ceramic scaffolds for bone repair mimic mucosal tissues, Soft Matter 6 (2010) 551–557.
and regeneration, Int. J. Mol. Sci. 15 (2014) 4714–4732. [215] S. Ladet, L. David, A. Domard, Multi-membrane hydrogels, Nature 452 (2008)
[192] A.M. Ferreira, P. Gentile, V. Chiono, G. Ciardelli, Collagen for bone tissue 76–79.
regeneration, Acta Biomater. 8 (2012) 3191–3200. [216] H.H. Kim, J.B. Park, M.J. Kang, Y.H. Park, Surface-modified silk hydrogel
[193] J. Venkatesan, R. Nithya, P.N. Sudha, S.K. Kim, Role of alginate in bone tissue containing hydroxyapatite nanoparticle with hyaluronic acid-dopamine
engineering, Adv. Food Nutr. Res. 73 (2014) 45–57. conjugate, Int. J. Biol. Macromol. 70C (2014) 516–522.
[194] P. Gentile, V. Chiono, I. Carmagnola, P.V. Hatton, An overview of poly(lactic- [217] S.S. Silva et al., Silk hydrogels from non-mulberry and mulberry silkworm
co-glycolic) acid (PLGA)-based biomaterials for bone tissue engineering, Int. J. cocoons processed with ionic liquids, Acta Biomater. 9 (2013) 8972–8982.
Mol. Sci. 15 (2014) 3640–3659.

You might also like